Mechanisms of Ageing and Development 122 (2001) 1537– 1553 www.elsevier.com/locate/mechagedev Viewpoint Does functional depletion of stem cells drive aging? David Schlessinger a,*, Gary Van Zant b a Laboratory of Genetics, Gerontology Research Center, National Institute on Aging, NIH-NIA, 5600 Nathan Shock Dri6e, Baltimore, MD 21218, USA b Di6ision of Hematology/Oncology, Markey Cancer Center, Uni6ersity of Kentucky Medical Center, 800 Rose St., Lexington, KY 40536 -0093, USA Received 16 April 2001; received in revised form 17 May 2001; accepted 18 May 2001 Abstract The regenerative power of stem cells has raised issues about their relation to aging. We focus on the question of whether a decline in the function of stem cells may itself be a significant feature of aging. The question is implicitly two-fold: does functional depletion of stem cells affect the accumulation of aging-related deficits, and -- whether or not depletion is significant -- can activation of stem cells alleviate deficits? Two types of system are considered: 1) the exhaustible pool of ovarian follicles. The depletion of follicles leads to the aging-related phenomenon of menopause; and 2) the reserve of hematopoietic stem cells. Substantial numbers are sustained throughout life, but in mouse models, endogenous replicative activity has been shown to decline sharply with age. We discuss the possible implications of these observations for the rate of aging and the prospects for intervention. © 2001 Elsevier Science Ireland Ltd. All rights reserved. Keywords: Immortal cells; Ovarian follicles; Hematopoiesis; Lifespan * Corresponding author. Tel.: +1-410-558-8337; fax: +1-410-558-8331. E-mail address: schlessingerd@grc.nia.nih.gov (D. Schlessinger). 0047-6374/01/$ - see front matter © 2001 Elsevier Science Ireland Ltd. All rights reserved. PII: S 0 0 4 7 - 6 3 7 4 ( 0 1 ) 0 0 2 9 9 - 8 1538 D. Schlessinger, G. Van Zant / Mechanisms of Ageing and De6elopment 122 (2001) 1537–1553 1. The aging manifesto We are born from immortal cells. Yet all of us are chained by aging and mortality. How does this come about, and what can we do about it? Restated in standard scientific terms, there has been a fundamental paradox in biological thinking about aging and death compared with embryonic life. The paradox has been mentioned as relevant to the evolution of aging (Kirkwood and Austad, 2000), but recent discoveries about stem cells make it unavoidable to confront it in detail. The confrontation, discussed here, questions traditional assumptions about the nature of aging and the possibility of intervention in the aging process. 2. Germ cells versus ‘disposable’ somatic cells By definition, our hereditary potential is passed on by immortal, totipotent cells: that is, cells that can reproduce and initiate full development of new individuals in their progeny on an evolutionary time scale. An extra boost to survival is given by the production of a vast excess of germ cells, both eggs and sperm, compared with the number of progeny. In contrast to the immortal, totipotent germ cells, the rest of the body consists mainly of ‘mortal’, differentiated cells. This distinction has a special importance for theories of the mechanism of aging-related processes, for the substratum of aging has been taken to be the mortal cells and their integument products (Hayflick, 1994). Aging itself has often been ascribed to the accumulation of ‘damage’ of one kind or another in mortal cells— poorly-repaired damage of chromosomal DNA, stress-related mistakes in structural proteins or enzymes or turnover, deletions in mitochondrial DNA, etc. Based in part on such thinking, important discussions have ascribed aging to ‘disposable soma’ (Kirkwood, 1977; Kirkwood and Rose, 1991), with the notion that selection has chiefly operated to increase fertility, and the cells and tissues of the body need have only enough oomph to maintain function through child-bearing age. At that point the soma (that is, the individual) becomes evolutionary dead meat, and, therefore, unsurprisingly subject to insults that have no selective counterpressure to promote continued survival. This formulation has shown remarkable staying power, with masterful presentations and repetitions (Medawar, 1952; Charlesworth, 1993; Rose, 1991 Westendorp and Kirkwood, 1998; Kirkwood, 2001). But several considerations suggest that it may require modification. First, the distinction begs the question of what constitutes the difference between germ cells and ‘disposable’ cells. After all, germ cells should be subject to all the stresses and environmental changes and stochastic errors in DNA replication and deletion, etc. that are thought by various constituencies to result in aging. How do germ cells escape that formal cause, and if there are escape mechanisms, why don’t differentiated cells escape in a similar way? There has been some tendency to say that comparable immortality of somatic cells is linked to uncontrolled growth in D. Schlessinger, G. Van Zant / Mechanisms of Ageing and De6elopment 122 (2001) 1537–1553 1539 cancer (e.g. Kirkwood, 1977), but that assertion is controverted by recent research. For example, oligodendrocyte cells (Tang et al., 2001), glial cells (Mathon et al., 2001), and mammary epithelial cells (Romanov et al., 2001), which are clearly ‘disposable’, can apparently grow and divide indefinitely in culture without ‘aging’. So can a variety of stem cells (including embryonic stem cells, Suda et al., 1987), as discussed further below. The doctrine of disposable soma also assumes (or states as an inference) that there is no significant selection for ‘old age’ (or on the contrary, that there may be selection for genes that are positive at younger ages but have negative effects in old age, Williams, 1957). But within our own species, longevity has a clearly demonstrated genetic component, particularly for survival to ‘centenarian’ status (Perls et al., 1998, 2000). Therefore, at the very least, some individuals have a more disposable soma than others. The notion of disposability is further weakened if one takes into account the possibility (Schlessinger and Ko, 1998), fostered by versions of group selection, that there may be positive selection for only small numbers of very old individuals, with driving forces that might be negative for the bulk of a population. 3. Germ cells and soma, meet stem cells The notion of disposable soma has also been sustained by the traditional assumption that aging is a post-developmental process, occurring in a compartment of life that starts in late maturity (Hayflick, 1994; Miller, 2001). But this view is challenged by the findings with a third category of cells, ‘stem cells’, that retain developmental potential. Recent studies have sharply increased the repertoire of astonishing properties of stem cells. These cells are not in themselves ‘germ cells’, and are clearly differentiated, but they have long been known to retain the possibility of self-renewal as well as further differentiation into end-stage cells. Successive adoptive transfer experiments have demonstrated that mouse lympho-hematopoietic cells, for example, can go through immense numbers of productive divisions to repopulate depleted marrow (Harrison, 1972; Iscove and Nawa, 1997; Siminovitch et al., 1964). Furthermore, and more relevant to this discussion, it is now clear that the ‘differentiation’ of stem cells is not irreversible, and one kind of stem cells can substitute for others to repopulate the reserves of many tissues (Bjornson et al., 1999; Brazelton et al., 2000; Brustle et al., 1999; Ferrari et al., 1998; Gussoni et al., 1999; Jackson et al., 1999; Kopen et al., 1999; Lagasse et al., 2000; Lavker and Sun, 2000; Mezey et al., 2000; Orkin, 2000; Uchida et al., 2000; Watt and Hogan, 2000; Weissman, 2000). Thus, stem cells retain the capacity to grow and divide apparently without limit, and they can ‘dedifferentiate’ and ‘redifferentiate’ (or transdifferentiate directly) to a new stem cell lineage (Weissman, 2000). In extreme tests of potential, both stem cells and ‘ordinary’ somatic cells can contribute nuclei to enucleated eggs to clone mammals (Wilmut et al., 1997; Wakayama et al., 1999, 2000). Such cloning experiments demonstrate that the differentiation and limited potential of cells that 1540 D. Schlessinger, G. Van Zant / Mechanisms of Ageing and De6elopment 122 (2001) 1537–1553 has been linked to aging is contingent. Contrary to earlier thinking, ‘commitment’ is often intrinsically reversible. The a priori and experimental weakening of the neat distinction between germ cells and soma is not merely academic. There are definable differences between these types of cells and stem cells, which are linked to genetic mechanisms of aging; and depending on what those differences are, there may or may not be interventions that can change the rate of aging or make the survival of all individuals approximate the longest lived in an extant population— or even augment survival beyond current limits. One can then ask, what special abilities or mechanisms of stem cells can be regained by somatic cells?, to what extent can we capitalize on reversibility to regenerate aging cells or tissues? and more generally, what is the relation of stem cells to aging? 4. Embryonic life and stem cell dynamics We have pointed out elsewhere (Schlessinger and Ko, 1998) that aging-related phenomena have a profound dependence on earlier processes, many of them initiated in utero. Although, no one would argue that the loss of cells and function with age is in any way the reverse of development, it seems obvious that knowing more about the process of formation of cells and tissues, and the factors required, can help to prolong or even to regenerate failing components of the aging body. One can then consider how, and how well the pool of stem cells is adapted to needs at different ages; or whether damaged (or non-dividing ‘crisis’, Hayflick, 1994) cells that are resistant to or not accessible to stem cell replacement contribute to the fundamental course of aging. To answer such concerns we require information about three features of stem cells throughout life: their numbers, their accessibility/ mobilizability, and their capacity to replace or supplement already existing mature cells. Fig. 1 contrasts two fundamental alternatives for cell dynamics, exemplified by mammalian ovarian follicles (Fig. 1a) and hematopoietic stem cells (Fig. 1b). In both cases, the cells retain indefinite capacity for growth and division. But they differ in the extent to which they are called on or available to exercise that capacity. Follicles are progressively depleted, whereas blood stem cells are repeatedly expanded in bursts of activity. (It is notable that evolution has no rigid, invariant commitment to strategies. For example, the nematode has no ‘somatic’ stem cells; and in Drosophila (Xie and Spradling, 2000), in contrast to mammals, ovarian follicles are made continuously through life, much in the manner of Fig. 1b). 5. Menopause and ovarian follicle dynamics At the moment of birth, a mammalian female infant has all the ovarian follicles that she is destined to have (Fig. 1a, Fig. 2). Reproductive lifespan is then D. Schlessinger, G. Van Zant / Mechanisms of Ageing and De6elopment 122 (2001) 1537–1553 1541 determined by the progressive depletion of that pool through successive menstrual cycles (Faddy, 2000; Simpson and Rajkovic, 1999; Morita and Tilly, 1999). The critical embryonic events that yield the final pool are thus the balance of synthesis and turnover of cells. The process is marked by the formation of a very large Fig. 1. (a) Ovarian follicle dynamics. A fixed pool of follicles available at birth is depleted by atresia, and after puberty, by ovulation until the numbers fall too low and menopause ensues. (b) Hematopoietic stem cell dynamics. Periodic bursts of production of mature cells occur from a self-renewing compartment of stem cells (it remains unclear whether there is clonal succession, with stem cells called upon one or a few at a time, or if all stem cells contribute continuously to most or all bursts; contradictory data (Harrison et al., 1987; Jordan and Lemischka, 1990) may result because the transplant model usually studied deviates from the homeostatic condition in vivo). 1542 D. Schlessinger, G. Van Zant / Mechanisms of Ageing and De6elopment 122 (2001) 1537–1553 Fig. 2. Follicle dynamics during life. The production of high levels of germ cells in utero (yellow) is accompanied by massive atresia; only about 10% of oocytes are protected by follicular cells (blue) by birth. The ovarian follicles and their oocytes are then progressively depleted until menopause (see Faddy, 2000). number of germ cells (of the order of 10 000 000) in fetal life. These are then largely discarded during a phase of active atresia (apoptosis). At the same time, however, about 600 000 are surrounded by follicular cells and largely preserved at birth and until puberty initiates the cycles of ovulation (Fig. 1a, Fig. 2). One might expect that a variety of transcription factors are involved in setting the rate and extent of synthesis of follicles, and that correspondingly, a number of genes would be involved in setting the rate of atresia. In fact, atresia seems to work through the Bcl system, but with a number of specialized components that modify the relative stability of follicles (Hsu and Hsueh, 2000; Morita and Tilly, 1999; Morita et al., 1999a,b). A clue to the regulation of the final number of follicles at birth comes from recent investigations of premature ovarian failure (POF; Tibiletti et al., 2000). In contrast to the mean age of menopause of about 51 in all cultures and eras that have been studied, about 1% of all women either never go through menarche or undergo primary or secondary amenorrhea before age 40 (Coulam et al., 1986; Chang et al., 1992). A fraction of these women are easily ascertained, because they are affected by the linked, complex eyelid condition of Blepharophimosis, Epicanthus Inversus, Ptosis Syndrome (BEPS, c 605597 in Mendelian inheritance in man, National Center for Biotechnology Information). D. Schlessinger, G. Van Zant / Mechanisms of Ageing and De6elopment 122 (2001) 1537–1553 1543 The gene responsible for BEPS has been cloned from its chromosome three locus (Crisponi et al., 2001). It proves to be the FOXL2 gene, a winged-helix transcription factor. Consistent with the phenotypes of BEPS and premature ovarian failure, the gene is expressed essentially only in developing eyelids and in ovarian follicles; the loss of function of one of the chromosome three alleles leads to eyelid overgrowth and ovarian follicle deficiency. It should now be possible to determine whether the gene truly acts as a rheostat to determine follicle number, and whether that effect is exerted at follicle synthesis or maintenance (in Turner syndrome, another genetic source of POF, the sole X also is thought to be haploinsufficient, leading to streak gonads (Zinn et al., 1993). This has been related to excessive atresia in at least some patients (Schlessinger et al., 2001; Simpson and Rajkovic, 1999)). Certainly, once the pool of ovarian follicles is formed, the simplest ‘counting’ mechanism for depletion would be a ‘commuter ticket’ model, in which a repeated event involves the ordered, sequential loss of a critical component-either as a function of time or of cyclical activity. The depletion of follicles indeed generally occurs for some follicles each ovulation cycle until a threshold value is reached beyond which ovulation fails and menopause ensues. The cyclical needs of ovulation are met by periodic release of the cascade of hormones that pick out a follicle and send it on its way from the ovarian pool. The detailed mechanism is unknown, but we want to point out three vital features: (1) the starting size of the pool, (2) the rate of depletion, and (3) the need for a ‘threshold’ level. As discussed above, the initial number is set during early development. In normal biology, the rate of turnover is then fixed by the ovulation cycle. The third feature, the ‘threshold’, remains mysterious. The remaining cells could have accumulated damage, like DNA breaks or mitochondrial deletions, that prevents them from functioning— that is, they could have damage equivalent to ‘aging’ (see below), essentially eliminating them from the pool of stem cells. But there is also the alternative notion of failure of ‘mobilizability’: even if competent stem cells (follicles) remain, they may be in a location where stimulating hormones cannot reach them, or the number of follicles may have fallen to a level where they cannot sustain an adequate level of a paracrine hormone, or there may be a physical or cellular interaction that prevents them from division (gap junctions with follicle cells, stretching of fibers from extracellular matrix, etc.). Many of the parameters that support the possible existence of ‘immobilizable’ cells have been better assessed in other models, particularly for hematopoiesis. 6. Hematopoiesis and other stem cell systems: is functional depletion relevant? In sofar as follicles represent (totipotent!) stem cells, they demonstrate that depletion of a stem cell reserve can indeed lead to an irreversible age-related loss of function, in the classic instance of menopause. But how relevant is this example to other types of stem cells (hematopoietic, germ cell, neuronal, etc.)? The major difference (Fig. 1b) is that cell dynamics do not yield a final pool of fixed size that 1544 D. Schlessinger, G. Van Zant / Mechanisms of Ageing and De6elopment 122 (2001) 1537–1553 is irreversibly depleted. Instead, an added degree of flexibility is achieved in two ways to modify the stem cell pool. In one, ongoing cell differentiation is required, for example, to sustain the steady state of red cells in blood in the face of turnover of end-stage mature cells. In the other process affecting the pool, stimuli like the reduction of oxygen, with the red cell maturation response stimulated by erythropoietin, or blood loss from a wound, or challenge by an infectious agent, lead to additional waves of differentiation (Fig. 1b). The question then arises, is depletion of their numbers a significant feature of the fate of stem cells that follow the dynamics of Fig. 1b? Based on all we know, the answer is ‘no’. For example, repetitive protocols in which a small number of mouse stem cells are used to reconstitute the hematopoietic system of irradiated animals can be successfully carried through multiple generations, and only extreme conditions, involving prior treatment of mice with cyclophosphamide and cytokines, mimicking some treatment protocols for cancer, damages the capacity of marrow cells to contribute to the serial transplants (Hornung and Longo, 1992). Also, studies of aging in inbred strains show that the number of bone marrow stem cells is not appreciably diminished in old age. In fact, numbers are increased in both of two strains, DBA/2 and C57BL/6, studied in detail at middle-age (1 year) to 2–5-fold the level in 2-month-old animals (de Haan and Van Zant, 1999a). After 1 year, a strain-dependent difference in numbers is observed, but is less than 2-fold over most of the lifespan (de Haan and Van Zant, 1999a; Morrison et al., 1996). Thus, it is not surprising that clinical medicine rarely defines a syndrome caused by loss of blood cell precursors (aplastic anemia and lymphopenia are exceptions, though they are not generally thought to be strictly age-dependent). On the other hand, although the numbers of stem cells tend to be maintained, inter-strain variations in cell cycle kinetics of stem and progenitor cells of the hematopoietic system of mice have been identified. To determine whether interstrain variations are cell-autonomous and thus under the control of ‘stem cell genes’, or are extrinsically regulated by the environment, strains with large, quantifiable variations were selected, and chimeric mice were constructed by aggregating embryos of the strains and implanting them into foster mothers. The resulting mice are cellular chimeras in which all tissues, including the hematopoietic stem cell system, consist of a mixture of cells derived from the two embryos. Any genotype-restricted differences in stromal and humoral milieu that may affect stem cell function are thought to be obviated in the chimeras, with all stem cells exposed to the same environment. This permits one to attribute observed differences to cell-autonomous (genetic) causes. Fig. 3 shows the level of chimerism in the peripheral blood leukocyte populations of mice generated by aggregating DBA/2 and C57BL/6 embryos (Van Zant et al., 1990). DBA/2 stem cell contributions to lympho/hematopoiesis decline with age, and after about 2 years, blood cells are entirely of C57BL/6 origin. The results demonstrate that the inter-strain differences are cell autonomous. Furthermore, because cells of both the lymphoid and myeloid compartments are affected, the effects probably take hold at the level of the stem cell. D. Schlessinger, G. Van Zant / Mechanisms of Ageing and De6elopment 122 (2001) 1537–1553 1545 Nevertheless, although, there is a growing numerical advantage of C57BL/6 stem cells in old-aged chimeras, DBA/2 stem cells are not gone, large numbers of potentially active DBA/2 stem cells are still present and competent (Van Zant et al., 1992). This is demonstrated in Fig. 4, in which blood cell chimerism is shown in a single chimeric animal. DBA/2 chimerism was about 15% early in life but by 2 years of age had dropped to 0, and there was no longer any DBA/2 hematopoiesis evident. At 31 months of age, after 9 months of only C57BL/6-derived hematopoiesis, the mouse was sacrificed and its marrow cells were transplanted into lethally irradiated recipients. DBA/2 stem cells were reactivated and contributed robustly to engraftment at a disproportionately high level (40%) relative to chimerism in the early life of the donor chimera ( 15%). With time, however, the stem cells again become quiescent. This cycle of activation and subsequent quiescence was repeated in another round of transplantation to secondary recipients (Fig. 4). These data argue that during aging, DBA/2 stem cells do not disappear, but become less sensitive or refractory to those environmental cues that foster differentiation, and possibly stem cell replication, in young animals. In other words, the curve of Fig. 3 superficially resembles the decline phase of Fig. 2, but not because stem cells are being ‘used up’ like ovarian follicles. Rather, DBA/2 stem cells are tending to become quiescent. Reactivation of DBA/2 stem cells somehow occurs during the transplantation procedure, modifying the ‘mobilizability’ of stem cells. In other words, mobilizability declines during the aging of the mice, with functional but relatively mild numerical depletion of stem cells. The cell cycle kinetics in the hematopoietic compartments of DBA/2 and C57BL/6 mice have been studied, and observed differences are consistent with the Fig. 3. Quiescence of DBA/2 stem cells in C57BL/6l DBA/2 embryo-aggregated chimeric mice. Blood cell chimerism was measured in a group of 6 – 8 chimeric animals over 3 years, using glucose phosphate isomerase polymorphism as a genotype-specific marker. Note the complete quiescence of the DBA/2 stem cell pool after 30 months. 1546 D. Schlessinger, G. Van Zant / Mechanisms of Ageing and De6elopment 122 (2001) 1537–1553 Fig. 4. Quiescence and re-‘mobilization’ of DBA/2 stem cells through transplantation. Marrow was harvested at 31 months from a chimera in which DBA/2 stem cells had ceased blood cell production, and was transplanted into lethally irradiated recipients. Marrow was subsequently harvested from the group of primary recipients at 37 months and transplanted into a second group of lethally irradiated recipients. Note the repetitive cycles of ‘mobilization’ and quiescence of stem cells during transplantation and engraftment. results of transplantation of chimeric marrow (de Haan and Van Zant, 1999a,b). Cell cycling of progenitor cells in young animals, measured by a standard assessment of the fraction of cells that survive hydroxyurea treatment, is much greater in DBA/2 mice, but in both strains the replication rate drops dramatically with age until it is barely detectable in old animals of either strain. The initial competitive advantage of DBA/2 stem cells when transplanted may derive from the generally higher rate of proliferation in their progenitor cell compartment. But what accounts for the arousal and robust function of deeply quiescent DBA/2 stem cells transplanted from old chimeras? Indeed, what causes the deep quiescence of the DBA/2 stem cells at a time when in the same environment old C57BL/6 stem cells continue to function normally and provide all of the requisite blood cells of all lineages? What accounts for the loss of ‘mobilizability’? Based on the probable importance of ‘other’ (extrinsic) features of stem cell recruitment, there may be an age-related change in the environment of stem cells that renders the cells less accessible or responsive to activation. Activation of stem cells would be favored in transplantation experiments, either as a result of exposure to the abnormal cytokine milieu found in lethally irradiated transplant recipients, of dissociation from their native stroma in preparing a cell suspension, or of association with new stroma in the transplant recipient. These notions are not novel; the work of Bissell’s group (Schmeichel et al., 1998) has established the determinative D. Schlessinger, G. Van Zant / Mechanisms of Ageing and De6elopment 122 (2001) 1537–1553 1547 role that can be played by extracellular matrix in replication/differentiation pathways. Although, it has been argued that there is complete tolerance to cells from both parents in chimeras (Mintz and Silvers, 1967; Mintz and Palm, 1969), it is difficult to exclude effects of natural killer cells or other immune phenomena on the relative decline of stem cells from one parent (Fig. 3). However, in transplant experiments using chimeric marrow, treatment of the recipients with anti-asialo GM1 antibody to kill NK cells had no effect on either the quantitative or temporal contributions of C57BL/6 and DBA/2 stem cells to engraftment (Van Zant et al., 1990). In both the antibody-treated and control groups, DBA/2 stem cells made robust contributions to early engraftment but, as in the chimeras themselves, ultimately became quiescent. In the same vein, the comparable, age-related decline of the fraction of replicating stem cells in every strain examined (see below) is less likely to be immune-modulated. Furthermore, there are fragmentary but suggestive indications of similar progressive quiescence of other types of stem cells. In particular, there are already several reports indicating that during brain aging, neural stem cells are present at normal levels, but for some reason, stem cell proliferation declines (Kuhn et al., 1996; Montaron et al., 1999). Again in this case, immune modulation would be relatively unlikely, but remains conceivable. Whatever the mechanism for quiescence and declining mobilizability of stem cells during aging, a working hypothesis can be formulated from a look at the relationship between mouse longevity and cell cycle kinetics (Fig. 5). There is a strong negative correlation between the mean lifespan of eight commonly used inbred mouse lines and the fraction of hematopoietic progenitor cells in S-phase of the cell cycle (de Haan and Van Zant, 1997). The natural lifespan of DBA/2 mice is very nearly the time at which DBA/2 hematopoiesis ceased in the chimeras discussed above, indicating either an intriguing coincidence or a causal relationship. In Fig. 5. Negative correlation between hematopoietic progenitor cells in cell cycle and the mean lifespan of eight inbred strains of mice. The strains (from lowest to highest cell cycle kinetics) were 129/J, C57BL/6J, BALB/cJ, A/J, DBA/2J, C3H/HeJ, AKR/J, and CBA/J. Cell cycle activity was measured by the fraction of progenitor cells (cobblestone area-forming cells counted at day 7) killed by a 1-h incubation with hydroxyurea. The regression line has a correlation coefficient of 0.55 that is statistically significant (PB 0.01). 1548 D. Schlessinger, G. Van Zant / Mechanisms of Ageing and De6elopment 122 (2001) 1537–1553 Fig. 6. Short- and long-term defenses against aging deficits. Homeostasis is subject to various stresses that deplete or damage function. The short-term (immediate) responses that help to maintain/restore homeostasis include intracellular turnover and repair as well as stress responses to oxidation, heat, etc. In addition, germ cells and stem cells can provide longer-term renewal that replaces or regenerates cells and tissues. provocative terms, these results suggest that functional depletion of stem cells not only occurs, but may itself be a principal feature of aging, and possibly a driving force. 7. The future of aging In the life of a metazoon, as schematized in Fig. 6, the effects of stress and wear-and-tear on homeostasis can be seen as counteracted by two types of processes. One is immediate, based on constitutive repair and intracellular turnover supplemented by other stress responses (including, e.g. heat shock, panic, acute phase, and apoptotic responses). Other, longer-term responses to stress involve critical interactions with embryonic processes. To recapitulate, they are based on the regeneration and renewal of cells and tissues, which are dependent in turn on the setting of stem cell pool size and rates of turnover in early development. As reviewed here, functional depletion of fixed numbers dominates the subsequent fate of ovarian follicles, whereas turnover rates and mobilizability, with little drop in numbers, may determine the fate of most stem cells. The underlying mechanisms and genetic factors determining the numbers, turnover rate, and mobilizability of stem cells remain to be elucidated, though they seem to be related to cell cycle checkpoint controls that may be more or less stringent in different strains, but always lead to increasing quiescence (cf. Martin and Oshima, 2000). D. Schlessinger, G. Van Zant / Mechanisms of Ageing and De6elopment 122 (2001) 1537–1553 1549 One can explicitly note that the fundamental difference between germ cells and somatic cells, the paradox stated at the start of this discussion, is not resolved here. In fact, the formal cause of aging-oxidation, mitochondrial deletion, etc. remains undetermined. In organisms like nematodes, which lack stem cells, aging acts on somatic cells in an approximation of the ‘disposable soma’ formulation, and many have argued plausibly that a universal mechanism underlies aging in all species (see below). Furthermore, the insults that produce aging may occur in stem cells as well as somatic cells. In fact, the correlation between lifespan and stem cell activity in inbred mouse strains (Fig. 5) may be fortuitous. At present, mammalian lifespan is usually limited by specific diseases (heart disease, end-stage renal failure, etc.) that are not thought to involve a reduction in stem cell activity in their etiology. It is, therefore, highly speculative to think that organ-specific depletion of stem cell function could determine the fate of an organ, and thereby be involved in determining lifespan, and in that sense, the title to this essay is truly a question to be investigated, and not a claim. Nevertheless, regardless of the degree to which stem cells may be involved in particular disease processes, the existence and potential of stem cells make an important difference in discussing current prospects for aging. To be more precise: stem cell-based intervention is foreseeable. The flexibility of possible intervention is increased by two findings. First, as reviewed above, there is increasing evidence for the plasticity of stem cells: neuronal or other stem cells can repopulate a number of stem cell compartments in the body, apparently because their commitment is partial or reversible. Second, stem cell manipulation in vitro may remedy problems of mobilizability. In many scenarios of ‘regeneration’ or ‘supplementation’, the endogenous numbers and milieu of stem cells appear to be secondary. Instead, projections proceed on the basis of increasing the numbers of functioning stem cells in models like marrow transplantation or even egg transfer from donors. In other words, stem cells are in effect activated (‘remobilized’) in vitro and then provided back to a host. Such proposals are consistent with the general paradigm of a rate of aging that can be shifted by intervention, as inferred from the studies of caloric restriction in organisms ranging from yeast (Jazwinski, 2000) to nematode (Lakowski and Hekimi, 1998) to rodents (Lee et al., 2000) to monkeys (Lane, 2000). Both longevity and reserves against stress (Sohal and Weindruch, 1996; Mattson, 2000) are increased by limitations in caloric intake. In an independent line of research in humans, the work of Perls’ group (Hitt et al., 1999; Perls et al., 1998, 2000) has suggested that a ‘rate of aging’, under genetic control, is modified in supercentenarians. It should soon be known whether caloric restriction and supercentenarian advantage act on the same or different genetic substrata. In particular, it can be asked whether mouse strains with differential longevity or differential responses to caloric restriction have corresponding differences in stem cell kinetics and mobilizability. Already mouse studies show the inverse relation of longevity and stem cell kinetics (Fig. 5), and in a first hint about possible features of reserves in supercentenarians, Perls’ findings include the suggestive observation that supercentenarian women have a recorded mean age of last childbirth of 46.2 years, considerably in excess of peer female cohorts who survive menopause but die younger (Perls et al., 1997). 1550 D. Schlessinger, G. Van Zant / Mechanisms of Ageing and De6elopment 122 (2001) 1537–1553 In conclusion, it is early times in the study of stem cells, and one can mainly raise issues at this time. It seems increasingly likely, however, that an understanding of the determination of numbers, rate of turnover, and mobilization/activation of various types of stem cells will provide ways to prevent or alleviate many unwanted age-related phenomena. Stem cells are suspended in a limbo between an immortal state and irreversible, mortal commitment, and could in principle replace every structure, including ‘poorly renewable’ collagen, etc. They may thus be superimposed on any detailed mechanism of aging to determine the fate of the aging body. Returning to our initial statement, We have nothing to lose but our chains. Can the regulation of functional depletion of stem cells provide the key to unlock them? Acknowledgements This review and its formulation are dedicated to our teacher Gene Goldwasser, in the context of our own developments (‘mobilization’?) at the University of Chicago. We thank Dr Minoru Ko for drafting Fig. 1, Dr Antonino Forabosco for the formulation of Fig. 2, and Dr Mark Mattson and Dr Dan Longo for helpful suggestions. References Bjornson, C.R.R., Rietze, R.L., Reynolds, B.A., Magli, M.C., Vescovi, A.L., 1999. Turning brain into blood: a hematopoietic fate adopted by adult neural stem cells in vivo. Science 283, 534 – 537. Brazelton, T.R., Rossi, F.M.V., Keshet, G.I., Blau, H.M., 2000. From marrow to brain: expression of neuronal phenotypes in adult mice. Science 290, 1775 – 1779. Brustle, O., Jones, K.N., Learish, R.D., Karram, K., Choudhary, K., Wiestler, O.D., Duncan, I.D., McKay, R.D.G., 1999. Embryonic stem cell-derived glial precursors: a source of myelinating transplants. Science 285, 754 –756. Chang, J.S., Lee, J.Y., Moon, S.Y., Yoon, B.K., Kim, J.G., 1992. Diagnosis and management of premature ovarian failure. In: Fujimoto, S., Mizuno, M., Segal, S.J. (Eds.), Recent Advances in Ovarian Function: Basic and Clinical Researches, pp. 265 – 279. Charlesworth, B., 1993. Evolutionary mechanisms of senescence. Genetica 91, 11 – 19. Coulam, C.B., Adamson, S.C., Annegers, J.F., 1986. Incidence of premature ovarian failure. Obstet. Gynecol. 67, 604 –606. Crisponi, L., Deiana, M., Loi, A., Chiappe, F., Uda, M., Amati, P., Biscelia, L., Zelante, L., Nagaraja, R., Porcu, S., Ristaldi, M.S., Marzella, R., Rocchi, M., Nicolino, M., Lienhardt-Roussie, A., Nivelon, A., Verloes, A., Schlessinger, D., Gasparini, P., Bonneau, D., Cao, A., Pilia, G., 2001. The putative forkhead transcription factor FOXL2 is mutated in belopharophimosis/ptosis/epicanthus inversus syndrome. Nat. Genet. 27, 159 – 166. de Haan, G., Van Zant, G., 1997. Intrinsic and extrinsic control of hemopoietic stem cell numbers: mapping of a stem cell gene. J. Exp. Med. 186, 529 – 536. de Haan, G., Van Zant, G., 1999a. Dynamic changes in mouse hematopoietic stem cell numbers during aging. Blood 93, 3294 –3301. D. Schlessinger, G. Van Zant / Mechanisms of Ageing and De6elopment 122 (2001) 1537–1553 1551 de Haan, G., Van Zant, G., 1999b. Genetic analysis of hemopoietic cell cycling in mice suggests its involvement in organismal life span. FASEB J. 13, 707 – 713. Faddy, M.J., 2000. Follicle dynamics during ovarian ageing. Mol. Cell. Endocrinol. 163, 43 – 48. Ferrari, G., Cusella-De Angelis, G., Coletta, M., Paolucci, E., Stornaiuolo, A., Cossu, G., Mavilio, F., 1998. Muscle regeneration by bone marrow-derived myogenic progenitors. Science 279, 1528 – 1530. Gussoni, E., Soneoka, Y., Strickland, C.D., Buzney, E.A., Khan, M.K., Flint, A.F., Kunkel, L.M., Mulligan, R.C., 1999. Dystrophin expression in the mdx mouse restored by stem cell transplantation. Nature 401, 390 –394. Harrison, D.E., 1972. Normal function of transplanted mouse erythrocyte precursors for 21 months beyond donor life spans. Nat. New Biol. 237, 220 – 222. Harrison, D.E., Lerner, C., Hoppe, P.C., Carlson, G.A., Alling, D., 1987. Large numbers of primitive cells are active simultaneously in aggregated embryo chimeric mice. Blood 69, 773 –777. Hayflick, L., 1994. How and Why We Age. Ballantine Books, New York. Hitt, R., Young-Xu, Y., Silver, M., Perls, T., 1999. Centenarians: the older you get, the healthier you have been. Lancet 354, 652. Hornung, R.L., Longo, D.L., 1992. Hematopoietic stem cell depletion by restorative growth factor regimens during repeated high-dose cyclophosphamide therapy. Blood 80, 77 – 83. Hsu, S.Y., Hsueh, A.J.W., 2000. Tissue-specific Bcl-2 protein partners in apoptosis: an ovarian paradigm. Physiol. Rev. 80, 593 –614. Iscove, N., Nawa, K., 1997. Hematopoietic stem cells expand during serial transplantation in vivo without apparent exhaustion. Curr. Biol. 7, 805 – 808. Jackson, K.A., Mi, T.J., Goodell, M.A., 1999. Hematopoietic potential of stem cells isolated from murine skeletal muscle. Proc. Natl. Acad. Sci. USA 96, 14482 – 14486. Jazwinski, S.M., 2000. Metabolic mechanisms of yeast ageing. Exp. Gerontol. 35, 671 – 676. Jordan, C., Lemischka, I., 1990. Clonal and systemic analysis of long-term hematopoiesis in the mouse. Genes Dev. 4, 220 –232. Kirkwood, T.B., 1977. Evolution of ageing. Nature 270, 301 – 304. Kirkwood, T.B., 2001. Sex and ageing. Exp. Gerontol. 36, 413 – 418. Kirkwood, T.B.L., Rose, M.A., 1991. Evolution of senescence: late survival sacrificed for reproduction. Phil. Trans. R. Soc. Lond. B 332, 15 – 24. Kirkwood, T.B., Austad, S.N., 2000. Why do we age? Nature 408, 233 – 238. Kopen, G.C., Prockop, D.J., Phinney, D.G., 1999. Marrow stromal cells migrate throughout forebrain and cerebellum and they differentiate into astrocytes after injection into neonatal mouse brains. Proc. Natl. Acad. Sci. USA 96, 10711 –10716. Kuhn, H.G., Dickinson-Anson, H., Gage, F.H., 1996. Neurogenesis in the dentate gyrus of the adult rat: age-related decrease of neuronal progenitor proliferation. J. Neurosci. 16, 2027 – 2033. Lagasse, E., Connors, H., Al-Dhalmy, M., Reitsma, M., Dohse, M., Osborne, L., Wang, X., Finegold, M., Weissman, I.L., Grompe, M., 2000. Purified hematopoietic stem cells can differentiate into hepatocytes in vivo. Nat. Med. 6, 1229 – 1234. Lakowski, B., Hekimi, S., 1998. The genetics of caloric restriction in Caenorhabditis elegans. Proc. Natl. Acad. Sci. USA 95, 13091 –13096. Lane, M.A., 2000. Nonhuman primate models in biogerontology. Exp. Gerontol. 35, 533 – 541. Lavker, R.M., Sun, T.-T., 2000. Epidermal stem cells: properties, markers, and location. Proc. Natl. Acad. Sci. USA 97, 13473 –13475. Lee, C.K., Weindruch, R., Prolla, T.A., 2000. Gene-expression profile of the ageing brain in mice. Nat. Genet. 25, 294 –297. Martin, G.M., Oshima, J., 2000. Lessons from human progeroid syndromes. Nature 408, 263 – 266. Mathon, N.F., Malcolm, D.S., Harrisingh, M.C., Cheng, L., Lloyd, A.C., 2001. Lack of replicative senescence in normal rodent glia. Science 291, 872 – 875. Mattson, M.P., 2000. Neuroprotective signaling and the aging brain: take away my food and let me run. Brain Res. 886, 47 –53. Medawar, P.B., 1952. An Unsolved Problem of Biology. Lewis, London. 1552 D. Schlessinger, G. Van Zant / Mechanisms of Ageing and De6elopment 122 (2001) 1537–1553 Mezey, E., Chandross, K.J., Harta, G., Maki, R.A., McKercher, S.R., 2000. Turning blood into brain: cells bearing neuronal antigens generated in vivo from bone marrow. Science 290, 1779 – 1782. Miller, R.A., 2001. New paradigms for research on aging and late-life illness. Mech. Ageing Dev. 122, 130–132. Mintz, B., Silvers, W.K., 1967. ‘Intrinsic’ immunological tolerance in allophenic mice. Science 158, 1484–1486. Mintz, B., Palm, J., 1969. Gene control of hematopoiesis. I. erythrocyte mosaicism and permanent immunological tolerance in allophenic mice. J. Exp. Med. 129, 1013 – 1027. Montaron, M.F., Petry, K.G., Rogdriguez, J.J., Marinelli, M., Aurousseau, C., Rougon, G., Le Moal, M., Abrous, D.N., 1999. Adrenalectomy increases neurogeneisis but not PSA-NCAM expression in aged dentate gyrus. Eur. J. Neurosci. 11, 1479 – 1485. Morita, Y., Tilly, J.L., 1999. Oocyte apoptosis: like sand through an hourglass. Dev. Biol. 213, 1 – 17. Morita, Y., Manganaro, T.F., Tao, X.-J., Martimbeau, S., Donahoe, P.K., Tilly, J.L., 1999a. Requirement for phosphatidylinositol-3%-kinase in cytokine-mediated germ cell survival during fetal oogenesis in the mouse. Endocrinology 140, 941 – 949. Morita, Y., Perez, G.I., Maravei, D.V., Tilly, K.I., Tilly, J.L., 1999b. Targeted expression of Bcl-2 in mouse oocytes inhibits ovarian follicle atresia and prevents spontaneous and chemotherapy-induced oocyte apoptosis in vitro. Mol. Endocrinol. 13, 841 – 850. Morrison, S.J., Wandycz, A.M., Akashi, K., Globerson, A., Weissman, I.L., 1996. The aging of hematopoietic stem cells. Nat. Med. 2, 1011 – 1016. Orkin, S.H., 2000. Diversification of haematopoietic stem cells to specific lineages. Nat. Rev. Genet. 1, 57–64. Perls, T.T., Alpert, L., Fretts, R.C., 1997. Middle-age mothers live longer. Nature 389, 133. Perls, T.T., Bubrick, E., Wger, C.G., Vijg, J., Kruglyak, L., 1998. Siblings of centenarians live longer. Lancet 351, 1550. Perls, T., Shea-Drinkwater, M., Bowen-Flynn, J., Ridge, S.B., Kang, S., Joyce, E., Daly, M., Brewster, S.J., Kunkel, L., Puca, A.A., 2000. Exceptional familial clustering for extreme longevity in humans. J. Am. Geriatr. Soc. 48, 1483 –1485. Romanov, S.R., Kozakiewicz, B.K., Holst, C.R., Stampfer, M.R., Haupt, L.M., Tisty, T.D., 2001. Normal human mammary epithelial cells spontaneously escape senescence and acquire genomic changes. Nature 409, 633 –637. Rose, M.R., 1991. Evolutionary Biology of Ageing. Oxford University Press, New York. Schlessinger, D., Ko, M.S., 1998. Developmental genomics and its relation to aging. Genomics 52, 113–118. Schlessinger, D., Herrera, L., Crisponi, L., Mumm, S. Percesepe, A., Pellegrini, M., Pilia, G., Forabosco, A., 2001. Genes and translocations involved in premature ovarian failure, Am. J. Med. Genet., in press. Schmeichel, K.L., Weaver, V.M., Bissell, M.J., 1998. Structural cues from the tissue microenvironment are essential determinants of the human mammary epithelial cell phenotype. J. Mammary Gland Biol. Neoplasia 3, 201 –213. Siminovitch, L., Till, J.E., McCulloch, E.A., 1964. Decline in colony-forming ability of marrow cells subjected to serial transplantation into irradiated mice. J. Cell Comp. Physiol. 64, 23 – 31. Simpson, J.L., Rajkovic, A., 1999. Ovarian differentiation and gonadal failure. Am. J. Med. Genet. 89, 186–200. Sohal, R.S., Weindruch, R., 1996. Oxidative stress, caloric restriction, and aging. Science 273, 59 – 63. Suda, Y., Suzuki, M., Ikawa, Y., Aizawa, S., 1987. Mouse embryonic stem cells exhibit indefinite proliferative potential. J. Cell Physiol. 133, 197 – 201. Tang, D.G., Tokumoto, Y.M., Apperly, J.A., Lloyed, A.C., Raff, M.C., 2001. Lack of replicative senescence in cultured rat oligodendrocyte precursor cells. Science 291, 868 – 871. Tibiletti, M.G., Dalpra, L., Crosignani, P.G., 2000. Premature ovarian failure. Mol. Cell. Endocrinol. 161, 53 – 57. Uchida, N., Buck, D.W., He, D., Reitsma, M.J., Masek, M., Phan, T.V., Tsukamoto, A.S., Gage, F.H., Weissman, I.L., 2000. Direct isolation of human central nervous system stem cells. Proc. Natl. Acad. Sci. USA 97, 14720 –14725. D. Schlessinger, G. Van Zant / Mechanisms of Ageing and De6elopment 122 (2001) 1537–1553 1553 Van Zant, G., Holland, B.P., Eldridge, P.W., Chen, J.-J., 1990. Genotype-restricted growth and aging patterns in hematopoietic stem cell populations of allophenic mice. J. Exp. Med. 171, 1547 – 1565. Van Zant, G., Scott-Micus, K., Thompson, B.P., Fleischman, R.A., Perkins, S., 1992. Stem cell quiescence/activation is reversible by serial transplantation and is independent of stromal cell genotype in mouse aggregation chimeras. Exp. Hematol. 20, 470 – 475. Wakayama, T., Rodriguez, I., Perry, A.C., Yanagimachi, R., Mombaerts, P., 1999. Mice cloned from embryonic stem cells. Proc. Natl. Acad. Sci. USA 96, 14984 – 14989. Wakayama, T., Shinkai, Y., Tamashiro, K.L.K., Niida, H., Blanchard, D.C., Blanchard, R.J., Ogura, A., Tanemura, K., Tachibana, M., Perry, A.C.F., 2000. Cloning of mice to six generations. Nature 407, 318 –319. Watt, F.M., Hogan, B.L.M., 2000. Out of Eden: stem cells and their niches. Science 287, 1427 – 1430. Weissman, I.L., 2000. Stem cells: units of development, units of regeneration, and units in evolution. Cell 100, 157 –168. Westendorp, R.G., Kirkwood, T.B., 1998. Human longevity at the cost of reproductive success. Nature 396, 743 –746. Williams, G.C., 1957. Pleiotropy, natural selection and the evolution of senescence. Evolution 11, 398–411. Wilmut, I., Schnieke, A.E., McWhir, J., Kind, A.J., Campbell, K.H.S., 1997. Viable offspring derived from fetal and adult mammalian cells. Nature 385, 810 – 813. Xie, T., Spradling, A.C., 2000. A niche maintaining germ cell line stem cells in the Drosophila ovary. Science 290, 328 –330. Zinn, A.R., Page, D.C., Fisher, E.M., 1993. Turner syndrome: the case of the missing sex chromosome. Trends Genet. 9, 90 – 93. .