The proinflammatory cytokine interferon-γ (IFN-γ) is well known for its important role in innate and adaptive immunity against intracellular infections and for tumor control. Yet, it has become clear that IFN-γ also has a strong impact on bone marrow (BM) output during inflammation, as it affects the differentiation of most hematopoietic progenitor cells. Here, we review the impact of IFN-γ on hematopoiesis, including the function of hematopoietic stem cells (HSCs) and more downstream progenitors. We discuss which hematopoietic lineages are functionally modulated by IFN-γ and through which underlying molecular mechanism(s). We propose the novel concept that IFN-γ acts through upregulation of suppressor of cytokine signaling molecules, which impairs signaling of several cytokine receptors. IFN-γ has also gained clinical interest from different angles, and we discuss how chronic IFN-γ production can lead to the development of anemia and BM failure and how it is involved in malignant hematopoiesis. Overall, this review illustrates the wide-ranging effect of IFN-γ on the (patho-)physiological processes in the BM.

Normal tissue homeostasis and immune surveillance are highly dependent on stable production of blood cells, which is tightly orchestrated by the intricate process of hematopoiesis in the BM. Yet, infections disturb this homeostatic process and typically result in depletion of mature immune cells due to consumption of the cells fighting the invading pathogen. This requires a rapid compensatory response in hematopoiesis to provide the immune system with the appropriate type of cells during the infection and to restore cellular homeostasis in the blood system when the infection has been resolved. One of the clinically relevant consequences is that hematopoiesis is suppressed upon infection, which can lead to leucopenia and anemia. How these hematopoietic stress events are regulated is not fully understood, but it has become clear that functional crosstalk exists between the activated immune system and the hematopoietic compartment in the bone marrow (BM).1,2  An important role in this respect has been ascribed to interferon-γ (IFN-γ). This type II interferon is typically produced by T, natural killer, and natural killer T cells during an immune response to intracellular pathogens, like mycobacteria and viruses. Although IFN-γ is an important cytokine in orchestrating a vast array of immunologic responses, it is also historically known as a suppressor of hematopoiesis, and BM failure syndromes resulting from chronic inflammation are associated with IFN-γ production (reviewed in Mangan3  and Trinchieri et al4 ). Over the last decade, it has become evident which BM cell types are affected by IFN-γ (Figure 1) and through what molecular and cellular mechanisms. Here, we discuss these studies and lay out how they have contributed to our understanding of IFN-γ as a modulator of stress hematopoiesis.

Figure 1

Impact of IFN-γ on hematopoiesis. Overview of hematopoietic differentiation, in which is indicated whether IFN-γ has either a stimulating (thick red arrow) or inhibitory (dotted thin red arrow) effect on a particular lineage. IFN-γ receptor α (IFNγR) is expressed on all analyzed progenitors, including HSCs, MPPs, common myeloid progenitors (CMPs), GMPs, megakaryocyte-erythroid progenitors (MEPs), and common lymphoid progenitors (CLPs) (shown in green; unpublished observations; expression of IFNγR on fully differentiated cells is not indicated in this figure). LT-HSC, long-term HSC; NK, natural killer; ST-HSC, short-term HSC.

Figure 1

Impact of IFN-γ on hematopoiesis. Overview of hematopoietic differentiation, in which is indicated whether IFN-γ has either a stimulating (thick red arrow) or inhibitory (dotted thin red arrow) effect on a particular lineage. IFN-γ receptor α (IFNγR) is expressed on all analyzed progenitors, including HSCs, MPPs, common myeloid progenitors (CMPs), GMPs, megakaryocyte-erythroid progenitors (MEPs), and common lymphoid progenitors (CLPs) (shown in green; unpublished observations; expression of IFNγR on fully differentiated cells is not indicated in this figure). LT-HSC, long-term HSC; NK, natural killer; ST-HSC, short-term HSC.

Close modal

The impact of IFN-γ on HSCs

The first experimental evidence that IFN-γ can inhibit hematopoiesis dates back to the 1980s, when IFN-γ was shown to inhibit colony formation in vitro.5-7  However, it remained obscure whether hematopoietic stem cells (HSCs) are also affected by IFN-γ, because most studies addressed the effect of IFN-γ on hematopoiesis by using total BM, lineage-depleted cells, or enriched progenitors. Instead, Snoeck et al used a 2-step culture system to study the effect of IFN-γ on purified human CD34+CD38 HSCs.8  Single HSCs were first clonally expanded in liquid cultures with or without IFN-γ for 2 weeks, followed by assessing colony-forming unit (CFU) activity in subsequent semisolid cultures, which indicated that IFN-γ strongly diminishes expansion of HSCs in the primary liquid cultures.8  Likewise, by combining long-term culture-initiating cell (LTC-IC) assays with CFU assays on purified human CD34+ BM cells, Selleri et al also showed that exogenous IFN-γ, and particularly IFN-γ produced by stromal cells, has a potent inhibiting effect on the number of LTC-ICs, indicating functional impairment of HSCs.9  Chronic exposure of IFN-γ is required for the inhibiting effect, and it has been suggested that IFN-γ negatively affects proliferation and increases apoptosis.9  Correspondingly, IFN-γ inhibits expansion of cord blood–derived CD34+CD38 HSCs in liquid cultures and impairs the maintenance of LTC-ICs, as shown by CFU assays and reconstitution of nonobese diabetic/severe combined immunodeficient mice.10 

Next to these studies with human HSCs, IFN-γ also negatively affects the maintenance of murine HSCs. IFN-γ–producing stromal cells decrease CFU activity of murine BM cells and impair their long-term repopulating activity upon in vivo transfer.11  Moreover, we showed that maintenance of highly purified murine HSCs (identified as Linc-Kit+Sca1+CD150+CD48 BM cells) is also impaired by IFN-γ.12  IFN-γ does not affect cell-cycle entry, differentiation, or apoptosis of HSCs in these assays, but decreases the number of self-renewing cell divisions. This was attributed to the ability of IFN-γ to perturb thrombopoietin (TPO)-induced phosphorylation of signal transducer and activator of transcription 5 (STAT5), possibly through suppressor of cytokine signaling 1 (SOCS1) and thereby expression of STAT5-associated genes involved in the regulation of HSC proliferation, which provides a molecular explanation on how IFN-γ negatively affects HSC self-renewal.12  Interestingly, IFN-γ also regulates the reconstitution capacity of HSCs by modulating TPO-dependent interaction of the integrin αvβ3 with the extracellular matrix protein Nov/CCN3, indicating that TPO-mediated cellular interactions in the niche might also be modulated by IFN-γ.13 

The well-established notion that in vitro exposure to IFN-γ impairs HSC maintenance is substantiated by several studies that have shown in vivo suppressive effects of IFN-γ on HSC function. Overexpression of IFN-γ in IFN-γ–transgenic mice decreases the number of CFU-GEMM colonies derived from BM, indicating a loss of functional multipotent HSCs.14  Conversely, HSCs from mice that lack IFN-γ signaling have a better reconstitution capacity during the steady-state situation compared with wild-type (WT) mice, indicating that the basal IFN-γ tone is sufficient to affect HSC function.12,15  The negative impact of IFN-γ on HSC becomes more apparent upon immune challenge, as IFN-γ production during chronic infection with Mycobacterium avium decreases HSC engraftment potential, though it would enhance HSC proliferation.15  Using an infection model with lymphocytic choriomeningitis virus (LCMV), we found no evidence for enhanced proliferation of HSCs, but we did find that IFN-γ impairs HSC self-renewal. By generating WT:IFNγR1−/− mixed BM chimeric mice, we could demonstrate this was due to a direct effect of IFNγ on HSCs and that IFN-γ modulates expression of genes (CyclinD1, p57) involved in the regulation of HSC self renewal in vivo.12  So whereas the negative effect of IFN-γ on HSC reconstitution is not disputed, discrepancies exist on the impact of IFN-γ on HSC proliferation,12,15,16  which may be due to variations in HSC characterization and the experimental models employed. The context in which HSCs are exposed to IFN-γ is indeed important, as IFN-γ production by adoptively transferred CD8 T cells in a steady-state situation is not sufficient to influence HSCs, though it does modulate more downstream progenitors17  (see below). We postulate that the impact of IFN-γ on HSCs is dependent on the status of the HSC compartment; acute viral infections, such as with LCMV, can decimate the HSC compartment through the production of type I IFNs.18  As a result, HSC self-renewal is induced to restore HSC numbers after the infection, and this compensatory proliferation is an IFN-γ–sensitive process.12  However, if HSC numbers are stable and self-renewal is not enhanced, IFN-γ may have little effect on HSCs.17 

In conjunction with these context-dependent effects, it is of interest to also consider the impact of type I IFNs on HSC function. Production of interferon-α in a noninfectious setting is sufficient to activate quiescent HSCs and drive them into cell cycle,19,20  possibly through the induction of c-Myc protein expression.21  However, the impact of type I IFNs on HSCs is different between in vitro and in vivo settings and might even change depending on the time of exposure.16  The release of HSC quiescence and induction of proliferation upon initial IFN production is in fact followed by a return of HSCs to quiescence upon prolonged exposure, which protects them from IFN-induced apoptosis.16  It is thus important to consider that the impact of IFNs on HSCs is depending on the stimulatory condition. In line with this, mice deficient for adar1 or irgm1,22,23  genes induced by IFN-γ, have hyperproliferative HSCs with a decreased reconstitution capacity and elevated IFN signaling and IFN-γ serum levels, respectively. We suggest that like type I IFNs, IFN-γ can have stimulating and suppressing effects on HSC proliferation and reconstitution, and the outcome of this balance may be context dependent.

Effect of IFN-γ on multipotent progenitors and myelopoiesis

Next to its impact on HSCs, it has been suggested that IFN-γ produced upon infection or when intravenously injected expands the population of Linc-Kit+Sca-1+ cells, which contains both HSCs and more committed progenitor cells.24-27  However, in our opinion, these findings are difficult to interpret, as interferon-signaling induces Sca-1 expression on many hematopoietic cells2,28  and thereby leads to an overestimation of the number of Linc-Kit+Sca-1+ cells. IFN-γ has been shown to enhance differentiation of purified CD34+ hematopoietic stem and progenitor cells, CMPs, and granulocyte-macrophage progenitors (GMPs),12  indicating that IFN-γ can modulate lineage-specific myeloid differentiation. Indeed, a critical role for IFN-γ in infection-induced myelopoiesis has been demonstrated by infecting IFN-γ–deficient mice with Mycobacterium bovis, because these mice display a severe granulocytosis in BM, peripheral blood, and spleen.29  Similar increases in granulocytes have been reported after infection of IFN-γ–deficient mice with Mycobacterium tuberculosis30  and Toxoplasma gondii.31  Moreover, infection with either Ehrlichia muris or LCMV induces predominantly monopoiesis in WT mice, whereas neutrophil development is strongly increased in infected IFN-γ–deficient mice.32,33  This strong impact of IFN-γ on myelopoiesis is due to the fact that IFN-γ reciprocally regulates the production of neutrophils and monocytes. On one hand, IFN-γ induces expression of SOCS3 in GMPs and thereby inhibits granulocyte colony stimulating factor (G-CSF)-induced activation of STAT3,32  an essential transcription factor for emergency granulopoiesis.34  On the other hand, IFN-γ also elevates expression of monocyte-promoting transcription factors PU.1 and interferon responsive factor 8 (IRF-8) in the same myeloid progenitors.32  These findings corroborate a previous report demonstrating that IFN-γ stimulates monocytic colony formation from human progenitor cells, while G-CSF–induced granulocytic colony formation is impaired in the presence of IFN-γ.35  Interestingly, IFN-γ can also induce monocyte differentiation indirectly through production of interleukin-6 (IL-6) by BM mesenchymal stromal cells (MSCs), whereby IL-6 decreases expression of Runx1 and CEBPα in multipotent progenitors and thereby skews them toward the monocytic lineage.17  Importantly, T-cell–driven immunity and IFN-γ production can also induce the mobilization of MSCs from the BM,36  which further illustrates the complexity of the cellular dynamics in the BM during inflammation. Moreover, IFN-γ enhances the ability of MSCs to inhibit T-cell proliferation and proinflammatory cytokine production through upregulation of the immunoregulatory enzymes indoleamine 2,3-dioxygenase (IDO) and inducible nitric oxide synthase37-39  and may thus also play a role in restoring immune homeostasis in the BM.

Regarding the impact of IFN-γ on myelopoiesis, it is of interest that IFN-γ can also negatively affect eosinophil development upon viral infection or sensitization with an allergen. Mice deficient for IFN-γ signaling show accumulation of eosinophils in the lungs or brains upon infection with respiratory syncytial virus40  or Borna disease virus,41  respectively. Additionally, IL-12–mediated induction of IFN-γ signaling inhibits eosinophilia upon allergen challenge of sensitized mice42  or after parasite infection.43  Interestingly, IFN-γ has a direct inhibiting effect on the differentiation of eosinophils, as IFN-γ reduces expression of IL-5Rα and GATA-1 in progenitor cells, which are both essential factors in eosinophil development.44  Whereas IFN-γ impairs IL-5–mediated differentiation of myeloid progenitors to eosinophils, IFN-γ does not alter the response to granulocyte-macrophage colony stimulating factor–supported cultures, again demonstrating that IFN-γ has lineage-specific effects on myelopoiesis.44  Furthermore, malaria infection induces the generation of a novel type of progenitor cell, which is dependent on IFN-γ signaling. These cells express IL-7Rα, like lymphoid progenitors, but transplantation experiments revealed that these cells produce myeloid progeny in vivo, which contribute to the clearance of malaria-infected cells.45  In conclusion, IFN-γ production during immunologic challenges is very important for tightly orchestrating myelopoiesis by directly regulating growth factor responsiveness and transcription factor expression, as well as cytokine release from stromal cells in the BM

The impact of IFN-γ on erythropoiesis and thrombopoiesis

Chronic infections are notorious for inducing anemia, and a role for IFN-γ has been well recognized in this process. IFN-γ disturbs iron homeostasis and thereby the erythroid balance by inducing iron retention in macrophages.46  Furthermore, IFN-γ–activated macrophages contribute to red blood cell (RBC) loss by increased hemophagocytosis.47  However, erythroid colony formation of hematopoietic progenitors is also directly suppressed by IFN-γ,48,49  and IFN-γ primarily inhibits the earliest stages of erythroid differentiation and proliferation.50  Interestingly, it has also been suggested that IL-15 plays an important role for the IFN-γ–mediated inhibition of erythropoiesis, both in vitro and in vivo.51  Erythroid progenitors are obtained from megakaryocyte-erythroid progenitors, which, in contrast to myeloid progenitors, express low levels of PU.1 and high levels of GATA-1. Yet, exposure of erythroid progenitors to IFN-γ in vitro or in vivo is sufficient to increase PU.1 messenger RNA and protein expression.52  Using human erythroid progenitors, we established that IFN-γ–induced upregulation of PU.1 is dependent on IRF-1 and that knockdown of either IRF-1 or PU.1 expression is sufficient to overcome IFN-γ–induced reduction in erythroid colony formation.52  Because PU.1 can physically interact with GATA-1 and thereby inhibit its function in erythroid differentiation,53  we postulate that the IFN-γ–mediated increase of PU.1 alters the transcriptional profile of hematopoietic progenitors, thereby blocking their erythroid differentiation potential.52 

Strikingly, although IFN-γ diminishes TPO signaling in HSCs and impairs HSC proliferation, IFN-γ does not interfere with the strongly TPO-dependent process of megakaryopoiesis (Tsuji et al54 and A.M.d.B., unpublished data). A possible explanation for this divergence is the requirement of STAT1 for megakaryopoiesis, which is a downstream target of GATA1 but also activated by IFN-γ signaling.55  Moreover, the IFN-γ target genes IRF1 and IRF2 are involved in the regulation of megakaryopoiesis by inducing expression of the integrin CD41.55,56  Whereas IFN-γ is not required for megakaryopoiesis, IFN-γ–induced factors do not reduce but instead contribute to the process of platelet formation.

The effect of IFN-γ on B lymphopoiesis

The impact of IFN-γ on hematopoiesis is not restricted to the myeloid and erythroid lineage but extends to the lymphoid lineage. IFN-γ expression is not required for B-cell development but enhanced IFN-γ signaling in vivo is sufficient to inhibit B-cell differentiation, as was shown in mice overexpressing IFN-γ14  or human IRF157  or lacking SOCS1.58  Moreover, enhanced costimulation of T cells through CD27 by overexpression of its ligand CD70 strongly increases the production of effector T cells, which inhibits B lymphopoiesis in an IFN-γ–dependent manner.59 

The molecular mechanism by which IFN-γ inhibits B-cell development in these mice has not been unraveled, but there is strong evidence that this is related to IL-7 signaling, because IFN-γ inhibits IL-7–induced proliferation of primary pre-B cells in vitro.60  Whereas IFN-γ alone does not directly induce apoptosis of pre-B cells in vitro, increased IFN-γ–mediated cell death is observed in IL-7–supplemented cultures of these cells,60,61  which is rescued by overexpressing Bcl2.61  While IFN-γ does not affect IL-7 messenger RNA expression, a decreased binding of IL-7 to its receptor has been reported, which can explain the negative effect of IFN-γ on IL-7–induced survival and proliferation.60  Furthermore, IFN-γ also induces SOCS1 expression in immature B cells, thereby abrogating signaling through the IL-7 receptor and decreasing the responsiveness of these cells to IL-7.62 

Although decreased IL-7 signaling may explain the negative impact of IFN-γ on B-cell differentiation, it could also be that IFN-γ changes the transcriptional profile required for B-cell differentiation. The fact that IFN-γ can induce PU.1 expression in progenitor cells52  may also be relevant in this context, because low levels of PU.1 are required for B-cell differentiation, but high PU.1 levels induce myeloid and inhibit B-cell differentiation.63  Therefore, we hypothesize that next to an IFN-γ–mediated decreased responsiveness to IL-7, IFN-γ also increases PU.1 expression in B cells and thereby contributes to the block in B-cell differentiation.

The impact of IFN-γ on cytokine signaling and transcription factor expression

Many cytokines, including IFN-γ, induce expression of SOCS molecules, which stimulates a negative feedback to the receptor that is transmitting the signal in order to terminate the signaling process.64  We showed that IFN-γ induces SOCS1 and SOCS3 in hematopoietic progenitor cells, thereby perturbing respectively TPO12  and G-CSF responses.32  Furthermore, IFN-γ–induced SOCS1 expression was shown to abrogate IL-7 signaling in immature B cells.62  Thus, an emerging concept arises that IFN-γ negatively affects the differentiation of various hematopoietic lineages through SOCS-mediated inhibition of cytokine signaling at the level of STAT activation and thereby inhibits the proliferation and/or differentiation of the progenitors of that particular lineage (Figure 2). The question arises whether IFN-γ can in a similar manner also inhibit the signaling of other cytokines. It would be interesting to examine if responsiveness to erythropoietin (EPO) can be modulated in this fashion, because EPO acts through phosphorylation of STAT5 and induces its own negative feedback through SOCS1 and SOCS3.64,65  If so, this would imply that IFN-γ inhibits erythropoiesis at multiple levels, namely by impairing EPO signaling through SOCS and by inducing PU.1 expression through IRF1.52  Moreover, the antagonizing effect of PU.1 on GATA-153  may be relevant not only for erythropoiesis, but also for eosinophil differentiation, because GATA-1 is important in IL-5Rα+ eosinophil progenitors.66  It is therefore tempting to speculate (Figure 2) that PU.1-mediated inhibition of GATA-1 may also contribute to the IFN-γ–driven impairment of eosinophil differentiation,44  but this remains to be determined.

Figure 2

IFN-γ regulates function and expression of key hematopoietic transcription factors. IFN-γ impairs HSC self-renewal as well as differentiation of neutrophils and B cells by upregulating SOCS proteins, which block cytokine-induced STAT signaling. Furthermore, IFN-γ also induces expression of monocyte-inducing transcription factors IRF8 and PU.1. The latter inhibits the function of GATA1, which is important for the development of erythrocytes and eosinophilic granulocytes.

Figure 2

IFN-γ regulates function and expression of key hematopoietic transcription factors. IFN-γ impairs HSC self-renewal as well as differentiation of neutrophils and B cells by upregulating SOCS proteins, which block cytokine-induced STAT signaling. Furthermore, IFN-γ also induces expression of monocyte-inducing transcription factors IRF8 and PU.1. The latter inhibits the function of GATA1, which is important for the development of erythrocytes and eosinophilic granulocytes.

Close modal

IFN-γ production and BM failure

Although short-term exposure of hematopoietic progenitors to IFN-γ may be beneficial during viral infection due to its monocyte-skewing effects, the coinciding suppression of other lineages should not persist, as this will negatively affect blood cell homeostasis (Figure 3). The clinical relevance of this is evident in patients with aplastic anemia (AA), which have increased levels of IFN-γ in their circulation and impaired colony-forming potential of BM cells, which improves with anti–IFN-γ antibodies.67  Although oligoclonally expanded T cells reactive to hematopoietic progenitors are present in AA patients, the antigen to which they respond is unknown.68  IFN-γ production by circulating T cells is also increased in AA patients, which may be driven by increased levels of T-bet, and the presence of these IFN-γ–producing lymphocytes correlates well with the response to immunosuppressive therapy and subsequent recovery.69,70  Moreover, a polymorphism in the gene encoding IFN-γ that results in overproduction of IFN-γ is associated with the risk of AA.71  Prolonged suppression of several hematopoietic lineages by IFN-γ is important in disease progression, but IFN-γ–dependent upregulation of Fas, resulting in increased cell death of hematopoietic progenitors, may also contribute to a decline in hematopoietic output in AA.72,73 

Figure 3

Chronic production of IFN-γ induces BM failure, monocytosis, and AA. Chronic IFN-γ production during persistent viral infection or chronic inflammatory diseases has a dramatic impact on BM output, as it impairs HSC self-renewal, increases monopoiesis, and inhibits development of B cells, granulocytes, and RBCs (cellular identity according to Figure 1). Adapted from King et al2  with permission.

Figure 3

Chronic production of IFN-γ induces BM failure, monocytosis, and AA. Chronic IFN-γ production during persistent viral infection or chronic inflammatory diseases has a dramatic impact on BM output, as it impairs HSC self-renewal, increases monopoiesis, and inhibits development of B cells, granulocytes, and RBCs (cellular identity according to Figure 1). Adapted from King et al2  with permission.

Close modal

Although most information regarding the role of IFN-γ in BM failure in human disease comes from studies of AA,74  chronic immune activation is a major underlying cause of IFN-γ–associated BM failure. Decreased numbers of IFN-γ–producing lymphocytes have been associated with hematologic improvement following immunosuppression in patients with hypoplastic myelodysplasia.75  IFN-γ is also associated with hematopoietic suppression observed in patients with Fanconi anemia and HIV.76,77  In a mouse model for graft-versus-host disease, it has been shown that IFN-γ produced by CD4 T cells is responsible for the development of severe anemia and BM aplasia when the recipients receive sublethal irradiation. However, when the recipients are lethally irradiated (followed by BM transplantation), IFN-γ production is rather protective.78  Using IFN-γ–reporter mice, it has also been shown that allogeneic Th1 cells generated during graft-versus-host disease can traffic to hematopoietic sites and induce BM failure via IFN-γ–mediated toxicity.79 

From a therapeutic perspective it is interesting that anti–IFN-γ antibodies improve the in vitro capacity of hematopoietic progenitors derived from patients with AA.67,80  Moreover, in a mouse model of T-cell–induced BM failure, characterized by severe pancytopenia and BM hypoplasia, anti–IFN-γ can even enhance survival.81  Antibodies to IFN-γ have been clinically tested in Th1-driven diseases,82  and it would be interesting to investigate whether such a treatment is sufficient to reduce hematopoietic suppression in patients with AA or other forms of inflammation-induced BM failure.

Role of IFN-γ in malignant hematopoiesis

IFN-γ not only has a strong impact on normal hematopoiesis, but also influences malignant hematopoietic cells, though both growth limiting and promoting effects have been reported. IFN-γ inhibits colony formation and expansion of tumor cells obtained from patients with acute myeloid leukemia (AML),83  chronic myeloid leukemia (CML),84  and multiple myeloma (MM).85  Next to growth-limiting and cytotoxic effects of IFN-γ on leukemic cells, IFN-γ induces differentiation of immature myeloid blasts from AML and CML patients.86  Interestingly, these effects seem to be dependent on other cytokines, because IFN-γ enhances colony formation of IL-3–dependent AML cell lines, but inhibits the growth of IL-3–independent cell lines. Remarkably, both the inhibitory and stimulatory effects of IFN-γ can be reduced by blocking tumor necrosis factor α.87  Furthermore, IL-6 is a major cytokine for MM cells, and IFN-γ inhibits IL-6–dependent proliferation of MM cells by interfering with IL-6 signaling85 ; whether this effect is SOCS dependent is not known. Thus, the effect of IFN-γ on malignant cells seems context dependent, which is also evident from the observation that the effect of IFN-γ on AML cells depends on the cytokines added and the stromal subset that is used in cocultures.88  It could thus be that the in vivo effect of IFN-γ on growth of tumor cells depends on the local cytokine milieu created by supporting niche cells and by autocrine cytokine signaling from the malignant cells. In addition, tumor-specific genomic alteration of leukemic cells might also be involved in regulating the response to IFN-γ signaling. Activating mutations in FLT3, which occur in 20% to 25% of AML patients, enable direct phosphorylation of STAT5. Although SOCS1 is also upregulated in these cells, SOCS1 is not able to inhibit this cytokine-independent activation of STAT5. Aberrant FLT3-induced SOCS1 even prevents the inhibitory effect of IFN-γ on colony formation of transformed cells,89  demonstrating that activated oncogenic pathways can modulate the effect of IFN-γ on growth of malignant cells.

Next to regulation of cytokine responses, IFN-γ upregulates expression of Fas on MM cells, sensitizing them to Fas-mediated apoptosis.90  In contrast, IFN-γ induces PD-L1 expression in AML,91  CML,92  and MM93  cells, thereby reducing cytotoxic T-cell–mediated lysis. Using a CML mouse model, it has been shown that PD-1/PDL-1 interaction impairs the immune control of CML in vivo94  and that IFN-γ enhances leukemic outgrowth.92  Although it was claimed that IFN-γ promotes tumor progression by inducing proliferation of leukemic stem cells, it might be interesting to investigate in more detail the direct role of IFN-γ in PD-L1–dependent immune escape of transformed cells. It is also suggested that IFN-γ enables immune escape of leukemic cells by inducing IDO in AML cells, which suppresses antitumor activity of T cells.95  On the other hand, whereas IFN-γ does not affect IDO expression in MM cells, IDO levels in MSCs increase after stimulation with IFN-γ, resulting in apoptosis of cocultured MM cells.96 

Altogether, IFN-γ might act as a double-edged sword in malignant hematopoiesis in which its pro- or antitumor activity is dependent on the nature of the hematologic malignancy and the interplay between a variety of molecular, cellular, and microenvironmental factors. However, as IFN-γ affects malignant cells, immune cells, and nonhematopoietic niche cells, it will be challenging to translate results of in vitro experiments to more complex in vivo situations. Therefore, in-depth studies on the in vivo effects of IFN-γ on malignant hematopoiesis are required to delineate its effect on cytokine signaling, immune escape, and the leukemic niche in the BM.

Altogether, IFN-γ has strong and diverse effects on various levels of hematopoiesis, and these are clinically relevant. Although IFN-γ has subtle effects on steady-state hematopoiesis, it profoundly regulates blood cell production during immunologic challenges, indicating that IFN-γ is an important factor in controlling BM output during stress situations. There is also emerging evidence that MSCs are important players in mediating these effects of IFN-γ on hematopoiesis in the BM.

We postulate that the evolutionary beneficial role of IFN-γ is that it enhances the formation of the appropriate immune cells to combat an invading pathogen. Myeloid cells are the first line of defense against pathogens, and in contrast to RBCs, have a very limited lifespan. A transient decrease in erythropoiesis will therefore not directly affect oxygen transport, while decreased competition for growth factors and space in the BM may facilitate expansion of myeloid cells. IFN-γ is typically produced in immune responses against intracellular pathogens and induces monopoiesis at the cost of neutrophil and eosinophil formation, which are less relevant during such infections and more important to clear extracellular pathogens. Yet, it is interesting to speculate that this disbalance in myeloid cells may contribute to the increased susceptibility of virally infected patients to subsequent bacterial infections.97  Prolongation of IFN-γ production during chronic inflammation will lead to a disbalance in blood cell homeostasis and cause anemia. Better understanding of the underlying molecular mechanism may facilitate the development of successful strategies to tackle inflammation-induced anemia and modulate disease progression in case of malignant hematopoiesis.

The authors thank Dr Howard A. Young (Laboratory of Experimental Immunology, National Cancer Institute, Frederick, MD) and Dr Sacha Zeerleder (Department of Hematology, Academic Medical Center, Amsterdam, The Netherlands) for critical reading of the manuscript and D. Egberts for support. For this review, the authors were limited in the number of words and references they could use, and given the breadth of the topics discussed, they were obliged to make a selection of the papers that they would refer to. Hence, the authors apologize to those whose relevant work was not cited in this review.

C.V. and M.A.N. are both supported by the Landsteiner Foundation for Blood Transfusion Research (LSBR fellowships 1101 and 1014, respectively).

Contribution: A.M.d.B., C.V., and M.A.N. jointly designed the figures and wrote the paper.

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

Correspondence: Martijn A. Nolte, Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory AMC/UvA, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; e-mail: m.nolte@sanquin.nl.

1
Takizawa
 
H
Boettcher
 
S
Manz
 
MG
Demand-adapted regulation of early hematopoiesis in infection and inflammation.
Blood
2012
, vol. 
119
 
13
(pg. 
2991
-
3002
)
2
King
 
KY
Goodell
 
MA
Inflammatory modulation of HSCs: viewing the HSC as a foundation for the immune response.
Nat Rev Immunol
2011
, vol. 
11
 
10
(pg. 
685
-
692
)
3
Mangan
 
KF
Immune disregulation of hematopoiesis.
Annu Rev Med
1987
, vol. 
38
 (pg. 
61
-
70
)
4
Trinchieri
 
G
Murphy
 
M
Perussia
 
B
Regulation of hematopoiesis by T lymphocytes and natural killer cells.
Crit Rev Oncol Hematol
1987
, vol. 
7
 
3
(pg. 
219
-
265
)
5
Klimpel
 
GR
Fleischmann
 
WR
Klimpel
 
KD
Gamma interferon (IFN gamma) and IFN alpha/beta suppress murine myeloid colony formation (CFU-C)N: magnitude of suppression is dependent upon level of colony-stimulating factor (CSF).
J Immunol
1982
, vol. 
129
 
1
(pg. 
76
-
80
)
6
Broxmeyer
 
HE
Lu
 
L
Platzer
 
E
Feit
 
C
Juliano
 
L
Rubin
 
BY
Comparative analysis of the influences of human gamma, alpha and beta interferons on human multipotential (CFU-GEMM), erythroid (BFU-E) and granulocyte-macrophage (CFU-GM) progenitor cells.
J Immunol
1983
, vol. 
131
 
3
(pg. 
1300
-
1305
)
7
Zoumbos
 
NC
Djeu
 
JY
Young
 
NS
Interferon is the suppressor of hematopoiesis generated by stimulated lymphocytes in vitro.
J Immunol
1984
, vol. 
133
 
2
(pg. 
769
-
774
)
8
Snoeck
 
HW
Van Bockstaele
 
DR
Nys
 
G
, et al. 
Interferon gamma selectively inhibits very primitive CD342+CD38- and not more mature CD34+CD38+ human hematopoietic progenitor cells.
J Exp Med
1994
, vol. 
180
 
3
(pg. 
1177
-
1182
)
9
Selleri
 
C
Maciejewski
 
JP
Sato
 
T
Young
 
NS
Interferon-gamma constitutively expressed in the stromal microenvironment of human marrow cultures mediates potent hematopoietic inhibition.
Blood
1996
, vol. 
87
 
10
(pg. 
4149
-
4157
)
10
Yang
 
L
Dybedal
 
I
Bryder
 
D
, et al. 
IFN-gamma negatively modulates self-renewal of repopulating human hemopoietic stem cells.
J Immunol
2005
, vol. 
174
 
2
(pg. 
752
-
757
)
11
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
)
12
de Bruin
 
AM
Demirel
 
Ö
Hooibrink
 
B
Brandts
 
CH
Nolte
 
MA
Interferon-γ impairs proliferation of hematopoietic stem cells in mice.
Blood
2013
, vol. 
121
 
18
(pg. 
3578
-
3585
)
13
Ishihara
 
J
Umemoto
 
T
Yamato
 
M
, et al. 
Nov/CCN3 regulates long-term repopulating activity of murine hematopoietic stem cells via integrin αvβ3.
Int J Hematol
2014
, vol. 
99
 
4
(pg. 
393
-
406
)
14
Young
 
HA
Klinman
 
DM
Reynolds
 
DA
, et al. 
Bone marrow and thymus expression of interferon-gamma results in severe B-cell lineage reduction, T-cell lineage alterations, and hematopoietic progenitor deficiencies.
Blood
1997
, vol. 
89
 
2
(pg. 
583
-
595
)
15
Baldridge
 
MT
King
 
KY
Boles
 
NC
Weksberg
 
DC
Goodell
 
MA
Quiescent haematopoietic stem cells are activated by IFN-gamma in response to chronic infection.
Nature
2010
, vol. 
465
 
7299
(pg. 
793
-
797
)
16
Pietras
 
EM
Lakshminarasimhan
 
R
Techner
 
JM
, et al. 
Re-entry into quiescence protects hematopoietic stem cells from the killing effect of chronic exposure to type I interferons.
J Exp Med
2014
, vol. 
211
 
2
(pg. 
245
-
262
)
17
Schürch
 
CM
Riether
 
C
Ochsenbein
 
AF
Cytotoxic CD8+ T cells stimulate hematopoietic progenitors by promoting cytokine release from bone marrow mesenchymal stromal cells.
Cell Stem Cell
2014
, vol. 
14
 
4
(pg. 
460
-
472
)
18
Binder
 
D
Fehr
 
J
Hengartner
 
H
Zinkernagel
 
RM
Virus-induced transient bone marrow aplasia: major role of interferon-alpha/beta during acute infection with the noncytopathic lymphocytic choriomeningitis virus.
J Exp Med
1997
, vol. 
185
 
3
(pg. 
517
-
530
)
19
Essers
 
MA
Offner
 
S
Blanco-Bose
 
WE
, et al. 
IFNalpha activates dormant haematopoietic stem cells in vivo.
Nature
2009
, vol. 
458
 
7240
(pg. 
904
-
908
)
20
Sato
 
T
Onai
 
N
Yoshihara
 
H
Arai
 
F
Suda
 
T
Ohteki
 
T
Interferon regulatory factor-2 protects quiescent hematopoietic stem cells from type I interferon-dependent exhaustion.
Nat Med
2009
, vol. 
15
 
6
(pg. 
696
-
700
)
21
Ehninger
 
A
Boch
 
T
Uckelmann
 
H
, et al. 
Posttranscriptional regulation of c-Myc expression in adult murine HSCs during homeostasis and interferon-α-induced stress response.
Blood
2014
, vol. 
123
 
25
(pg. 
3909
-
3913
)
22
Feng
 
CG
Weksberg
 
DC
Taylor
 
GA
Sher
 
A
Goodell
 
MA
The p47 GTPase Lrg-47 (Irgm1) links host defense and hematopoietic stem cell proliferation.
Cell Stem Cell
2008
, vol. 
2
 
1
(pg. 
83
-
89
)
23
Hartner
 
JC
Walkley
 
CR
Lu
 
J
Orkin
 
SH
ADAR1 is essential for the maintenance of hematopoiesis and suppression of interferon signaling.
Nat Immunol
2009
, vol. 
10
 
1
(pg. 
109
-
115
)
24
MacNamara
 
KC
Jones
 
M
Martin
 
O
Winslow
 
GM
Transient activation of hematopoietic stem and progenitor cells by IFNγ during acute bacterial infection.
PLoS ONE
2011
, vol. 
6
 
12
pg. 
e28669
 
25
Singh
 
P
Yao
 
Y
Weliver
 
A
Broxmeyer
 
HE
Hong
 
SC
Chang
 
CH
Vaccinia virus infection modulates the hematopoietic cell compartments in the bone marrow.
Stem Cells
2008
, vol. 
26
 
4
(pg. 
1009
-
1016
)
26
Zhang
 
Y
Jones
 
M
McCabe
 
A
Winslow
 
GM
Avram
 
D
MacNamara
 
KC
MyD88 signaling in CD4 T cells promotes IFN-γ production and hematopoietic progenitor cell expansion in response to intracellular bacterial infection.
J Immunol
2013
, vol. 
190
 
9
(pg. 
4725
-
4735
)
27
Zhao
 
X
Ren
 
G
Liang
 
L
, et al. 
Brief report: interferon-gamma induces expansion of Lin(-)Sca-1(+)C-Kit(+) Cells.
Stem Cells
2010
, vol. 
28
 
1
(pg. 
122
-
126
)
28
Snapper
 
CM
Yamaguchi
 
H
Urban
 
JF
Finkelman
 
FD
Induction of Ly-6A/E expression by murine lymphocytes after in vivo immunization is strictly dependent upon the action of IFN-alpha/beta and/or IFN-gamma.
Int Immunol
1991
, vol. 
3
 
9
(pg. 
845
-
852
)
29
Murray
 
PJ
Young
 
RA
Daley
 
GQ
Hematopoietic remodeling in interferon-gamma-deficient mice infected with mycobacteria.
Blood
1998
, vol. 
91
 
8
(pg. 
2914
-
2924
)
30
Pearl
 
JE
Saunders
 
B
Ehlers
 
S
Orme
 
IM
Cooper
 
AM
Inflammation and lymphocyte activation during mycobacterial infection in the interferon-gamma-deficient mouse.
Cell Immunol
2001
, vol. 
211
 
1
(pg. 
43
-
50
)
31
Norose
 
K
Naoi
 
K
Fang
 
H
Yano
 
A
In vivo study of toxoplasmic parasitemia using interferon-gamma-deficient mice: absolute cell number of leukocytes, parasite load and cell susceptibility.
Parasitol Int
2008
, vol. 
57
 
4
(pg. 
447
-
453
)
32
de Bruin
 
AM
Libregts
 
SF
Valkhof
 
M
Boon
 
L
Touw
 
IP
Nolte
 
MA
IFNγ induces monopoiesis and inhibits neutrophil development during inflammation.
Blood
2012
, vol. 
119
 
6
(pg. 
1543
-
1554
)
33
MacNamara
 
KC
Oduro
 
K
Martin
 
O
, et al. 
Infection-induced myelopoiesis during intracellular bacterial infection is critically dependent upon IFN-γ signaling.
J Immunol
2011
, vol. 
186
 
2
(pg. 
1032
-
1043
)
34
Zhang
 
H
Nguyen-Jackson
 
H
Panopoulos
 
AD
Li
 
HS
Murray
 
PJ
Watowich
 
SS
STAT3 controls myeloid progenitor growth during emergency granulopoiesis.
Blood
2010
, vol. 
116
 
14
(pg. 
2462
-
2471
)
35
Snoeck
 
HW
Lardon
 
F
Lenjou
 
M
Nys
 
G
Van Bockstaele
 
DR
Peetermans
 
ME
Interferon-gamma and interleukin-4 reciprocally regulate the production of monocytes/macrophages and neutrophils through a direct effect on committed monopotential bone marrow progenitor cells.
Eur J Immunol
1993
, vol. 
23
 
5
(pg. 
1072
-
1077
)
36
Koning
 
JJ
Kooij
 
G
de Vries
 
HE
Nolte
 
MA
Mebius
 
RE
Mesenchymal stem cells are mobilized from the bone marrow during inflammation.
Front Immunol
2013
, vol. 
4
 pg. 
49
 
37
Ren
 
G
Zhang
 
L
Zhao
 
X
, et al. 
Mesenchymal stem cell-mediated immunosuppression occurs via concerted action of chemokines and nitric oxide.
Cell Stem Cell
2008
, vol. 
2
 
2
(pg. 
141
-
150
)
38
Meisel
 
R
Zibert
 
A
Laryea
 
M
Göbel
 
U
Däubener
 
W
Dilloo
 
D
Human bone marrow stromal cells inhibit allogeneic T-cell responses by indoleamine 2,3-dioxygenase-mediated tryptophan degradation.
Blood
2004
, vol. 
103
 
12
(pg. 
4619
-
4621
)
39
François
 
M
Romieu-Mourez
 
R
Li
 
M
Galipeau
 
J
Human MSC suppression correlates with cytokine induction of indoleamine 2,3-dioxygenase and bystander M2 macrophage differentiation.
Mol Ther
2012
, vol. 
20
 
1
(pg. 
187
-
195
)
40
Boelen
 
A
Kwakkel
 
J
Barends
 
M
de Rond
 
L
Dormans
 
J
Kimman
 
T
Effect of lack of interleukin-4, interleukin-12, interleukin-18, or the interferon-gamma receptor on virus replication, cytokine response, and lung pathology during respiratory syncytial virus infection in mice.
J Med Virol
2002
, vol. 
66
 
4
(pg. 
552
-
560
)
41
Hausmann
 
J
Pagenstecher
 
A
Baur
 
K
Richter
 
K
Rziha
 
HJ
Staeheli
 
P
CD8 T cells require gamma interferon to clear borna disease virus from the brain and prevent immune system-mediated neuronal damage.
J Virol
2005
, vol. 
79
 
21
(pg. 
13509
-
13518
)
42
Rais
 
M
Wild
 
JS
Choudhury
 
BK
, et al. 
Interleukin-12 inhibits eosinophil differentiation from bone marrow stem cells in an interferon-gamma-dependent manner in a mouse model of asthma.
Clin Exp Allergy
2002
, vol. 
32
 
4
(pg. 
627
-
632
)
43
Finkelman
 
FD
Madden
 
KB
Cheever
 
AW
, et al. 
Effects of interleukin 12 on immune responses and host protection in mice infected with intestinal nematode parasites.
J Exp Med
1994
, vol. 
179
 
5
(pg. 
1563
-
1572
)
44
de Bruin
 
AM
Buitenhuis
 
M
van der Sluijs
 
KF
van Gisbergen
 
KP
Boon
 
L
Nolte
 
MA
Eosinophil differentiation in the bone marrow is inhibited by T cell-derived IFN-gamma.
Blood
2010
, vol. 
116
 
14
(pg. 
2559
-
2569
)
45
Belyaev
 
NN
Brown
 
DE
Diaz
 
AI
, et al. 
Induction of an IL7-R(+)c-Kit(hi) myelolymphoid progenitor critically dependent on IFN-gamma signaling during acute malaria.
Nat Immunol
2010
, vol. 
11
 
6
(pg. 
477
-
485
)
46
Weiss
 
G
Iron metabolism in the anemia of chronic disease.
Biochim Biophys Acta
2009
, vol. 
1790
 
7
(pg. 
682
-
693
)
47
Zoller
 
EE
Lykens
 
JE
Terrell
 
CE
, et al. 
Hemophagocytosis causes a consumptive anemia of inflammation.
J Exp Med
2011
, vol. 
208
 
6
(pg. 
1203
-
1214
)
48
Raefsky
 
EL
Platanias
 
LC
Zoumbos
 
NC
Young
 
NS
Studies of interferon as a regulator of hematopoietic cell proliferation.
J Immunol
1985
, vol. 
135
 
4
(pg. 
2507
-
2512
)
49
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
)
50
Wang
 
CQ
Udupa
 
KB
Lipschitz
 
DA
Interferon-gamma exerts its negative regulatory effect primarily on the earliest stages of murine erythroid progenitor cell development.
J Cell Physiol
1995
, vol. 
162
 
1
(pg. 
134
-
138
)
51
Mullarky
 
IK
Szaba
 
FM
Kummer
 
LW
, et al. 
Gamma interferon suppresses erythropoiesis via interleukin-15.
Infect Immun
2007
, vol. 
75
 
5
(pg. 
2630
-
2633
)
52
Libregts
 
SF
Gutiérrez
 
L
de Bruin
 
AM
, et al. 
Chronic IFN-γ production in mice induces anemia by reducing erythrocyte life span and inhibiting erythropoiesis through an IRF-1/PU.1 axis.
Blood
2011
, vol. 
118
 
9
(pg. 
2578
-
2588
)
53
Rekhtman
 
N
Radparvar
 
F
Evans
 
T
Skoultchi
 
AI
Direct interaction of hematopoietic transcription factors PU.1 and GATA-1: functional antagonism in erythroid cells.
Genes Dev
1999
, vol. 
13
 
11
(pg. 
1398
-
1411
)
54
Tsuji
 
K
Muraoka
 
K
Nakahata
 
T
Interferon-gamma and human megakaryopoiesis.
Leuk Lymphoma
1998
, vol. 
31
 
1-2
(pg. 
107
-
113
)
55
Huang
 
Z
Richmond
 
TD
Muntean
 
AG
Barber
 
DL
Weiss
 
MJ
Crispino
 
JD
STAT1 promotes megakaryopoiesis downstream of GATA-1 in mice.
J Clin Invest
2007
, vol. 
117
 
12
(pg. 
3890
-
3899
)
56
Masumi
 
A
Hamaguchi
 
I
Kuramitsu
 
M
, et al. 
Interferon regulatory factor-2 induces megakaryopoiesis in mouse bone marrow hematopoietic cells.
FEBS Lett
2009
, vol. 
583
 
21
(pg. 
3493
-
3500
)
57
Yamada
 
G
Ogawa
 
M
Akagi
 
K
, et al. 
Specific depletion of the B-cell population induced by aberrant expression of human interferon regulatory factor 1 gene in transgenic mice.
Proc Natl Acad Sci USA
1991
, vol. 
88
 
2
(pg. 
532
-
536
)
58
Marine
 
JC
Topham
 
DJ
McKay
 
C
, et al. 
SOCS1 deficiency causes a lymphocyte-dependent perinatal lethality.
Cell
1999
, vol. 
98
 
5
(pg. 
609
-
616
)
59
Arens
 
R
Tesselaar
 
K
Baars
 
PA
, et al. 
Constitutive CD27/CD70 interaction induces expansion of effector-type T cells and results in IFNgamma-mediated B cell depletion.
Immunity
2001
, vol. 
15
 
5
(pg. 
801
-
812
)
60
Garvy
 
BA
Riley
 
RL
IFN-gamma abrogates IL-7-dependent proliferation in pre-B cells, coinciding with onset of apoptosis.
Immunology
1994
, vol. 
81
 
3
(pg. 
381
-
388
)
61
Grawunder
 
U
Melchers
 
F
Rolink
 
A
Interferon-gamma arrests proliferation and causes apoptosis in stromal cell/interleukin-7-dependent normal murine pre-B cell lines and clones in vitro, but does not induce differentiation to surface immunoglobulin-positive B cells.
Eur J Immunol
1993
, vol. 
23
 
2
(pg. 
544
-
551
)
62
Corfe
 
SA
Rottapel
 
R
Paige
 
CJ
Modulation of IL-7 thresholds by SOCS proteins in developing B lineage cells.
J Immunol
2011
, vol. 
187
 
7
(pg. 
3499
-
3510
)
63
DeKoter
 
RP
Singh
 
H
Regulation of B lymphocyte and macrophage development by graded expression of PU.1.
Science
2000
, vol. 
288
 
5470
(pg. 
1439
-
1441
)
64
Krebs
 
DL
Hilton
 
DJ
SOCS proteins: negative regulators of cytokine signaling.
Stem Cells
2001
, vol. 
19
 
5
(pg. 
378
-
387
)
65
Sasaki
 
A
Yasukawa
 
H
Shouda
 
T
Kitamura
 
T
Dikic
 
I
Yoshimura
 
A
CIS3/SOCS-3 suppresses erythropoietin (EPO) signaling by binding the EPO receptor and JAK2.
J Biol Chem
2000
, vol. 
275
 
38
(pg. 
29338
-
29347
)
66
Iwasaki
 
H
Mizuno
 
S
Mayfield
 
R
, et al. 
Identification of eosinophil lineage-committed progenitors in the murine bone marrow.
J Exp Med
2005
, vol. 
201
 
12
(pg. 
1891
-
1897
)
67
Zoumbos
 
NC
Gascon
 
P
Djeu
 
JY
Young
 
NS
Interferon is a mediator of hematopoietic suppression in aplastic anemia in vitro and possibly in vivo.
Proc Natl Acad Sci USA
1985
, vol. 
82
 
1
(pg. 
188
-
192
)
68
Risitano
 
AM
Maciejewski
 
JP
Green
 
S
Plasilova
 
M
Zeng
 
W
Young
 
NS
In-vivo dominant immune responses in aplastic anaemia: molecular tracking of putatively pathogenetic T-cell clones by TCR beta-CDR3 sequencing.
Lancet
2004
, vol. 
364
 
9431
(pg. 
355
-
364
)
69
Sloand
 
E
Kim
 
S
Maciejewski
 
JP
Tisdale
 
J
Follmann
 
D
Young
 
NS
Intracellular interferon-gamma in circulating and marrow T cells detected by flow cytometry and the response to immunosuppressive therapy in patients with aplastic anemia.
Blood
2002
, vol. 
100
 
4
(pg. 
1185
-
1191
)
70
Solomou
 
EE
Keyvanfar
 
K
Young
 
NS
T-bet, a Th1 transcription factor, is up-regulated in T cells from patients with aplastic anemia.
Blood
2006
, vol. 
107
 
10
(pg. 
3983
-
3991
)
71
Dufour
 
C
Capasso
 
M
Svahn
 
J
, et al. 
Associazione Italiana di Emato-Oncologia Pediatrica (AIEOP); Department of Hematology, Ospedale S. Martino, Genoa, Italy
Homozygosis for (12) CA repeats in the first intron of the human IFN-gamma gene is significantly associated with the risk of aplastic anaemia in Caucasian population.
Br J Haematol
2004
, vol. 
126
 
5
(pg. 
682
-
685
)
72
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
)
73
Sato
 
T
Selleri
 
C
Anderson
 
S
Young
 
NS
Maciejewski
 
JP
Expression and modulation of cellular receptors for interferon-gamma, tumour necrosis factor, and Fas on human bone marrow CD34+ cells.
Br J Haematol
1997
, vol. 
97
 
2
(pg. 
356
-
365
)
74
Young
 
NS
Scheinberg
 
P
Calado
 
RT
Aplastic anemia.
Curr Opin Hematol
2008
, vol. 
15
 
3
(pg. 
162
-
168
)
75
Selleri
 
C
Maciejewski
 
JP
Catalano
 
L
, et al. 
Effects of cyclosporine on hematopoietic and immune functions in patients with hypoplastic myelodysplasia: in vitro and in vivo studies.
Cancer
2002
, vol. 
95
 
9
(pg. 
1911
-
1922
)
76
Fuchs
 
D
Zangerle
 
R
Artner-Dworzak
 
E
, et al. 
Association between immune activation, changes of iron metabolism and anaemia in patients with HIV infection.
Eur J Haematol
1993
, vol. 
50
 
2
(pg. 
90
-
94
)
77
Dufour
 
C
Corcione
 
A
Svahn
 
J
, et al. 
TNF-alpha and IFN-gamma are overexpressed in the bone marrow of Fanconi anemia patients and TNF-alpha suppresses erythropoiesis in vitro.
Blood
2003
, vol. 
102
 
6
(pg. 
2053
-
2059
)
78
Welniak
 
LA
Blazar
 
BR
Anver
 
MR
Wiltrout
 
RH
Murphy
 
WJ
Opposing roles of interferon-gamma on CD4+ T cell-mediated graft-versus-host disease: effects of conditioning.
Biol Blood Marrow Transplant
2000
, vol. 
6
 
6
(pg. 
604
-
612
)
79
Chewning
 
JH
Zhang
 
W
Randolph
 
DA
Swindle
 
CS
Schoeb
 
TR
Weaver
 
CT
Allogeneic Th1 cells home to host bone marrow and spleen and mediate IFNγ-dependent aplasia.
Biol Blood Marrow Transplant
2013
, vol. 
19
 
6
(pg. 
876
-
887
)
80
Laver
 
J
Castro-Malaspina
 
H
Kernan
 
NA
, et al. 
In vitro interferon-gamma production by cultured T-cells in severe aplastic anaemia: correlation with granulomonopoietic inhibition in patients who respond to anti-thymocyte globulin.
Br J Haematol
1988
, vol. 
69
 
4
(pg. 
545
-
550
)
81
Bloom
 
ML
Wolk
 
AG
Simon-Stoos
 
KL
Bard
 
JS
Chen
 
J
Young
 
NS
A mouse model of lymphocyte infusion-induced bone marrow failure.
Exp Hematol
2004
, vol. 
32
 
12
(pg. 
1163
-
1172
)
82
Skurkovich
 
S
Skurkovich
 
B
Anticytokine therapy, especially anti-interferon-gamma, as a pathogenetic treatment in TH-1 autoimmune diseases.
Ann N Y Acad Sci
2005
, vol. 
1051
 (pg. 
684
-
700
)
83
Takanashi
 
M
Motoji
 
T
Oshimi
 
K
Mizoguchi
 
H
Effect of recombinant interferons on colony formation of blast progenitors in acute myeloblastic leukemia.
Exp Hematol
1987
, vol. 
15
 
9
(pg. 
946
-
951
)
84
McGlave
 
P
Mamus
 
S
Vilen
 
B
Dewald
 
G
Effect of recombinant gamma interferon on chronic myelogenous leukemia bone marrow progenitors.
Exp Hematol
1987
, vol. 
15
 
4
(pg. 
331
-
335
)
85
Portier
 
M
Zhang
 
XG
Caron
 
E
Lu
 
ZY
Bataille
 
R
Klein
 
B
gamma-Interferon in multiple myeloma: inhibition of interleukin-6 (IL-6)-dependent myeloma cell growth and downregulation of IL-6-receptor expression in vitro.
Blood
1993
, vol. 
81
 
11
(pg. 
3076
-
3082
)
86
Geissler
 
K
Tricot
 
G
Leemhuis
 
T
Walker
 
E
Broxmeyer
 
HE
Differentiation-inducing effect of recombinant human tumor necrosis factor alpha and gamma-interferon in vitro on blast cells from patients with acute myeloid leukemia and myeloid blast crisis of chronic myeloid leukemia.
Cancer Res
1989
, vol. 
49
 
11
(pg. 
3057
-
3062
)
87
Murohashi
 
I
Hoang
 
T
Interferon-gamma enhances growth factor-dependent proliferation of clonogenic cells in acute myeloblastic leukemia.
Blood
1991
, vol. 
78
 
4
(pg. 
1085
-
1095
)
88
Ersvaer
 
E
Skavland
 
J
Ulvestad
 
E
Gjertsen
 
BT
Bruserud
 
Ø
Effects of interferon gamma on native human acute myelogenous leukaemia cells.
Cancer Immunol Immunother
2007
, vol. 
56
 
1
(pg. 
13
-
24
)
89
Reddy
 
PN
Sargin
 
B
Choudhary
 
C
, et al. 
Study Alliance Leukemia (SAL)
SOCS1 cooperates with FLT3-ITD in the development of myeloproliferative disease by promoting the escape from external cytokine control.
Blood
2012
, vol. 
120
 
8
(pg. 
1691
-
1702
)
90
Spets
 
H
Georgii-Hemming
 
P
Siljason
 
J
Nilsson
 
K
Jernberg-Wiklund
 
H
Fas/APO-1 (CD95)-mediated apoptosis is activated by interferon-gamma and interferon- in interleukin-6 (IL-6)-dependent and IL-6-independent multiple myeloma cell lines.
Blood
1998
, vol. 
92
 
8
(pg. 
2914
-
2923
)
91
Berthon
 
C
Driss
 
V
Liu
 
J
, et al. 
In acute myeloid leukemia, B7-H1 (PD-L1) protection of blasts from cytotoxic T cells is induced by TLR ligands and interferon-gamma and can be reversed using MEK inhibitors.
Cancer Immunol Immunother
2010
, vol. 
59
 
12
(pg. 
1839
-
1849
)
92
Schürch
 
C
Riether
 
C
Amrein
 
MA
Ochsenbein
 
AF
Cytotoxic T cells induce proliferation of chronic myeloid leukemia stem cells by secreting interferon-γ.
J Exp Med
2013
, vol. 
210
 
3
(pg. 
605
-
621
)
93
Liu
 
J
Hamrouni
 
A
Wolowiec
 
D
, et al. 
Plasma cells from multiple myeloma patients express B7-H1 (PD-L1) and increase expression after stimulation with IFN-gamma and TLR ligands via a MyD88-, TRAF6-, and MEK-dependent pathway.
Blood
2007
, vol. 
110
 
1
(pg. 
296
-
304
)
94
Mumprecht
 
S
Schürch
 
C
Schwaller
 
J
Solenthaler
 
M
Ochsenbein
 
AF
Programmed death 1 signaling on chronic myeloid leukemia-specific T cells results in T-cell exhaustion and disease progression.
Blood
2009
, vol. 
114
 
8
(pg. 
1528
-
1536
)
95
Folgiero
 
V
Goffredo
 
BM
Filippini
 
P
, et al. 
Indoleamine 2,3-dioxygenase 1 (IDO1) activity in leukemia blasts correlates with poor outcome in childhood acute myeloid leukemia.
Oncotarget
2014
 
5(8):2052-2064
96
Pfeifer
 
S
Schreder
 
M
Bolomsky
 
A
, et al. 
Induction of indoleamine-2,3 dioxygenase in bone marrow stromal cells inhibits myeloma cell growth.
J Cancer Res Clin Oncol
2012
, vol. 
138
 
11
(pg. 
1821
-
1830
)
97
Mills
 
EL
Viral infections predisposing to bacterial infections.
Annu Rev Med
1984
, vol. 
35
 (pg. 
469
-
479
)
Sign in via your Institution