Sunteți pe pagina 1din 34

Nature 506, 316321 (20 February 2014)

Go J. Yoshida M.D.,Ph.D. (E-mail:goscm@tmd.ac.jp)


Dept. of Stem Cell Biology, Medical Research Institute
Tokyo Medical and Dental University, Tokyo, Japan
Senescence regulation in stem cells
Senescence is a state in which a cell no longer has
the ability to proliferate.
Senescence accompanies changes in nuclear
morphology and formation of a distinct chromatin
structure, called senescence-associated
heterochromatic foci (SAHF).
Cell. !!"#$$"%&!"-&$'.
p16
Ink4a
promotes SAHF formation
(pigenetic regulation of the p16
INK4a
locus in
senescent cells
)n senescent cells, polycomb proteins are lost or displaced from the INK4A locus by a
number of established mechanisms, including (!) (*H downregulation resulting in the
loss of H"+& methylation, (e) the histone demethylase ,-,.", which acti/ely remo/es
the H"+&me" mar0 and (f) the S1)2S3F chromatin remodelling comple4, which is
proposed to displace polycombs from the INK4A locus. 1e also propose a no/el
mechanism (") by which polycomb proteins may be displaced by transcriptional
acti/ators.
5ncogene. !$$#"!%6!$-$$.
7he p8b pathway and SAHF formation in senescent cells
#$ere is a in%erse! re&ation
bet'een p16(N)4a an! p*+b
p8b protein functions to
recruit repressi/e en9ymatic
acti/ities to (F-regulated
target gene promoters.
7he remar0ably stable state of
cellular senescence is thought
to be maintained by
compacted heterochromatic
or :closed; chromatin, called
SAHF, which accumulates
around genes encoding cell
cycle and proliferation
proteins.
5ncogene. !$$#"!%6!$-$$.
Aging Cell. !!#$%$"-6.
7he arrows indicate satellite cells stained
positi/ely for 3-CA- (pero4idase, brown)
whereas the arrowheads gi/e e4amples of
myonuclei (blue).
Cold Spring Harb.!$$#&'%$!$<$$$.
Age-associated decline in muscle regeneration
Aged muscle (right, = months of
age) shows a significant deficit in
the number of regenerating fibers
and an increase in necrotic fibers
compared with young muscle.
>re/ention of age-related increase in fibrosis during
muscle regeneration by heterochronic parabiosis
established for $month
muscle tissue damage (free9e in?uries)
" days later, @rdA was administered.
days later, muscle tissue was fi4ed.
Science. !!&#"$&%B!&-$!.
'month =month
7he effects of the aged en/ironment on myogenic
progenitor cell fate and muscle regeneration are
mediated by the 1nt signaling pathway.
Science. !!&#"$&%B!&-$!.
Satellite Cells
S0eletal -uscle Stem Cells
to e4ist close pro4imity to s0eletal muscle fibers
to e4hibit regenerati/e acti/ity of s0eletal muscle
to be reCuired for early post-natal muscle growth
and for repair of catastrophic muscle in?ury
SC mar0ers% >a4&, -yfD, -yo., Ca/$, 3-CA-etc.
>hysiol 8e/. !$"#6"%"-'&.
Stem Cells )nt. !$"#!$"%=!$'=.
8egulatory switch between
Cuiescent and acti/ated states
of satellite cells
3otch and 1nt signaling antagoni9e
each other to define the state of
satellite cells.
Stem Cells )nt. !$"#!$"%=!$'=. 3otch-predominant 1nt-predominant
Functional dysregulation of stem cells during
aging% a focus on s0eletal muscle stem cells
F(@S ,. !$"#B!%=!D$<=!'.
.e/ @iol. !!'#6=%D!-''.
% sarcopenia
Eoss of muscle mass% sarcopenia
Eoss of self-renewal% senescence
"ero,on%ersion
Bmi1 plays a crucial role in the maintenance of the
stem cell pool in postnatal skeletal muscle and is
essential for efficient muscle regeneration after injury,
possibly through its actions on p16
ink4a
and p19
arf
Regenerating fibers express fetal myosin
heavy chain (MyHC) initially and then switch
to adult isoforms while they mature.
>EoS 5ne. !$$#'($$)%e&$$'.
@mi-$-2- -(F underwent premature aging.
3ature. $666#"6&%$'=-B.
SA-./ pre*mature a"in" mo!e&
sarcopenia in senescence-accelerated mice (SA->B)
(4p Ferontol. !!D#=!%D'-&.
type ( fiber type ((A fiber
SA->B strains e4hibit
accelerated muscle aging
as compared with SA-8$.
senescence-prone inbred
strains (SA->)
senescence-resistant inbred
strains (SA-8)
7ype-) fiber si9e was not
different between SA-
strains.
-a0or Hypot$esis
7he geriatric satellite stem cells show the irre/ersible
switch of Cuiescence-to-senescence ("ero,on%ersion) and
lose self-renewal potential /ia >8C$2p$'
)3+=a
28b2(F a4is.
young% <" months of age
adult% D-' months of age
old% !<= months of age
geriatric% B-" months of age
young% <" months of age
adult% D-' months of age
old% !<= months of age
geriatric% B-" months of age
A reduced number of satellite cells in aged mice is not
responsible for the significant regenerati/e decline.
(n0e,tion of ,ar!ioto1in (2#3
1310
*5
-) to tibia&is anterior
mus,&e
After $days
e-HC (embryonic myosin hea/y chain) is a differentiation mar0er of s0eletal muscle.
>a4&-positi/e satellite
cells in resting phase
Satellite-cell-intrinsic potential of regeneration $wee0
after transplantation into young recipient mice
(Cual numbers of sorted satellite cells labelled with
FF> were transplanted into muscles of young mice.
>a4&G Cuiescent satellite cells from geriatric mice
showed a reduced acti/ation rate early after in?ury
compared to adult2old cells.
.a145-yo6 !oub&e*positi%e sate&&ite ,e&&s
are !efine! as a,ti%ate! sate&&ite ,e&&s
Huiescent geriatric satellite cells e4hibited a
defecti/e acti/ation capacity =hr after
transplantation into a young host
7he only significantly up-regulated senescence
gene in cluster F$ was the master regulator of
senescence p16
INK4a
(Cdkn2a)
group of genes with increased
e4pression in geriatric satellite cells
HAub mar0 in the INK4a locus was significantly
reduced in Cuiescent satellite cells from geriatric mice
>olycomb repressi/e comple4 $
(>8C$)% (" ubiCuitin ligase comple4
that is specific for histone HA.
.+21 ,omp&e possess H2A*)117
ubi8uitin 93 &i"ase a,ti%ities
2anoni,a& .+21
,omp&e1
3ature !!=#="$%B&"-B&B.
-ol Cell. !!D#!%B=D-D=.
HAub% histone HA mono-ubiCuitination at
lysine $$6 (.+21 repressi%e mar:er)
8. domain is the main binding site for @mi$ of
p$'
)3+=a
locus.
p16
INK4a
e4pression changed the satellite cell acti/ation
ectopic p16
INK4a
o/ere4pression in
Cuiescent young satellite cells
pre/ented their acti/ation
p16
INK4a
silencing rescued the permanent
cell-cycle arrest and allowed geriatric
satellite cell acti/ation
p16
INK4a
silencing in geriatric satellite cells
before engraftment into young mice
p16
INK4a
silencing in geriatric satellite cells
reco/ered the self-renewal potential
Self-renewing satellite cells# >a4&(G)2 +i-'&(-)
p$'
)3+=a
o/ere4pression reduced the
capacity of young :reser/e; satellite cells
to reacti/ate.
Acti/ation capacity of satellite cells were
reduced after successi/e myogenesis rounds
(repeated in?uries).
Feriatric satellite cells underwent an accelerated
entry into senescence under proliferati/e pressure
7ransplanted geriatric satellite cells also displayed
signs of deep senescence at times of ma4imal
proliferati/e e4pansion after in?ury
Feriatric satellite cells e4posed to proliferati/e
conditions presented high p$'
)3+=a
e4pression le/els
correlating with reduced phosphorylated 8b protein
p16
INK4a
interference reduced gerocon/ersion in
geriatric satellite cells
Ex vivo culture of muscle satellite cells
, Stem Cell 8es 7her !$#S$$%!!".
adult% "! years of age
old% 'D years of age
geriatric% 6' years of age
Ex vivo ana&ysis
Acti/e p$'
)3+=a
28b a4is in human geriatric stem cells
Conclusion
Feriatric age induces intrinsic alterations in
muscle stem-cell regenerati/e functions, which
cannot be reco/ered by a young host en/ironment.
p$'
)3+=a
e4pression is specifically induced in
geriatric satellite cells and dri/es gerocon/ersion
at the e4pense of proliferation.
p$'
)3+=a
seems to be positi/ely associated with
reduced myogenic potential and increased cellular
senescence in human satellite cells from geriatric
indi/iduals with sarcopenia.

S-ar putea să vă placă și