干细胞之家 - 中国干细胞行业门户第一站

 

 

搜索
朗日生物

免疫细胞治疗专区

欢迎关注干细胞微信公众号

  
查看: 320464|回复: 245
go

Impaired Hematopoietic Stem Cell Functioning After Serial Transplantation and Du [复制链接]

Rank: 7Rank: 7Rank: 7

积分
威望
0  
包包
483  
楼主
发表于 2009-3-5 10:52 |只看该作者 |倒序浏览 |打印
ABSTRACT1 q5 n4 s+ M1 z

& M: J, m% W, XAdult somatic stem cells possess extensive self-renewal capacity, as their primary role is to replenish aged and functionally impaired tissues. We have previously shown that the stem cell pool in short-lived DBA/2 (D2) mice is reduced during aging, in contrast to long-lived C57BL/6 (B6) mice. This suggests the existence of a genetically determined mitotic clock operating in stem cells, which possibly limits organismal aging. In the study reported here, unfractionated bone marrow (BM) cells or highly purified Lin–Sca-1 c-kit  (LSK) cells were serially transplanted in lethally irradiated D2 and B6 mice. In both strains, serial transplantation resulted in a substantial loss of stem cell activity. However, as we estimate that in B6 mice, the maximum number of population doublings of primitive cells is approximately 30, in D2 mice this is only approximately 20, resulting in a 1,000–fold difference in expansion potential, irrespective of whether whole bone marrow or purified hematopoietic stem cells (HSCs) were transplanted. Interestingly, recipients reconstituted with serially transplanted BM cells were able to accept a freshly isolated graft without any further conditioning. Finally, we show that whereas transplantation of BM cells into healthy, nonconditioned, young B6 recipients does not lead to engraftment, young BM cells do engraft and provide multilineage reconstitution in nonirradiated aged mice. Our data clearly establish the relevance of an intrinsic, genetically controlled program associated with impaired stem cell functioning during aging.& i" h3 q1 ~' u+ ^) p

2 h& ]- n7 m4 i6 q# T( SIINTRODUCTION. R+ H( M5 \; y( C+ P1 e
. V% v6 z5 M+ j; _% f1 L( _2 Z
Hematopoieticstemcells (HSCs) sustain lifelong production of mature blood cells. In fact, it has been documented that HSCs can even outlive their original donor upon repeated serial transplantation in lethally irradiated recipients [1], the most widely used model to study the process of HSC exhaustion. However, multiple studies have shown that serial transplantation is limited, suggesting stem cell exhaustion [1–10]. It has been documented that serial transplantation results in a permanent loss of self-renewal capacity, which is cell dose dependent [10–12]. Also, functional decline increases with repeated serial transfers [2, 3, 6, 8, 13, 14]. In a competitive repopulation assay, serially transplanted stem cells showed impaired self-renewal, after the first transplantation [8, 15]. HSCs showed limited self-renewal when either purified HSCs [15] or unfractionated bone marrow (BM) cells were used [8, 9]. Most of these studies have suggested that the limit to serial transplantation is caused by exhaustion of stem cells, but this finding has been challenged by more recent experiments in which it was argued that the engraftment defect results from an increasingly smaller number of stem cells transplanted (i.e., in vivo dilution of stem cells) [10]. In addition, it has been proposed that artifacts of the serial transplantation procedure, such as residual injury caused by removal of the HSCs from their natural environment [8, 9], might cause the decline in self-renewal capacity. This hypothesis was challenged by experiments in which animals, except for one hind limb, were repeatedly irradiated. Due to repeated stress caused by repopulation of irradiated marrow spaces, self-renewal capacity of the shielded bone marrow remained depressed and did not recover with time [16]. However, it is possible that, in conjunction with repeated proliferative stress in these shielding studies, forced cell migration and potential loss of stem cell niches contributed to decreased stem cell functioning.
8 ?- @7 D: C: h/ M- Z% ?9 g2 v& G- f9 J, [; A  Z6 g' K
In the study reported here, we assessed whether an intrinsic, genetic program exists that leads to stem cell exhaustion in serial transplantation models. To this end, we estimated the maximal number of cobblestone area–forming cell (CAFC) d35 population doublings in two genetically distinct mouse strains, C57BL/6 (B6) and DBA/2 (D2) mice, which differ with respect to numerous HSC traits [17–20]. During normal aging, the number of stem cells and their repopulating ability after transplant decreased in D2 mice, whereas these parameters increased in B6 mice [17, 19, 21–27]. Intriguingly, D2 mice have a shorter life span than B6 animals have [28]. The presence of a mitotic clock operating in proliferating cells and potentially limiting the lifespan of cells in culture has been a topic of much debate ever since it was first postulated by Hayflick [29, 30]. This applies, in particular, to the relevance of such a putative clock in limiting organismal lifespan. Since adult somatic stem cells have a key role in replenishing tissues, it is interesting to speculate that conservation of stem cell functioning during normal aging extends lifespan.
6 M& |5 z- b% S5 J5 N% B
4 r. Q8 R% m/ n* j% |) Y5 s3 BIn this study we used two distinct transplantation models, unfractionated bone marrow cells or highly purified Lin–Sca-1 c-kit  (LSK) cells, to estimate the maximal number of CAFC d35 population doublings following serial transplantation. Using unfractionated BM, the number of transplanted stem cells can only be assessed retrospectively, increasing the likelihood that HSC dose varies from one transplant to the other, potentially resulting in stem cell dilution. This problem is partly controlled by using highly purified stem cells, although it has been shown (and was confirmed in the current study) that functional activity per stem cell, based on phenotype (Lin–Sca-1 c-kit ) declines in serial transplantations. Finally, we tested whether BM cells isolated from a young donor were able to engraft in recipients that were previously reconstituted with serially transplanted BM cells or to engraft in nonconditioned older recipients.
1 ?$ Y; p' {% T7 X/ `: v" Y# `1 @. z/ }. H6 ], E
MATERIALS AND METHODS$ W7 g  u4 |2 h, }& |$ P* L9 g
9 w' C% |7 T# I7 U6 ~& l3 V
Animal* j9 P( w+ i# m+ r3 z

& y% N* i! M; l& SSerial transplantations were carried out using C57BL/60laHsD (B6) and DBA/20laHsD (D2) female recipients and male donors. Animals were purchased from Harlan (Horst, The Netherlands; http://www.harlan.com) and were used at an age of 6–8 weeks. The initial transplantations were carried out using male donors and female recipients. Although a mild immune response potentially can be elicited against the H-Y antigen [31], this is not observed in severely conditioned recipients (we used 9.5 Gy total body irradiation). Enhanced green fluorescent protein (eGFP) transgenic C57BL/6 mice [32, 33] were originally purchased from the Jackson Laboratory (Bar Harbor, ME; http://www.jax.org), and were further bred under specific pathogen–free conditions in the Central Animal Facility of the University of Groningen.- }+ M2 N3 f& }% ~0 R
3 O0 X6 C4 p/ o
Serial Transplantations
( N7 E+ T  x; N+ X. Y1 w! ^
  s3 B  N& Y4 x9 oBone marrow cells were isolated from both femora. Single-cell suspensions were obtained by flushing each femur three times, and the total number of nucleated cells was assessed using a Coulter Counter (Coulter Electronics, Dunstable, England). Prior to purification of HSCs, a standard NH4Cl erythrocyte lysis was performed. Subsequently, cells were incubated with 5% normal rat serum for 15 minutes at 4~C, after which cells were stained with biotinylated lineage-specific antibodies (anti-B220, anti-Gr-1, anti-Mac-1, anti-TER-119, and anti-CD3e), fluorescein isothiocyanate (FITC)–anti-Sca-1, and Allophycocyanin (APC)–anti-c-kit (all antibodies from Pharmingen, San Diego, CA; http://www.bdbiosciences.com) for 40 minutes at 4~C. Cells were subsequently incubated with Streptavidin-PE (Pharmingen) for 40 minutes at 4~C and strained through a 35-μm cell strainer (Becton, Dickinson, Bedford, MA; http://www.bd.com), after which cells were purified using a MoFlo flow cytometer (Dako Cytomation, Fort Collins, CO; http://www.dakocytomation.com). The clearly distinct Sca-1 c-kit  population among the 5% of the BM cells showing the least phycoerythrin (PE) fluorescence intensity was selected. Sorting gates were determined based on normal BM cells and were not changed when serially transplanted samples were analyzed. Recipients were lethally irradiated (9.5 Gy) using an IBL 637 Cesium-137  source (CIS Biointernational, Gifsur-Yvette, France, http://www.cisbiointernational.fr/) 24 hours prior to transplantation of either 4 x 106 unfractionated BM cells or 1,500 sorted Lin–Sca-1 c-kit  (LSK) cells. Two independent sets of experiments were performed with LSK cells. At 4–6 months after each transplantation, recipients were sacrificed, bone marrow was collected, and the entire procedure was repeated with transplantations in a new group of lethally irradiated recipients. Both transplantation models were initiated with 6–12 recipients. Due to lack of cells and loss of survival of recipients, in selected groups, fewer recipients were used in later transplantations.
) D( C4 g, y* H" H, b0 T9 Q& a1 Z: w# I* @1 q7 j) M. B6 [5 ~9 s
CAFC Assay
& _; f- _) ~0 Y. c5 x
0 u; G) e1 B- E0 P8 QIn vitro limiting dilution-type long-term BM cultures were used to assess clonogenic activity in unfractionated and purified cell fractions. To this end, the CAFC assay was performed, as described previously [17, 34].% \+ u0 N  [+ s  x  @
  G' G% O: x- o8 T5 i+ Q
Where as committed progenitors form colonies 7–14 days after culture initiation, the most primitive hematopoietic cell subsets appear late (i.e., after 35 days) in culture [35].
( c. `" s$ h% k' u: \
! X% _. t) ~2 M' a9 v. wDetermination of Donor Cell Contribution after Serial Transplant! S+ @- B/ v2 s9 x

  _# E% w$ }$ L" rTo verify donor-derived hematopoiesis, the presence of the Y-chromosome in individual progenitor cell colonies was assessed at various time points. To this end, methylcellulose colony-forming unit granulocyte-macrophage (CFU-GM) cultures were initiated. After 7 days of culture, DNA was isolated from individual colonies, as described previously [19]. To assess the presence of the Y-chromosome, a 20-μ1 polymerase chain reaction (PCR) (10 minutes at 95~C; 50 cycles of 30 seconds at 94~C, 30 seconds at 60~C, and 30 seconds at 72~C; 10 minutes at 72~C) was performed using a Perk-inElmer GeneAmp PCR system 9700 (PerkinElmer Corp., Norwalk, CT; http://www.perkinelmer.com) and Y-chromosome–specific primers (Isogen Bioscience B.V., Maarssen, The Netherlands; http://www.isogen-lifescience.com); YMT2/B1 5'-CTG GAG CTC TAC AGT GAT GA-3' and YMT2/B2 5'-CAG TTA CCA ATC AAC ACA TCA C-3', amplifying a 342-bp fragment. PCR products were analyzed using a 1% ethidium bromide-stained agarose gel.  \/ E, G+ j/ S2 e; Y5 U# h% m

7 s" }  J! H  P. O' qCompetitive Repopulating Assay
8 Y; O3 ?7 g9 x; _" q  e4 a+ q# p3 d. Q
- J& |2 o8 N0 ?; A, m6 e, ETo determine in vivo reconstituting potential, peripheral white blood cell numbers were counted using a Coulter Counter (Coulter Electronics), and, after serial transplantation was performed, hematocrits were obtained.' c, o9 u( v8 J. V8 ?. T5 ~: H# n+ t

6 ]7 l5 [( ~6 V+ k% k/ u' `5 dCompetitive repopulating assays were carried out in two distinct settings. First, 4 x 106 B6 BM cells that had been serially transplanted five times were mixed with an equal number of BM cells that were freshly isolated from young eGFP  transgenic B6 donors [32, 33], and both cell populations were co-transplanted in lethally irradiated B6 recipients. At various timepoints after transplantation, the levels of eGFP  leukocytes in the peripheral blood was quantified using fluorescence-activated cell sorter (FACS) analysis (FACS Calibur; Becton, Dickinson, Palo Alto, CA; http://www.bd.com). Second, to address the issue whether the engraftment defect of serially transplanted BM was due to a homing defect, lethally irradiated B6 recipients were reconstituted with 5x serially transplanted BM cells alone, and after 3 months the freshly isolated BM cells from young eGFP  B6 donors were infused, without additional conditioning.
7 k6 }0 n) B  {" z' a8 a
# d7 F: L) Q- v( a; t; j! @Bone Marrow Transplantations in Nonirradiated Young and Aged Recipients
# Q: J( F4 d; e+ D# U0 A& u8 L6 e0 S6 W2 }$ |7 x: D' O/ h
Four to five million unfractionated BM cells isolated from 6- to 8-week-old female eGFP  donors were infused intravenously in nonirradiated, normal, healthy, young (16 months old) wild-type B6 recipients. Transplantations were repeated every 2 months. Prior to each transplant, the presence of eGFP  leukocytes in the peripheral blood was assessed by flowcytometry.! y4 z% J% f: u9 K; T
! `/ Y1 t( i/ R) C0 @# q' o
Survival of old, transplanted recipients was compared with a cohort of aged-matched non-transplanted animals, housed under the same conditions.  Z- m) e) R& }; }

4 i9 a# q+ E- _+ X8 w* lQuantification of Expansion and the Number of Stem Cell Population Doublings
/ O2 ~4 f- ]7 T2 ^5 G  m* f' \
# z3 q: ?- I* \, CThe total number of CAFC d35 present in the whole animal at the moment of sacrifice was calculated from the number of CAFC d35 present in one femur, assuming that one femur represents 6% of the total marrow [36, 37]. The fold expansion of the CAFC d35 was calculated by assessing the total number of CAFC d35 present per animal and dividing this number by the number of infused CAFC d35 some 4–6 months earlier. The actual number of CAFC d35 population doublings was calculated by the algorithm: (2n = fold expansion), in which n = the number of population doublings [38].
$ P) y7 p8 U, J  T- @6 W! c
  L, N- S& Y& z; H7 K( @) qStatistical Analysis
4 M" Q, ^8 ], l% ?: L% D; `  R
9 X3 }/ y& u6 V  i$ ~During serial transplantation, BM cells of recipients were pooled and CAFC cultures were initiated. The entire set of serial transplantations using purified HSCs was repeated twice. Values of the two independent experiments of LSK transplantations were averaged, and the SEM was calculated. Kaplan-Meier survival analysis was done of aged mice that received repetitive eGFP  transplantations and of the non-treated controls. Differences in survival were tested for significance using a log-rank test. Statistical analysis was performed using the SPSS statistical package. The Student’s t-test was used to assess significant differences in expansion of D2 and B6 unfractionated BM cell and LSK transplantations.
2 \& r! i- B( L& H9 B( @: N$ {
. [; q( ]1 l1 H/ F8 _1 aRESULTS; A2 E0 n) H6 G7 A& i$ X# Q: ^

2 `7 i+ [! ?, sSurvival after Serial Transplantation
' m. _; M' Q8 q8 q; |" u! G3 V. Q4 A; [4 g# g4 N( z! a6 W2 K
Purified HSCs failed to sustain long-term survival of lethally irradiated recipients after four transplantations (Table 1). Survival was better in recipients transplanted with unfractionated BM cells. The transplantations of highly purified HSCs ended after the fourth step; due to survival of too few recipients, isolation of sufficient numbers of LSK cells for further transplantation could not be achieved.2 {* s' I( ^) M3 m* ^; n; L! H

; S. a* Z$ S: t7 m4 u. ?Evaluation of Donor Contribution; Z7 h0 A( r* G) V  L- C& n9 o( y
/ m( S/ N3 r% C9 t2 o& m
When purified HSCs are serially transplanted in B6 mice, it has been shown that donor-derived stem cells are abruptly lost after the 4th transplantation [39]. Although in our current study, we did not directly quantify donor stem cell chimerism in animals transplanted with purified cells, severe loss of stem cell quality after four transplantations was also evident, suggesting stem cell exhaustion (Table 1).; B. V5 H& {3 N0 U& }- n. W! Y1 P

1 R. T# ^2 b# a, Q: h/ OIn recipients transplanted with unfractionated BM cells, we did assess the percentage of original male donor cells by detection of Y-chromosome–specific sequences in individually isolated CFU-GM colonies after the third and sixth transplantation. After the third transplantation, >95% of the progenitors were of donor (male) origin in both D2 and B6 mice (Fig. 1A). In contrast, after the sixth transplantation, only few colonies (
+ i3 Y' F' k8 ?) B) q3 g0 a5 k" p! F1 V( L1 u2 u( A+ X, \4 N
Such a gradual loss of donor-derived stem cells and recurrence of recipient hematopoiesis after serial transplantations, as we report here, has been observed in previous studies in which unfractionated BM cells were used [10].
3 H5 s$ I3 \  f7 x  z3 s: G9 K8 c  X9 O) Z$ p  ?. b
As no substantial part of the hematopoietic system was derived from the original male donor after the sixth transplantation, the ones using unfractionated BM were ended after the fifth transplant.
0 s% z+ {! [+ \8 [' n' v/ O2 D0 X, f( ?) v/ Q
Stem Cell Numbers after Serial Transplantations
. ~' X+ ^4 H$ }3 t/ N7 `: |8 D
( u/ {& s; D) @1 gWhen 1,500 purified HSCs were transplanted, a continuous decrease of CAFC d35 frequencies in D2 mice was observed (Fig. 2A). In contrast, frequencies in B6 mice increased at first, and only after three transplantations was a severe decline noted. Using unfractionated BM, CAFC d35 frequencies decreased in B6 mice, but, again, more dramatically in D2 animals after the first three transplantations (Fig. 2B). Hereafter, the frequency continued to decline for D2 animals but stabilized in B6 mice. The decline of CAFC d35 frequency in combination with lack of donor contribution clearly indicates functional exhaustion of stem cells.
& ~; t5 R1 T" I7 W  }2 M% W# Z/ q8 n4 D
In conjunction with a decline in cobblestone area–forming ability, a gradual decrease of the percentage of LSK cells as identified by FACS was observed (Fig. 2C). No difference was seen between the frequency of LSK cells in D2 and B6. The decline of the percentage of LSK cells in the BM of the recipients resulted in a gradual reduction of the total LSK pool size per animal (data not shown). In addition, in B6 mice a minor decrease was observed for the clonogenic CAFC d35 activity of the purified cells, until a sudden drop after the fourth transplant (Fig. 2D). This suggests severe functional impairment of these serially transplanted HSCs. In D2 mice, the clonogenic activity returned to normal levels after a small decline after the first and second transplant.
7 e' P9 O6 T7 f! O7 A+ q, ^- W* d
2 x& ]4 H* G  H! S7 xCompetitive In Vivo Assays. J0 F, \7 P: q, \! D

9 N! T4 N8 z( d' zAlthough it has been well documented that during normal, unperturbed hematopoiesis there is a strong overlap between LSK FACS phenotype, in vitro functional CAFC activity, and in vivo long-term repopulating ability, this correlation is not always evident in specific experimental conditions in which the system is perturbed, such as serial transplantations [15]. Therefore, to confirm functional stem cell decline measured by FACS (Fig. 2C) or CAFC potential (Fig. 2A–B, D), we mixed 5x serially transplanted unfractionated B6 BM cells with an equal number of freshly isolated eGFP  young BM cells, and co-transplanted both cell populations in lethally irradiated B6 recipients. Although 5x serially transplanted BM, which consist of a mixture of original donor cells and irradiated recipients, possessed substantial numbers of CAFC d35 (Fig. 2B), there was no evidence of any in vivo stem cell activity when transplanted with similar numbers of CAFC d35 derived from young competitors (Fig. 3A). However, when 5x serially transplanted BM was transplanted without competitors (i.e., a 6th transplant), all recipients survived, clearly documenting that remaining stem cells were present and engrafted (Fig. 3B). To further illustrate that after serial transplantations stem cells have acquired a dramatic competitive defect when compared with freshly isolated BM that is not due to a difference in homing, we transplanted, without further conditioning, eGFP  unfractionated BM cells in animals that had received a transplant of 5x serially transplanted BM cells 3 months earlier. Freshly isolated BM cells engrafted very rapidly in these recipients, and hematopoiesis converted fully to an eGFP  genotype (Fig. 3B), which confirms a competitive disadvantage of serially transplanted cells.. _& r" J, x8 W. r* c- V

) f; R( b( I  L' pCalculating Stem Cell Expansion9 r, t3 }- v3 N$ E
  e' w/ W$ U! j7 N2 g) k# x
One of the aims of this study was to estimate the total number of population doublings of CAFC d35. Clearly, such calculations require assumptions and are affected by choice of the assay. It has been shown previously (and this was confirmed in the present study, Figure 2C–D) that the FACS phenotype is not very appropriate, as functional activity of phenotypically identical cells is highly affected by serial transplantations [15]. In vivo competitive repopulation assays have been used frequently and are very informative. However, in our current study we show that although serially transplanted BM cells may have no competitive repopulating potential, they may still contain stem cells, as they repopulate recipients when transplanted alone (Fig. 3A–B), and stem cell activity in these cells was detected by the CAFC assay (Fig. 2B). Therefore, although we do not claim that the CAFC assay is superior to other assay models in all circumstances, it was deemed valuable in the context of the current experiments.
6 p7 p" C9 i" g# v2 c8 h9 ?  c+ |! O2 Z0 M# Z5 {& z
The actual expansion of CAFC d35 after each transplant was calculated by dividing the total number of CAFC d35 present in the bone marrow in the entire mouse 4–6 months after transplantation, by the number of CAFC d35 infused (Fig. 4). Expansion of D2 CAFC d35 was stable (approximately 30-fold) during each serial transplantation of 1,500 LSK cells (Fig. 4A). Expansion of B6 CAFC d35 started at a much higher value (approximately 700-fold) but decreased rapidly with each transplant (Fig. 4A). During serial transplantation of unfractionated BM, the expansion of D2 CAFC d35 was again stable (approximately 15-fold), similar to transplanting purified LSK cells (Fig. 4B). Expansion of B6 CAFC d35 increased during the first three transplantations from 23-fold to 114-fold but dropped after the third transplant to ultimately 81-fold during the fifth transplant. The total cumulative expansion, which is the product of the expansion after each transplant, was dramatically higher (>1,000-fold) in B6 mice than in D2 mice, but similar whether purified LSK cells or unfractionated BM was used. (Fig. 4C–D).
/ ]) `! @. l* C! `3 o: U8 B; \  F- i/ o  D
The number of CAFC d35 self-renewing population doublings was estimated after three (a timepoint when stem cells were >95% donor-derived) and five transplantations (Fig. 5). Remarkably, the same maximum number of population doublings was reached when either highly purified HSC or unfractionated BM was used for serial transplantation. However, whereas the maximum number of population doublings for B6 was approximately 30, D2 stem cells could undergo only approximately 20 population doublings. The strain differences were already apparent after three serial transplantations of unfractionated BM cells.* f, \) a: }" Z$ J

' B& C+ w: @( f  U( b: s% K( r% YStem Cell Functioning during Normal Aging
* q' E; {6 R# X% `: o
: \2 M' L1 i. l+ c/ [0 H3 F& ?Most, if not all, HSC aging studies have tested the effect of replicative history on HSCs in transplantation assays, which require irradiation of the recipient. In fact, it has been suggested previously that an important part of the defect of serially transplanted HSCs is a direct result of the transplant procedure as such [9]. To determine whether a stem cell defect could also be observed during normal aging, we transplanted freshly isolated eGFP  young BM cells, harvested from 2-month-old donors into normal, unconditioned young (16 months) recipients. As expected, this strategy did not result in detectable levels of engraftment in the peripheral blood in nonconditioned young recipients. In contrast, however, eGFP  donor-derived leukocytes were readily detectable, albeit at low levels, in the majority of the old recipients (Fig. 6A). Repeated transplantations resulted in further increases of donor-type engraftment (Fig. 6B), which was multilineage (Gr-1 , Mac1 , B220 , CD3 ; data not shown).
" Z/ Q' k* O( L) k7 H* [8 v
6 q- u! u* |8 a$ p' t9 `DISCUSSION6 c5 R% [! V+ l3 l! X  ]2 {
: f  Q! ~( l* P+ u$ z/ c
In this study we assessed whether there is a limit to the number of population doublings that CAFC d35 can undergo. To this end, serial transplantations were performed using either unfractionated BM cells or highly purified HSCs. We observed exhaustion of stem cells, decreased stem cell clonogenic activity, reduced competitive repopulating activity, and impaired peripheral blood cell recovery, ultimately leading to decreased survival of recipients. We estimate that B6 stem cells (CAFC d35) are able to undergo 30 population doublings, whereas D2 cells are exhausted after 20 population doublings. This ultimately results in a 1,000-fold difference in expansion potential. Our results show that the maximum number of stem cell self-renewal divisions is finite and genetically determined.
: P3 T" e- Y% I3 W2 O! z; ?6 p  c* x# ]- C0 d: ]
Clearly, these values are an approximate estimate of stem cell self-renewal capacity. First, we have shown that recipient cells contribute to hematopoiesis in the final serial transplantations, which interferes with our calculation. Second, our estimate is assay-dependent and holds true when CAFC d35 are used as endpoints. This is illustrated by lack of engraftment in competitive assay while CAFC d35 activity was still observed. Finally, our estimate only applies to the entire population of cells, and we cannot rule out that individual, single HSCs can exceed this threshold. Importantly, the first two arguments imply that our calculations are an overestimate of the true self-renewal potential of the HSC population. It is unlikely that these limits of the amplification potential are reached during normal aging. To put this into perspective, in a study where B6 stem cells were labeled in vivo by BrdU, it has been shown that it takes ~2 months before >99% of all HSCs have undergone a cell division [40].This would suggest that during their entire lifespan (mean lifespan ~ 28 months) HSCs undergo ~14 divisions.
* q% S! u! U( P
" g% F3 E. K7 q% L3 jNonetheless, our data clearly reveal that young BM cells provide multilineage engraftment in aged (>16 months) recipients, as well as in recipients of five time serially transplanted BM cells. As this was not observed in young recipients, we believe that these engraftment levels indicate a functional defect of endogenous stem cells in old mice and in serially transplanted BM cells. However, this stem cell defect is very mild in aged recipients, and, consequently, the levels of engraftment are modest. Therefore, we were not surprised that repeated cell transfer did not extend lifespan, although the trend that we observed is intriguing. Much larger groups of mice will be needed to rigorously test the lifespan-extending potential of HSC transplantations. In addition, B6 mice may not be the most optimal model to test this, as these mice are relatively age-resistant. Interestingly, it has recently been shown that caloric restriction, currently the only effective way to extend lifespan in rodents, preserves stem cell function in old BALB/c mice [41].
6 _  B0 b$ c6 P  @% [; i3 _& e0 ~4 z8 z9 h# D# s
A number of potential explanations for stem cell exhaustion may be coined, among which the hypothesis of telomere loss during cell division is the most frequently supported one. Decreased telomere length of stem cell chromosomes after each transplant is a potential mechanism that could limit the maximum number of self-renewal divisions. It has been postulated that loss of telomeric DNA with age might function as a mitotic clock during normal aging [42, 43], as well as during serial transplantation of HSCs [44]. In addition, an association has been made between the self-renewal potential of HSCs and telomerase activity [45]. Importantly, stem cells isolated from telomerase (mTERT)–deficient mice can only be serially transplanted twice. This is accompanied by a rapid decline in telomere shortening in HSCs and reduction of long-term repopulating capacity [39, 46]. However, although HSCs overexpressing mTERT showed maintenance of telomere length after serial transplantation, no extended replicative capacity was observed, compared with wild-type mice [47]. Recently, it has been suggested that telomere length as such is less important for the process of aging than specific molecular alterations in telomere structure during aging [48, 49]. The role of telomere shortening and telomerase activity in HSCs of mice is further obscured as mice have longer telomeres than humans have [50]. With respect to our current results, it is unlikely that telomere erosion contributes to the reduced self-renewal ability of D2 HSCs as D2 animals have longer telomeres than B6 mice have [51].
* r2 s+ z8 l% _1 [; f% Z2 z2 S8 H# b' e9 @4 x& B; P
Another process that might affect in vivo stem cell self-renewal is proliferative activity of hematopoietic cells, which is consistent with a previously suggested model that proliferation leads to loss of stem cell quality [52]. The higher cycling activity of D2 progenitor cells might lead to more rapid exhaustion of D2 HSCs [17]. However, although D2 CAFC d35 made fewer self-renewing population doublings, we did not observe faster exhaustion of these cells, so other mechanisms are involved as well. Theoretically, HSCs have the option to either differentiate, self-renew, or die. During steady-state hematopoiesis the self-renewal probability is .5. We speculate that D2 HSCs, during serial transplantation, have a preference to differentiate rather than to self-renew. This would explain that, although there is a lower expansion per transplant and a lower number of maximal population doublings, the white blood cells and hematocrit in D2 mice are stable for a sustained period of time. Therefore, a reduced self-renewal probability (p ! n: C( F/ M  d

% J$ K" h# {. _$ x' Y9 N2 F/ wIn conclusion, our data clearly establish that maximum self-renewal capacity of HSCs is genetically controlled. The genetic balance between differentiation and stem cell self-renewal may affect stem cell aging and may possibly affect organ lifespan.
0 ~- h/ b5 g6 ?; P1 L2 q& L) `" d. X
ACKNOWLEDGMENTS7 W- Y' I8 D/ i4 J& `! d: \# R! [! ~

  [% I  o6 X- T  `3 F& [& z& eThis study was supported by the Netherlands Organization for Scientific Research (NWO) (grant number 901-08-339) and the Dutch Cancer Society (RUG 2000-2182). We thank Geert Mesander and Henk Moes for excellent assistance with flow cytometry.1 C0 N/ X4 X) r% x3 o5 ~

; g3 f, C" ?9 J! T% M& I/ j& gREFERENCES
' ~7 o1 o/ m4 V
* N) F' Z% F, I  [3 zHarrison DE. Mouse erythropoietic stem cell lines function normally 100 months: loss related to number of transplantations. Mech Ageing Dev 1979;9:427–433.7 F- `( H8 F6 q
/ P) H# v0 E8 B, ?% T
Siminovitch L, Till JE, McCulloch EA. Decline in colony-forming ability of marrow cells subjected to serial transplantion into irradiated mice. J Cell Physiol 1964;64:23–31.) e6 T7 R# _& z, O, N, ^
6 e# t- V! y+ e$ P5 O
Cudkowicz G, Upton AC, Shearer GM et al. Lymphocyte content and proliferative capacity of serially transplanted mouse bone marrow. Nature 1964;201:165–167.- E; J8 ?0 v+ ?! s% w

, A* J( b1 S; R8 c# `; c8 X2 `" bMicklem HS, Ogden DA. Ageing of haematopoietic cell populations in the mouse. In: Cairnie AB, Lala PK, Osmond DG, eds. Stem Cells of Renewing Cell Populations. New York: Academic Press, 1976:331–341.1 g5 I* ]& V! A& x" f: A

8 \3 S/ _/ M% m, p: tOgden DA, Mickliem HS. The fate of serially transplanted bone marrow cell populations from young and old donors. Transplantation 1976;22:287–293
/ [. l7 F! p3 j4 F8 Y/ a; ^! e9 r, ^% W- y6 y# T
Harrison DE, Astle CM, Delaittre JA. Loss of proliferative capacity in immunohemopoietic stem cells caused by serial transplantation rather than aging. J Exp Med 1978;147:1526–1531.
' u0 @4 b: L3 m# }3 c; Y+ v: ]& J; F6 ]* a
Hellman S, Botnick LE, Hannon EC et al. Proliferative capacity of murine hematopoietic stem cells. Proc Natl Acad Sci U S A 1978;75:490–494.4 ]8 t) N" x1 G7 N) z* \3 \
$ m% b) l! O& n# s* V( Y
Ross EA, Anderson N, Micklem HS. Serial depletion and regeneration of the murine hematopoietic system. Implications for hematopoietic organization and the study of cellular aging. J Exp Med 1982;155:432–444." n, K# Z; f7 z- F" L6 M2 \6 r( g2 N
: y; e. ?: @8 k9 o. n/ C; V
Harrison DE, Astle CM. Loss of stem cell repopulating ability upon transplantation: effects of donor age, cell number, and transplantation procedure. J Exp Med 1982;156:1767–1779.1 g5 G7 I" d6 }6 S
1 P% v9 h: n7 {/ y/ u9 o
Iscove NN, Nawa K. Hematopoietic stem cells expand during serial transplantation in vivo without apparent exhaustion. Curr Biol 1997;7:805–808., F# D; }3 [* e# n) g% p

6 k0 y7 D1 B% i! u% mMauch P, Hellman S. Loss of hematopoietic stem cell self-renewal after bone marrow transplantation. Blood 1989;74:872–875.
: l: @0 ?# @: M* o
% m$ L3 u+ ^# D9 B4 F; k1 Rde Haan G, Nijhof W, Van Zant G. Mouse strain-dependent changes in frequency and proliferation of hematopoietic stem cells during aging: correlation between lifespan and cycling activity. Blood 1997;89:1543–1550.
4 `3 D& n$ {1 ^% b) l
- Z9 I4 u3 p- I4 l9 [de Haan G, Szilvassy SJ, Meyerrose TE et al. Distinct functional properties of highly purified hematopoietic stem cells from mouse strains differing in stem cell numbers. Blood 2000;96:1374–1379./ k. }, K' P6 F: E! @  J; t. Q

$ b& Y; J3 S6 c4 a! [/ }6 ?+ RKamminga LM, Akkerman I, Weersing E et al. Autonomous behavior of hematopoietic stem cells. Exp Hematol 2000;28:1451–1459.2 m7 o; V) q3 J1 _
8 @, b  C3 a4 T" U
Müller-Sieburg-Christa E, Cho-Rebecca H, Sieburg-Hans B et al. Genetic control of hematopoietic stem cell frequency in mice is mostly cell autonomous. Blood 2000;95:2446–2448.6 j: J: t) D. p$ _
+ T  w7 ?% A, m6 K4 S; y9 e
Hayflick L, Moorhead PS. The serial cultivation of human diploid cell strains. Exp Cell Res 1961;25:585–621.' i, O& G3 S* s- h6 b
2 r) i! f% X) }3 c7 r% |
Hayflick L. The limited in vitro lifetime of human diploid cell strains. Exp Cell Res 1965; 37:614–636.
0 Z$ z2 i# s" z$ S5 P. _
. {0 G9 H+ ]5 {- T( fPole D, Simpson E. Genetic control and effector cells in host-versus-graft responses to H-Y antigen in mice. Transplantation 1983;36:546–551.1 [% z" w6 C  n4 X; Y, y

* f+ z5 m& V& o  N& d+ mIkawa M, Yamada S, Nakanishi T et al. "Green mice" and their potential usage in biological research. FEBS Lett 1998;430:83–87.' E  N% N! L( c9 X6 h

4 W6 [  @5 L4 x1 P, t/ BOkabe M, Ikawa M, Kominami K et al. "Green mice" as a source of ubiquitous green cells. FEBS Lett 1997;407:313–319.& j2 Z2 s2 {& Q* s+ A

5 ]2 v9 C8 F' N; Z1 I7 |de Haan G, Ploemacher R. The cobblestone-area-forming cell assay. In: Klug CA, Jordan CT, eds. Hematopoietic Stem Cell Protocols. Towata, NJ: Humana Press, 2002:143–152.6 K6 f2 ~8 l3 G$ p2 x+ C

+ l2 o- r6 r! H4 X. JPloemacher RE, van der Sluijs JP, van Beurden CA et al. Use of limiting-dilution type long-term marrow cultures in frequency analysis of marrow-repopulating and spleen colony-forming hematopoietic stem cells in the mouse. Blood 1991;78:2527–2533.# \4 [( e0 W6 r  \8 c3 Z& r

  _* d% m5 K# g: }Chervenick PA, Boggs DR, Marsh JC et al. Quantitative studies of blood and bone marrow neutrophils in normal mice. Am J Physiol 1968;215:353–360.
0 L. X4 k1 C& R  l. [0 S5 _
$ R: c  Q- F6 O/ }& W% L9 t" iBoggs DR. The total marrow mass of the mouse: a simplified method of measurement. Am J Hematol 1984;16:277–286.
) ^* h* t( c0 A9 B) B% [# f5 y- v1 d' {1 ^
Blasco MA, Lee HW, Hande MP et al. Telomere shortening and tumor formation by mouse cells lacking telomerase RNA9 z2 y1 L% Q7 ]! m

. S8 e# r1 \7 VAllsopp RC, Morin GB, DePinho R et al. Telomerase is required to slow telomere shortening and extend replicative lifespan of HSC during serial transplantation. Blood 2003; 102(2):517–520
) S0 B" ]% Z. v5 o2 T! v0 |, _" e- U7 F; f1 u# P( F4 Y4 ^% }+ i
Cheshier SH, Morrison SJ, Liao X et al. In vivo proliferation and cell cycle kinetics of long-term self-renewing hematopoietic stem cells. Proc Natl Acad Sci U S A 1999;96:3120–3125.3 F1 n" r$ _7 ?0 p5 p, C& B& X: a& t

5 w2 V( [4 q  y% T! j/ zChen J, Astle CM, Harrison DE. Hematopoietic senescence is postponed and hematopoietic stem cell function is enhanced by dietary restriction. Exp Hematol 2003;31:1097–1103.
" M9 m1 c/ F# C' ?/ k
" j3 G' c! H' o5 D( f( z. l- oVaziri H, Dragowska W, Allsopp RC et al. Evidence for a mitotic clock in human hematopoietic stem cells: loss of telomeric DNA with age. Proc Natl Acad Sci U S A 1994;91:9857–9860, s9 s) n: v( Q6 [
& W' y/ M; f) q& S, U. T+ Y/ y
Notaro R, Cimmino A, Tabarini D et al. In vivo telomere dynamics of human hematopoietic stem cells. Proc Natl Acad Sci U S A 1997;94:13782–13785.) W) M3 u( J' e* R, ~
; }) [/ p+ K( N" b* U9 A7 y+ `
Allsopp RC, Cheshier S, Weissman IL. Telomere shortening accompanies increased cell cycle activity during serial transplantation of hematopoietic stem cells. J Exp Med 2001;193:917–924.* c+ @% Q1 G- i: @

! i) l5 Z: ~# F8 |+ C0 T: RMorrison SJ, Prowse KR, Ho P et al. Telomerase activity in hematopoietic cells is associated with self-renewal potential. Immunity 1996;5:207–216.& R( k3 M: u. a2 M
7 N) f9 H: g$ l- m
Samper E, Fernandez P, Eguia R et al. Long-term repopulating ability of telomerase-deficient murine hematopoietic stem cells. Blood 2002;99:2767–2775.4 x- J" R1 D, T# N! Z% Z) `9 ~
2 e" b6 l  P' Y
Allsopp RC, Morin GB, Horner JW et al. Effect of TERT over-expression on the long-term transplantation capacity of hematopoietic stem cells. Nat Med 2003;9:369–371.+ m, d% E) R( Z
: P. V" P9 `0 S, f* s$ Z
Karlseder J, Smogorzewska A, de Lange T. Senescence induced by altered telomere state, not telomere loss. Science 2002;295:2446–2449.
# x4 L/ A2 u1 \1 t1 X6 f* A) X3 m$ k( [" u
Stewart SA, Ben Porath I, Carey VJ et al. Erosion of the telomeric single-strand overhang at replicative senescence. Nat Genet 2003;33:492–496.
( C  ^% C: X  R; Z8 L- @$ q) `) L) d( n3 E( M9 k' S
Kipling D, Cooke HJ. Hypervariable ultra-long telomeres in mice. Nature 1990;347:400–402.
" ]7 P6 [  R+ G3 ~0 {1 ?, V
/ l2 y+ A+ S, t* I, m# i2 LManning EL, Crossland J, Dewey MJ et al. Influences of inbreeding and genetics on telomere length in mice. Mamm Genome 2002;13:234–238.
& x- H! a/ a3 S/ w5 ~: N
2 q! ~1 T) ~6 c9 Y8 ^Van Zant G, De-Haan G, Rich IN. Alternatives to stem cell renewal from a developmental viewpoint. Exp Hematol 1997;1997:187–192.
; c; o9 h. a: L! R1 L) T% ~; ^
$ F* t5 V6 M& yvan Os R, Robinson S, Sheridan T et al. Granulocyte colony-stimulating factor enhances bone marrow stem cell damage caused by repeated administration of cytotoxic agents. Blood 1998;92:1950–1956# S% E- L( W$ ?  Q' ~2 `) l9 d
3 t7 O, Q, |  b+ U- k$ V
De-Haan G, Van Zant G. Dynamic changes in mouse hematopoietic stem cell numbers during aging. Blood 1999;93:3294–3301.(Leonie M. Kammingaa, Rona)

Rank: 1

积分
威望
0  
包包
9  
沙发
发表于 2009-3-21 09:14 |只看该作者
还不错

Rank: 1

积分
威望
0  
包包
7  
藤椅
发表于 2009-4-1 08:31 |只看该作者
感谢胞友分享

Rank: 7Rank: 7Rank: 7

积分
1530 
威望
1530  
包包
7769  

新闻小组成员 小小研究员 博览群书 帅哥研究员 优秀会员 金话筒 热心会员 精华勋章 积极份子

板凳
发表于 2013-8-5 13:37 |只看该作者
干细胞之家微信公众号
本帖最后由 ambassador 于 2013-8-5 13:37 编辑
! ]) P2 Y  I1 q( H
* l, d1 z# n/ m! n$ ^( E2 T6 }文章在哪里?

Rank: 1

积分
威望
0  
包包
1  
报纸
发表于 2013-10-16 15:25 |只看该作者
顶顶顶

Rank: 2

积分
72 
威望
72  
包包
1942  
地板
发表于 2015-5-29 10:29 |只看该作者
我的啦嘿嘿  

Rank: 2

积分
80 
威望
80  
包包
1719  
7
发表于 2015-6-25 10:10 |只看该作者
感谢党和人民的关爱~~~  

Rank: 2

积分
132 
威望
132  
包包
1727  
8
发表于 2015-7-1 16:43 |只看该作者
水至清则无鱼,人至贱则无敌!  

Rank: 2

积分
72 
威望
72  
包包
1942  
9
发表于 2015-7-8 21:48 |只看该作者
不错,看看。  

Rank: 2

积分
116 
威望
116  
包包
1832  
10
发表于 2015-7-20 21:27 |只看该作者
回答了那么多,没有加分了,郁闷。。  
‹ 上一主题|下一主题
你需要登录后才可以回帖 登录 | 注册
验证问答 换一个

Archiver|干细胞之家 ( 吉ICP备2021004615号-3 )

GMT+8, 2024-11-1 07:41

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.