Secretion of ligands of the tumor necrosis factor (TNF) superfamily is a conserved response of parenchymal tissues to injury and inflammation that commonly perpetuates elimination of dysfunctional cellular components by apoptosis. The same signals of tissue injury that induce apoptosis in somatic cells activate stem cells and initiate the process of tissue regeneration as a coupling mechanism of injury and recovery. Hematopoietic stem and progenitor cells upregulate the TNF family receptors under stress conditions and are transduced with trophic signals. The progeny gradually acquires sensitivity to receptor-mediated apoptosis along the differentiation process, which becomes the major mechanism of negative regulation of mature proliferating hematopoietic lineages and immune homeostasis. Receptor/ligand interactions of the TNF family are physiological mechanisms transducing the need for repair, which may be harnessed in pathological conditions and transplantation. Because these interactions are physiological mechanisms of injury, neutralization of these pathways has to be carefully considered in disorders that do not involve intrinsic aberrations of excessive susceptibility to apoptosis.

The hallmark of stem and progenitor cell activity in the adult is reconstitution of cells, tissues, and organs under stress conditions such as disease and injury. The immense regenerative capacity of the immunohematopoietic system is best emphasized by spontaneous recovery after radiochemotherapy and therapeutic transplantation of reconstituting progenitors in oncology patients, with perspectives of much wider implementation in the treatment of immune disorders, autoimmunity, and correction of congenital disorders. High turnover rates of bone marrow (BM) hematopoiesis, replacing various lineages at frequencies of 1 day to 12 weeks, render this system excessively sensitivity to disease, irradiation, and toxic agents. Efforts to harness the reconstituting capacity of stem cells for regenerative purposes focused interest in the mechanisms of survival and apoptosis. One of the best characterized signaling systems involves receptors and ligands of the tumor necrosis factor (TNF) superfamily involved in survival, differentiation, proliferation, and function of immunohematopoietic cells.1,2  Negative regulation by TNF receptors controls the size of expanding hematopoietic and immune clones in the terminal stages of differentiation to make way to the activity of other hematopoietic stem and progenitor cells (HSPC).3,4  It is possible that this mode of regulation of clonal expansion maintains hematopoietic heterogeneity, responds to frequent changes in demand, and contains mutagenesis associated with the burst of activity of individual precursors.5  In parallel, all immune cell types upregulate TNF family receptors upon activation and become susceptible to physical elimination by apoptosis,6  a functional characteristic that regulates the intensity of immune reactions through activation-induced cell death (AICD). The present discussion of the involvement of the TNF superfamily in hematopoiesis focuses on 3 main receptor/ligand interactions: (1) Fas is the common executioner of apoptosis in the family and requires trimerization by membrane-bound Fas-ligand, (2) TNFα signals through 2 receptors but only TNF-R1 contains a death domain, and (3) TNF-related apoptosis-induced ligand (TRAIL) signals apoptosis through receptors 1 and 2 among 5 cognate receptors.

Several competing hypotheses have emerged from characterization of the mechanisms of negative regulation in the hematopoietic and immune systems. The first concept has adoptively transferred the dominant role of TNF family receptors in negative regulation from the distal to proximal stages of hematopoietic differentiation. Favoring a negative regulatory role of the Fas and the TNF receptors is deterioration of progenitor viability and clonogenic activity in culture,7,8  and physical and functional suppression of donor progenitor activity by residual host cells.9,10  Consequently, cytokine-induced upregulation of Fas was used to demonstrate inhibition of progenitors in cultures supplemented with supporting growth factors and chemokines.11-13  The second concept suggests that TNF family receptors have dual supportive activity in progenitors and mediate apoptosis in differentiated hematopoietic cells.14-17  Hierarchical susceptibility of immunohematopoietic cells to injury is evident from nonmyeloablative radiochemotherapy, which causes depletion of mature lineages, and spontaneous recovery of surviving endogenous HSPC ensures reconstitution.18,19  According to this scenario, the causes of relative insensitivity of progenitors to apoptosis were derived from somatic cells: (1) low levels of TNF family receptors, (2) upregulated expression of individual antiapoptotic factors, and (3) detrimental consequences of TNF family receptor activation.

Both concepts attribute injury, signaled in part by the TNF family ligands, a potential negative impact on HSPC activity, but they do not account for several features of stress hematopoiesis. A certain degree of damage is rather necessary for functional engraftment in the transplant setting because reconstitution is diminished when progenitor grafting is delayed by several days, a period sufficient for stromal recovery from conditioning-induced injury.20,21  In fact, stem and progenitor cells should be able to fulfill their role as units of regeneration under harsh environments that cause death of somatic cells. Some injury signals such as TNFα and IL-1α are rather protective and enhance recovery of endogenous progenitors from radiation-induced injury.22-24  These features led to the formulation of a third hypothesis, suggesting that insensitivity of the most primitive units of regeneration to signals that cause death of somatic cells is a prerequisite of their reconstituting activity.

The majority of grafted progenitors upregulate the TNF family receptors early after transplantation10,25-27  and under virtually all conditions of stress hematopoiesis. This is a conserved response of progenitors to stress similar to activation of mature immune cells, which may be related to transcriptional programs of gene expression induced by the nuclear factor of activated T cells (NFAT) and nuclear factor-κB (NF-κB).28,29  Acute upregulation of the receptors is most characteristic of progenitors that home successfully to the bone marrow and is affected by donor-host antigen disparity and the nature of hosting parenchymal stroma.25  Furthermore, the possibility of autocrine signaling arises from co-expression of the cognate ligands,25  which is easier to determine for Fas and membrane-bound Fas-ligand (FasL) but is also valid for soluble TNFα.30  This is an apparent paradoxic phenomenon that renders donor cells susceptible to functional suppression9  and physical elimination,10  because the most significant transplant-related early event is protection of HSPC viability by interaction with the marrow stroma.31  Most intriguing is the coordinated ubiquitous upregulation of the TNF family receptors in most primitive murine hematopoietic precursors characterized by absent lineage markers and co-expression of c-Kit and SCA-1 (KLS), as well as the small-sized subset of long-term reconstituting cells isolated by elutriation,25,32  suggesting physiological involvement of the TNF family receptors in the engraftment process. These considerations refute the contention that HSPC are unresponsive to TNF signaling: reconstituting units are highly responsive to these signals under stress conditions such as radiochemotherapy, transplantation, infection, and bleeding.

Donor lineage–negative, KLS, and elutriated murine HSPC harvested from the BM of recipients are remarkably insensitive to an apoptotic challenge despite ubiquitous expression of death receptors.25-27,32  Likewise, human CD34+ progenitors derived from umbilical cord blood (UCB), bone marrow (BMC), and mobilized peripheral blood (MPB) are resistant to apoptosis triggered by FasL, TNFα, and TRAIL.26,29,33,34  Profiling of human progenitors derived from the 3 main sources—UCB, BMC, and MPB—emphasized robust transcription of components of receptor-related and mitochondrial apoptotic signaling pathways.29,35-38  Functional mitochondrial apoptotic pathway might be anticipated from the extreme metabolic stress experienced during burst in progenitor activity at the onset of differentiation and proliferation, with suicidal consequences in case of energetic compromise. Survival of progenitors under stress conditions of ex vivo culture has been attributed to a series of antiapoptotic factors, which are found at higher levels in more primitive hematopoietic precursors. In most cases, equilibrium between pro- and antiapoptotic factors favors survival of human UCB progenitors, such as high Bcl-2/Bax protein ratios39,40  that sustain viability in the presence of functional Fas signaling.41,42  Likewise, high FLIP/caspase-8 protein ratios are considered to preserve progenitor viability,43,44  along with increased transcriptional and translational upregulation of survivin, a member of the inhibitor of apoptosis (IAP) family.45  Expression of these factors is regulated by NF-κB, a pathway associated with preservation of progenitor viability under stress conditions.38,46,47  Although these factors may be significant in preservation of progenitor viability, transcriptional profiles demonstrate ample and developed mechanisms of protection from apoptosis. For example, similar to death induced by the topoisomerase II inhibitor etoposide,35  apoptosis triggered by Fas crosslinking was associated with low Bcl-2/Bax ratios, primarily as a result of transcriptional upregulation of Bax and, to a lesser extent, downregulation of Bcl-2.29  Progenitors resistant to Fas crosslinking also display low transcriptional FLIP/caspase-8 ratios along high and stable members of the IAP family, questioning whether these are the dominant regulators of cell viability.29 

Survival of Fas crosslinking was specifically associated with an inherent transcriptional pattern unfavorable to the transduction of apoptotic signals, including marked upregulation of constituents of the NF-κB pathways. Both canonical (adaptive) and noncanonical (developmental) pathways are upregulated from the level of the triggering receptors to the end products, corresponding to dimers of the NF-κB1 and 2 translocation factors and the partially homologous RelA and RelB transactivation domains.29,38  The NF-κB pathways are involved primarily in sustained clonogenic activity48  and, to a lesser extent, in progenitor commitment to differentiation49 ; thus their earliest role is to sustain cell viability before engagement in developmental programs. The reason for transcriptional upregulation of a significant number of target genes of NF-κB in mitotically quiescent and inactive progenitors is intermittent shuttling of the NF-κB1/RelA signaling unit conjugated to its physiological inhibitor IκB to the nucleus.50  Rather vast patterns of transcription29,38  and translation46,47  may be a wasteful mode of regulation of apoptosis and survival; however, they endow stem and progenitor cells with flexible capacity in adjusting viability by the processing of multiple receptor–mediated environmental cues and integration of signals of energetic sufficiency. A burst in stem and progenitor cell function is preempted by inherent dual transcriptional upregulation of pro- and antiapoptotic pathways, along significant buffering capacity for deterministic triggers of cell death and products of the NF-κB gene expression programs. It is tempting to speculate that the transcriptomes and proteasomes of mitotically and functionally quiescent HSPC affect the nature of response: the default mechanism of stem cells to extreme stress is differentiation, whereas somatic cells (mature progeny) are induced into apoptosis. The capacity to regulate survival is particularly significant under conditions that deviate from the optional linear differentiation, including progenitor dedifferentiation and transient engagement of stem cells in various differentiation traits.51-53 

Continuous reconstitution of developmental systems from progenitors involves elaborate mechanisms of activation, differentiation, proliferation, and incorporation in the injured tissue. Toxic chemokines and inflammatory cytokines are released by the injured BM stroma and hematopoietic cells, activating extrinsic pathways of programed cell death to ensure deletion of defective cellular components that might have survived injury. Dysfunctional cellular elements should be eliminated and replaced to resume proper tissue function through activation of progenitors. Some of the factors released by dead cells are unknown, such as those that suppress clonogenic activity in culture.29,54  Among the identified factors that contribute to compensatory regeneration are TNF family receptor/ligand interactions, which enhance reconstitution by activation of stem and progenitor cells. It is therefore not surprising that hematopoietic engraftment depends to a certain extent on injury18,19  that signals the need to terminate the state of stem cell dormancy,55  questioning the consequences of TNF family receptor signaling in modulation of such acute responses. Trophic activities of TNF family receptors have been recognized in several vital organs, for example neural and glial cells are resistant to apoptosis triggered by Fas and TRAIL-R2 and are sensitive to apoptosis induced by TNF-R1,56-58  but these receptors also induce neural progenitor growth.59,60  This functional duality raises debate over the potential therapeutic efficiency of neutralization of these receptors to reduce cell death in neurodegenerative disorders, which concomitantly may block their trophic activities.57  Likewise, hepatocytes are excessively sensitive to Fas-mediated apoptosis, yet liver regeneration depends on functional Fas signaling.61  The murine and human hematopoietic system displays similar differential responses to TNF family receptors: trophic signaling in stem and progenitor cells and induction of apoptosis in the differentiated and mature progeny.26,27,33,34,54  As growth factors, the TNF family members couple injury with mechanisms of cellular repair.

The TNF family receptors do not induce particular traits of differentiation, but rather they activate HSPC and synergize with other growth factors in initiating clonogenic activity. TNF-R1 and TRAIL-R1 synergize with granulocyte-macrophage colony stimulating factor (GM-CSF) and erythropoietin (EPO) and substantially increase the number of human colonies by ∼50%.27,33,54  In addition to support of committed human and murine progenitors in semisolid cultures, myeloid differentiation is also enhanced in xenochimeric mice after pretransplant exposure of human progenitors to TNFα, affirmed by secondary clonogenic assays and transplants.27,29,33,34,54  This surrogate assay evaluates more primitive hematopoietic precursors, possibly associated with the human reconstituting cells, emphasizing that acute TNF receptor activation is memorized during the very long period of human cell development under conditions of partial interspecies chemokine compatibility. Trophic signaling of the TNFα and TRAIL receptors may be mediated by termination of the state of dormancy and/or diversion of signaling toward growth, which might represent an autocrine mechanism of activation because TNFα induces both secretion of growth factors, such as IL-3 and GM-CSF, and concomitantly upregulates the cognate receptors.62  The molecular mechanisms of trophic signaling by the TNF family receptors remain elusive. TNF-R1 and TRAIL-R1 potentiate recruitment and initiation of the activity of colony-forming units in cultures stimulated by SCF, IL-3, and GM-CSF, whereas Fas indirectly suppresses clonogenic activity through apoptosis of sensitive cells. A distinct checkpoint is involvement of caspase-8 in trophic activity of TNF-R1, which is not required for signaling by Fas and TRAIL-R1, whereas effector caspase-3 is dispensable in trophic activities of all receptors.26,29,54,63  Susceptibility to receptor-mediated apoptotic signaling develops gradually along the differentiation process, resulting in decreased size of colonies mediated predominantly by Fas and TNF-R1.25-27,29,54 

Trophic activities are not equally distributed among members of the TNF superfamily. TRAIL sustains viability of human HSPC, enhances engraftment in xenogeneic chimeras after extended ex vivo culture, and is the most potent stimulant of clonogenic activity (Figure 1).26,64  The ligand induces expression of both cognate receptors without affecting the proliferation of human progenitors. The TRAIL receptors operate in a compensatory mode, with TRAIL-R1 supporting activation and TRAIL-R2 suppressing HSPC activity, and concurrent signaling of both receptors converges to induce a marked increase in the size of myeloid colonies.26  The only murine TRAIL-2 receptor expressed in one-third of fresh BM progenitors is upregulated within several days after transplantation, including universal expression in KLS cells and assumes both apoptotic and trophic activities, resulting in a substantial 50% increase in the number of colonies in semisolid cultures.26  Human and murine progenitor stimulation requires high concentrations of TRAIL, usually one order of magnitude higher than other ligands within the TNF family.

Figure 1

Interactions between TNF family receptors. (A) Stem and progenitor cells; (B) the differentiated progeny. The impact of receptor ligation by the cognate ligands and crosstalk are presented in reference to proliferation, induced expression, apoptosis, survival, and trophic signaling. Joint activation of Fas/TNF-R1 and Fas/TRAIL-R1 results in superior survival, with dominant influence of the TNFα and TRAIL receptors.

Figure 1

Interactions between TNF family receptors. (A) Stem and progenitor cells; (B) the differentiated progeny. The impact of receptor ligation by the cognate ligands and crosstalk are presented in reference to proliferation, induced expression, apoptosis, survival, and trophic signaling. Joint activation of Fas/TNF-R1 and Fas/TRAIL-R1 results in superior survival, with dominant influence of the TNFα and TRAIL receptors.

Close modal

Fas is the common executioner of apoptosis within the TNF family, unique in its requirement of receptor trimerization by oligomers of Fas-ligand, usually bound to cell membrane,28  whereas the soluble isoform blocks receptor trimerization and neutralizes this mechanism.65,66  Both human and murine progenitors are insensitive to Fas-induced apoptosis, whereas the differentiating progeny acquires sensitivity to negative regulation (Figure 1). Trophic activities seen in semisolid cultures of murine BM progenitors and in vivo32  are less evident in human progenitors, although engraftment is enhanced in xenogeneic chimeras.29 

The functions of TNF receptors are conserved in human and murine progenitors, with dual apoptotic and trophic activity of TNF-R1 and no detectable effect of TNF-R2 on viability and differentiation.27,54  TNF-R1 supports murine and human progenitor viability, augments clonogenic activity, enhances hematopoietic reconstitution, and fosters early myeloid recovery after pretransplant exposure to TNFα. The only significant impact of TNF-R2 in unstimulated UCB cells is suppression of proliferation (Figure 1)64  of CD34+TNF-R2+ progenitors that cycle at faster rates than progenitors expressing TNF-R1.54,67  However, impaired competitiveness and durable reconstitution of progenitors deficient in either one of the TNF receptors attribute TNF-R2, a significant, yet unidentified role in hematopoiesis.27  It is possible that some of the features of TNF-R2 are evaluated in suboptimal surrogate assays, because it is preferentially activated by membrane-bound TNFα, whereas TNF-R1 is activated by the soluble ligand.68,69 

Interpretation of the activities of TNF family receptors has to consider inductive crosstalk, such as the classical inductive effect of TNFα on Fas,7,11,12  which is mediated by both TNF receptors.64  In contrast to the association of Fas crosslinking with impaired function of chemokine-stimulated progenitors, we found that TNF-R1 is dominant over Fas and TNF-R2 in protecting survival of progenitors in the absence of supporting chemokines.33,64  Fas indirectly suppresses clonogenic activities of progenitors by induction of apoptosis in sensitive cells, as demonstrated by the restrictive impact of dead cells in semisolid cultures.29,54  This phenomenon might bias the interpretation of in vitro assays as direct negative regulation of progenitor viability and function, and suggests that chemokine supplements sensitize to Fas-induced apoptosis and abolish the dominant protective effect of TNF-R1.70,71 

Additional inductive interactions between receptors include Fas-mediated upregulation of TRAIL-R1 and reversal of slow proliferation of CD34+TRAIL-R1+ progenitors,26  which result in decreased fractional apoptosis.64  These patterns of crosstalk are interesting because both TRAIL receptors and Fas are common targets of gene transcription induced by NF-κB, yet only TRAIL-R1 is transcribed and translated, whereas transcription of TRAIL-R2 is not induced by Fas crosslinking.64  Furthermore, TRAIL is the only TNF family member that upregulates both cognate receptors 1 and 2 in hematopoietic progenitors (Figure 1). If upregulated expression of the cognate receptors by TRAIL and of Fas by TNFα was mediated solely by NF-κB translocation, it is unexplained why the transcription program is only partially performed: TRAIL does not induce Fas and TNFα does not induce the TRAIL receptors.

The best characterized model of hematopoietic progenitor activation is the acute response to infection and inflammation,72  mediated by a series of receptors to cytokines and chemokines expressed by HSPC.29,35,38  The influence of growth factors has been long recognized,73  with subsequent identification of the involvement of toll-like receptors74  and cytokines such as interferons α and γ, which suppress and induce hematopoietic progenitor function upon tonic and phasic exposure, respectively.75,76  Similar differential effects are observed for short and sustained TNF receptor activation,77,78  which induces transcription of different sets of genes controlled by NF-κB and has distinct consequences under various experimental conditions.7-17,70,72  Identification of multiple functions of TNF superfamily receptor/ligand interactions in hematopoietic cells questions the physiological significance. Engraftment of murine lineage–negative progenitors deficient in Fas, TNF-R1, and TNF-R2 is markedly deficient, with transient early support of hematopoiesis but reduced competitive durable reconstitution.27,79  The activity of these receptors is therefore essential rather than complementary in the process of hematopoietic reconstitution from stem cells. An apparent confounding observation is superior competitive engraftment of donor cells deficient in either one or both TNF receptors after transplantation of whole BM cells.80  The higher levels of chimerism observed in mice reconstituted with cells deficient in TNF receptors might be caused by extended survival of the progeny in the periphery, rather than the reduced engraftment caused by sensitivity to TNFα.81  Several physiological mechanisms have been identified thus far with direct and indirect influences on immunogenic and nonimmunogenic events in the course of hematopoietic reconstitution.

The first mechanism involves trophic signals that support HSPC activity and foster repopulation of the immune-hematopoietic system, mediated by autocrine and/or paracrine signaling in progenitors displaying joint upregulation of TNF family receptors and ligands under stress conditions. Autocrine signaling was evident from the markedly improved engraftment when ectopic metalloproteinase-resistant FasL protein was adsorbed onto the surface of murine BM progenitors.25,32  Paracrine signaling was evident from impaired engraftment under conditions of incompetent Fas/FasL interaction caused by both the absence of receptors in donor cells (lpr) and the ligand in recipients (gld).79  In both cases, trophic signaling was mediated by functional Fas trimerization because neutralizing antibodies and the soluble FasL isoform reversed the beneficial effects on progenitor activity in culture.32,65  Furthermore, human progenitors responsive to TNFα displayed enhanced clonogenic activity and myeloid differentiation in culture54,82  and myelomonocytic reconstitution in xenogeneic chimeras,33,54,83  possibly mediated by activation of the NF-κB pathway.29,48  TNFα is abundantly secreted as a consequence of tissue injury and is also produced by hematopoietic progenitors and immune cells.30,83,84 

The second mechanism involves self-defense of the hematopoietic progenitors from host versus graft rejection in nonmyeloablative transplants. Physiological and pharmacologic overexpression of FasL improve the chances of hematopoietic progenitors to engraft in the host BM.25,85  FasL is inherently expressed in approximately one-third of most primitive hematopoietic stem cells79  endowed with long-term reconstituting potential and nonhematopoietic differentiation capacity,86,87  conferring immune privilege to progenitors in hostile environments. Furthermore, murine KLS progenitors ubiquitously upregulate FasL after transplantation, as a mechanism of immune privilege and defense from residual recipient immunity.25  A similar veto effect is imposed by TNFα to ensure survival of human progenitors and protect from rejection.88,89  In the transplant setting, polarized insensitivity of stem cells and sensitivity of committed and more differentiated progenitors to apoptosis endows the more primitive precursors with a competitive advantage in homing to and seeding in the marrow niches.25  This prioritization mechanism favors engraftment of units of regeneration over cells with more limited reconstituting capacity.

The third mechanism involves bidirectional interactions of hematopoietic progenitors with the niches, which govern and direct the activity of progenitors at distinct sites of residence within the marrow space through inductive and restrictive signals emanating from the stroma, osteoblasts, and extracellular matrix.90  An interesting mechanism is the activation of stromal Fas by the cognate ligand expressed in hematopoietic progenitors.79  Impaired engraftment of wild-type progenitors in Fas-deficient recipients (lpr) and of FasL-deficient progenitors (gld) in wild-type recipients was reversed in the latter case by overexpression of ectopic FasL protein, demonstrating functional signaling in the process of engraftment. Reduced engraftment in the absence of competent stromal Fas signaling was validated in chimeras expressing Fas only in the BM, removing the trophic consequences of Fas signaling in the engrafting progenitors. Likewise, myeloablative transplants showed that the mechanism of stromal signaling is independent of the immune privilege conferred to the grafted progenitors by FasL, particularly because cell-stroma interactions in the BM are not restricted by antigenic barriers.91  Reverse stromal signaling through constitutively expressed Fas92  may be involved in potentiation of the BM to accept additional cells after initial colonization by donor progenitors, resulting in a virtually unlimited seeding space.91 

Resistance to apoptotic signaling is a fundamental characteristic of hematopoietic stem and progenitor cells regulated at the transcriptional level, which has several potential therapeutic implications. Pretransplant exposure of the hematopoietic cell graft to apoptotic ligands accomplishes concomitant independent mechanisms that converge to improve the outcome of transplants. First, an apoptotic challenge is a simple physiological tool for collection of larger numbers of viable HSPC unrestricted by particular phenotypic identifiers, because cellular grafts can be safely processed without impairing the viability of the units of reconstitution.26,29,33,34,54  Receptor-mediated apoptosis can also be used to deplete differentiated progeny under the influence of growth factors in extended cultures of UCB expansion.33  Second, pretransplant exposure of human progenitors to FasL, TNFα, and TRAIL trigger trophic signals that improve subsequent myeloid reconstitution in vivo.26,29,54  Third, equally important is the nature of cells depleted by the apoptotic challenge, including donor T cells that mediate potentially lethal graft-versus-host disease (GVHD). Functional depletion and inactivation of donor T cells may be a better approach to GVHD prophylaxis93  in view of the limited success of phenotypic deletion, because this immune reaction is mediated by redundant effector activity of numerous T-cell subsets instructed by antigen-presenting cells of donor and host origin.94  Depletion of T cells sensitive to apoptosis without host-specific sensitization reduces significantly graft-versus-host reactivity in murine and human grafts, while preserving facilitation of progenitor engraftment and graft-vs-tumor reactivity.33,95  Fourth, some types of malignant cells display excessive sensitivity to apoptosis and can be effectively purged by short graft processing before autologous hematopoietic reconstitution after aggressive radiochemotherapy in oncology patients.96,97  An intriguing possibility is malignant transformation that occurs in more primitive hematopoietic precursors, where inherited resistance to apoptosis might cause aggressive behavior and poor response to therapy.98-100 

Differential trophic responses elicited by the individual receptors allow flexible implementation to modulate several transplant-related parameters. For example, an algorithm of the limitations of various types of transplants can be applied to consider donor-host antigen disparity, the source of HSPC, conditioning, and the anticipated complications in individual patients. Autologous reconstitution in oncology patients requires prompt purging of residual tumor cells using FasL, TNFα, and TRAIL, whereas UCB transplants are limited by the low numbers of cells and delayed engraftment, which may be improved by negative selection and simultaneous trophic signaling by TNFα and TRAIL. Allogeneic and haplo-identical transplants using MPB grafts containing significant numbers of donor T cells present high risk of severe GVHD, which may be ameliorated by exposure to FasL and TNFα. Transplants are frequently performed in heavily pretreated patients with dormant and subclinical infections that might flare in the presence of immunosuppression; thus the use of additional cytotoxic agents to suppress GVHD is unwarranted.

The multifaceted involvement of TNF family receptors in hematopoietic stem and progenitor cell function calls for extreme care in neutralization of these signaling pathways. Apoptotic activities of these receptor/ligand interactions are in fact a mechanism of defense from lymphoproliferation, immunodeficiency, and malignancy, conditions triggered by prolonged administration of immunosuppressive agents. Although members of the TNF family act as cytotoxic pathways in severe GVHD reactions,93  and initial clinical studies suggested that early deletion of TNF-R1+ T cells may be beneficial depending on conditioning,101  randomized studies showed little benefit of this approach to treat severe GVHD.102  Likewise, individual members of the TNF family might participate in particular aspects of dysfunctional hematopoiesis in aplastic anemia and myelodysplastic syndromes.103-106  In perspective of the trophic activities of the TNF superfamily in hematopoiesis, receptors and ligands are expected to increase as a feature of the effort to recover from hypoplasia; thus therapeutic neutralization has to be carefully considered. It remains to be determined which types of dysfunctional hematopoiesis are caused by inherent hypersensitivity of hematopoietic progenitors to apoptosis mediated by specific receptors and/or aberrant responses to certain environments, and whether neutralization or supplementation are of therapeutic potential.103-107 

Contribution: K.M. and N.A. prepared the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Nadir Askenasy, Frankel Laboratory, Center for Stem Cell Research, Schneider Children’s Medical Center of Israel, 14 Kaplan St, Petach Tikva, Israel 49202; e-mail: anadir@012.net.il.

1
Aggarwal
 
BB
Signalling pathways of the TNF superfamily: a double-edged sword.
Nat Rev Immunol
2003
, vol. 
3
 
9
(pg. 
745
-
756
)
2
Aggarwal
 
BB
Nuclear factor-kappaB: the enemy within.
Cancer Cell
2004
, vol. 
6
 
3
(pg. 
203
-
208
)
3
Greil
 
R
Anether
 
G
Johrer
 
K
Tinhofer
 
I
Tuning the rheostat of the myelopoietic system via Fas and TRAIL.
Crit Rev Immunol
2003
, vol. 
23
 
4
(pg. 
301
-
322
)
4
Testa
 
U
Apoptotic mechanisms in the control of erythropoiesis.
Leukemia
2004
, vol. 
18
 
7
(pg. 
1176
-
1199
)
5
Traver
 
D
Akashi
 
K
Weissman
 
IL
Lagasse
 
E
Mice defective in two apoptosis pathways in the myeloid lineage develop acute myeloblastic leukemia.
Immunity
1998
, vol. 
9
 
1
(pg. 
47
-
57
)
6
Aggarwal
 
BB
Gupta
 
SC
Kim
 
JH
Historical perspectives on tumor necrosis factor and its superfamily: 25 years later, a golden journey.
Blood
2012
, vol. 
119
 
3
(pg. 
651
-
665
)
7
Broxmeyer
 
HE
Williams
 
DE
Lu
 
L
, et al. 
The suppressive influences of human tumor necrosis factors on bone marrow hematopoietic progenitor cells from normal donors and patients with leukemia: synergism of tumor necrosis factor and interferon-gamma.
J Immunol
1986
, vol. 
136
 
12
(pg. 
4487
-
4495
)
8
Wisniewski
 
D
Strife
 
A
Atzpodien
 
J
Clarkson
 
BD
Effects of recombinant human tumor necrosis factor on highly enriched hematopoietic progenitor cell populations from normal human bone marrow and peripheral blood and bone marrow from patients with chronic myeloid leukemia.
Cancer Res
1987
, vol. 
47
 
18
(pg. 
4788
-
4794
)
9
Vinci
 
G
Chouaib
 
S
Autran
 
B
Vernant
 
JP
Evidence that residual host cells surviving the conditioning regimen to allogeneic bone marrow transplantation inhibit donor hematopoiesis in vitro—the role of TNF-alpha.
Transplantation
1991
, vol. 
52
 
3
(pg. 
406
-
411
)
10
Saheki
 
K
Fujimori
 
Y
Takemoto
 
Y
Kakishita
 
E
Increased expression of Fas (APO-1, CD95) on CD34+ haematopoietic progenitor cells after allogeneic bone marrow transplantation.
Br J Haematol
2000
, vol. 
109
 
2
(pg. 
447
-
452
)
11
Maciejewski
 
J
Selleri
 
C
Anderson
 
S
Young
 
NS
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
, vol. 
85
 
11
(pg. 
3183
-
3190
)
12
Nagafuji
 
K
Shibuya
 
T
Harada
 
M
, et al. 
Functional expression of Fas antigen (CD95) on hematopoietic progenitor cells.
Blood
1995
, vol. 
86
 
3
(pg. 
883
-
889
)
13
Selleri
 
C
Sato
 
T
Anderson
 
S
Young
 
NS
Maciejewski
 
JP
Interferon-gamma and tumor necrosis factor-alpha suppress both early and late stages of hematopoiesis and induce programmed cell death.
J Cell Physiol
1995
, vol. 
165
 
3
(pg. 
538
-
546
)
14
Backx
 
B
Broeders
 
L
Bot
 
FJ
Löwenberg
 
B
Positive and negative effects of tumor necrosis factor on colony growth from highly purified normal marrow progenitors.
Leukemia
1991
, vol. 
5
 
1
(pg. 
66
-
70
)
15
Caux
 
C
Favre
 
C
Saeland
 
S
, et al. 
Potentiation of early hematopoiesis by tumor necrosis factor-alpha is followed by inhibition of granulopoietic differentiation and proliferation.
Blood
1991
, vol. 
78
 
3
(pg. 
635
-
644
)
16
Rusten
 
LS
Jacobsen
 
FW
Lesslauer
 
W
Loetscher
 
H
Smeland
 
EB
Jacobsen
 
SE
Bifunctional effects of tumor necrosis factor alpha (TNF alpha) on the growth of mature and primitive human hematopoietic progenitor cells: involvement of p55 and p75 TNF receptors.
Blood
1994
, vol. 
83
 
11
(pg. 
3152
-
3159
)
17
Jacobsen
 
FW
Veiby
 
OP
Stokke
 
T
Jacobsen
 
SE
TNF-alpha bidirectionally modulates the viability of primitive murine hematopoietic progenitor cells in vitro.
J Immunol
1996
, vol. 
157
 
3
(pg. 
1193
-
1199
)
18
Ploemacher
 
RE
van Os
 
R
van Beurden
 
CA
Down
 
JD
Murine haemopoietic stem cells with long-term engraftment and marrow repopulating ability are more resistant to gamma-radiation than are spleen colony forming cells.
Int J Radiat Biol
1992
, vol. 
61
 
4
(pg. 
489
-
499
)
19
Down
 
JD
Boudewijn
 
A
van Os
 
R
Thames
 
HD
Ploemacher
 
RE
Variations in radiation sensitivity and repair among different hematopoietic stem cell subsets following fractionated irradiation.
Blood
1995
, vol. 
86
 
1
(pg. 
122
-
127
)
20
Down
 
JD
Tarbell
 
NJ
Thames
 
HD
Mauch
 
PM
Syngeneic and allogeneic bone marrow engraftment after total body irradiation: dependence on dose, dose rate, and fractionation.
Blood
1991
, vol. 
77
 
3
(pg. 
661
-
669
)
21
van Os
 
R
Thames
 
HD
Konings
 
AW
Down
 
JD
Radiation dose-fractionation and dose-rate relationships for long-term repopulating hemopoietic stem cells in a murine bone marrow transplant model.
Radiat Res
1993
, vol. 
136
 
1
(pg. 
118
-
125
)
22
Neta
 
R
Oppenheim
 
JJ
Douches
 
S
Interdependence of the radioprotective effects of human recombinant interleukin 1 alpha, tumor necrosis factor alpha, granulocyte colony-stimulating factor, and murine recombinant granulocyte-macrophage colony-stimulating factor.
J Immunol
1988
, vol. 
140
 
1
(pg. 
108
-
111
)
23
Slørdal
 
L
Muench
 
MO
Warren
 
DJ
Moore
 
MA
Radioprotection by murine and human tumor-necrosis factor: dose-dependent effects on hematopoiesis in the mouse.
Eur J Haematol
1989
, vol. 
43
 
5
(pg. 
428
-
434
)
24
Moreb
 
J
Zucali
 
JR
The therapeutic potential of interleukin-1 and tumor necrosis factor on hematopoietic stem cells.
Leuk Lymphoma
1992
, vol. 
8
 
4-5
(pg. 
267
-
275
)
25
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
, vol. 
35
 
10
(pg. 
1601
-
1612
)
26
Mizrahi
 
K
Stein
 
J
Pearl-Yafe
 
M
Kaplan
 
O
Yaniv
 
I
Askenasy
 
N
Regulatory functions of TRAIL in hematopoietic progenitors: human umbilical cord blood and murine bone marrow transplantation.
Leukemia
2010
, vol. 
24
 
7
(pg. 
1325
-
1334
)
27
Pearl-Yafe
 
M
Mizrahi
 
K
Stein
 
J
, et al. 
Tumor necrosis factor receptors support murine hematopoietic progenitor function in the early stages of engraftment.
Stem Cells
2010
, vol. 
28
 
7
(pg. 
1270
-
1280
)
28
Askenasy
 
N
Yolcu
 
ES
Yaniv
 
I
Shirwan
 
H
Induction of tolerance using Fas-ligand: a double-edged immunomodulator.
Blood
2005
, vol. 
105
 
4
(pg. 
1396
-
1404
)
29
Mizrahi
 
K
Kagan
 
S
Stein
 
J
Yaniv
 
I
Zipori
 
D
Askenasy
 
N
Resistance of hematopoietic progenitors to Fas-mediated apoptosis is actively sustained by NFκB with a characteristic transcriptional signature.
Stem Cells Dev
2014
, vol. 
23
 
6
(pg. 
676
-
686
)
30
Majka
 
M
Janowska-Wieczorek
 
A
Ratajczak
 
J
, et al. 
Numerous growth factors, cytokines, and chemokines are secreted by human CD34(+) cells, myeloblasts, erythroblasts, and megakaryoblasts and regulate normal hematopoiesis in an autocrine/paracrine manner.
Blood
2001
, vol. 
97
 
10
(pg. 
3075
-
3085
)
31
Askenasy
 
N
Yolcu
 
ES
Shirwan
 
H
Stein
 
J
Yaniv
 
I
Farkas
 
DL
Characterization of adhesion and viability of early seeding hematopoietic cells in the host bone marrow in vivo and in situ.
Exp Hematol
2003
, vol. 
31
 
12
(pg. 
1292
-
1300
)
32
Pearl-Yafe
 
M
Stein
 
J
Yolcu
 
ES
, et al. 
Fas transduces dual apoptotic and trophic signals in hematopoietic progenitors.
Stem Cells
2007
, vol. 
25
 
12
(pg. 
3194
-
3203
)
33
Mizrahi
 
K
Ash
 
S
Peled
 
T
Yaniv
 
I
Stein
 
J
Askenasy
 
N
Negative selection by apoptosis enriches progenitors in naïve and expanded human umbilical cord blood grafts.
Bone Marrow Transplant
 
2014. (in press)
34
Mizrahi
 
K
Yaniv
 
I
Ash
 
S
Stein
 
J
Askenasy
 
N
Apoptotic signaling through Fas and TNF receptors ameliorates GVHD in mobilized peripheral blood grafts.
Bone Marrow Transplant
2014
, vol. 
49
 
5
(pg. 
640
-
648
)
35
Tao
 
W
Hangoc
 
G
Hawes
 
JW
Si
 
Y
Cooper
 
S
Broxmeyer
 
HE
Profiling of differentially expressed apoptosis-related genes by cDNA arrays in human cord blood CD34+ cells treated with etoposide.
Exp Hematol
2003
, vol. 
31
 
3
(pg. 
251
-
260
)
36
Ng
 
YY
van Kessel
 
B
Lokhorst
 
HM
, et al. 
Gene-expression profiling of CD34+ cells from various hematopoietic stem-cell sources reveals functional differences in stem-cell activity.
J Leukoc Biol
2004
, vol. 
75
 
2
(pg. 
314
-
323
)
37
Jaatinen
 
T
Hemmoranta
 
H
Hautaniemi
 
S
, et al. 
Global gene expression profile of human cord blood-derived CD133+ cells.
Stem Cells
2006
, vol. 
24
 
3
(pg. 
631
-
641
)
38
Panepucci
 
RA
Calado
 
RT
Rocha
 
V
Proto-Siqueira
 
R
Silva
 
WA
Zago
 
MA
Higher expression of transcription targets and components of the nuclear factor-kappaB pathway is a distinctive feature of umbilical cord blood CD34+ precursors.
Stem Cells
2007
, vol. 
25
 
1
(pg. 
189
-
196
)
39
Peters
 
R
Leyvraz
 
S
Perey
 
L
Apoptotic regulation in primitive hematopoietic precursors.
Blood
1998
, vol. 
92
 
6
(pg. 
2041
-
2052
)
40
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
, vol. 
191
 
2
(pg. 
253
-
264
)
41
Bárcena
 
A
Park
 
SW
Banapour
 
B
Muench
 
MO
Mechetner
 
E
Expression of Fas/CD95 and Bcl-2 by primitive hematopoietic progenitors freshly isolated from human fetal liver.
Blood
1996
, vol. 
88
 
6
(pg. 
2013
-
2025
)
42
Maurillo
 
L
Del Poeta
 
G
Venditti
 
A
, et al. 
Quantitative analysis of Fas and bcl-2 expression in hematopoietic precursors.
Haematologica
2001
, vol. 
86
 
3
(pg. 
237
-
243
)
43
Kim
 
H
Whartenby
 
KA
Georgantas
 
RW
Wingard
 
J
Civin
 
CI
Human CD34+ hematopoietic stem/progenitor cells express high levels of FLIP and are resistant to Fas-mediated apoptosis.
Stem Cells
2002
, vol. 
20
 
2
(pg. 
174
-
182
)
44
Ratajczak
 
J
Kucia
 
M
Reca
 
R
Zhang
 
J
Machalinski
 
B
Ratajczak
 
MZ
Quiescent CD34+ early erythroid progenitors are resistant to several erythropoietic ‘inhibitory’ cytokines; role of FLIP.
Br J Haematol
2003
, vol. 
123
 
1
(pg. 
160
-
169
)
45
Fukuda
 
S
Pelus
 
LM
Regulation of the inhibitor-of-apoptosis family member survivin in normal cord blood and bone marrow CD34(+) cells by hematopoietic growth factors: implication of survivin expression in normal hematopoiesis.
Blood
2001
, vol. 
98
 
7
(pg. 
2091
-
2100
)
46
Pyatt
 
DW
Stillman
 
WS
Yang
 
Y
Gross
 
S
Zheng
 
JH
Irons
 
RD
An essential role for NF-kappaB in human CD34(+) bone marrow cell survival.
Blood
1999
, vol. 
93
 
10
(pg. 
3302
-
3308
)
47
Kerzic
 
PJ
Pyatt
 
DW
Zheng
 
JH
Gross
 
SA
Le
 
A
Irons
 
RD
Inhibition of NF-kappaB by hydroquinone sensitizes human bone marrow progenitor cells to TNF-alpha-induced apoptosis.
Toxicology
2003
, vol. 
187
 
2-3
(pg. 
127
-
137
)
48
Bugarski
 
D
Krstic
 
A
Mojsilovic
 
S
, et al. 
Signaling pathways implicated in hematopoietic progenitor cell proliferation and differentiation.
Exp Biol Med (Maywood)
2007
, vol. 
232
 
1
(pg. 
156
-
163
[Maywood]
49
De Molfetta
 
GA
Lucíola Zanette
 
D
Alexandre Panepucci
 
R
Dos Santos
 
AR
da Silva
 
WA
Antonio Zago
 
M
Role of NFKB2 on the early myeloid differentiation of CD34+ hematopoietic stem/progenitor cells.
Differentiation
2010
, vol. 
80
 
4-5
(pg. 
195
-
203
)
50
Ghosh
 
S
Hayden
 
MS
New regulators of NF-kappaB in inflammation.
Nat Rev Immunol
2008
, vol. 
8
 
11
(pg. 
837
-
848
)
51
Zipori
 
D
The stem state: plasticity is essential, whereas self-renewal and hierarchy are optional.
Stem Cells
2005
, vol. 
23
 
6
(pg. 
719
-
726
)
52
Askenasy
 
N
Yaniv
 
I
Stein
 
J
Sharkis
 
SJ
Our perception of developmental plasticity: esse est percipi (to be is to be perceived)?
Curr Stem Cell Res Ther
2006
, vol. 
1
 
1
(pg. 
85
-
94
)
53
Shoshani
 
O
Zipori
 
D
Mammalian cell dedifferentiation as a possible outcome of stress.
Stem Cell Rev
2011
, vol. 
7
 
3
(pg. 
488
-
493
)
54
Mizrahi
 
K
Stein
 
J
Yaniv
 
I
Kaplan
 
O
Askenasy
 
N
TNF-α has tropic rather than apoptotic activity in human hematopoietic progenitors: involvement of TNF receptor-1 and caspase-8.
Stem Cells
2013
, vol. 
31
 
1
(pg. 
156
-
166
)
55
Trumpp
 
A
Essers
 
M
Wilson
 
A
Awakening dormant haematopoietic stem cells.
Nat Rev Immunol
2010
, vol. 
10
 
3
(pg. 
201
-
209
)
56
Song
 
JH
Bellail
 
A
Tse
 
MC
Yong
 
VW
Hao
 
C
Human astrocytes are resistant to Fas ligand and tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis.
J Neurosci
2006
, vol. 
26
 
12
(pg. 
3299
-
3308
)
57
Haase
 
G
Pettmann
 
B
Raoul
 
C
Henderson
 
CE
Signaling by death receptors in the nervous system.
Curr Opin Neurobiol
2008
, vol. 
18
 
3
(pg. 
284
-
291
)
58
Harry
 
GJ
Lefebvre d’Hellencourt
 
C
McPherson
 
CA
Funk
 
JA
Aoyama
 
M
Wine
 
RN
Tumor necrosis factor p55 and p75 receptors are involved in chemical-induced apoptosis of dentate granule neurons.
J Neurochem
2008
, vol. 
106
 
1
(pg. 
281
-
298
)
59
Lambert
 
C
Landau
 
AM
Desbarats
 
J
Fas-beyond death: a regenerative role for Fas in the nervous system.
Apoptosis
2003
, vol. 
8
 
6
(pg. 
551
-
562
)
60
Kajiwara
 
K
Ogata
 
S
Tanihara
 
M
Promotion of neurite outgrowth from fetal hippocampal cells by TNF-alpha receptor 1-derived peptide.
Cell Transplant
2005
, vol. 
14
 
9
(pg. 
665
-
672
)
61
Desbarats
 
J
Newell
 
MK
Fas engagement accelerates liver regeneration after partial hepatectomy.
Nat Med
2000
, vol. 
6
 
8
(pg. 
920
-
923
)
62
Jacobsen
 
SE
Ruscetti
 
FW
Dubois
 
CM
Keller
 
JR
Tumor necrosis factor alpha directly and indirectly regulates hematopoietic progenitor cell proliferation: role of colony-stimulating factor receptor modulation.
J Exp Med
1992
, vol. 
175
 
6
(pg. 
1759
-
1772
)
63
Secchiero
 
P
Gonelli
 
A
Mirandola
 
P
, et al. 
Tumor necrosis factor-related apoptosis-inducing ligand induces monocytic maturation of leukemic and normal myeloid precursors through a caspase-dependent pathway.
Blood
2002
, vol. 
100
 
7
(pg. 
2421
-
2429
)
64
Mizrahi
 
K
Askenasy
 
N
Activation and crosstalk between TNF family receptors in umbilical cord blood cells is not responsible for loss of engraftment capacity following culture.
Am J Stem Cells
2013
, vol. 
2
 
3
(pg. 
155
-
164
)
65
Bárcena
 
A
Muench
 
MO
Song
 
KS
Ohkubo
 
T
Harrison
 
MR
Role of CD95/Fas and its ligand in the regulation of the growth of human CD34(++)CD38(-) fetal liver cells.
Exp Hematol
1999
, vol. 
27
 
9
(pg. 
1428
-
1439
)
66
Josefsen
 
D
Myklebust
 
JH
Lynch
 
DH
Stokke
 
T
Blomhoff
 
HK
Smeland
 
EB
Fas ligand promotes cell survival of immature human bone marrow CD34+CD38- hematopoietic progenitor cells by suppressing apoptosis.
Exp Hematol
1999
, vol. 
27
 
9
(pg. 
1451
-
1459
)
67
Warren
 
DJ
Slørdal
 
L
Moore
 
MA
Tumor-necrosis factor induces cell cycle arrest in multipotential hematopoietic stem cells: a possible radioprotective mechanism.
Eur J Haematol
1990
, vol. 
45
 
3
(pg. 
158
-
163
)
68
Grell
 
M
Douni
 
E
Wajant
 
H
, et al. 
The transmembrane form of tumor necrosis factor is the prime activating ligand of the 80 kDa tumor necrosis factor receptor.
Cell
1995
, vol. 
83
 
5
(pg. 
793
-
802
)
69
Müller
 
S
Rihs
 
S
Schneider
 
JM
, et al. 
Soluble TNF-alpha but not transmembrane TNF-alpha sensitizes T cells for enhanced activation-induced cell death.
Eur J Immunol
2009
, vol. 
39
 
11
(pg. 
3171
-
3180
)
70
Stahnke
 
K
Hecker
 
S
Kohne
 
E
Debatin
 
KM
CD95 (APO-1/FAS)-mediated apoptosis in cytokine-activated hematopoietic cells.
Exp Hematol
1998
, vol. 
26
 
9
(pg. 
844
-
850
)
71
Dybedal
 
I
Yang
 
L
Bryder
 
D
Aastrand-Grundstrom
 
I
Leandersson
 
K
Jacobsen
 
SE
Human reconstituting hematopoietic stem cells up-regulate Fas expression upon active cell cycling but remain resistant to Fas-induced suppression.
Blood
2003
, vol. 
102
 
1
(pg. 
118
-
126
)
72
Takizawa
 
H
Boettcher
 
S
Manz
 
MG
Demand-adapted regulation of early hematopoiesis in infection and inflammation.
Blood
2012
, vol. 
119
 
13
(pg. 
2991
-
3002
)
73
Cheers
 
C
Haigh
 
AM
Kelso
 
A
Metcalf
 
D
Stanley
 
ER
Young
 
AM
Production of colony-stimulating factors (CSFs) during infection: separate determinations of macrophage-, granulocyte-, granulocyte-macrophage-, and multi-CSFs.
Infect Immun
1988
, vol. 
56
 
1
(pg. 
247
-
251
)
74
Nagai
 
Y
Garrett
 
KP
Ohta
 
S
, et al. 
Toll-like receptors on hematopoietic progenitor cells stimulate innate immune system replenishment.
Immunity
2006
, vol. 
24
 
6
(pg. 
801
-
812
)
75
Gardner
 
RV
Interferon-gamma (IFN-gamma) as a potential radio- and chemo-protectant.
Am J Hematol
1998
, vol. 
58
 
3
(pg. 
218
-
223
)
76
Yu
 
JM
Emmons
 
RV
Hanazono
 
Y
Sellers
 
S
Young
 
NS
Dunbar
 
CE
Expression of interferon-gamma by stromal cells inhibits murine long-term repopulating hematopoietic stem cell activity.
Exp Hematol
1999
, vol. 
27
 
5
(pg. 
895
-
903
)
77
Hoffmann
 
A
Levchenko
 
A
Scott
 
ML
Baltimore
 
D
The IkappaB-NF-kappaB signaling module: temporal control and selective gene activation.
Science
2002
, vol. 
298
 
5596
(pg. 
1241
-
1245
)
78
Nelson
 
DE
Ihekwaba
 
AE
Elliott
 
M
, et al. 
Oscillations in NF-kappaB signaling control the dynamics of gene expression.
Science
2004
, vol. 
306
 
5696
(pg. 
704
-
708
)
79
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
, vol. 
25
 
6
(pg. 
1448
-
1455
)
80
Pronk
 
CJ
Veiby
 
OP
Bryder
 
D
Jacobsen
 
SE
Tumor necrosis factor restricts hematopoietic stem cell activity in mice: involvement of two distinct receptors.
J Exp Med
2011
, vol. 
208
 
8
(pg. 
1563
-
1570
)
81
Rebel
 
VI
Hartnett
 
S
Hill
 
GR
Lazo-Kallanian
 
SB
Ferrara
 
JL
Sieff
 
CA
Essential role for the p55 tumor necrosis factor receptor in regulating hematopoiesis at a stem cell level.
J Exp Med
1999
, vol. 
190
 
10
(pg. 
1493
-
1504
)
82
Lardon
 
F
Snoeck
 
HW
Berneman
 
ZN
, et al. 
Generation of dendritic cells from bone marrow progenitors using GM-CSF, TNF-alpha, and additional cytokines: antagonistic effects of IL-4 and IFN-gamma and selective involvement of TNF-alpha receptor-1.
Immunology
1997
, vol. 
91
 
4
(pg. 
553
-
559
)
83
Rezzoug
 
F
Huang
 
Y
Tanner
 
MK
, et al. 
TNF-alpha is critical to facilitate hemopoietic stem cell engraftment and function.
J Immunol
2008
, vol. 
180
 
1
(pg. 
49
-
57
)
84
Umland
 
O
Heine
 
H
Miehe
 
M
Marienfeld
 
K
Staubach
 
KH
Ulmer
 
AJ
Induction of various immune modulatory molecules in CD34(+) hematopoietic cells.
J Leukoc Biol
2004
, vol. 
75
 
4
(pg. 
671
-
679
)
85
Whartenby
 
KA
Straley
 
EE
Kim
 
H
, et al. 
Transduction of donor hematopoietic stem-progenitor cells with Fas ligand enhanced short-term engraftment in a murine model of allogeneic bone marrow transplantation.
Blood
2002
, vol. 
100
 
9
(pg. 
3147
-
3154
)
86
Jones
 
RJ
Collector
 
MI
Barber
 
JP
, et al. 
Characterization of mouse lymphohematopoietic stem cells lacking spleen colony-forming activity.
Blood
1996
, vol. 
88
 
2
(pg. 
487
-
491
)
87
Jang
 
YY
Sharkis
 
SJ
Stem cell plasticity: a rare cell, not a rare event.
Stem Cell Rev
2005
, vol. 
1
 
1
(pg. 
45
-
51
)
88
Rachamim
 
N
Gan
 
J
Segall
 
H
, et al. 
Tolerance induction by “megadose” hematopoietic transplants: donor-type human CD34 stem cells induce potent specific reduction of host anti-donor cytotoxic T lymphocyte precursors in mixed lymphocyte culture.
Transplantation
1998
, vol. 
65
 
10
(pg. 
1386
-
1393
)
89
Gur
 
H
Krauthgamer
 
R
Bachar-Lustig
 
E
, et al. 
Immune regulatory activity of CD34+ progenitor cells: evidence for a deletion-based mechanism mediated by TNF-alpha.
Blood
2005
, vol. 
105
 
6
(pg. 
2585
-
2593
)
90
Wilson
 
A
Trumpp
 
A
Bone-marrow haematopoietic-stem-cell niches.
Nat Rev Immunol
2006
, vol. 
6
 
2
(pg. 
93
-
106
)
91
Askenasy
 
N
Farkas
 
DL
Antigen barriers or available space do not restrict in situ adhesion of hemopoietic cells to bone marrow stroma.
Stem Cells
2002
, vol. 
20
 
1
(pg. 
80
-
85
)
92
Lepri
 
E
Delfino
 
DV
Migliorati
 
G
Moraca
 
R
Ayroldi
 
E
Riccardi
 
C
Functional expression of Fas on mouse bone marrow stromal cells: upregulation by tumor necrosis factor-alpha and interferon-gamma.
Exp Hematol
1998
, vol. 
26
 
13
(pg. 
1202
-
1208
)
93
Blazar
 
BR
Murphy
 
WJ
Abedi
 
M
Advances in graft-versus-host disease biology and therapy.
Nat Rev Immunol
2012
, vol. 
12
 
6
(pg. 
443
-
458
)
94
Ho
 
VT
Soiffer
 
RJ
The history and future of T-cell depletion as graft-versus-host disease prophylaxis for allogeneic hematopoietic stem cell transplantation.
Blood
2001
, vol. 
98
 
12
(pg. 
3192
-
3204
)
95
Askenasy
 
N
Mizrahi
 
K
Ash
 
S
Askenasy
 
EM
Yaniv
 
I
Stein
 
J
Depletion of naïve lymphocytes with fas ligand ex vivo prevents graft-versus-host disease without impairing T cell support of engraftment or graft-versus-tumor activity.
Biol Blood Marrow Transplant
2013
, vol. 
19
 
2
(pg. 
185
-
195
)
96
Ashkenazi
 
A
Targeting the extrinsic apoptosis pathway in cancer.
Cytokine Growth Factor Rev
2008
, vol. 
19
 
3-4
(pg. 
325
-
331
)
97
Droin
 
N
Guéry
 
L
Benikhlef
 
N
Solary
 
E
Targeting apoptosis proteins in hematological malignancies.
Cancer Lett
2013
, vol. 
332
 
2
(pg. 
325
-
334
)
98
Qin
 
Y
Camoretti-Mercado
 
B
Blokh
 
L
Long
 
CG
Ko
 
FD
Hamann
 
KJ
Fas resistance of leukemic eosinophils is due to activation of NF-kappa B by Fas ligation.
J Immunol
2002
, vol. 
169
 
7
(pg. 
3536
-
3544
)
99
de Totero
 
D
Montera
 
M
Rosso
 
O
, et al. 
Resistance to CD95-mediated apoptosis of CD40-activated chronic lymphocytic leukemia B cells is not related to lack of DISC molecules expression.
Hematol J
2004
, vol. 
5
 
2
(pg. 
152
-
160
)
100
Romano
 
C
De Fanis
 
U
Sellitto
 
A
, et al. 
Induction of CD95 upregulation does not render chronic lymphocytic leukemia B-cells susceptible to CD95-mediated apoptosis.
Immunol Lett
2005
, vol. 
97
 
1
(pg. 
131
-
139
)
101
Choi
 
SW
Stiff
 
P
Cooke
 
K
, et al. 
TNF-inhibition with etanercept for graft-versus-host disease prevention in high-risk HCT: lower TNFR1 levels correlate with better outcomes.
Biol Blood Marrow Transplant
2012
, vol. 
18
 
10
(pg. 
1525
-
1532
)
102
Alousi
 
AM
Weisdorf
 
DJ
Logan
 
BR
, et al. 
Blood and Marrow Transplant Clinical Trials Network
Etanercept, mycophenolate, denileukin, or pentostatin plus corticosteroids for acute graft-versus-host disease: a randomized phase 2 trial from the Blood and Marrow Transplant Clinical Trials Network.
Blood
2009
, vol. 
114
 
3
(pg. 
511
-
517
)
103
Welsh
 
JP
Rutherford
 
TR
Flynn
 
J
Foukaneli
 
T
Gordon-Smith
 
EC
Gibson
 
FM
In vitro effects of interferon-gamma and tumor necrosis factor-alpha on CD34+ bone marrow progenitor cells from aplastic anemia patients and normal donors.
Hematol J
2004
, vol. 
5
 
1
(pg. 
39
-
46
)
104
Zeng
 
W
Miyazato
 
A
Chen
 
G
Kajigaya
 
S
Young
 
NS
Maciejewski
 
JP
Interferon-gamma-induced gene expression in CD34 cells: identification of pathologic cytokine-specific signature profiles.
Blood
2006
, vol. 
107
 
1
(pg. 
167
-
175
)
105
Kakagianni
 
T
Giannakoulas
 
NC
Thanopoulou
 
E
, et al. 
A probable role for trail-induced apoptosis in the pathogenesis of marrow failure. Implications from an in vitro model and from marrow of aplastic anemia patients.
Leuk Res
2006
, vol. 
30
 
6
(pg. 
713
-
721
)
106
Platzbecker
 
U
Kurre
 
P
Guardiola
 
P
, et al. 
Fanconi anemia type C-deficient hematopoietic cells are resistant to TRAIL (TNF-related apoptosis-inducing ligand)-induced cleavage of pro-caspase-8.
Exp Hematol
2004
, vol. 
32
 
9
(pg. 
815
-
821
)
107
Milsom
 
MD
Schiedlmeier
 
B
Bailey
 
J
, et al. 
Ectopic HOXB4 overcomes the inhibitory effect of tumor necrosis factor-alpha on Fanconi anemia hematopoietic stem and progenitor cells.
Blood
2009
, vol. 
113
 
21
(pg. 
5111
-
5120
)
Sign in via your Institution