117
Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection Jens Dinter, Berlin 2015

Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Processing and presentation of epitopes by

dendritic cells and macrophages in HIV

infection

Jens Dinter, Berlin 2015

Page 2: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing
Page 3: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Processing and presentation of epitopes by

dendritic cells and macrophages in HIV infection

vorgelegt von Dipl.-Ing.

Jens Dinter geb. in Berlin

von der Fakultät III - Prozesswissenschaften der Technischen Universität Berlin

zur Erlangung des akademischen Grades

Doktor der Naturwissenschaften - Dr. rer. nat. -

genehmigte Dissertation

Promotionsausschuss:

Vorsitzender: Professor Dr. Juri Rappsilber Gutachter: Professor Dr. Roland Lauster Gutachter: Professor Dr. Jens Kurreck Gutachterin: Professor Dr. Sylvie Le Gall Tag der wissenschaftlichen Aussprache: 27. März 2015

Berlin 2015

Page 4: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Abstract

Human immunodeficiency virus (HIV)-1 infection causes a chronic infection associated with a

progressive CD4+ T cell depletion and dysregulation of the immune system. HIV-1 infects

several CD4-expressing immune cell subsets, including CD4+ T cells, monocytes, dendritic cells

(DCs) and macrophages (Møs). Recognition and killing of these cell subsets by HIV-specific

CD8+ T cells (CTLs) requires the presentation of viral epitopes by MHC I. Whether all HIV-

infectable cell subsets equally process and present the same HIV-1 epitopes remains open, but

is critical for the identification of protective HIV-specific CTL responses that should recognize

and kill all HIV-infectable cell subsets and should be part of a HIV-1 vaccine.

In this study we focus on the processing and presentation of HIV-1 epitopes by DCs and Møs,

which are important in priming and activation of anti-viral immune responses, but also

targeted by HIV-1. We show that proteolytic activities and expression levels of cytosolic

proteases involved in antigen processing significantly differ between monocyte-derived DCs

and Møs matured with TLR ligands or upon HIV-1 infection. These differences affect the

amount and the kinetics of epitope production and lead to differences in CTL responses, which

may contribute to the unequal capacity of HIV-specific CTLs to control viral load.

HIV-1 can enter DCs and Møs by fusion at the plasma membrane for delivery into the cytosol

or by internalization into endo-lysosomal vesicles. How trafficking of HIV-1 through various cell

compartments affects the degradation patterns of HIV-1 peptides and the recognition of

infected cells by immune cells is not understood. Using DC and Mø cell extracts as a source of

proteases from various cell compartments to degrade long HIV-1 peptides, we identify by mass

spectrometry cell-specific and compartment-specific degradation patterns. We show a

preferential production and a superior intracellular stability of peptides containing

immunodominant epitopes corresponding to stronger CTL responses, which may contribute to

the establishment of immunodominance patterns during HIV-1 infection.

During disease progression constant immune pressure on immunodominant epitopes results in

the appearance of frequently detected HLA-associated escape mutations to avoid CTL

recognition. We show for the first time that processing of escape mutations in cross-

presentation-competent cell compartments reduces intracellular stability and impairs epitope

production, which leads to less epitope available for presentation and thus immune escape.

Together these results demonstrate the impact of protein degradation patterns in subcellular

compartments on epitope production for CTL recognition and their contribution to

immunodominance patterns and immune escape during HIV-1 infection.

Page 5: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Zusammenfassung

Eine Infektion durch das humane Immundefizienz-Virus (HIV)-1 führt zu einer chronischen

Infektion einhergehend mit einer kontinuierlichen Reduktion der CD4+ T-Zellen und einer

Fehlregulation des Immunsystems. HIV-1 infiziert mehrere CD4-exprimierende Immunzellen,

unter anderem CD4+ T-Zellen, Monozyten, Dendritische Zellen und Makrophagen. Die

Erkennung und Lyse von HIV-infizierten Zellen durch CD8+ T-Zellen erfordert die Präsentation

viraler Epitope auf MHC I Molekülen. Es ist bisher nicht geklärt, ob alle HIV-infizierten Zellen

gleichermaßen diese viralen Epitope prozessieren und präsentieren, obwohl dies ein wichtiger

Aspekt in der Identifizierung von protektiven CD8+ T-Zellantworten ist, die alle infizierten

Zellen erkennen und lysieren sollen und deswegen Bestandteil eines HIV-Vaccines sein sollten.

Der Fokus dieser Arbeit liegt in der Prozessierung und Präsentation von HIV-1 Epitopen durch

Dendritische Zellen und Makrophagen. Diese Zellen spielen eine wichtige Rolle in der

Aktivierung der Immunantwort gegen HIV-1, werden allerdings auch durch HIV-1 infiziert. Wir

zeigen, dass wichtige Proteasen, die in der Epitop-Prozessierung involviert sind, ihre

Aktivitäten und Expressionslevel durch TLR-Aktivierung oder HIV-1 Infektion von Dendritischen

Zellen und Makrophagen verändern. Diese Veränderungen führen zu unterschiedlichen

Produktionen von HIV-1 Epitopen und dadurch zu unterschiedlichen CD8+ T-Zellantworten.

Dies könnte dazu führen, dass CD8+ T-Zellen HIV-infizierte Dendritische Zellen und

Makrophagen nicht gleichermaßen erkennen und lysieren können und somit nicht die

Ausbreitung der Infektion verhindern können.

HIV-1 kann durch direkte Fusion mit der Plasmamembran oder durch die Aufnahme in Endo-

und Lysosomen in Dendritische Zellen und Makrophagen gelangen. Wie HIV-1 Peptide in

diesen verschiedenen Zellkompartimenten abgebaut werden, ist bisher nicht bekannt. Wir

zeigen mit Hilfe von massenspektrometrischen Analysen, dass Proteasen aus verschiedenen

Zellkompartimenten HIV-1 Peptide unterschiedlich abbauen, wobei Unterschiede zwischen

Zelltypen und Zellkompartimenten zu erkennen sind. Der Abbau von Peptiden bevorzugt die

Produktion immundominanter Epitope unabhängig von dem Zelltyp oder dem

Zellkompartiment und führt dadurch zu einer erhöhten CD8+ T-Zellantwort spezifisch für diese

Epitope. Dies könnte zu der Etablierung immundominater CD8+ T-Zellantworten während einer

HIV-1 Infektion beitragen.

Die konstante Erkennung immundominanter Epitope durch HIV-spezifische CD8+ T-Zellen, führt

zum Auftreten von Mutationen, die dazu führen, dass CD8+ T-Zellen diese Epitope nicht mehr

erkennen können und diese somit der Immunantwort entkommen. Wir zeigen hier zum ersten

Mal, dass die Prozessierung dieser Mutationen in Endo- und Lysosomen zu einer reduzierten

Page 6: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Epitop-Produktion und Epitop-Stabilität führen. Dies führt wiederum dazu, dass weniger Epitop

zur Präsentation zur Verfügung steht und dies eine CD8+ T-Zellantwort verhindert.

Diese Ergebnisse demonstrieren, dass die Prozessierung von HIV-1 Proteinen in verschiedenen

Zellkompartimenten auf die Etablierung von immundominanten CD8+ T-Zellantworten sowie

auf das Entkommen dieser immundominanten Epitope nach Einbau von Mutationen einen

wichtigen Einfluss hat.

Page 7: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Publications

Dinter J., Duong E., Lai N. Y., Berberich M. J., Kourjian G., Bracho-Sanchez E., Chu D., Su H.,

Zhang S. C., Le Gall S. Variable processing and cross-presentation of HIV by dendritic cells and

macrophages shapes CTL immunodominance and immune escape; PLoS Pathogens 2015 Mar

17; 11(3):e1004725

Dinter J.*, Gourdain P.*, Lai N. Y.*, Duong E., Bracho-Sanchez E., Rucevic M., Liebesny P. H.,

Xu Y., Shimada M., Ghebremichael M., Kavanagh D. G. and Le Gall S. Different antigen-

processing activities in dendritic cells, macrophages, and monocytes lead to uneven

production of HIV epitopes and affect CTL recognition; Journal of Immunology 2014 Nov 1; 193

(9):4322-34

Rucevic M., Boucau J., Dinter J., Kourjian G., Le Gall S. Mechanisms of HIV protein degradation

into epitopes: implications for vaccine design; Viruses 2014 Aug 21; 6(8):3271-92

Mattison C., Dinter J., Berberich M., Chung S.-Y., Reed S., Le Gall S., Grimm C. In-vitro

evaluation of digestive and endolysosomal enzymes to cleave CML-modified Ara h 1 peptides;

Food Science & Nutrition 2015, in press

Conference contributions

8th International Workshop on Antigen Processing and Presentation in Philadelphia, USA (June

2014) Poster: “Differential cross-presentation of HIV epitopes by dendritic cells and

macrophages identifies variable intracellular degradation rates of HIV peptides”

Ragon Symposium on Humanized Mouse HIV Vaccine Initiatives in Boston, USA (February

2014) Talk: “Processing of peptide antigens by human antigen presenting cells”

13th Annual Meeting of the Federation of Clinical Immunology Societies (FOCIS) in Boston, USA

(June 2013) Talk: “Variable Cross-Presentation of HIV Epitopes by Dendritic Cells and

Macrophages to CD8 T Cells Uncovers Differences in Intracellular Peptide Degradation Rates”

Keystone Symposium: Understanding dendritic cell biology to advance disease therapies in

Keystone, USA (March 2013) Poster: “Variable cross-presentation of HIV epitopes by dendritic

cells and macrophages to CD8 T cells uncovers differences in peptide degradation rates”

AIDS Vaccine 2012 in Boston, USA (September 2012) Poster: “Variable processing and

presentation of HIV epitopes in dendritic cells and macrophages to CD8 T cells”

99th Annual Meeting of the American Association of Immunologists in Boston, USA (May 2012)

Poster: “Variable processing and presentation of HIV epitopes in HIV-infectable cell subsets”

Page 8: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing
Page 9: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Table of Contents

List of Figures .......................................................................................................................... 4

List of Supplemental Figures .............................................................................................. 6

1. Introduction ..................................................................................................................... 7

1.1. HIV-1 infection ...................................................................................................................... 7

1.2. HIV-1-infectable cell subsets ........................................................................................... 8

1.3. Processing of HIV-1 peptides ........................................................................................ 10

1.3.1. The direct presentation pathway ...................................................................................... 11

1.3.2. The cross-presentation pathways .................................................................................... 13

1.4. Immune response to HIV-1 infection ......................................................................... 16

1.5. Immunodominance patterns in HIV-1 infection .................................................... 17

1.6. Viral escape mutations .................................................................................................... 19

1.7. HIV-1 vaccine design ........................................................................................................ 19

2. Motivation ...................................................................................................................... 21

3. Materials and Methods .............................................................................................. 23

3.1. Materials .............................................................................................................................. 23

3.1.1. Instruments ................................................................................................................................ 23

3.1.2. Chemicals, Solutions ............................................................................................................... 23

3.1.3. Buffers, Media ............................................................................................................................ 24

3.2. Study participants ............................................................................................................. 24

3.3. Cell culture .......................................................................................................................... 25

3.3.1. Generation of monocyte-derived DCs and Møs ........................................................... 25

3.3.2. Culture of epitope-specific CD8+ T cell clones ............................................................. 25

3.4. Analysis of the antigen processing machinery in DCs and Møs ........................ 25

3.4.1. Preparation of cell extracts .................................................................................................. 25

3.4.2. Fluorescent measurement of hydrolytic activities .................................................... 26

3.4.3. Analysis of the antigen processing machinery by Western blot .......................... 28

3.5. In vitro processing of HIV peptides ............................................................................ 29

3.5.1. In vitro peptide degradation assay ................................................................................... 30

3.5.2. Analysis of the antigenicity of the degradation products ....................................... 30

3.5.3. Identification and semi-quantification of degradation products by MS ........... 30

3.5.4. Intracellular peptide stability assay ................................................................................. 31

3.5.5. Quantification of epitope degradation by reversed-phase HPLC analysis ....... 31

Page 10: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Table of Contents 2

3.6. Cross-presentation of exogenous HIV proteins by DCs and Møs ..................... 33

3.6.1. ELISPOT cross-presentation assay ................................................................................... 33

3.6.2. P24 uptake assay ..................................................................................................................... 33

3.7. Statistical analysis ............................................................................................................ 34

4. Results ............................................................................................................................. 35

4.1. Hydrolytic activities of cytosolic proteases involved in antigen processing in

DCs and Møs .................................................................................................................................... 35

4.1.1. Proteasomal hydrolytic activities in DCs and Møs change differently upon

maturation with TLR4 or TLR7/8 ligands ..................................................................................... 35

4.1.2. Hydrolytic activities of cytosolic aminopeptidases and TPP II are comparable

in DCs and Møs, whereas TOP activities are significantly higher in Møs .......................... 36

4.1.3. TLR-mediated maturation of DCs and Møs results in early changes of

proteasomal activities and reflect changes detected upon HIV infection ........................ 38

4.2. Expression of cytosolic and ER-resident proteases involved in antigen

processing in DCs and Møs ........................................................................................................ 40

4.2.1. Higher expression levels of the catalytical and structural subunits of the

constitutive and immunoproteasome in Møs compared with DCs ..................................... 40

4.2.2. Møs have higher expression levels of cytosolic TOP and ER-resident ERAP1

and ERAP2 compared to DCs .............................................................................................................. 42

4.3. HIV-1 epitope processing by cytosolic peptidases in DCs and Møs................. 43

4.3.1. Proteasomal activities in cell extracts of DCs and Møs reflect those measured

in intact, live cells ..................................................................................................................................... 43

4.3.2. The kinetics and amounts of optimal HIV-1 epitopes produced in cytosolic

extracts of DCs and Møs vary among epitopes ............................................................................ 44

4.4. Processing of HIV-1 peptides by cytosolic and endo-lysosomal peptidases in

DCs and Møs .................................................................................................................................... 50

4.4.1. Lysosomal activities in live intact DCs and Møs correlate with activities

measured in cell extracts ...................................................................................................................... 50

4.4.2. Preferential production of immunodominant epitopes KF11 and TW10 in

cytosolic and endo-lysosomal cell extracts ................................................................................... 52

4.5. Cross-presentation of HIV-1 proteins by DCs and Møs ........................................ 56

4.5.1. Cross-presentation of the immunodominant Gag p24 B57-TW10 and B57-

KF11 epitopes is more efficient than that of subdominant B57-ISW9 epitope.............. 56

4.5.2. Protease activities in cross-presentation competent cell compartments

differently affect processing of HIV-1 epitopes in DCs and Møs .......................................... 58

Page 11: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Table of Contents 3

4.5.3. Low degradation of RK9-containing fragments results in high amounts of

HLA-A03 RK9 epitope cross-presented by DCs and Møs ........................................................ 60

4.6. Intracellular stability of HIV-1 epitopes in cytosolic and lysosomal cell

extracts ............................................................................................................................................. 62

4.6.1. Intracellular epitope stability of HIV-1 epitopes in cytosolic and lysosomal

cell extracts follows CTL responses hierarchy ............................................................................. 62

4.7. The impact of HIV-1 escape mutations on epitope production and peptide

stability in the cross-presentation pathway ....................................................................... 64

4.7.1. Frequent escape mutations in immunodominant epitopes reduce epitope

production and peptide stability in the cross-presentation pathway ................................ 64

5. Discussion ...................................................................................................................... 67

5.1. Antigen processing in HIV-infectable cell subsets ................................................ 67

5.2. Viral entry and intracellular trafficking of antigens ............................................ 69

5.3. Viral recognition and cell activation upon HIV-1 infection ............................... 70

5.4. Epitope processing in different subcellular compartments .............................. 72

5.5. Cross-presentation contributes to immunodominance patterns .................... 74

5.6. The impact of viral escape mutants on cross-presentation............................... 75

5.7. Impact of antigen processing and cross-presentation on vaccine design .... 76

6. References ..................................................................................................................... 80

7. Supplemental Figures .............................................................................................. 100

Page 12: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

List of Figures 4

List of Figures

Figure 1. Overview of the HIV-1 replication cycle. ........................................................................ 8

Figure 2. Trafficking of antigens for processing and presentation by MHC I or MHC II molecules.

............................................................................................................................................ 11

Figure 3. Regulation of the pH during maturation of early endosomes to lysosomes. ............... 14

Figure 4. HIV-1 infection and disease progression. ..................................................................... 16

Figure 5. Representation of the fluorescence based protease activity assay. ............................ 26

Figure 6. Schematic representation of the in vitro degradation assay. ...................................... 29

Figure 7. Defining the intracellular stability of peptides by RP-HPLC quantification. ................. 32

Figure 8. Proteasomal activities in monocyte-derived DCs and Møs change differently upon

maturation with TLR4 or TLR7/8 ligands. ........................................................................... 36

Figure 9. DCs and Møs have comparable aminopeptidases and TPP II activities, whereas TOP

activities are higher in Møs. ............................................................................................... 37

Figure 10. Changes in antigen processing activities in maturing monocyte-derived DCs reflect

changes observed upon HIV infection. ............................................................................... 39

Figure 11. Higher expression levels of proteins involved in antigen processing in Møs compared

to DCs. ................................................................................................................................ 40

Figure 12. Higher expression levels of catalytically active and structural proteasome subunits in

Møs compared to DCs. ....................................................................................................... 41

Figure 13. Comparable expression levels of cytosolic post-proteasomal activities in DCs and

Møs. .................................................................................................................................... 42

Figure 14. Changes in proteasomal activities in cell extracts reflect those observed in live, intact

DCs and Møs. ...................................................................................................................... 44

Figure 15. Mø cell extracts produce HLA-A11-ATK9 epitope more efficiently than DCs. ............ 45

Figure 16. Mø cell extracts produce more A11-ATK9 epitope and epitope precursors compared

to DC cell extracts. .............................................................................................................. 46

Figure 17. Higher amounts of HLA-A11 ATK9 produced by Mø cell extracts resulted in higher

lysis of target cells by ATK9-specific CTLs. .......................................................................... 47

Figure 18. Extracts from DCs and Møs generate similar amounts of Nef-derived antigenic

peptides at similar rates. .................................................................................................... 49

Figure 19. Lysosomal activities in DC and Mø cell extracts reflect activities in live, intact cells

and can be activated at different pH values....................................................................... 51

Page 13: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

List of Figures 5

Figure 20. The immunodominant epitope B57-KF11 is more efficiently produced in cross-

presentation-competent compartments than subdominant epitope B57-ISW9. ............... 53

Figure 21. Major cleavage sites within p24-35mer destroy the B57-ISW9 epitope. ................... 54

Figure 22. Slow degradation of TW10-containing peptides results in high amounts of B57-TW10

available for direct or cross-presentation. ......................................................................... 55

Figure 23. The immunodominant HLA-B57-restricted TW10 and KF11 epitopes are more

efficiently cross-presented than subdominant ISW9 epitope. ............................................ 57

Figure 24. The amount of exogenous p24 protein internalized by DCs and Møs affects the

amount of epitope cross-presented to CTLs. ...................................................................... 58

Figure 25. Inhibition of proteasomes increases B57-KF11 and B57-ISW9 epitope cross-

presentation by DCs but not Møs. ...................................................................................... 59

Figure 26. The RK9-containing fragment shows minor cleavage sites by cytosolic or endo-

lysosomal proteases. .......................................................................................................... 60

Figure 27. Limited degradation of RK9-containing fragments results in cross-presentation of

high amounts of HLA-A03 RK9 epitope. ............................................................................. 61

Figure 28. Variable stability of B57-KF11 and A03-RK9 in cytosolic and lysosomal cell extracts of

immature DCs and Møs. ..................................................................................................... 62

Figure 29. The intracellular stability of HIV-1 epitopes in the cytosol and lysosomes follows CTL

responses hierarchy. ........................................................................................................... 63

Figure 30. Escape mutations in immunodominant epitopes reduce epitope production in the

cross-presentation-competent cell compartments. ........................................................... 65

Figure 31. Reduced intracellular stability of TW10 escape mutants in cytosolic and lysosomal

cell extracts. ........................................................................................................................ 66

Page 14: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

List of Supplemental Figures 6

List of Supplemental Figures

Supplemental Figure 1. Phenotypic analysis of DCs and Møs cell populations......................... 100

Supplemental Figure 2. Infection of immature DCs and Møs with VSVg ΔEnv NL4.3. .............. 101

Supplemental Figure 3. Degradation patterns of a HIV-1 RT 16-mer fragment in cytosolic

extracts of immature and mature DCs and Møs. ............................................................. 102

Supplemental Figure 4. Degradation of a HIV-1 Gag p24 35mer in DC cell extracts at pH4.0,

pH5.5 and pH7.4. .............................................................................................................. 103

Supplemental Figure 5. Variable production of 16 HIV-1 epitopes in cytosolic and endo-

lysosomal cell extracts of DCs and Møs. ........................................................................... 104

Supplemental Figure 6. Minor cleavage sites within p24-31mer results in less degradation of

B57-TW10-containing peptides. ....................................................................................... 105

Page 15: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Introduction 7

1. Introduction

1.1. HIV-1 infection

According to estimates by WHO and UNAIDS, 35 million people were living with human

immunodeficiency virus (HIV)-1 globally at the end of 2013. That same year, approximately 2.1

million people became newly infected, and 1.5 million died of AIDS-related causes.

HIV-1 infects several CD4-expressing immune cell subsets, including CD4+ T cells, monocytes,

Møs and DCs. HIV-1 enters cells by specific interactions of the virus and the receptor CD4 and

co-receptors CCR5 and CXCR4 on the target cell. After binding to co-receptors, viral and

cellular membranes fuse and the viral core is released into the cytoplasm of the cell, where

viral RNA is retro-transcribed and provirus is integrated into the cellular genome. Both viral-

and host-cell machineries are necessary for transcription of the viral genes using the

integrated provirus as template. Upon viral protein synthesis, the virus assembles and budding

of new virions occurs (Figure 1). Alternatively, viral fusion via the endosomal pathway has been

shown to constitute a possible mode of virus entry and may depend on the cell type and the

rate of endocytosis (1, 2).

The clinical course of HIV-1 infection in the absence of treatment shows an initial high peak of

plasma viral loads within the first 6 weeks of transmission, which spontaneously decreases as

HIV-1-specific immune responses develop. Already at this early time point of infection, HIV-1

disseminates to the lymphoid organs, and establishes viral reservoirs and latency throughout

the body. In the majority of HIV-1 infected individuals, the immune system ultimately fails to

control the infection, resulting in loss of CD4+ T cells, increasing plasma viral loads and

development of AIDS.

Multiple drugs have been developed to inhibit HIV-1 viral replication and to reverse the decline

in CD4+ T cell counts in HIV-1 infected individuals. Several steps in the HIV-1 replication cycle

are targeted by this highly active antiretroviral therapy (HAART). The three major groups of

drugs being used in clinics are inhibitors of the reverse transcriptase (nucleoside/nucleotide,

NRTI, and non-nucleoside, nNRTI), integrase inhibitors and protease inhibitors (Figure 1, shown

in green). In the majority of HIV-1 infected individuals treated with antiretroviral drugs plasma

HIV-1 viral loads drop significantly and CD4+ T cell counts increase, which can be maintained

for years. However, HAART does not fully restore the immune system and is associated with

considerable side effects and the development of resistances. Therefore, the design of an

effective HIV-1 vaccine is still the only way to control viral infection in the absence of HAART.

Page 16: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Introduction 8

Figure 1. Overview of the HIV-1 replication cycle.

HIV-1 binds to CD4 and the appropriate co-receptor (CCR5 or CXCR4) resulting in fusion of the

viral envelope and the cellular membrane and the release of viral nucleocapsid into the

cytoplasm. Following the uncoating viral RNA is reverse transcribed by the reverse

transcriptase, integrates into the cellular DNA by the integrase, transcribed by the cellular RNA

polymerase II and the resulting mRNA is exported to the cytoplasm and translated into viral

proteins. Viral proteins and genomic RNA are transported to the cellular membrane and

assembled. Immature virions are released. Polypeptide precursors are processed by the viral

Protease to produce mature viral particles. Several host derived restriction factors can inhibit

HIV-1 replication at different stages of the replication cycle (shown in red). HIV-1 has evolved to

counteract these innate antiviral factors by expression of viral proteins: the viral capsid for

TRIM5α, Vpu/Nef/Env for tetherin, Vif for APOBEC-3G, and Vpx for SAMHD1 (shown in blue).

The major families of antiretroviral drugs and the step during the replication cycle that they

block are indicated (shown in green) (3).

1.2. HIV-1-infectable cell subsets

CD4+ T cells, Møs and DCs patrolling the mucosal surface are the first immune cells to

encounter HIV-1. CD4+ T cells can be divided into several subpopulations with diverse cytokine

secretion patterns, which recruit and activate other immune cells including B cells, CD8+ T cells

and Møs (4). CD4+ T cell subpopulations reside within the gastrointestinal tract, the lymph

nodes, other lymphatic tissues and the blood. During the acute phase of HIV-1 infection a

dramatic loss of CD4+ CCR5+ T cells in the gastrointestinal tract can be detected (5), while the

chronic phase of infection, characterized by a massive production of proinflammatory

cytokines, is characterized by a gradual loss of peripheral CD4+ T cells. Despite being a main

target by HIV-1, previous studies showed that HIV-1 Gag-specific CD4+ T cell responses

Page 17: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Introduction 9

increased after acute HIV-1 infection in individuals who control HIV-1 infection (6) and were

significantly associated with control of viral replication (7, 8). However, it has been shown that

these HIV-specific CD4+ T cells are preferentially infected, rather than CD4+ T cells specific for

other antigens (9).

DCs represent an important subset in HIV-1 infection, as they affect viral transmission and

presentation of HIV-1 antigens. DCs can modulate antiviral immune responses by presentation

of MHC I and MHC II epitopes to CD8+ and CD4+ T cells and secret pro-inflammatory cytokines

and interferons to alter T cell proliferation and differentiation. DCs express high levels of the

HIV-1 entry receptors CCR5 and CXCR4, as well as low levels of CD4, allowing gp120 binding

and attachment of HIV-1 virions. Moreover, specific DC subsets express several receptors that

can bind HIV-1 gp120 (10), such as Langerin on Langerhans cells in the skin, DC

immunoreceptor (DCIR) on conventional DCs in the lamina propria, or DC-specific intercellular

adhesion molecule-grabbing nonintegrin (DC-SIGN) and the mannose receptor on dermal DCs.

DCs express high levels of restriction factors, such as SAMHD1, thus only a small proportion of

cells is productively infected (11). Besides productive infection, replication and formation of

new virions for infection of CD4+ T cells (termed cis-infection) DCs can promote infection to

CD4+ T cells in trans. C-type lectins expressed by DCs capture HIV-1 and migrate to CD4+ T cell

enriched lymphoid tissues, where HIV-1 trans-infection of active CD4+ T cells occurs and

facilitates viral dissemination. This direct non-replicative trans-infection across the infectious

synapse allows virions to be rapidly transferred when DCs and CD4+ T cells are in close contact

at the infectious synapse.

Møs can take up viral particles or virions for processing and presentation by MHC II to CD4+ T

cells or cross-presentation by MHC I to CD8+ T cells, which may be crucial for activation of

antiviral immune responses. A significant proportion of Møs at the mucosal surface is

productively infected with HIV-1 (12) and infection results in only limited virus production. In

contrast to CD4+ T cells, Møs are more resistant to the cytopathic effects of HIV-1 and better

able to evade the host immune response. The two major cellular reservoirs for HIV-1 are

therefore latently infected resting CD4+ T cells and Møs. Especially long-lived Møs accumulate

replication-competent HIV-1 for prolonged periods, even in patients receiving HAART (13).

Monocytes are composed of several subpopulations with the majority of monocytes in healthy

individuals expressing CD14 but not CD16. During infection the number of circulating

CD14+CD16+ monocytes increases dramatically. This CD16+ monocytes subset is preferentially

targeted by HIV-1 (14). Relative to CD4+ T cells, CD14+ monocytes are more resistant to

productive HIV-1 infection as a result of differential expression levels of several restriction

Page 18: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Introduction 10

factors, including SAMHD1 or APOBEC-3G (15). In contrast, CD16+ monocytes express less

APOBEC-3G in line with higher infectivity of this subset.

Understanding which HIV-1 epitopes are presented by all infectable cell subsets is essential to

evaluate the capacity of immunogens to generate T cell responses specific for those epitopes

in order to recognize and clear all infected cells.

1.3. Processing of HIV-1 peptides

All nucleated cells express MHC I molecules on their surface, while professional antigen-

presenting cells, including DCs, Møs and B cells, additionally express MHC II molecules on their

surface. The recognition and killing of HIV-infected cells requires the presentation of adequate

amounts of peptide-loaded MHC I molecules to HIV-specific CTLs. Therefore, it is important to

analyze the capacity of all HIV-infectable cell subsets to process and present MHC I-restricted

epitopes. Viral epitopes originate from HIV-1 proteins undergoing intracellular processing.

Endogenous antigens within the cytoplasm are mainly processed by the proteasome before

transportation into the endoplasmic reticulum (ER) via the transporter associated with antigen

processing (TAP). In the ER peptides can be further trimmed before loading onto MHC I

molecules for presentation to CD8+ T cells. MHC I peptides are usually 8-11 amino acids long

and confined by binding groove interactions at both the N- and C-termini.

In contrast, exogenous antigens are internalized into the endosomal/lysosomal compartment,

where antigens are processed by different cathepsins and other proteases. Peptides of usually

12-15 amino acids in length can then be loaded onto MHC II molecules (and even overlay the

MHC II binding groove) and presented to CD4+ T cells. Given the important role of CD4+ T cells

in providing help for CD8+ T cells and B cells (16) or even direct antiviral effects (17) it is crucial

to understand the processing of MHC II peptides in the endo-lysosomal compartments.

Despite the differences in the source of antigen and the processing pathways, exogenous

antigens can be translocated into the cytosol for further processing and presentation onto

MHC I, a process known as cross-presentation (Figure 2).

Page 19: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Introduction 11

Figure 2. Trafficking of antigens for processing and presentation by MHC I or MHC II

molecules.

Cytosolic proteins, such as viral or endogenous proteins, alternative transcripts or defective

ribosomal products (DRiPs), are processed primarily by the proteasome into shorter peptides.

These peptides can be further trimmed by cytosolic post-proteasomal peptidases before

transportation into the endoplasmic reticulum (ER) by the transporter associated with antigen

processing (TAP) for subsequent trimming and assembly by MHC I molecules. In professional

antigen-presenting cells (APCs) exogenous antigens can be internalized into endosomes and

lysosomes for processing by various cathepsins and presentation by MHC II to CD4+ T cells.

Alternatively, exogenous antigens can be translocated into the cytoplasm for further processing

by cytosolic proteases and MHC I loading in the ER, a phenomenon known as cross-

presentation. Loading of MHC II occurs in mature or late endosomal compartments, specialized

in processing and loading onto MHC II molecules.

1.3.1. The direct presentation pathway

In the cytosolic antigen-processing pathway proteins destined for presentation on MHC I are

primarily degraded by the proteasome (18-20). Different sources of antigen can be degraded,

such as cryptic transcriptional products (21), defective ribosomal products (DRiPs) (22, 23),

endogenous mature proteins, or exogenous antigens preprocessed in endo- and lysosomes.

The 26S proteasome is composed of a core cylinder, the 20S proteasome, which is capped at

each end by a 19S complex. The 20S proteasome consists of a barrel-shaped core structure

containing four stacked rings of seven subunits each. The outer rings are composed of alpha-

subunits and the middle two beta-subunits, three of which beta1 (chymotrypsin-like), beta2

(tryptic-like), and beta5 (caspase-like) constitute the active proteolytic components. The 19S

component has deubiquitinase and an unfoldase activity that allows the targeted proteins to

Page 20: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Introduction 12

enter the channel in the center of the barrel where the beta-subunit active sites reside (24).

The hydrolytic activities within the 26S proteasome cleave after hydrophobic, acidic, or basic

residues, respectively and therefore cleave the protein into many diverse oligopeptides,

although the precise location of the cleavages vary widely with each molecule degraded.

Whereas 26S proteasomes degrade mostly polyubiquitinated substrates, 20S proteasomes

capped with regulatory PA28/11S lids are able to degrade non-polyubiquitinated substrates.

Therefore, hydrolysis of proteins by proteasomes is the key step in the generation of most

antigenic peptides (24).

HIV-1 infection triggers the massive production of proinflammatory cytokines and type I and II

interferons (25, 26). Upon interferon-gamma, TNF-alpha or LPS stimulation, the three catalytic

beta-subunits of the constitutive proteasome are replaced by immunoproteasome subunits,

beta1i, beta2i, and beta5i, which have been shown to differ in cleavage specificities compared

to the constitutive subunits (27). Immunoproteasomes can also be constitutively expressed in

lymphoid tissues. Immunoproteasomes exhibit increased rates of substrate turnover with

enhanced cleavage after hydrophobic and basic amino acids, correlating to the preferred C-

terminal residues of many MHC I ligands (28, 29) and generate more N-terminally extended

peptides (30). The presence of both proteasomes and immunoproteasomes in the same cell is

associated with a greater variety of peptides available for presentation (31). Even though

constitutive and immunoproteasomes generate peptides with appropriate C-terminals they

actually destroy more epitopes than they generate (28, 32).

N-terminally extended peptides produced by the proteasomes require trimming by cytosolic

post-proteasomal peptidases, such as leucine aminopeptidase, tripeptidyl peptidase II (TPPII),

thimet oligopeptidases (TOP), puromycin sensitive aminopeptidase (PSA) and several others

(33) before transportation into the ER. Cytosolic peptidases cannot unfold proteins and

therefore degrade post-proteasomal degradation products. TPPII has been shown to generate

epitopes in a proteasome-independent manner by removing three residues at a time from the

N-terminus of peptides of more than 15 residues in lengths (34). Despite the contribution of

several cytosolic aminopeptidases to the generation of individual epitopes, these peptidases

also have an overall destructive effect on epitope generation (35, 36). A small fraction of the

peptides produced by proteasomes and post-proteasomal peptidases escape destruction in

the cytosol and are transported by TAP into the ER for loading onto MHC I. Thus, peptide

stability is an important parameter that has been shown to contribute to the amount of

epitope available for presentation (37, 38).

TAP transports peptides that range in length from about 7 to more than 20 amino acids and

can therefore import both mature epitopes and longer precursors into the ER (39). TAP

Page 21: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Introduction 13

deficient cells showed decreased MHC I surface expression levels indicating the importance of

TAP in antigen presentation and MHC surface expression. In the ER, ER-resident

aminopeptidases, such as ERAP1 and ERAP2 are able to remove from the N-terminus of

peptides nearly all amino acids except those followed by proline prior to MHC I loading and

presentation (40-43). Thus, N-terminal trimming occurs in both, the cytoplasm and the ER.

Peptide-MHC (pMHC) class I complexes are then transporter to the cell surface for

presentation and activation of CD8+ T cells with a cognate receptor.

Together, differences in the antigen-processing machinery among HIV-infectable cell subsets

may affect the timing and the amount of epitopes presented to HIV-specific CTLs, which will

affect the capacity of CTLs to recognize and kill all infected cells.

1.3.2. The cross-presentation pathways

Cross-presentation has been demonstrated to play an important role in the induction of

immune responses against viruses that do not infect APCs, viruses that impair direct

presentation of antigens upon infection or viruses that exhibit strict tissue tropism. HIV-1 can

enter cells by direct fusion at the plasma membrane or by endocytosis into the endocytic

system. In addition, vaccines, including viral vectors, peptides bound to nanoparticles or viral

particles, enter DCs by endocytosis. Whether processing of antigens by the cross-presentation

pathway versus the direct presentation pathway results in the presentation of the same

epitopes with comparable amounts and kinetics is still unknown. This aspect is important for

the design of a HIV vaccine, as processing of vaccines in the cross-presentation pathway should

result in the presentation of epitopes that match those presented by all HIV-infectable cell

subsets. In general, the cross-presentation pathway can be differentiated into two main

pathways: The cytosolic pathway and the vacuolar pathway.

1.3.2.1. The cytosolic cross-presentation pathway

Exogenous antigens, such as proteins, apoptotic cells or antibody-coated viruses can be

internalized by multiple mechanisms, including receptor-mediated endocytosis via clathrin-

coated vesicles, macropinocytosis, or phagocytosis. In the endocytic system antigens undergo

gradual proteolytic degradation by various cathepsins and other proteases including insulin-

regulated aminopeptidase (IRAP) (44, 45) along their journey from early endosomes and

phagosomes to lysosomes. In the cytosolic cross-presentation pathway, antigens are

translocated from endosomes or phagosomes into the cytosol for further degradation by the

proteasome (46, 47). After proteasomal degradation, antigen-derived peptides are transported

by TAP into the ER for further trimming and loading onto MHC I.

Page 22: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Introduction 14

Considering that DCs pick up antigen in the peripheral tissue and migrate for several hours to

lymph nodes (48), cross-priming of naïve CD8+ T cells in lymph nodes requires continuous

cross-presentation of epitopes. Thus prolonged antigen stability and low degradation is an

important prerequisite. It has been shown that limited antigen degradation correlates with

efficient cross-presentation (49, 50), which has been supported by findings showing that

artificially delivered antigens to highly degradative, late endocytic compartments by

lipososomes are presented on MHC II more efficiently than on MHC I (51).

Most lysosomal proteases have an optimal proteolytic activity between pH5.5 and 6.5 (52).

After internalization of exogenous antigens phagosomes fuse first with early and late

endosomes to acquire the lysosomal proteases and the proteins responsible for acidification

(Figure 3).

Figure 3. Regulation of the pH during maturation of early endosomes to lysosomes.

Exogenous antigens are internalized into early endosomes, which display a high pH and a low

degradation environment. These are probably the compartments that allow antigen escape to

the cytosol and most likely the site of peptide loading onto MHC I in the vacuolar pathway. As

endosomes and phagosomes mature, the pH drops and the levels of proteolysis increase. The

compartments become incompetent for cross-presentation, but enable peptide loading onto

MHC II.

In DCs several mechanisms have been described to preserve and retain antigen in the

endocytic system. First, DCs express lower levels of most lysosomal proteases than Møs (49)

and are less recruited to phagosomes (53). As a result, DCs degrade internalized antigens at

much lower rates than other phagocytes. Second, it has been shown that the pH in

phagosomes and endosomes in DCs is less acidic than in Møs, which results in lower activation

Page 23: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Introduction 15

of lysosomal proteases thus increasing cross-presentation (54). In DCs phagosomal pH is

approximately pH7.5 in contrast to Møs, which rapidly acidify their phagosomes, reaching

values of 4.5-5 following phagocytosis of inert latex beads (55). In this acidic and highly

proteolytic environment it is very unlikely that peptides are loaded onto MHC I. Endosome

acidification is mediated by the vacuolar ATPase (V-ATPase), which transports protons from

the cytosol into the endosomes. In DCs low levels of V-ATPase activity due to its incomplete

assembly (56), results in a neutral endosomal pH, which prevents rapid antigen degradation

and enhanced cross-presentation. In addition, high levels of activity of NADPH oxidase 2

(NOX2) in DCs, which mediates the generation of reactive oxygen species (ROS) causes the

consumption of protons and thereby the alkalinization of the endocytic compartments (49,

55). Taken together, prolonged MHC I-restricted presentation requires antigen transport into

specialized cell compartments, where high pH and low endocytic protease activity protects

antigens from extensive degradation and allows continuous processing of peptides for loading

onto MHC I.

1.3.2.2. The vacuolar pathway

In the vacuolar pathway, which is also termed TAP-independent cross-presentation pathway,

internalized antigens remain in endosomes and lysosomes, where they are degraded by

endocytic proteases such as cathepsin S. After degradation antigenic peptides are loaded onto

MHC I, which recycles from the cell membrane (57, 58) (Figure 3). The acidic environment in

these endosomes might allow peptides to dissociate from the MHC I molecules, enabling the

peptides processed in the endosomes to bind to MHC I molecules. Within endosomes, IRAP

has been shown to be involved in the trimming of antigens before loading onto MHC I (44).

IRAP displays a broader pH optimum compared to the ER-resident aminopeptidase ERAP,

allowing IRAP activity at a slightly acidic pH (59). IRAP activity might ensure that antigen-

derived peptides are trimmed to their optimal size for loading onto MHC I molecules.

This indicates that trafficking of an antigen through subcellular compartments harboring

different proteases may affect the degradation patterns and thus the amount of epitope

available for presentation.

Page 24: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Introduction 16

1.4. Immune response to HIV-1 infection

Figure 4. HIV-1 infection and disease progression.

HIV-1 infection leads to high levels of HIV-1 replication within the first 6 weeks of infection.

These high viral loads result in the generation of HIV-1-specific CD4+ and CD8+ T cell responses

within 4-6 weeks of infection. Within 1-3 months post infection the appearance of HIV-specific

antibodies indicates sero-conversion. During the subsequent asymptomatic phase of infection

blood CD4+ T cell counts decline slowly, even though successful HAART treatment can partially

reverse this loss. The onset of AIDS is characterized by a progressive loss in CD4+ T cells and

high plasma virus loads. Adapted from (60).

The majority of individuals infected with HIV-1 develop an acute viral syndrome within the first

three weeks of infection, characterized by fever, lymphadenopathy, and cutaneous rash in the

presence of very high levels of HIV-1 replication (61). These high viral loads result in the

generation of innate and adaptive immune responses, including the appearance of HIV-1-

specific CD4+ and CD8+ T cell responses within 4-6 weeks of HIV-1 infection and HIV-1-specific

antibodies, indicating sero-conversion, typically within 1-3 months after HIV-1 infection.

Subsequent to the development of HIV-specific immune responses, HIV-1 RNA levels decline

and eventually stabilize at the so-called viral set point. This viral set point is a strong predictor

for HIV-1 disease progression (62). The time between HIV-1 infection and the establishment of

the viral set point, usually between 3-6 months, is termed primary HIV-1 infection. With

reduction in plasma viral load, CD4+ and CD8+ T cell counts start to normalize, but neither

return to pre-infection levels as CD4+ T cell counts remain reduced and CD8+ T cell counts

remain elevated. However, HIV-specific immune responses fail to eliminate the virus leading to

a chronic infection in most individuals during an asymptomatic period that may persist for

months or years. During the chronic phase of infection, the blood CD4+ T cell counts decline

slowly, even though this loss can be partially reversed by successful HAART, until progressive

CD4+ T cell decline results in the onset of AIDS (Figure 4). The appearance of HIV-1-specific

CD8+ T cell responses has been shown to be temporally associated with the initial decline of

Page 25: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Introduction 17

HIV-1 viral load during primary infection, the resolution of clinical symptoms of the acute viral

syndrome and the rapid selection of viral CTL escape variants. These findings demonstrate an

important role of HIV-specific CD8+ T cells in the control of HIV-1 replication and indicate that

HIV-specific CD8+ T cells will most likely be important for HIV-1 vaccines, if we can identify

protective immune responses that can recognize and kill all HIV-infectable cell subsets.

1.5. Immunodominance patterns in HIV-1 infection

A central feature of many anti-viral T cell responses is the phenomenon of immunodominance.

Despite the expression of 3-6 different MHC I molecules in APCs and the potential generation

of thousands of distinct viral peptides, a large proportion of the anti-viral CTL responses tend

to be dominated by certain class I alleles presenting only a small number of viral peptides.

These CTL responses can be ordered into highly reproducible hierarchies as shown for HIV-1

(62-64), vaccinia virus (65), cytomegalovirus (CMV) (66) and West Nile virus (67).

Immunodominance is generally defined by ELISPOT assays as the most frequent responses in a

population sharing the same HLA allele or in an individual, as the response that induces the

strongest production of IFN-gamma (68, 69). In acute HIV-1 infection a predictable hierarchy of

narrow CD8+ T cell responses can be detected with consistent recognition of certain epitopes

and lack of recognition of others, whereas a broader range of responses has been identified in

chronic infection (63, 70, 71). Previous studies indicated that the immunodominance patterns

of HIV-specific CTL responses developed in primary HIV-1 infection affect the initial control

over viral replication (63, 64, 72) and strongly associate with the subsequent set point of viral

replication (62). However, rising HIV-1 replication in the chronic phase of infection is usually

paralleled by a disintegration of the original CD8+ T cell immunodominance patterns (73-75).

Moreover, other studies showed that immunodominant CTL responses were not always

associated with protection whereas some subdominant responses were associated with lower

viral load (76). Therefore, an effective HIV-1 vaccine should aim to break immunodominance

patterns observed in HIV-1 infection rather than mimicking them. This requires a better

understanding of the mechanisms that contribute to immunodominance. Several factors have

been implicated in shaping immunodominance patterns. Immunodominance can be associated

with the kinetics of protein production, as viral proteins expressed early after infection may

offer an earlier source of epitopes than viral proteins expressed at a later stage. However, in

HIV-1 infection, proteins synthesized later during the viral replication cycle, such as Gag, have

been shown to contribute to dominant CTL responses, although other studies showed that Gag

from incoming virions can be degraded prior to viral protein synthesis and thus lead to Gag-

specific CTL responses (77). Moreover, previous studies with HIV-1 have shown that the

Page 26: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Introduction 18

efficiency of HIV-epitope processing contributes to immunodominance. Degradation of long

peptides in PBMC extracts and purified proteasomes led to the preferential production of

dominant epitopes and its precursors whereas subdominant epitopes were degraded (78, 79).

This preferential processing has been shown to be sequence-dependent, which suggest that

the capacity of proteins to generate epitopes is governed by specific motifs. More recent

studies reported that the absence of proteasomal components is detrimental on the pool of

peptides generated and led to different production of viral peptide pools in comparison to WT

mice during LCMV infection (80). Furthermore, mice deficient in proteasomal catalytic

subunits, LMP2 and MECL1, showed reduced anti-viral CTL responses to immunodominant

influenza epitopes (81), whereas in LMP7-/- mice the overall presentation of this

immunodominant epitope was not affected. These findings indicated a direct impact of the

processing of peptides by proteasomes on the generation of immunodominant epitopes. If

processing of antigens in the cross-presentation pathway contributes to immunodominance

remains open.

TAP-dependent transport of peptides into the ER also plays an important role in

immunodominance. Peptides missing the correct C-terminal amino acid or being longer than

the preferred length for TAP binding are not transported into the ER lumen. Moreover,

mutations within epitopes or flanking sequences have been demonstrated to affect binding

affinity to TAP leading to an overall reduced anti-viral CTL response to peptides that would

have been normally immunodominant (78). Furthermore, studies in ERAP-1-/- mice have shown

the generation of different LCMV-specific CTL responses compared to WT mice infected with

the same virus. The production of ERAP-1 dependent epitopes was impaired, whereas ERAP-1-

independent epitopes or those usually destroyed by ERAP-1 increased in presentation to CTLs,

which affected immunodominance hierachies (82). Similar results for immunodominant HIV-

derived epitopes were observed in other studies (78).

The expression of specific MHC I molecule combinations and their level of expression

determine which epitopes will be recognized by specific CTLs and therefore contribute to

immunodominance. In HIV-1 infection, immunodominant responses are preferentially

restricted by HLA B alleles, especially HLA-B27 or HLA-B57 (64, 83). In this case CTL responses

restricted by other alleles than HLA-B27 or -B57 are always subdominant even though they

would be dominant in persons lacking HLA-B27 or -B57 (62, 84). Moreover, peptides with

highest binding affinity for a given HLA allele may favor peptide loading and a longer display of

epitopes at the cell surface and thus an earlier and prolonged interaction with CD8+ T cells (85,

86). Binding affinity of the peptide-MHC complex for the TCR may affect interaction with CD8+

T cells and contributes to immunodominance patterns. Previous studies demonstrated that

Page 27: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Introduction 19

high affinity of epitope-MHC I complexes for the TCR may prolong the interaction with

epitope-specific CTLs and contribute to immunodominance (86, 87). The number of T cell

precursors and the ability of these cells to proliferate and persist over time may also

contribute to immunodominance as shown previously (88).

1.6. Viral escape mutations

An important factor limiting the success of HIV-specific CTL responses to clear infection is the

capacity of HIV-1 to mutate targeted epitopes to evade CTL recognition (89). Viral escape

mutations occur during the acute and chronic phase of HIV-1 infection (90, 91). Escape

mutations can affect several steps during epitope production and presentation to CTLs.

Impaired production of epitope-containing peptides by the proteasome or by cytosolic

aminopeptidases may lead to less antigen available for presentation (92-94), while reduced

binding of the mutant to MHC I, or altered recognition by the TCR (95) affects epitope

presentation and recognition by CTLs. Other steps during the antigen processing pathway can

also be affected, such as reduced binding of peptides to TAP for transportation into the ER or

impaired trimming of precursors into antigenic peptides for loading onto MHC I molecules in

the ER (78, 93). Whether processing of escape mutants in the cross-presentation pathway

contributes to immune escape is not known.

While CTL escape mutants enable the evasion of host immune responses and have been

associated with a shift in immunodominance patterns and even the loss of HIV-1 control (89)

the requirement to preserve the viral fitness may limit the appearance of these mutants.

Indeed, previous studies showed that some CTL escape mutations diminished HIV-1 replication

and reverted back to WT sequences upon transmission to an HLA-mismatched individual (96,

97), while other studies demonstrated that the replication capacity of viruses containing CTL

escape mutations that compromised viral fitness could be restored by a compensatory

mutation within the epitope or in flanking sequences (98). These findings suggest that escape

mutants that disrupt functionally important regions of the virus may not sustain and that a

barrier for CTL escape may therefore contribute to containment. This may suggest that

simultaneous delivery of immunogens containing wild-type sequences and escape variants

may represent an effective approach to increase immune pressure on the virus, thus pushing

the virus to mutants that affect viral fitness (99, 100).

1.7. HIV-1 vaccine design

Although treatment of infected individuals with HAART provides successful viral control, HIV-1

remains persistent and is not eradicated. Moreover, HAART does not fully restore the immune

Page 28: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Introduction 20

system and is associated with considerable side effects, thus remaining a life-long

commitment. Therefore, the design of an effective HIV-1 vaccine is still the only way to control

viral infection in the absence of HAART. An effective vaccine against HIV-1 should ideally elicit

humoral and adaptive immunity, such as efficient antibody responses to prevent or limit HIV-1

entry into cells and a potent T cell response that will allow clearance of infected cells (101).

Challenges for the design of a HIV-1 vaccine include the absence of protective immunity in HIV-

1 infected patients, the genetic diversity of HIV-1, the ability of HIV-1 to form viral reservoirs

that cannot be eliminated with HAART, and the lack of B cells making specific neutralizing

antibodies.

HIV-1 infection leads to a predictable hierarchy of immunodominant and subdominant T cell

responses for each HLA allele. Since HIV-1 is not spontaneously cleared by the immune

responses developed during infection it is likely that a vaccine eliciting the same

immunodominance patterns as in natural HIV-1 infection will most likely not be successful at

preventing or clearing HIV-1 infection. Thus, an alternative approach may aim to break natural

immunodominance patterns during vaccination. Besides immunogen design, it is important to

identify specific APCs that can internalize the immunogen for efficient processing and

presentation. Another important aspect in vaccine design is the choice of vectors and

adjuvants, which can alter conditions in which peptides are processed.

Regardless of the immunogen and adjuvant used for vaccination, the HIV-1 immunogen has to

be processed in APCs for priming of immune responses. Therefore, it is important to better

understand how HIV-1 immunogens are degraded into peptides within different cell subsets

and subcellular compartments used for antigen trafficking to evaluate their capacity to

generate peptides compatible with loading onto a variety of MHC I or MHC II molecules.

Page 29: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Motivation 21

2. Motivation

HIV-specific CD8+ T cells play a major role in reducing viral load in acute HIV-1 infection and in

the spontaneous control of HIV-1 infection, and therefore will most likely become a critical

part of an effective HIV-1 vaccine. However multiple lines of evidence indicate that not all CTL

responses have equivalent capacity to kill HIV-infected cells and reduce viral load. Therefore it

is critical to define criteria leading to an efficient CTL response against all HIV-infectable cell

subsets.

1) Are all HIV-infectable cell subsets able to equally process and present the same epitopes?

Several CD4-expressing cell subsets, including CD4+ T cells, monocytes, Møs and DCs, are

permissive to HIV-1 infection. The identification of epitopes presented by all infectable cell

subsets is crucial for the identification and priming of protective CTL responses that should

recognize and kill all infected cell subsets. Whether all HIV-infectable cell subsets process and

present equivalent amounts of epitopes remains open. Differences in the kinetics or amounts

of epitopes displayed by any of these cell subsets may impair CTL recognition hence allow viral

spreading.

1.1) How does the antigen processing machinery in DCs and Møs affect the processing and

presentation of HIV-1 peptides?

1.2) Do DCs and Møs similarly process and present epitopes, which match those presented by

all other HIV-infectable cell subsets?

1.3) How does maturation of both cell subsets with TLR ligands, which are often used as

adjuvants during vaccination or present during HIV-1 infection, affect protease activities and

processing of HIV-1 peptides?

2) Does processing in the cross-presentation pathway versus the direct presentation

pathway lead to similar epitope presentation?

DCs and Møs have the ability to process and present MHC I epitopes from endogenous and

exogenous antigens via two main pathways: the cytosolic/direct pathway and the cross-

presentation pathway. Here we analyze the impact of antigen trafficking through different cell

compartments containing different proteases on the degradation patterns of HIV-1 peptides.

2.1) How does trafficking of antigens/immunogens through different cell compartments affect

its degradation patterns, the amount and the timing of epitope presentation?

2.2) Does processing of antigens/immunogens in the cross-presentation pathway result in

similar epitope presentation compared to the direct presentation pathway?

Page 30: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Motivation 22

3) What is the impact of cross-presentation on HIV-1 immunodominance patterns and

immune escape?

Multiple studies have shown that a narrow repertoire of epitopes is targeted during HIV-1

infection, which results in the formation of immunodominance patterns. The appearance of

escape mutations during disease progression has been shown to result in viral evasion from

the host’s immune responses. Escape mutations in immunodominant and subdominant

epitopes result in a shift in immunodominance patterns and can lead to loss of viral control.

HIV-1 can enter DCs and Møs by fusion at the plasma membrane for delivery into the cytosol,

or by internalization into endolysosomal vesicles. Therefore, the focus of this study is to

analyze the impact of the cross-presentation pathway on immunodominance patterns and

immune escape.

3.1) What is the contribution of cross-presentation to the establishment of immunodominance

patterns during HIV-1 infection?

3.2) Do escape mutants affect the processing of epitopes in cell compartments involved in

cross-presentation?

4) What are the implications for immunogen design?

An effective vaccine against HIV-1 should ideally elicit humoral and adaptive immunity, such as

efficient antibody responses to prevent or limit HIV-1 entry into cells and potent CD8+ T cells

for clearance of infected cells as well as CD4+ T cell help required for priming. Vaccines,

including peptides, viral vectors or peptides bound to nanoparticles, enter DCs by receptor-

dependent or -independent endocytosis. To evaluate the capacity of proteins/immunogens to

generate peptides compatible with loading onto a variety of MHC I or MHC II molecules

independent of a given HLA, it is necessary to better understand how HIV proteins are

degraded into peptides within different cell subsets and subcellular compartments used for

antigen/immunogen trafficking.

Page 31: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Materials & Methods 23

3. Materials and Methods

3.1. Materials

3.1.1. Instruments

Instruments

Centrifuge Sorvall Legend XTR Thermo Scientific, USA

CO2 incubator Galaxy 170R Eppendorf, Germany

CTL-ImmunoSpot S6 Macro Analyzer CTL, USA

ELISA Sunrise microplate reader Tecan, Switzerland

Flow cytometer, FACSCalibur BD Biosciences, USA

Flow cytometer, LSR II BD Biosciences, USA

HPLC system Waters, USA

Labquake Tube Shaker Thermo Scientific, USA

LTQ Orbitrap Mass Spectrometer Thermo Scientific, USA

Microcentrifuge 5417R Eppendorf, Germany

Microscope BH2 Olympus, USA

Microscope EVOS XL Life Technologies, USA

Nano-HPLC Eksigent, USA

NucleoCounter NC-200 Chemometec, Denmark

Odyssey CLx Infrared Imaging System LI-COR Biosciences, USA

PowerPac Universal Power Supply Bio-Rad, USA

Trans-Blot SD Semi-Dry Transfer Cell Bio-Rad, USA

VICTOR X5 Multilabel Plate Reader PerkinElmer, USA

Vortex-Genie 2 Scientific Industries, USA

XCell SureLock Mini-Cell Electrophoresis System Life Technologies, USA

3.1.2. Chemicals, Solutions

Chemicals, Solutions

Acetonitrile, 99.8%, for HPLC Fisher Scientific, USA

Acetonitrile, Optima LC/MS Fisher Scientific, USA

Adenosine 5’-triphosphate disodium salt hydrate (102) Sigma-Aldrich, USA

Page 32: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Materials & Methods 24

Chemicals, Solutions

Digitonin Sigma-Aldrich, USA

Dimethyl Sulfoxide (DMSO) Fisher Scientific, USA

DL-Dithiothreitol (DTT) Sigma-Aldrich, USA

Ethylenediaminetetraacetic acid disodium salt solution (EDTA) Sigma-Aldrich, USA

Fetal Bovine Serum, Heat inactivated (FBS) Sigma-Aldrich, USA

Formic acid, 99+% Thermo Scientific, USA

Glycerol Sigma-Aldrich, USA

Human Serum AB Gemini Bio-Products, USA

Hydrochloric acid, HCl Fisher Scientific, USA

Magnesium chloride, MgCl2 Sigma-Aldrich, USA

Penicillin-Streptomycin Solution, 50X Corning, USA

Potassium phosphate monobasic, KH2PO4 Sigma-Aldrich, USA

Potassium acetate, C2H3KO2 Sigma-Aldrich, USA

Sodium chloride, NaCl Sigma-Aldrich, USA

Trichloroacetic acid, ≥ 99.0% Sigma-Aldrich, USA

Trifluoroacetic acid, 99% Sigma-Aldrich, USA

Water, Optima LC/MS Fisher Scientific, USA

3.1.3. Buffers, Media

Buffers, Media

AIM-V media Invitrogen, USA

Dulbecco’s Phosphate Buffered Saline (PBS) Sigma-Aldrich, USA

Hank’s balanced salt solution Sigma-Aldrich, USA

HEPES, 1M solution Corning, USA

RPMI-1640 Medium, without L-glutamine Sigma-Aldrich, USA

Trizma hydrochloride buffer solution Sigma-Aldrich, USA

3.2. Study participants

HLA-typed blood donors were recruited at the Massachusetts General Hospital (MGH) in

Boston. All participants provided written informed consent for participation in the study.

Anonymous buffy coats were purchased from the MGH. This study was approved for use by

the Partners Human Research Committee under protocol 2005P001218 (Boston, MA).

Page 33: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Materials & Methods 25

3.3. Cell culture

3.3.1. Generation of monocyte-derived DCs and Møs

Human peripheral blood mononuclear cells (PBMCs) were freshly isolated from HLA-typed

blood donors or anonymous buffy coats by Ficoll-Hypaque (Sigma-Aldrich, USA) density

centrifugation. Monocytes were enriched from PBMCs using CD14+ magnetic isolation kits

according to the manufacturer’s instructions (StemCell, Canada). Briefly, PBMCs were

incubated with human anti-CD14 antibodies followed by incubation with dextran-coated

magnetic particles in PBS, 2% human serum AB and 2mM EDTA. Labeled CD14+ monocytes

were subsequently isolated by magnetic separation. The purity of CD14+ monocytes was

>98%. DCs were differentiated from monocytes during a 6-day culture at 106 cells/mL in AIM-V

medium, supplemented with 1% HEPES, 1% human serum AB, 20ng/mL IL-4 (CellGenix,

Germany) and 10ng/mL GM-CSF (CellGenix, Germany). On day 2 and 4, fresh IL-4 and GM-CSF

were added. Møs were differentiated from monocytes during a 6-day culture at 106 cells/mL

in ultra low attachment plates (Corning, USA) in AIM-V medium supplemented with 1% HEPES

and 10% human serum AB. Where indicated, maturation of DCs and Møs was induced by TLR

ligand stimulation with 2µg/mL LPS (TLR4 ligand), 1µg/mL CL097, or 1µg/mL R848 (TLR 7/8

ligands) (Invivogen, USA) for 2 days.

To assess the purity of DCs and Møs populations, cells were stained using human anti-CD11c,

anti-CD14, and anti-BDCA1 antibodies (BD Biosciences, USA) and analyzed by flow cytometry

(Supplemental Figure 1).

3.3.2. Culture of epitope-specific CD8+ T cell clones

HLA-B57-restricted-ISW9-, B57-KF11-, B57-TW10-, A03-RK9-, B35-QY9- and A11-ATK9-specific

CTL clones were maintained in the presence of 50U/mL IL-2, using 0.1µg/mL CD3-specific

monoclonal antibody 12F6, and irradiated feeder cells as stimulus for T cell proliferation.

EBV-immortalized B cell lines were maintained in RPMI medium, 10% FBS, 1% HEPES, 2mM L-

glutamine, 50U/mL penicillin, and 50μg/mL streptomycin (CellGro, USA).

3.4. Analysis of the antigen processing machinery in DCs and Møs

3.4.1. Preparation of cell extracts

Cells were permeabilized using digitonin to effectively release the contents of the cytosol and

endo-lysosomes. DCs and Møs were permeabilized in ice-cold lysis buffer containing 0.125%

digitonin (50mM HEPES, 50mM potassium acetate, 5mM magnesium chloride, 1mM DTT, 1mM

ATP, 0.5mM EDTA, 10% Glycerol, pH 7.4), followed by 13,000 rpm centrifugation for 15

Page 34: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Materials & Methods 26

minutes at 4ºC to remove cell debris. The protein concentration was measured by Bio-Rad

protein assay according to the manufacturer’s instructions (Bio-Rad, USA).

3.4.2. Fluorescent measurement of hydrolytic activities

The hydrolytic activities of multiple cytosolic and endo-lysosomal proteases were measured

using fluorogenic peptide substrates. These substrates consist of three to four amino acid

residues attached to a fluorogenic reporter group at the C terminus (Figure 5A). The hydrolytic

activity of the protease cleaves the bond between an amino acid and the reporter group,

resulting in release of a highly fluorescent product, such as 7-amino-4-methylcoumarin (AMC).

The increase in fluorescence upon substrate cleavage can be measured on a fluorescence

plate reader over time and the maximum slope of the curve correlates to the hydrolytic

activity of the protease (Figure 5B). To check the specificity of the reaction, cells or cell

extracts were incubated with specific protease inhibitors before addition of the fluorogenic

substrates. Substrate incubated in the absence of cells or cell extracts were used to measure

the background of the reaction.

Figure 5. Representation of the fluorescence based protease activity assay.

A. Protease-specific substrates consist of a peptide residue linked to a fluorogenic reporter

group at the C terminus. Upon cleavage between the peptide residue and the fluorogenic

reporter group a highly fluorescent product is released.

B. Proteasomal tryptic activity was measured in immature DCs using Boc-LRR-AMC as a

fluorogenic substrate. Increase in fluorescence, which is proportional to the hydrolytic tryptic

activity, was measured with a fluorescence plate reader every 5 minutes. Cells pretreated with

MG132, a proteasome inhibitor, were used to check the specificity of the reaction. To measure

the background of the reaction, fluorogenic substrate was added to PBS without cells.

Page 35: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Materials & Methods 27

To measure cytosolic protease activities in live, intact cells, 2x104 DCs or Møs were plated per

well of a black 96-well plate in PBS/0.0025% digitonin in the presence or absence of relevant

inhibitors and incubated for 30 minutes before addition of the fluorogenic substrates.

Addition of low concentrations of digitonin slightly permeabilized the cell membrane and

enabled diffusion of the fluorogenic substrates into the cytosol. To measure proteolytic

activities of endo-lysosomal proteases, cells were incubated with the relevant substrates in

the absence of digitonin to ensure that the fluorogenic substrate is only taken up into

endosomes and lysosomes.

Cytosolic protease activities in whole cell extracts were measured using equivalent amounts of

extracts as determined by actin normalization in reaction buffer at pH7.4 (50mM HEPES,

50mM potassium acetate, 5mM magnesium chloride, 1mM DTT, 1mM ATP, 0.5mM EDTA). To

measure hydrolytic activities of endo-lysosomal proteases in whole cell extracts, the same

amount of cell extracts was used in reaction buffer at pH4.0, pH5.5 and pH7.4, respectively

(50mM sodium chloride, 50mM potassium phosphate, 2mM DTT, 2mM EDTA). The rate of

fluorescence emission, which is proportional to the kinetics of proteolytic activities, was

measured every 5 minutes at 37°C in a Victor-3 Plate Reader (Perkin Elmer, USA) (103).

Table 1. List of protease specific fluorogenic peptide substrates used to analyze hydrolytic

activities of cytosolic and endo-lysosomal peptidases.

Peptidase Substrate Concentration (µM) Company

Cells Extracts

Proteasome

Caspase-like Z-LLE-AMC 50µM 75μM EMD Millipore

Tryptic Boc-LRR-AMC 50μM 2.5μM Bachem

Chymotryptic Suc-LLVY-AMC 50μM 100μM Bachem

Post-proteasomal peptidases

Aminopeptidases H-Leu-AMC 5μM 12.5μM Bachem

TOP Mca-PLGPK-DNP 20μM 5μM Bachem

TPPII H-AAF-AMC 100μM 100μM Bachem

Endo-lysosomal peptidases

Cathepsin S Z-VVR-AMC 50µM 10μM Enzo Life Sciences

OmniCathepsins (Cathepsin B, L, S)

Z-FR-AMC 50µM 50μM Enzo Life Sciences

Cathepsin B Z-RR-AMC - 50μM Bachem

Cathepsin D&E Mca-GKPILFFRLK-Dnp

- 10µM Enzo Life Sciences

Page 36: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Materials & Methods 28

Table 2. List of protease specific inhibitors used to check the specificity of reactions.

Peptidase Inhibitor Concentration (µM) Company

Cells Extracts

Proteasome

Caspase-like

MG132 10µM 10µM Enzo Life Sciences Tryptic

Chymotryptic

Post-proteasomal peptidases

Aminopeptidases Bestatin 12μM - Sigma-Aldrich

TOP N-[(RS)-1-Carboxy -3-phenyl-propyl] -AAF-4-Abz-OH

10μM - Bachem

TPPII Butabindide 10μM - Tocris

Endo-lysosomal peptidases

Cathepsin S Z-FL-COCHO 10µM 10µM Calbiochem

OmniCathepsins (Cathepsin B, L, S)

E64 50µM 50µM Enzo Life Sciences

Cathepsin B Z-RLV-azagly-IV-OMe

- 10μM Bachem

Cathepsin D&E Pepstatin A - 100µM Enzo Life Sciences

3.4.3. Analysis of the antigen processing machinery by Western blot

Proteins in cell extracts were separated on a 4-12% Bis-Tris gradient gel (Invitrogen, USA)

according to their molecular weight and transferred to PVDF membranes (Millipore, USA). To

minimize non-specific binding of the primary antibodies, membranes were blocked with

Odyssey Blocking Buffer (Li-cor Biosciences, USA), and subsequently probed with antibodies

specific for β-actin, the subunit of the proteasome lid S1, the cytosolic leucine aminopeptidase

LAP (Abcam, USA), the proteasome activator subunit PA28α, the proteasome core subunit α7,

the catalytically active subunits of the constitutive and immunoproteasome β1, β2, β5, β1i, β5i

(Enzo Life Sciences, USA), the chaperone Hsp90, the cytosolic peptidase TOP, the ER-resident

aminopeptidases ERAP1 (Santa Cruz, USA), ERAP2 (R&D Systems, USA), and the cytosolic

peptidase TPPII (ProteinTech, USA). After overnight incubation with the primary antibodies,

membranes were washed multiple times and incubated with IRDye secondary antibodies.

Protein bands were visualized by dual infrared fluorophore Odyssey Infrared Imaging System,

and densitometric quantification was performed (Li-cor Biosciences, USA) (104). Equivalent

amounts of cell extracts, as determined by actin normalization, were used to measure

peptidase activities in cell extracts and to degrade HIV peptides using an in vitro degradation

assay.

Page 37: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Materials & Methods 29

3.5. In vitro processing of HIV peptides

To analyze and compare the degradation of peptides into epitopes in different subcellular

compartments, we used a previously developed in vitro degradation assay (105, 106). This

assay uses whole cell extracts of DCs and Møs as a source of peptidases from different cell

compartments to degrade synthetic HIV-1 peptides. In order to activate compartment-specific

proteases the pH value of the degradation buffer was adjusted to pH7.4, pH5.5 or pH4.0. At

these pH values proteases specific for the cytosol, endosomes, and lysosomes were activated

(Figure 3), which allowed us to analyze the processing of HIV peptides in these cell

compartments from the same cell (105). Degradation products at different time points were

identified by mass spectrometry (MS) and their antigenicity was tested by a 51Cr release assay.

To test the intracellular stability of optimal MHC I epitopes, synthetic peptides were incubated

in whole cell extracts. At different time points during the degradation the amount of

remaining epitope was quantified by RP-HPLC analysis (37) (Figure 6).

Figure 6. Schematic representation of the in vitro degradation assay.

DC or Mø cell extracts were used as a source of proteases to degrade synthetic HIV peptides or

optimal MHC I epitopes. Degradation products were identified by mass spectrometry and their

antigenicity was tested by a 51Cr release assay. To determine the intracellular stability of an

epitope the amount of remaining epitope at different time points during degradation was

quantified by RP-HPLC analysis.

Page 38: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Materials & Methods 30

3.5.1. In vitro peptide degradation assay

Highly purified peptides (>98% pure) (MGH peptide core facility or from Bio-Synthesis, USA)

were degraded in whole cell extracts at 37ºC in degradation buffer (50mM Tris-HCl, 137mM

potassium acetate, 1mM magnesium chloride, 1mM ATP; pH7.4, pH5.5, or pH4.0). At different

time points during degradation, aliquots of the reaction were taken and the reaction was

stopped by 5% formic acid to inactivate proteases. Peptide fragments present in the digestion

mix were purified by 5% trichloroacetic acid precipitation for 10 minutes on ice and then

centrifuged at 14,000 rpm for 30 minutes. Degradation samples were stored at -80ºC for

antigenicity assays and MS analyses.

3.5.2. Analysis of the antigenicity of the degradation products

Peptide fragments present in the digestion mix were purified by 5% trichloroacetic acid

precipitation and diluted in RPMI without serum. The pH was re-adjusted to pH7.4 with

NaOH. Chromium-51 (51Cr)-labeled HLA-matched B cells were pulsed with the purified

digestion products (diluted to 0.4μg/mL for 5-ATK9-2 and 0.8μg/mL for 13-QY9-6) for 30

minutes at 37°C in the absence of serum and used as targets in a 4-hour 51Cr release assay

with HLA-matched, epitope-specific CTL clones at a 4:1 effector-to-target ratio. The lysis %

were compared with the lysis of B cells pulsed with undigested long peptides and optimal

epitope titrations (104, 106).

3.5.3. Identification and semi-quantification of degradation products by MS

The identity of the peptides generated during in vitro degradation was determined by tandem

MS. Equal amounts of peptide degradation samples generated at different time points were

injected into a Nano-HPLC (Eksigent) coupled to an Orbitrap MS (LTQ Orbitrap Discovery,

Thermo) at a 400nL/min flow rate. A Nano cHiPLC trap column (200µm x 0.5mm ChromXP

c18-CL 5µm 120Å; Eksigent, USA) was used to remove salts in the sample buffer. Peptides

were separated in a Nano cHiPLC column (75µm x 15cm ChromXP c18-CL 5µm 300Å; Eksigent,

USA) with 0.1% formic acid in water (buffer A) and 0.1% formic acid in acetonitrile (buffer B).

The conditions for the HPLC separation prior to MS analysis can be found in table 3. Mass

spectra were recorded in the range of 370 to 2000Daltons. In tandem MS mode, the eight

most intense ions were selected with a 1Da window for fragmentation in a collision cell using

Helium with 35V as collision voltage. MS fragments were searched against source peptide

databases with Protein Discoverer 1.4 (Thermo Scientific, USA). For semi-quantification the

integrated area under a peak generated by a given peptide is proportional to the abundance

of that peptide. Each degradation time point was run on the MS at least twice.

Page 39: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Materials & Methods 31

Table 3. HPLC gradient used to fractionate degradation products prior to MS analysis.

Time (min) % buffer A % buffer B

0.0 98.0 2.0

0.1 98.0 2.0

5.0 98.0 2.0

100.0 40.0 60.0

100.5 2.0 98.0

107.5 2.0 98.0

108.0 98.0 2.0

125.0 98.0 2.0

125.1 98.0 2.0

3.5.4. Intracellular peptide stability assay

To analyze the intracellular stability of optimal MHC I epitopes, highly purified peptides (>98%

pure) were degraded with 15μg of whole cell extracts at 37°C in 50μL of degradation buffer

(50mM Tris-HCl, 137mM potassium acetate, 1mM magnesium chloride, 1mM ATP; pH7.4 or

pH4.0) (56, 105). Aliquots were taken at 0, 10, 30, and 60 minutes, and the reaction was

stopped with 5% trifluoroacetic acid to inactivate proteases. Peptides incubated in

degradation buffer without cell extracts were used as controls.

3.5.5. Quantification of epitope degradation by reversed-phase HPLC analysis

To quantify the amount of remaining peptide in the digestion mix at each time point, we used

reversed-phase high-performance liquid chromatography (RP-HPLC; Waters, USA) analyses.

RP-HPLC involves the separation of molecules on the basis of their hydrophobicity. The

separation depends on the hydrophobic interaction of molecules from the mobile phase to

the immobilized hydrophobic ligands attached to the stationary phase. The digestion mix is

initially applied to the stationary phase in the presence of aqueous buffers. To elute the

peptides in order of increasing hydrophobicity, increasing amounts of organic solvents over

time are applied.

Peptides were fractionated on a Biosuite 4.6mmx50mm C18 column or a xBridge

4.6mmx150mm C18 column (Waters, USA) in water/0.01% formic acid and

acetonitrile/0.005% formic acid (gradient 1) or water/0.05% trifluoroacetic acid and 50%

acetonitrile/0.04% trifluoroacetic acid (gradient 2). The degradation of the original peptide

resulted in the reduction of its peak and the appearance of additional peaks corresponding to

truncated peptides (Figure 7A). The amount of peptide at each time point was calculated by

the integration of the area under each peptide peak. The identity of the peak corresponding to

the original peptide was determined by comparing its elution time to that of pure peptide in

degradation buffer without cell extracts. The relative amount of each peptide at each time

Page 40: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Materials & Methods 32

point was plotted over time, whereby 100% represents the amount of peptide detected at

time 0 minutes. The time at which 50% of the peptide was degraded defines its half-life

(Figure 7B, left panel). A stability rate of each peptide was calculated by a nonlinear regression

(one-phase exponential decay) of the degradation profile obtained over the 60-minute

incubation (Figure 7B, right panel).

Figure 7. Defining the intracellular stability of peptides by RP-HPLC quantification.

A. The peptides present in the digestion mix at each time point were quantified by RP-HPLC

analysis. Each peptide in the digestion mix corresponded to a distinct peak. The degradation of

the original peptide resulted in the reduction of its peak and the appearance of additional

peaks corresponding to truncated peptides. The amount of each peptide was calculated by the

integration of the area under the peptide peak. The identity of each peak was determined by

comparing its elution time to that of known synthetic peptides.

B. The relative amount of each peptide at each time point was plotted against the time. 100%

represents the amount of peptide detected at time 0 minutes. The time at which 50% of the

peptide was degraded defines its half-life. A stability rate of each peptide was calculated by a

nonlinear regression (one-phase exponential decay) of the degradation profile obtained over

the 60-minute incubation (blue line).

Table 4. HPLC gradient 1 to analyze the degradation products of B57-ISW9, B57-KF11, B57-

TW10, B57-FF9 and A11-ATK9.

Time (min) % buffer A % buffer B

0.00 95.0 5.0

4.00 90.0 10.0

23.00 30.0 70.0

23.50 10.0 90.0

27.00 10.0 90.0

28.00 100.0 0.0

Page 41: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Materials & Methods 33

Table 5. HPLC gradient 2 to analyze the degradation products of A03-RK9 and A03-KK9.

Time (min) % buffer A % buffer B

0.00 100.0 0.0

2.00 100.0 0.0

16.50 10.0 90.0

19.00 0.0 100.0

19.50 95.0 5.0

30.0 95.0 5.0

3.6. Cross-presentation of exogenous HIV proteins by DCs and Møs

3.6.1. ELISPOT cross-presentation assay

To measure HIV-specific CD8+ T cell responses to cross-presenting DCs and Møs we used the

enzyme-linked immunospot (ELISpot) assay. This assay detects antigen-induced secretion of

interferon-gamma (IFN-y) by CD8+ T cells trapped by an immobilized antibody, which is

visualized by an enzyme-coupled secondary antibody.

Immature DCs and Møs were incubated with recombinant HIV-1 p24-protein or HIV-1 p55-

protein (Protein Sciences Corporation, USA) for 1hr at 37ºC. As a control, cells were exposed

to the same concentrations of control protein (Protein Sciences Corporation, USA). Where

indicated, immature DCs or Møs were incubated with inhibitors for proteasome (10µM

MG132; Enzo Life Sciences, USA) or cysteine proteases (5µM E64; Sigma-Aldrich, USA) for 45

minutes at 37ºC before addition of recombinant proteins. Cells pulsed with equivalent molar

concentrations of the optimal epitope HLA-B57 ISW9, HLA-B57 KF11, HLA-B57 TW10, and HLA-

A03 RK9 were used as controls for the antigen presentation and CD8+ T cell clone specificity.

Following incubation with proteins, DCs and Møs were thoroughly washed with ice-cold PBS

and co-cultured overnight with epitope-specific CD8+ T cell clones at a 2:1 effector-to-target

ratio in 96-well plates (Millipore, USA) coated with anti-IFN-y mAb 1-D1K (Mabtech, USA). On

the next day, ELISPOT plates were washed and developed, and the detection of spot forming

cells (SFC) was performed as described previously (107). The number of SFC was calculated by

subtracting the number of spots in the negative-control wells from the number of spots in

each experimental well. Responses were regarded as positive if they had at least three times

the mean number of SFC in the three negative-control wells. The magnitude of the epitope-

specific response was reported as the number of SFC per million cells.

3.6.2. P24 uptake assay

To assess the amount of exogenous p24 protein taken up by immature DCs and Møs we used

an enzyme-linked immunosorbent assay (ELISA) to quantify the amount of intracellular p24

Page 42: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Materials & Methods 34

protein. This assay is based on the binding of an antigen to its specific antibody, which allows

the detection of very small amounts of antigen. Briefly, an immobilized antibody captures the

p24 antigen. The captured antigen is complexed with a biotinylated polyclonal antibody to the

p24 antigen, followed by a streptavidin-horseradish peroxidase (HRP) conjugate. The resulting

complex is detected by incubation with ortho-phenylenediamine-HCl (ODP), which produces a

yellow color that is directly proportional to the amount of HIV-1 p24 captured. The

absorbance of each microplate well is determined using a microplate reader and calibrated

against the absorbance of an HIV-1 p24 antigen standard curve.

Immature DCs or immature Møs (1x106cells/mL) were exposed to the following cocktail of

protease inhibitors for 45 minutes at 37ºC to reduce intracellular degradation of internalized

protein before cell lysis: 10µM MG132 (Enzo Life Sciences, USA), 5µM E64 (Sigma-Aldrich,

USA), 10µM cathepsin S inhibitor Z-FL-COCHO (Calbiochem, USA), 10µM leupeptin (Enzo Life

Sciences, USA), 120µM bestatin (Sigma-Aldrich, USA). Following incubation with inhibitors,

DCs and Møs were exposed to different concentrations of recombinant HIV-1 p24 protein and

incubated for 1hr at 37ºC or 4ºC. Afterwards, all samples were thoroughly washed with ice-

cold PBS and immediately treated with 3mg/mL pronase E (Sigma-Aldrich, USA) in AIM-V

medium without serum for 10 minutes on ice. Cells were washed once in AIM-V medium

supplemented with 10% HSA and three times in ice-cold PBS to eliminate pronase E and then

resuspended in 0.5% Triton X-100 containing lysis buffer. The amount of p24 protein was

determined using a HIV-1 p24 antigen ELISA kit according to the manufacturer’s instructions

(Perkin Elmer, USA). The absorbance of each microplate well was determined using a

microplate reader (Tecan, Switzerland).

3.7. Statistical analysis

Spearman’s rank correlation coefficient was used to examine bivariate associations. The

paired t-test, Wilcoxon signed rank test, and Kruskal-Wallis test were used to compare

measurements between groups. All p values are two-sided and p values of <0.05 were

considered significant. In figures, p-value criteria are assigned as *p<0.05, **p<0.01 and

***p<0.001. Statistical analyses were conducted using GraphPad Prism (GraphPad Prism

Software, La Jolla, CA).

Page 43: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 35

4. Results

4.1. Hydrolytic activities of cytosolic proteases involved in antigen

processing in DCs and Møs

We first analyzed the hydrolytic activities of several cytosolic proteases involved in the direct

antigen-processing pathway in immature and mature monocyte-derived dendritic cells (iDCs,

mDCs) and macrophages (iMøs, mMøs). Maturation of cells was achieved by addition of LPS

(TLR4 agonist), CL097 or R848 (TLR7/8 agonists) for two days. Proteasomal catalytic subunits

(caspase-like, tryptic, and chymotryptic) and post-proteasomal activities (aminopeptidases,

TPP II, and TOP) in live, intact DCs and Møs were measured using protease-specific fluorogenic

peptidic substrates, and fluorescence emission upon peptide hydrolysis was monitored over

time as previously done (106, 108, 109).

4.1.1. Proteasomal hydrolytic activities in DCs and Møs change differently upon

maturation with TLR4 or TLR7/8 ligands

In immature DCs and Møs we detected comparable proteasome caspase-like, tryptic and

chymotryptic activities. Maturation of cells for 2 days with LPS or R848, resulted in significant

differences in proteasomal hydrolytic activities between mMøs and mDCs (1.8-, 2.1-, and 1.5-

fold higher in LPS-matured Møs than DCs, and 1.5-, 1.8-, and 1.3-fold higher in R848-matured

Møs, respectively) (Figure 8A). Similarly, in CL097-matured Møs significant higher proteasomal

hydrolytic activities were observed compared to CL097-matured DCs (1.2-, 2.0-, and 2.1-fold

higher in CL097-matured Møs than DCs) (Figure 8B).

To compare how each TLR agonist altered cytosolic antigen processing activities in DCs or Møs,

we calculated a ratio of activities in mature cells over their immature counterpart (data not

shown). Both TLR4 and TLR7/8 stimulations increased proteasomal caspase-like, tryptic, and

chymotryptic hydrolytic activities in Møs by 20% to 30%. In contrast, matured DCs showed

decreased proteasomal activities upon maturation with a 30% lower caspase-like activity, and

a 40% to 50% lower tryptic activity in LPS- and R848-matured DCs. We then plotted the

hydrolytic activity of each subunit in immature and mature DCs against the % of CD86+ CD83+

DCs. The expression of both markers is upregulated upon maturation of DCs with various TLR

ligands. Caspase-like, tryptic, and chymotryptic activities inversely correlated with the

expression of the maturation markers CD83+ and CD86+ after LPS-, R848- or CL097-induced

maturation (rs=-0.4191, p=0.042; rs=-0.4200, p=0.029; and rs=-0.6018, p=0.002, respectively),

Page 44: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 36

which demonstrated decreased proteasomal activities upon maturation of DCs (Figure 8C).

Together, these results indicated that TLR-induced maturation triggered significant changes in

all three proteasomal hydrolytic activities of DCs and Møs in divergent ways.

Figure 8. Proteasomal activities in monocyte-derived DCs and Møs change differently upon

maturation with TLR4 or TLR7/8 ligands.

A. Proteasomal caspase-like, tryptic, and chymotryptic activities were measured with

peptidase-specific fluorogenic substrates in live, intact iDC (), iMø (), mDC LPS (), mMø

LPS (), mDC R848 () and mMø R848 (). Results are from n>10 healthy donors.

B. The same activities as in (A) were measured in iDC (), iMø (), mDC LPS (), mMø LPS (),

mDC CL097 (), and mMø CL097 (). n>10 healthy donors.

C. Proteasomal activities in iDC and mDCs were plotted versus the percentage of CD86+ CD83+

DCs for each experiment. Surface expression was analyzed by flow cytometry. Comparison by

Spearman test is indicated. n>23 measurements.

4.1.2. Hydrolytic activities of cytosolic aminopeptidases and TPP II are comparable

in DCs and Møs, whereas TOP activities are significantly higher in Møs

Proteasomal degradation products can be further trimmed by cytosolic peptidases before

translocation into the ER for loading onto MHC I or they can be degraded into fragments that

Page 45: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 37

are too short to enter the ER (33). To assess the hydrolytic activity of cytosolic

aminopeptidases, thimet oligopeptidase (TOP), and tripeptidyl peptidase II (TPP II) we used

protease-specific fluorogenic peptide substrates (Figure 9). We detected comparable

aminopeptidases hydrolytic activities between immature and mature DCs and Møs, whereas

TOP activities were approximately 2-fold higher in immature, LPS- and R848-matured Møs

compared to their DCs counterparts (Figure 9A). Similar findings were observed in CL097-

matured Møs (Figure 9B). Hydrolytic activities of TPP II were comparable between immature,

LPS- and CL097-matured DCs and Møs (Figure 9B). In contrast to proteasomal activities, we

did not detect any changes in amínopeptidases, TOP, and TPP II activities between the

immature and mature state of each cell subset.

These results indicated that TLR-induced maturation did not result in differences in post-

proteasomal hydrolytic activities between DCs and Møs.

Figure 9. DCs and Møs have comparable aminopeptidases and TPP II activities, whereas TOP

activities are higher in Møs.

A. Post-proteasomal activities for aminopeptidases and TOP were measured in iDC (), iMø

(), mDC LPS (), mMø LPS (), mDC R848 () and mMø R848 ().

B. Aminopeptidases, TOP and TPP II activities were measured in iDC (), iMø (), mDC LPS (),

mMø LPS (), mDC CL097 (), and mMø CL097 (). For (A-B) results are from n>7 healthy

donors.

Page 46: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 38

4.1.3. TLR-mediated maturation of DCs and Møs results in early changes of

proteasomal activities and reflect changes detected upon HIV infection

To assess how TLR stimulation changes protease activities in maturing DCs, we measured

proteasome and aminopeptidases activities at 5h, 24h, and 48h post stimulation with LPS or

CL097. A ratio of activities in maturing cells over their immature counterpart at each time

point was calculated (Figure 10). In line with our previous results, LPS- and CL097-maturing

DCs showed 30% decreased proteasomal caspase-like and tryptic activities as early as 24h

post stimulation (Figure 10A). Further maturation with both TLR ligands decreased caspase-

like activities by 40% and tryptic activities up to 50% compared to immature cells.

Chymotryptic activities were increased as early as 5h upon maturation with LPS but were not

affected by CL097 stimulation at any time point (Figure 10A).

We next analyzed whether HIV-1 infection of DCs and Møs affected the antigen processing

activities. Single-round infection with VSVg-pseudotyped lentivirus expressing HIV-1 NL4.3

Env resulted in infection rates of 14.4% and 23.7% of DCs and Møs 6 days post infection, as

measured by the expression of GFP (Supplemental Figure 2). Addition of SIV viral protein X

(Vpx) increased the infection rate of Møs (34.7% vs. 23.7% infected Møs in the presence or

absence of Vpx), whereas no effect was observed in DCs (Supplemental Figure 2). Previous

studies demonstrated increased infection rates of human monocyte-derived DCs after

incubation with Vpx, which targets the restriction factor SAMHD1 for degradation by

proteasomes (110). However, differences in the concentration of Vpx used in this study and

the incubation time of Vpx with cells prior to the addition of the virus may explain the

different infection rates observed.

In line with results from TLR-matured cells, we detected 20% lower caspase-like activity, and

30% lower tryptic activities in infected DCs (Figure 10B). Chymotryptic activities in DCs did not

change during the first three days of infection and showed a 30% increase after 6 days post

infection. In Møs, we detected 10-20% increased proteasomal caspase-like, tryptic, and

chymotryptic hydrolytic activities upon infection (data not shown). Aminopeptidases activities

in DCs and Møs did not change upon maturation with LPS or CL097 or HIV-1 infection (Figure

10C and data not shown).

These results indicated that TLR-induced maturation affected proteasomal activities within

24h post stimulation and resulted in significant differences between DCs and Møs, which

reflected changes seen upon HIV-1 infection.

Page 47: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 39

Figure 10. Changes in antigen processing activities in maturing monocyte-derived DCs reflect

changes observed upon HIV infection.

A. Ratios of proteasomal activities in TLR-maturing DCs over immature cells of the same donor

were calculated and represented as mean ± SD at the indicated time points post stimulation.

B. Immature DCs were infected with VSVg-ΔEnv NL4.3 in the presence or absence of Vpx for the

indicated time points. At each time point proteasomal caspase-like, tryptic and chymotryptic

activities were measured using protease-specific fluorogenic substrates. Ratios of activities in

infected cells over non-infected cells from the same donor were calculated and represented as

mean ± SD.

C. Ratios of aminopeptidases activities in TLR-maturing DCs (left panel) or infected DCs (right

panel) over immature or non-infected cells of the same donor were calculated and represented

as mean ± SD at the indicated time points post stimulation or infection. For (A-C) results are

from n=2 healthy donors.

Page 48: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 40

4.2. Expression of cytosolic and ER-resident proteases involved in

antigen processing in DCs and Møs

To examine whether the elevated hydrolytic activities observed in mature Møs compared to

mature DCs were due to different expression levels of peptidases, we analyzed cytosolic

extracts by dual infrared fluorophore Western blotting for the expression of structural and

catalytically active proteasomal subunits, cytosolic post-proteasomal peptidases, the

proteasome activator, the ER-resident aminopeptidases ERAP1 and ERAP2 and the chaperone

Hsp90. This technique allows for multiplex detection of signals over a wider quantifiable linear

range than chemiluminescence (111) (Figure 11).

Figure 11. Higher expression levels of proteins involved in antigen processing in Møs

compared to DCs.

Cell extracts from immature, LPS-matured, or CL097-matured DCs and Møs were probed in

Western blots for the expression of chaperone Hsp90, constitutive proteasome catalytic

subunits β1, β2, and β5, proteasomal lid S1, ER-resident aminopeptidase ERAP1, proteasome

regulator PA28α, and proteasomal core α7 using actin as a loading control.

4.2.1. Higher expression levels of the catalytical and structural subunits of the

constitutive and immunoproteasome in Møs compared with DCs

The expression levels of the constitutive proteasomal subunits responsible for caspase-like

(β1), tryptic (β2), and chymotryptic activities (β5) were 2.4-fold higher in immature Møs

compared to immature DCs (Figure 12A). Similar differences were detected between LPS-,

CL097-, and R848-matured Møs and DCs (2.3-, 2.8-, and 2.4-fold in LPS-matured Møs, and 1.8-,

2.5-, and 2.4-fold in CL097-matured Møs, and 2.4-, 3.6-, and 2.0-fold in R848-matured Møs,

respectively). In immature and LPS-matured Møs, approximately 2-fold and 1.6-fold higher

expression levels of immunoproteasome catalytic subunits β1i and β5i were detected (Figure

12B), whereas the expression levels of the proteasome activator subunit PA28α were

comparable between immature and mature DCs and Møs. The lack of a sensitive anti-β2i

Page 49: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 41

antibody prevented us from analyzing the tryptic immunoproteasome subunit. Similar to the

constitutive proteasomal subunits, the expression levels of proteasomal core α7 were

significantly higher in immature and mature Møs compared to DCs, whereas the expression of

the lid S1 tended to be higher in Møs compared to DCs, but these differences were not always

significant (Figure 12C).

These results demonstrated that, regardless of cell maturation states and variability among

donors, the higher expression of the catalytic subunits of proteasomes in Møs likely

contributed to the elevated hydrolytic activities observed in Møs compared to DCs.

Figure 12. Higher expression levels of catalytically active and structural proteasome subunits

in Møs compared to DCs.

Densitometric quantification normalized to actin was performed for the expression of (A)

constitutive proteasomal catalytic subunits β1, β2, and β5; (B) the proteasomal activator

PA28α, and for immunoproteasome catalytic subunit β1i and β5i; (C) proteasomal lid S1 and

proteasomal core α7. Paired t-tests were performed between matched iDC (), iMø (), mDC

LPS (), mMø LPS (), mDC CL097 (), mMø CL097 (), mDC R848 () and mMø R848 ().

Maturation was induced for 48h with the indicated TLR ligands. Mean ± SD is shown.

Page 50: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 42

4.2.2. Møs have higher expression levels of cytosolic TOP and ER-resident ERAP1

and ERAP2 compared to DCs

Analysis of the expression levels of post-proteasomal TPP II and leucine aminopeptidases

(LAP) showed comparable expression levels between immature and mature DCs and Møs.

Higher TOP expression levels were detected in immature and mature Møs compared to DCs,

without reaching statistical significance (1.9-fold in immature Møs, 2.0-fold in LPS-matured

Møs, and 1.7-fold in R848-matured Møs) (Figure 13A). The expression levels of the chaperone

Hsp90 were similar between DCs and Møs in the immature and mature state. The expression

of the ER-resident aminopeptidases ERAP1 and ERAP2 was higher in Møs compared to DCs,

specifically for immature and LPS-matured cells (1.3- and 1.5-fold in immature Møs; 1.6- and

2.2-fold in LPS-matured mMøs for ERAP1 and ERAP2, respectively) (Figure 13B).

These results indicated that higher TOP expression levels detected in immature and mature

Møs might contribute to the higher TOP activities measured in Møs compared to DCs.

Figure 13. Comparable expression levels of cytosolic post-proteasomal activities in DCs and

Møs.

Densitometric quantification normalized to actin was performed for the expression of (A) post-

proteasomal cytosolic proteases TOP, TPP II, and LAP and (B) cytosolic chaperone Hsp90, and

ER-resident aminopeptidases, ERAP1 and ERAP2. Paired t-tests were performed between

matched iDC (), iMø (), mDC LPS (), mMø LPS (), mDC CL097 (), mMø CL097 (), mDC

R848 () and mMø R848 (). Maturation was induced for 48h with the indicated TLR ligands.

Mean ± SD is shown.

Page 51: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 43

4.3. HIV-1 epitope processing by cytosolic peptidases in DCs and Møs

We next aimed to determine whether higher expression levels and proteolytic activities of the

antigen processing machinery in Møs compared to DCs led to differences in the processing of

HIV-derived epitopes. We previously developed an in vitro degradation assay to analyze

epitope processing in cytosolic extracts (79, 106). This assay uses cell extracts as a source of

cytosolic proteases to degrade synthetic HIV-1 peptides. Peptides generated during the

degradation were identified by MS and their antigenicity was analyzed using a 51Cr release

assay. This assay allowed us to compare the degradation of peptides by cytosolic proteases

from DCs and Møs in order to analyze the production of multiple epitopes located within a

peptide independent of the HLA of the donor.

4.3.1. Proteasomal activities in cell extracts of DCs and Møs reflect those measured

in intact, live cells

A prerequisite to using this assay was to confirm that the differences in peptidase activities

measured in live, intact DCs and Møs were replicable in cytosolic extracts used for the epitope

processing assay. To measure proteasomal caspase-like, tryptic, and chymotryptic activities in

cell extracts from DCs and Møs we used equal amounts of extracts normalized to actin levels

(Figure 14). In line with our previous results, we observed in cytosolic cell extracts from Møs

matured with TLR4 and TLR7/8 ligands significant higher proteasomal activities compared to

cell extracts from mature DCs. In cell extracts from LPS-matured Møs we detected 5.4-fold,

6.5-fold, and 2.8-fold higher caspase-like, tryptic, and chymotryptic activities, respectively,

compared to cell extracts from LPS-matured DCs. Similar trends were observed in cytosolic

extracts from R848- and CL097-matured DCs and Møs (Figure 14A-14B). Differences in values

of peptidase activities between intact, live cells and cell extracts may be due to variations in

uptake of substrates between DCs and Møs, differences in ratios between peptidase and

substrate between extracts and intracellular volumes of live cells, and the fact that

proteasome activities measured in live cells include both cytosolic and nuclear proteasomes.

Despite differences in values, all three proteasomal activities measured in cells correlated

significantly with their activities measured in cytosolic cell extracts (rs=0.32, p=0.002, caspase-

like; rs=0.30, p=0.010, tryptic; rs=0.23, p=0.025, chymotryptic; Figure 14C).

Together these results demonstrated that alterations of peptidase activities induced by TLR

ligands and infection occurred similarly in live cells and cell extracts and therefore validated

the use of cytosolic extracts to compare epitope processing in DCs and Møs using our in vitro

degradation assay.

Page 52: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 44

Figure 14. Changes in proteasomal activities in cell extracts reflect those observed in live,

intact DCs and Møs.

A. Proteasomal caspase-like, tryptic, and chymotryptic activities were measured in cytosolic

extracts from iDC (), iMø (), mDC LPS (), mMø LPS (), mDC R848 () and mMø R848 ()

using protease-specific fluorogenic substrates. Results are from n>16 healthy donors.

B. Proteasomal caspase-like, tryptic, and chymotryptic activities were measured as described in

(A) including cytosolic extracts from mDC CL097 (), and mMø CL097 (). Results are from

n>13 healthy donors and show mean ± SD. Paired t-tests and Wilcoxon signed rank test were

performed (*p<0.05, **p<0.01, ***p<0.001).

C. The proteasomal hydrolytic activities measured in live, intact immature or mature DCs ()

and Møs () were plotted against their activities in corresponding cell extracts. A partial

correlation on Spearman ranked data was performed to control for cell type-dependent effects.

Results are from n>73 measurements.

4.3.2. The kinetics and amounts of optimal HIV-1 epitopes produced in cytosolic

extracts of DCs and Møs vary among epitopes

We first used cytosolic extracts from DCs and Møs to degrade a synthetic 16-mer peptide

representing HIV-1 reverse transcriptase (RT), 5-ATK9-2 (WKGSPAIFQSSMTKIL, aa 153-168),

which contains the subdominant HLA-A03/A11-restricted ATK9 epitope (AIFQSSMTK, aa 158-

166) (112). At different time points during the degradation, generated peptides were

Page 53: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 45

identified by MS (79, 106, 109). The degradation of 5-ATK9-2 led to the production of various

fragments representing peptides containing ATK9 with extensions at the N- and C-terminus,

peptides with N-terminal extensions that could be further trimmed into optimal ATK9, the

optimal ATK9 epitope, and also fragments lacking intact ATK9, called antitopes (Figure 15). Mø

cell extracts produced a large variety of fragments containing ATK9 with N-terminal extensions

at each degradation time point and optimal epitope ATK9 was detectable within 3 minutes,

whereas it appeared after 10 minutes in DC cell extracts. Similar results were observed with

cell extracts of immature and mature DCs and Møs (Supplemental Figure 3).

Figure 15. Mø cell extracts produce HLA-A11-ATK9 epitope more efficiently than DCs.

Four nmol of 5-ATK9-2 (aa 153-168 in HIV-1 RT) were degraded with 30µg of cytosolic extracts

from CL097-matured DCs (A) or Møs (B) for 3, 10, 30, 60 and 120 minutes. Peptides containing

epitope HLA-A11-restricted ATK9 (black bars), or no epitope, called antitopes (white bars) were

identified by MS. Optimal epitope ATK9 () is indicated. The data are representative of three

independent experiments from different donors.

Page 54: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 46

To assess and compare the production of peptides in both cell subsets over time, we

calculated for each time point the contribution of each category of peptides (epitope,

antitopes, extended epitopes, original fragment 5-ATK9-2) to the total degradation products

(Figure 16). Each peptide is defined by a peak at a specific retention time and an area under

the peak, which we previously showed to be proportional to the amount of the corresponding

peptide (94, 113). In cell extracts of Møs matured with CL097, optimal ATK9 and fragments

containing N-extended ATK9 represented the major products detected as early as 3 minutes

and throughout the degradation reaction, whereas in cell extracts of DCs similarly matured,

longer peptides were produced in accordance with lower hydrolytic activities. Moreover,

regardless of the maturation state of the cells, Mø cell extracts always produced more N-

extended ATK9-containing fragments and optimal ATK9 than DC cell extracts.

Figure 16. Mø cell extracts produce more A11-ATK9 epitope and epitope precursors

compared to DC cell extracts.

All degradation products of 5-ATK9-2 generated in cell extracts from CL097-matured DC and

Mø were identified by MS at each time point. The peak area of each identified peptide was

determined using Proteome Discoverer 1.4. Peptides were grouped into antitopes that are

lacking intact ATK9 (blue), optimal ATK9 epitope (black), fragments containing N-terminal

(white), C-terminal (red) or N- and C-terminal extensions (green) of ATK9, and the original

fragment (gray). The contribution of each category of peptides to the total intensity of all

degradation products is shown at each time point.

The area of the peak corresponding to ATK9 detected by MS increased over the course of 210

minutes of degradation in Mø cell extracts to up to 11.4-fold higher than during the

degradation in DC cell extracts (Figure 17A). To independently confirm the difference in

production of ATK9 epitope between DCs and Møs, we measured the antigenicity of the

degradation peptides produced over 210 minutes as previously done (79, 106, 109).

Degradation peptides were purified and pulsed onto an HLA-A11 B cell line used as target cells

in a killing assay with ATK9-specific CTLs. Degradation peptides generated in Møs became

increasingly antigenic over the course of the experiment, with lysis % plateauing around 120

Page 55: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 47

minutes at 38% lysis for CL097-matured Mø digests, whereas it reached only 3% lysis for

CL097-matured DC digests – a 12.6-fold difference in antigenicity of the degradation products

(Figure 17B). The antigenicity of the degradation products measured by CTL killing assay

correlated strongly with the production of optimal ATK9 epitope detected by MS (rS=0.6068,

p<0.0001), providing additional validation of the semi-quantification of epitope production by

MS (Figure 17C).

These results showed that the processing of ATK9 epitope from HIV-1 RT-derived precursor

peptide, 5-ATK9-2, was faster and yielded more antigenic peptides in Mø than in DC cytosolic

extracts independent of the maturation state.

Figure 17. Higher amounts of HLA-A11 ATK9 produced by Mø cell extracts resulted in higher

lysis of target cells by ATK9-specific CTLs.

A. Kinetics of optimal ATK9 production by iDC (), iMø (), mDC LPS (), mMø LPS (), mDC

CL097 (), and mMø CL097 () detected by MS.

B. 5-ATK9-2 degradation products were purified and pulsed onto HLA-A03/A11 B cell targets.

ATK9-specific CTL responses were assessed by 51Cr release assay. For (A-B), data are

representative of three independent experiments with extracts from different healthy donors.

C. ATK9 peak areas in DCs () or Møs () detected by MS in three independent experiments

were plotted against the target cell lysis values generated by the corresponding degradation

peptides. Spearman correlation test was performed; n=106 data points. For (A-C) the 30

minutes degradation time points for iMø were lost.

Page 56: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 48

To analyze whether differences in the processing of HLA-A11 ATK9 in cytosolic extracts of DCs

and Møs reflected differences seen upon HIV-1 infection of both cell subsets, we infected

immature DCs and Møs with VSVg-ΔEnv NL4.3 alone or in combination with Vpx and analyzed

the activation of HLA-A11 ATK9-specific CTLs by IFN-gamma ELISPOT assay (data not shown).

Due to low infectivity of DCs and Møs as shown before and insufficient amount of cells to sort

infected cells we were not able to directly compare the endogenous processing and

presentation of HLA-A11 ATK9 by DCs and Møs to A11-ATK9-specific CTL clones.

We next analyzed the processing of a 28-mer Nef-derived precursor peptide 13-QY9-6

(AQEEEEVGFPVTPQVPLRPMTYKAAVDL, aa 60-87 in Nef) which contains HLA-B35-restricted

QY9 and 16 other epitopes (114). Cell extracts from Møs and DCs similarly degraded 13-QY9-6

at a slow rate with 85% of the total peak intensity contributed by peptides longer than 26

amino acids after 10 minutes and approximately 40% after 210 minutes (Figure 18A). In

contrast to A11-ATK9, HLA-B35-restricted QY9 epitope (QVPLRPMTY, aa 73-81) was efficiently

generated by both cell subsets (Figure 18B). There was little difference in the antigenicity of

peptides generated in cell extracts from DCs and Møs (26% and 22% lysis for CL097-matured

DCs and Møs, respectively) (Figure 18C), and no significant differences were observed among

TLR4, 7 and 8 stimulated cells (data not shown). The antigenicity of the degradation products

correlated strongly with the detection of optimal QY9 epitope by MS, again validating the use

of MS to semi-quantify epitope production (rS=0.8329, p<0.0001; results from two

independent experiments) (Figure 18D).

Moreover, we analyzed the production of the 16 well described HIV-1 CD8+ T cell epitopes and

epitope precursors (defined as peptides with the correct C-terminus and extended by up to

three residues at the N-terminus that could be further trimmed in the ER) located in this 28-

mer. Three epitopes (19%) were produced in DC and Mø cell extract at each time point during

degradation, whereas three epitopes (19%) were only produced at later time points during

degradation. Furthermore, seven epitopes (44%) were not produced in either DC or Mø cell

extracts at any time point, while three epitopes (19%) were only produced in cell extracts

from Møs (data not shown).

Thus due to heterogeneity in peptidase activities among DCs and Møs, peptides presented by

HIV-infectable subsets may differ in their timing of presentation, amount and length (optimal

or extended epitopes), all of which could affect recognition by HIV-specific CD8+ T cells.

Page 57: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 49

Figure 18. Extracts from DCs and Møs generate similar amounts of Nef-derived antigenic

peptides at similar rates.

A. Two nmol of 13-QY9-6 (aa 60-87 in Nef) were degraded with 45µg of cytosolic extracts from

mDC CL097 and mMø CL097 for 10, 120, and 210 minutes. Degradation products identified by

MS were grouped according to their lengths of fragments: longer than 26 aa (blue), 19-25 aa

(orange), 13-18 aa (gray), 8-12 aa (red) and fragments equal or shorter than 7 aa (light gray).

The contribution of each category of peptides to the total intensity of all degradation products

is shown at each time point.

B. QY9 epitope production in extracts from mDC CL097 () and mMø CL097 () detected by

MS at the different time points.

C. The 13-QY9-6 degradation products were purified and pulsed onto HLA-B35 B cell targets.

QY9-specific CTL responses were assessed by a 51Cr release assay. For (A-C), data are

representative of three independent experiments with different healthy donors.

D. MS peak areas of peptide QY9 produced in extracts from mDC CL097 () and mMø CL097

() from two independent experiments were plotted against their respective target cell lysis

values from the 51Cr release assay. Spearman correlation test was performed; n=27 data points.

Page 58: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 50

4.4. Processing of HIV-1 peptides by cytosolic and endo-lysosomal

peptidases in DCs and Møs

To assess how degradation of HIV-1 peptides along the cross-presentation pathway of

immature and mature DCs and Møs affected the degradation patterns, we used the previously

described degradation assay recapitulating degradation in cell compartments involved in

cross-presentation (105). This assay allowed the simultaneous analysis of degradation

products by cytosolic, endosomal and lysosomal peptidases from the same cells using MS.

4.4.1. Lysosomal activities in live intact DCs and Møs correlate with activities

measured in cell extracts

To confirm that hydrolytic activities of endo-lysosomal proteases in cell extracts used for

degradation of HIV-1 peptides reflected activities in live, intact DCs and Møs, we first analyzed

the hydrolytic activities of cathepsin S and omni cathepsins, which measures the combined

activities of cathepsins B, L, and S in live, intact DCs and Møs. In line with previous studies we

detected lower omni cathepsins and cathepsin S activities in live, intact DCs compared with

Møs (49). Similar to proteasomal activities in mature DCs, we demonstrated that hydrolytic

activities of cathepsin S and omni cathepsins decreased in DCs upon maturation as shown by

an inverse correlation between both activities and the % of CD86+ and CD83+ DCs (Figure 19A).

We then analyzed the same activities in cell extracts from DCs and Møs at pH4.0 to specifically

activate cathepsins. In line with our previous data using cytosolic extracts, we found that

cathepsin S and omni cathepsins activities in live, intact cells significantly correlated to their

counterparts in corresponding cell extracts, which validated the use of cell extracts to analyze

the degradation of HIV-1 peptides by endo-lysosomal proteases in vitro (Figure 19B).

Maturation of phagosomes to endosomes and lysosomes correlates with a drop in the pH and

the sequential activation of different cathepsins (53). To confirm that different cathepsins

activities can be activated in vitro at different pH values, we measured the proteolytic activities

of omni cathepsins, cathepsin S, cathepsin B, and cathepsin D in cell extracts from immature

DCs at pH4.0, pH5.5, and pH7.4, respectively (Figure 19C). All analyzed cathepsins showed

higher hydrolytic activities at acidic pH values. However, whereas cathepsin D was more active

at pH4.0, cathepsin B showed higher activity at pH5.5, demonstrating different activation of

cathepsins during maturation of mild acidic early endosomes to strong acidic lysosomes.

Similar results were observed using cell extracts from Møs (data not shown).

Together these results demonstrated that different pH values activated cathepsins from

endosomes and lysosomes, which allowed us to analyze the processing of HIV-1 peptides by

endo-lysosomal proteases in vitro.

Page 59: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 51

Figure 19. Lysosomal activities in DC and Mø cell extracts reflect activities in live, intact cells

and can be activated at different pH values.

A. Omni cathepsins and cathepsin S activities in immature and TLR-matured DCs were plotted

against the % of CD86+ CD83+ DCs for each experiment. Surface expression was analyzed by

flow cytometry. Comparison by Spearman test is indicated. n≥26 measurements.

B. Omni cathepsins and cathepsin S hydrolytic activities measured in live, intact immature or

mature DCs () or Møs () were plotted against their activities in corresponding cell extracts

at pH4.0. A partial correlation on Spearman ranked data was performed to control for cell type-

dependent effects. Results are from n≥30 measurements.

C. Omni cathepsins activities (combined cathepsin B, L, S activities), cathepsin S, cathepsin B,

and cathepsin D were measured with specific fluorogenic substrates in whole cell extracts of

immature DCs at pH4.0, pH5.5, and pH7.4, respectively. n≥5 independent donors.

Page 60: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 52

4.4.2. Preferential production of immunodominant epitopes KF11 and TW10 in

cytosolic and endo-lysosomal cell extracts

We first analyzed the degradation of a synthetic 35-mer peptide containing the HLA-B57-

restricted subdominant epitope ISW9 and the immunodominant epitope KF11

(MVHQAISPRTLNAWVKVVEEKAFSPEVIPMFAALS, aa 10-44 in Gag p24) (104-106) by cytosolic

(pH7.4) and endosomal and lysosomal (pH5.5 and pH4.0) proteases. Degradation of this

fragment resulted in the production of peptides of variable lengths at the different pH values

over time indicating the degradation by different proteases with different cleavage

preferences (Supplemental Figure 4). To assess and compare the production of peptides in

each cell subset and cell compartment, we used the peak area under each of the MS identified

peptides. Peptides were grouped according to their lengths and the contribution of each

category of peptides to the total degradation products was calculated for each time point.

Degradation at pH4.0 in cell extracts from immature DCs yielded shorter fragments compared

with degradation at pH7.4, with majority of fragments being 8-12 and 13-18 aa long and

contributing to 45% and 51% of total peptide intensity at 120 minutes, respectively. At pH7.4,

fragments of 8-12 and 13-18 aa length contributed to only 26% and 37% of the total peptide

intensity, whereas fragments of 19-25 aa length represented 31% of total peptide intensity at

120 minutes (Figure 20A, upper left panel). Then we grouped all degradation products

according to their epitope content and showed that the degradation of fragments containing

both epitopes (ISW9+/KF11+) resulted in the preferential production of B57-KF11 epitope-

containing fragments (ISW9-/KF11+), and only small amounts of B57-ISW9 epitope-containing

fragments (ISW9+/KF11-) in extracts of immature DCs at all pH values tested (Figure 20B, upper

left panel). KF11- and ISW9-containing fragments were produced more efficiently at pH7.4

than at pH5.5 and pH4.0, suggesting that both epitopes may most likely be presented in the

direct presentation pathway or if epitope precursors may escape from endo-lysosomes into

the cytosol quickly after internalization. Similar results were observed for immature Møs

(Figure 20A/B, lower left panel), in line with comparable endocytic hydrolytic activities in cell

extracts from immature monocyte-derived DCs and Møs (115). We then analyzed the

degradation of the 35-mer in extracts from LPS-matured DCs and Møs and detected similar

degradation patterns with fragments of comparable lengths and higher amounts of fragments

containing immunodominant epitope B57-KF11 (Figure 20A/B, right panel).

To analyze the cleavage patterns by proteases active in different cell compartments involved

in cross-presentation, we grouped degradation products identified by MS according to their N-

terminus or C-terminus and determined the contribution of each group of peptides to the

total intensity of all degradation products (Figure 21, upper or lower graph of each panel).

Page 61: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 53

Figure 20. The immunodominant epitope B57-KF11 is more efficiently produced in cross-

presentation-competent compartments than subdominant epitope B57-ISW9.

A. Two nmol of p24-35mer (MVHQAISPRTLNAWVKVVEEKAFSPEVIPMFAALS, aa 10-44 in Gag

p24) were degraded in 15µg of whole cell extracts from immature and mature DCs and Møs for

10, 30, 60, and 120 minutes in degradation buffer at pH7.4, pH5.5 or pH4.0. Degradation

products identified by MS were grouped according to their lengths of fragments: equal or

longer than 26 aa (blue), 19-25 aa (orange), 13-18 aa (gray), 8-12 aa (red), and fragments

equal or shorter than 7 aa (light gray). The peak area of each identified peptide was

determined using Proteome Discoverer 1.4 and the contribution of each category of peptides to

the total intensity of all degradation products is shown at each time point.

B. All degradation products of p24-35mer were identified as described in (A). Peptides were

grouped into fragments containing B57-ISW9 and B57-KF11 epitopes (black), containing only

B57-KF11 epitope (red), containing only B57-ISW9 epitope (blue), or neither epitope (gray). For

(A-B) data are representative of three independent experiments with three different donors.

After 30 minutes of degradation at pH7.4 several minor cleavage sites produced ISW9- and

KF11-containing fragments, whereas at pH5.5 and pH4.0 major cleavage sites within ISW9 and

KF11 destroyed the epitopes. Further N- and C-terminal trimming resulted in the appearance

of new cleavage sites, which still preserved KF11-containing peptides at pH7.4, whereas at

pH5.5 and pH4.0 both epitopes were further destroyed.

Together these data indicated that this p24 35-mer was sensitive to degradation by proteases

active in all three cell compartments involved in cross-presentation in DCs and Møs and

favored the production of dominant epitope B57-KF11 over that of B57-ISW9.

Page 62: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 54

Figure 21. Major cleavage sites within p24-35mer destroy the B57-ISW9 epitope.

Cleavage patterns of p24-35mer incubated with whole cell extracts from immature DCs for 30

minutes (upper panel) or 120 minutes (lower panel) at pH7.4, pH5.5, and pH4.0 are shown as

the contribution of each cleavage site, presented as cleavage N-terminal or C-terminal to a

specific amino acid, to the total intensity of all degradation products. Data are representative

of three independent experiments with three different donors.

Moreover, we analyzed the production of 16 well described HIV-1 CD8+ and CD4+ T cell

epitopes and epitope precursors located in this 35-mer (116). Epitope precursors, defined as

peptides with the correct C-terminus and extended by up to three residues at the N- terminus

that could be further trimmed in the ER (Supplemental Figure 5). Peptides were produced in

extracts of both cell subsets at all pH values tested (B57-KF11, B15-HL9), preferentially

produced at pH7.4 (B57-ISW9, A02-TV9), or at pH5.5 and pH4.0 (B45-VI11, B15-VF9, B44-EV9)

or not produced at any time (B07-SV9). These results highlighted a variable production or

degradation of epitopes at different rates in different cell compartments which might affect

their capacity to activate CD8+ or CD4+ T cells during infection or cross-presentation.

We next extended the analysis to another HIV-1 Gag p24-derived peptide containing the HLA-

B57-restricted epitope TW10 (GSDIAGTTSTLQEQIGWMTNNPPIPVGGEIY, aa 101-131 in Gag

p24) (Figure 22). In contrast to p24-35-mer, the majority of degradation products identified

after 10 to 120 minutes in extracts from immature DCs and Møs at pH7.4, pH5.5 and pH4.0

represented the original fragment or long fragments of mostly >26aa (Figure 22A), containing

the TW10 epitope with N- and C-terminal extensions (Figure 22B). Similarly, degradation in

Page 63: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 55

extracts from mature DCs and Møs showed comparable kinetics of degradation and resulted

in the production of fragments with similar lengths.

Figure 22. Slow degradation of TW10-containing peptides results in high amounts of B57-

TW10 available for direct or cross-presentation.

A. Two nmol of p24-31mer (GSDIAGTTSTLQEQIGWMTNNPPIPVGGEIY, aa 101-131 in Gag p24)

were degraded in 15µg of whole cell extracts from immature and mature DCs and Møs for 10,

30, 60, and 120 minutes in degradation buffer at pH7.4, pH5.5 or pH4.0. Degradation products

identified by MS were grouped according to their lengths of fragments as described before.

B. All degradation products of p24-31mer were grouped into fragments containing B57-TW10

(black), B57-TW10 epitope with N-terminal extensions (white), B57-TW10 epitope with C-

terminal extensions (red), B57-TW10 epitope with N- and C-terminal extensions (green),

antitopes defined as fragments lacking B57-TW10 (blue), or the original peptide (gray),

respectively. For (A-B) the contribution of each category of peptides to the total intensity of all

degradation products is shown at each time point.

In line with the slow degradation of this fragment, the few cleavage sites identified after 30 or

120 minutes were mostly located outside B57-TW10 at all three pH values (Supplemental

Figure 6). The relative resistance of the B57-TW10-containing fragment to intracellular

degradation contrasted with the rapid degradation of the p24 fragment containing B57-ISW9

and B57-KF11 which may contribute to a higher amount of TW10-containing peptide available

for presentation by direct or cross-presentation, thus contributing to the dominance of TW10-

specific CTL responses during acute HIV-1 infection.

Page 64: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 56

4.5. Cross-presentation of HIV-1 proteins by DCs and Møs

We next analyzed whether differences in the processing of HIV-1 peptides observed in cell

extracts from DCs and Møs at pH4.0, pH5.5 and pH7.4 reflected differences in the actual cross-

presentation of the epitopes by live, intact DCs and Møs. We therefore incubated cells with

exogenous recombinant HIV-1 proteins and quantified epitope-specific CTLs responses to

cross-presenting DCs and Møs by IFN-gamma ELISPOT assay.

4.5.1. Cross-presentation of the immunodominant Gag p24 B57-TW10 and B57-KF11

epitopes is more efficient than that of subdominant B57-ISW9 epitope

We first analyzed the cross-presentation of the three optimally defined HLA-B57 restricted HIV

epitopes originating from HIV-1 p24 protein by immature monocyte-derived DCs and Møs:

subdominant HLA-B57-restricted ISW9 (ISPRTLNAW, aa 15-23 in Gag p24), dominant HLA-B57-

restricted KF11 (KAFSPEVIPMF, aa 30-40 in Gag p24), and dominant HLA-B57-restricted TW10

(TSTLQEQIGW, aa 108-117 in Gag p24) (116, 117).

We detected 28-fold and 94-fold lower B57-ISW9-specific CTL responses to cross-presenting

DCs compared with B57-KF11 and B57-TW10-specific responses, respectively. In addition, B57-

KF11 specific CD8+ T cell responses to cross-presenting DCs were 3-fold lower compared with

B57-TW10 without reaching significance (Figure 23A, left panel). Similar results were observed

with cross-presenting Møs, with 47-fold lower B57-ISW9-specific CTL responses compared

with B57-KF11 and B57-TW10-specific CTL responses (Figure 23A, right panel). We then

analyzed the capacity of DCs and Møs pulsed with increasing amounts of the synthetic optimal

B57-ISW9 and B57-TW10 epitopes to activate epitope-specific CTLs. Similar activation of B57-

ISW9 and B57-TW10-specific CTLs were detected at each epitope concentration (Figure 23B).

Previous studies showed comparable affinities of ISW9, KF11 and TW10 for HLA-B57 (118),

suggesting that differences in CTL responses to cross-presenting DCs and Møs were not due to

differences in peptide avidity among the clones, but due to differences in the intracellular

processing of epitope-containing fragments as shown by our in vitro degradation assay.

These findings were in line with our previous results in cell extracts from DCs and Møs

demonstrating higher production of TW10- and KF11-containing peptides compared to ISW9

in all cell compartments involved in cross-presentation. Moreover, these results indicated that

processing of peptides in the cross-presentation pathway might contribute to

immunodominance patterns.

Page 65: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 57

Figure 23. The immunodominant HLA-B57-restricted TW10 and KF11 epitopes are more

efficiently cross-presented than subdominant ISW9 epitope.

A. Immature DCs (left panel) and Møs (right panel) were incubated with recombinant HIV-1 p24

protein and used as antigen presenting cells in an overnight IFN-gamma ELISPOT assay with

HLA-B57 ISW9-specific (), KF11-specific (), and TW10-specific () CTL clones as effector

cells. CTL responses in form of SFCs are shown for n≥10 donors.

B. CTL responses to immature DCs (left panel) and Møs (right panel) pulsed with different

concentrations of optimal epitope B57-ISW9 () or B57-TW10 () were measured. Data are

representative of two independent experiments with different donors.

To ensure that epitopes cross-presented by DCs and Møs were endogenously processed we

measured the intracellular concentrations of HIV-1 p24 protein after uptake at 37ºC or 4ºC. In

both cell subsets the intracellular concentration of HIV-1 p24 increased with the amount of

p24 used for uptake at 37ºC whereas the uptake at 4ºC was minimal (Figure 24A). The

intracellular p24 concentrations in immature Møs were at least 5-fold lower compared with

DCs, which may indicate a faster degradation of internalized protein by Møs (49). Moreover,

B57-KF11-specific CTL responses increased with the amount of exogenous p24 protein added

to cells, in accordance with higher amount of intracellular p24 leading to higher amount of

peptide presentation (Figure 24B). Together, these data showed that the higher CTL responses

against dominant TW10 and KF11 epitopes after uptake of HIV-1 p24 by DCs and Møs were

due to cross-presentation of higher amounts of both peptides compared with subdominant

ISW9.

Page 66: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 58

Figure 24. The amount of exogenous p24 protein internalized by DCs and Møs affects the

amount of epitope cross-presented to CTLs.

A. Immature DCs (red bars) and immature Møs (blue bars) were pre-treated with a cocktail of

protease inhibitors to reduce intracellular degradation before addition of different

concentrations of recombinant HIV-1 p24 protein at 37ºC or 4ºC. Internalized p24 protein was

determined by a standard p24 ELISA assay using whole cell extracts from lysed DCs and Møs.

Results are from three independent experiments with different donors. Mean ± SD is shown.

B. Immature DCs (red bars) or immature Møs (blue bars) were incubated with different

concentrations of recombinant HIV-1 p24 protein and used as antigen-presenting cells to B57-

KF11-specific CTLs. Responses in form of SFC are shown. Data are representative of two

independent experiments with different donors.

4.5.2. Protease activities in cross-presentation competent cell compartments

differently affect processing of HIV-1 epitopes in DCs and Møs

We next aimed to identify factors contributing to the production or destruction of the three

epitopes in live, intact DCs and Møs along the cross-presentation pathway. Incubation of

immature DCs with proteasome inhibitor MG132 resulted in a 43-fold and 4-fold increased

presentation of B57-ISW9 and B57-KF11 epitopes, respectively, suggesting that proteasomal

degradation of epitope-containing peptides limits the amount of ISW9 and KF11 available for

presentation (Figure 25A). Incubation of DCs with epoxomicin, which selectively inhibits the

proteasome, showed a 11-fold increase in B57-KF11-specific CTL responses (data not shown).

Page 67: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 59

In contrast, inhibition of cysteine proteases by E64 had no effect on the cross-presentation of

both epitopes, indicating that fragments escape early into the cytosol before trafficking to

compartments with high cysteine protease activity. B57-TW10-specific CTL responses to cross-

presenting immature DCs decreased approximately 3-fold upon inhibition of proteasomes,

suggesting that proteasomal processing is required for efficient presentation of TW10. In

contrast to DCs, the cross-presentation of B57-ISW9 by immature Møs was not affected upon

inhibition of proteasomes, suggesting that the cross-presentation of ISW9 in Møs is

proteasome-independent (Figure 25B). B57-KF11 and B57-TW10 CTL responses decreased 2-

and 3-fold respectively upon proteasome inhibition with MG132 or epoxomycin in Møs,

suggesting that the processing of both epitopes requires proteasome processing in Møs.

Inhibition of cysteine proteases in Møs did not affect the cross-presentation of ISW9, KF11

and TW10. Together, these results indicated that cross-presentation of HIV-1 p24 involved

distinct proteases in DCs and Møs, which might be essential or detrimental for the processing

of epitopes.

Figure 25. Inhibition of proteasomes increases B57-KF11 and B57-ISW9 epitope cross-

presentation by DCs but not Møs.

Immature DCs (A) and immature Møs (B) were pretreated with proteasome inhibitor (MG132)

or a broad inhibitor of cysteine proteases (E64) before addition of recombinant HIV-1 p24

protein. CTL responses specific to HLA-B57-ISW9 (), HLA-B57-KF11 (), and HLA-B57-TW10

() were measured as described in Figure 23. Results are from n≥5 different donors.

Page 68: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 60

4.5.3. Low degradation of RK9-containing fragments results in high amounts of HLA-

A03 RK9 epitope cross-presented by DCs and Møs

We next analyzed the processing of another immunodominant epitope located in a different

HIV-1 protein and restricted by a different HLA allele in the cross-presentation pathway. The

A03-RK9 epitope (RLRPGGKKK, aa 20-28 in Gag p17) has been shown to be efficiently

produced by cytosolic peptidases in the endogenous processing pathway and presented to

A03-RK9-specific CTLs (37, 79).

We first analyzed the degradation of a HIV-1 p17-derived peptide (RWEKIRLRPGGKKKYKL, aa

15-31 in p17) containing the epitope A03-RK9 by proteases active at pH7.4, pH5.5, and pH4.0.

The degradation patterns at all pH values demonstrated minimal degradation of the epitope

(Figure 26). These data indicated that the low degradation of this peptide in all compartments

involved in cross-presentation may result in more fragments available for loading onto MHC I

and therefore better cross-presentation compared with B57-ISW9 and B57-KF11 epitopes.

Figure 26. The RK9-containing fragment shows minor cleavage sites by cytosolic or endo-

lysosomal proteases.

Cleavage patterns of p17-17mer incubated with whole cell extracts from immature DCs for 120

minutes at pH7.4, pH5.5, and pH4.0 are shown as the contribution of each cleavage site,

presented as cleavage N-terminal or C-terminal to a specific amino acid, to the total intensity of

all degradation products. Data are representative of two independent experiments with

different donors.

To test the cross-presentation of the A03-RK9 epitope by immature DCs and Møs, we

incubated cells with recombinant HIV-1 p55 protein and measured the activation of A03-RK9-

specific CTLs to cross-presenting DCs and Møs. Indeed, A03-RK9 CTL responses to cross-

presenting cells were as strong as cells exogenously pulsed with 1.2 ug/ml RK9 (Figure 27A-B).

Moreover, pre-incubation of DCs and Møs with inhibitors of the proteasome or cysteine

Page 69: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 61

proteases, showed no effect on A03-RK9 cross-presentation, in line with the limited sensitivity

of proteasome and cathepsin-mediated degradation resulting in high amounts of peptide

available for maximum A03-RK9-specific CTL stimulation (Figure 27A). Similar to our previous

findings with HIV-1 p24 protein, the incubation of DCs or Møs with different concentrations of

HIV-1 p55 protein resulted in concentration-dependent A03-RK9-specific CTL responses

(Figure 27C).

Together, these findings indicated that the high stability of A03-RK9-containing peptides

against degradation by proteases from cross-presentation competent cell compartments

resulted in high amounts of epitope available for cross-presentation.

Figure 27. Limited degradation of RK9-containing fragments results in cross-presentation of

high amounts of HLA-A03 RK9 epitope.

A. Immature DCs (left panel) or Møs (right panel) were incubated with recombinant HIV-1 p55

protein and used as antigen presenting cells in an overnight IFN-gamma ELISPOT assay with

HLA-A03 RK9-specific CTLs as effector cells. CTL responses as SFC are shown for n≥5 donors.

B. CTL responses to immature DCs and immature Møs pulsed with different concentrations of

optimal epitope A03-RK9 were measured as described before.

C. Immature DCs (left panel) or Møs (right panel) were incubated with different concentrations

of recombinant HIV-1 p55 protein and used as antigen presenting cells in an overnight IFN-

gamma ELISPOT assay with HLA-A03 RK9-specific CTLs as effector cells. CTL responses in form

of SFC are shown.

Page 70: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 62

4.6. Intracellular stability of HIV-1 epitopes in cytosolic and lysosomal

cell extracts

The variable stability of epitopes in cytosolic extracts of PBMCs contributes to the amount of

peptide available for presentation to T cells (37, 38, 113). We hypothesized that peptide

stability in cytosol and endo-lysosomes of DCs and Møs may contribute to the relative

efficiency of cross-presentation of immunodominant epitopes.

4.6.1. Intracellular epitope stability of HIV-1 epitopes in cytosolic and lysosomal cell

extracts follows CTL responses hierarchy

We first measured the stability of A03-RK9, a dominant epitope in the acute phase, and B57-

KF11, a dominant epitope in the chronic phase, in whole cell extracts of immature DCs and

Møs at pH7.4 and pH4.0. In cell extracts of immature DCs at pH7.4, we detected a more rapid

degradation of B57-KF11 epitope compared to the A03-RK9 epitope (approximate half-lives of

20 minutes versus 61 minutes). In cell extracts of immature DCs at pH4.0, we detected an 11-

fold faster degradation of B57-KF11 epitope (half-life: 1.9 minutes), whereas the A03-RK9

epitope showed a 20-fold higher half-life (half-life: 1223 minutes) (Figure 28, left panel).

Similar results were observed in cell extracts from immature Møs (Figure 28, right panel).

Figure 28. Variable stability of B57-KF11 and A03-RK9 in cytosolic and lysosomal cell extracts

of immature DCs and Møs.

One nmol of highly purified HLA-B57-restricted KF11 () and HLA-A03-restricted RK9 () were

degraded in 15µg of immature DC extracts (left panel) or Mø extracts (right panel) in

degradation buffer at pH7.4 (solid line) or pH4.0 (dashed line). Degradation products were

analyzed by RP-HPLC after 10, 30, and 60 minutes. 100% represents the amount of peptide

detected at time 0, calculated as the surface area under the peptide peak. Data are

representative of three independent experiments using extracts from different healthy donors.

We next compared the intracellular stability of seven well-defined dominant and subdominant

MHC I epitopes located in HIV Gag p24, Gag p17 and RT, and examined whether their stability

Page 71: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 63

corresponded to immunodominance patterns observed in HIV-1 infection. To rank epitopes

we calculated for each epitope a stability rate as done before (37, 113). In cell extracts of

immature DCs at pH7.4 the dominant epitopes A03-RK9 and B57-TW10 showed approximately

5-fold and 4-fold higher stability rates compared with the subdominant epitopes B57-ISW9

and A11-ATK9, respectively (Figure 29, upper left panel). At pH4.0 we detected dramatically

reduced stability rates for B57-ISW9, B57-KF11, B57-FF9, and A11-ATK9, indicating a more

rapid proteolysis by endo-lysosomal proteases. However, the observed stability rates of all

epitopes formed the same hierarchy as seen at pH7.4. Similar results were observed in cell

extracts of immature Møs (Figure 29, lower left panel) and in cell extracts from LPS-matured

DCs and Møs (Figure 29, upper and lower right panel).

Figure 29. The intracellular stability of HIV-1 epitopes in the cytosol and lysosomes follows

CTL responses hierarchy.

The stability rate of optimal epitopes ISW9, KF11, FF9, TW10, ATK9, RK9, and KK9 at pH7.4 and

pH4.0 was calculated by a nonlinear regression (one-phase exponential decay) of the

degradation profile obtained over a 60-minute incubation in extracts of immature and mature

DCs (upper panel) and Møs (lower panel). N=3 and mean ± SD is shown.

Together, these results showed that the intracellular stability of optimal HIV epitopes was

highly variable in DCs and Møs and in different cell compartments, but followed similar

hierarchies and might contribute to differences seen in cross-presentation and

immunodominance patterns observed in HIV-1 infection.

Page 72: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 64

4.7. The impact of HIV-1 escape mutations on epitope production and

peptide stability in the cross-presentation pathway

Immune pressure exerted by T cell immune responses leads to predictable mutations within

and outside epitopes altering viral fitness or epitope presentation. Many mutations altering

proteasomal or aminopeptidase peptide processing lead to immune escape (37). However the

impact of HLA-restricted mutations on the processing of epitopes in the cross-presentation

pathway and immune escape is not known despite being a compartment involved in virus

uptake.

4.7.1. Frequent escape mutations in immunodominant epitopes reduce epitope

production and peptide stability in the cross-presentation pathway

In HLA-B57+ patients TW10 rapidly mutates at residue 3 and 9 during acute infection (119,

120) and the dominant TW10 CTL response wanes and KF11-specific CTL response becomes

dominant (64, 121). To assess the impact of escape mutations on degradation patterns in the

cross-presentation pathway, peptides containing the TW10 epitope or its naturally occurring

variants TW10 T3N or TW10 T3N/G9A were degraded in whole cell extracts from immature

DCs and Møs at pH7.4 and pH4.0. Degradation at pH7.4 showed comparable kinetics with 67-

87% of original peptide and N- and C-extended TW10 fragments left after 60 minutes (Figure

30A). However, both mutants produced less N- and C-extended TW10-containing peptides

than the WT at pH7.4, with peptides contributing to 23% of the total peptide intensity in

TW10 T3N variant, 19% in the TW10 T3N/G9A variant and 45% in the TW10 WT at 60 minutes,

respectively. Degradation at pH4.0 demonstrated faster kinetics and generated a majority of

fragments lacking part of the epitope, called antitopes. After 10 minutes of degradation

antitopes contributed to 39% of the total peptide intensity in TW10 WT, whereas in T3N and

T3N/G9A mutants antitopes contributed to 65% and 99% of total peptide intensity.

The analysis of the N- and C-terminal cleavage sites showed that TW10 WT and TW10 T3N

sequences were spared from degradation at pH7.4 after 10 minutes, whereas a cleavage site

between tryptophane and alanine destroyed the TW10 T3N/G9A variant within 10 minutes of

degradation (Figure 30B, upper panel). At pH4.0, in line with the faster degradation of the

peptides into antitopes, two major cleavage sites produced fragments with an isoleucine at

the N-terminus or a glutamine at the C-terminus thereby destroying TW10 and its variants.

These cleavage sites were 2.8-fold and 2.6-fold higher in the TW10 T3N/G9A variant compared

with the TW10 WT and the TW10 T3N variant (Figure 30B, lower panel). Similar results were

observed with cell extracts from immature Møs (data not shown).

Page 73: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 65

Figure 30. Escape mutations in immunodominant epitopes reduce epitope production in the

cross-presentation-competent cell compartments.

A. Two nmol of 5-TW10-3 (DIAGTTSTLQEQIGWMTN, aa 103-120 in Gag p24), 5-TW10-3 T3N

(DIAGTTSNLQEQIGWMTN), and 5-TW10-3 T3N/G9A (DIAGTTSNLQEQIAWMTN) were degraded

in 15µg of whole cell extracts from immature DCs and Møs for 10, 30 or 60 minutes in

degradation buffer at pH7.4 and pH4.0. (Continued on next page)

Page 74: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Results 66

Degradation products identified by MS were grouped into fragments containing the optimal

epitope TW10 or its mutant (black), the epitope/mutant with N-terminal extensions (white), the

epitope/mutant with C-terminal extensions (red), the epitope/mutant with N- and C-terminal

extensions (green), antitopes defined as fragments lacking part of the epitope/mutant (blue),

or the original peptide (gray), respectively. The contribution of each category of peptides to the

total intensity of all degradation products is shown at each time point.

B. Cleavage patterns of 5-TW10-3, 5-TW10-3 T3N, and 5-TW10-3 T3N/G9A incubated with

whole cell extracts from immature DCs for 10 minutes at pH7.4 (upper panel) or at pH4.0

(lower panel) are shown as the contribution of each cleavage site, presented as cleavage N-

terminal or C-terminal to a specific amino acid, to the total intensity of all degradation

products. For (A-B) data are representative of two independent experiments with different

donors.

We then compared the cytosolic and lysosomal stability of TW10 epitope and its variants in

DCs and Møs (Figure 31). Similar intracellular stabilities were observed for B57-TW10 and

TW10 T3N in each compartment whereas the stability of TW10 T3N/G9A was reduced by 7- to

8-fold in both compartments, in line with the faster degradation of the fragment containing

the TW10 T3N/G9A mutant.

Figure 31. Reduced intracellular stability of TW10 escape mutants in cytosolic and lysosomal

cell extracts.

One nmol of highly purified HLA-B57-restricted TW10, TW10-T3N or TW10-T3N/G9A mutants

were degraded in 15µg of immature DCs or Møs extracts (right and left panel) in degradation

buffer at pH7.4 or pH4.0. A stability rate was calculated as described before. Bars represent the

mean ± SD of three independent experiments with extracts from different healthy donors.

These results demonstrated that the low stability of TW10 T3N/G9A variant might reduce the

epitope presentation in both direct and cross-presentation pathways in line with our previous

findings in cells infected with TW10 variants (37). This represents the first demonstration of an

escape mutation affecting the cross-presentation of an HIV-1 epitope.

Page 75: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Discussion 67

5. Discussion

In this study we comprehensively analyzed and compared the processing of HIV-derived

epitopes by monocyte-derived DCs and Møs in the two antigen processing pathways: the

direct/endogenous pathway and the cross-presentation pathway.

We found significant differences in the hydrolytic activities and expression levels of the

cytosolic antigen processing machinery in monocyte-derived DCs and Møs upon maturation

with TLR4 and TLR7/8 ligands or HIV-1 infection, with mature Møs having highest activities.

These differences led to variations in the kinetics and the amount of HIV-1 epitopes produced

and affected the recognition by HIV-specific CTLs. DCs and Møs have the ability to internalize

exogenous antigens, such as virions, antibody-coated particles or viral vectors, into endosomes

and lysosomes for processing and presentation by MHC II to CD4+ T cells or cross-presentation

by MHC I to CD8+ T cells. Here, we showed that degradation patterns in the cross-presentation

pathway of APCs favored the production of HIV-1 immunodominant MHC I-restricted epitopes,

which led to highest CTL responses to these epitopes cross-presented by DCs and Møs. During

disease progression, the appearance of escape mutations can impair epitope production and

presentation, which results in shifts in immunodominance patterns and less efficient clearance

of infected cells. In this study we provided the first demonstration of antigen processing

mutations affecting the cross-presentation of HIV-1 epitopes by DCs and Møs to CTLs.

Together, in the context of HIV infection the processing of an antigen will most likely be

affected by (i) the processing machinery in different HIV-infectable cell subsets, (ii) the mode

of viral entry and the pathway used by the antigen to traffic through subcellular

compartments, (iii) the recognition of viruses by pathogen-recognition receptors and the

subsequent activation of cells, (iv) the hydrolytic activities of proteases located in different cell

compartments harboring specific cleavage site preferences, and (v) the cleavability of HIV-1

sequences by these proteases. All these aspects will affect the presentation of peptides by

MHC I or MHC II for recognition and activation of HIV-specific CD8+ or CD4+ T cells. A better

understanding of each of these factors will be crucial for HIV-1 vaccine design.

5.1. Antigen processing in HIV-infectable cell subsets

HIV-1 infects several CD4-expressing cell subsets, including CD4+ T cells, monocytes, Møs and

DCs, which should be targeted by HIV-specific CD8+ T cells for clearance of infection. Whether

all HIV-infectable cell subsets process and present equivalent amounts of epitopes and are

equally recognized by HIV-specific CTLs is poorly defined. If all HIV-infectable cell subsets

display sufficient amount of peptides upon infection, they may be recognized and cleared by

Page 76: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Discussion 68

HIV-specific CD8+ T cells, and this broader recognition of infected targets may contribute to the

superior antiviral function of these CD8+ T cells - independently of their poly-functionality,

avidity, and TCR (122-124). However, if peptide presentation by one subset is slower or below

threshold of detection for a specific TCR, these cells may produce and propagate virus before

immune recognition occurs. The processing and presentation of MHC I epitopes is a multistep

pathway including several proteases in different cell compartments (125, 126), which can

produce or destroy epitope-containing peptides. Previously, it has been shown that significant

higher proteasome and aminopeptidases activities in monocytes compared to CD4+ T cells

from the same donor affected the processing of several HIV-1 epitopes and resulted in clear

differences in the kinetics and the amount of antigenic peptides produced (106). In line with

these findings, other studies demonstrated differences in the processing of LCMV proteins by

fibroblasts and DCs in mice (127) or during degradation of HIV-1 epitopes by purified

proteasomes from DCs or CD4+ T cells (128). In this study we show that differences in the

expression levels and proteolytic activities of proteases involved in the processing of antigens

in immature and mature DCs and Møs affect the production of several HIV-1 epitopes and lead

to uneven recognition by epitope-specific CTLs (104). Elevated levels of immunoproteasomes

can lead to faster kinetics and higher amounts of epitope-containing fragments available for

presentation (129-131). As we detect more ATK9-containing fragments in Møs compared to

DCs it is tempting to link the production of ATK9 to higher proteolytic activities of the

constitutive proteasome or immunoproteasome. This would be in line with previous findings in

monocytes, which demonstrated higher proteasomal activities compared to CD4+ T cells and

produced more ATK9 epitope (106). However, processing of peptides by proteasomes may also

lead to higher degradation of peptides, as we show for the B57-ISW9 and B57-KF11 epitopes

during cross-presentation by DCs, thus reducing the amount of epitope available for

presentation (28, 32) and the processing of some epitopes has been shown to be less efficient

by immunoproteasomes (132). Moreover, differences in other cytosolic or ER-resident

peptidases, such as TOP or ERAP1 or 2 (which are higher in Møs compared to DCs) may lead to

different epitope processing. Due to low infectivity of DCs and Møs, we were not able to

directly compare the endogenous processing and presentation of the HLA-A11-restricted ATK9

epitope by both cell subsets to A11-ATK9-specific CTL clones. However, the similar changes in

antigen-processing activities observed in DCs and Møs upon TLR-stimulation or HIV infection

suggest that our findings using the in vitro degradation assay are relevant to epitope

production and presentation in the context of HIV-1 infection.

Human DCs represent a very heterogeneous population that consists of different DC subsets,

each with distinct functional characteristics. In the blood at least three subsets can be

Page 77: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Discussion 69

distinguished (BDCA1+ DCs, BDCA3+ DCs and pDCs) and in the skin at least two main subsets

(Langerhans cells and CD14 DCs). Similarly, multiple Møs subsets (133), monocyte subsets

(134), and CD4+ T cell subsets (4) have been identified, which may have different capacities to

process HIV-1 epitopes. In addition, cell-specific peptidases such as a serine protease uniquely

expressed in monocytes (135), may lead to cell-specific processing of epitopes and may

contribute to the differential epitope presentation and priming of immune responses by

different DC subsets shown previously (136, 137). Further transcriptomics (138, 139),

proteomics and functional analyses are needed to identify additional cell subset-specific

peptidases that may shape epitope presentation by various APC subsets.

Besides differences in the proteolytic activities between different cell subsets, the presence of

natural peptides, usually five to six amino acids long, has been shown to modulate the

proteasome activity and may contribute to differences in peptide processing (140). Further

studies are required to evaluate the relevance of these peptides in different cell subsets during

HIV-1 infection.

Together, differences in the antigen processing activities between HIV-infectable cell subsets

may result in differences in the timing and the efficiency of epitope production, which may

lead to unequal recognition and killing by HIV-specific CTLs.

5.2. Viral entry and intracellular trafficking of antigens

Viral fusion may occur either at the cell membrane or from within the endosomes, which may

be dependent on the cell subset that is targeted by HIV-1. The relative contribution of either

route to infection of target cells may be affected by the activity of endocytic proteases (higher

endocytic activities will lead to higher degradation of incoming virions), the pH within endo-

and lysosomes (more acidic pH will lead to the activation of multiple endo-lysosomal

proteases) and the fusogenicity of the virus. Thus, the same virus may fuse primarily at the

plasma membrane if endocytosis rates are low or from within the endosomes when

endocytosis activities are high.

The route of virus entry determines the intracellular trafficking of antigens and thus the

proteases that the antigen will encounter. The presence of different proteases in various cell

compartments in HIV-infectable cell subsets may affect the degradation patterns of antigens.

Previous studies showed different degradation patterns by proteases located in endo-

lysosomes, the cytosol or the ER (78, 105, 141). Internalization of virions into endocytic

compartments will lead to degradation by compartment-specific peptidases (different

cathepsins, IRAP, asparaginyl endopeptidase, gamma-interferon-inducible lysosomal thiol

reductase). Processed antigens can then enter the cytosol for further processing by the

Page 78: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Discussion 70

proteasome and eventually cross-presentation by MHC I. In contrast, viral fusion at the cell

membrane may lead to the transport of the same viral antigens into the cytoplasm where the

proteasome will degrade the antigen and thus may result in different cleavage patterns (141).

Degradation of HIV-1 peptides in cytosolic and endo-lysosomal cell extracts shown in this study

demonstrate that some MHC I epitopes are similarly processed in both cell compartments

while others are better processed in one or the other compartment as shown before (105).

The trafficking of antigens in DCs and Møs may also depend on the receptor used to internalize

the antigen. Different receptors can transport their cargo into different endocytic

compartments, such as early or late endosomes or lysosomes, where peptides can be loaded

onto MHC II or escape into the cytosol for cross-presentation by MHC I.

Whether differences in the intracellular trafficking of antigens (direct presentation vs. cross-

presentation) affect the kinetics of epitope presentation remains open, despite the importance

of early immune recognition and killing of infected cells before viral spreading occurs. Our in

vitro degradation data show different kinetics and amounts of MHC I and MHC II peptides,

which may lead to differences in the kinetics of epitope presentation to CD8+ or CD4+ T cells.

To address this question a previously developed real-time cytotoxicity assay can be used (142).

This assay measures the presentation of epitopes by live, intact APCs to epitope-specific CTLs

or CD4+ T cells and subsequent killing or activation in real-time.

Together, the route of virus entry into the cell, either by membrane fusion and release of viral

RNA into the cytoplasm or by endocytosis of viral particles into endosomes, will affect (i) the

efficiency of infection of target cells (143), (ii) the route of antigen trafficking through various

subcellular compartments and (iii) may affect the receptors involved in pathogen sensing and

therefore the signaling cascade utilized to activate cells.

5.3. Viral recognition and cell activation upon HIV-1 infection

DCs and Møs express a variety of TLRs for recognition of pathogen-associated molecular

patterns (PAMPs), such as glycoproteins, lipopolysaccharides, and nucleic acid motifs. From

the first contact of HIV-1 with its receptors at the plasma membrane to its replication in the

cytoplasm, viral proteins and nucleic acids can be detected and trigger activation of immune

cells (144). Different DC subsets express distinct TLRs, which determine their responses to

products derived from pathogens (145, 146) and lead to changes in the capacity to internalize

antigens (147), modify the intracellular antigen processing machinery (104) and shape the

adaptive immune response by producing different cytokines (148, 149). All DC subsets express

TLR7 and/or TLR8, which are expressed in endosomes and can sense single-stranded RNA

oligonucleotides from HIV-1. Upon viral recognition a downstream signaling cascade activates

Page 79: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Discussion 71

the expression of genes required for inflammation and adaptive immune activation (150). We

show that TLR4 and TLR7/8 activation of DCs and Møs leads to significant changes in the

hydrolytic activities of various cytosolic and endo-lysosomal proteases between both cell

subsets (104), which modifies the kinetics and the amount of epitope production and leads to

differences in CTL activation. The presence of multiple DCs and Møs subsets in vivo with

different TLR ligands and different susceptibilities to HIV-1 infection, suggest that there are

most likely multiple maturation phenotypes and/or infection states of cell subsets (immature

cells, mature cells, and cells undergoing maturation), which will affect their capacity to

produce and present epitopes. Thus, we cannot rule out that the heterogeneous populations

of both cell subsets in vivo would result in more complex patterns of epitope production and

presentation.

The activation of TLR7/8 requires the endocytosis of extracellular viral particles or the cellular

process of autophagy, which transports cytosolic viral RNA into lysosomes (151). In order to

recognize viral RNA in the cytoplasm many cells express RIG-I-like receptors (RLRs), which

utilize the same signaling cascade as TLR7 and TLR8 (152). Whether sensing through RLRs

results in similar kinetics of cell activation and changes in antigen processing activities as

shown in this study using TLR ligands needs to be evaluated in future studies.

During HIV-1 infection a massive production of pro-inflammatory cytokines and type I and II

interferons can be detected (25, 26). Interferon-gamma, LPS and TNF-alpha can induce the

expression of catalytically active immunoproteasome subunits and several other cytosolic and

ER-resident peptidases in cells (153-155), which can affect the processing of HIV-1 epitopes

and thus modifies CTL recognition. Recent studies reported of intermediate proteasomes, in

which only one or two catalytic subunits were replaced and which displayed additional

cleavage specificity, even enlarging the repertoire of antigens presented on MHC I (156).

Moreover, changes in the protease composition in different cell subsets may not only affect

the processing of HIV-1 peptides, but also of peptides from other pathogens or co-infections

thereby affecting the hierarchy of immune responses against these pathogens. Previous

studies have shown that even the self-peptidome displayed by HIV-1 infected cells can change

during HIV-1 infection (157). In addition, activation of CD4+ T cells by TCR stimulation can affect

protease activities and modify the processing of HIV-1 peptides, which may affect the capacity

of HIV-specific CD8+ T cells to recognize and kill these cells.

Moreover, treatment of HIV-1 infected patients with antiretroviral drugs can also affect

antigen processing. HIV protease inhibitors, which target HIV-1 protease to prevent virus

maturation, have been shown to alter proteasome and aminopeptidases activities (113, 158,

Page 80: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Discussion 72

159). These changes modified the degradation patterns of HIV-1 peptides and the production

and presentation of epitopes to HIV-specific CTLs.

Thus, besides differences in the expression levels and hydrolytic activities of proteases

involved in antigen processing between different HIV-infectable cell subsets, the recognition of

PAMPs and the subsequent production of pro-inflammatory cytokines as well as extrinsic

factors such as drugs used to treat HIV-infected individuals can affect the processing of

epitopes and therefore the activation of CD8+ and CD4+ T cells.

5.4. Epitope processing in different subcellular compartments

DCs and Møs express a variety of antigen capture and sensing receptors, which will most likely

affect their capacity to acquire and process internalized antigens (160). Previous studies

showed that targeting antigens to specific receptors will result in the transport of its cargo to

different cell compartments and results in presentation by MHC I or MHC II (51, 161, 162).

Various endocytic proteases, including cysteine cathepsins, serine proteases, and aspartic

proteases are located in endocytic vesicles, which are tightly regulated by the pH. DCs have

been shown to maintain a more alkaline pH within endosomes and phagosomes compared to

Møs (55), which results in lower hydrolytic activities of various cathepsins. In line with these

findings, we show that different cathepsins can be activated at different pH values and

cathepsins activities in intact, live DCs are lower than in Møs. However, using whole cell

extracts as a source of proteases from endosomes and lysosomes activated at pH5.5 and pH4.0

may simplify the actual processing of antigens in different cell compartments of DCs and Møs.

Nevertheless, as antigen processing involves multiple proteases, the use of cell extracts for

protein degradation allows us to consider all these peptidases present in a given compartment,

even though we cannot exclude that by lysing cells and disrupting the cell structure, proteases

from different compartments will affect the degradation patterns. Despite these limitations,

we show that the degradation patterns observed in our in vitro degradation assay correspond

to the cross-presentation patterns of the epitopes by intact, live DCs and Møs, validating the

use of our in vitro degradation assay to analyze processing of peptides in the cross-

presentation pathway. Moreover, other studies showed similar correlations between

degradation of peptides in cytosolic extracts and epitope presentation and killing of infected

cells by CTLs (37, 79, 113). Ideally, different cell compartments, such as early and late

endosomes or lysosomes, would be separated at different time points during cross-

presentation and analyzed for the presence of degradation products. This approach is limited

by the large number of cells required to separate sufficient amounts of different cell

compartments and the fast sample preparation to avoid further peptide degradation. Thus,

Page 81: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Discussion 73

our method represents a simple and fast approach to analyze the degradation of antigens by

proteases from different cell compartments at different time points.

Since antigens intended for cross-presentation traffic through the endocytic system and the

cytosol, the stability of an antigen contributes to the amount of peptide available for

presentation. Indeed, we observed poor cross-presentation of the B57-ISW9 epitope in DCs

and Møs compared to the A03-RK9 epitope, due to a low intracellular stability of the optimal

epitope B57-ISW9 and multiple cleavage sites within ISW9-containing peptides during

degradation of long HIV-1 peptides. This suggests that cross-presentation requires high

stability against degradation in phagosomes to preserve potential MHC I binding epitopes.

However, some degree of degradation is required for cross-presentation, probably because

small molecules are exported to the cytosol with a higher efficiency than large ones (163).

The export of antigens from endosomes and lysosomes into the cytosplasm may be an

additional step determining the efficiency of cross-presentation and whether antigens are

presented by the vacuolar pathway or the cytosolic cross-presentation pathway. Previous

studies showed that induction of macropinocytosis in Møs promotes antigen export to the

cytoplasm, whereas DCs that macropinocytose constitutively also showed constitutive export

of antigens into the cytoplasm (163-165). In line with these findings, we observed an increased

presentation of B57-ISW9 and B57-KF11 epitopes by DCs upon inhibition of the proteasome,

indicating the export of epitope-containing peptides into the cytoplasm whereas in Møs the

processing of both epitopes seemed to be proteasome-independent. However, even though

cross-presentation of some epitopes requires proteasomal processing and TAP transport,

there is no clear evidence for peptide import and loading in the ER during cross-presentation.

The detection of MHC I peptide complexes in phagosomes and endosomes (166, 167), the

localization of TAP to ER membranes (168) and the identification of an endosomal N-terminal

peptidase (IRAP) (44) indicate that after antigen export to the cytoplasm and processing by the

proteasome, peptides may translocate back into endosomes and phagosomes by TAP for

loading onto MHC I. Therefore, we cannot exclude that loading of B57-ISW9 and B57-KF11

onto HLA-molecules during cross-presentation in DCs may occur in endocytic compartments.

To confirm the transport of B57-ISW9- and B57-KF11-containing peptides into the ER during

cross-presentation by DCs, either TAP or ERAP need to be inhibited. Moreover, as different

receptors transport exogenous antigens into different subcellular compartments, it is most

likely that even the same epitope can be loaded onto MHC I in endosomes or the ER

depending on the trafficking of the antigen after internalization. However, it is still very

difficult to evaluate the relevance and the relative contribution of these two pathways to

cross-priming in vivo.

Page 82: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Discussion 74

5.5. Cross-presentation contributes to immunodominance patterns

During acute and chronic HIV infection a predictable hierarchy of CTL responses can be

identified, demonstrating consistent recognition of specific epitopes, whereas others are not

recognized. In line with previous data using cytosolic cell extracts from PBMCs (79) or purified

proteasomes (78), we observe a preferential production of peptides containing

immunodominant epitopes in all cell compartments involved in cross-presentation, which

leads to highest CTL responses to immunodominant epitopes cross-presented by DCs and Møs.

Previous studies showed that the number of peptide/MHC-complexes needed to target a cell

for lysis by epitope-specific CTLs may be fewer than 10 complexes per cell (169, 170), whereas

the amount of peptide/MHC-complexes required to stimulate proliferation of CD8+ T cells is

much greater (171, 172). The higher production and cross-presentation of B57-TW10

compared to B57-ISW9 may lead to better priming of naïve CD8+ T cells and thus to

immunodominance of B57-TW10 observed during acute HIV-1 infection. Besides differences in

the amount of epitope produced, we find highly variable intracellular stabilities of multiple

optimal MHC I epitopes in cytosolic and lysosomal cell extracts with immunodominant

epitopes having highest stabilities. Higher intracellular stability may result in more epitope

available for presentation and thus higher amounts of peptide/MHC-complexes available for

priming of naïve CD8+ T cells. These data demonstrate that the production of peptides during

cross-presentation as well as the intracellular stability may contribute to immunodominance

patterns. However, as immunodominance patterns also depend on the binding affinity of the

epitope to MHC I, the binding of the TCR to the peptide/MHC I-complex as well as the

frequency of T cell precursors, the establishment of immunodominant CD8+ T cell responses

may be determined by the contribution of each of these factors.

Moreover, differences in endo-lysosomal activities in DCs and Møs in vivo (115) as well as

different capacities to cross-present exogenous antigens by different DCs subsets (173, 174)

may affect the ability of each subset to prime CD8+ T cells in vivo. Whether the source of

antigen, such as viral particles, cell-associated antigens from apoptotic infected cells or

antibody-conjugated antigens, will result in similar cross-presentation patterns as shown in

this study requires further analysis.

Priming of naïve CD8+ T cells depends on CD4+ T cell help. Therefore it is important to better

understand the processing and presentation of MHC II epitopes during the generation of anti-

viral immune responses. Previous studies showed that degradation of HIV-1 peptides in

cytosolic extracts affected the kinetics of epitope production (79, 104, 113), and its efficiency

was defined by intraepitopic and flanking motifs (78, 79, 94, 175). Current studies analyze

whether similar motifs can be identified for the processing of peptides in endo-lysosomes,

Page 83: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Discussion 75

which will be important for the induction of CD4+ T cell responses during priming of naïve CD8+

T cells. Together, these data highlight the importance of degradation patterns and intracellular

epitope stability in the cross-presentation pathway to immunodominance patterns.

5.6. The impact of viral escape mutants on cross-presentation

HLA-restricted escape mutations can be frequently detected in the acute and chronic phase of

HIV-1 infection (83, 91, 176). Constant immune pressure on selected epitopes results in

variants that impair epitope processing, reduce binding to MHC I or to the TCR and thus avoid

CTL recognition (90, 95).

This study provides the first demonstration that a frequently detected HLA-restricted mutation

during acute HIV-1 infection affects the cross- and direct presentation of an epitope in DCs and

Møs. While the wild-type epitope B57-TW10, dominant in the acute phase, is efficiently

processed and highly stable in endo-lysosomes and the cytosol of DCs and Møs its mutant

TW10 T242N/G248A is faster degraded. The lower amount of epitope available for

presentation may lead to less epitope presented to T cells and therefore results in immune

escape. Whether all known escape mutants impair epitope processing in the cross-

presentation pathway is currently studied. If an escape mutant impairs the processing of an

epitope in the cross-presentation pathway and in the direct presentation pathway it will not be

presented by any HIV-infectable cell subset. However, if an escape mutant impairs the

processing by cytosolic proteases, but is produced in endo-lysosomes, it may still be presented

by DCs and Møs in the vacuolar pathway. This may suggest that DCs can prime new CD8+ T cell

responses against these mutants – assuming the mutant can bind to MHC I and is recognized

by a TCR. In line with this hypothesis, previous studies showed that some mutants did not

affect epitope presentation and primed novel CTL responses (91, 177). Whether these novel

CTL responses can recognize only cell subsets able to internalize and process escape mutants

in the vacuolar pathway or also recognize cells that transport mutants from the cytosol into

the vacuolar pathway by macroautophagy requires further studies.

Some mutants occurring outside of defined CTL epitopes may correspond to compensatory

mutations enabling the virus to achieve improved fitness after a deleterious mutation arising

elsewhere (98). Previous studies showed that escape mutants in flanking sequences affect

MHC I epitope production in the cytosol or the ER (92-94). Whether these mutants affect the

processing of MHC I epitopes in endo-lysosomes and therefore the cross-presentation of these

mutants is currently investigated.

Due to their loose interaction with the MHC II binding groove, MHC II epitopes containing

mutants may still bind to MHC II molecules and get presented. However, if MHC I-restricted

Page 84: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Discussion 76

escape or compensatory mutants insert major cleavage sites into overlapping MHC II peptides

this would result in impaired MHC II peptide production and represent a new mechanism for

MHC II-restricted escape. Thus, mutants in MHC I epitopes may not only affect their own

processing but also the production of overlapping MHC II epitopes.

Previous studies showed that transmission of the TW10 T242N/G248A variant to HLA-B57

negative individuals resulted in reversion of the T242N mutation to the wild-type, whereas the

G248A mutant persisted and became another’s person wild-type virus indicating different

impacts on the viral fitness (96). The fast degradation of peptides containing the G248A

mutation, as shown in this study, may affect the processing and stability of other MHC I-

restricted epitopes overlapping this mutant. If an immunodominant epitope in the newly

infected individual overlaps with the G248A mutant this may lead to impaired epitope

production of that epitope and hence affect the CTL recognition and immunodominance

patterns in the newly infected individual. Therefore it is important to detect the accumulation

of mutations that do not impact viral fitness cost and are transmitted during HIV-1 infection, as

these mutations can potentially impact the processing and presentation of other MHC I or

MHC II epitopes.

5.7. Impact of antigen processing and cross-presentation on vaccine

design

Vaccines are classified into two main types: preventative and therapeutic vaccines.

Preventative vaccines typically elicit the generation of humoral and adaptive immune

responses, which block the spread of pathogens. Therapeutic vaccines are designed as a

treatment to eradicate the cause of disease by activating or inducing CTLs to eliminate virally

infected cells.

The critical role of DCs to prime CD8+ T cells makes them an attractive target for vaccination

against HIV-1 and other viruses for which cellular immunity seems to be a crucial part of the

immune response. One approach for therapeutic vaccination is the administration of

autologous monocyte-derived DCs loaded ex vivo with a variety of antigens (178, 179).

However, this approach is laborious and expensive and therefore probably not useful in the

context of the HIV-1 epidemic. Alternatively, the selective targeting of antigens to DC-specific

endocytic receptors by monoclonal antibodies coupled to the desired antigen presents a very

efficient alternative and offers a way for antigen presentation on both MHC I and MHC II to

CD8+ and CD4+ T cells (180). Depending on the receptor targeted, immunogens may be taken

up into different subcellular compartments in which different proteases result in different

degradation patterns of peptides. To evaluate the capacity of proteins/immunogens to

Page 85: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Discussion 77

generate peptides compatible with loading onto a variety of MHC I or MHC II molecules

independent of a given HLA it is necessary to better understand how HIV-1 proteins are

degraded into peptides within cells. Our in vitro degradation data demonstrate that MHC I and

MHC II epitopes are variably produced in different subcellular compartments with different

kinetics and different amounts, which will affect the presentation of epitopes to CD8+ and CD4+

T cells. To prime an effective HIV-specific CD8+ T cell response, DCs also have to present MHC II

peptides to generate CD4+ T cell help required for priming. This suggests that vaccines should

target different receptors on DCs to transport antigens either to the MHC I presentation

pathway or into the MHC II pathway. This strategy would also allow to balance the

presentation of MHC I and MHC II epitopes and to minimize the activation of CD4+ T cells to

avoid HIV-1 spreading.

HIV-1 infection leads to a predictable hierachy of T cell responses with narrow

immunodominance of a few T cell responses for each HLA type (64, 83, 84). Since these

immune responses are not able to clear infection, a vaccine leading to the same

immunodominance patterns as natural HIV-1 infection will most likely not clear infection as

shown in previous vaccine trials (181, 182). These data suggest that breaking natural

immunodominance patterns may be an interesting alternative. To break immunodominance

patterns mutations can be inserted into epitope sequences or into regions flanking epitopes to

increase the production of protective epitopes and decrease the production of non-protective

epitopes (37, 79). Understanding the cleavage patterns in various subcellular compartments

will help to design immunogens that are efficiently processed in these compartments and lead

to sufficient amounts of epitopes for presentation. Moreover, the usage of chimeric proteins

that contain only portions of HIV-1 proteins, thus eliminating non-protective or “non-

functional” parts of the protein may be another alternative. However, the success of this

approach requires the identification of protective CD8+ and CD4+ T cell responses, which is still

limited (8, 183).

During transition from acute to chronic infection escape mutations within and outside of

immunodominant epitopes result in changes of CTL response hierachies. This may suggest that

simultaneous delivery of wild-type sequences and escape variants in form of a mosaic

immunogen may represent an effective approach to increase immune pressure on the virus,

thus pushing the virus to mutants that affect viral fitness (99, 100). Understanding the

degradation of escape mutants during cross-presentation will be critical for the design of

immunogens containing frequently detected escape mutants that will traffic through various

cell compartments where efficient processing is required for the induction of effective immune

responses. Moreover, this strategy requires further identification of all HLA-restricted escape

Page 86: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Discussion 78

mutations within an epitope and its flanking residues, which is difficult due to the continuous

evolving of escape mutants to evade the host’s immune response.

Another way to escape immune recognition used by HIV-1 is the expression of viral proteins

that interfere with antigen processing and presentation (184). Vaccination with complete HIV

proteins or with vectors that induce the expression of these proteins will most likely result in

interference of viral proteins with the antigen processing machinery of the APC. Cross-

presentation of exogenous HIV-1 proteins may circumvent this effect by pre-processing

proteins in endosomes and lysosomes before transport into the cytoplasm. However, as shown

in this study cross-presentation of exogenous complete HIV-1 proteins results in

immunodominance patterns similar to those detected during acute HIV-1 infection, which do

not result in clearance of HIV-1 infection.

Recent studies in rhesus macaques showed that immunization with a CMV vector expressing

complete SIV proteins led to clearance of SIV infection in 50% of the animals and led to broad

CD4+ and CD8+ T cell responses, highlighting the importance to generate MHC I and MHC II

epitopes with an immunogen. Interestingly, most SIV-specific CD8+ T cell responses recognized

epitopes presented by MHC II (185-187). Further studies need to analyze the mechanisms

leading to these unconventional CD8+ T cell responses and whether this approach is applicable

to humans. In addition, future studies need to analyze the impact of viral vectors used for

vaccination on the antigen processing machinery in the APCs targeted and whether these

vectors elicit immune responses, which may affect the overall hierarchy of immune responses.

Based on these data, it would be interesting to test whether conjugation of SIV proteins to DC-

specific antibodies would result in similar CD8+ and CD4+ T cell responses.

Previous studies showed that HIV Gag-specific CD4+ T cell responses increased after acute HIV

infection in individuals who control HIV-1 infection (6) and were significantly associated with

control of viral replication (7, 8). These findings suggest, that HIV-specific CD4+ T cell responses

are required for efficient HIV-specific CD8+ T cell responses (188) and should be included in an

immunogen. However, in the context of HIV-1 infection, the activation of CD4+ T cells can

always result in increased infection of CD4+ T cells and thus disease progression.

The type of adjuvant used for vaccination will trigger changes in the hydrolytic activities of

proteases and affect epitope production and presentation, as shown in this study, and will

affect the quality and type of immune response generated. Therefore it is important to analyze

the impact of different adjuvants on antigen processing in subcellular compartments to better

understand the production of MHC I and MHC II epitopes for presentation to CD8+ and CD4+ T

cells.

Page 87: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Discussion 79

Together, various factors impact the processing of immunogens, including (i) the antigen

processing machinery in the different APC subsets, (ii) the pathway used by the antigen to

traffic through subcellular compartments in which different proteases with different

proteolytic activities can cleave the antigen, (iii) the presence or absence of cleavable motifs in

the antigen, and (iv) the presence of TLR ligands used as adjuvants. Future studies need to (i)

identify protective immune responses covering various HLA types across multiple clades, (ii)

design immunogens encompassing these responses, and (iii) identify intracellular pathways

that lead to proper processing and presentation of these MHC I and MHC II epitopes in order

to induce effective immune responses able to recognize and kill all infected cell subsets.

Page 88: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

References 80

6. References

1. Miyauchi, K., Y. Kim, O. Latinovic, V. Morozov, and G. B. Melikyan. 2009. HIV enters

cells via endocytosis and dynamin-dependent fusion with endosomes. Cell 137:433-

444.

2. Daecke, J., O. T. Fackler, M. T. Dittmar, and H. G. Krausslich. 2005. Involvement of

clathrin-mediated endocytosis in human immunodeficiency virus type 1 entry. J Virol

79:1581-1594.

3. Barre-Sinoussi, F., A. L. Ross, and J. F. Delfraissy. 2013. Past, present and future: 30

years of HIV research. Nat Rev Microbiol 11:877-883.

4. Geginat, J., M. Paroni, S. Maglie, J. S. Alfen, I. Kastirr, P. Gruarin, M. De Simone, M.

Pagani, and S. Abrignani. 2014. Plasticity of human CD4 T cell subsets. Front Immunol

5:630.

5. Mehandru, S., M. A. Poles, K. Tenner-Racz, A. Horowitz, A. Hurley, C. Hogan, D. Boden,

P. Racz, and M. Markowitz. 2004. Primary HIV-1 infection is associated with

preferential depletion of CD4+ T lymphocytes from effector sites in the gastrointestinal

tract. J Exp Med 200:761-770.

6. Soghoian, D. Z., H. Jessen, M. Flanders, K. Sierra-Davidson, S. Cutler, T. Pertel, S.

Ranasinghe, M. Lindqvist, I. Davis, K. Lane, J. Rychert, E. S. Rosenberg, A. Piechocka-

Trocha, A. L. Brass, J. M. Brenchley, B. D. Walker, and H. Streeck. 2012. HIV-specific

cytolytic CD4 T cell responses during acute HIV infection predict disease outcome. Sci

Transl Med 4:123ra125.

7. Schieffer, M., H. K. Jessen, A. F. Oster, F. Pissani, D. Z. Soghoian, R. Lu, A. B. Jessen, C.

Zedlack, B. T. Schultz, I. Davis, S. Ranasinghe, E. S. Rosenberg, G. Alter, R. R. Schumann,

and H. Streeck. 2014. Induction of Gag-specific CD4 T cell responses during acute HIV

infection is associated with improved viral control. J Virol 88:7357-7366.

8. Ranasinghe, S., M. Flanders, S. Cutler, D. Z. Soghoian, M. Ghebremichael, I. Davis, M.

Lindqvist, F. Pereyra, B. D. Walker, D. Heckerman, and H. Streeck. 2012. HIV-specific

CD4 T cell responses to different viral proteins have discordant associations with viral

load and clinical outcome. J Virol 86:277-283.

9. Douek, D. C., J. M. Brenchley, M. R. Betts, D. R. Ambrozak, B. J. Hill, Y. Okamoto, J. P.

Casazza, J. Kuruppu, K. Kunstman, S. Wolinsky, Z. Grossman, M. Dybul, A. Oxenius, D.

A. Price, M. Connors, and R. A. Koup. 2002. HIV preferentially infects HIV-specific CD4+

T cells. Nature 417:95-98.

Page 89: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

References 81

10. Turville, S. G., P. U. Cameron, A. Handley, G. Lin, S. Pohlmann, R. W. Doms, and A. L.

Cunningham. 2002. Diversity of receptors binding HIV on dendritic cell subsets. Nat

Immunol 3:975-983.

11. Laguette, N., B. Sobhian, N. Casartelli, M. Ringeard, C. Chable-Bessia, E. Segeral, A.

Yatim, S. Emiliani, O. Schwartz, and M. Benkirane. 2011. SAMHD1 is the dendritic- and

myeloid-cell-specific HIV-1 restriction factor counteracted by Vpx. Nature 474:654-657.

12. Shen, R., H. E. Richter, R. H. Clements, L. Novak, K. Huff, D. Bimczok, S. Sankaran-

Walters, S. Dandekar, P. R. Clapham, L. E. Smythies, and P. D. Smith. 2009.

Macrophages in vaginal but not intestinal mucosa are monocyte-like and permissive to

human immunodeficiency virus type 1 infection. J Virol 83:3258-3267.

13. Carter, C. A., and L. S. Ehrlich. 2008. Cell biology of HIV-1 infection of macrophages.

Annu Rev Microbiol 62:425-443.

14. Ellery, P. J., E. Tippett, Y. L. Chiu, G. Paukovics, P. U. Cameron, A. Solomon, S. R. Lewin,

P. R. Gorry, A. Jaworowski, W. C. Greene, S. Sonza, and S. M. Crowe. 2007. The CD16+

monocyte subset is more permissive to infection and preferentially harbors HIV-1 in

vivo. J Immunol 178:6581-6589.

15. Berger, G., S. Durand, G. Fargier, X. N. Nguyen, S. Cordeil, S. Bouaziz, D. Muriaux, J. L.

Darlix, and A. Cimarelli. 2011. APOBEC3A is a specific inhibitor of the early phases of

HIV-1 infection in myeloid cells. PLoS Pathog 7:e1002221.

16. Chevalier, M. F., B. Julg, A. Pyo, M. Flanders, S. Ranasinghe, D. Z. Soghoian, D. S. Kwon,

J. Rychert, J. Lian, M. I. Muller, S. Cutler, E. McAndrew, H. Jessen, F. Pereyra, E. S.

Rosenberg, M. Altfeld, B. D. Walker, and H. Streeck. 2011. HIV-1-specific interleukin-

21+ CD4+ T cell responses contribute to durable viral control through the modulation

of HIV-specific CD8+ T cell function. J Virol 85:733-741.

17. Soghoian, D. Z., and H. Streeck. 2010. Cytolytic CD4(+) T cells in viral immunity. Expert

Rev Vaccines 9:1453-1463.

18. Seeger, G., D. F. Braus, M. Ruf, U. Goldberger, and M. H. Schmidt. 2002. Body image

distortion reveals amygdala activation in patients with anorexia nervosa -- a functional

magnetic resonance imaging study. Neurosci Lett 326:25-28.

19. Goldberg, C. J., M. C. Fallon, D. P. Moore, E. E. Fogarty, and F. E. Dowling. 2002.

Growth patterns in children with congenital vertebral anomaly. Spine (Phila Pa 1976)

27:1191-1201.

20. Rock, K. L., I. A. York, T. Saric, and A. L. Goldberg. 2002. Protein degradation and the

generation of MHC class I-presented peptides. Adv Immunol 80:1-70.

Page 90: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

References 82

21. Bansal, A., J. Carlson, J. Yan, O. T. Akinsiku, M. Schaefer, S. Sabbaj, A. Bet, D. N. Levy, S.

Heath, J. Tang, R. A. Kaslow, B. D. Walker, T. Ndung'u, P. J. Goulder, D. Heckerman, E.

Hunter, and P. A. Goepfert. 2010. CD8 T cell response and evolutionary pressure to

HIV-1 cryptic epitopes derived from antisense transcription. J Exp Med 207:51-59.

22. Yewdell, J. W., U. Schubert, and J. R. Bennink. 2001. At the crossroads of cell biology

and immunology: DRiPs and other sources of peptide ligands for MHC class I

molecules. J Cell Sci 114:845-851.

23. Yewdell, J. W., and H. D. Hickman. 2007. New lane in the information highway:

alternative reading frame peptides elicit T cells with potent antiretrovirus activity. J

Exp Med 204:2501-2504.

24. Kloetzel, P. M. 2004. Generation of major histocompatibility complex class I antigens:

functional interplay between proteasomes and TPPII. Nat Immunol 5:661-669.

25. Cossarizza, A., C. Ortolani, C. Mussini, V. Borghi, G. Guaraldi, N. Mongiardo, E. Bellesia,

M. G. Franceschini, B. De Rienzo, and C. Franceschi. 1995. Massive activation of

immune cells with an intact T cell repertoire in acute human immunodeficiency virus

syndrome. J Infect Dis 172:105-112.

26. Norris, P. J., B. L. Pappalardo, B. Custer, G. Spotts, F. M. Hecht, and M. P. Busch. 2006.

Elevations in IL-10, TNF-alpha, and IFN-gamma from the earliest point of HIV Type 1

infection. AIDS Res Hum Retroviruses 22:757-762.

27. Griffin, T. A., D. Nandi, M. Cruz, H. J. Fehling, L. V. Kaer, J. J. Monaco, and R. A. Colbert.

1998. Immunoproteasome assembly: cooperative incorporation of interferon gamma

(IFN-gamma)-inducible subunits. J Exp Med 187:97-104.

28. Toes, R. E., A. K. Nussbaum, S. Degermann, M. Schirle, N. P. Emmerich, M. Kraft, C.

Laplace, A. Zwinderman, T. P. Dick, J. Muller, B. Schonfisch, C. Schmid, H. J. Fehling, S.

Stevanovic, H. G. Rammensee, and H. Schild. 2001. Discrete cleavage motifs of

constitutive and immunoproteasomes revealed by quantitative analysis of cleavage

products. J Exp Med 194:1-12.

29. Strehl, B., K. Textoris-Taube, S. Jakel, A. Voigt, P. Henklein, U. Steinhoff, P. M. Kloetzel,

and U. Kuckelkorn. 2008. Antitopes define preferential proteasomal cleavage site

usage. J Biol Chem 283:17891-17897.

30. Cascio, P., C. Hilton, A. F. Kisselev, K. L. Rock, and A. L. Goldberg. 2001. 26S

proteasomes and immunoproteasomes produce mainly N-extended versions of an

antigenic peptide. EMBO J 20:2357-2366.

Page 91: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

References 83

31. Zanker, D., J. Waithman, J. W. Yewdell, and W. Chen. 2013. Mixed proteasomes

function to increase viral peptide diversity and broaden antiviral CD8+ T cell responses.

J Immunol 191:52-59.

32. Kisselev, A. F., T. N. Akopian, K. M. Woo, and A. L. Goldberg. 1999. The sizes of

peptides generated from protein by mammalian 26 and 20 S proteasomes.

Implications for understanding the degradative mechanism and antigen presentation. J

Biol Chem 274:3363-3371.

33. Rock, K. L., I. A. York, and A. L. Goldberg. 2004. Post-proteasomal antigen processing

for major histocompatibility complex class I presentation. Nat Immunol 5:670-677.

34. Seifert, U., C. Maranon, A. Shmueli, J. F. Desoutter, L. Wesoloski, K. Janek, P. Henklein,

S. Diescher, M. Andrieu, H. de la Salle, T. Weinschenk, H. Schild, D. Laderach, A. Galy,

G. Haas, P. M. Kloetzel, Y. Reiss, and A. Hosmalin. 2003. An essential role for tripeptidyl

peptidase in the generation of an MHC class I epitope. Nat Immunol 4:375-379.

35. Saric, T., J. Beninga, C. I. Graef, T. N. Akopian, K. L. Rock, and A. L. Goldberg. 2001.

Major histocompatibility complex class I-presented antigenic peptides are degraded in

cytosolic extracts primarily by thimet oligopeptidase. J Biol Chem 276:36474-36481.

36. York, I. A., A. X. Mo, K. Lemerise, W. Zeng, Y. Shen, C. R. Abraham, T. Saric, A. L.

Goldberg, and K. L. Rock. 2003. The cytosolic endopeptidase, thimet oligopeptidase,

destroys antigenic peptides and limits the extent of MHC class I antigen presentation.

Immunity 18:429-440.

37. Lazaro, E., C. Kadie, P. Stamegna, S. C. Zhang, P. Gourdain, N. Y. Lai, M. Zhang, S. A.

Martinez, D. Heckerman, and S. Le Gall. 2011. Variable HIV peptide stability in human

cytosol is critical to epitope presentation and immune escape. J Clin Invest 121:2480-

2492.

38. Herberts, C. A., J. J. Neijssen, J. de Haan, L. Janssen, J. W. Drijfhout, E. A. Reits, and J. J.

Neefjes. 2006. Cutting edge: HLA-B27 acquires many N-terminal dibasic peptides:

coupling cytosolic peptide stability to antigen presentation. J Immunol 176:2697-2701.

39. Momburg, F., J. Roelse, G. J. Hammerling, and J. J. Neefjes. 1994. Peptide size selection

by the major histocompatibility complex-encoded peptide transporter. J Exp Med

179:1613-1623.

40. Saric, T., S. C. Chang, A. Hattori, I. A. York, S. Markant, K. L. Rock, M. Tsujimoto, and A.

L. Goldberg. 2002. An IFN-gamma-induced aminopeptidase in the ER, ERAP1, trims

precursors to MHC class I-presented peptides. Nat Immunol 3:1169-1176.

41. Saveanu, L., O. Carroll, V. Lindo, M. Del Val, D. Lopez, Y. Lepelletier, F. Greer, L.

Schomburg, D. Fruci, G. Niedermann, and P. M. van Endert. 2005. Concerted peptide

Page 92: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

References 84

trimming by human ERAP1 and ERAP2 aminopeptidase complexes in the endoplasmic

reticulum. Nat Immunol 6:689-697.

42. Serwold, T., F. Gonzalez, J. Kim, R. Jacob, and N. Shastri. 2002. ERAAP customizes

peptides for MHC class I molecules in the endoplasmic reticulum. Nature 419:480-483.

43. York, I. A., S. C. Chang, T. Saric, J. A. Keys, J. M. Favreau, A. L. Goldberg, and K. L. Rock.

2002. The ER aminopeptidase ERAP1 enhances or limits antigen presentation by

trimming epitopes to 8-9 residues. Nat Immunol 3:1177-1184.

44. Saveanu, L., O. Carroll, M. Weimershaus, P. Guermonprez, E. Firat, V. Lindo, F. Greer, J.

Davoust, R. Kratzer, S. R. Keller, G. Niedermann, and P. van Endert. 2009. IRAP

identifies an endosomal compartment required for MHC class I cross-presentation.

Science 325:213-217.

45. Riese, R. J., R. N. Mitchell, J. A. Villadangos, G. P. Shi, J. T. Palmer, E. R. Karp, G. T. De

Sanctis, H. L. Ploegh, and H. A. Chapman. 1998. Cathepsin S activity regulates antigen

presentation and immunity. J Clin Invest 101:2351-2363.

46. Ackerman, A. L., C. Kyritsis, R. Tampe, and P. Cresswell. 2003. Early phagosomes in

dendritic cells form a cellular compartment sufficient for cross presentation of

exogenous antigens. Proc Natl Acad Sci U S A 100:12889-12894.

47. Kovacsovics-Bankowski, M., and K. L. Rock. 1995. A phagosome-to-cytosol pathway for

exogenous antigens presented on MHC class I molecules. Science 267:243-246.

48. Kamath, A. T., S. Henri, F. Battye, D. F. Tough, and K. Shortman. 2002. Developmental

kinetics and lifespan of dendritic cells in mouse lymphoid organs. Blood 100:1734-

1741.

49. Delamarre, L., M. Pack, H. Chang, I. Mellman, and E. S. Trombetta. 2005. Differential

lysosomal proteolysis in antigen-presenting cells determines antigen fate. Science

307:1630-1634.

50. Accapezzato, D., V. Visco, V. Francavilla, C. Molette, T. Donato, M. Paroli, M. U.

Mondelli, M. Doria, M. R. Torrisi, and V. Barnaba. 2005. Chloroquine enhances human

CD8+ T cell responses against soluble antigens in vivo. J Exp Med 202:817-828.

51. Belizaire, R., and E. R. Unanue. 2009. Targeting proteins to distinct subcellular

compartments reveals unique requirements for MHC class I and II presentation. Proc

Natl Acad Sci U S A 106:17463-17468.

52. Claus, V., A. Jahraus, T. Tjelle, T. Berg, H. Kirschke, H. Faulstich, and G. Griffiths. 1998.

Lysosomal enzyme trafficking between phagosomes, endosomes, and lysosomes in

J774 macrophages. Enrichment of cathepsin H in early endosomes. J Biol Chem

273:9842-9851.

Page 93: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

References 85

53. Lennon-Dumenil, A. M., A. H. Bakker, R. Maehr, E. Fiebiger, H. S. Overkleeft, M.

Rosemblatt, H. L. Ploegh, and C. Lagaudriere-Gesbert. 2002. Analysis of protease

activity in live antigen-presenting cells shows regulation of the phagosomal proteolytic

contents during dendritic cell activation. J Exp Med 196:529-540.

54. Hotta, C., H. Fujimaki, M. Yoshinari, M. Nakazawa, and M. Minami. 2006. The delivery

of an antigen from the endocytic compartment into the cytosol for cross-presentation

is restricted to early immature dendritic cells. Immunology 117:97-107.

55. Savina, A., C. Jancic, S. Hugues, P. Guermonprez, P. Vargas, I. C. Moura, A. M. Lennon-

Dumenil, M. C. Seabra, G. Raposo, and S. Amigorena. 2006. NOX2 controls phagosomal

pH to regulate antigen processing during crosspresentation by dendritic cells. Cell

126:205-218.

56. Trombetta, E. S., M. Ebersold, W. Garrett, M. Pypaert, and I. Mellman. 2003. Activation

of lysosomal function during dendritic cell maturation. Science 299:1400-1403.

57. Shen, L., L. J. Sigal, M. Boes, and K. L. Rock. 2004. Important role of cathepsin S in

generating peptides for TAP-independent MHC class I crosspresentation in vivo.

Immunity 21:155-165.

58. Bertholet, S., R. Goldszmid, A. Morrot, A. Debrabant, F. Afrin, C. Collazo-Custodio, M.

Houde, M. Desjardins, A. Sher, and D. Sacks. 2006. Leishmania antigens are presented

to CD8+ T cells by a transporter associated with antigen processing-independent

pathway in vitro and in vivo. J Immunol 177:3525-3533.

59. Georgiadou, D., A. Hearn, I. Evnouchidou, A. Chroni, L. Leondiadis, I. A. York, K. L. Rock,

and E. Stratikos. 2010. Placental leucine aminopeptidase efficiently generates mature

antigenic peptides in vitro but in patterns distinct from endoplasmic reticulum

aminopeptidase 1. J Immunol 185:1584-1592.

60. Munier, M. L., and A. D. Kelleher. 2007. Acutely dysregulated, chronically disabled by

the enemy within: T-cell responses to HIV-1 infection. Immunol Cell Biol 85:6-15.

61. Kahn, J. O., and B. D. Walker. 1998. Acute human immunodeficiency virus type 1

infection. N Engl J Med 339:33-39.

62. Streeck, H., J. S. Jolin, Y. Qi, B. Yassine-Diab, R. C. Johnson, D. S. Kwon, M. M. Addo, C.

Brumme, J. P. Routy, S. Little, H. K. Jessen, A. D. Kelleher, F. M. Hecht, R. P. Sekaly, E. S.

Rosenberg, B. D. Walker, M. Carrington, and M. Altfeld. 2009. Human

immunodeficiency virus type 1-specific CD8+ T-cell responses during primary infection

are major determinants of the viral set point and loss of CD4+ T cells. J Virol 83:7641-

7648.

Page 94: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

References 86

63. Cao, J., J. McNevin, S. Holte, L. Fink, L. Corey, and M. J. McElrath. 2003. Comprehensive

analysis of human immunodeficiency virus type 1 (HIV-1)-specific gamma interferon-

secreting CD8+ T cells in primary HIV-1 infection. J Virol 77:6867-6878.

64. Altfeld, M., E. T. Kalife, Y. Qi, H. Streeck, M. Lichterfeld, M. N. Johnston, N. Burgett, M.

E. Swartz, A. Yang, G. Alter, X. G. Yu, A. Meier, J. K. Rockstroh, T. M. Allen, H. Jessen, E.

S. Rosenberg, M. Carrington, and B. D. Walker. 2006. HLA Alleles Associated with

Delayed Progression to AIDS Contribute Strongly to the Initial CD8(+) T Cell Response

against HIV-1. PLoS Med 3:e403.

65. Flesch, I. E., W. P. Woo, Y. Wang, V. Panchanathan, Y. C. Wong, N. L. La Gruta, T.

Cukalac, and D. C. Tscharke. 2010. Altered CD8(+) T cell immunodominance after

vaccinia virus infection and the naive repertoire in inbred and F(1) mice. J Immunol

184:45-55.

66. Munks, M. W., A. K. Pinto, C. M. Doom, and A. B. Hill. 2007. Viral interference with

antigen presentation does not alter acute or chronic CD8 T cell immunodominance in

murine cytomegalovirus infection. J Immunol 178:7235-7241.

67. Zlatkovic, J., K. Stiasny, and F. X. Heinz. 2011. Immunodominance and functional

activities of antibody responses to inactivated West Nile virus and recombinant

subunit vaccines in mice. J Virol 85:1994-2003.

68. Yewdell, J. W., and M. Del Val. 2004. Immunodominance in TCD8+ responses to

viruses: cell biology, cellular immunology, and mathematical models. Immunity 21:149-

153.

69. Yewdell, J. W. 2006. Confronting complexity: real-world immunodominance in antiviral

CD8+ T cell responses. Immunity 25:533-543.

70. Yu, X. G., M. M. Addo, E. S. Rosenberg, W. R. Rodriguez, P. K. Lee, C. A. Fitzpatrick, M.

N. Johnston, D. Strick, P. J. Goulder, B. D. Walker, and M. Altfeld. 2002. Consistent

patterns in the development and immunodominance of human immunodeficiency

virus type 1 (HIV-1)-specific CD8+ T-cell responses following acute HIV-1 infection. J

Virol 76:8690-8701.

71. Goulder, P. J., M. A. Altfeld, E. S. Rosenberg, T. Nguyen, Y. Tang, R. L. Eldridge, M. M.

Addo, S. He, J. S. Mukherjee, M. N. Phillips, M. Bunce, S. A. Kalams, R. P. Sekaly, B. D.

Walker, and C. Brander. 2001. Substantial differences in specificity of HIV-specific

cytotoxic T cells in acute and chronic HIV infection. J Exp Med 193:181-194.

72. Borrow, P., H. Lewicki, B. H. Hahn, G. M. Shaw, and M. B. Oldstone. 1994. Virus-specific

CD8+ cytotoxic T-lymphocyte activity associated with control of viremia in primary

human immunodeficiency virus type 1 infection. J Virol 68:6103-6110.

Page 95: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

References 87

73. Edwards, B. H., A. Bansal, S. Sabbaj, J. Bakari, M. J. Mulligan, and P. A. Goepfert. 2002.

Magnitude of functional CD8+ T-cell responses to the gag protein of human

immunodeficiency virus type 1 correlates inversely with viral load in plasma. J Virol

76:2298-2305.

74. Draenert, R., C. L. Verrill, Y. Tang, T. M. Allen, A. G. Wurcel, M. Boczanowski, A.

Lechner, A. Y. Kim, T. Suscovich, N. V. Brown, M. M. Addo, and B. D. Walker. 2004.

Persistent recognition of autologous virus by high-avidity CD8 T cells in chronic,

progressive human immunodeficiency virus type 1 infection. J Virol 78:630-641.

75. Addo, M. M., X. G. Yu, A. Rathod, D. Cohen, R. L. Eldridge, D. Strick, M. N. Johnston, C.

Corcoran, A. G. Wurcel, C. A. Fitzpatrick, M. E. Feeney, W. R. Rodriguez, N. Basgoz, R.

Draenert, D. R. Stone, C. Brander, P. J. Goulder, E. S. Rosenberg, M. Altfeld, and B. D.

Walker. 2003. Comprehensive epitope analysis of human immunodeficiency virus type

1 (HIV-1)-specific T-cell responses directed against the entire expressed HIV-1 genome

demonstrate broadly directed responses, but no correlation to viral load. J Virol

77:2081-2092.

76. Frahm, N., P. Kiepiela, S. Adams, C. H. Linde, H. S. Hewitt, K. Sango, M. E. Feeney, M.

M. Addo, M. Lichterfeld, M. P. Lahaie, E. Pae, A. G. Wurcel, T. Roach, M. A. St John, M.

Altfeld, F. M. Marincola, C. Moore, S. Mallal, M. Carrington, D. Heckerman, T. M. Allen,

J. I. Mullins, B. T. Korber, P. J. Goulder, B. D. Walker, and C. Brander. 2006. Control of

human immunodeficiency virus replication by cytotoxic T lymphocytes targeting

subdominant epitopes. Nat Immunol 7:173-178.

77. Buseyne, F., S. Le Gall, C. Boccaccio, J. P. Abastado, J. D. Lifson, L. O. Arthur, Y. Riviere,

J. M. Heard, and O. Schwartz. 2001. MHC-I-restricted presentation of HIV-1 virion

antigens without viral replication. Nat Med 7:344-349.

78. Tenzer, S., E. Wee, A. Burgevin, G. Stewart-Jones, L. Friis, K. Lamberth, C. H. Chang, M.

Harndahl, M. Weimershaus, J. Gerstoft, N. Akkad, P. Klenerman, L. Fugger, E. Y. Jones,

A. J. McMichael, S. Buus, H. Schild, P. van Endert, and A. K. Iversen. 2009. Antigen

processing influences HIV-specific cytotoxic T lymphocyte immunodominance. Nat

Immunol 10:636-646.

79. Le Gall, S., P. Stamegna, and B. D. Walker. 2007. Portable flanking sequences modulate

CTL epitope processing. J Clin Invest 117:3563-3575.

80. Kincaid, E. Z., J. W. Che, I. York, H. Escobar, E. Reyes-Vargas, J. C. Delgado, R. M. Welsh,

M. L. Karow, A. J. Murphy, D. M. Valenzuela, G. D. Yancopoulos, and K. L. Rock. 2012.

Mice completely lacking immunoproteasomes show major changes in antigen

presentation. Nat Immunol 13:129-135.

Page 96: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

References 88

81. Pang, K. C., M. T. Sanders, J. J. Monaco, P. C. Doherty, S. J. Turner, and W. Chen. 2006.

Immunoproteasome subunit deficiencies impact differentially on two

immunodominant influenza virus-specific CD8+ T cell responses. J Immunol 177:7680-

7688.

82. York, I. A., M. A. Brehm, S. Zendzian, C. F. Towne, and K. L. Rock. 2006. Endoplasmic

reticulum aminopeptidase 1 (ERAP1) trims MHC class I-presented peptides in vivo and

plays an important role in immunodominance. Proc Natl Acad Sci U S A 103:9202-

9207.

83. Kiepiela, P., A. J. Leslie, I. Honeyborne, D. Ramduth, C. Thobakgale, S. Chetty, P.

Rathnavalu, C. Moore, K. J. Pfafferott, L. Hilton, P. Zimbwa, S. Moore, T. Allen, C.

Brander, M. M. Addo, M. Altfeld, I. James, S. Mallal, M. Bunce, L. D. Barber, J. Szinger,

C. Day, P. Klenerman, J. Mullins, B. Korber, H. M. Coovadia, B. D. Walker, and P. J.

Goulder. 2004. Dominant influence of HLA-B in mediating the potential co-evolution of

HIV and HLA. Nature 432:769-775.

84. Streeck, H., Z. L. Brumme, M. Anastario, K. W. Cohen, J. S. Jolin, A. Meier, C. J.

Brumme, E. S. Rosenberg, G. Alter, T. M. Allen, B. D. Walker, and M. Altfeld. 2008.

Antigen load and viral sequence diversification determine the functional profile of HIV-

1-specific CD8+ T cells. PLoS Med 5:e100.

85. Chen, W., L. C. Anton, J. R. Bennink, and J. W. Yewdell. 2000. Dissecting the

multifactorial causes of immunodominance in class I-restricted T cell responses to

viruses. Immunity 12:83-93.

86. Sette, A., A. Vitiello, B. Reherman, P. Fowler, R. Nayersina, W. M. Kast, C. J. Melief, C.

Oseroff, L. Yuan, J. Ruppert, J. Sidney, M. F. del Guercio, S. Southwood, R. T. Kubo, R.

W. Chesnut, H. M. Grey, and F. V. Chisari. 1994. The relationship between class I

binding affinity and immunogenicity of potential cytotoxic T cell epitopes. J Immunol

153:5586-5592.

87. Trautmann, L., M. Rimbert, K. Echasserieau, X. Saulquin, B. Neveu, J. Dechanet, V.

Cerundolo, and M. Bonneville. 2005. Selection of T cell clones expressing high-affinity

public TCRs within Human cytomegalovirus-specific CD8 T cell responses. J Immunol

175:6123-6132.

88. La Gruta, N. L., K. Kedzierska, K. Pang, R. Webby, M. Davenport, W. Chen, S. J. Turner,

and P. C. Doherty. 2006. A virus-specific CD8+ T cell immunodominance hierarchy

determined by antigen dose and precursor frequencies. Proc Natl Acad Sci U S A

103:994-999.

Page 97: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

References 89

89. Goulder, P. J., R. E. Phillips, R. A. Colbert, S. McAdam, G. Ogg, M. A. Nowak, P.

Giangrande, G. Luzzi, B. Morgan, A. Edwards, A. J. McMichael, and S. Rowland-Jones.

1997. Late escape from an immunodominant cytotoxic T-lymphocyte response

associated with progression to AIDS. Nat Med 3:212-217.

90. Jones, N. A., X. Wei, D. R. Flower, M. Wong, F. Michor, M. S. Saag, B. H. Hahn, M. A.

Nowak, G. M. Shaw, and P. Borrow. 2004. Determinants of human immunodeficiency

virus type 1 escape from the primary CD8+ cytotoxic T lymphocyte response. J Exp

Med 200:1243-1256.

91. Allen, T. M., X. G. Yu, E. T. Kalife, L. L. Reyor, M. Lichterfeld, M. John, M. Cheng, R. L.

Allgaier, S. Mui, N. Frahm, G. Alter, N. V. Brown, M. N. Johnston, E. S. Rosenberg, S. A.

Mallal, C. Brander, B. D. Walker, and M. Altfeld. 2005. De novo generation of escape

variant-specific CD8+ T-cell responses following cytotoxic T-lymphocyte escape in

chronic human immunodeficiency virus type 1 infection. J Virol 79:12952-12960.

92. Allen, T. M., M. Altfeld, X. G. Yu, K. M. O'Sullivan, M. Lichterfeld, S. Le Gall, M. John, B.

R. Mothe, P. K. Lee, E. T. Kalife, D. E. Cohen, K. A. Freedberg, D. A. Strick, M. N.

Johnston, A. Sette, E. S. Rosenberg, S. A. Mallal, P. J. Goulder, C. Brander, and B. D.

Walker. 2004. Selection, transmission, and reversion of an antigen-processing

cytotoxic T-lymphocyte escape mutation in human immunodeficiency virus type 1

infection. J Virol 78:7069-7078.

93. Draenert, R., S. Le Gall, K. J. Pfafferott, A. J. Leslie, P. Chetty, C. Brander, E. C. Holmes,

S. C. Chang, M. E. Feeney, M. M. Addo, L. Ruiz, D. Ramduth, P. Jeena, M. Altfeld, S.

Thomas, Y. Tang, C. L. Verrill, C. Dixon, J. G. Prado, P. Kiepiela, J. Martinez-Picado, B. D.

Walker, and P. J. Goulder. 2004. Immune selection for altered antigen processing leads

to cytotoxic T lymphocyte escape in chronic HIV-1 infection. J Exp Med 199:905-915.

94. Zhang, S. C., E. Martin, M. Shimada, S. B. Godfrey, J. Fricke, S. Locastro, N. Y. Lai, P.

Liebesny, J. M. Carlson, C. J. Brumme, O. A. Ogbechie, H. Chen, B. D. Walker, Z. L.

Brumme, D. G. Kavanagh, and S. Le Gall. 2012. Aminopeptidase substrate preference

affects HIV epitope presentation and predicts immune escape patterns in HIV-infected

individuals. J Immunol 188:5924-5934.

95. Iversen, A. K., G. Stewart-Jones, G. H. Learn, N. Christie, C. Sylvester-Hviid, A. E.

Armitage, R. Kaul, T. Beattie, J. K. Lee, Y. Li, P. Chotiyarnwong, T. Dong, X. Xu, M. A.

Luscher, K. MacDonald, H. Ullum, B. Klarlund-Pedersen, P. Skinhoj, L. Fugger, S. Buus, J.

I. Mullins, E. Y. Jones, P. A. van der Merwe, and A. J. McMichael. 2006. Conflicting

selective forces affect T cell receptor contacts in an immunodominant human

immunodeficiency virus epitope. Nat Immunol 7:179-189.

Page 98: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

References 90

96. Leslie, A. J., K. J. Pfafferott, P. Chetty, R. Draenert, M. M. Addo, M. Feeney, Y. Tang, E.

C. Holmes, T. Allen, J. G. Prado, M. Altfeld, C. Brander, C. Dixon, D. Ramduth, P. Jeena,

S. A. Thomas, A. St John, T. A. Roach, B. Kupfer, G. Luzzi, A. Edwards, G. Taylor, H. Lyall,

G. Tudor-Williams, V. Novelli, J. Martinez-Picado, P. Kiepiela, B. D. Walker, and P. J.

Goulder. 2004. HIV evolution: CTL escape mutation and reversion after transmission.

Nat Med 10:282-289.

97. Li, B., A. D. Gladden, M. Altfeld, J. M. Kaldor, D. A. Cooper, A. D. Kelleher, and T. M.

Allen. 2007. Rapid reversion of sequence polymorphisms dominates early human

immunodeficiency virus type 1 evolution. J Virol 81:193-201.

98. Schneidewind, A., M. A. Brockman, R. Yang, R. I. Adam, B. Li, S. Le Gall, C. R. Rinaldo, S.

L. Craggs, R. L. Allgaier, K. A. Power, T. Kuntzen, C. S. Tung, M. X. LaBute, S. M. Mueller,

T. Harrer, A. J. McMichael, P. J. Goulder, C. Aiken, C. Brander, A. D. Kelleher, and T. M.

Allen. 2007. Escape from the dominant HLA-B27-restricted cytotoxic T-lymphocyte

response in Gag is associated with a dramatic reduction in human immunodeficiency

virus type 1 replication. J Virol 81:12382-12393.

99. Barouch, D. H., K. E. Stephenson, E. N. Borducchi, K. Smith, K. Stanley, A. G. McNally, J.

Liu, P. Abbink, L. F. Maxfield, M. S. Seaman, A. S. Dugast, G. Alter, M. Ferguson, W. Li,

P. L. Earl, B. Moss, E. E. Giorgi, J. J. Szinger, L. A. Eller, E. A. Billings, M. Rao, S.

Tovanabutra, E. Sanders-Buell, M. Weijtens, M. G. Pau, H. Schuitemaker, M. L. Robb, J.

H. Kim, B. T. Korber, and N. L. Michael. 2013. Protective efficacy of a global HIV-1

mosaic vaccine against heterologous SHIV challenges in rhesus monkeys. Cell 155:531-

539.

100. Fischer, W., S. Perkins, J. Theiler, T. Bhattacharya, K. Yusim, R. Funkhouser, C. Kuiken,

B. Haynes, N. L. Letvin, B. D. Walker, B. H. Hahn, and B. T. Korber. 2007. Polyvalent

vaccines for optimal coverage of potential T-cell epitopes in global HIV-1 variants. Nat

Med 13:100-106.

101. Burton, D. R., R. Ahmed, D. H. Barouch, S. T. Butera, S. Crotty, A. Godzik, D. E.

Kaufmann, M. J. McElrath, M. C. Nussenzweig, B. Pulendran, C. N. Scanlan, W. R.

Schief, G. Silvestri, H. Streeck, B. D. Walker, L. M. Walker, A. B. Ward, I. A. Wilson, and

R. Wyatt. 2012. A Blueprint for HIV Vaccine Discovery. Cell Host Microbe 12:396-407.

102. Adhikari, N. K., A. Bashir, F. Lamontagne, S. Mehta, N. D. Ferguson, Q. Zhou, L. Hand, K.

Czarnecka, D. J. Cook, J. T. Granton, J. O. Friedrich, A. Freitag, I. Watpool, and M. O.

Meade. 2011. High-frequency oscillation in adults: a utilization review. Crit Care Med

39:2631-2644.

Page 99: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

References 91

103. Kisselev, A. F., and A. L. Goldberg. 2005. Monitoring activity and inhibition of 26S

proteasomes with fluorogenic peptide substrates. Methods Enzymol 398:364-378.

104. Dinter, J., P. Gourdain, N. Y. Lai, E. Duong, E. Bracho-Sanchez, M. Rucevic, P. H.

Liebesny, Y. Xu, M. Shimada, M. Ghebremichael, D. G. Kavanagh, and S. Le Gall. 2014.

Different Antigen-Processing Activities in Dendritic Cells, Macrophages, and

Monocytes Lead to Uneven Production of HIV Epitopes and Affect CTL Recognition. J

Immunol 193:4322-4334.

105. Vaithilingam, A., N. Y. Lai, E. Duong, J. Boucau, Y. Xu, M. Shimada, M. Gandhi, and S. Le

Gall. 2013. A simple methodology to assess endolysosomal protease activity involved

in antigen processing in human primary cells. BMC Cell Biol 14:35.

106. Lazaro, E., S. B. Godfrey, P. Stamegna, T. Ogbechie, C. Kerrigan, M. Zhang, B. D. Walker,

and S. Le Gall. 2009. Differential HIV epitope processing in monocytes and CD4 T cells

affects cytotoxic T lymphocyte recognition. J Infect Dis 200:236-243.

107. Streeck, H., N. Frahm, and B. D. Walker. 2009. The role of IFN-gamma Elispot assay in

HIV vaccine research. Nat Protoc 4:461-469.

108. Lazaro, E., C. Kadie, P. Stamegna, S. C. Zhang, P. Gourdain, N. Y. Lai, M. Zhang, S. M.

Martinez, D. Heckerman, and S. Le Gall. 2011. Variable HIV peptide stability in human

cytosol is critical to epitope presentation and immune escape. J. Clin. Invest. 121:2480-

2492.

109. Zhang, S. C., E. Martin, M. Shimada, S. B. Godfrey, J. Fricke, S. Locastro, N. Y. Lai, P.

Liebesny, J. M. Carlson, C. J. Brumme, O. A. Ogbechie, H. Chen, B. D. Walker, Z. L.

Brumme, D. G. Kavanagh, and S. Le Gall. 2012. Aminopeptidase Substrate Preference

Affects HIV Epitope Presentation and Predicts Immune Escape Patterns in HIV-Infected

Individuals. J Immunol.

110. Pertel, T., C. Reinhard, and J. Luban. 2011. Vpx rescues HIV-1 transduction of dendritic

cells from the antiviral state established by type 1 interferon. Retrovirology 8:49.

111. Mathews, S. T., E. P. Plaisance, and T. Kim. 2009. Imaging systems for westerns:

chemiluminescence vs. infrared detection. Methods Mol Biol 536:499-513.

112. Walker, B. D., C. Flexner, K. Birch-Limberger, L. Fisher, T. J. Paradis, A. Aldovini, R.

Young, B. Moos, and R. T. Scholey. 1989. Long-term culture and fine specificity of

human cytotoxic T-lymphocyte clones reactive with human immunodeficiency virus

type 1. Proc Natl Acad Sci U S A 86:9514-9518.

113. Kourjian, G., Y. Xu, I. Mondesire-Crump, M. Shimada, P. Gourdain, and S. Le Gall. 2014.

Sequence-specific alterations of epitope production by HIV protease inhibitors. J

Immunol 192:3496-3506.

Page 100: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

References 92

114. Llano, A., A. Williams, A. Olvera, S. Silva-Arrieta, and C. Brander. 2013. Best-

Characterized HIV-1 CTL Epitopes: The 2013 Update. In HIV Molecular Immunology

2013. K. Yusim, B. Korber, C. Brander, D. H. Barouch, R. de Boer, B. F. Haynes, R. A.

Koup, J. P. Moore, B. D. Walker, and D. I. Watkins, eds. Los Alamos National

Laboratory, Theoretical Biology and Biophysics, Los Alamos, New Mexico. 3-25.

115. McCurley, N., and I. Mellman. 2010. Monocyte-derived dendritic cells exhibit increased

levels of lysosomal proteolysis as compared to other human dendritic cell populations.

PLoS One 5:e11949.

116. Yusim, K., B. T. M. Korber, C. Brander, D. Barouch, R. de Boer, B. F. Haynes, R. Koup, J.

P. Moore, B. D. Walker, and D. I. Watkins. 2013. HIV Molecular Immunology 2013. Los

Alamos National Laboratory, Theoretical Biology and Biophysics, Los Alamos, New

Mexico.

117. Goulder, P. J., M. Bunce, P. Krausa, K. McIntyre, S. Crowley, B. Morgan, A. Edwards, P.

Giangrande, R. E. Phillips, and A. J. McMichael. 1996. Novel, cross-restricted,

conserved, and immunodominant cytotoxic T lymphocyte epitopes in slow progressors

in HIV type 1 infection. AIDS Res Hum Retroviruses 12:1691-1698.

118. Friedrich, D., E. Jalbert, W. L. Dinges, J. Sidney, A. Sette, Y. Huang, M. J. McElrath, and

H. Horton. 2011. Vaccine-induced HIV-specific CD8+ T cells utilize preferential HLA

alleles and target-specific regions of HIV-1. J Acquir Immune Defic Syndr 58:248-252.

119. Brockman, M. A., A. Schneidewind, M. Lahaie, A. Schmidt, T. Miura, I. Desouza, F.

Ryvkin, C. A. Derdeyn, S. Allen, E. Hunter, J. Mulenga, P. A. Goepfert, B. D. Walker, and

T. M. Allen. 2007. Escape and compensation from early HLA-B57-mediated cytotoxic T-

lymphocyte pressure on human immunodeficiency virus type 1 Gag alter capsid

interactions with cyclophilin A. J Virol 81:12608-12618.

120. Miura, T., M. A. Brockman, A. Schneidewind, M. Lobritz, F. Pereyra, A. Rathod, B. L.

Block, Z. L. Brumme, C. J. Brumme, B. Baker, A. C. Rothchild, B. Li, A. Trocha, E. Cutrell,

N. Frahm, C. Brander, I. Toth, E. J. Arts, T. M. Allen, and B. D. Walker. 2009. HLA-

B57/B*5801 human immunodeficiency virus type 1 elite controllers select for rare gag

variants associated with reduced viral replication capacity and strong cytotoxic T-

lymphocyte [corrected] recognition. J Virol 83:2743-2755.

121. Pereyra, F., D. Heckerman, J. M. Carlson, C. Kadie, D. Z. Soghoian, D. Karel, A.

Goldenthal, O. B. Davis, C. E. DeZiel, T. Lin, J. Peng, A. Piechocka, M. Carrington, and B.

D. Walker. 2014. HIV control is mediated in part by CD8+ T-cell targeting of specific

epitopes. J Virol.

Page 101: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

References 93

122. Almeida, J. R., D. A. Price, L. Papagno, Z. A. Arkoub, D. Sauce, E. Bornstein, T. E. Asher,

A. Samri, A. Schnuriger, I. Theodorou, D. Costagliola, C. Rouzioux, H. Agut, A. G.

Marcelin, D. Douek, B. Autran, and V. Appay. 2007. Superior control of HIV-1

replication by CD8+ T cells is reflected by their avidity, polyfunctionality, and clonal

turnover. J Exp Med 204:2473-2485.

123. Chen, H., Z. M. Ndhlovu, D. Liu, L. C. Porter, J. W. Fang, S. Darko, M. A. Brockman, T.

Miura, Z. L. Brumme, A. Schneidewind, A. Piechocka-Trocha, K. T. Cesa, J. Sela, T. D.

Cung, I. Toth, F. Pereyra, X. G. Yu, D. C. Douek, D. E. Kaufmann, T. M. Allen, and B. D.

Walker. 2012. TCR clonotypes modulate the protective effect of HLA class I molecules

in HIV-1 infection. Nat Immunol 13:691-700.

124. Dong, T., G. Stewart-Jones, N. Chen, P. Easterbrook, X. Xu, L. Papagno, V. Appay, M.

Weekes, C. Conlon, C. Spina, S. Little, G. Screaton, A. van der Merwe, D. D. Richman, A.

J. McMichael, E. Y. Jones, and S. L. Rowland-Jones. 2004. HIV-specific cytotoxic T cells

from long-term survivors select a unique T cell receptor. J Exp Med 200:1547-1557.

125. Blum, J. S., P. A. Wearsch, and P. Cresswell. 2013. Pathways of antigen processing.

Annu Rev Immunol 31:443-473.

126. Lev, A., M. F. Princiotta, D. Zanker, K. Takeda, J. S. Gibbs, C. Kumagai, E. Waffarn, B. P.

Dolan, A. Burgevin, P. Van Endert, W. Chen, J. R. Bennink, and J. W. Yewdell. 2010.

Compartmentalized MHC class I antigen processing enhances immunosurveillance by

circumventing the law of mass action. Proc Natl Acad Sci U S A 107:6964-6969.

127. Butz, E. A., and M. J. Bevan. 1998. Differential presentation of the same MHC class I

epitopes by fibroblasts and dendritic cells. J Immunol 160:2139-2144.

128. Steers, N. J., J. R. Currier, G. H. Kijak, R. C. di Targiani, A. Saxena, M. A. Marovich, J. H.

Kim, N. L. Michael, C. R. Alving, and M. Rao. 2011. Cell type-specific proteasomal

processing of HIV-1 Gag-p24 results in an altered epitope repertoire. J Virol 85:1541-

1553.

129. Deol, P., D. M. Zaiss, J. J. Monaco, and A. J. Sijts. 2007. Rates of processing determine

the immunogenicity of immunoproteasome-generated epitopes. J Immunol 178:7557-

7562.

130. Sijts, A. J., T. Ruppert, B. Rehermann, M. Schmidt, U. Koszinowski, and P. M. Kloetzel.

2000. Efficient generation of a hepatitis B virus cytotoxic T lymphocyte epitope

requires the structural features of immunoproteasomes. J Exp Med 191:503-514.

131. Sijts, A. J., S. Standera, R. E. Toes, T. Ruppert, N. J. Beekman, P. A. van Veelen, F. A.

Ossendorp, C. J. Melief, and P. M. Kloetzel. 2000. MHC class I antigen processing of an

Page 102: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

References 94

adenovirus CTL epitope is linked to the levels of immunoproteasomes in infected cells.

J Immunol 164:4500-4506.

132. Morel, S., F. Levy, O. Burlet-Schiltz, F. Brasseur, M. Probst-Kepper, A. L. Peitrequin, B.

Monsarrat, R. Van Velthoven, J. C. Cerottini, T. Boon, J. E. Gairin, and B. J. Van den

Eynde. 2000. Processing of some antigens by the standard proteasome but not by the

immunoproteasome results in poor presentation by dendritic cells. Immunity 12:107-

117.

133. Galli, S. J., N. Borregaard, and T. A. Wynn. 2011. Phenotypic and functional plasticity of

cells of innate immunity: macrophages, mast cells and neutrophils. Nat Immunol

12:1035-1044.

134. Ziegler-Heitbrock, L., P. Ancuta, S. Crowe, M. Dalod, V. Grau, D. N. Hart, P. J. Leenen, Y.

J. Liu, G. MacPherson, G. J. Randolph, J. Scherberich, J. Schmitz, K. Shortman, S.

Sozzani, H. Strobl, M. Zembala, J. M. Austyn, and M. B. Lutz. 2010. Nomenclature of

monocytes and dendritic cells in blood. Blood 116:e74-80.

135. Chateau, M. T., V. Robert-Hebmann, C. Devaux, J. B. Lazaro, B. Canard, and O. Coux.

2001. Human monocytes possess a serine protease activity capable of degrading HIV-1

reverse transcriptase in vitro. Biochem Biophys Res Commun 285:863-872.

136. Dudziak, D., A. O. Kamphorst, G. F. Heidkamp, V. R. Buchholz, C. Trumpfheller, S.

Yamazaki, C. Cheong, K. Liu, H. W. Lee, C. G. Park, R. M. Steinman, and M. C.

Nussenzweig. 2007. Differential antigen processing by dendritic cell subsets in vivo.

Science 315:107-111.

137. Burster, T., A. Beck, E. Tolosa, P. Schnorrer, R. Weissert, M. Reich, M. Kraus, H.

Kalbacher, H. U. Haring, E. Weber, H. Overkleeft, and C. Driessen. 2005. Differential

processing of autoantigens in lysosomes from human monocyte-derived and

peripheral blood dendritic cells. J Immunol 175:5940-5949.

138. Lindstedt, M., K. Lundberg, and C. A. Borrebaeck. 2005. Gene family clustering

identifies functionally associated subsets of human in vivo blood and tonsillar dendritic

cells. J Immunol 175:4839-4846.

139. Duclos, S., G. Clavarino, G. Rousserie, G. Goyette, J. Boulais, V. Camossetto, E. Gatti, S.

LaBoissiere, P. Pierre, and M. Desjardins. 2011. The endosomal proteome of

macrophage and dendritic cells. Proteomics 11:854-864.

140. Papapostolou, D., O. Coux, and M. Reboud-Ravaux. 2002. Regulation of the 26S

proteasome activities by peptides mimicking cleavage products. Biochem Biophys Res

Commun 295:1090-1095.

Page 103: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

References 95

141. Steers, N. J., S. Ratto-Kim, M. S. de Souza, J. R. Currier, J. H. Kim, N. L. Michael, C. R.

Alving, and M. Rao. 2012. HIV-1 envelope resistance to proteasomal cleavage:

implications for vaccine induced immune responses. PLoS One 7:e42579.

142. Gourdain, P., J. Boucau, G. Kourjian, N. Y. Lai, E. Duong, and S. Le Gall. 2013. A real-

time killing assay to follow viral epitope presentation to CD8 T cells. J Immunol

Methods 398-399:60-67.

143. Marechal, V., F. Clavel, J. M. Heard, and O. Schwartz. 1998. Cytosolic Gag p24 as an

index of productive entry of human immunodeficiency virus type 1. J Virol 72:2208-

2212.

144. Iwasaki, A., and R. Medzhitov. 2010. Regulation of adaptive immunity by the innate

immune system. Science 327:291-295.

145. Schreibelt, G., J. Tel, K. H. Sliepen, D. Benitez-Ribas, C. G. Figdor, G. J. Adema, and I. J.

de Vries. 2010. Toll-like receptor expression and function in human dendritic cell

subsets: implications for dendritic cell-based anti-cancer immunotherapy. Cancer

Immunol Immunother 59:1573-1582.

146. Jarrossay, D., G. Napolitani, M. Colonna, F. Sallusto, and A. Lanzavecchia. 2001.

Specialization and complementarity in microbial molecule recognition by human

myeloid and plasmacytoid dendritic cells. Eur J Immunol 31:3388-3393.

147. Weck, M. M., F. Grunebach, D. Werth, C. Sinzger, A. Bringmann, and P. Brossart. 2007.

TLR ligands differentially affect uptake and presentation of cellular antigens. Blood

109:3890-3894.

148. Kwissa, M., H. I. Nakaya, H. Oluoch, and B. Pulendran. 2012. Distinct TLR adjuvants

differentially stimulate systemic and local innate immune responses in nonhuman

primates. Blood 119:2044-2055.

149. Coffman, R. L., A. Sher, and R. A. Seder. 2010. Vaccine adjuvants: putting innate

immunity to work. Immunity 33:492-503.

150. Akira, S., and K. Takeda. 2004. Toll-like receptor signalling. Nat Rev Immunol 4:499-

511.

151. Lee, H. K., J. M. Lund, B. Ramanathan, N. Mizushima, and A. Iwasaki. 2007. Autophagy-

dependent viral recognition by plasmacytoid dendritic cells. Science 315:1398-1401.

152. Kato, H., O. Takeuchi, S. Sato, M. Yoneyama, M. Yamamoto, K. Matsui, S. Uematsu, A.

Jung, T. Kawai, K. J. Ishii, O. Yamaguchi, K. Otsu, T. Tsujimura, C. S. Koh, C. Reis e Sousa,

Y. Matsuura, T. Fujita, and S. Akira. 2006. Differential roles of MDA5 and RIG-I helicases

in the recognition of RNA viruses. Nature 441:101-105.

Page 104: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

References 96

153. Reis, J., X. Q. Guan, A. F. Kisselev, C. J. Papasian, A. A. Qureshi, D. C. Morrison, C. W.

Van Way, 3rd, S. N. Vogel, and N. Qureshi. 2011. LPS-induced formation of

immunoproteasomes: TNF-alpha and nitric oxide production are regulated by altered

composition of proteasome-active sites. Cell Biochem Biophys 60:77-88.

154. Link, C., R. Gavioli, T. Ebensen, A. Canella, E. Reinhard, and C. A. Guzman. 2004. The

Toll-like receptor ligand MALP-2 stimulates dendritic cell maturation and modulates

proteasome composition and activity. Eur J Immunol 34:899-907.

155. Beninga, J., K. L. Rock, and A. L. Goldberg. 1998. Interferon-gamma can stimulate post-

proteasomal trimming of the N terminus of an antigenic peptide by inducing leucine

aminopeptidase. J Biol Chem 273:18734-18742.

156. Guillaume, B., J. Chapiro, V. Stroobant, D. Colau, B. Van Holle, G. Parvizi, M. P.

Bousquet-Dubouch, I. Theate, N. Parmentier, and B. J. Van den Eynde. 2010. Two

abundant proteasome subtypes that uniquely process some antigens presented by

HLA class I molecules. Proc Natl Acad Sci U S A 107:18599-18604.

157. Hickman, H. D., A. D. Luis, W. Bardet, R. Buchli, C. L. Battson, M. H. Shearer, K. W.

Jackson, R. C. Kennedy, and W. H. Hildebrand. 2003. Cutting edge: class I presentation

of host peptides following HIV infection. J Immunol 171:22-26.

158. Piccinini, M., M. T. Rinaudo, N. Chiapello, E. Ricotti, S. Baldovino, M. Mostert, and P. A.

Tovo. 2002. The human 26S proteasome is a target of antiretroviral agents. AIDS

16:693-700.

159. Andre, P., M. Groettrup, P. Klenerman, R. de Giuli, B. L. Booth, Jr., V. Cerundolo, M.

Bonneville, F. Jotereau, R. M. Zinkernagel, and V. Lotteau. 1998. An inhibitor of HIV-1

protease modulates proteasome activity, antigen presentation, and T cell responses.

Proc Natl Acad Sci U S A 95:13120-13124.

160. Heath, W. R., and F. R. Carbone. 2009. Dendritic cell subsets in primary and secondary

T cell responses at body surfaces. Nat Immunol 10:1237-1244.

161. Burgdorf, S., A. Kautz, V. Bohnert, P. A. Knolle, and C. Kurts. 2007. Distinct pathways of

antigen uptake and intracellular routing in CD4 and CD8 T cell activation. Science

316:612-616.

162. Chatterjee, B., A. Smed-Sorensen, L. Cohn, C. Chalouni, R. Vandlen, B. C. Lee, J. Widger,

T. Keler, L. Delamarre, and I. Mellman. 2012. Internalization and endosomal

degradation of receptor-bound antigens regulate the efficiency of cross presentation

by human dendritic cells. Blood 120:2011-2020.

Page 105: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

References 97

163. Rodriguez, A., A. Regnault, M. Kleijmeer, P. Ricciardi-Castagnoli, and S. Amigorena.

1999. Selective transport of internalized antigens to the cytosol for MHC class I

presentation in dendritic cells. Nat Cell Biol 1:362-368.

164. Norbury, C. C., B. J. Chambers, A. R. Prescott, H. G. Ljunggren, and C. Watts. 1997.

Constitutive macropinocytosis allows TAP-dependent major histocompatibility

complex class I presentation of exogenous soluble antigen by bone marrow-derived

dendritic cells. Eur J Immunol 27:280-288.

165. Norbury, C. C., L. J. Hewlett, A. R. Prescott, N. Shastri, and C. Watts. 1995. Class I MHC

presentation of exogenous soluble antigen via macropinocytosis in bone marrow

macrophages. Immunity 3:783-791.

166. Houde, M., S. Bertholet, E. Gagnon, S. Brunet, G. Goyette, A. Laplante, M. F. Princiotta,

P. Thibault, D. Sacks, and M. Desjardins. 2003. Phagosomes are competent organelles

for antigen cross-presentation. Nature 425:402-406.

167. Guermonprez, P., L. Saveanu, M. Kleijmeer, J. Davoust, P. Van Endert, and S.

Amigorena. 2003. ER-phagosome fusion defines an MHC class I cross-presentation

compartment in dendritic cells. Nature 425:397-402.

168. Huang, A. Y., A. T. Bruce, D. M. Pardoll, and H. I. Levitsky. 1996. In vivo cross-priming of

MHC class I-restricted antigens requires the TAP transporter. Immunity 4:349-355.

169. Sykulev, Y., M. Joo, I. Vturina, T. J. Tsomides, and H. N. Eisen. 1996. Evidence that a

single peptide-MHC complex on a target cell can elicit a cytolytic T cell response.

Immunity 4:565-571.

170. Kageyama, S., T. J. Tsomides, Y. Sykulev, and H. N. Eisen. 1995. Variations in the

number of peptide-MHC class I complexes required to activate cytotoxic T cell

responses. J Immunol 154:567-576.

171. Valitutti, S., S. Muller, M. Dessing, and A. Lanzavecchia. 1996. Different responses are

elicited in cytotoxic T lymphocytes by different levels of T cell receptor occupancy. J

Exp Med 183:1917-1921.

172. Faroudi, M., C. Utzny, M. Salio, V. Cerundolo, M. Guiraud, S. Muller, and S. Valitutti.

2003. Lytic versus stimulatory synapse in cytotoxic T lymphocyte/target cell

interaction: manifestation of a dual activation threshold. Proc Natl Acad Sci U S A

100:14145-14150.

173. Tel, J., G. Schreibelt, S. P. Sittig, T. S. Mathan, S. I. Buschow, L. J. Cruz, A. J. Lambeck, C.

G. Figdor, and I. J. de Vries. 2013. Human plasmacytoid dendritic cells efficiently cross-

present exogenous Ags to CD8+ T cells despite lower Ag uptake than myeloid dendritic

cell subsets. Blood 121:459-467.

Page 106: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

References 98

174. Segura, E., M. Durand, and S. Amigorena. 2013. Similar antigen cross-presentation

capacity and phagocytic functions in all freshly isolated human lymphoid organ-

resident dendritic cells. J Exp Med 210:1035-1047.

175. Steers, N. J., J. R. Currier, O. Jobe, S. Tovanabutra, S. Ratto-Kim, M. A. Marovich, J. H.

Kim, N. L. Michael, C. R. Alving, and M. Rao. 2014. Designing the epitope flanking

regions for optimal generation of CTL epitopes. Vaccine 32:3509-3516.

176. Karlsson, A. C., A. K. Iversen, J. M. Chapman, T. de Oliviera, G. Spotts, A. J. McMichael,

M. P. Davenport, F. M. Hecht, and D. F. Nixon. 2007. Sequential broadening of CTL

responses in early HIV-1 infection is associated with viral escape. PLoS One 2:e225.

177. Lichterfeld, M., D. G. Kavanagh, K. L. Williams, B. Moza, S. K. Mui, T. Miura, R.

Sivamurthy, R. Allgaier, F. Pereyra, A. Trocha, M. Feeney, R. T. Gandhi, E. S. Rosenberg,

M. Altfeld, T. M. Allen, R. Allen, B. D. Walker, E. J. Sundberg, and X. G. Yu. 2007. A viral

CTL escape mutation leading to immunoglobulin-like transcript 4-mediated functional

inhibition of myelomonocytic cells. J Exp Med 204:2813-2824.

178. Lu, W., L. C. Arraes, W. T. Ferreira, and J. M. Andrieu. 2004. Therapeutic dendritic-cell

vaccine for chronic HIV-1 infection. Nat Med 10:1359-1365.

179. Nestle, F. O., S. Alijagic, M. Gilliet, Y. Sun, S. Grabbe, R. Dummer, G. Burg, and D.

Schadendorf. 1998. Vaccination of melanoma patients with peptide- or tumor lysate-

pulsed dendritic cells. Nat Med 4:328-332.

180. Bonifaz, L., D. Bonnyay, K. Mahnke, M. Rivera, M. C. Nussenzweig, and R. M. Steinman.

2002. Efficient targeting of protein antigen to the dendritic cell receptor DEC-205 in

the steady state leads to antigen presentation on major histocompatibility complex

class I products and peripheral CD8+ T cell tolerance. J Exp Med 196:1627-1638.

181. Hanke, T. 2008. STEP trial and HIV-1 vaccines inducing T-cell responses. Expert Rev

Vaccines 7:303-309.

182. Horton, H., C. Havenar-Daughton, D. Lee, E. Moore, J. Cao, J. McNevin, T. Andrus, H.

Zhu, A. Rubin, T. Zhu, C. Celum, and M. J. McElrath. 2006. Induction of human

immunodeficiency virus type 1 (HIV-1)-specific T-cell responses in HIV vaccine trial

participants who subsequently acquire HIV-1 infection. J Virol 80:9779-9788.

183. Pereyra, F., D. Heckerman, J. M. Carlson, C. Kadie, D. Z. Soghoian, D. Karel, A.

Goldenthal, O. B. Davis, C. E. DeZiel, T. Lin, J. Peng, A. Piechocka, M. Carrington, and B.

D. Walker. 2014. HIV Control Is Mediated in Part by CD8+ T-Cell Targeting of Specific

Epitopes. J Virol 88:12937-12948.

Page 107: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

References 99

184. Schwartz, O., V. Marechal, S. Le Gall, F. Lemonnier, and J. M. Heard. 1996. Endocytosis

of major histocompatibility complex class I molecules is induced by the HIV-1 Nef

protein. Nat Med 2:338-342.

185. Hansen, S. G., J. C. Ford, M. S. Lewis, A. B. Ventura, C. M. Hughes, L. Coyne-Johnson, N.

Whizin, K. Oswald, R. Shoemaker, T. Swanson, A. W. Legasse, M. J. Chiuchiolo, C. L.

Parks, M. K. Axthelm, J. A. Nelson, M. A. Jarvis, M. Piatak, Jr., J. D. Lifson, and L. J.

Picker. 2011. Profound early control of highly pathogenic SIV by an effector memory T-

cell vaccine. Nature 473:523-527.

186. Hansen, S. G., M. Piatak, Jr., A. B. Ventura, C. M. Hughes, R. M. Gilbride, J. C. Ford, K.

Oswald, R. Shoemaker, Y. Li, M. S. Lewis, A. N. Gilliam, G. Xu, N. Whizin, B. J. Burwitz, S.

L. Planer, J. M. Turner, A. W. Legasse, M. K. Axthelm, J. A. Nelson, K. Fruh, J. B. Sacha, J.

D. Estes, B. F. Keele, P. T. Edlefsen, J. D. Lifson, and L. J. Picker. 2013. Immune

clearance of highly pathogenic SIV infection. Nature 502:100-104.

187. Hansen, S. G., C. Vieville, N. Whizin, L. Coyne-Johnson, D. C. Siess, D. D. Drummond, A.

W. Legasse, M. K. Axthelm, K. Oswald, C. M. Trubey, M. Piatak, Jr., J. D. Lifson, J. A.

Nelson, M. A. Jarvis, and L. J. Picker. 2009. Effector memory T cell responses are

associated with protection of rhesus monkeys from mucosal simian immunodeficiency

virus challenge. Nat Med 15:293-299.

188. Boaz, M. J., A. Waters, S. Murad, P. J. Easterbrook, and A. Vyakarnam. 2002. Presence

of HIV-1 Gag-specific IFN-gamma+IL-2+ and CD28+IL-2+ CD4 T cell responses is

associated with nonprogression in HIV-1 infection. J Immunol 169:6376-6385.

Page 108: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Supplemental Figures 100

7. Supplemental Figures

Supplemental Figure 1. Phenotypic analysis of DCs and Møs cell populations.

Immature DCs or Møs were phenotypically analyzed by flow cytometry. Cells were stained with

antibodies against human CD11c, BDCA-1 and CD14 (black lines) or isotype controls (gray lines)

to assess the purity of each cell population. The percentage of cells expressing each marker is

indicated. The level of surface expression of each marker is indicated as mean fluorescence

intensity (MFI) (upper right corner).

Page 109: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Supplemental Figures 101

Supplemental Figure 2. Infection of immature DCs and Møs with VSVg ΔEnv NL4.3.

Immature DCs (A) or Møs (B) were infected with VSVg ΔEnv NL4.3 in the presence of Vpx. On

day 1, 3, and 6 post infection the infection rate, measured by the expression of GFP, was

analyzed by flow cytometry.

(C) Bars represent the mean ± SD of two independent experiments from different donors.

Page 110: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Supplemental Figures 102

Supplemental Figure 3. Degradation patterns of a HIV-1 RT 16-mer fragment in cytosolic

extracts of immature and mature DCs and Møs.

Four nmol of 5-ATK9-2 (aa 153-168 in HIV-1 RT) were degraded with 30µg of cytosolic extracts

from iDCs, LPS-, or CL097-matured DCs (A) or iMøs, LPS-, or CL097-matured Møs (B) for 120

minutes. Peptides containing (black bars) or lacking intact ATK9 (white bars) were identified by

MS. Optimal ATK9 () is indicated. The data are representative of three independent

experiments from different donors.

Page 111: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Supplemental Figures 103

Supplemental Figure 4. Degradation of a HIV-1 Gag p24 35mer in DC cell extracts at pH4.0,

pH5.5 and pH7.4.

The HIV-1 peptide p24-35mer (MVHQAISPRTLNAWVKVVEEKAFSPEVIPMFAALS, aa 10-44 in Gag

p24) was degraded in 15µg of whole cell extracts from immature DCs for 60 minutes in

degradation buffer at pH4.0, pH5.5 or pH7.4. Peptides containing the epitopes B57-ISW9 and

B57-KF11 (black bars), B57-ISW9 epitope (blue bars), B57-KF11 epitope (red bars) or lacking

both epitopes (gray bars) were identified by MS. Optimal B57-ISW9 (blue star) and B57-KF11

(red star) are indicated. Data represent one of three independent experiments from different

donors.

Page 112: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Supplemental Figures 104

Supplemental Figure 5. Variable production of 16 HIV-1 epitopes in cytosolic and endo-

lysosomal cell extracts of DCs and Møs.

A. The map shows the location of 12 MHC I epitopes (black arrows) and 4 MHC II epitopes (gray

arrows) within the sequence of Gag p24-35mer (aa 10-44).

B. Summary of the relative amount of optimal epitopes and corresponding N-terminal

extensions detected by MS after 10, 30, 60, and 120 minutes degradation in extracts of

immature DCs, LPS-matured DCs, immature Møs, LPS-matured Møs at pH7.4, pH5.5 and pH4.0.

Epitope precursors, defined as peptides with the correct C-terminus and extended by up to

three residues at the N-terminus, could be further trimmed in the ER. Numbers represent the

contribution of optimals and N-extended optimals to the total intensity of all degradation

products at each time point. The presence of optimal epitopes is indicated (*). Data represent

one of three MS analyses from independent experiments.

Page 113: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Supplemental Figures 105

Supplemental Figure 6. Minor cleavage sites within p24-31mer results in less degradation of

B57-TW10-containing peptides.

Cleavage patterns of p24-31mer incubated with whole cell extracts from immature DCs for 30

minutes (left panel) or 120 minutes (right panel) at pH7.4, pH5.5, and pH4.0 are shown as the

contribution of each cleavage site, presented as cleavage N-terminal or C-terminal to a specific

amino acid, to the total intensity of all degradation products. Data are representative of three

independent experiments with three different donors.

Page 114: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

Abbreviations

AMC 7-amino-4-methylcoumarin

APC Antigen presenting cell

ATP Adenosine triphosphate

BDCA-1 Blood Dendritic Cell Antigen 1

CCR Chemokine receptor

CMV Cytomegalovirus

51Cr Chromium 51 isotope

CTL Cytotoxic T lymphocyte

DC Dendritic cell

DC-SIGN Dendritic cell-specific intercellular adhesion molecule-3-grabbing non-

integrin

DRiPs Defective ribosomal products

DTT Dithiothreitol

EBV Epstein-Barr virus

EDTA Ethylenediaminetetraacetic acid

ELISA Enzyme-Linked ImmunoSorbent assay

ELISPOT Enzyme-Linked ImmunoSpot

Env Envelope

ER Endoplasmic reticulum

ERAP Endoplasmic reticulum aminopeptidase

FBS Fetal bovine serum

Gag Group-specific antigen

GM-CSF Granulocyte macrophage colony-stimulating factor

HAART Highly active antiretroviral therapy

HEPES 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid

HIV Human immunodeficiency virus

HLA Human leukocyte antigen

HRP Horseradish peroxidase

HSA Human serum albumin

Hsp90 Heat shock protein 90

IFN-γ Interferon-gamma

IL Interleukin

IRAP Insulin-regulated aminopeptidase

Page 115: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing

LAP Leucine aminopeptidase

LCMV Lymphocytic choriomeningitis

LPS Lipopolysaccharide

Mø Macrophage

MS Mass spectrometer

NOX2 NADPH oxidase 2

ODP o-Phenylenediamine

PA28 Proteasome activator

PAMPs Pathogen-associated molecular patterns

pDC Plasmacytoid dendritic cell

PBMCs Peripheral blood mononuclear cells

PBS Phosphate Buffered Saline

PSA Puromycin-sensitive aminopeptidase

PVDF Polyvinylidene fluoride

RLR RIG-I-like receptor

RP-HPLC Reversed-phase high-performance liquid chromatography

SAMHD1 SAM domain and HD domain-containing protein 1

SFC Spot-forming cell

TAP Transporter associated with antigen processing

TCA Trichloroacetic acid

TCR T-cell receptor

TFA Trifluoroacetic acid

TLR Toll-like receptor

TOP Thimet oligopeptidase

TPPII Tripeptidyl-peptidase II

Page 116: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing
Page 117: Processing and presentation of epitopes by dendritic cells ......Processing and presentation of epitopes by dendritic cells and macrophages in HIV infection vorgelegt von Dipl.-Ing