10
In eukaryotes, the process of transcriptional regula- tion, DNA recombination, condensation and repli- cation relies on changes in chromatin structure that do not involve alterations in the DNA sequence but are nevertheless inherited 1 . Such regulation is said to be beyond genetics (epigenetics) and has been exten- sively studied in recent years, starting a hunt for fac- tors that act as modulators of epigenetic information. In the nucleus of every eukaryotic cell, a module of approximately 147 bp of DNA is wrapped around an octamer of core histone proteins, forming a nucleo- some, which is the basic repetitive unit of chromatin (FIG. 1). Histone 1 (H1) binds to the DNA loop between two neighbouring nucleosomes. Histones and DNA can undergo several types of modification, which can be faithfully transmitted to cell progeny. The Allis laboratory proposed a hypothesis known as the histone code, in which post-translational modifications (PTMs) of histones act in a combinatorial or sequential man- ner to specify unique downstream activities that affect transcription activation, chromatin condensation and signalling for DNA repair 2 . The code is ‘written’ by histone-modifying enzymes and ‘read’ by histone-binding proteins (FIG. 2). Trypanosoma brucei is a unicellular eukaryotic para- site that causes sleeping sickness in humans and nagana in cattle in sub-Saharan Africa. Sleeping sickness is a vector-borne disease that affects 70 million people and is responsible for approximately 50,000 deaths annu- ally 3 . The study of this substantial economical and medical threat has been facilitated by the development of genetic tools that allow functional studies and genetic screens (BOX 1). Therefore, T. brucei is a useful model organism to study basic biological processes of other Kinetoplastida and of higher eukaryotes. T. brucei inhabits the bloodstream and interstitial spaces of the mammalian host and is covered by a dense coat of a glycosylphosphatidylinositol (GPI)-anchored variant surface glycoprotein (VSG) (TABLE 1). Throughout its life cycle, T. brucei changes forms and must adapt to different host environments. By unknown means, a fraction of the proliferating slender bloodstream form commits to a differentiation process that generates a quiescent, cell cycle-arrested population (stumpy form) that is reactivated after ingestion by their insect vector, the tsetse. In the midgut of the fly, the parasite seems to sense both the drop in temperature and a chemical cue that trigger its differentiation into a proliferative, procyclic form 4,107 . The procyclic form does not express VSGs, but its surface is covered by GPI-anchored proteins called EP (rich in Glu-Pro repeats) or GPEET (rich in Gly-Pro-Glu-Glu-Thr repeats) procyclins. After migrat- ing to the salivary glands, the parasites further differen- tiate into and proliferate as epimastigotes, the surface of which seems to consist of alanine-rich protein (BARP) 5 . When epimastigotes differentiate into the non-replicative metacyclic form, BARP is replaced by VSG. When the tsetse takes another meal, the metacyclics are injected into the mammal, where they differentiate into the blood- stream form. Throughout its life cycle, T. brucei alternates in a tightly regulated manner between cell proliferation and cell differentiation. Differentiation in T. brucei is prob- ably a result of responses to different environments and involves a coordinated cascade of signalling events and transcriptional and post-transcriptional machiner- ies. These features probably exist in all biological systems. *Laboratory of Molecular Parasitology, The Rockefeller University, 1230 York Avenue, New York, New York 10065, USA. University of Munich, Biocenter, Department of Genetics, Groβhardener Straβe 2‑4, 82152, Martinsried, Germany. Correspondence to L.M.F. e‑mail: luisa.figueiredo@ rockefeller.edu doi:10.1038/nrmicro2149 Post-translational modification An enzymatic modification of an amino acid residue that occurs after a protein is synthesized, and often modifies the function or lifespan of a protein. Variant surface glycoprotein (VSG). The most abundant protein (approximately 10 million identical copies on individual cells) at the surface of the bloodstream slender, stumpy and metacyclic life cycle stages of Trypanosoma brucei. Periodic change of the VSG surface coat is a crucial part of the evasion mechanism known as antigenic variation. Epigenetic regulation in African trypanosomes: a new kid on the block Luisa M. Figueiredo*, George A. M. Cross* and Christian J. Janzen Abstract | Epigenetic regulation is important in many facets of eukaryotic biology. Recent work has suggested that the basic mechanisms underlying epigenetic regulation extend to eukaryotic parasites. The identification of post-translational histone modifications and chromatin-modifying enzymes is beginning to reveal both common and novel functions for chromatin in these parasites. In this Review, we compare the role of epigenetics in African trypanosomes and humans in several biological processes. We discuss how the study of trypanosome chromatin might help us to better understand the evolution of epigenetic processes. REVIEWS 504 | JULY 2009 | VOLUME 7 www.nature.com/reviews/micro © 2009 Macmillan Publishers Limited. All rights reserved.

Epigenetic regulation in African trypanosomes: a new kid on the block

Embed Size (px)

Citation preview

Page 1: Epigenetic regulation in African trypanosomes: a new kid on the block

In eukaryotes, the process of transcriptional regula­tion, DNA recombination, condensation and repli­cation relies on changes in chromatin structure that do not involve alterations in the DNA sequence but are nevertheless inherited1. Such regulation is said to be beyond genetics (epigenetics) and has been exten­sively studied in recent years, starting a hunt for fac­tors that act as modulators of epigenetic information. In the nucleus of every eukaryotic cell, a module of approximately 147 bp of DNA is wrapped around an octamer of core histone proteins, forming a nucleo­some, which is the basic repetitive unit of chromatin (FIG. 1). Histone 1 (H1) binds to the DNA loop between two neighbouring nucleosomes. Histones and DNA can undergo several types of modification, which can be faithfully transmitted to cell progeny. The Allis labora tory proposed a hypothesis known as the histone code, in which post-translational modifications (PTMs) of histones act in a combinatorial or sequential man­ner to specify unique downstream activities that affect transcription activation, chromatin condensation and signalling for DNA repair2. The code is ‘written’ by histone­modifying enzymes and ‘read’ by histone­binding proteins (FIG. 2).

Trypanosoma brucei is a unicellular eukaryotic para­site that causes sleeping sickness in humans and nagana in cattle in sub­Saharan Africa. Sleeping sickness is a vector­borne disease that affects 70 million people and is responsible for approximately 50,000 deaths annu­ally3. The study of this substantial economical and medical threat has been facilitated by the development of genetic tools that allow functional studies and genetic screens (BOX 1). Therefore, T. brucei is a useful model

organism to study basic biological processes of other Kinetoplastida and of higher eukaryotes.

T. brucei inhabits the bloodstream and interstitial spaces of the mammalian host and is covered by a dense coat of a glycosylphosphatidylinositol (GPI)­anchored variant surface glycoprotein (VSG) (TABLE 1). Throughout its life cycle, T. brucei changes forms and must adapt to different host environments. By unknown means, a fraction of the proliferating slender bloodstream form commits to a differentiation process that generates a quiescent, cell cycle­arrested population (stumpy form) that is reactivated after ingestion by their insect vector, the tsetse. In the midgut of the fly, the parasite seems to sense both the drop in temperature and a chemical cue that trigger its differentiation into a proliferative, pro cyclic form4,107. The procyclic form does not express VSGs, but its surface is covered by GPI­anchored proteins called EP (rich in Glu­Pro repeats) or GPEET (rich in Gly­Pro­Glu­Glu­Thr repeats) procyclins. After migrat­ing to the salivary glands, the parasites further differen­tiate into and proliferate as epimastigotes, the surface of which seems to consist of alanine­rich protein (BARP)5. When epimastigotes differentiate into the non­replicative metacyclic form, BARP is replaced by VSG. When the tsetse takes another meal, the metacyclics are injected into the mammal, where they differentiate into the blood­stream form. Throughout its life cycle, T. brucei alternates in a tightly regulated manner between cell proliferation and cell differentiation. Differentiation in T. brucei is prob­ably a result of responses to different environments and involves a coordinated cascade of signalling events and transcriptional and post­transcriptional machiner­ies. These features probably exist in all biological systems.

*Laboratory of Molecular Parasitology, The Rockefeller University, 1230 York Avenue, New York, New York 10065, USA. ‡University of Munich, Biocenter, Department of Genetics, Groβhardener Straβe 2‑4, 82152, Martinsried, Germany. Correspondence to L.M.F. e‑mail: [email protected]:10.1038/nrmicro2149

Post-translational modificationAn enzymatic modification of an amino acid residue that occurs after a protein is synthesized, and often modifies the function or lifespan of a protein.

Variant surface glycoprotein(VSG). The most abundant protein (approximately 10 million identical copies on individual cells) at the surface of the bloodstream slender, stumpy and metacyclic life cycle stages of Trypanosoma brucei. Periodic change of the VSG surface coat is a crucial part of the evasion mechanism known as antigenic variation.

Epigenetic regulation in African trypanosomes: a new kid on the blockLuisa M. Figueiredo*, George A. M. Cross* and Christian J. Janzen‡

Abstract | Epigenetic regulation is important in many facets of eukaryotic biology. Recent work has suggested that the basic mechanisms underlying epigenetic regulation extend to eukaryotic parasites. The identification of post-translational histone modifications and chromatin-modifying enzymes is beginning to reveal both common and novel functions for chromatin in these parasites. In this Review, we compare the role of epigenetics in African trypanosomes and humans in several biological processes. We discuss how the study of trypanosome chromatin might help us to better understand the evolution of epigenetic processes.

R E V I E W S

504 | july 2009 | VoluME 7 www.nature.com/reviews/micro

© 2009 Macmillan Publishers Limited. All rights reserved.

Page 2: Epigenetic regulation in African trypanosomes: a new kid on the block

Nature Reviews | Microbiology

Nucleosome

Histones

DNA

Spacer DNA

Histoneoctamerplus 147 bp of DNA

H1

TsetseThe insect vector (Glossina spp.) that ensures the transmission of Trypanosoma brucei between two mammalian hosts. The word tsetse means fly and it originates from the Tswana language.

ProcyclinThe most abundant protein at the surface of the procyclic life cycle form. There are two types of procyclin: EP and GPEET. These glycosylated proteins are not subject to allelic exclusion.

EpimastigoteThe Trypanosoma brucei life cycle stage that colonizes the tsetse salivary glands.

MetacyclicThe Trypanosoma brucei life cycle stage in the tsetse salivary glands that reinfects the mammalian host.

Determination of stem cell fate6 and T cell development7 are two well­known examples.

The genome of T. brucei contains approximately 8,000 genes that are mostly transcribed polycistroni­cally by RNA polymerase II (Pol II)8. unlike bacterial operons, most genes in a polycistronic unit are not func­tionally related, and gene regulation occurs mainly at the post­transcriptional level9. The only known exceptions are procyclin and VSG genes, which are regulated at the transcriptional level. These genes are transcribed by Pol I, which is used exclusively for the transcription of ribosomal DNA (rDNA) in other organisms.

Given that Pol II transcription in T. brucei seems to be essentially constitutive and regulation is mainly post­transcriptional, it might be expected that, in contrast to the situation in higher eukaryotes, there will be a pau­city of histone PTMs and chromatin­modifying and chromatin­remodelling enzymes dedicated to repress­ing Pol II transcription. Consistent with this prediction, T. brucei has a smaller set of histone PTMs10,11 and fewer proteins that write and read the histone code. Therefore, T. brucei is a useful model system in which to study epigenetic phenomena. In this Review, we discuss the current knowledge of the role of epigenetic mechanisms in T. brucei, specifically in the regulation of Pol I transcription, cell differentiation and the cell cycle.

The repertoire of epigenetic regulatorsThe physiological role of chromatin at a particular locus depends on the activity of a set of epigenetic regulators that act at different levels, from nucleosomal positioning and remodelling to writing and reading histone modifica­tions of single amino acids. DNA nucleotide modifications also carry epigenetic information.

Chromatin-remodelling enzymes. Chromatin remod­ellers are specialized complexes that alter the struc­ture, composition and positioning of nucleosomes and thereby facilitate access of the transcription machinery to nucleosomal DNA12. Remodellers are multiprotein complexes containing a core ATPase that functions as a motor to alter histone–DNA contacts. Most eukaryo­tes have several classes of remodellers, including the SWI/SNF (switch/sucrose non­fermentable) complex, and the ISWI (imitation SWI), NuRD/MI2/CHD, INo80, SWR1 and RAD54­related proteins. These complexes are involved in a wide range of biological functions, includ­ing gene expression, splicing, DNA repair or signalling12. SWI/SNF and ISWI are the best­studied remodellers and, in general, SWI/SNF remodellers alter nucleosome positioning to promote transcription factor binding and transcription13, whereas members of the ISWI family primarily mediate transcriptional repression14.

In each class of remodeller, the catalytic subunit has a SNF2­like domain and a unique domain that interacts with specific chromatin substrates. SWI/SNF has a bromo domain, CHD has a chromodomain and ISWI has a SANT and/or SlIDE domain15. T. brucei has 13 genes with a putative SNF2 domain (TABLE 2), of which two have been characterized: ISWI16 and jBP2 (base j binding protein 2)17. ISWI has a SlIDE motif,

previously labelled as a MyB­like domain by earlier versions of the 3D­jIGSAW database (G. Rudenko, personal communication). jBP2 lacks any recognizable chromatin­binding domain, but sequence homology of the SNF2 domain suggests that jBP2 belongs to the RAD54 class (R. Sabatini, personal communication). As discussed below, both proteins affect the chroma­tin structure of specialized subtelomeric loci. A second RAD54­related enzyme has been recently identified on the basis of sequence homology.

Histone-modifying enzymes. Every eukaryote has a conserved set of enzymes that methylate, acetylate, phosphorylate and ubiquitylate histones. Genome searches suggest that African trypanosomes have a smaller set of histone­modifying enzymes than mam­malian cells (TABLE 2). All putative genes were named according to new nomenclature18; previously character­ized genes will be referred to using both new and old nomenclature.

Five lysine acetyltransferases (KATs) and seven histone deacetylases (HDACs) have been found in T. brucei19–21. The KATs belong to two classes: three are members of the MyST family (HAT1–3; also known as KAT1–3), which localize in the nucleus20,21, and two are uncharacterized homologues of the human histone acetyltransferase ElP3 (ElP3a (also known as KAT9a) and ElP3b (also known as KAT9b)). T. brucei HDACs share homology with class I (HDAC1–2) or class II (HDAC3–4) histone deactetylases found in yeast and mammals19. SIR2­related histone deacetylases are also present in T. brucei. SIR2rp1 possesses histone deacety­lase activity and localizes in the nucleus, whereas SIR2rp2 and SIR2rp3 are mitochondrial proteins22,23.

Figure 1 | Chromatin structure. The nucleosomes are the basic units of chromatin. Each nucleosome is composed of approximately 147 bp of DNA wrapped around an octamer of histones (two copies of H2A, H2B, H3 and H4). Histone H1 binds to the DNA that links the two nucleosomes.

R E V I E W S

NATuRE REVIEWS | MiCrobiology VoluME 7 | july 2009 | 505

© 2009 Macmillan Publishers Limited. All rights reserved.

Page 3: Epigenetic regulation in African trypanosomes: a new kid on the block

Me

Nature Reviews | Microbiology

NH

O

O

NH3+

Non-modifiedlysine (H3K76me0)

NH

O

O

NH2+H3C

Monomethyl lysine(H3K76me1)

NH

O

O

NH+

CH3

H3C

Dimethyl lysine(H3K76me2)

NH

O

O

N+

CH3

CH3H3C

Trimethyl lysine(H3K76me3)

a

b

KMT KMT KMT

Me

Me

Edman degradationA method developed by P. Edman to sequence peptides by sequential labelling and cleavage of single residues from the amino-terminal end of polypeptides.

In humans and trypanosomes, there are two classes of lysine methyltransferase (KMT) that can add one, two or three methyl groups onto lysine residues: those that contain the SET (su(var)3–9, enhancer of zeste, trithorax) domain and the DoT1 (disruptor of telo­meric silencing) homologues. Twenty to twenty­seven genes encoding a SET domain can be found in the T. brucei genome, but none has been characterized. Two DoT1 (also known as KMT4) homologues have been described in T. brucei, DoT1A (also known as KMT4A) and DoT1B (also known as KMT4B), the substrate specificity and function of which will be addressed below. Mammals have two types of lysine demethylase: the jumonji family and the lSD1 homo­logues24. Although lSD1 homologues have not been found in T. brucei, four genes have been identified that encode putative jmjC domain­containing proteins (jumonji family).

Arginine methylation occurs in various proteins and is controlled by protein arginine methyltransferases (PRMTs)25. In histones, arginine methylation has been associated with both transcriptional activation and repression26. Five candidate PRMTs are present in the T. brucei genome, three of which are homologues of mammalian PRMT1, PRMT5 and PRMT7 (REFS 27–29). Although recombinant forms of the three PRMTs from T. brucei methylate arginines of bovine histones in vitro,

arginine methylation of trypanosome histones has not been detected. Therefore, it remains unclear whether arginine methylation has a role in epigenetic regulation in T. brucei.

Histone modifications. The histone repertoire of T. brucei consists of the four canonical histones (H2A, H2B, H3 and H4) and four histone variants (H2Az, H2Bv, H3v and H4v)30–32. As in other eukaryo­tes, T. brucei also has divergent H1 linker histones (FIG. 1). Phylogenetic analyses and the structure and function of trypanosome core histones and their variants have been described in detail elsewhere32,33. Although core histones are among the most evolutionarily conserved proteins, there are substantial differences between trypanosome histones and those of higher eukaryotes, especially in the highly modified amino­terminal tails11,34. A survey of T. brucei PTMs using Edman degradation and mass spectrometry revealed some unexpected results: a strik­ing absence of many well­conserved modifications and some unusual and apparently trypanosome­specific PTMs10,11 (FIG. 3). Similar observations were made for histone H4 in a closely related species, Trypanosoma cruzi 35. However, some PTMs are conserved between T. brucei and humans and might represent an ancient basal repertoire of histone modifications. As T. brucei mainly regulates gene expression post­transcriptionally, the functions of the few PTMs that have been described and whether they can be predicted by analogy to higher eukaryotes are important areas of research.

In T. brucei, the first amino acid (alanine) of histone H2A can be either methylated or acetylated, whereas the serine at position 1 in human H2A can be phosphory­lated, although the function of these modifications is unknown. Perhaps the most surprising differences between human and T. brucei H2A are the modifications found at the carboxyl terminus, where T. brucei H2A is heavily acetylated in a complex pattern. The function (or functions) of these acetylation marks remains a mystery, but they might contribute to various levels of accessibil­ity of the modified chromatin for protein complexes to promote processes such as transcription, replication or recombination. Homologous modifications to the other human H2A marks have not been identified to date.

of the trypanosome histones, H2B is one of the least conserved, and only four PTMs have been detected: A1 is methylated and K4, K12 and K16 are acetylated. No clear homologues of these PTMs can be found in human H2B, suggesting that these marks have T. brucei­specific functions. Surprisingly, T. brucei H2B lacks one of the most conserved lysine residues, K120, which is ubiquitylated in various organisms. Saccharomyces cer-evisiae H2BK123ub (monoubiquitylated histone H2B on lysine 123) is a well­investigated example of PTMs that influence the level of other modifications, so­called PTM crosstalk36, because it is required for the efficient meth­ylation of H3K4 and H3K79 (REFS 37,38). Although there seems to be a homologous lysine in the trypanosome H2B variant (H2BvK129), it was not observed to be ubiquitylated, and mutation of K129 caused no change in phenotype. H2Bv, however, specifically associates

Figure 2 | The histone code hypothesis. a | The amino-terminal histone tails protrude from the core of nucleosomes and undergo multiple post-translational modifications (PTMs), which define the role of chromatin for that genomic locus. The enzymes that write this code are called histone-modifying enzymes. For example, a lysine methyltransferase (KMT; light blue) transfers a methyl group (yellow) from a donor molecule (green) to a lysine in the histone tail. Some PTMs are read by a histone-binding protein (purple), which will subsequently interact with other factors to produce the appropriate biological response. b | Lysines can be mono-, di- or trimethylated by the same or different KMTs. Part b adapted, with permission, from Nature Reviews Molecular Cell Biology REF. 105 (2007) Macmillan Publishers Ltd. All rights reserved.

R E V I E W S

506 | july 2009 | VoluME 7 www.nature.com/reviews/micro

© 2009 Macmillan Publishers Limited. All rights reserved.

Page 4: Epigenetic regulation in African trypanosomes: a new kid on the block

EuchromatinA form of chromatin that was first defined as lightly stained nuclear regions by light microscopy and is usually associated with transcriptionally competent chromosome loci.

HeterochromatinA more compact form of chromatin that, when using light or electron microscopy, appears as darker regions of the nucleus. Heterochromatin is usually associated with transcriptionally silent chromosome loci.

with H3 that was enriched in H3K4 and H3K76 tri­methylation39, suggesting that trypanosomes regulate H3K4 and H3K76 methylation by incorporating H2Bv into nucleosomes.

In humans, the N­terminal tail of histone H3 has almost two dozen PTMs, many of which are involved in transcriptional regulation. In T. brucei, only a few amino acids of the N­terminal tail of human H3 are conserved. H3K4 is trimethylated in T. brucei and, although H3K4 methylation is a hallmark of trans­criptional activation in many organisms, the function of this PTM in T. brucei is unknown. Homologues of well­conserved amino acids, such as K9 and S10, seem to be absent from T. brucei H3, and the only other PTMs detected in the N­terminal tail are acetyla­tion of K23 and trimethylation of K32. on the basis of sequence alignment, the T. brucei homologues of human H3K23, H3K27 and H3K36 are H3K19, H3K23 and H3K32, respectively. Acetylation of H3K27 is associated with transcription in several organisms and is catalysed by GCN5 (also known as KAT2A) in humans40. Human H3K36 can be methylated by SET2 (also known as KMT3) and might be involved in tran­scriptional elongation of Pol II41. Despite the presence of acetylated H3K23, homologues of GCN5 or SET2 have not been described in trypanosomes, and the function of the corresponding PTMs is unknown. one of the best­characterized PTMs in T. brucei is methyl­ation of H3K76, the homologue of H3K79, which is mainly monomethylated or dimethylated in humans. Methylation of H3K79 is mediated by DoT1 methyl­transferases and is associated with transcribed chro­matin42. Furthermore, DoT1 is involved in meiotic checkpoint control and the DNA damage response43,44. In T. brucei, methylation of H3K76 is mediated by two enzymes: DoT1A and DoT1B, which are exclu­sively responsible for dimethylation or trimethyl­ation, respectively45. Although the two enzymes and their target seem to be conserved, trypanosomes have developed additional functions for this PTM. one of the main functions of DoT1A and DoT1B may be to influence the trypanosome cell cycle by regulating the level of H3K76 methylation (see below). Additionally, cells in which DoT1B has been depleted have defec­tive VSG switching and differentiation (see below). In addition to these PTMs, mass spectrometry has revealed the presence of other modification states on

the lysine­rich N­terminal tail of H3, but pinpoint­ing their localization to individual lysine residues was not possible11.

H4 is the most conserved trypanosome histone and, as for H2A and H2B, H4 is also highly methylated at A1 (REFS 10,11). H4K2 can be either methylated or acetylated, which could possibly have antagonistic functions, as is the case for H3K9me (methylated H3K9) and H3K9ac (acetylated H3K9) in humans46,47. H4K4 of T. brucei is acetylated by HAT3 (also known as KAT3), and might be the homologue of H4K5 in other organisms (the acetyla­tion of H4K5 promotes the incorporation of newly syn­thesized histones into chromatin48). Although H4K4 is highly acetylated (80–100%), the deletion of HAT3 and the subsequent loss of K4 acetylation, with no compen­satory change in the modification status of K5, has no impact on viability21. Another well­described and con­served PTM is the acetylation of H4K12, for which the T. brucei homologue may be H4K10. Acetylation of K12 is mediated by different KATs in humans (HAT1 (also known as KAT1), TIP60 (also known as PlIP or KAT5) and HBo1 (also known as MyST2 or KAT7) and is involved in histone deposition, DNA repair and trans­criptional activation (reviewed in REF. 49). In T. brucei, H4K10 is acetylated exclusively by HAT2 (also known as KAT2), which is essential for growth. RNA interference (RNAi)­mediated depletion of HAT2 results in an accu­mulation of cells before cytokinesis20. Genome­wide studies revealed that this PTM is enriched at probable Pol II transcription start sites, defining an open chro­matin structure that may be necessary for initiating transcription50. The fact that trypanosomes use distinct acetyltransferases for H4K4 and H4K10 suggests little redundancy in acetyltransferase activity and supports the idea of a simplified non­redundant histone code. There seems to be a T. brucei homologue (H4K14ac) of human H4K16ac, another euchromatin marker in humans, but neither the corresponding acetyltrans­ferase nor the function of this PTM are known in trypanosomes. There is also a clear T. brucei homo­logue (H4K18me3) of human H4K20me3. In mammals, H4K20 is dimethy lated or trimethylated by SuV420H1 (also known as KMT5B) and SuV420H2 (also known as KMT5C) and is involved in heterochromatin forma­tion, DNA repair, gene regulation and development51–53. However, a T. brucei KMT that is responsible for H4K18 methylation has not been found. Identification of this enzyme should be straightforward, given the few poten­tial SET domain­containing proteins in the T. brucei genome (TABLE 2).

Readers of the histone code. The histone code suggests that at least some of the histone PTMs are read by effector molecules and their associated complexes, which define a functional state of the chromatin and eventually trigger a physiological response. Effector proteins can alter the properties of chromatin by interconnecting two or more nucleosomes, by promoting the recruitment of RNA polymerase and transcription factors, by recruiting chromatin remodellers or by promoting downstream chemical modfications54. Readers of the

Box 1 | Genetic tools to study Trypanosoma brucei

T. brucei has several properties that make it amenable to genetic manipulation.• It is easy to culture and clone in liquid medium93 or on agar plates94.

• Homologous recombination is efficient and specific, which allows deletion or epitope tagging of endogenous genes after transfection by electroporation.

• T. brucei has an intrinsic RNAi system that can be exploited for mRNA knockdown95,96.

• Several markers for drug resistance are available for positive and negative selection.

• The ‘cre-lox’ system works efficiently and can be used for various purposes, principally to allow a marker for drug resistance to be removed and reused, or to tag genes without materially affecting upstream or downstream regulatory sequences97.

• Inducible expression can be used to obtain conditional gene knockouts.

R E V I E W S

NATuRE REVIEWS | MiCrobiology VoluME 7 | july 2009 | 507

© 2009 Macmillan Publishers Limited. All rights reserved.

Page 5: Epigenetic regulation in African trypanosomes: a new kid on the block

histone code use different domains to recognize and bind to specific PTMs55. Bromodomains bind to acetyl groups and are usually associated with active chromatin. Chromodomains, MBT (malignant brain tumour) fingers and PHD (plant homeo domain) bind to methyl lysine, and Tudor domains recognize methylated arginine or lysine. T. brucei has three classes of putative histone reader: bromodomain­, PHD finger domain­ and Tudor domain­containing proteins (TABLE 2), none of which has been characterized. unlike many proteins in higher eukaryotes54, all putative T. brucei readers seem to have a single PTM­binding domain. The absence of complex binding properties further supports the idea of a simplified histone code.

DNA modifications. DNA methylation is one of the best­characterized epigenetic markers. It occurs in higher eukaryotes, bacteria and archaea but is absent from some organisms, including S. cerevisiae and Caenorhabditis elegans56. DNA methylation plays an important part in the control of gene expression, but it is also involved in the regulation of DNA replication, repair and transposition, and in chromatin packag­ing57. Although DNA methylation has been observed in T. cruzi 58, for many years it was thought that DNA methylation was absent from T. brucei. Prompted by the discovery of a putative cytosine­5 DNA methyl­transferase gene, 5­methylcytosine was recently identified in nuclear DNA of T. brucei in both the bloodstream and procyclic form59.

The bloodstream form of T. brucei and other related species have a unique DNA modification: β­d­glucopyranosyloxymethyluracil or base j60,61. This is a bulkier DNA modification than 5­methylcytosine (FIG. 4), and it is found predominantly at telomeric repeats and, to a lesser extent, in other repetitive sequences. It also occurs in silent telomeric VSG genes, but the importance of this observation remains unclear61. Two base j­binding pro­teins (jBP1 and jBP2) were recently shown to be necessary for the synthesis of base j62,63. Because jBP2 requires the SNF2 ATPase domain for its activity, it has been proposed that this potential remodelling domain might recognize distinct chromatin elements and thereby determine the site specificity of base j deposition17. Base j is absent from jBP1 and jBP2 null cells64, but the functional con­sequences of a lack of base j remain to be determined. By contrast, base j is essential in Leishmania tarentolae65, another member of the Kinetoplastida.

Monoallelic expression of surface proteinsIn most eukaryotes, Pol I exclusively transcribes ribo­somal RNA (rRNA) genes. In T. brucei, Pol I also trans­cribes the genes encoding two major life cycle­specific surface proteins: VSGs and procyclins.

The rRNA gene family in humans contains hun­dreds to thousands of copies, and their transcription is under epigenetic control; individual genes exist in either an active or silent transcriptional state, and this transcriptional state is inheritable and revers­ible66. Recent studies have revealed that the transcrip­tional state of rDNA depends on a complex interplay of DNA­ and histone­modifying enzymes that act together with chromatin­remodelling complexes (reviewed in REF. 66).

T. brucei has eight rRNA genes67 dispersed among four chromosomes, and indirect evidence suggests that they are not all equivalent. When ectopic induc­ible genes are introduced into the non­transcribed region of T. brucei rDNA, the expression of the induc­ible gene is tightly regulated in some but not all cell lines68. This variability may reflect different epigenetic states of adjacent rRNA genes. Species of Plasmodium, the eukaryotic parasite that causes malaria, also have a small rRNA gene family (four genes). Remarkably, these genes belong to two distinct classes, the expres­sion of which is regulated in a stage­specific man­ner69,70. It remains to be seen whether T. brucei rRNA genes also fall into differently regulated categories.

Trypanosomes process RNA transcripts in a fun­damentally different way to most other eukaryotes. The mRNA molecule has a 39­nucleotide spliced­leader capped RNA at the 5′ end, which is transferred from a longer precursor by trans­splicing (reviewed in REF. 9). Therefore, capping and mRNA synthesis are uncoupled in trypanosomes, in contrast to most eukaryotes, in which the mRNA capping machinery associates with Pol II71. This feature enables T. brucei to efficiently process Pol I transcripts that encode proteins such as VSGs and procyclins72,73 (TABLE 1).

VSGs are key molecules for antigenic variation, which allows the parasite to escape the mammalian immune

Table 1 | Developmental stages of the Trypanosoma brucei life cycle*

life cycle and parasitic form Surface glycoprotein

gene rNA polymerase

Allelic exclusion?

Bloodstream of mammalian host

Slender (proliferative)

VSG Pol I Yes

Stumpy (non-dividing)

VSG Not applicable

Not applicable

Midgut and salivary gland of insect vector (tsetse)

Procyclic (proliferative)

GPEET, EP procyclins

Pol I No

Epimastigote (proliferative)

BARP Unknown Unknown

Metacyclic (non-dividing)

VSG Pol I Yes

* T. brucei is extracellular in both the bloodstream of its mammalian host and in its transmission vector, the tsetse (Glossina species). Its life cycle alternates between proliferating stages and non-proliferating stages that precede the switch between host and vector. The parasite surface in each stage consists of characteristic glycosylphosphatidylinositol-anchored proteins, the genes of which are mainly transcribed by RNA polymerase I (Pol I). Although transcription is strongly reduced in stumpy forms106, it is unclear whether the variant surface glycoprotein (VSG) detected at the surface originates from weak de novo transcription or if it remains from the previous life cycle stage. For simplicity, the developmental stages in the proventriculus of the insect vector are not included. BARP, alanine-rich protein.

R E V I E W S

508 | july 2009 | VoluME 7 www.nature.com/reviews/micro

© 2009 Macmillan Publishers Limited. All rights reserved.

Page 6: Epigenetic regulation in African trypanosomes: a new kid on the block

Antigenic variationThe process by which an infectious organism alters its surface to evade the host immune response; common in several pathogens, such as Trypanosoma brucei, the malaria parasite Plasmodium falciparum and Giardia lamblia.

Monoallelic expressionThe expression of a single allele from a gene family.

system by periodically switching the make­up of its sur­face coat proteins. Although there are hundreds of VSG genes in the genome, only one is transcribed at any time from a specialized subtelomeric locus called a blood­stream expression site (BES)74. This monoallelic expression occurs in a specialized non­nucleolar structure called the expression site body75,76 (BOX 2). The other BESs are excluded from this compartment, which may be neces­sary to keep them silent. Allelic exclusion is also used for regulating the expression of VAR genes in the malaria parasite and for regulating the odorant receptor (OR) gene family in olfactory sensory neurons77,78. Therefore, understanding the mechanism of monoallelic expres­sion would be a fundamental achievement not only in the trypanosome field, but also for research into other infectious diseases and neurobiology.

Transcription of the VSG gene family needs to be tightly controlled, not only to achieve monoallelic expression, but also to ensure developmental regula­tion during the life cycle of the parasite (TABLE 1). So far, only one Pol I trans cription complex has been identified, and it is essential for both rRNA and VSG transcription79. VSG transcriptional control is inherited from one generation to the next, and VSG switching

does not necessarily involve DNA rearrangements, suggesting that it is regulated epigenetically. As in higher eukaryotes, histone PTMs are thought to carry the epigenetic information necessary for VSG regula­tion. Genome database searches in T. brucei have led to the identification of candidate epigenetic factors that might have a role in the regulation of Pol I transcription. Some of them belong to the chromatin­remodelling or histone­modifying enzyme families (TABLE 2).

ISWI is an essential gene that encodes the only ISWI family member of chromatin remodellers found in T. brucei 16. The T. brucei ISWI has an N­terminal ATPase domain (although it is not known whether this domain is important for the function of ISWI) and a SlIDE domain at the C terminus, which is thought to be involved in the interaction with DNA and histone tails80. ISWI has been shown to have a repressive role in other systems, and its RNAi­mediated depletion in T. brucei leads to a derepression of the promoter­proximal region of a silent BES. Derepression ranges from 30­fold to 60­fold in the bloodstream form and from 10­fold to 17­fold in the procyclic form, sug­gesting that BES silencing requires ISWI in both stages of the life cycle.

Table 2 | Chromatin-remodelling enzymes, histone-modifying and histone-binding proteins*

Class Name Number of putative homologues in humans‡

Chromatin-remodelling families

ISWI ISWI15 Pfam, 2

RAD54 relative JBP2 (REF. 14) and a RAD54 homologue

Pfam, 0; SMART, 0

Other SNF2 domain-containing homologues

Ten candidate genes Pfam, 91

Histone-modifying enzymes

Lysine acetyltransferase (KAT) HAT1–3 (MYST family)20,21 Pfam, 14 of MYST family; (5 characterized)

Elp3a–b (1 characterized)

Lysine deacetylase (HDAC) HDAC1–4 (REF. 19) Pfam, 47; (11 characterized)

SIR2rp1–3 (REFS 22,23) Pfam, 11; (7 characterized)

Lysine methyltransferase (KMT) DOT1A–B45,83 Pfam, 1

20–27 SET domain-containing proteins (KMTs)

Pfam, 99; SMART, 62

Lysine demethylases (KDM) KDM1–4 (JmjC domain) Pfam, 53; SMART, 38

Arginine methyltransferases PRMT1 (REF.27), PRMT5 (REF.28) and PRMT7 (REF.28) and two other candidate genes

(9 highly related members)

Histone-binding proteins

Bromodomain BDF1–4 Pfam, 132; SMART, 82

PHD finger PHD1–5 Pfam, 214; SMART, 132

Tudor TDR1 Pfam, 35; SMART, 28

*List of candidate chromatin-remodelling enzymes, histone-modifying and histone-binding proteins found in the Trypanosoma brucei and human genomes or predicted by Pfam or SMART databases. ‡The number of characterized human homologues is indicated in brackets. DOT1, disruptor of telomeric silencing protein 1; Elp3, elongator protein 3; HDAC, histone deacetylase; JBP2, J base binding protein 2; JmjC, Jumonji C; ISWI, imitation SWI (switching deficient); MYST, human MOZ (monocytic leukaemia zinc finger protein), yeast Ybf2 (renamed Sas3; something about silencing 3), yeast Sas2 and mammalian TIP60 (HIV Tat-interacting protein 60 kDa); PHD, plant homeo domain; PRMT, protein arginine methyltransferase; RAD54, radiation sensitive 54; SET, su(var)3–9, enhancer of zeste, trithorax; SIR2rp1–3, silencing information regulator 2-related proteins (also known as sirtuins) 1–3; SNF2, sucrose non-fermenting 2.

R E V I E W S

NATuRE REVIEWS | MiCrobiology VoluME 7 | july 2009 | 509

© 2009 Macmillan Publishers Limited. All rights reserved.

Page 7: Epigenetic regulation in African trypanosomes: a new kid on the block

Nature Reviews | Microbiology

Human H2A

T. brucei H2A 1

1

4

4

4 9 23 27 36

3219 23

**

**

*

5

* *

K115

K119

K120

K122

K125

K128

134

129

125

112

136

132

102

99

120

K120

41

***

12 16

Human H2B

T. brucei H2B

Human H3

T. bruceiH3

Human H4

T. bruceiH4

α1 α2 α3

α1 α2 α3

α1 α2

α2

α3

α1

α1

α1

α2

α2

α3

α3

α3

α1 α2 α3

α1 α2 α3

K79

K76

5 12 16 20

181714105421

Acetylation

Methylation

Phosphorylation

Ubiquitylation

Triton acid urea gel electrophoresisA polyacrylamide gel-based electrophoresis technique that allows the efficient separation of core histones as a result of their association with Triton X-100.

Among the ten histone­modifying enzymes that have been characterized so far in T. brucei (HAT1–3, HDAC1–4, SIR2rp1, DoT1A and DoT1B), only a subset has been tested for a potential role in Pol I regu­lation in the bloodstream form. HAT1, SIR2rp1 and DoT1B are all involved in either telomeric silencing and/or VSG trans cription. The depletion of HAT1 by RNAi results in the derepression of a telomeric reporter promoter, but does not affect transcription from a BES promoter20. SIR2rp1 is a histone deacetylase that is involved in maintaining subtelomeric regions that are silent in S. cerevisiae81 and in the malaria para­site Plasmodium falciparum82. In SIR2rp1 mutants of T. brucei, although a Pol I reporter construct becomes derepressed in both the bloodstream and pro cyclic stages of the life cycle, VSG genes are not derepressed. Moreover, the frequency of transcriptional switch­ing between two BESs is not altered, suggesting that antigenic variation is not dramatically affected by the absence of SIR2rp1 (REF. 22).

The only histone­modifying enzyme that is clearly involved in VSG transcriptional regulation and BES switching is DoT1B, which is responsible for tri­methylation of H3K76 (REF. 83). Deletion of this gene results in a tenfold derepression of various silent VSG genes. DoT1B is also necessary for rapid transcrip­tional switching between two BESs. In a DoT1B null mutant, switching between two BESs occurred more slowly than in wild­type cells, and stable intermedi­ates expressing two VSGs simultaneously at the sur­face could be detected. In these intermediates, the two BESs are transcribed from the promoter through the subtelomeric VSG, but only one BES is transcribed at the normal level, suggesting that other epigenetic fac­tors are responsible for ensuring that only one BES is maximally transcribed. Despite the clear reliance on

DoT1B, the precise mechanistic role of H3K76me3 in VSG regulation remains unclear.

The proximity of BES to telomeres suggests that telomeric chromatin is involved in VSG regulation. However, the first two telomere­binding proteins described in T. brucei (H3 variant30 and TRF84) did not reveal a role in VSG transcription. By contrast, deple­tion of RAP1, a TRF­interacting protein, resulted in an approximately 100­fold derepression of silent VSGs85. Moreover, genes located close to the telomeres were found to be more derepressed than genes located further upstream, suggesting that telomere structure is essential for silencing. Even though mechanistic details are lack­ing, it is clear that telomeric proteins play an important part in the regulation of surface protein expression. Future studies should identify other factors, which will help us to understand how this regulation is achieved.

Cell differentiationDifferentiation of mammalian cells is accompanied by substantial alterations of chromatin structure (reviewed in REF. 86). Is there any evidence that chromatin struc­ture is involved in regulating gene expression during the trypanosome life cycle? Studies during the early 1990s that compared histones from two T. brucei life cycle stages using Triton acid urea gel electrophoresis revealed differences in the number and pattern of bands of the core histones and linker histone H1, suggesting stage­specific PTM differences87,88. Studies of chromatin con­densation in vitro showed that the level of condensation was higher in the bloodstream than in the procyclic form87. These observations were confirmed by electron microscopy, which revealed that the electron­dense (heterochromatic) regions in the nucleus of the blood­stream form are large and mostly associated with the envelope, whereas nuclei in the procyclic form contain a

Figure 3 | Sequence alignment and histone modifications of Trypanosoma brucei and human core histones. Important regions of homology are shaded by grey boxes. Residues that might be modified are marked with circles, colour-coded as indicated. Modifications that were found only in the bloodstream form are indicated by asterisks. The three helices of the histone core are shown as coloured boxes. The post-translational modification (PTM) inventory of human histones is not complete. Only abundant or well-characterized PTMs are shown.

R E V I E W S

510 | july 2009 | VoluME 7 www.nature.com/reviews/micro

© 2009 Macmillan Publishers Limited. All rights reserved.

Page 8: Epigenetic regulation in African trypanosomes: a new kid on the block

Nature Reviews | Microbiology

N

NH2

ON

O

O

O

NH

O

ON

O

O

O

O

OHO

HO OH

HO

5-methylcytosine Base J

KaryokinesisThe process that partitions of the nucleus into the daughter cells during cell division.

higher number of small heterochromatin areas that are scattered throughout the nucleus89.

Is there any evidence that histone PTMs are responsible for these stage­specific differences in chromatin structure, and do they have a role in differentiation? There are few differences in the inventory of histone PTMs between the bloodstream and procyclic forms of T. brucei 10,11 and, although abundant PTMs are present in similar amounts, there are few interesting differences. For exam­ple, H4K18me3 seems to be enriched almost fourfold in the procyclic form. As H4K18me3 may be the homologue of the mammalian heterochromatin marker H4K20me3, it might be interesting to explore a possible role for H4K18 methylation in stage­dependent heterochromatin forma­tion in trypanosomes. other PTMs that were found in the bloodstream form but not in the procyclic form were acetylation of H2A (K4 and K122), H2B (K4, K12 and K16), H3 (K23) and H4 (K2, K5 and K14) and methyla­tion of H3K32 and H4K2. However, these variations might be attributable to technical differences between the stud­ies of the bloodstream and procyclic forms. For example, different protocols needed to be used to isolate and purify histones from different life cycle stages and different mass

spectrometry techniques were applied to analyse PTMs.DoT1B is the only histone­modifying enzyme that

has been studied in the context of differentiation from the bloodstream form to the procyclic form in vitro45. When wild­type cells from the bloodstream form are exposed to differentiation signals in vitro, they differenti­ate to the procyclic form and grow normally. By contrast, DoT1B­deficient cells died several days after attempt­ing differentiation. Cell cycle profiles of ∆DoT1B cells, determined by flow cytometry, were indistinguishable from those of proliferating wild­type cells, suggesting that growth was not arrested at a specific phase of the cell cycle. A cell cycle phenotype during differentiation is of special interest because only G1­arrested trypano­somes can enter the differentiation process90. Therefore, there seems to be an unknown connection between cell cycle control and commitment to differentiation. For reasons of tractability, however, these experiments were performed in a T. brucei strain, for which the differentia­tion kinetics differ from the synchronous transition that occurs in less laboratory­adapted strains. Experiments to unravel the role of chromatin structure during devel­opmental differentiation of T. brucei are still in the early stages, and it remains to be seen what can be learnt from differentiation processes in other eukaryotic cells.

Cell cycle controlCell cycle control in trypanosomes is characterized by several unusual features. Mitosis is closed, meaning that the nuclear envelope persists during all stages of the cell cycle, and the chromosomes do not condense91. Although many orthologues of conserved cell cycle reg­ulators are identifiable in the T. brucei genome, some key enzymes appear to be missing and trypanosomes seem to use different checkpoints from those of other model eukaryotes (reviewed in REF. 92). What is known about the role of PTMs in cell cycle control? Genetic manipu­lations of several histone­modifying enzymes result in cell cycle defects in trypanosomes. For example, HDAC4 is required for normal cell cycle progression19. In both bloodstream and procyclic forms, HDAC4­depleted cells have a mild growth defect that is caused by a delay in the G2­ to M­phase transition. By contrast, cell cycle analysis of cells with depleted HAT1 and HAT2 showed an increase in the number of post­mitotic cells20. It is not yet known how different acetylation levels influence cell cycle progression. The most striking effects on cell cycle regulation were obtained by depleting DoT1A and DoT1B45. Dimethylated H3K76 can be detected only during mitosis and karyokinesis, whereas trimethylated H3K76 is present throughout the cell cycle. Deletion of DoT1B leads to cell cycle­independent dimethyla­tion of H3K76, suggesting that the mitosis­specific reg­ulation of H3K76me2 is mediated by the presence or absence of DoT1B. Depletion of DoT1A by RNAi gen­erates a population of cells that contain half the normal (diploid) amount of DNA, suggesting that H3K76me2 has an important role in the regulation of cell cycle pro­gression. The exact mechanism by which PTMs regulate cell cycle progression in trypanosomes remains to be determined. Better understanding of the regulation of

Figure 4 | Structure of the DNA modifications in Trypanosoma brucei. The genomes of both bloodstream and procyclic forms of T. brucei contain 5-methylcytosine.At least 1 in every 10,000 cytosines is methylated. Base J, (β-d-glucopyranosyloxymethyluracil) is found only in the bloodstream form, but its absence is not lethal. Two proteins (JBP1 and JBP2) have been found that bound to base J in the context of DNA, and either catalysed or modulated the synthesis of base J.

Box 2 | Monoallelic expression: a gene regulation dilemma

Some multigene families need to be tightly regulated because only one allele must be expressed at a time. The best-studied examples are the VSG gene family in Trypanosoma brucei98, the VAR gene family in the human malaria parasite Plasmodium falciparum99 and the odorant receptor (OR) gene family in the mammalian olfactory sensory neurons100. To achieve monoallelic expression, cells must successfully perform several steps: choose which member is going to be activated, count the number of actively transcribed genes and, perhaps, retain some form of cellular memory to remember which members were recently activated. We still know very little about the molecular machineries that mediate these processes, although recent data raised testable working hypotheses: one model suggests that the active allele locates to a unique subcompartment in the nucleus75,101; a second model proposes that chromatin plays a part in maintaining the epigenetic state of each member of the family16,83,102; a third model proposes that cis-regulatory elements within members of the gene family may determine which allele is activated103; and a fourth model suggests a role for cis-linked promoter activity in maintaining all except one allele silent104.

R E V I E W S

NATuRE REVIEWS | MiCrobiology VoluME 7 | july 2009 | 511

© 2009 Macmillan Publishers Limited. All rights reserved.

Page 9: Epigenetic regulation in African trypanosomes: a new kid on the block

histone­modifying enzymes and identification of PTM­binding proteins should help to unravel fundamental biological processes.

ConclusionIn the past few years, efforts to find and characterize histone PTMs and the corresponding chromatin­ modifying enzymes have been initiated in trypanosomes. It is already apparent that trypanosomes have a smaller enzymatic repertoire than higher eukaryotes and, presum­ably, a less complicated histone code. Considering that trypanosomes diverged several hundred million years ago from the main eukaryotic lineage, better understanding of T. brucei epigenetic regulation could help to answer three basic questions in the chromatin field: what does a simple histone code look like, what are the minimum essential functions of chromatin and what drives histone­modifying

enzymes to acquire new chromatin­related functions? Answering these questions will help us to understand the evolution of the histone code and its functions. Because Pol II transcription in trypanosomes seems to be consti­tutive, there are probably few PTMs that are dedicated to Pol II silencing. T. brucei is therefore a potentially use­ful model organism in which to study the PTMs that regulate only Pol I transcription and other evolutionarily conserved chromatin­mediated processes. Future studies have to focus on the dynamic aspects of PTMs and chro­matin structure in trypanosomes. For example, how is the deposition and removal of these marks and the activity of histone­modifying enzymes regulated? Furthermore, PTM­binding proteins must be identified to unravel how different histone modifications mediate important bio­logical processes such as transcriptional regulation and cell cycle control in this fascinating organism.

1. Kouzarides, T. Chromatin modifications and their function. Cell 128, 693–705 (2007).

2. Strahl, B. D. & Allis, C. D. The language of covalent histone modifications. Nature 403, 41–45 (2000).

3. World Health Organization. The world health report 2004 — changing history (WHO, Geneva, 2004).

4. Engstler, M. & Boshart, M. Cold shock and regulation of surface protein trafficking convey sensitization to inducers of stage differentiation in Trypanosoma brucei. Genes Dev. 18, 2798–2811 (2004).

5. Urwyler, S., Studer, E., Renggli, C. K. & Roditi, I. A family of stage-specific alanine-rich proteins on the surface of epimastigote forms of Trypanosoma brucei. Mol. Microbiol. 63, 218–228 (2007).

6. Welstead, G. G., Schorderet, P. & Boyer, L. A. The reprogramming language of pluripotency. Curr. Opin. Genet. Dev. 18, 123–129 (2008).

7. Rothenberg, E. V., Moore, J. E. & Yui, M. A. Launching the T-cell-lineage developmental programme. Nature Rev. Immunol. 8, 9–21 (2008).

8. Palenchar, J. B. & Bellofatto, V. Gene transcription in trypanosomes. Mol. Biochem. Parasitol. 146, 135–141 (2006).

9. Clayton, C. E. Life without transcriptional control? From fly to man and back again. EMBO J. 21, 1881–1888 (2002).

10. Janzen, C. J. et al. Unusual histone modifications in Trypanosoma brucei. FEBS Lett. 580, 2306–2310 (2006).This first comprehensive study of the histone PTMs in T. brucei showed that this parasite has a simplified histone code and some unusual modifications.

11. Mandava, V. et al. Histone modifications in Trypanosoma brucei. Mol. Biochem. Parasitol. 156, 41–50 (2007).

12. Saha, A., Wittmeyer, J. & Cairns, B. R. Chromatin remodelling: the industrial revolution of DNA around histones. Nature Rev. Mol. Cell Biol. 7, 437–447 (2006).

13. Martens, J. A. & Winston, F. Recent advances in understanding chromatin remodeling by Swi/Snf complexes. Curr. Opin. Genet. Dev. 13, 136–142 (2003).

14. Deuring, R. et al. The ISWI chromatin-remodeling protein is required for gene expression and the maintenance of higher order chromatin structure in vivo. Mol. Cell 5, 355–365 (2000).

15. de la Serna, I. L., Ohkawa, Y. & Imbalzano, A. N. Chromatin remodelling in mammalian differentiation: lessons from ATP-dependent remodellers. Nature Rev. Genet. 7, 461–473 (2006).

16. Hughes, K. et al. A novel ISWI is involved in VSG expression site downregulation in African trypanosomes. EMBO J. 26, 2400–2410 (2007).Although chromatin-remodelling activity was not definitively shown, T. brucei ISWI was the first candidate remodeller shown to be necessary to maintain fully silenced VSGs in two stages of the T. brucei life cycle.

17. Dipaolo, C., Kieft, R., Cross, M. & Sabatini, R. Regulation of trypanosome DNA glycosylation by a SWI2/SNF2-like protein. Mol. Cell 17, 441–451 (2005).

18. Allis, C. D. et al. New nomenclature for chromatin-modifying enzymes. Cell 131, 633–636 (2007).

19. Ingram, A. K. & Horn, D. Histone deacetylases in Trypanosoma brucei: two are essential and another is required for normal cell cycle progression. Mol. Microbiol. 45, 89–97 (2002).This pioneering study of histone-modifying enzymes in T. brucei showed that, despite substantial evolutionary divergence, four histone deacetylases in T. brucei have conserved features with those of higher eukaryotes.

20. Kawahara, T. et al. Two essential MYST-family proteins display distinct roles in histone H4K10 acetylation and telomeric silencing in trypanosomes. Mol. Microbiol. 69, 1054–1068 (2008).

21. Siegel, T. N. et al. Acetylation of histone H4K4 is cell cycle regulated and mediated by HAT3 in Trypanosoma brucei. Mol. Microbiol. 67, 762–771 (2008).

22. Alsford, S., Kawahara, T., Isamah, C. & Horn, D. A sirtuin in the African trypanosome is involved in both DNA repair and telomeric gene silencing but is not required for antigenic variation. Mol. Microbiol. 63, 724–736 (2007).

23. Garcia-Salcedo, J. A., Gijon, P., Nolan, D. P., Tebabi, P. & Pays, E. A chromosomal SIR2 homologue with both histone NAD-dependent ADP-ribosyltransferase and deacetylase activities is involved in DNA repair in Trypanosoma brucei. EMBO J. 22, 5851–5862 (2003).

24. Cloos, P. A., Christensen, J., Agger, K. & Helin, K. Erasing the methyl mark: histone demethylases at the center of cellular differentiation and disease. Genes Dev. 22, 1115–1140 (2008).

25. Bedford, M. T. & Clarke, S. G. Protein arginine methylation in mammals: who, what, and why. Mol. Cell 33, 1–13 (2009).

26. Wysocka, J., Allis, C. D. & Coonrod, S. Histone arginine methylation and its dynamic regulation. Front. Biosci. 11, 344–355 (2006).

27. Pelletier, M., Pasternack, D. A. & Read, L. K. In vitro and in vivo analysis of the major type I protein arginine methyltransferase from Trypanosoma brucei. Mol. Biochem. Parasitol. 144, 206–217 (2005).

28. Pasternack, D. A., Sayegh, J., Clarke, S. & Read, L. K. Evolutionarily divergent type II protein arginine methyltransferase in Trypanosoma brucei. Eukaryot. Cell 6, 1665–1681 (2007).

29. Fisk, J. C. et al. A type III protein arginine methyltransferase from the protozoan parasite, Trypanosoma brucei. J. Biol. Chem. 284, 11590–11600 (2009).

30. Lowell, J. E. & Cross, G. A. A variant histone H3 is enriched at telomeres in Trypanosoma brucei. J. Cell Sci. 117, 5937–5947 (2004).

31. Lowell, J. E., Kaiser, F., Janzen, C. J. & Cross, G. A. Histone H2AZ dimerizes with a novel variant H2B and is enriched at repetitive DNA in Trypanosoma brucei. J. Cell Sci. 118, 5721–5730 (2005).This was the first time that the histone variant H2AZ was shown to associate with a novel variant H2B.

32. Alsford, S. & Horn, D. Trypanosomatid histones. Mol. Microbiol. 53, 365–372 (2004).

33. Sullivan, W. J. Jr, Naguleswaran, A. & Angel, S. O. Histones and histone modifications in protozoan parasites. Cell. Microbiol. 8, 1850–1861 (2006).

34. Thatcher, T. H. & Gorovsky, M. A. Phylogenetic analysis of the core histones H2A, H2B, H3, and H4. Nucleic Acids Res. 22, 174–179 (1994).

35. da Cunha, J. P., Nakayasu, E. S., de Almeida, I. C. & Schenkman, S. Post-translational modifications of Trypanosoma cruzi histone H4. Mol. Biochem. Parasitol. 150, 268–277 (2006).

36. Latham, J. A. & Dent, S. Y. Cross-regulation of histone modifications. Nature Struct. Mol. Biol. 14, 1017–1024 (2007).

37. Briggs, S. D. et al. Gene silencing: trans-histone regulatory pathway in chromatin. Nature 418, 498 (2002).

38. Sun, Z. W. & Allis, C. D. Ubiquitination of histone H2B regulates H3 methylation and gene silencing in yeast. Nature 418, 104–108 (2002).

39. Mandava, V., Janzen, C. J. & Cross, G. A. Trypanosome H2Bv replaces H2B in nucleosomes enriched for H3 K4 and K76 trimethylation. Biochem. Biophys. Res. Commun. 368, 846–851 (2008).

40. Hyland, E. M. et al. Insights into the role of histone H3 and histone H4 core modifiable residues in Saccharomyces cerevisiae. Mol. Cell. Biol. 25, 10060–10070 (2005).

41. Shilatifard, A. Chromatin modifications by methylation and ubiquitination: implications in the regulation of gene expression. Annu. Rev. Biochem. 75, 243–269 (2006).

42. Steger, D. J. et al. DOT1L/KMT4 recruitment and H3K79 methylation are ubiquitously coupled with gene transcription in mammalian cells. Mol. Cell. Biol. 28, 2825–2839 (2008).

43. San-Segundo, P. A. & Roeder, G. S. Role for the silencing protein Dot1 in meiotic checkpoint control. Mol. Biol. Cell 11, 3601–3615 (2000).

44. Huyen, Y. et al. Methylated lysine 79 of histone H3 targets 53BP1 to DNA double-strand breaks. Nature 432, 406–411 (2004).

45. Janzen, C. J., Hake, S. B., Lowell, J. E. & Cross, G. A. M. Selective di- or trimethylation of histone H3 lysine 76 by two DOT1 homologs is important for cell cycle regulation in Trypanosoma brucei. Mol. Cell 23, 497–507 (2006).This paper is the first study of the enzymatic activity of KMTs in trypanosomes and also describes the importance of the DOT1 homologues in cell cycle control.

46. Grant, P. A. & Berger, S. L. Histone acetyltransferase complexes. Semin. Cell Dev. Biol. 10, 169–177 (1999).

47. Tachibana, M. et al. G9a histone methyltransferase plays a dominant role in euchromatic histone H3 lysine 9 methylation and is essential for early embryogenesis. Genes Dev. 16, 1779–1791 (2002).

48. Brownell, J. E. & Allis, C. D. Special HATs for special occasions: linking histone acetylation to chromatin assembly and gene activation. Curr. Opin. Genet. Dev. 6, 176–184 (1996).

R E V I E W S

512 | july 2009 | VoluME 7 www.nature.com/reviews/micro

© 2009 Macmillan Publishers Limited. All rights reserved.

Page 10: Epigenetic regulation in African trypanosomes: a new kid on the block

49. Sterner, D. E. & Berger, S. L. Acetylation of histones and transcription-related factors. Microbiol. Mol. Biol. Rev. 64, 435–459 (2000).

50. Siegel, T. N. et al. Four histone variants mark the boundaries of polycistronic transcription units in Trypanosoma brucei. Genes Dev. 23, 1063–1076 (2009).

51. Schotta, G. et al. A silencing pathway to induce H3-K9 and H4-K20 trimethylation at constitutive heterochromatin. Genes Dev. 18, 1251–1262 (2004).

52. Schotta, G. et al. A chromatin-wide transition to H4K20 monomethylation impairs genome integrity and programmed DNA rearrangements in the mouse. Genes Dev. 22, 2048–2061 (2008).

53. Donati, G. et al. An NF-Y-dependent switch of positive and negative histone methyl marks on CCAAT promoters. PLoS ONE 3, e2066 (2008).

54. Ruthenburg, A. J., Li, H., Patel, D. J. & Allis, C. D. Multivalent engagement of chromatin modifications by linked binding modules. Nature Rev. Mol. Cell Biol. 8, 983–994 (2007).

55. Taverna, S. D., Li, H., Ruthenburg, A. J., Allis, C. D. & Patel, D. J. How chromatin-binding modules interpret histone modifications: lessons from professional pocket pickers. Nature Struct. Mol. Biol. 14, 1025–1040 (2007).

56. Colot, V. & Rossignol, J. L. Eukaryotic DNA methylation as an evolutionary device. Bioessays 21, 402–411 (1999).

57. Robertson, K. D. DNA methylation and human disease. Nature Rev. Genet. 6, 597–610 (2005).

58. Rojas, M. V. & Galanti, N. DNA methylation in Trypanosoma cruzi. FEBS Lett. 263, 113–116 (1990).

59. Militello, K. T. et al. African trypanosomes contain 5-methylcytosine in nuclear DNA. Eukaryot. Cell 7, 2012–2016 (2008).

60. Gommers-Ampt, J. H. et al. β-d-glucosyl-hydroxymethyluracil: a novel modified base present in the DNA of the parasitic protozoan T. brucei. Cell 75, 1129–1136 (1993).This paper describes the chemical structure of base J, a DNA modification that has not been found in other organisms.

61. Borst, P. & Sabatini, R. Base J: discovery, biosynthesis, and possible functions. Annu. Rev. Microbiol. 62, 235–251 (2008).

62. Kieft, R. et al. JBP2, a SWI2/SNF2-like protein, regulates de novo telomeric DNA glycosylation in bloodstream form Trypanosoma brucei. Mol. Biochem. Parasitol. 156, 24–31 (2007).

63. Yu, Z. et al. The protein that binds to DNA base J in trypanosomatids has features of a thymidine hydroxylase. Nucleic Acids Res. 35, 2107–2115 (2007).

64. Cliffe, L. J. et al. JBP1 and JBP2 are two distinct thymidine hydroxylases involved in J biosynthesis in genomic DNA of African trypanosomes. Nucleic Acids Res. 37, 1452–1462 (2009).

65. Genest, P. A. et al. Formation of linear inverted repeat amplicons following targeting of an essential gene in Leishmania. Nucleic Acids Res. 33, 1699–1709 (2005).

66. McStay, B. & Grummt, I. The Epigenetics of rRNA genes: from molecular to chromosome biology. Annu. Rev. Cell Dev. Biol. 24, 131–157 (2008).

67. Berriman, M. et al. The genome of the African trypanosome Trypanosoma brucei. Science 309, 416–422 (2005).

68. Alsford, S., Kawahara, T., Glover, L. & Horn, D. Tagging a T. brucei RRNA locus improves stable transfection efficiency and circumvents inducible expression position effects. Mol. Biochem. Parasitol. 144, 142–148 (2005).

69. Waters, A. P., Syin, C. & McCutchan, T. F. Developmental regulation of stage-specific ribosome populations in Plasmodium. Nature 342, 438–440 (1989).

70. Waters, A. P. et al. Species-specific regulation and switching of transcription between stage-specific ribosomal RNA genes in Plasmodium berghei. J. Biol. Chem. 272, 3583–3589 (1997).

71. McCracken, S. et al. The C-terminal domain of RNA polymerase II couples mRNA processing to transcription. Nature 385, 357–361 (1997).

72. Kooter, J. M. & Borst, P. a-amanitin-insensitive transcription of variant surface glycoprotein genes provides further evidence for discontinuous transcription in trypanosomes. Nucleic Acids Res. 12, 9457–9472 (1984).

73. Rudenko, G., Bishop, D., Gottesdiener, K. & Van der Ploeg, L. H. a-amanitin resistant transcription of protein coding genes in insect and bloodstream form Trypanosoma brucei. EMBO J. 8, 4259–4263 (1989).

74. Hertz-Fowler, C. et al. Telomeric expression sites are highly conserved in Trypanosoma brucei. PLoS ONE 3, e3527 (2008).

75. Navarro, M. & Gull, K. A pol I transcriptional body associated with VSG mono-allelic expression in Trypanosoma brucei. Nature 414, 759–763 (2001).The observation that the actively transcribed VSG localizes in a discrete non-nucleolar structure that contains RNA polymerase I and is resistant to DNase I treatment allowed the emergence of a new model in which allelic exclusion was achieved by limiting the location of VSG genes in the nuclear space.

76. Landeira, D. & Navarro, M. Nuclear repositioning of the VSG promoter during developmental silencing in Trypanosoma brucei. J. Cell Biol. 176, 133–139 (2007).

77. Scherf, A. et al. Antigenic variation in malaria: in situ switching, relaxed and mutually exclusive transcription of var genes during intra-erythrocytic development in Plasmodium falciparum. EMBO J. 17, 5418–5426 (1998).

78. Chess, A., Simon, I., Cedar, H. & Axel, R. Allelic inactivation regulates olfactory receptor gene expression. Cell 78, 823–834 (1994).

79. Brandenburg, J. et al. Multifunctional class I transcription in Trypanosoma brucei depends on a novel protein complex. EMBO J. 26, 4856–4866 (2007).

80. Grune, T. et al. Crystal structure and functional analysis of a nucleosome recognition module of the remodeling factor ISWI. Mol. Cell 12, 449–460 (2003).

81. Sandell, L. L., Gottschling, D. E. & Zakian, V. A. Transcription of a yeast telomere alleviates telomere position effect without affecting chromosome stability. Proc. Natl Acad. Sci. USA 91, 12061–12065 (1994).

82. Freitas-Junior, L. H. et al. Telomeric heterochromatin propagation and histone acetylation control mutually exclusive expression of antigenic variation genes in malaria parasites. Cell 121, 25–36 (2005).

83. Figueiredo, L. M., Janzen, C. J. & Cross, G. A. A histone methyltransferase modulates antigenic variation in African trypanosomes. PLoS Biol 6, e161 (2008).This paper showed that DOT1B is necessary for rapid VSG switching, demonstrating for the first time the importance of a histone-modifying enzyme for antigenic variation in African trypanosomes.

84. Li, B., Espinal, A. & Cross, G. A. M. Trypanosome telomeres are protected by a homologue of mammalian TRF2. Mol. Cell. Biol. 25, 5011–5021 (2005).

85. Yang, X., Figueiredo, L. M., Espinal, A., Okubo, E. & Li, B. RAP1 is essential for silencing telomeric Variant Surface Glycoprotein genes in Trypanosoma brucei. Cell 137, 99–109 (2009).RAP1 is the first telomeric protein that, when disrupted, shows a phenotype clearly related to VSG gene regulation, confirming the importance of telomeres during antigenic variation.

86. Margueron, R., Trojer, P. & Reinberg, D. The key to development: interpreting the histone code? Curr. Opin.t Genet. Dev. 15, 163–176 (2005).

87. Schlimme, W., Burri, M., Bender, K., Betschart, B. & Hecker, H. Trypanosoma brucei brucei: differences in the nuclear chromatin of bloodstream forms and procyclic culture forms. Parasitology 107, 237–247 (1993).Hecker et al. present some groundbreaking findings on chromatin structure in trypanosomes, including the biochemical properties of chromatin in two stages of the T. brucei life cycle.

88. Burri, M., Schlimme, W., Betschart, B. & Hecker, H. Characterization of the histones of Trypanosoma brucei brucei bloodstream forms. Acta Trop. 58, 291–305 (1994).

89. Rout, M. P. & Field, M. C. Isolation and characterization of subnuclear compartments from Trypanosoma brucei. Identification of a major repetitive nuclear lamina component. J. Biol. Chem. 276, 38261–38271 (2001).

90. Ziegelbauer, K., Quinten, M., Schwarz, H., Pearson, T. W. & Overath, P. Synchronous differentiation of Trypanosoma brucei from bloodstream to procyclic forms in vitro. Eur. J. Biochem. 192, 373–378 (1990).

91. Ogbadoyi, E., Ersfeld, K., Robinson, D., Sherwin, T. & Gull, K. Architecture of the Trypanosoma brucei nucleus during interphase and mitosis. Chromosoma 108, 501–513 (2000).

92. Hammarton, T. C. Cell cycle regulation in Trypanosoma brucei. Mol. Biochem. Parasitol. 153, 1–8 (2007).

93. Hirumi, H. & Hirumi, K. Continuous cultivation of Trypanosoma brucei blood stream forms in a medium containing a low concentration of serum protein without feeder cell layers. J. Parasitol. 75, 985–989 (1989).

94. Carruthers, V. B. & Cross, G. A. High-efficiency clonal growth of bloodstream- and insect-form Trypanosoma brucei on agarose plates. Proc. Natl Acad. Sci. USA 89, 8818–8821 (1992).

95. LaCount, D. J., Bruse, S., Hill, K. L. & Donelson, J. E. Double-stranded RNA interference in Trypanosoma brucei using head-to-head promoters. Mol. Biochem. Parasitol. 111, 67–76 (2000).

96. Wang, Z., Morris, J. C., Drew, M. E. & Englund, P. T. Inhibition of Trypanosoma brucei gene expression by RNA interference using an integratable vector with opposing T7 promoters. J. Biol. Chem. 275, 40174–40179 (2000).

97. Scahill, M. D., Pastar, I. & Cross, G. A. CRE recombinase-based positive-negative selection systems for genetic manipulation in Trypanosoma brucei. Mol. Biochem. Parasitol. 157, 73–82 (2008).

98. Pays, E. Regulation of antigen gene expression in Trypanosoma brucei. Trends Parasitol. 21, 517–520 (2005).

99. Scherf, A., Lopez-Rubio, J. J. & Riviere, L. Antigenic variation in Plasmodium falciparum. Annu. Rev. Microbiol. 62, 445–470 (2008).

100. Buck, L. & Axel, R. A novel multigene family may encode odorant receptors: a molecular basis for odor recognition. Cell 65, 175–187 (1991).

101. Voss, T. S. et al. A var gene promoter controls allelic exclusion of virulence genes in Plasmodium falciparum malaria. Nature 439, 1004–1008 (2006).

102. Lopez-Rubio, J. J. et al. 5′ flanking region of var genes nucleate histone modification patterns linked to phenotypic inheritance of virulence traits in malaria parasites. Mol. Microbiol. 66, 1296–1305 (2007).

103. Nguyen, M. Q., Zhou, Z., Marks, C. A., Ryba, N. J. & Belluscio, L. Prominent roles for odorant receptor coding sequences in allelic exclusion. Cell 131, 1009–1017 (2007).

104. Frank, M. et al. Strict pairing of var promoters and introns is required for var gene silencing in the malaria parasite Plasmodium falciparum. J. Biol. Chem. 281, 9942–9952 (2006).

105. Klose, R. J. & Zhang, Y. Regulation of histone methylation by demethylimination and demethylation. Nature Rev. Mol. Cell Biol. 8, 307–318 (2007).

106. Amiguet-Vercher, A. et al. Loss of the mono-allelic control of the VSG expression sites during the development of Trypanosoma brucei in the bloodstream. Mol. Microbiol. 51, 1577–1588 (2004).

107. Dean, S., Marchetti, R., Kirk, K. & Matthews, K. R. A surface transporter family conveys the trypanosome differentiation signal. Nature 459, 213–217 (2009).

AcknowledgementsThe authors thank S. Hake and N. Siegel for their critical read-ing of the manuscript; G. Rudenko and R. Sabatini for helpful comments; and P. Bastin and B. Rotureau for providing micro-graphs of the different stages of the T. brucei life cycle, which were used, in part, to draw the cartoons in Table 1.

DATABASESEntrez Genome Project: http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=genomeprjCaenorhabditis elegans | Saccharomyces cerevisiae | Trypanosoma bruceiUniProtKB: http://www.uniprot.orgELP3 | GCN5 | H2BK123ub | HAT1 | HAT2 | HBO1 | JBP1 | PRMT1 | PRMT5 | PRMT7 | RAD54 | SUV420H1 | SUV420H2 | TIP60 | TRF

FURTHER INFORMATIONGeorge A. M. Cross’s homepage: http://tryps.rockefeller.eduPfam: http://pfam.sanger.ac.ukPlasmoDB: http://plasmodb.org/plasmoSmart: http://smart.embl-heidelberg.deTriTrypDB: http://tritrypdb.org/tritrypdb

All liNkS Are ACTive iN The oNliNe pDf

R E V I E W S

NATuRE REVIEWS | MiCrobiology VoluME 7 | july 2009 | 513

© 2009 Macmillan Publishers Limited. All rights reserved.