1
MOUSE MODEL OF WERNER SYNDROME Table 1. Summary of phenotypes observed in the Wrn +/+ Terc -/- and Wrn -/- Terc -/- mouse models HUMAN WS MOUSE MODELS Wrn -/- Terc -/- Long telomeres Wrn +/+ Terc -/- Control* Wrn -/- Terc -/- Short telomeres Osteoporosis Cataracts Type II diabetes Skin defects Hypogonadism Atherosclerosis Genome instability Mesenchymal tumours No No No No No No No + No No No ++ ++ No +++ + ++++ ++++ ++++ ++++ ++++ No ++++ ++++ *Telomerase null animals display a subset of aging phenotypes but do not display many of the symptoms associated with human aging. 1. Telomere biology differences 2. Telomerase activity 3. Redundant function of murine WRN 4. Inter-individual differences 10-15kb 40-80kb somatic cell telomerase telomerase somatic cell WRN Other RecQ helicases WRN Other RecQ helicases WS fibroblast Normal fibroblast Chromosomal fusions and non-reciprocal chromosomal translocations mTerc deletion Mutation rate Premature loss of proliferative capacity Wrn -/- NO aging phenotype Premature aging phenotype Wrn -/- Terc -/- DNA damage response Premature replicative senescence Sister telomere loss (STL) Dysfunctional telomere Normal telomere Lagging Leading 5’ 5’ 5’ 5’ 5’ 5’ 5’ 5’ 3’ 3’ 3’ 3’ 3’ 3’ 3’ 3’ 3’ FISH analysis of WS cells. Telomeres were hybridized with a TRITC-[TTAGGG] 4 probe and are shown in the red channel. Arrows indicate missing telomeric staining from single sister chromatids. WS patient Slow growth rates Premature senescence Genome instability Telomere dysfunction S G 1 G 2 M telomeres STALLED REPLICATION FORK G G WRN -/- Lagging Lagging Leading Leading 5’ 5’ 3’ TELOMERE DYSFUNCTION CONTRIBUTES TO WS PATHOLOGY WRN TRF1/2 and POT1 POT1 D-loop T-loop WRN Uncapping/ recapping POT1 ROS ROS TRF1 TRF2 Repair of oxidative damage WRN TRF1 TRF2 POT1 TRF2 TRF1 NHEJ WRN Ku and DNA-PKcs Telomere T loop D loop Shelterin Sh DNA replication Apoptosis DNA repair Transcription Recombination Telomere ends WRN Ku DNA- PKcs DSB End recognition Processing Ligation WRN INTERACTS WITH RESIDENT TELOMERIC PROTEINS Werner Syndrome (WS) is a rare genetic disorder that mimics the characteristics of normal human aging. Since aging is likely caused by numerous genetic and environmental factors acting simultaneously, it is very difficult to dissect the roles of individual genes in this process. In order to simplify such complexity, the goal of this review is focused on the relationship between three key components: aging, telomeres and the enzyme telomerase, taking Werner syndrome as a model of human aging. WERNER SYNDROME AS A MODEL OF AGING: THE ROLE OF WRN AT TELOMERES AND THE IMPACT OF TELOMERASE Anna Esteve Garcia. Facultat de Biociències. Curs acadèmic 2013-2014. Universitat Autònoma de Barcelona (Catalunya, Espanya) WRN TRF1 TRF2 TRF1 TRF2 TRF1 TRF2 TRF1 TRF2 TRF1 TRF2 TRF1 TRF2 TRF1 TRF2 TRF1 TRF2 TRF1 TRF2 Both shortened telomeres and a lack of telomerase are required, at least in mice, for WS disease manifestation. FUTURE APPROACHES Increased telomerase expression WS aging associated phenotypes INCREASED HEALTHSPAN WS fibroblasts WS fibroblasts DIFFERENT STUDIES EXEMPLIFY THIS INTERDEPENDENT FUNCTION BETWEEN WRN AND TELOMERASE: STL rescued Sister telomere signal G strand C strand Expression of exogenous TERT or WRN WS patient DNA induced lesions reversion 4NQO WS fibroblasts Sensitivity reversed Colony forming capacity High sensitivity Colony forming capacity loss Premature aging protection Restoration of telomere function in WS iPSCs WS fibroblasts WS iPSCs WS fibroblasts WS fibroblasts Helicase-deficient WRN WRN WS phenotypes WRN preserves the genome integrity of WS fibroblasts, protecting telomeres erosion BOTH WRN AND TELOMERASE RESCUE DEFICIENCIES IN WS FIBROBLASTS Extension of telomeres TERT mTERT TELOMERE DYSFUNCTION GENOME INSTABILITY ? CHROMOSOME FUSIONS STL replication fusion STL replication replication breakage breakage fusion Fusion with telomeric DNA at fusion site Fusion without telomeric DNA at fusion site Telomere loss (STL) Unprotected telomere ends Controls cells WS cells BOTH CONTROL AND WS CELLS WITH CHROMOSOMAL ABERRATIONS DNA at fusion s DRUG INTERVENTION IN PREVENTING ACCELERATED AGING IN WS Telomere dysfunction & chromosomal aberration Decreased cellular lifespan of WS fibroblasts SENESCENCE Activation of p38 WRN gene mutation Oxidative stress ACCELERATED PREMATURE AGING PREVENTION Because of their G-rich nature, telomeres are fraught with potential obstacles to DNA replication that block replication forks. WRN is capable of unwinding such structures, preventing collapse in telomere forks. In its absence, telomeric signals are only detected at a single chromatid, phenotype termed sister telomere loss (STL). WRN participates in several important DNA metabolic pathways. These include DNA repair, replication, recombination and telomere maintenance. WRN plays a role at telomeres as it interactions with other resident telomeric proteins. Elevated stress response correlates with the activation of the stress-associated mitogen-activated protein kinase (MAPK) p38. Activation of p38 stabilizes p21 and promotes entry into cellular senescence. SB203580 is a drug that targets p38 activity. This drug treatment extends the replicative lifespan of WS fibroblasts in vitro. Detected by FISH ? ? ? WERNER SYNDROME AS A MODEL OF HUMAN AGING Main features Molecular level Bilateral cataracts Greying hair and hair loss Short stature Scleroderma-like skin Atherosclerosis Ischemic heart disease Osteoporosis Type II diabetes mellitus Hypogonadism Cancer predisposition (sarcomas) Bloom Syndrome Cockayne Syndrome Xeroderma Pigmentosum Rothmund-Thomson Syndromes Hutchinson-Guilford Progeria Syndrome WERNER SYNDROME Progeroid aging syndromes cr.8 WRN Human RecQ helicase family Exonuclease and helicase activities Multiple functions Normal human aging New potent and selective inhibitors WRN WRN loss induces the wide range of premature aging phenotypes seen in Werner Syndrome. The key event, caused by WRN mutations, is the dysfunction of telomeres. Thus, the cascade started by telomere dysfunction explains the majority of the WS pathological phenotypes. Telomerase phenotypes recruitment as well as the WS mouse model generation reinforce this hypothesis. The SB203580 drug treatment may lead to novel therapies involving MAPK.

Werner Syndrome as a model of aging - UAB Barcelona · 2014. 11. 25. · Werner Syndrome (WS) is a rare genetic disorder that mimics the characteristics of normal human aging. Since

  • Upload
    others

  • View
    4

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Werner Syndrome as a model of aging - UAB Barcelona · 2014. 11. 25. · Werner Syndrome (WS) is a rare genetic disorder that mimics the characteristics of normal human aging. Since

MOUSE MODEL OF

WERNER SYNDROME

Table 1. Summary of phenotypes observed in the Wrn+/+

Terc-/- and Wrn-/- Terc-/- mouse models

HUMAN WS

MOUSE MODELS

Wrn-/- Terc-/-

Long

telomeres

Wrn+/+ Terc-/-

Control*

Wrn-/- Terc-/-

Short

telomeres

Osteoporosis

Cataracts

Type II diabetes

Skin defects

Hypogonadism

Atherosclerosis

Genome instability

Mesenchymal tumours

No

No

No

No

No

No

No

+

No

No

No

++

++

No

+++

+

++++

++++

++++

++++

++++

No

++++

++++

*Telomerase null animals display a subset of aging phenotypes but do

not display many of the symptoms associated with human aging.

1. Telomere biology differences

2. Telomerase activity

3. Redundant function of murine WRN

4. Inter-individual differences

10-15kb 40-80kb

ACCCAC 3’ 5’

somatic cell

telomerase

ACCCAC 3’ 5’

ACCCAC3’ 5’

telomerase

somatic

cell

WRN

Other RecQ

helicases

WRN

Other RecQ helicases

WS

fibroblast

Normal

fibroblast

Chromosomal

fusions and

non-reciprocal

chromosomal

translocations

mTerc

deletion

� Mutation rate

Premature loss

of proliferative

capacity

Wrn-/-

NO aging phenotype

Premature aging phenotype

Wrn-/- Terc-/-

DNA damage

response

Premature

replicative

senescence

Sister

telomere

loss (STL) Dysfunctional telomere

Normal telomere

Lagging

Leading

5’

5’

5’ 5’

5’

5’

5’

5’

3’

3’

3’

3’

3’

3’ 3’

3’ 3’

FISH analysis of WS cells.

Telomeres were hybridized with a

TRITC-[TTAGGG]4 probe and are

shown in the red channel. Arrows

indicate missing telomeric staining

from single sister chromatids.

WS patient

Slow growth rates

Premature senescence

Genome instability

Telomere dysfunction

S

G1

G2

M telomeres

STALLED

REPLICATION FORK

G

G

WRN-/-

Lagging

Lagging

Leading

Leading

5’

5’

3’

TELOMERE DYSFUNCTION CONTRIBUTES TO WS PATHOLOGY

WRN – TRF1/2 and POT1

POT1

D-loop

T-loop

WRN

Uncapping/

recapping

POT1

ROS ROS

TR

F1

TR

F2

Repair of

oxidative

damage

WRN

TR

F1

TR

F2

POT1

TR

F2

TR

F1

loopNHEJ

WRN – Ku and DNA-PKcs

Telomere

T loop

D loop

Shelterin Shelte

DNA

replication

Apoptosis

DNA repair

Transcription

Recombination

Telomere ends

WRN

Ku

DNA-

PKcs

DSB

End

recognition

Processing

Ligation

WRN INTERACTS WITH RESIDENT TELOMERIC PROTEINS

Werner Syndrome (WS) is a rare genetic disorder that mimics

the characteristics of normal human aging. Since aging is likely

caused by numerous genetic and environmental factors acting

simultaneously, it is very difficult to dissect the roles of individual

genes in this process. In order to simplify such complexity, the

goal of this review is focused on the relationship between three

key components: aging, telomeres and the enzyme telomerase, taking Werner syndrome as a model of human aging.

WERNER SYNDROME AS A MODEL OF AGING:

THE ROLE OF WRN AT TELOMERES AND THE IMPACT OF TELOMERASE

Anna Esteve Garcia. Facultat de Biociències. Curs acadèmic 2013-2014. Universitat Autònoma de Barcelona (Catalunya, Espanya)

WRN

TR

F1

TR

F2

TR

F1

TR

F2

TR

F1

TR

F2

TR

F1

TR

F2

TR

F1

TR

F2

T

RF

1

TR

F2

TR

F1

TR

F2

TR

F1

TR

F2

TR

F1

TR

F2

Both shortened telomeres

and a lack of telomerase

are required, at least in

mice, for WS disease

manifestation.

FUTURE APPROACHES

Increased

telomerase

expression

WS aging

associated

phenotypes

INCREASED

HEALTHSPAN

WS fibroblasts

WS fibroblasts

DIFFERENT STUDIES EXEMPLIFY THIS INTERDEPENDENT FUNCTION BETWEEN WRN AND TELOMERASE:

STL rescued Sister telomere

signal

G strand C strand

Expression of

exogenous

TERT or WRN

WS

patient DNA induced lesions reversion

4NQO

WS fibroblasts

Sensitivity reversed

Colony forming

capacity

High sensitivity

Colony forming

capacity loss

Premature aging protection

Restoration

of telomere

function in

WS iPSCs WS

fibroblasts

WS

iPSCs

WS fibroblasts WS fibroblasts

Helicase-deficient

WRN WRN

WS

phenotypes

WRN preserves the

genome integrity of WS

fibroblasts, protecting

telomeres erosion

BOTH WRN AND TELOMERASE RESCUE DEFICIENCIES IN WS FIBROBLASTS

Extension of

telomeres

TERT

mTERT

TELOMERE

DYSFUNCTION

GENOME

INSTABILITY

? CHROMOSOME

FUSIONS

ST

L

replic

ation

fusio

n

ST

L

replic

atio

n

replic

ation

bre

akag

e

bre

akag

e

fusio

n

Fusion with telomeric

DNA at fusion site

Fusion without telomeric

DNA at fusion site

Telomere loss

(STL)

Unprotected

telomere ends

Controls cells WS cells

BOTH CONTROL

AND WS CELLS

WITH

CHROMOSOMAL

ABERRATIONS

DNA at fusion si

DRUG INTERVENTION IN PREVENTING ACCELERATED

AGING IN WS

Telomere dysfunction &

chromosomal aberration

Decreased cellular

lifespan of WS

fibroblasts

SENESCENCE

Activation

of p38

WRN gene

mutation

Oxidative

stress

ACCELERATED

PREMATURE

AGING

PREVENTION

Because of their G-rich nature,

telomeres are fraught with

potential obstacles to DNA

replication that block replication

forks. WRN is capable of

unwinding such structures,

preventing collapse in telomere

forks. In its absence, telomeric

signals are only detected at a

single chromatid, phenotype

termed sister telomere loss

(STL).

WRN participates in several important DNA metabolic pathways. These include

DNA repair, replication, recombination and telomere maintenance. WRN plays a

role at telomeres as it interactions with other resident telomeric proteins.

Elevated stress response

correlates with the activation

of the stress-associated

mitogen-activated protein

kinase (MAPK) p38.

Activation of p38 stabilizes

p21 and promotes entry into

cellular senescence.

SB203580 is a drug that

targets p38 activity. This drug

treatment extends the

replicative lifespan of WS

fibroblasts in vitro.

Detected

by FISH

? ?

?

WERNER SYNDROME AS A MODEL OF HUMAN AGING Main features Molecular level Bilateral cataracts

Greying hair and hair loss

Short stature

Scleroderma-like skin

Atherosclerosis

Ischemic heart disease

Osteoporosis

Type II diabetes mellitus

Hypogonadism

Cancer predisposition

(sarcomas)

Bloom Syndrome

Cockayne Syndrome

Xeroderma Pigmentosum

Rothmund-Thomson Syndromes

Hutchinson-Guilford Progeria Syndrome

WERNER

SYNDROME

geri Synd

Progeroid aging

syndromes

cr.8

WRN

Human RecQ helicase family

Exonuclease and helicase activities

Multiple

functions

Normal human

aging

New potent and

selective inhibitors

WRN

WRN loss induces the wide range of premature aging phenotypes seen in Werner Syndrome.

The key event, caused by WRN mutations, is the dysfunction of telomeres. Thus, the cascade started by

telomere dysfunction explains the majority of the WS pathological phenotypes.

Telomerase phenotypes recruitment as well as the WS mouse model generation reinforce this hypothesis.

The SB203580 drug treatment may lead to novel therapies involving MAPK.