الخلايا الجذعية للدكتور فارس العنزي

سبق نشر المقال قبل شهرين بمجله جامعه ييل الامريكيه للعلوم و الطب (وأعتذر لعدم أمكانيه ترجمتها لكثره مشاغلي
Abstract:
Self-renewal and apoptosis of haemopoietic stem cells (HSC) represents major factors that determine the size of the haemopoietic cell mass. Changes in self-renewal above or below the steady state value of 0.5 will result in either bone marrow expansion or aplasia. Despite the growing body of research that describes the potential role of the HSC, there is still a very little information on the mechanisms that govern the HSC self renewal and apoptosis. Considerable insight into the role of the HSC in many diseases has been gained in recent years. In the light of their crucial importance, this article reviews recent developments in the understanding of the molecular, biological and physiological characteristics of haemopoietic stem cells.
1. Stem cell: Background and properties:
The most primitive haemopoietic cells are the haemopoietic stem cells (HSC). They are defined as cells with a high potential for self-renewal, and possess the capacity for dividing into identical copies of themselves without forming any newly differentiated features. Because most mature blood cells have a very short life span, the importance of HSC in sustaining the life the mammal i.e., through its ability to self-renew is very critical. Stem cells in both embryonic and adult tissues are defined by their ability to undergo self renewal and differentiation in a balanced state without depleting the stem cell pool. If a progenitor can divide (and in certain circumstances can generate a secondary colony) it does not mean that it is capable of self-renewal. All myeloid progenitor cells, except HSC, will terminally differentiate within 2 months or sooner. In contrast, HSC are capable of maintaining haematopoiesis during the life of the animal or longer if transplanted. So, self-renewal implies immortality at least within a reasonably long period of time, even as far as lifespan. All other progenitors which eventually extinguish do not self-renew: all their daughters are of a progressively decreased quality (in terms of proliferative potential) and therefore cannot be considered as copies of the original cell. This topic is currently under huge debate.
HSC are capable of differentiating into at least eight cell lines. The balance between self-renewal and differentiation is considered to be critical to the maintenance of stem cell numbers (1-3). More importantly, stem cells are proposed to have a major role in curing many degenerative diseases and cancers (4-7). Significant efforts have been made in recent years in understanding the mechanism governing HSC generation, self renewal, proliferation and commitment. Yet understanding the overall process is still far from complete and largely hypothetical. A growing body of research suggests that pathways regulating the self-renewal of normal stem cells are dysregulated in cancer stem cells resulting in continuous expansion of self-renewal and tumour development, and therefore give hope that new cancer therapies may emerge via this approach (4-10). Most stem cells are in G0 phase of the cell cycle, and therefore only a small number of stem cells are responsible for stem cell maintenance and for producing mature cells at any specific time (3,11). Experimental work has indicated that a single primitive progenitor may survive in a quiescent state for more than two weeks in culture before it divides (1-2, 12-14).
Stem cells are characterized by the expression of CD34 and Thy1, and absence of CD38, CD33 and HLA-DR (15-17). These cells also lack of expression of a great number of markers that are expressed on mature blood cells (lineage negative), and that lineage negativity is as important as other criteria for identifying and isolating these cells. Human HSC are CD34+ while murine HSC are CD34-. CD34 is a transmembrane glycoprotein (mucin), expressed in immature haemopoietic cells, fibroblasts, vascular endothelium and high endothelial venules (HEV) (18-19). CD34 is a stem and progenitor cell marker in humans. It contains two sites for serine/threonine phosphorylation by protein kinase C (PKC) and a tyrosine phosphorylation site, implying a possible role in signalling (20). In addition, endothelial CD34 binds to the lectin-like adhesion molecule L-selectin (21). Surprisingly, however, experimental work on CD34 deficient mice has revealed no major abnormalities either in haemopoiesis or in interactions of haemopoietic progenitor cells with stromal cells (22). It is well established that even more primitive progenitor cells are present within the CD34 negative fraction (23). Additionally, haemopoietic progenitor cells express adhesion molecules such as L-selectin (24), integrins (25), and homing-associated cell adhesion molecule (H-CAM) (26). Alternative methods to isolate HSC, including dye efflux activity such as side population activity (SP) or rhodamine efflux, and also other new markers that have been emerged in recent years such as Slamf1 (CD150), or endoglin.
2. Committed progenitor cells: Background and properties:
This compartment includes stem cell progeny which have been identified by their ability to form colonies of morphologically recognizable cells in semi-solid cultures. In addition, progenitor cells are characterized phenotypically by expression of CD38, CD33 and HLA-DR and a greater proportion of them are in an active cell cycle compared with stem cells. It is generally assumed that progenitor cells can not self renew. However, considerable self-renewal has recently been demonstrated experimentally in vitro by replating CFU-GM into secondary cultures (27). This has now provided an informative clonogenic assay for demonstrating self renewal capacity (3). The size of the progenitor’s cell compartment is governed by the balance between the cell gain (self renewal) and the cell loss (apoptosis). Therefore, an imbalance in either of these parameters will result in an elevation or a fall in the progenitor cell mass. The colony-forming units granulocyte, erythroid, monocyte and megakaryocyte (CFU-GEMM) are intermediate between stem cells and single lineage precursors. They are capable of producing granulocytes, erythrocytes, monocytes and megakaryocytes. The more committed progenitor cells have only single lineage potential. These are colony-forming unit granulocyte (CFU-G), colony-forming unit macrophage (CFU-M), burst-forming unit erythroid (BFU-E) and colony-forming unit megakaryocyte (CFU-Meg) which produce granulocytes, macrophages, erythrocytes or megakaryocytes respectively (28-30).
3. Models of HSC self-renewal:
3.i Stochastic theory
Stochastic is a term applied to processes appear random but have associated underlying probability distribution. In the context of haemopoiesis, the fate of individual cells can not be predicted. An important feature of stochastic processes is that they are associated with an overall probability of an event. Therefore in haemopoiesis, there is a probability of self-renewal (pSR) and a probability of differentiation (pDiff). When cells divide the values of pSR and pDiff will be 0.5 for steady state haemopoiesis (31). There is often a misconception that stochastic processes are random and therefore that stem cells can not respond to environmental factors. It is sometimes assumed that stochastic processes are regulated intrinsically.
In 1961, Till and McCulloch developed an assay that involved inbred strains of mice, where the donor and recipient are genetically identical (32). The recipient mice are irradiated to ablate all haemopoietic cells. Bone marrow cells from donor mice are then injected into the recipient mice. After 10 days, the mice are sacrificed and the number of spleen colonies are counted. The number of colonies forming on the spleen of lethally irradiated mice correlated with the number of bone marrow (BM) cells transplanted, and different lineages were represented in the developing colonies. It is known from other works that not all CFU-S are capable of giving rise to secondary colonies. Other workers found that replating of erythroid colonies and blast cell colonies showed a similar distribution to that seen in CFU-S (33-34). More recently, Abkowitz and colleagues have shown that stochastic effects may occur in haemopoiesis in vivo (35).
3.ii Deterministic model of haemopoiesis:
This model suggests that decisions to self renew or differentiate are entirely controlled and that responses to a particular set of circumstances will be predetermined (12,36). Morrison and Weissmann suggested a model based on the purification of three multipotent progenitor cells which had different self-renewal capacities. The progenitors from the most primitive population showed a greater self-renewal capacity than those from less primitive progenitors. They concluded that self-renewal is a deterministic event based on the fact that cell separation could predict self-renewal potential (37). In the deterministic model, the haemopoiesis process is controlled by extrinsic factors such as cell-cell interactions within the haempoietic micro-environment or signalling by cytokines and therefore, the cell fate is determined. These obstacles have been reviewed by Metcalf and Enver, Heyworth and Dexter (38-39).
Gordon and Blackett have proposed that the stochastic model with observations that cells with different self-renewal capacities could be separated into subpopulations. They proposed that self-renewal was a stochastic event but the probability of this process could be modified by extrinsic factors (11).
4. Kinetic models of stem cell regulation:
Two theories have been proposed to explain the ability of stem cells to maintain lifelong haemopoiesis. The first suggests that the embryo has sufficient stem cells to maintain haemopoiesis throughout life. However, this concept fails to explain the ability of a bone marrow transplant (consisting of a small fraction of the total marrow) to restore haemopoiesis to normal levels after the recipient has been exposed to myeloablative therapy (40). The second theory is that a small number of stem cells are able to sustain lifelong haemopoiesis because they are capable of self-renewal when they divide. For steady state haemopoiesis, the probability of self-renewal must be 0.5 and the probability of differentiation and/or loss by apoptosis must also be 0.5 (13-14) (Figures 1,2). Models of stem cell division with a self-renewal probability equal to 0.5 can be seen in Figure 3a & 3b. Figure (3a) shows asymmetric division when a stem cell divides into one new stem cell and one differentiated cell. Figure (3b) shows symmetrical divisions where one stem cell divides into two stem cells and another divides into two differentiated cells. Both models can account for steady state haemopoiesis. However, only the model shown in Figure 3b can account for expansion or recovery of the stem cell pool following damage. As indicated in Figure 3c, this may be accomplished by increasing the proportions of stem cells which self-renew (i.e. increasing the probability of self-renewal) (14).
Several investigators have shown that the probability of stem/progenitor cell self renewal is not fixed. Metcalf demonstrated that G-CSF decreases the replating ability of the WEHI-3B colony-forming cells (41). Additionally, Lewis and colleagues have reported that cytokines modify the self-renewal kinetics of primary granulocytic and erythroid progenitor cells (42-44). As shown in figure 3b, loss by apoptosis, as well as by differentiation, from the stem cell population may contribute to the control of stem cell numbers. However, there is a paucity of information on this point. Despite the wide acceptance and being publicised repeatedly at scientific meetings, still there is a need to re-examine some of the basic concepts in haemopoiesis. Quesenberry and Lowry suggested previously the important need to bring current models of stem cell self-renewal and differentiation into line with experimental observations (45-46). This necessity is further emphasised by the discovery that haemopoitic stem cells may differentiate into non-haemopoitic lineages such as muscle and neural cells (47-48).
The concept of stem cell compartmentalisation is a cornerstone of HSC self renewal (49). There are various known factors that contribute to stem cell functionality, including second messenger systems, transcr iption factors and the type and number of growth factor receptors expressed. The interaction of these factors will determine the potential responses of those cells to both internal and external signals. Probability of self-renewal is decreased as HSC progress towards maturity. Ogawa and colleagues (50) pointed out that the heterogeneity of HSC and the inability to measure the self-renewal probability of an individual HSC remain among the most significant limitations. It is impossible to measure precisely the self-renewal capacity of an individual HSC at the same time as measuring its differentiation capacity. Therefore, the focus should not be on individual cells but perhaps rather on populations of stem cells by using techniques that have been developed by Jankovic et al (51). Whilst progenitor cells do not have the same self-renewal capacity as the parent stem cells, they retain some capacity for self-renewal. Therefore, the CFU-GM and BFU-E progenitor assays are very useful tools for studying haemopoiesis, and considerable evidence has been accumulated that this might be the case (52-53). Till and colleagues developed a technique to detect HSC and it is widely believed that it detects not only an early but also more mature progenitor cells (32). It has been demonstrated that replating colonies from murine blast cell colonies and committed progenitors such as CFU-GM and BFU-E results in secondary colony formation (27,33-34). Additionally, murine B cell progenitors can undergo self-renewal in the presence of stromal cells and interlukin-7 (IL-7) (ref 54).
5. Intrinsic mechanisms governing apoptosis of HSC
HSCs are defined by their ability both to self-renew and to give rise to any of the hematopoietic cell lineages throughout the lifetime of an animal (Nebel A 2006). To maintain hematopoietic homeostasis in a host, the HSC numbers need to be precisely regulated. The fate decisions, including life and death and self-renewal and differentiation, of HSCs are important processes that regulate the numbers and lifespan of the HSC pool in a host. Defects in these processes can contribute to hematopoietic insufficiencies and the development of hematopoietic malignancies (55). Failure of HSCs to self-renew during aging is believed to depend on several intrinsic (cell-autonomous) and extrinsic (non-cell-autonomous) factors. Products of numerous genes that are involved in either DNA-damage responses or longevity-related signaling contribute to the maintenance of the HSC self-renewal capacity. Several intrinsic factors have been identified as potential mechanisms underlying the self renewal of HSC. These factors include telomerase activity that control telomere length, intracellular pool of reactive oxygen species (ROS), Flt3, Foxo3a, BMI1, Zfx, and Notch.
Cell cycle checkpoints induced by telomere dysfunction represent one of the major in vivo tumor suppressor mechanisms provoking age dependent decline in self-renewal and regeneration of tissues and organs. Since stem cells are continuously proliferating throughout lifetime, most stem cell compartments express telomerase. In stem cells, however, the level of telomerase activity is not sufficient to maintain telomere length during aging. Exhaustion of replicative potential of telomerase appears to be at least partially dependent on the cell cycle regulatory component of the DNA damage response. To overcome its telomere activity, stem cells appear to have tighter DNA damage checkpoint control in comparison to somatic cells. These enhanced checkpoint responses may have a detrimental impact on stem cell function, by causing increased sensitivity towards senescence or apoptosis induced by telomere shortening (56). Stresses on stem and progenitor cell pools, in the form of telomere shortening or other genome maintenance failures have been shown to lower tissue renewal capacity and accelerate the appearance of senescence. Therefore, long-term stem and progenitor cell potential depends on both the genome maintenance mechanisms that counter DNA damage and the cell cycle checkpoint responses to damage (57-58).
It has been demonstrated that long-term self-renewing HSCs normally possess low levels of intracellular ROS. If this intracellular ROS levels become excessive, under pathological conditions, they cause senescence or apoptosis of stem cells and a failure of their self-renewal. Correction of the intracellular levels of ROS in HSC by treatment with an antioxidant that scavenges ROS can rescue HSC self-renewal (59). Defining the molecular mechanisms that govern the ROS regulation and strategies that can control the excess levels of ROS could lead to the significant improvement in designing novel therapeutics approaches for hematopoietic diseases, regenerative medicine, and the prevention of leukemia.
Flt3 is a member of the receptor tyrosine kinase family, which plays a critical role in maintenance of hematopoietic homeostasis; Recently, it has been found that the human HSC in both the bone marrow and the cord blood that are capable of long-term reconstitution in xenogeneic hosts uniformly express Flt3. Detailed analysis of Flt3 expression show that it is expressed not only in early lymphoid progenitors, but also in progenitors continuously along the granulocyte/macrophage pathway, including the common myeloid progenitor and the granulocyte/macrophage progenitor (60). These studies showed further that intact Flt3 signaling pathway has been found to be required for prevention of HSC stem and progenitors from spontaneous apoptotic cell death. This role of Flt3 has been suggested, at least in part, to the up-regulating of Mcl-1, which is an indispensable survival factor for hematopoiesis.
Foxo3a is a forkhead transcr iption factor that acts downstream of the PTEN/PI3K/Akt pathway. It has been found in mice deficient in Foxo3a that: (1) the frequency of HSC is less than normal; (2) the number of colony-forming cells is lowered and the ability of HSC to support long-term reconstitution of hematopoiesis is impaired; (3) HSC showed increased phosphorylation of p38MAPK, an upregulation of ROS; (4) defective maintenance of quiescence; and (5) increased sensitivity to cell-cycle-specific myelotoxic injury (61). Taken together, these results demonstrate that Foxo3a is an important intrinsic factor role for maintaining the HSC pool.
Cdk2 is a major regulator of S phase entry, is activated by mitogenic cytokines, and has been suggested to be involved in antigen-induced apoptosis of T lymphocytes. Analysis of the HSC compartment in mice deficient in Cdk2 revealed normal proportions of stem cells and progenitors. Furthermore, a competitive graft experiment on HSC deficient in Cdk2 showed normal renewal and multilineage differentiation (62), indicating that Cdk2 is not required for proliferation and differentiation of HSC in vivo. In vitro analyses, however, consider Cdk2 to be a major player in proliferation and apoptosis in HSCs. Therefore, further studies are required to analyze the contribution of this factor and a potential target for therapy.
Recent studies have shown that deletion of the transcr iption factor Zfx in murine HSC and embryonic stem cells (ESC) impairs their self-renewal, resulting in increased apoptosis and upregulation of stress-inducible genes. By contrast, Zfx directly activated common target genes, including ESC self-renewal regulators Tbx3 and Tcl1, in ESC and HSC (63). These studies identify Zfx as another factor that is critical for the self-renewal in embryonic and adult stem cells.
Notch signaling regulates diverse cell fate decisions during development and is reported to promote murine HSC self-renewal. Recent in vitro studies have shown that constitutive expression of active human Notch 1 intracellular domain in human blood CD34+ cells induced a reduction in the proliferation the number of CD34+ cell populations, coinciding with inhibited cell cycle kinetics and upregulation of p21 mRNA expression and induced apoptosis (64). The results of this study show that activation of the Notch signaling pathway play an important role in regulation of the proliferation and survival of adult stem cells.
6. Apoptosis in Haemopoiesis:
6.i Evidence for apoptosis of progenitor cells in vivo:
A possible role for apoptosis is illustrated by the relationship between erythropoietin (Epo) levels and erythropoietic activity in vivo in mice. When the Epo concentration is elevated due to an increase in demand (such as anaemia) many Epo-dependent-progenitor cells that would otherwise rapidly undergo apoptosis will survive. If Epo levels are decreased by hyper-transfusion, Epo-dependent progenitor cells that would normally survive will die rapidly and erythrocyte production is reduced (65).This hypothesis is supported by results using hyper-transfused mice which shows continual production of erythroid progenitor cells, but with no increase in their number following the cessation of erythropoiesis, and thus indicating the direct involvement of cell death (66).
6.ii Evidence for apoptosis of progenitor cells in vitro:
A group led by Dexter demonstrated that both GM-CSF dependent and G-CSF dependent cell lines undergo rapid cell death after removal of the relevant CSF and that this was accompanied by a typical pattern of DNA fragmentation (67). Peschle’s group has demonstrated the possible involvement of Fas and Fas ligand (FasL) in the regulation of erythropoiesis, and immunohistochemistry of normal bone marrow (NBM) samples and determined that several immature erythroblasts undergo apoptosis in vivo. These results showed that erythroid blast express Fas, whereas, more mature cells express FasL. These findings thus suggest the existence of a negative regulatory feedback between mature and immature erythroid cells. Accordingly, the interaction of Fas and FasL might represent an apoptotic control mechanism for erythropoiesis, contributing to the regulation of red blood cell homeostasis (68). Furthermore, we have shown that Fas, FasL, and caspase activation are likely to play an important role in the regulation of myelopoiesis (69).
7. Relationship between Apoptosis and self renewal (i.e., proliferation):
Evidence has accumulated concerning the participation of certain proliferation promoting factors (such as oncogenes) that are also able to act as potent triggers of apoptosis. Recent experimental evidence has also suggested that there may be a positive relationship between apoptosis and proliferation in normal haemopoiesis. For example, Traycoff et al (70) showed that cord blood, and NBM CD34+ cells with a high number of cell divisions short term cultures in vitro, are associated with an increase in the percentage of apoptotic CD34+cells. On the other hand, models proposed by Koury (71) imply that a reduction in apoptosis might result in an increase in proliferation, potentially resulting in an inverse relationship between cell death and cell division. Evidence for a link between apoptosis and proliferation (i.e., self-renewal) in haemopoiesis remains unclear; we hypothesize that cell proliferation (i.e., self-renewal) and cell apoptosis are linked in normal haemopoietic tissues and that this relationship may be abnormal in chronic myeloid leukaemia (CML).
We investigated the role of p21 in normal HSC from p21 knockout mice. We found that p21 deletion increases the growth rate of colonies and the multiplication of haemopoietic progenitor cells in vitro. We also found also an increase in apoptotic percentage on primary hemopoietic cells from mice (unpublished observations). Therefore, in order for a transformed haemopietic cell to launch a clone and result in leukaemia, it is of importance for some defect to provide the cell with an increased probability of self-renewal. This is because a self-renewal probability greater than 0.5 will not result in clonal expansion, rather it will attain maintenance of cell number (SR=0.5) or result in clonal loss (SR<0.5). These preliminary unpublished results suggest that p21 has the potential to act as a tumour suppressor gene in the myeloid lineage. Additionally, it is now confirmed that p21 is attributed with a role in regulating self renewal (i.e. proliferation) and apoptosis.
Alenzi and colleagues (72) showed greater frequencies of myeloid progenitor cells (CFU-GM) in lpr and gld mice BM (Fas and FasL knockout mice respectively) compared to wild-type (WT) mice marrow (p=0.0008). The self-renewal (i.e., proliferation capacity) was also significantly greater for lpr and gld CFU-GM compared to WT CFU-GM. Retroviral-mediated gene transfer (RMGT) of Fas (apoptotic gene) into lpr marrow reduced CFU-GM self renewal (proliferative capacity) to WT levels. Therefore, Fas is likely to play an important role in regulating myeloid progenitor cell self renewal. This raises the possibility that Fas/FasL are linked to self renewal (i.e., proliferation capacity) in mice. Therefore, our results suggest that the Fas/FasL pathway and caspase activation inhibit progenitor cell proliferation and promote differentiation. They support proposals that caspase activation may have non-apoptotic functions in the regulation of haemopoiesis (73-74). Thus, it can be inferred that the presence of fully functional genes that regulate both cell proliferation (i.e., self-renewal) and apoptosis, will maintain the balance between the rate of cell division and apoptosis of any (cell) population in vivo. Therefore, malfunction in, or loss of, any of these genes (or inappropriate DNA or RA splicing) may lead to an increase in their self-replication (i.e., self renewal).
We investigated whether changes in the level of apoptosis due to Fas/FasL deficiencies cause changes in the number and kinetics of HSC. For this, mice with mutations in Fas or FasL were used to investigate how this change may affect the self renewal of clonogenic haemopoietic cells. It has been shown previously that haemopoietic activity is increased in lpr mice which have Fas mutations (75), but this was done only at the level of the number of CFU-GM and not in terms of their proliferative activity. The results obtained differ from those of Schneider et al (76), who did not show a significant increase in the CFU-GM frequency in the bone marrow of lpr mice, although they found no difference in BM cellularity. Instead, they found increased levels of extramedullary haemopoiesis, especially in the spleen (which is a site of haemopoiesis in normal mice). Our results are in line with those of Traver et al (77) who showed that lpr marrow contains a significantly greater number of myeloid progenitor cells compared to WT mice.
Our results concerning the self-renewal (i.e., proliferation) by NBM progenitors are in line with Marley and colleagues (78) showed that the self renewal values of CML progenitors are significantly greater than those of normal marrow (p=0.0001). There was a relatively low frequency of apoptotic CD34+ cells in progenitors grown from NBM with a significant difference between the NBM and CML samples. We have accumulated a larger number of CML samples and found a significantly increased level of self renewal (i.e., proliferation capacity) in CML compared to NBM (n=100, p=0.01). There was an evidence of a difference in the self renewal (i.e., proliferation capacity) between progenitors grown from NBM or CML samples. The frequency of apoptotic cells in CML progenitors was dramatically increased compared to normal samples which agree with the fact that apoptosis is reduced in CML. These data are consistent with those of Thiele et al (79-85) and others who found a considerable level of apoptosis in CD34+ cells harvested from CML patients. The correlation between apoptosis and proliferation was fairly strongly positive (unpublished observations).
Domen et al produced Bcl-2 transgenic mice that overexpressed Bcl-2. They found that the HSC from WT mice died after growth factor withdrawal whereas HSC from Bcl-2 transgenic mice remained viable. More importantly, HSC from Bcl-2 transgenic mice proliferated more rapidly and extensively (in the presence of a cocktail of factors including IL-1, IL-3, IL-6, SCF, Flt3L) than those of WT. Additionally, there was a delay in cell cycle entry. The most dramatic difference between WT and Bcl-2 transgenic mice was revealed when HSCs were cultured in the presence of SCF. Only 20% of WT HSC remained viable after one week, whereas, HSC from Bcl-2 transgenic mice showed enhanced survival and more vigorous proliferation. Bcl-2 over-expression and SCF/c-kit signalling was found to be sufficient for HSC proliferation although, it should be noted that proliferation also resulted in differentiation of myeloid progenitor cells (86-87).
8. Non-apoptotic role of Fas
Although several in vivo and in vitro studies have demonstrated variable sensitivities of hematopoietic precursors to Fas-mediated apoptosis, HSC, at least in murine model, that induced to express Fas after treatment with TNF-a showed reduction in their engraftment potential (88). The induced Fas expression also decreased the self-renewal of highly purified progenitors (88). Furthermore, murine HSC with best hematopoietic reconstituting potential have been found to be express Fas, where ~50% of the colony-forming cells in spleen-derived lineage-negative (lin–) progenitors were resistant to Fas ligation (88). These data suggest that Fas expression in HSC is not a negative factor in HSC apoptosis. Recent experiments uggest, however, that Fas can improve the engraftment of HSC. With this regard, it was found that hematopoietic progenitors that homed successfully to the BM showed a marked upregulation in the expression of the Fas receptor and Fas ligand (FasL), and were more resistant to induction of apoptosis (89-90). It was found also in a recent study that Fas is capable of transducing growth signals in hematopoietic progenitors, after trimerization of this receptor (91). Therefore identification of factors that can upregulate Fas expression in HSC can be of particular importance to HSC transplantation. Inflammatory cytokines, in particular TNF-a and IFN-g induced in response to environmental stress factors, are a potent inducer of Fas expression on the HSC and haematopietic progenitors (92-95). The acquisition of HSC to apoptotic signals upon Fas upregulation can help sustain viability of progenitors under stress conditions in case of allogeneic hematopoietic grafts into myeloablative recipients. Ectopic expression of FasL in HSC also showed better survival. For example, it has been reported that FasL-modified lin(-) BM can kill Fas-expressing T cells in vitro and that transplanting of allogeneic FasL(+) lin(-) BM into recipient mice treated with nonmyeloablative conditioning regimen resulted in an enhanced short-term engraftment (96).
Conclusions:
Cell population size is determined by a balance between cell loss (apoptosis and differentiation) and cell gain (proliferation and mitosis). In normal haemopoiesis, these factors are controlled so that steady-state kinetics are preserved. In contrast, in myeloid leukemias, myeloid expansion can be explained by an increase in proliferation (self-renewal), and reduced apoptosis. There have been huge recent advances in research into stem cells. We are currently in a phase where these developments may be used precisely towards the successful attainment of the use of “intelligent” therapeutics for many diseases. Further studies are needed to complete understanding of how these two pathways are regulated in vivo and in vitro.
References:
1. McCulloch EA. Stem cells in normal and leukemic hemopoiesis. Blood. 1983; 62: 1-13.
2. Ploemacher R. Stem cells: characterization and measurement. Baillieres Clin Haematol. 1997; 10: 429-44.
3. Gordon MY. Human haemopoietic stem cell assays. Blood Rev. 1993; 7: 190-7.
4. Al-Hajj M, Becker MW, Wicha M, Weissman I, Clarke MF. Therapeutic implications of cancer stem cells. Curr Opin Genet Dev. 2004;14:43-7.
5. Crowe DL, Parsa B, Sinha UK. Relationships between stem cells and cancer stem cells. Histol Histopathol. 2004;19:505-9.
6. Sikora MA, Olszewski WL. Stem cells-biology and therapeutic application. Postepy Hig Med Dosw. 2004;58:202-8.
7. Mayhall EA, Paffett-Lugassy N, Zon LI. The clinical potential of stem cells. Curr Opin Cell Biol. 2004;16:713-20.
8. Al-Hajj M, Clarke MF.Self-renewal and solid tumor stem cells.
Oncogene. 2004;23:7274-82.
9. Dontu G, Al-Hajj M, Abdallah WM, Clarke MF, Wicha MS. Stem cells in normal breast development and breast cancer. Cell Prolif. 2003; 36:59-72.
10. Galderisi U, Cipollaro M, Giordano A. Stem cells and brain cancer.
Cell Death Differ. 2005 Aug 26; .
11. Gordon MY, Blackett NM. Routes to repopulation-a unification of the stochastic model and separation of stem-cell subpopulations. Leukemia. 1994; 8: 1068-72.
12. Novak JP, Stewart CC. Stochastic versus deterministic in haemopoiesis: what is what? Br J Haematol. 1991; 78: 149-54.
13. Ogawa M. Differentiation and proliferation of hematopoietic stem cells. Blood. 1993; 81: 2844-53.
14. Morrison SJ, Shah NM, Anderson DJ. Regulatory mechanisms in stem cell biology. Cell. 1997; 88: 287-98.
15. Baum CM, Weissman IL, Tsukamoto AS, Buckle AM, Peault B. Isolation of a candidate human hematopoietic stem-cell population. Proc Natl Acad Sci USA. 1992; 87: 2804-8.
16. Craig W, Kay R, Cutler RL, Lansdorp PM. Expression of Thy-1 on human hematopoietic progenitor cells J Exp Med. 1993; 177: 1331-42.
17. Sutherland HJ, Eaves CJ, Eaves AC, Dragowska W, Lansdorp PM. Characterization and partial purification of human marraw cells capable of initiating long term hematopoiesis in vitro. Blood.1989;74:1563.
18. Fina L, Molgaard HV, Robertson D, Bradley NJ, Monaghan P, Delia D, Sutherland DR, Baker MA, Greaves M. Expression of the CD34 gene in vascular endothelial cells. Blood. 1990; 75: 2417-26.
19. Krause DS, Fackler MJ, Civin CI, May WS. CD34: structure, biology, and clinical utility. Blood. 1996; 87: 1-13
20. Fackler MJ, Krause DS, Smith OM, Civin CI, May WS. Full-length but not truncated CD34 inhibits hematopoietic cell differentiation of M1 cells. Blood. 1995; 85: 3040-7.
21. Baumhueter S, Dybdal N, Kyle C, Lasky LA. Global vascular expression of murine CD34, a sialomucin-like endothelial ligand for L-selectin. Blood. 1994; 8: 2554-65.
22. Suzuki A, Andrew DP, Gonzalo JA, Fukumoto M, Spellberg J, Hashiyama M, Takimoto H, Gerwin N, Webb I, Molineux G, Amakawa R, Tada Y, Wakeham A, Brown J, McNiece I, Ley K, Butcher EC, Suda T, Gutierrez-Ramos JC, Mak TW. CD34-deficient mice have reduced eosinophil accumulation after allergen exposure and show a novel crossreactive 90-kD protein. Blood. 1996; 87: 3550-62.
23. Osawa M, Hanada K, Hamada H, Nakauchi H. Long-term lympho-hematopoietic reconstitution by a single CD34-low/negative hematopoietic stem cell. Science. 1996; 273: 242-5.
24. Sackstein R. Expression of an L-selectin ligand on hematopoietic progenitor cells. Acta-Haematol. 1997; 97: 22-8.
25. Coulombel L, Auffray I, Gaugler MH, Rosemblatt M. Expression and function of integrins on hematopoietic progenitor cells. Acta Haematol. 1997; 97: 13-21.
26. Deguchi T, Komada Y, Sugiyama K, Zhang XL, Azuma E, Yamamoto H, Sakurai M. Expression of homing-associated cell adhesion molecule (H-CAM/CD44) on human CD34+ hematopoietic progenitor cells. Exp Hematol. 1999; 27: 542-52.
27. Gordon MY, Marley SB, Lewis JL, Davidson RJ, Nguyen DX, Grand FH, Amos TA, Goldman JM. Treatment with interferon-alpha preferentially reduces the capacity for amplification of granulocyte-macrophage progenitors (CFU-GM) from patients with chronic myeloid leukemia but spares normal CFU-GM. J Clin Invest.1998; 102: 710-5.
28. Metcalf D. Control of granulocytes and macrophages: molecular, cellular, and clinical aspects. Science.1991; 254: 529-33.
29. Eaves AC, Eaves CJ. Erythropoiesis in culture. Clin Haematol.1984; 13: 371-91
30. Hoffman R. Regulation of megakaryocytopoiesis. Blood 74: 1196, 1989.
31. Gordon MY; Amos TA. Stochastic effects in hemopoiesis. Stem Cells. 1994; 12: 175-9.
32. Till JE and McCulloch EA: A direct measurement of the radiation sensitivity of normal mouse bone marrow cells. Radiation Research.1961:14,213.
33. Humphries RK, Eaves AC, Eaves CJ. Self-renewal of hemopoietic stem cells during mixed colony formation in vitro. Proc Natl Acad Sci USA. 1981; 78: 3629-33.
34. Nakahata T, Ogawa M. Identification in culture of a class of hemopoietic colony-forming units with extensive capability to self-renew and generate multipotential hemopoietic colonies. Proc Natl Acad Sci USA. 1982; 79: 3843-7.
35. Abkowitz JL, Catlin SN, Guttorp P. Evidence that hematopoiesis may be a stochastic process in vivo. Nat Med. 1996; 2: 190-7.
36. Nicola NA, Johnson GR. The production of committed hemopoietic colony-forming cells from multipotential precursor cells in vitro. Blood. 1982; 60: 1019-29.
37. Morrison SJ, Weissman IL. The long term repopulating subset of hematopoietic stem cells is deterministic and isolatable by phenotype. Immunity. 1994; 1: 661-73.
38. Metcalf D. Lineage commitment and maturation in hematopoietic cells: the case for extrinsic regulation. Blood. 1998; 92: 345-7.
39. Enver T, Heyworth CM, Dexter TM. Do stem cells play dice?. Blood. 1998 ; 92: 348-51.
40. Siminoyitch L, McCulloch A, Till JE. The identification of colony-forming cells among spleen colonies. J Cell Comp Physiol. 1963;62:327.
41. Metcalf D. Regulator-induced suppression of myelomonocytic leukemic cells: clonal analysis of early cellular events. Int J Cancer. 1982; 30: 203-10.
42. Humphries RK, Eaves AC, Eaves CJ. Self-renewal of hemopoietic stem cell during mixed colony formation in vitro. Proc Natl Acad Sci USA.1981;78:3629-33.
43. Lewis JL, Marley S, Davidson RJ, Nguyen DX. Chinswangwatanakul W, Goldman JM, and Gordon MY. Protein tyrosine kinase receptor signalling decreases progenitor cell self replication. Br J Haematol. 1998;102:23.
44. Lewis JL, Marley SB, Blackett NM, Szydlo R, Goldman JM, Gordon MY. Interleukin 3 (IL-3), but not stem cell factor (SCF) increases self-renewal by human erythroid burst-forming units (BFU-E) in vitro. Cytokine. 1998; 10: 49-54
45. Quesenberry PJ. The blueness of stem cells. Exp Hematol. 1991; 19: 725-8.
46. Lowry PA. Botanical musings. Exp Hematol. 1992; 20: 540.
47. Eglitis MA, Mezey E. Hematopoietic cells differentiate into both microglia and macroglia in the brains of adult mice. Proc Natl Acad Sci USA. 1997; 94: 4080-5
48. Ferrari G, Cusella De Angelis G, Coletta M, Paolucci E, Stornaiuolo A, Cossu G, Mavilio F. Muscle regeneration by bone marrow-derived myogenic progenitors Science. 1998; 279: 1528-30.
49. Till JE, McCulloch EA, and Siminovitch L. A stochastic model of stem cell proliferation, based on the growth of spleen colony-forming units. Proc Natl Acad Sci USA.1964, 51,29.
50. Suda J, Suda T and Ogawa M. Analysis of differentiation of mouse hemopoietic stem cell in cultures by sequential replating of paired progenitors. Blood.1983, 64, 393.
51. Jankovic GM, Colovic MO, Petrovic MO, Rolovic ZN, Janosevic S. The stochastic blueness of leukemic stem cells in experimental antineoplastic therapy. Exp Hematol. 1993; 21: 1399-400.
52. Lewis JL, Blackett NM, Gordon MY. The kinetics of colony formation by CFU-GM in vitro. Br J Haematol. 1994; 88: 440-2.
53. Marley SB, Amos TA, Gordon MY. Kinetics of colony formation by BFU-E grown under different culture conditions in vitro. Br J Haematol. 1996; 92: 559-61.
54. Rolink A, Kudo A, Karasuyama H, Kikuchi Y, Melchers F. Long-term proliferating early pre B cell lines and clones with the potential to develop to surface Ig-positive, mitogen reactive B cells in vitro and in vivo. EMBO J. 1991; 10: 327-36.
55. Oguro H, Iwama A. Life and death in hematopoietic stem cells. Curr Opin Immunol. 2007 Oct;19(5):503-9.
56. Ju Z, Rudolph KL. Telomeres and Telomerase in Stem Cells during Aging and Disease. Genome Dyn. 2006;1:84-103.
57. Ruzankina Y, Asare A, Brown EJ. Replicative stress, stem cells and aging. Mech Ageing Dev. 2008 Jul-Aug;129(7-8):460-6.
58. Krishnamurthy J, Sharpless NE. Stem cells and the rate of living. Cell Stem Cell. 2007 Jun 7;1(1):9-11.
59. Naka K, Muraguchi T, Hoshii T, Hirao A. Regulation of reactive oxygen species and genomic stability in hematopoietic stem cells. Antioxid Redox Signal. 2008 Nov;10(11):1883-94
60. Yoshikane Kikushige, Goichi Yoshimoto , Toshihiro Miyamoto, Tadafumi Iino, Yasuo Mori, Hiromi Iwasaki Hiroaki Niiro Katsuto Takenaka*, Koji Nagafuji*, Mine Harada*, Fumihiko Ishikawa and Koichi Akashi: Human Flt3 is expressed at the hematopoietic stem cell and the granulocyte/macrophage progenitor stages to maintain cell survival. J Immunology 2008 Jun 1;180(11):7358-67
61. Miyamoto K, Miyamoto T, Kato R, Yoshimura A, Motoyama N, Suda T. FoxO3a regulates hematopoietic homeostasis through a negative feedback pathway in conditions of stress or aging. Blood. 2008 Sep 17.
62. Berthet C, Rodriguez-Galan MC, Hodge DL, Gooya J, Pascal V, Young HA, Keller J, Bosselut R, Kaldis P. Hematopoiesis and thymic apoptosis are not affected by the loss of Cdk2. Mol Cell Biol. 2007 Jul;27(14):5079-89.
63. Galan-Caridad JM, Harel S, Arenzana TL, Hou ZE, Doetsch FK, Mirny LA, Reizis B. Zfx controls the self-renewal of embryonic and hematopoietic stem cells. Cell. 2007 Apr 20;129(2):345-57.
64. Chadwick N, Nostro MC, Baron M, Mottram R, Brady G, Buckle AM. Notch signaling induces apoptosis in primary human CD34+ hematopoietic progenitor cells. Stem Cells. 2007 Jan;25(1):203-10
65. Koury MJ. Programmed cell death (apoptosis) in hematopoiesis. Exp Hematol. 1992; 20: 391-4.
66. Testa N. Erythroid progenitor cells: their relevance for the study of haematological disease. Clin Hematol. 1979; 8,311-33.
67. Williams GT, Smith CA, Spooncer E, Dexter TM, and Taylor DR. Haemopoietic colony stimulating factors promote cell survival by suppressing apoptosis. Nature.1990;343:76-9.
68. De Maria R, Testa U, Luchetti L, Zeuner A, Stassi G, Pelosi E, Riccioni R, Felli,-N, Samoggia P, Peschle C. Apoptotic role of Fas/Fas ligand system in the regulation of erythropoiesis. Blood. 1999; 93: 796-803.
69. Alenzi FQB, Tamimi W, Al-Sebiany A, El-Nashar E, El-Tounsi E, Bamaga MS, Al-Enazi M, Al-Amri A, Al-Sheikh I. Apoptosis role of Fas/Fas Ligand System in the Regulation of Myelopoesis. Yale Journal of Biology and Medicine (2005)
70. Traycoff CM, Orazi A, Ladd AC, Rice S, McMahel J, Srour EF. Proliferation-induced decline of primitive hematopoietic progenitor cell activity is coupled with an increase in apoptosis of ex vivo expanded CD34+ cells. Exp Hematol.1998; 26: 53-62.
71. Koury MJ. Programmed cell death (apoptosis) in hematopoiesis. Exp Hematol.1992; 20: 391-4.
72. Alenzi FQB, Marley SB, Chandrashekran A, Warrens A, Goldman JM and Gordon MY. Regulation of haemopoietic progenitor cell number by the Fas/FasL apoptotic mechanism. Exp Hematol.2002;30: 1428-33.
73. De Maria R, Testa U, Luchetti L, Zeuner A, Stassi G, Pelosi E, Riccioni R, Felli N, Samoggia P, Peschle C. Apoptotic role of Fas/Fas ligand system in the regulation of erythropoiesis. Blood.1999; 93: 796-803.
74. De Maria R, Zeuner A, Eramo A, Domenichelli C, Bonci D, Grignani F, Srinivasula SM, Alnemri ES, Testa U, Peschle C. Negative regulation of erythropoiesis by caspase-mediated cleavage of GATA-1. Nature.1999; 401:489-93.
75. Schneider E, Moreau G, Arnould A, Vasseur F, Khodabaccus N, Dy M, Ezine S. Increased fetal and extramedullary hematopoiesis in Fas-deficient C57BL/6-lpr/lpr mice. Blood.1999; 94: 2613-21.
76. Yin C, Knudson CM, Korsmeyer SJ, Van Dyke T.Bax suppresses tumorigenesis and stimulates apoptosis in vivo. Nature.1997; 385:637-40.
77. Traver D, Akashi K, Weissman IL, Lagasse E. Mice defective in two apoptosis pathways in the myeloid lineage develop acute myeloblastic leukemia. Immunity. 1998; 9: 47-57.
78. Marley SB, Lewis JL, Zheng B, Davidson RJ, Alenzi FQB, Goldman JM Gordon MY. Peripheral blood progenitor cell mobilisation alters myeloid, but not erythroid progenitor cell self-renewal kinetics. Bone Marrow Transplant.2001; 27: 241-8.
79. Thiele J, Zirbes T, Lorenzen J, Kvasnicka HM, Dresbach S, Manich B, Leder LD, Niederle N, Diehl V, Fischer R. Apoptosis and proliferation (PCNA labelling) in CML, a comparative immunohistological study on bone marrow biopsies following interferon and busulfan therapy. J Pathol.1997; 181: 316-22.
80. Selleri C, Maciejewski JP, Pane F, Luciano L, Raiola AM, Mostarda I, Salvatore F, Rotoli B. Fas-mediated modulation of BCR-ABL in chronic myelogenous leukemia results in differential effects on apoptosis. Blood.1998; 92: 981-9.
81. Hariharan IK, Adams JM, Cory S. BCR-ABL oncogene renders myeloid cell line factor independent: potential autocrine mechanism in chronic myeloid leukemia. Oncogene Res.1988; 3: 387-99.
82. Mandanas RA, Boswell HS, Lu L, Leibowitz D. BCR-ABL confers growth factor independence upon a murine myeloid cell line. Leukemia.1992; 6: 796-800.
83. Sirard C, Laneuville P, Dick J. Expression of bcr-abl abrogates factor-dependent growth of human hematopoietic M07E cells by an autocrine mechanism. Blood 1994; 83: 1575-85.
84. Cortez D, Kadlec L, Pendergast AM. Structural and signaling requirements for BCR-ABL-mediated transformation and inhibition of apoptosis. Mol Cell Biol 1995; 15: 5531-41.
85. Daley GQ and Baltimore D. Transfection of an interleukin 3-dependent hematopoietic cell line by the chronic myelogenous leukemia-specific P210 protein. Proc Natl Acad Sci USA.1988; 85: 9312-6.
86. Domen J, Weissman IL. Hematopoietic stem cells need two signals to prevent apoptosis; BCL-2 can provide one of these, Kitl/c-Kit signaling the other. J Exp Med.2000; 192:1707-18
87. Domen J, Cheshier SH, Weissman IL.The role of apoptosis in the regulation of hematopoietic stem cells: Overexpression of Bcl-2 increases both their number and repopulation potential. J Exp Med. 2000; 191:253-64
88. Bryder D, Ramsfjell V, Dybedal I et al. Self-renewal of multipotent long-term repopulating hematopoietic stem cells is negatively regulated by Fas and tumor necrosis factor receptor activation. J Exp Med 2001;194:941–952
89. Pearl-Yafe M, Yolcu ES, Stein J et al. Expression of Fas and Fas-ligand in donor hematopoietic stem and progenitor cells is dissociated from the sensitivity to apoptosis. Exp Hematol 2007;35:1601–161
90. Pearl-Yafe M, Yolcu ES, Stein J et al. Fas ligand enhances hematopoietic cell engraftment through abrogation of alloimmune responses and nonimmunogenic interactions. STEM CELLS 2007;25:1448–1455
91. Michal Pearl-Yafe, Jerry Steina, Esma S. Yolcu, Daniel L. Farkas, Haval Shirwan, Isaac Yaniv, Nadir Askenasy Fas transduces dual apoptotic and trophic signals in hematopoietic progenitors. Stem Cells. 2007 Dec;25(12):3194-203.
92. Jacobsen SE, Ruscetti FW, Dubois CM et al. Tumor necrosis factor alpha directly and indirectly regulates hematopoietic progenitor cell proliferation: Role of colony-stimulating factor receptor modulation. J Exp Med 1992;175:1759–1772.
93. Maciejewski J, Selleri C, Anderson S et al. Fas antigen expression on CD34+ human marrow cells is induced by interferon gamma and tumor necrosis factor alpha and potentiates cytokine-mediated hematopoietic suppression in vitro. Blood 1995;85:3183–3190.
94. Selleri C, Sato T, Anderson S et al. Interferon-gamma and tumor necrosis factor-alpha suppress both early and late stages of hematopoiesis and induce programmed cell death. J Cell Physiol 1995;165:538–546.
95. Lepri E, Delfino DV, Migliorati G et al. Functional expression of Fas on mouse bone marrow stromal cells: Upregulation by tumor necrosis factor. Exp Hematol 1998;26:1202–1208.
96. Katharine A. Whartenby, Erin E. Straley, Heeje Kim, Frederick Racke, Vivek Tanavde, Kevin S. Gorski, Linzhao Cheng, Drew M. Pardoll, and Curt I. Civin. Transduction of donor hematopoietic stem-progenitor cells with Fas ligand enhanced short-term engraftment in a murine model of allogeneic bone marrow transplantation. Blood, 1 November 2002, Vol. 100, No. 9, pp. 3147-3154.