The bone marrow (BM) is responsible for generating and maintaining lifelong output of blood and immune cells. In addition to its key hematopoietic function, the BM acts as an important lymphoid organ, hosting a large variety of mature lymphocyte populations, including B cells, T cells, natural killer T cells, and innate lymphoid cells. Many of these cell types are thought to visit the BM only transiently, but for others, like plasma cells and memory T cells, the BM provides supportive niches that promote their long-term survival. Interestingly, accumulating evidence points toward an important role for mature lymphocytes in the regulation of hematopoietic stem cells (HSCs) and hematopoiesis in health and disease. In this review, we describe the diversity, migration, localization, and function of mature lymphocyte populations in murine and human BM, focusing on their role in immunity and hematopoiesis. We also address how various BM lymphocyte subsets contribute to the development of aplastic anemia and immune thrombocytopenia, illustrating the complexity of these BM disorders and the underlying similarities and differences in their disease pathophysiology. Finally, we summarize the interactions between mature lymphocytes and BM resident cells in HSC transplantation and graft-versus-host disease. A better understanding of the mechanisms by which mature lymphocyte populations regulate BM function will likely improve future therapies for patients with benign and malignant hematologic disorders.

Hematopoietic stem cells (HSCs) reside in specialized bone marrow (BM) niches that regulate their maintenance to ensure lifelong blood cell production. Numerous cell types contribute to the formation and function of such niches, which exist in various compositions and locations and support diverse functional states of HSCs and hematopoietic progenitors. Niches provide anchorage and survival signals, regulate HSC quiescence, self-renewal, differentiation, proliferation, homing, and mobilization, and modulate BM output.1  Sinusoidal endothelial cells and perivascular C-X-C motif chemokine ligand 12 (CXCL12)-abundant reticular (CAR) cells are important constituents of HSC niches, but they also constitute a topologically complex network to which many other BM cells connect2,3  (Figure 1). In addition to classical niche cells that have been identified, it has become clear that mature hematopoietic cells also regulate hematopoiesis through various mechanisms. Lymphocytes are of particular interest, because their motile behavior and dedicated role in immunity enables them to transfer information from the periphery back to the BM. Many mature lymphocyte subsets can enter the BM, and an attractive hypothesis is that these cells provide feedback about an ongoing infection, which enables a tailored hematopoietic response against the invading pathogen. Resting BM T cells already produce HSC-supportive cytokines,4,5  whereas their transient activation boosts the production of cytokines that skew hematopoietic differentiation.6  However, prolonged T-cell activation suppresses hematopoiesis, causing the development of BM failure diseases such as aplastic anemia (AA) by impairing HSC maintenance.7 

Figure 1.

Localization and function of mature lymphocytes in the BM. Sinusoidal endothelial cells and perivascular C-X-C motif chemokine ligand 12 (CXCL12)-abundant reticular (CAR) cells are important constituents of HSC niches and form a topologically complex network to which many other BM cells connect. (A) Plasmablasts (PB) colocalize with perisinusoidal CAR cells. Here, they receive important survival signals such as B lymphocyte stimulator (BLyS), interleukin-6 (IL-6), and a proliferation-inducing ligand (APRIL), which enable them to differentiate into long-lived immunoglobulin-secreting plasma cells (PCs). Plasma cells are closely associated with BM regulatory T cells (TREGS), dendritic cells (DCs), and eosinophils (Eos). During various clinical conditions, the BM can contain germinal centers, complete with follicular DCs (FDCs) and T follicular helper (TFH) cells. The BM contains significant numbers of memory B cells (BMEM), the majority of which are noncirculating BM resident cells that dock onto VCAM-1+ stromal cells. (B) Scattered naïve CD8+ T cells (CD8+ Tnaïve) cluster at a 10:1 ratio around antigen-presenting cells (APCs) presenting their cognate antigen. Both naïve and memory CD8+ T cells (CD8+ TMEM) locate close to the sinusoidal vessel and perivascular VCAM-1+ stromal cells that support their survival and maintenance by producing IL-7 and IL-15. Memory CD8+ T cells produce tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ), which can regulate or disrupt HSC maintenance, depending on the amount produced. Memory CD4+ T cells (CD4+ TMEM) colocalize with VCAM-1+ stromal cells, which also produce IL-7. They secrete cytokines necessary for HSC maintenance, such as IL-3 and granulocyte-macrophage colony-stimulating factor (GM-CSF). Tregs preserve normal hematopoiesis because of their T-cell suppressive function and possibly because of their IL-10 secretion. CD150hi Tregs and CD4+ T cells localize near HSCs in the perivascular niche and provide immune privilege. Extracellular adenosine generated via CD39 expressed on those lymphocyte subsets maintains HSC quiescence. (C-D) The BM also contains mature γδ T cells, natural killer (NK) cells, NK T cells (NKT cells), and innate lymphoid cells (ILCs), but little is known about their precise localizations and functions. (C) BM NKT cells can be activated by CD1d-expressing HSCs and enhance myelopoiesis by secreting GM-CSF. BM NKT cells also secrete IFN-γ, IL-10, and IL-4, suggesting that they regulate hematopoiesis. Whether γδ T cells regulate hematopoiesis, as has been shown for αβ T cells, is not known. (D) The presence of ILC2s and ILC3s in the BM has been reported for mice and humans, respectively. ILC3s in human BM may be harbored in a stromal cell niche, but systematic analyses of the presence, localization, and function of mature ILCs in the BM are still lacking. In humans and mice, BM NK cells are a major source of cytokines that can regulate hematopoiesis, including IFN-γ, TNF-α, and GM-CSF. In a mouse Toxoplasma gondii infection model, BM NK cells triggered by systemic IL-12 produced IFN-γ, promoting an immune regulatory program in monocytes (Mono).

Figure 1.

Localization and function of mature lymphocytes in the BM. Sinusoidal endothelial cells and perivascular C-X-C motif chemokine ligand 12 (CXCL12)-abundant reticular (CAR) cells are important constituents of HSC niches and form a topologically complex network to which many other BM cells connect. (A) Plasmablasts (PB) colocalize with perisinusoidal CAR cells. Here, they receive important survival signals such as B lymphocyte stimulator (BLyS), interleukin-6 (IL-6), and a proliferation-inducing ligand (APRIL), which enable them to differentiate into long-lived immunoglobulin-secreting plasma cells (PCs). Plasma cells are closely associated with BM regulatory T cells (TREGS), dendritic cells (DCs), and eosinophils (Eos). During various clinical conditions, the BM can contain germinal centers, complete with follicular DCs (FDCs) and T follicular helper (TFH) cells. The BM contains significant numbers of memory B cells (BMEM), the majority of which are noncirculating BM resident cells that dock onto VCAM-1+ stromal cells. (B) Scattered naïve CD8+ T cells (CD8+ Tnaïve) cluster at a 10:1 ratio around antigen-presenting cells (APCs) presenting their cognate antigen. Both naïve and memory CD8+ T cells (CD8+ TMEM) locate close to the sinusoidal vessel and perivascular VCAM-1+ stromal cells that support their survival and maintenance by producing IL-7 and IL-15. Memory CD8+ T cells produce tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ), which can regulate or disrupt HSC maintenance, depending on the amount produced. Memory CD4+ T cells (CD4+ TMEM) colocalize with VCAM-1+ stromal cells, which also produce IL-7. They secrete cytokines necessary for HSC maintenance, such as IL-3 and granulocyte-macrophage colony-stimulating factor (GM-CSF). Tregs preserve normal hematopoiesis because of their T-cell suppressive function and possibly because of their IL-10 secretion. CD150hi Tregs and CD4+ T cells localize near HSCs in the perivascular niche and provide immune privilege. Extracellular adenosine generated via CD39 expressed on those lymphocyte subsets maintains HSC quiescence. (C-D) The BM also contains mature γδ T cells, natural killer (NK) cells, NK T cells (NKT cells), and innate lymphoid cells (ILCs), but little is known about their precise localizations and functions. (C) BM NKT cells can be activated by CD1d-expressing HSCs and enhance myelopoiesis by secreting GM-CSF. BM NKT cells also secrete IFN-γ, IL-10, and IL-4, suggesting that they regulate hematopoiesis. Whether γδ T cells regulate hematopoiesis, as has been shown for αβ T cells, is not known. (D) The presence of ILC2s and ILC3s in the BM has been reported for mice and humans, respectively. ILC3s in human BM may be harbored in a stromal cell niche, but systematic analyses of the presence, localization, and function of mature ILCs in the BM are still lacking. In humans and mice, BM NK cells are a major source of cytokines that can regulate hematopoiesis, including IFN-γ, TNF-α, and GM-CSF. In a mouse Toxoplasma gondii infection model, BM NK cells triggered by systemic IL-12 produced IFN-γ, promoting an immune regulatory program in monocytes (Mono).

Close modal

Here, we discuss the diversity, localization, and (patho)physiology of mature lymphocyte populations in the BM (supplemental Table 1, available on the Blood Web site). We focus on B cells, T cells, and innate lymphoid cell (ILC) populations and address their roles in homeostasis, upon immune challenge, and in non-neoplastic hematologic disorders, including AA, immune thrombocytopenia (ITP), HSC transplantation (HSCT), and graft-versus-host disease (GVHD). The role of lymphocytes in hematologic malignancies and antitumor therapies is important, but it has been thoroughly reviewed elsewhere8,9  and will not be covered here.

B cells and plasma cells

The BM acts as a primary lymphoid organ by accommodating B-cell development, and it also harbors the most differentiated B cells, the plasma cells. Long-lived plasma cells are the first and best described mature lymphocyte subset residing in the BM, and their accumulation makes the BM a major site of antibody production.10  Plasmablasts (plasma cell precursors that develop during a humoral immune response in secondary lymphoid tissues [SLTs]) express C-X-C motif chemokine receptor 3 (CXCR3) and CXCR4 and home to the BM via endothelial cells expressing CXCL9, CXCL10, and CXCL12.11  Upon BM entry, plasmablasts co-localize with CAR cells, where they receive important survival signals, such as interleukin-6 (IL-6), B lymphocyte stimulator (BLyS), and a proliferation-inducing ligand (APRIL), which enable their differentiation into long-lived plasma cells12-14  (Figure 1A). This process is important for long-term immunity after infection and vaccination and also for the pathophysiology of multiple myeloma.15  Interestingly, although plasma cells and HSCs are both highly responsive to CXCL12 and therefore reside close to CAR cells,16  there is, to our knowledge, no evidence for functional crosstalk between plasma cells and HSCs. In contrast, plasma cells are closely associated with BM regulatory T cells (Tregs), dendritic cells (DCs), and eosinophils, and depletion of Tregs significantly reduces BM plasma cell numbers17,18  (Figure 1A).

As with SLTs, B-cell activation and differentiation can also occur inside the BM.19  Microscopic analyses in autoimmune disorders, infections, lymphomas, and other malignancies revealed that BM can contain reactive germinal centers (GCs), complete with follicular DCs and surrounding T cells.20  This is supported by flow cytometric detection of GC B cells,21  and T follicular helper cells.22,23  Therefore, not only B-cell activation, but also subsequent affinity maturation, isotype switching, and plasmablast development, can occur in the BM24  (Figure 1A). Moreover, the BM contains significant numbers of memory B cells, which can rapidly differentiate into plasma cells upon local activation.25,26  Distinct subsets of BM memory B cells have recently been identified, the majority of which are noncirculating and resident to the BM, docking onto vascular cell adhesion molecule 1 (VCAM-1+) stromal cells.27  These observations indicate that the BM serves not only as the generative organ for B-cell development but also serves as an SLT with great importance to systemic humoral immunity.

CD8+ T cells

Next to B cells and plasma cells, a large part of the mature lymphocyte pool in BM consists of T cells (supplemental Table 1). The percentage of CD8+ T cells is higher than the percentage of CD4+ T cells, which is different from the ratios in peripheral blood (PB) and SLTs (CD4:CD8 ratios in BM and PB are 0.7 and 2.0, respectively).28,29  In immunologically unchallenged mice, a large fraction (∼50% to 65%) of BM CD8+ T cells are naïve, whereas the remainder are central memory and effector memory cells.30,31  Importantly, this composition can change substantially upon immunologic challenge32,33 ; the frequency of effector memory CD8+ T cells in murine BM was shown to increase from ∼10% to ∼60% after recovery from lymphocytic choriomeningitis virus infection.30  In this model, virus-specific BM memory CD8+ T cells greatly outnumber those in the spleen, which is also a result of the large amount of BM distributed throughout the body.30 

Adoptively transferred naïve CD8+ T cells migrate not only to SLTs but also to BM where they maintain a naïve phenotype34  and cluster around antigen-presenting cells (APCs)31,34  (Figure 1B). Ex vivo isolated central memory CD8+ T cells also rapidly home to BM as do effector memory CD8+ T cells, albeit to a lesser extent.32  Entry of CD8+ T cells into the BM is dependent on CXCR4, which is highly expressed on both naïve and memory CD8+ T cells.35,36  Contrary to plasmablasts,10  memory CD8+ T cells do not depend on their high expression of CXCR3 to enter the BM, despite local production of CXCL9 and CXCL10.35  BM memory CD8+ T cells are fully functional and mount recall responses when transferred into naïve mice.37  BM naïve CD8+ T cells can be activated by BM-resident DCs,34  which again demonstrates that BM acts as SLT. Although the presented antigens may be taken up from the blood,31,34  it has also been shown that neutrophils can transport viral antigens from the dermis to the BM, resulting in polyclonal CD8+ T-cell priming via APCs.38  Interestingly, these BM-primed CD8+ T cells exhibited differential effector functions and gene-expression profiles compared with their SLT-primed counterparts.38  This is consistent with a study in humans showing that BM CD8+ T cells have a different homing phenotype than those in other organs and that the BM selectively accumulates certain virus-specific memory CD8+ T-cell populations.39 

Even though BM can serve as SLT, intravital imaging revealed that both naïve and memory CD8+ T cells are surprisingly immobile in the BM compared with SLT, and they interact with perivascular stromal cells that support their survival.35  This suggests that the BM is also important for maintaining CD8+ T cells in the absence of cognate antigen and that they temporarily reside in the BM to recharge on cytokines.40  Indeed, BM memory CD8+ T cells localize close to VCAM-1+ stromal cells, which produce high levels of IL-7, a cytokine critical for their survival.33,41  Moreover, these stromal niches also produce IL-15,35,42  which drives proliferation of memory CD8+ T cells43,44  (Figure 1B). Whether maintenance of BM memory CD8+ T cells depends on homeostatic proliferation or survival is heavily debated.45,46  Furthermore, although it is unclear how long these cells reside in the BM, most BM CD8+ T cells are in equilibrium with PB.47  However, a small pool of memory CD8+ T cells with specificities against a variety of peripheral and systemic antigens is truly resident and remains inside the BM.48  These data corroborate the notion that the BM is a key organ for maintenance of CD8+ T-cell–mediated immunologic memory. It should be noted that memory CD8+ T cells not only passively recharge in the BM but also produce soluble factors that enhance HSC self-renewal and maintenance5  (Figure 1B). This supportive function is altered when an infection induces CD8+ T-cell activation and production of interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α), promoting HSC differentiation toward mature immune cells that eliminate the pathogen.49,50 

CD4+ T cells

Naïve CD4+ T cells compose ∼60% of the total BM CD4+ T-cell pool,51  and like their CD8+ counterparts, they can be locally activated to generate robust immune responses.34  Not much is known about their localization, but they probably colocalize in clusters with local APCs like naïve CD8+ T cells.52  BM localization of memory CD4+ T cells has been studied in more detail; they are found in VCAM-1+IL-7+ stromal cell niches,53  similar to memory CD8+ T cells and B cells (Figure 1B). Importantly, BM CD4+ T cells do not homeostatically proliferate53  but are reactivated by cognate antigen, whereupon they proliferate and form clusters with B cells.54  These clusters are distinct from classical GCs, because the CD4+ T cells in them do not develop into T follicular helper cells, and the B cells do not acquire a GC phenotype.54 

When activated, CD4+ T cells can produce many hematopoiesis-regulating cytokines, such as IL-3, IL-4, IL-6, IL-17, granulocyte-macrophage colony-stimulating factor (GM-CSF), IFN-γ, and TNF-α55-57  (Figure 1B). BM CD4+ T cells have a key role in maintaining normal hematopoiesis, because T-cell–deficient mice show defective terminal differentiation of myeloid cells, leading to an accumulation of immature myeloid cells in the BM and peripheral neutropenia.58  Normal granulopoiesis is restored upon reconstitution with CD4+, but not CD8+, T cells and is dependent on the activation state of the CD4+ T cells.58  Experiments with T-cell–specific deficiency of signal transducer and activator of transcription 4 (STAT-4) or STAT-6 showed that effector CD4+ T cells can also actively regulate hematopoietic progenitor cell homeostasis, possibly by secreting oncostatin M.4 

The BM also contains a high fraction of Tregs compared with PB and SLTs, which make up ∼25% to 45% of total BM CD4+ T cells in humans and mice59-61  (supplemental Table 1). Tregs also depend on CXCR4/CXCL12 signaling for BM localization and are mobilized upon administration of granulocyte-CSF (G-CSF), which decreases BM CXCL12 levels.60,61  BM Tregs are important for preserving normal hematopoiesis,61,62  most likely because of their T-cell suppressive functions, and possibly because of their secretion of IL-10, which may stimulate HSC self-renewal.63 

γδ T cells

γδ T cells constitute a small percentage of circulating T cells that participate in immune surveillance and innate immune responses because they recognize foreign and stress-induced self-antigens without previous antigen exposure.64  Unlike αβ T cells, γδ T cells are activated independently of major histocompatibility complex (MHC); they recognize lipids presented on CD1-family molecules and butyrophilin-like ligands.65  They mainly localize to epithelial and mucosal tissues, with T-cell receptor (TCR) differentiation and adaptations unique to their tissue environment.66  Subsets of γδ T cells can also follow an αβ-like adaptive pathway, including delayed activation, expansion, and establishment of memory.67  The 2 main subsets of γδ T cells, Vδ1+ and Vδ2+, are found in the BM (Figure 1C). Vγ9+Vδ2+ cells usually participate in innate-like immune responses, whereas Vγ9Vδ1+ cells have been associated with adaptive-like responses.68,69  In murine BM, ∼2% of cells are γδ T cells.70  In human BM, ∼6% of T cells are γδ T cells, and the majority of these are Vγ9+Vδ2+, pointing to a high constitution of innate-like γδ T cells.71  Of these, 30% display a naïve, 30% a central memory, and 40% an effector memory phenotype72  (supplemental Table 1). Vγ9+Vδ2+ cells can act as innate effector cells and can kill tumor cells upon activation with aminobisphosphonates.73  Whether γδ T cells can also affect hematopoietic progenitor cells, as has been shown for αβ T cells, is unknown.

NKT cells

NKT cells are unconventional T cells that recognize glycolipids presented on CD1d. Type I NKT cells express an invariant TCR, whereas type II NKT cells express diverse TCRs. NKT cells lack CD8, but some cells do express CD4.74  Although NKT cells constitute only a minority of all BM T cells (supplemental Table 1), they can be activated by CD1d-expressing HSCs and modulate myelopoiesis by secreting GM-CSF and IFN-γ. NKT cell–deficient mice have significantly fewer leukocytes and platelets, lower BM cellularity, and fewer HSCs, suggesting that NKT cells positively regulate hematopoiesis75  (Figure 1C).

NK cells are cytotoxic lymphoid cells that recognize and lyse virus-infected and cancer cells that display distress signals and/or lack self-signals such as MHC-I and regulate immune responses through interactions with T cells, DCs, macrophages, and endothelial cells.76,77  NK cells are widely distributed throughout tissues in both humans and mice and constitute between 2% and 4% of the total lymphocytes in the BM77,78  (supplemental Table 1). BM NK cells are a major source of cytokines that regulate myelopoiesis, including IFN-γ and TNF-α.8  Upon gastrointestinal infection of mice with Toxoplasma gondii, mucosa-associated lymphoid tissue–resident DCs produce systemic levels of IL-12, which stimulate BM-resident NK cells to produce IFN-γ and form clusters with developing monocytes. This induces an immune regulatory program in the monocytes before their egress into the blood and recruitment to the site of infection79  (Figure 1D).

ILCs

The family of noncytotoxic (helper) ILCs comprises 3 subsets: ILC1s, ILC2s, and ILC3s.80  ILCs arise from HSCs via lymphoid-primed multipotent progenitors, common lymphoid progenitors, and different committed ILC precursors (ILCPs), such as α-lymphoid precursors.81,82 Cxcl12 deletion studies showed that lymphoid-primed multipotent progenitors, common lymphoid progenitors, and B-cell progenitors depend on and live in close proximity to CXCL12-secreting osteoblasts and perivascular stromal cells.83  In contrast, localization and niche requirements for more committed ILCPs and mature ILCs remain obscure. In adult humans, committed CD34+RORγt+ ILC3 progenitors are not found in the BM, but rather in tonsils and intestinal lamina propria, indicating that final maturation occurs at the definite site where ILC3s are required.84  This is consistent with recent findings that SLT-resident human ILCPs locally give rise to helper ILCs and cytotoxic NK cells.85  Importantly, these ILCPs were found in SLTs, but not in PB or BM. Yet, another study showed that certain ILCPs do circulate through PB and give rise to IFN-γ–producing ILC1s, IL-13–producing ILC2s, and IL-22–, but not IL-17–producing ILC3s within specific tissues.86  There are reports that ILC2s can be found in murine BM and ILC3s in human BM (supplemental Table 1), but systematic analyses of the presence, localization, and function of mature ILCs in the BM are still lacking (Figure 1D).

Non-neoplastic BM failure diseases are characterized by BM hypocellularity in ≥1 blood lineages and the resultant peripheral cytopenias. BM failure is found in several inherited syndromes, including Fanconi anemia, but it can also be acquired in diseases such as AA and ITP. In most patients with AA, immune-mediated injury to HSCs leads to BM hypocellularity in all 3 lineages, resulting in serious pancytopenia. HSCT is curative but is associated with substantial short- and long-term adverse effects; elderly patients are often ineligible because of comorbidities, and there is a worldwide shortage of HSC donors. Therefore, most patients with AA are treated with immunosuppressive therapies.7  In contrast to AA, ITP affects the megakaryocyte lineage only, leading to low platelet counts and the risk of bleeding. ITP is associated with polymorphisms in immunity-related genes, dysregulated T-cell responses to platelet surface antigens, and antiplatelet autoantibodies, resulting in increased platelet apoptosis and phagocytosis.87 

Mechanistically, chronic exposure of HSCs and hematopoietic progenitors to inflammatory cytokines is associated with impaired self-renewal, HSC loss, and BM failure,50,88  as seen in AA and ITP89,90  (Figure 2). Because there is no convincing evidence for the recognition of exogenous antigens, a prevailing theory is that AA is mediated by autoreactive T cells.91  Activated T cells preferentially infiltrate residual hematopoietic areas in otherwise hypocellular BM biopsies from patients with AA, suggesting site-directed homing of autoreactive T cells in the pathogenesis of AA.92  Patients with AA exhibit significantly higher frequencies of activated CD8+ T cells in their BM than do healthy people. Those cells express high levels of CX3CR1,93  which mediates leukocyte chemotaxis and adhesion. Indeed, patients with AA have significantly higher fractalkine (CX3CL1) levels in BM plasma, which promotes chemotaxis and recruitment of CX3CR1+ T cells to BM and contributes to AA pathogenesis.93  BM T cells from patients with AA also express elevated levels of TNF-α and IFN-γ,89  which impedes HSC survival by disrupting suppressor of cytokine signaling 2 (SOCS2) and STAT pathways,94  impairing thrombopoietin-MPL signaling,88,95  and promoting apoptosis by upregulating Fas expression.96  CD8+ T cells also use perforin- and granzyme-mediated mechanisms to directly target HSCs97  (Figure 2B). Flow cytometry, spectratyping of complementarity-determining region 3 length skewing, and TCR Vβ sequencing revealed oligoclonal expansions of dysregulated effector-memory CD8+ T cells, suggesting that they respond to an HSC-expressed antigen.91,98 

Figure 2.

Pathophysiology of mature BM lymphocytes in autoimmune cytopenias. (A) In healthy BM, mature lymphocyte populations regulate HSC self-renewal, maintenance and hematopoietic output by producing soluble factors and likely by (as yet undefined) direct cell-cell interactions. (B-C) In AA and ITP, this delicate regulatory balance is distorted. For example, chronic exposure to high levels of inflammatory cytokines results in HSC loss, as seen in AA, and megakaryocyte (Mk) malfunction, as seen in ITP. (B) In AA, the patients’ BM is infiltrated by activated autoreactive (auto-)CD8+ T cells that express high levels of CX3CR1 and are recruited by CX3CL1 present in AA BM. BM auto-CD8+ T cells secrete high amounts of TNF-α and IFN-γ, and also use perforin- and granzyme-mediated mechanisms to directly target HSCs. Polyclonal BM auto-CD4+ T-cell numbers are increased in AA, and Th17-cell expansion is characteristic. Elevated levels of IL-17 impair hematopoiesis by depleting Tregs, directly inhibiting HSCs, and recruiting additional Th1 cells. IL-17 also promotes the secretion of TNF-α by macrophages (Mϕs), which in turn stimulates IFN-γ secretion by T cells. Patients with AA have reduced NK- and NKT-cell numbers in their BM. Whereas the significance of this finding for NKT cells is unclear, the reduction of BM NK cells in patients with AA may contribute to the excess of auto-CD8+ T cells as result of decreased T-cell killing by NK cells. (C) In patients with ITP, low platelet (plt) numbers are the predominant clinical feature. Presumably, BM auto-CD8+ T cells recognize megakaryocyte-derived antigens presented on platelets via MHC-I and can lyse platelets directly or induce platelet apoptosis. Moreover, suppression of megakaryocyte apoptosis by BM auto-CD8+ T cells directly inhibits platelet formation. BM auto-CD4+ T cells induce the production of anti-platelet autoantibodies (anti-plt abs) involved in disease pathogenesis. Patients with ITP have lower total numbers of CD4+ T cells in the BM than healthy controls. The disproportional loss of BM Th2 cells, as well as Tregs, results in a Th1 shift that contributes to autoantibody-mediated platelet destruction and T-cell cytotoxicity. The localizations and functions of BM NK and NKT cells in ITP are largely unknown.

Figure 2.

Pathophysiology of mature BM lymphocytes in autoimmune cytopenias. (A) In healthy BM, mature lymphocyte populations regulate HSC self-renewal, maintenance and hematopoietic output by producing soluble factors and likely by (as yet undefined) direct cell-cell interactions. (B-C) In AA and ITP, this delicate regulatory balance is distorted. For example, chronic exposure to high levels of inflammatory cytokines results in HSC loss, as seen in AA, and megakaryocyte (Mk) malfunction, as seen in ITP. (B) In AA, the patients’ BM is infiltrated by activated autoreactive (auto-)CD8+ T cells that express high levels of CX3CR1 and are recruited by CX3CL1 present in AA BM. BM auto-CD8+ T cells secrete high amounts of TNF-α and IFN-γ, and also use perforin- and granzyme-mediated mechanisms to directly target HSCs. Polyclonal BM auto-CD4+ T-cell numbers are increased in AA, and Th17-cell expansion is characteristic. Elevated levels of IL-17 impair hematopoiesis by depleting Tregs, directly inhibiting HSCs, and recruiting additional Th1 cells. IL-17 also promotes the secretion of TNF-α by macrophages (Mϕs), which in turn stimulates IFN-γ secretion by T cells. Patients with AA have reduced NK- and NKT-cell numbers in their BM. Whereas the significance of this finding for NKT cells is unclear, the reduction of BM NK cells in patients with AA may contribute to the excess of auto-CD8+ T cells as result of decreased T-cell killing by NK cells. (C) In patients with ITP, low platelet (plt) numbers are the predominant clinical feature. Presumably, BM auto-CD8+ T cells recognize megakaryocyte-derived antigens presented on platelets via MHC-I and can lyse platelets directly or induce platelet apoptosis. Moreover, suppression of megakaryocyte apoptosis by BM auto-CD8+ T cells directly inhibits platelet formation. BM auto-CD4+ T cells induce the production of anti-platelet autoantibodies (anti-plt abs) involved in disease pathogenesis. Patients with ITP have lower total numbers of CD4+ T cells in the BM than healthy controls. The disproportional loss of BM Th2 cells, as well as Tregs, results in a Th1 shift that contributes to autoantibody-mediated platelet destruction and T-cell cytotoxicity. The localizations and functions of BM NK and NKT cells in ITP are largely unknown.

Close modal

Activated CD4+ T cells also have the potential to disrupt hematopoiesis, and polyclonal expansion of dysregulated CD4+ T cells is implied in AA pathogenesis.99  T helper 1 (Th1)-, Th2-, and Th17-cell expansions are characteristic of AA.100  Elevated IL-17 levels may impair hematopoiesis by depleting Tregs,100  directly inhibiting HSCs,101  recruiting Th1 cells,100  and/or by inducing macrophages to secrete TNF-α.102  TNF-α in turn stimulates IFN-γ secretion from T cells,103  accelerating HSC impairment. Oligoclonal or monoclonal expansion of Th1 cells,104  increased T-bet (a T-box transcription factor) expression, and abnormally high production of IFN-γ, TNF-α, and IL-2 are also typical for AA, indicating a Th1 response–based pathogenesis105,106  (Figure 2B).

Autoreactive CD8+ T cells are also implicated in the pathophysiology of ITP. In patients with ITP, BM T cells express more VLA-4 and CX3CR1 and make up 81% of BM lymphocytes (vs 55% in healthy controls), suggesting that increased BM T-cell recruitment is part of ITP pathogenesis.107  Most of those CD8+ T cells are cytotoxic, expressing high levels of TNF, FASLG, PRF1, and GZMB messenger RNAs, and they can lyse platelets directly or induce platelet apoptosis108,109  (Figure 2C). The antigens recognized by CD8+ T cells in ITP have not yet been identified, but they might recognize megakaryocyte- or pathogen-derived antigens presented on platelets via MHC-I. ITP BM CD8+ T cells can also inhibit platelet formation by suppressing megakaryocyte apoptosis.110 

CD4+ T cells also display a Th1 shift in ITP, and glycoprotein IIb/IIIa-autoreactive CD4+ T cells induce the production of antiplatelet autoantibodies involved in disease pathogenesis111  (Figure 2C). The Th1 shift in ITP is a result of a decrease in Th2 cells,112  suggesting a different mechanism than the T-bet induction observed in AA. In addition, and contrary to AA, the fraction of CD4+ T cells in ITP BM is lower than that in healthy controls (40% vs 61%, respectively).110 

Treg malfunction is associated with both AA and ITP. Circulating CD4+CD25+ T cells in patients with AA have significantly lower levels of FoxP3 than those in healthy controls, and significantly decreased or absent levels of NFAT1, the master regulator for FoxP3 expression.113  Patients with AA also display an inverse correlation of Th17 and Treg numbers,100  the differentiation of which is reinforced by specific cytokine milieus.114  This indicates that the immune system is polarized toward a Th1/Th17 response, with Treg deficiency contributing to the development of AA.100  Treg deficiency is also implicated in the pathogenesis of ITP; patients with ITP have fewer BM Tregs than do healthy controls (5% vs 11% of CD4+ cells, respectively). Lower levels of Tregs and Treg-secreted factors such as transforming growth factor β (TGF-β) contribute to destruction of platelets by antibodies and T cell–mediated cytotoxicity in ITP107  (Figure 2C).

Little is known about the role of γδ T cells in AA. γδ T cells in the BM of patients with AA are significantly decreased compared with those in healthy controls,115  but they are increased in patients with acquired pure red cell aplasia.116  In pediatric ITP, significant increases in polyclonally expanded Vγ9+Vδ2+ T cells (to up to 48% of all T cells) were observed in PB, and reductions in these cells directly correlated with recovery, implicating a role for γδ T cells in ITP pathogenesis. γδ T cells may induce platelet destruction by secreting cytokines such as IL-2, GM-CSF, and IFN-γ by enhancing antibody production by autoreactive B cells, by direct cytolysis, or via antibody-dependent cell-mediated cytotoxicity.117,118  Moreover, ITP can present as the first symptom of hepatosplenic γδ T-cell lymphoma as a result of BM involvement by malignant γδ T cells.119  This is seen in approximately two-thirds of patients, typically with a sinusoidal infiltration pattern that can compose up to 30% of all BM cells.120 

Patients with AA exhibit reduced BM NKT cells, which display a more diverse, polyclonal TCR repertoire, but the significance thereof remains unclear.121  Stimulating NKT cells from AA mice with α-galactosylceramide inhibits IFN-γ production and enhances IL-4 secretion, which restores hematopoietic colony formation in vitro.122  The role of NKT cells in ITP is less clear. One study reported increased NKT cell numbers in patients with severe ITP, suggesting that NKT cells may drive ITP pathogenesis.123  Other studies reported that NKT cell function was reduced in untreated patients with ITP124  and that NKT cell activation suppressed platelet-induced T-cell proliferation in vitro,125  suggesting a protective role for NKT cells in ITP. Similar to NKT cells, NK cells in PB and BM of patients with AA are significantly reduced, and their cytotoxic functions are impaired; these parameters are restored upon immunosuppressive therapy.126,127  Because NK cells regulate CD8+ T cells via direct killing,128  impaired NK cell cytotoxicity and the resulting excess of autoreactive CD8+ T cells likely contribute to AA pathogenesis129  (Figure 2B). In contrast, in ITP, NK cells can lyse platelets directly and contribute to disease pathogenesis.130  However, the role of BM NK cells in this process is largely unknown because most studies on the role of NK cells in ITP focused on PB or spleen, and results are contradictory. In 1 study, PB NK cell frequencies were approximately twice as high in patients with ITP compared with those in healthy controls.131  Another study reported largely unaltered NK cell frequencies in PB and spleen; however, their cytotoxic functions were impaired132  as in AA.126 

ILCs are altered in their distribution and function in the spleens of patients with ITP, with increased numbers of IFN-γ– and IL-2–producing ILCs, although no data on BM were reported.133  On the basis of their comparable cytokine profile, it is tempting to speculate that ILCs could have a similar impact on HSCs and progenitors as TCR-expressing lymphocytes.

To summarize, the efficacy of immunosuppressive therapies in AA and ITP provides a strong pathophysiological link to immune-mediated mechanisms, likely executed by BM-resident mature lymphocytes. Antithymocyte globulin and cyclosporin, in combination with the MPL agonist eltrombopag, are effective in most patients with AA, which is in line with the mainly T-cell–driven autoimmune process in AA.134  Nowadays, patients with ITP are most commonly treated with corticosteroids, intravenous immunoglobulins, or rituximab, an anti-CD20 monoclonal antibody; splenectomy is reserved for refractory cases.135,136  The efficacy of rituximab confirms that B cells and autoantibodies are involved in the pathophysiology of ITP; this is corroborated by a report showing that the proteasome inhibitor bortezomib, which targets long-lived plasma cells, also alleviated ITP.137  Thus, targeting BM plasma cells may be an interesting therapeutic strategy for autoimmune cytopenias as well.

HSCT is a cornerstone therapy for numerous hematologic diseases. Mature lymphocyte populations support HSC engraftment and execute graft-versus-tumor effects, but they are also key players in GVHD, a major cause of morbidity and mortality after HSCT. GVHD is induced by donor-derived T cells as well as persisting tissue-resident recipient T cells that are activated by donor APCs.138  In contrast, Tregs are known to alleviate GVHD and promote immune reconstitution after HSCT.139,140  Although many functions of different T-cell subsets in HSCT have been elucidated (Figure 3A), much still needs to be learned about the role of other lymphocyte populations such as ILCs and the localization and contribution of their specific niches.

Figure 3.

Pathophysiology of mature BM lymphocytes in HSCT. (A) In HSCT, donor BM CD8+ T cells support HSC engraftment by facilitating HSC entry into the BM, inhibiting their exit, and suppressing host-derived alloreactive (allo-) BM CD4+ and CD8+ T cells. NKT cells are resistant to irradiation, a common conditioning regimen in HSCT, leading to an altered NKT-cell/T-cell balance in the BM and IL-4 production by host BM NKT cells. This results in Th2-polarization of donor BM T cells and induction of IL-10–producing donor BM Tregs, which improve HSC engraftment. Moreover, donor BM Tregs preferentially accumulate around donor HSCs and provide immune protection. In addition, they promote B-cell reconstitution by facilitating IL-7 production from BM perivascular stromal cells. High numbers of donor-derived CD27+ γδ T cells post-HSCT correlate with longer disease-free survival, but the exact localization of those cells, and whether they home to the BM, is not known. (B) GVHD is mediated by activated donor T cells and is a major cause of morbidity and mortality after HSCT. High levels of naïve and central-memory allo-CD8+ T cells correlate with increased GVHD risk. Donor BM allo-CD8+ T cells can induce host plasma cell destruction. In addition, activated donor CD8+ T cells likely negatively affect HSCs in GVHD through secretion of proinflammatory cytokines, including TNF-α and IFN-γ. Moreover, donor-derived alloantibodies, presumably secreted by plasma cells in BM, can promote cutaneous chronic GVHD. Host BM NKT cells can ameliorate GVHD by inhibiting the proliferation and preventing the activation of allo-CD8+ and CD4+ T cells. Finally, donor-derived CD8+ γδ T cells are associated with higher incidences of GVHD, but the role of BM in this process is unknown.

Figure 3.

Pathophysiology of mature BM lymphocytes in HSCT. (A) In HSCT, donor BM CD8+ T cells support HSC engraftment by facilitating HSC entry into the BM, inhibiting their exit, and suppressing host-derived alloreactive (allo-) BM CD4+ and CD8+ T cells. NKT cells are resistant to irradiation, a common conditioning regimen in HSCT, leading to an altered NKT-cell/T-cell balance in the BM and IL-4 production by host BM NKT cells. This results in Th2-polarization of donor BM T cells and induction of IL-10–producing donor BM Tregs, which improve HSC engraftment. Moreover, donor BM Tregs preferentially accumulate around donor HSCs and provide immune protection. In addition, they promote B-cell reconstitution by facilitating IL-7 production from BM perivascular stromal cells. High numbers of donor-derived CD27+ γδ T cells post-HSCT correlate with longer disease-free survival, but the exact localization of those cells, and whether they home to the BM, is not known. (B) GVHD is mediated by activated donor T cells and is a major cause of morbidity and mortality after HSCT. High levels of naïve and central-memory allo-CD8+ T cells correlate with increased GVHD risk. Donor BM allo-CD8+ T cells can induce host plasma cell destruction. In addition, activated donor CD8+ T cells likely negatively affect HSCs in GVHD through secretion of proinflammatory cytokines, including TNF-α and IFN-γ. Moreover, donor-derived alloantibodies, presumably secreted by plasma cells in BM, can promote cutaneous chronic GVHD. Host BM NKT cells can ameliorate GVHD by inhibiting the proliferation and preventing the activation of allo-CD8+ and CD4+ T cells. Finally, donor-derived CD8+ γδ T cells are associated with higher incidences of GVHD, but the role of BM in this process is unknown.

Close modal

Donor CD8+ T cells support HSC engraftment by inactivating the host response against donor MHC-I alloantigens141  and suppressing host-derived immune cells.142  CD8+ T cells also facilitate BM entry and inhibit BM exit of donor HSCs,143,144  potentially by modulating phosphotyrosine-mediated signaling in response to CXCL12.144  However, although higher frequencies of donor CD8+ T cells support HSC engraftment, they are also associated with a higher likelihood of GVHD.145  Naïve and, to a lesser extent, central-memory (but not effector-memory) CD8+ T cells mediated GVHD in mice in both MHC-mismatched and MHC-matched recipients.146  Therefore, naïve T-cell depletion has been investigated as a means of reducing GVHD in HSCT.147,148  In addition, based on the pathophysiological changes that take place in AA, ITP, and viral infections, it is likely that activated donor CD8+ T cells modulate HSCs and hematopoiesis in GVHD through production of proinflammatory cytokines (Figure 3B).

The role of CD4+ T cells in HSCT has mostly been addressed in view of Tregs and GVHD. In patients with severe GVHD, there is an imbalance between Tregs and effector CD4+ T cells, mainly because of an overrepresentation of effector-memory CD4+ T cells.149,150  Although the positive functions of effector CD4+ T cells on HSC engraftment are somewhat clear, more is known about the role of Tregs. After HSCT in mice, donor-derived Tregs preferentially localize to the endosteal surface, accumulating around donor HSCs and providing immune protection by preventing allorejection. Therefore, Treg depletion resulted in a loss of 75% to 90% of donor HSCs.59  A distinct subset of CD150hi Tregs localizes in perivascular HSC niches, where they provide immune privilege and maintain HSCs quiescence.61  Cotransplantation of niche-associated CD150hi Tregs improved HSC engraftment to a greater extent than did transplantation of CD150low Tregs.61  Recently, CD4+CD150hiFoxP3 T cells were shown to play a similar role in regulating HSCs. It was postulated that CD150hi Tregs and CD4+CD150hiFoxP3 T cells generate extracellular adenosine in a coordinated fashion via CD39/CD73, maintaining HSC quiescence and improving engraftment by protecting HSCs from oxidative stress.151  Tregs are also important in B-cell reconstitution after HSCT. Treg depletion diminished the production of IL-7, an important B-cell survival factor, by ICAM+ perivascular stromal cells, possibly as a result of the inflammatory cytokine milieu induced by autoreactive T cells.62 

B cells, plasma cells, and antibodies play an important and complex role in GVHD.152  Although the precise role of BM herein is less clear, donor B-cell–derived antibodies can promote cutaneous chronic GVHD.153  Moreover, gradual destruction of host plasma cells by GVHD is an important reason why host antidonor isohemagglutinins gradually disappear after ABO-mismatched HSCT.154,155  Therapeutic efficacy of donor plasma cell depletion for treatment of chronic GVHD is being tested in a phase 2 clinical trial using pomalidomide, an immunomodulatory drug for multiple myeloma (clinicaltrials.gov identifier: NCT01688466). This is in line with the proven efficacy of the proteasome inhibitor bortezomib, which also targets long-lived plasma cells and thereby alleviates GVHD.156,157 

Recent studies suggest a favorable role for γδ T cells in HSCT.158  After HSCT, reconstitution of γδ T cells is largely the result of clonal expansion of donor-derived cells.159,160  High numbers of CD27+ γδ T cells post-HSCT correlated with longer disease-free survival.71  In addition, CD8+ γδ T cells were associated with a higher incidence of GVHD, whereas the frequency of CD8 γδ T cells tended to correlate with lower GVHD rates, suggesting a possible immunoregulatory role.71  Studies in leukemia patients who had undergone HSCT with αβ T cell-depleted grafts indicated that γδ T cells facilitate the graft-versus-leukemia effect without causing GVHD.161  This phenomenon may be explained by γδ T cells not being HLA-restricted, which reduces the chance of GVHD triggered by HLA-mismatch.158 

The importance of NK cells and NKT cells has also been documented in HSCT. NK cells were shown to facilitate the engraftment of HSCs in HSCT models using reduced myeloablative conditioning regimens and IL-15162  as well as in cord blood HSCT.163  The resistance of NKT cells to irradiation leads to an altered NKT cell/T cell balance after conditioning and IL-4 production by host NKT cells, resulting in Th2 polarization of donor T cells and induction of IL-10–producing donor Tregs, which ameliorates GVHD74,164  (Figure 3B). Tracking studies of ILCs in patients with acute myeloid leukemia who are receiving chemotherapy and HSCT revealed that abundance of ILC before and reconstitution of ILC after induction chemotherapy was associated with protection from GVHD. This was mediated by donor ILC3s expressing CD69 and tissue-homing markers for the gut and skin.165  A possible mechanism for GVHD protection is ILC3-derived IL-22, a cytokine associated with epithelial homeostasis. Indeed, ILC3s proliferate and upregulate IL-22 production when cocultured with BM-derived mesenchymal stem cells,166  suggesting that ILC3s may harbor in an mesenchymal stem cell–containing BM niche.

BM is a highly dynamic organ with complex, intertwined functions necessary for hematopoiesis and immunity, many of which can be regulated by mature lymphocytes residing within it. Thoroughly characterizing different BM lymphocyte populations, their diverse states and microenvironments, and the cells they interact with will improve our understanding of homeostatic and disordered hematopoiesis and possibly lead to novel therapies for diseases such as AA and ITP. Future studies of BM lymphocyte populations should therefore combine functional assays with high-dimensional analysis technologies, including single-cell RNA sequencing, mass cytometry, multiplexed tissue imaging, and 3D microscopy.3,167-169  This will enable a deeper understanding of the diversity, localization, and (patho)physiology of these different cell types and how they interact with hematopoietic and nonhematopoietic cells.

The online version of this article contains a data supplement.

The authors thank Marieke Goedhart (Sanquin Research, Amsterdam, The Netherlands) for giving key input to this review and Katherine MacNamara (Albany Medical College, Albany, NY) for critically reading the manuscript and providing constructive feedback. Parts of the figures were created with BioRender.com.

C.M.S. was supported by an Advanced Postdoc Mobility Fellowship from the Swiss National Science Foundation (P300PB_171189 and P400PM_183915) and an International Award for Research in Leukemia from the Lady Tata Memorial Trust (London, United Kingdom).

Contribution: C.M.S. conceived of the review; C.C. created the figures; and all authors contributed equally to the study by writing, editing, and approving the final version of the review.

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

Correspondence: Martijn A. Nolte, Sanquin Research, Plesmanlaan 125, Room Y308, 1066CX, Amsterdam, The Netherlands; e-mail: m.nolte@sanquin.nl; and Christian M. Schürch, Department of Pathology, Liebermeisterstrasse 8, Room 125, 72076 Tübingen, Germany; e-mail: christian.schuerch@med.uni-tuebingen.de.

1.
Crane
GM
,
Jeffery
E
,
Morrison
SJ
.
Adult haematopoietic stem cell niches
.
Nat Rev Immunol
.
2017
;
17
(
9
):
573
-
590
.
2.
Nombela-Arrieta
C
,
Manz
MG
.
Quantification and three-dimensional microanatomical organization of the bone marrow
.
Blood Adv
.
2017
;
1
(
6
):
407
-
416
.
3.
Gomariz
A
,
Helbling
PM
,
Isringhausen
S
, et al
.
Quantitative spatial analysis of haematopoiesis-regulating stromal cells in the bone marrow microenvironment by 3D microscopy
.
Nat Commun
.
2018
;
9
(
1
):
2532
.
4.
Broxmeyer
HE
,
Bruns
HA
,
Zhang
S
, et al
.
Th1 cells regulate hematopoietic progenitor cell homeostasis by production of oncostatin M
.
Immunity
.
2002
;
16
(
6
):
815
-
825
.
5.
Geerman
S
,
Brasser
G
,
Bhushal
S
, et al
.
Memory CD8+ T cells support the maintenance of hematopoietic stem cells in the bone marrow
.
Haematologica
.
2018
;
103
(
6
):
e230
-
e233
.
6.
Libregts
SFWM
,
Nolte
MA
.
Parallels between immune driven-hematopoiesis and T cell activation: 3 signals that relay inflammatory stress to the bone marrow
.
Exp Cell Res
.
2014
;
329
(
2
):
239
-
247
.
7.
Shallis
RM
,
Ahmad
R
,
Zeidan
AM
.
Aplastic anemia: Etiology, molecular pathogenesis, and emerging concepts
.
Eur J Haematol
.
2018
;
101
(
6
):
711
-
720
.
8.
Riether
C
,
Schürch
CM
,
Ochsenbein
AF
.
Regulation of hematopoietic and leukemic stem cells by the immune system
.
Cell Death Differ
.
2015
;
22
(
2
):
187
-
198
.
9.
Lamble
AJ
,
Lind
EF
.
Targeting the immune microenvironment in acute myeloid leukemia: A focus on T cell immunity
.
Front Oncol
.
2018
;
8
:
213
.
10.
Slifka
MK
,
Antia
R
,
Whitmire
JK
,
Ahmed
R
.
Humoral immunity due to long-lived plasma cells
.
Immunity
.
1998
;
8
(
3
):
363
-
372
.
11.
Hauser
AE
,
Debes
GF
,
Arce
S
, et al
.
Chemotactic responsiveness toward ligands for CXCR3 and CXCR4 is regulated on plasma blasts during the time course of a memory immune response
.
J Immunol
.
2002
;
169
(
3
):
1277
-
1282
.
12.
Cassese
G
,
Arce
S
,
Hauser
AE
, et al
.
Plasma cell survival is mediated by synergistic effects of cytokines and adhesion-dependent signals
.
J Immunol
.
2003
;
171
(
4
):
1684
-
1690
.
13.
O’Connor
BP
,
Raman
VS
,
Erickson
LD
, et al
.
BCMA is essential for the survival of long-lived bone marrow plasma cells
.
J Exp Med
.
2004
;
199
(
1
):
91
-
98
.
14.
Belnoue
E
,
Pihlgren
M
,
McGaha
TL
, et al
.
APRIL is critical for plasmablast survival in the bone marrow and poorly expressed by early-life bone marrow stromal cells
.
Blood
.
2008
;
111
(
5
):
2755
-
2764
.
15.
Kawano
Y
,
Moschetta
M
,
Manier
S
, et al
.
Targeting the bone marrow microenvironment in multiple myeloma
.
Immunol Rev
.
2015
;
263
(
1
):
160
-
172
.
16.
Tokoyoda
K
,
Egawa
T
,
Sugiyama
T
,
Choi
B-I
,
Nagasawa
T
.
Cellular niches controlling B lymphocyte behavior within bone marrow during development
.
Immunity
.
2004
;
20
(
6
):
707
-
718
.
17.
Glatman Zaretsky
A
,
Konradt
C
,
Dépis
F
, et al
.
T regulatory cells support plasma cell populations in the bone marrow
.
Cell Rep
.
2017
;
18
(
8
):
1906
-
1916
.
18.
Zehentmeier
S
,
Roth
K
,
Cseresnyes
Z
, et al
.
Static and dynamic components synergize to form a stable survival niche for bone marrow plasma cells
.
Eur J Immunol
.
2014
;
44
(
8
):
2306
-
2317
.
19.
Cariappa
A
,
Chase
C
,
Liu
H
,
Russell
P
,
Pillai
S
.
Naive recirculating B cells mature simultaneously in the spleen and bone marrow
.
Blood
.
2007
;
109
(
6
):
2339
-
2345
.
20.
Agbay
RLMC
,
Medeiros
LJ
,
Khoury
JD
,
Salem
A
,
Bueso-Ramos
CE
,
Loghavi
S
.
Characteristics and clinical implications of reactive germinal centers in the bone marrow
.
Hum Pathol
.
2017
;
68
:
7
-
21
.
21.
Dilosa
RM
,
Maeda
K
,
Masuda
A
,
Szakal
AK
,
Tew
JG
.
Germinal center B cells and antibody production in the bone marrow
.
J Immunol
.
1991
;
146
(
12
):
4071
-
4077
.
22.
Jiang
H
,
Cui
N
,
Yang
L
, et al
.
Altered follicular helper T cell impaired antibody production in a murine model of myelodysplastic syndromes
.
Oncotarget
.
2017
;
8
(
58
):
98270
-
98279
.
23.
Wang
Q
,
Li
J
,
Yu
T-S
, et al
.
Disrupted balance of CD4+ T-cell subsets in bone marrow of patients with primary immune thrombocytopenia
.
Int J Biol Sci
.
2019
;
15
(
13
):
2798
-
2814
.
24.
Tew
JG
,
DiLosa
RM
,
Burton
GF
, et al
.
Germinal centers and antibody production in bone marrow
.
Immunol Rev
.
1992
;
126
(
1
):
99
-
112
.
25.
Paramithiotis
E
,
Cooper
MD
.
Memory B lymphocytes migrate to bone marrow in humans
.
Proc Natl Acad Sci USA
.
1997
;
94
(
1
):
208
-
212
.
26.
McHeyzer-Williams
LJ
,
Cool
M
,
McHeyzer-Williams
MG
.
Antigen-specific B cell memory: Expression and replenishment of a novel b220(-) memory b cell compartment
.
J Exp Med
.
2000
;
191
(
7
):
1149
-
1166
.
27.
Riedel
R
,
Addo
R
,
Ferreira-Gomes
M
, et al
.
Discrete populations of isotype-switched memory B lymphocytes are maintained in murine spleen and bone marrow
.
Nat Commun
.
2020
;
11
(
1
):
2570
.
28.
Zeng
D
,
Hoffmann
P
,
Lan
F
,
Huie
P
,
Higgins
J
,
Strober
S
.
Unique patterns of surface receptors, cytokine secretion, and immune functions distinguish T cells in the bone marrow from those in the periphery: impact on allogeneic bone marrow transplantation
.
Blood
.
2002
;
99
(
4
):
1449
-
1457
.
29.
Westermann
J
,
Pabst
R
.
Distribution of lymphocyte subsets and natural killer cells in the human body
.
Clin Investig
.
1992
;
70
(
7
):
539
-
544
.
30.
Geerman
S
,
Hickson
S
,
Brasser
G
,
Pascutti
MF
,
Nolte
MA
.
Quantitative and qualitative analysis of bone marrow CD8(+) T cells from different bones uncovers a major contribution of the bone marrow in the vertebrae
.
Front Immunol
.
2016
;
6
:
660
.
31.
Milo
I
,
Sapoznikov
A
,
Kalchenko
V
, et al
.
Dynamic imaging reveals promiscuous crosspresentation of blood-borne antigens to naive CD8+ T cells in the bone marrow
.
Blood
.
2013
;
122
(
2
):
193
-
208
.
32.
Mazo
IB
,
Honczarenko
M
,
Leung
H
, et al
.
Bone marrow is a major reservoir and site of recruitment for central memory CD8+ T cells
.
Immunity
.
2005
;
22
(
2
):
259
-
270
.
33.
Sercan Alp
Ö
,
Durlanik
S
,
Schulz
D
, et al
.
Memory CD8(+) T cells colocalize with IL-7(+) stromal cells in bone marrow and rest in terms of proliferation and transcription
.
Eur J Immunol
.
2015
;
45
(
4
):
975
-
987
.
34.
Feuerer
M
,
Beckhove
P
,
Garbi
N
, et al
.
Bone marrow as a priming site for T-cell responses to blood-borne antigen
.
Nat Med
.
2003
;
9
(
9
):
1151
-
1157
.
35.
Goedhart
M
,
Gessel
S
,
van der Voort
R
, et al
.
CXCR4, but not CXCR3, drives CD8+ T-cell entry into and migration through the murine bone marrow
.
Eur J Immunol
.
2019
;
49
(
4
):
576
-
589
.
36.
Chaix
J
,
Nish
SA
,
Lin
W-HW
, et al
.
Cutting edge: CXCR4 is critical for CD8+ memory T cell homeostatic self-renewal but not rechallenge self-renewal
.
J Immunol
.
2014
;
193
(
3
):
1013
-
1016
.
37.
Slifka
MK
,
Whitmire
JK
,
Ahmed
R
.
Bone marrow contains virus-specific cytotoxic T lymphocytes
.
Blood
.
1997
;
90
(
5
):
2103
-
2108
.
38.
Duffy
D
,
Perrin
H
,
Abadie
V
, et al
.
Neutrophils transport antigen from the dermis to the bone marrow, initiating a source of memory CD8+ T cells
.
Immunity
.
2012
;
37
(
5
):
917
-
929
.
39.
Palendira
U
,
Chinn
R
,
Raza
W
, et al
.
Selective accumulation of virus-specific CD8+ T cells with unique homing phenotype within the human bone marrow
.
Blood
.
2008
;
112
(
8
):
3293
-
3302
.
40.
Di Rosa
F
,
Gebhardt
T
.
Bone marrow T cells and the integrated functions of recirculating and tissue-resident memory T cells
.
Front Immunol
.
2016
;
7
:
51
.
41.
Kieper
WC
,
Tan
JT
,
Bondi-Boyd
B
, et al
.
Overexpression of interleukin (IL)-7 leads to IL-15-independent generation of memory phenotype CD8+ T cells
.
J Exp Med
.
2002
;
195
(
12
):
1533
-
1539
.
42.
Herndler-Brandstetter
D
,
Landgraf
K
,
Jenewein
B
, et al
.
Human bone marrow hosts polyfunctional memory CD4+ and CD8+ T cells with close contact to IL-15-producing cells
.
J Immunol
.
2011
;
186
(
12
):
6965
-
6971
.
43.
Lodolce
JP
,
Boone
DL
,
Chai
S
, et al
.
IL-15 receptor maintains lymphoid homeostasis by supporting lymphocyte homing and proliferation
.
Immunity
.
1998
;
9
(
5
):
669
-
676
.
44.
Zhang
X
,
Sun
S
,
Hwang
I
,
Tough
DF
,
Sprent
J
.
Potent and selective stimulation of memory-phenotype CD8+ T cells in vivo by IL-15
.
Immunity
.
1998
;
8
(
5
):
591
-
599
.
45.
Nolte
MA
,
Goedhart
M
,
Geginat
J
.
Maintenance of memory CD8 T cells: Divided over division
.
Eur J Immunol
.
2017
;
47
(
11
):
1875
-
1879
.
46.
Di Rosa
F
.
Maintenance of memory T cells in the bone marrow: survival or homeostatic proliferation?
Nat Rev Immunol
.
2016
;
16
(
4
):
271
.
47.
Klonowski
KD
,
Williams
KJ
,
Marzo
AL
,
Blair
DA
,
Lingenheld
EG
,
Lefrançois
L
.
Dynamics of blood-borne CD8 memory T cell migration in vivo
.
Immunity
.
2004
;
20
(
5
):
551
-
562
.
48.
Pascutti
MF
,
Geerman
S
,
Collins
N
, et al
.
Peripheral and systemic antigens elicit an expandable pool of resident memory CD8+ T cells in the bone marrow
.
Eur J Immunol
.
2019
;
49
(
6
):
853
-
872
.
49.
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
;
14
(
4
):
460
-
472
.
50.
Pronk
CJH
,
Veiby
OP
,
Bryder
D
,
Jacobsen
SEW
.
Tumor necrosis factor restricts hematopoietic stem cell activity in mice: involvement of two distinct receptors
.
J Exp Med
.
2011
;
208
(
8
):
1563
-
1570
.
51.
Price
PW
,
Cerny
J
.
Characterization of CD4+ T cells in mouse bone marrow. I. Increased activated/memory phenotype and altered TCR Vbeta repertoire
.
Eur J Immunol
.
1999
;
29
(
3
):
1051
-
1056
.
52.
Feuerer
M
,
Beckhove
P
,
Mahnke
Y
, et al
.
Bone marrow microenvironment facilitating dendritic cell: CD4 T cell interactions and maintenance of CD4 memory
.
Int J Oncol
.
2004
;
25
(
4
):
867
-
876
.
53.
Tokoyoda
K
,
Zehentmeier
S
,
Hegazy
AN
, et al
.
Professional memory CD4+ T lymphocytes preferentially reside and rest in the bone marrow
.
Immunity
.
2009
;
30
(
5
):
721
-
730
.
54.
Siracusa
F
,
McGrath
MA
,
Maschmeyer
P
, et al
.
Nonfollicular reactivation of bone marrow resident memory CD4 T cells in immune clusters of the bone marrow
.
Proc Natl Acad Sci USA
.
2018
;
115
(
6
):
1334
-
1339
.
55.
Migliaccio
G
,
Migliaccio
AR
,
Visser
JW
.
Synergism between erythropoietin and interleukin-3 in the induction of hematopoietic stem cell proliferation and erythroid burst colony formation
.
Blood
.
1988
;
72
(
3
):
944
-
951
.
56.
Greenberger
JS
,
Krensky
AM
,
Messner
H
,
Burakoff
SJ
,
Wandl
U
,
Sakakeeny
MA
.
Production of colony-stimulating factor(s) for granulocyte-macrophage and multipotential (granulocyte/erythroid/megakaryocyte/macrophage) hematopoietic progenitor cells (CFU-GEMM) by clonal lines of human IL-2-dependent T-lymphocytes
.
Exp Hematol
.
1984
;
12
(
9
):
720
-
727
.
57.
Luckheeram
RV
,
Zhou
R
,
Verma
AD
,
Xia
B
.
CD4+T cells: Differentiation and functions
.
Clin Dev Immunol
.
2012
;
2012
:
925135
.
58.
Monteiro
JP
,
Benjamin
A
,
Costa
ES
,
Barcinski
MA
,
Bonomo
A
.
Normal hematopoiesis is maintained by activated bone marrow CD4+ T cells
.
Blood
.
2005
;
105
(
4
):
1484
-
1491
.
59.
Fujisaki
J
,
Wu
J
,
Carlson
AL
, et al
.
In vivo imaging of Treg cells providing immune privilege to the haematopoietic stem-cell niche
.
Nature
.
2011
;
474
(
7350
):
216
-
219
.
60.
Zou
L
,
Barnett
B
,
Safah
H
, et al
.
Bone marrow is a reservoir for CD4+CD25+ regulatory T cells that traffic through CXCL12/CXCR4 signals
.
Cancer Res
.
2004
;
64
(
22
):
8451
-
8455
.
61.
Hirata
Y
,
Furuhashi
K
,
Ishii
H
, et al
.
CD150high bone marrow Tregs maintain hematopoietic stem cell quiescence and immune privilege via adenosine
.
Cell Stem Cell
.
2018
;
22
(
3
):
445
-
453.e5
.
62.
Pierini
A
,
Nishikii
H
,
Baker
J
, et al
.
Foxp3+ regulatory T cells maintain the bone marrow microenvironment for B cell lymphopoiesis
.
Nat Commun
.
2017
;
8
:
15068
.
63.
Couper
KN
,
Blount
DG
,
Riley
EM
.
IL-10: the master regulator of immunity to infection
.
J Immunol
.
2008
;
180
(
9
):
5771
-
5777
.
64.
Bonneville
M
,
O’Brien
RL
,
Born
WK
.
Gammadelta T cell effector functions: a blend of innate programming and acquired plasticity
.
Nat Rev Immunol
.
2010
;
10
(
7
):
467
-
478
.
65.
Adams
EJ
,
Gu
S
,
Luoma
AM
.
Human gamma delta T cells: Evolution and ligand recognition
.
Cell Immunol
.
2015
;
296
(
1
):
31
-
40
.
66.
Khairallah
C
,
Chu
TH
,
Sheridan
BS
.
Tissue adaptations of memory and tissue-resident gamma delta T cells
.
Front Immunol
.
2018
;
9
:
2636
.
67.
Davey
MS
,
Willcox
CR
,
Baker
AT
,
Hunter
S
,
Willcox
BE
.
Recasting human Vδ1 lymphocytes in an adaptive role
.
Trends Immunol
.
2018
;
39
(
6
):
446
-
459
.
68.
Gaballa
A
,
Arruda
LCM
,
Rådestad
E
,
Uhlin
M
.
CD8+γδ T cells are more frequent in CMV seropositive bone marrow grafts and display phenotype of an adaptive immune response
.
Stem Cells Int
.
2019
;
2019
:
6348060
.
69.
Davey
MS
,
Willcox
CR
,
Hunter
S
,
Oo
YH
,
Willcox
BE
.
Vδ2+ T cells-Two subsets for the price of one
.
Front Immunol
.
2018
;
9
:
2106
.
70.
Sykes
M
.
Unusual T cell populations in adult murine bone marrow. Prevalence of CD3+CD4-CD8- and alpha beta TCR+NK1.1+ cells
.
J Immunol
.
1990
;
145
(
10
):
3209
-
3215
.
71.
Gaballa
A
,
Stikvoort
A
,
Önfelt
B
, et al
.
T-cell frequencies of CD8+ γδ and CD27+ γδ cells in the stem cell graft predict the outcome after allogeneic hematopoietic cell transplantation
.
Bone Marrow Transplant
.
2019
;
54
(
10
):
1562
-
1574
.
72.
Castella
B
,
Foglietta
M
,
Sciancalepore
P
, et al
.
Anergic bone marrow Vγ9Vδ2 T cells as early and long-lasting markers of PD-1-targetable microenvironment-induced immune suppression in human myeloma
.
OncoImmunology
.
2015
;
4
(
11
):
e1047580
.
73.
Kunzmann
V
,
Bauer
E
,
Feurle
J
,
Weissinger
F
,
Tony
HP
,
Wilhelm
M
.
Stimulation of gammadelta T cells by aminobisphosphonates and induction of antiplasma cell activity in multiple myeloma
.
Blood
.
2000
;
96
(
2
):
384
-
392
.
74.
Kohrt
HE
,
Pillai
AB
,
Lowsky
R
,
Strober
S
.
NKT cells, Treg, and their interactions in bone marrow transplantation
.
Eur J Immunol
.
2010
;
40
(
7
):
1862
-
1869
.
75.
Kotsianidis
I
,
Silk
JD
,
Spanoudakis
E
, et al
.
Regulation of hematopoiesis in vitro and in vivo by invariant NKT cells
.
Blood
.
2006
;
107
(
8
):
3138
-
3144
.
76.
Vivier
E
,
Tomasello
E
,
Baratin
M
,
Walzer
T
,
Ugolini
S
.
Functions of natural killer cells
.
Nat Immunol
.
2008
;
9
(
5
):
503
-
510
.
77.
Grégoire
C
,
Chasson
L
,
Luci
C
, et al
.
The trafficking of natural killer cells
.
Immunol Rev
.
2007
;
220
(
1
):
169
-
182
.
78.
Yang
C
,
Siebert
JR
,
Burns
R
, et al
.
Heterogeneity of human bone marrow and blood natural killer cells defined by single-cell transcriptome
.
Nat Commun
.
2019
;
10
(
1
):
3931
.
79.
Askenase
MH
,
Han
S-J
,
Byrd
AL
, et al
.
Bone-marrow-resident NK cells prime monocytes for regulatory function during infection
.
Immunity
.
2015
;
42
(
6
):
1130
-
1142
.
80.
Artis
D
,
Spits
H
.
The biology of innate lymphoid cells
.
Nature
.
2015
;
517
(
7534
):
293
-
301
.
81.
Klose
CSN
,
Flach
M
,
Möhle
L
, et al
.
Differentiation of type 1 ILCs from a common progenitor to all helper-like innate lymphoid cell lineages
.
Cell
.
2014
;
157
(
2
):
340
-
356
.
82.
Constantinides
MG
,
McDonald
BD
,
Verhoef
PA
,
Bendelac
A
.
A committed precursor to innate lymphoid cells
.
Nature
.
2014
;
508
(
7496
):
397
-
401
.
83.
Ding
L
,
Morrison
SJ
.
Haematopoietic stem cells and early lymphoid progenitors occupy distinct bone marrow niches
.
Nature
.
2013
;
495
(
7440
):
231
-
235
.
84.
Montaldo
E
,
Teixeira-Alves
LG
,
Glatzer
T
, et al
.
Human RORγt(+)CD34(+) cells are lineage-specified progenitors of group 3 RORγt(+) innate lymphoid cells
.
Immunity
.
2014
;
41
(
6
):
988
-
1000
.
85.
Scoville
SD
,
Mundy-Bosse
BL
,
Zhang
MH
, et al
.
A progenitor cell expressing transcription factor RORγt generates all human innate lymphoid cell subsets
.
Immunity
.
2016
;
44
(
5
):
1140
-
1150
.
86.
Lim
AI
,
Li
Y
,
Lopez-Lastra
S
, et al
.
Systemic human ILC precursors provide a substrate for tissue ILC differentiation
.
Cell
.
2017
;
168
(
6
):
1086
-
1100.e10
.
87.
Swinkels
M
,
Rijkers
M
,
Voorberg
J
,
Vidarsson
G
,
Leebeek
FWG
,
Jansen
AJG
.
Emerging concepts in immune thrombocytopenia
.
Front Immunol
.
2018
;
9
:
880
.
88.
de Bruin
AM
,
Demirel
Ö
,
Hooibrink
B
,
Brandts
CH
,
Nolte
MA
.
Interferon-γ impairs proliferation of hematopoietic stem cells in mice
.
Blood
.
2013
;
121
(
18
):
3578
-
3585
.
89.
Dufour
C
,
Corcione
A
,
Svahn
J
,
Haupt
R
,
Battilana
N
,
Pistoia
V
.
Interferon γ and tumour necrosis factor α are overexpressed in bone marrow T lymphocytes from paediatric patients with aplastic anaemia
.
Br J Haematol
.
2001
;
115
(
4
):
1023
-
1031
.
90.
Mouzaki
A
,
Theodoropoulou
M
,
Gianakopoulos
I
,
Vlaha
V
,
Kyrtsonis
M-C
,
Maniatis
A
.
Expression patterns of Th1 and Th2 cytokine genes in childhood idiopathic thrombocytopenic purpura (ITP) at presentation and their modulation by intravenous immunoglobulin G (IVIg) treatment: their role in prognosis
.
Blood
.
2002
;
100
(
5
):
1774
-
1779
.
91.
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 β-CDR3 sequencing
.
Lancet
.
2004
;
364
(
9431
):
355
-
364
.
92.
Melenhorst
JJ
,
van Krieken
JH
,
Dreef
E
,
Landegent
JE
,
Willemze
R
,
Fibbe
WE
.
T cells selectively infiltrate bone marrow areas with residual haemopoiesis of patients with acquired aplastic anaemia
.
Br J Haematol
.
1997
;
99
(
3
):
517
-
519
.
93.
Ren
J
,
Hou
XY
,
Ma
SH
, et al
.
Elevated expression of CX3C chemokine receptor 1 mediates recruitment of T cells into bone marrow of patients with acquired aplastic anaemia
.
J Intern Med
.
2014
;
276
(
5
):
512
-
524
.
94.
Krebs
DL
,
Hilton
DJ
.
SOCS proteins: negative regulators of cytokine signaling
.
Stem Cells
.
2001
;
19
(
5
):
378
-
387
.
95.
Alvarado
LJ
,
Andreoni
A
,
Huntsman
HD
,
Cheng
H
,
Knutson
JR
,
Larochelle
A
.
Heterodimerization of TPO and IFNγ impairs human hematopoietic stem/progenitor cell signaling and survival in chronic inflammation [abstract]
.
Blood
.
2017
;
130
(
suppl 1
).
Abstract 4
.
96.
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
;
85
(
11
):
3183
-
3190
.
97.
Zeng
W
,
Kajigaya
S
,
Chen
G
,
Risitano
AM
,
Nunez
O
,
Young
NS
.
Transcript profile of CD4+ and CD8+ T cells from the bone marrow of acquired aplastic anemia patients
.
Exp Hematol
.
2004
;
32
(
9
):
806
-
814
.
98.
Giudice
V
,
Feng
X
,
Lin
Z
, et al
.
Deep sequencing and flow cytometric characterization of expanded effector memory CD8+CD57+ T cells frequently reveals T-cell receptor Vβ oligoclonality and CDR3 homology in acquired aplastic anemia
.
Haematologica
.
2018
;
103
(
5
):
759
-
769
.
99.
Risitano
AM
,
Kook
H
,
Zeng
W
,
Chen
G
,
Young
NS
,
Maciejewski
JP
.
Oligoclonal and polyclonal CD4 and CD8 lymphocytes in aplastic anemia and paroxysmal nocturnal hemoglobinuria measured by V β CDR3 spectratyping and flow cytometry
.
Blood
.
2002
;
100
(
1
):
178
-
183
.
100.
de Latour
RP
,
Visconte
V
,
Takaku
T
, et al
.
Th17 immune responses contribute to the pathophysiology of aplastic anemia
.
Blood
.
2010
;
116
(
20
):
4175
-
4184
.
101.
Broxmeyer
HE
,
Starnes
T
,
Ramsey
H
, et al
.
The IL-17 cytokine family members are inhibitors of human hematopoietic progenitor proliferation
.
Blood
.
2006
;
108
(
2
):
770
.
102.
Gu
Y
,
Hu
X
,
Liu
C
,
Qv
X
,
Xu
C
.
Interleukin (IL)-17 promotes macrophages to produce IL-8, IL-6 and tumour necrosis factor-alpha in aplastic anaemia
.
Br J Haematol
.
2008
;
142
(
1
):
109
-
114
.
103.
Sun
W
,
Wu
Z
,
Lin
Z
, et al
.
Macrophage TNF-α licenses donor T cells in murine bone marrow failure and can be implicated in human aplastic anemia
.
Blood
.
2018
;
132
(
26
):
2730
-
2743
.
104.
Kordasti
S
,
Marsh
J
,
Al-Khan
S
, et al
.
Functional characterization of CD4+ T cells in aplastic anemia
.
Blood
.
2012
;
119
(
9
):
2033
-
2043
.
105.
Giannakoulas
NC
,
Karakantza
M
,
Theodorou
GL
, et al
.
Clinical relevance of balance between type 1 and type 2 immune responses of lymphocyte subpopulations in aplastic anaemia patients
.
Br J Haematol
.
2004
;
124
(
1
):
97
-
105
.
106.
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
;
107
(
10
):
3983
-
3991
.
107.
Olsson
B
,
Ridell
B
,
Carlsson
L
,
Jacobsson
S
,
Wadenvik
H
.
Recruitment of T cells into bone marrow of ITP patients possibly due to elevated expression of VLA-4 and CX3CR1
.
Blood
.
2008
;
112
(
4
):
1078
-
1084
.
108.
Olsson
B
,
Andersson
P-O
,
Jernås
M
, et al
.
T-cell-mediated cytotoxicity toward platelets in chronic idiopathic thrombocytopenic purpura
.
Nat Med
.
2003
;
9
(
9
):
1123
-
1124
.
109.
Zhang
F
,
Chu
X
,
Wang
L
, et al
.
Cell-mediated lysis of autologous platelets in chronic idiopathic thrombocytopenic purpura
.
Eur J Haematol
.
2006
;
76
(
5
):
427
-
431
.
110.
Li
S
,
Wang
L
,
Zhao
C
,
Li
L
,
Peng
J
,
Hou
M
.
CD8+ T cells suppress autologous megakaryocyte apoptosis in idiopathic thrombocytopenic purpura
.
Br J Haematol
.
2007
;
139
(
4
):
605
-
611
.
111.
Kuwana
M
,
Kaburaki
J
,
Kitasato
H
, et al
.
Immunodominant epitopes on glycoprotein IIb-IIIa recognized by autoreactive T cells in patients with immune thrombocytopenic purpura
.
Blood
.
2001
;
98
(
1
):
130
-
139
.
112.
Ogawara
H
,
Handa
H
,
Morita
K
, et al
.
High Th1/Th2 ratio in patients with chronic idiopathic thrombocytopenic purpura
.
Eur J Haematol
.
2003
;
71
(
4
):
283
-
288
.
113.
Solomou
EE
,
Rezvani
K
,
Mielke
S
, et al
.
Deficient CD4+ CD25+ FOXP3+ T regulatory cells in acquired aplastic anemia
.
Blood
.
2007
;
110
(
5
):
1603
-
1606
.
114.
Bettelli
E
,
Carrier
Y
,
Gao
W
, et al
.
Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells
.
Nature
.
2006
;
441
(
7090
):
235
-
238
.
115.
Mentzel
U
,
Vogt
H
,
Rossol
R
, et al
.
Analysis of lymphocyte subsets in patients with aplastic anemia before and during immunosuppressive therapy
.
Ann Hematol
.
1993
;
66
(
3
):
127
-
129
.
116.
Liu
M
,
Liu
T
,
Meng
W-T
,
Zhu
H-L
,
Cui
X
.
Role of gammadeltaT cells in pathogenesis of acquired pure red cell aplastic anemia (article in Chinese)
.
Zhongguo Shi Yan Xue Ye Xue Za Zhi
.
2007
;
15
(
1
):
142
-
146
.
117.
Ware
RE
,
Howard
TA
.
Elevated numbers of gamma-delta (gamma delta+) T lymphocytes in children with immune thrombocytopenic purpura
.
J Clin Immunol
.
1994
;
14
(
4
):
237
-
247
.
118.
Goette
NP
,
Glembotsky
AC
,
Lev
PR
, et al
.
Platelet apoptosis in adult immune thrombocytopenia: Insights into the mechanism of damage triggered by auto-antibodies
.
PLoS One
.
2016
;
11
(
8
):
e0160563
.
119.
Weidmann
E
.
Hepatosplenic T cell lymphoma. A review on 45 cases since the first report describing the disease as a distinct lymphoma entity in 1990
.
Leukemia
.
2000
;
14
(
6
):
991
-
997
.
120.
Garderet
L
,
Aoudjhane
M
,
Bonte
H
, et al
.
Immune thrombocytopenic purpura: first symptom of gamma/delta T -cell lymphoma
.
Am J Med
.
2001
;
111
(
3
):
242
-
243
.
121.
Zeng
W
,
Maciejewski
JP
,
Chen
G
, et al
.
Selective reduction of natural killer T cells in the bone marrow of aplastic anaemia
.
Br J Haematol
.
2002
;
119
(
3
):
803
-
809
.
122.
Qiao
X
,
Xie
X
,
Shi
W
,
Tang
J
,
Shao
Y
,
Li
F
.
OCH-mediated shift of Th1 and Th2 cytokines by NKT cells in mice with aplastic anemia
.
Med Oncol
.
2015
;
32
(
3
):
67
.
123.
Xu
R
,
Zheng
Z
,
Ma
Y
, et al
.
Elevated NKT cell levels in adults with severe chronic immune thrombocytopenia
.
Exp Ther Med
.
2014
;
7
(
1
):
149
-
154
.
124.
Johansson
U
,
Macey
MG
,
Kenny
D
,
Provan
D
,
Newland
AC
.
α-galactosylceramide-driven expansion of human natural killer T cells is inhibited by prednisolone treatment
.
Br J Haematol
.
2004
;
125
(
3
):
400
-
404
.
125.
Johansson
U
,
Macey
MG
,
Kenny
D
,
Provan
AB
,
Newland
AC
.
The role of natural killer T (NKT) cells in immune thrombocytopenia: is strong in vitro NKT cell activity related to the development of remission?
Br J Haematol
.
2005
;
129
(
4
):
564
-
565
.
126.
Gascón
P
,
Zoumbos
N
,
Young
N
.
Analysis of natural killer cells in patients with aplastic anemia
.
Blood
.
1986
;
67
(
5
):
1349
-
1355
.
127.
Myint
AA
,
Malkovska
V
,
Morgan
S
,
Luckit
J
,
Wonke
B
,
Gordon-Smith
EC
.
Antilymphocyte globulin therapy enhances impaired function of natural killer cells and lymphokine activated killer cells in aplastic anaemia
.
Br J Haematol
.
1990
;
75
(
4
):
578
-
584
.
128.
Crouse
J
,
Xu
HC
,
Lang
PA
,
Oxenius
A
.
NK cells regulating T cell responses: mechanisms and outcome
.
Trends Immunol
.
2015
;
36
(
1
):
49
-
58
.
129.
Chen
T
,
Zhang
T
,
Liu
C
, et al
.
NK cells suppress CD8+ T cell immunity via NKG2D in severe aplastic anemia
.
Cell Immunol
.
2019
;
335
:
6
-
14
.
130.
Wang
S
,
Liu
F
,
Zhao
H
.
Eosinophil granulocyte and NK cell-mediated platelet destruction in immune thrombocytopenia
.
Indian J Hematol Blood Transfus
.
2018
;
34
(
1
):
121
-
124
.
131.
Yoshimura
C
,
Nomura
S
,
Nagahama
M
,
Ozaki
Y
,
Kagawa
H
,
Fukuhara
S
.
Plasma-soluble Fas (APO-1, CD95) and soluble Fas ligand in immune thrombocytopenic purpura
.
Eur J Haematol
.
2000
;
64
(
4
):
219
-
224
.
132.
Ebbo
M
,
Audonnet
S
,
Grados
A
, et al
.
NK cell compartment in the peripheral blood and spleen in adult patients with primary immune thrombocytopenia
.
Clin Immunol
.
2017
;
177
:
18
-
28
.
133.
Audia
S
,
Moulinet
T
,
Ciudad-Bonté
M
, et al
.
Altered distribution and function of splenic innate lymphoid cells in adult chronic immune thrombocytopenia
.
J Autoimmun
.
2018
;
93
:
139
-
144
.
134.
Bacigalupo
A
.
How I treat acquired aplastic anemia
.
Blood
.
2017
;
129
(
11
):
1428
-
1436
.
135.
Cuker
A
,
Neunert
CE
.
How I treat refractory immune thrombocytopenia
.
Blood
.
2016
;
128
(
12
):
1547
-
1554
.
136.
Chaturvedi
S
,
Arnold
DM
,
McCrae
KR
.
Splenectomy for immune thrombocytopenia: down but not out
.
Blood
.
2018
;
131
(
11
):
1172
-
1182
.
137.
Li
G
,
Wang
S
,
Li
N
, et al
.
Proteasome inhibition with bortezomib induces apoptosis of long-lived plasma cells in steroid-resistant or relapsed immune thrombocytopaenia
.
Thromb Haemost
.
2018
;
118
(
10
):
1752
-
1764
.
138.
Divito
SJ
,
Aasebø
AT
,
Matos
TR
, et al
.
Peripheral host T cells survive hematopoietic stem cell transplantation and promote graft-versus-host disease
.
J Clin Invest
.
2020
;
130
(
9
):
4624
-
4636
.
139.
Nguyen
VH
,
Shashidhar
S
,
Chang
DS
, et al
.
The impact of regulatory T cells on T-cell immunity following hematopoietic cell transplantation
.
Blood
.
2008
;
111
(
2
):
945
-
953
.
140.
Hoffmann
P
,
Ermann
J
,
Edinger
M
,
Fathman
CG
,
Strober
S
.
Donor-type CD4(+)CD25(+) regulatory T cells suppress lethal acute graft-versus-host disease after allogeneic bone marrow transplantation
.
J Exp Med
.
2002
;
196
(
3
):
389
-
399
.
141.
Martin
PJ
.
Prevention of allogeneic marrow graft rejection by donor T cells that do not recognize recipient alloantigens: potential role of a veto mechanism
.
Blood
.
1996
;
88
(
3
):
962
-
969
.
142.
Nakamura
K
,
Inaba
M
,
Sugiura
K
, et al
.
Enhancement of allogeneic hematopoietic stem cell engraftment and prevention of GVHD by intra-bone marrow bone marrow transplantation plus donor lymphocyte infusion
.
Stem Cells
.
2004
;
22
(
2
):
125
-
134
.
143.
Russell
A
,
Malik
S
,
Litzow
M
,
Gastineau
D
,
Roy
V
,
Zubair
AC
.
Dual roles of autologous CD8+ T cells in hematopoietic progenitor cell mobilization and engraftment
.
Transfusion
.
2015
;
55
(
7
):
1758
-
1765
.
144.
Adams
GB
,
Chabner
KT
,
Foxall
RB
, et al
.
Heterologous cells cooperate to augment stem cell migration, homing, and engraftment
.
Blood
.
2003
;
101
(
1
):
45
-
51
.
145.
Geerman
S
,
Nolte
MA
.
Impact of T cells on hematopoietic stem and progenitor cell function: Good guys or bad guys?
World J Stem Cells
.
2017
;
9
(
2
):
37
-
44
.
146.
Zheng
H
,
Matte-Martone
C
,
Jain
D
,
McNiff
J
,
Shlomchik
WD
.
Central memory CD8+ T cells induce graft-versus-host disease and mediate graft-versus-leukemia
.
J Immunol
.
2009
;
182
(
10
):
5938
-
5948
.
147.
Boucault
L
,
Lopez Robles
MD
,
Thiolat
A
, et al
.
Transient antibody targeting of CD45RC inhibits the development of graft-versus-host disease
.
Blood Adv
.
2020
;
4
(
11
):
2501
-
2515
.
148.
Bleakley
M
.
Naive T-cell depletion in stem cell transplantation
.
Blood Adv
.
2020
;
4
(
19
):
4980
.
149.
Yamashita
K
,
Choi
U
,
Woltz
PC
, et al
.
Severe chronic graft-versus-host disease is characterized by a preponderance of CD4(+) effector memory cells relative to central memory cells
.
Blood
.
2004
;
103
(
10
):
3986
-
3988
.
150.
Matthews
K
,
Lim
Z
,
Afzali
B
, et al
.
Imbalance of effector and regulatory CD4 T cells is associated with graft-versus-host disease after hematopoietic stem cell transplantation using a reduced intensity conditioning regimen and alemtuzumab
.
Haematologica
.
2009
;
94
(
7
):
956
-
966
.
151.
Hirata
Y
,
Kakiuchi
M
,
Robson
SC
,
Fujisaki
J
.
CD150high CD4 T cells and CD150high regulatory T cells regulate hematopoietic stem cell quiescence via CD73
.
Haematologica
.
2019
;
104
(
6
):
1136
-
1142
.
152.
McManigle
W
,
Youssef
A
,
Sarantopoulos
S
.
B cells in chronic graft-versus-host disease
.
Hum Immunol
.
2019
;
80
(
6
):
393
-
399
.
153.
Jin
H
,
Ni
X
,
Deng
R
, et al
.
Antibodies from donor B cells perpetuate cutaneous chronic graft-versus-host disease in mice
.
Blood
.
2016
;
127
(
18
):
2249
-
2260
.
154.
Mielcarek
M
,
Leisenring
W
,
Torok-Storb
B
,
Storb
R
.
Graft-versus-host disease and donor-directed hemagglutinin titers after ABO-mismatched related and unrelated marrow allografts: evidence for a graft-versus-plasma cell effect
.
Blood
.
2000
;
96
(
3
):
1150
-
1156
.
155.
Griffith
LM
,
McCoy
JP
Jr.
,
Bolan
CD
, et al
.
Persistence of recipient plasma cells and anti-donor isohaemagglutinins in patients with delayed donor erythropoiesis after major ABO incompatible non-myeloablative haematopoietic cell transplantation
.
Br J Haematol
.
2005
;
128
(
5
):
668
-
675
.
156.
Pai
C-CS
,
Hsiao
H-H
,
Sun
K
, et al
.
Therapeutic benefit of bortezomib on acute graft-versus-host disease is tissue specific and is associated with interleukin-6 levels
.
Biol Blood Marrow Transplant
.
2014
;
20
(
12
):
1899
-
1904
.
157.
Pai
C-CS
,
Chen
M
,
Mirsoian
A
, et al
.
Treatment of chronic graft-versus-host disease with bortezomib
.
Blood
.
2014
;
124
(
10
):
1677
-
1688
.
158.
Minculescu
L
,
Sengeløv
H
.
The role of gamma delta T cells in haematopoietic stem cell transplantation
.
Scand J Immunol
.
2015
;
81
(
6
):
459
-
468
.
159.
Hirokawa
M
,
Horiuchi
T
,
Kawabata
Y
,
Kitabayashi
A
,
Miura
AB
.
Reconstitution of gammadelta T cell repertoire diversity after human allogeneic hematopoietic cell transplantation and the role of peripheral expansion of mature T cell population in the graft
.
Bone Marrow Transplant
.
2000
;
26
(
2
):
177
-
185
.
160.
Fujishima
N
,
Hirokawa
M
,
Fujishima
M
, et al
.
Skewed T cell receptor repertoire of Vdelta1(+) gammadelta T lymphocytes after human allogeneic haematopoietic stem cell transplantation and the potential role for Epstein-Barr virus-infected B cells in clonal restriction
.
Clin Exp Immunol
.
2007
;
149
(
1
):
70
-
79
.
161.
Godder
KT
,
Henslee-Downey
PJ
,
Mehta
J
, et al
.
Long term disease-free survival in acute leukemia patients recovering with increased gammadelta T cells after partially mismatched related donor bone marrow transplantation
.
Bone Marrow Transplant
.
2007
;
39
(
12
):
751
-
757
.
162.
Hu
B
,
Bao
G
,
Zhang
Y
, et al
.
Donor NK Cells and IL-15 promoted engraftment in nonmyeloablative allogeneic bone marrow transplantation
.
J Immunol
.
2012
;
189
(
4
):
1661
-
1670
.
163.
Escobedo-Cousin
M
,
Jackson
N
,
Laza-Briviesca
R
, et al
.
Natural killer cells improve hematopoietic stem cell engraftment by increasing stem cell clonogenicity in vitro and in a humanized mouse model
.
PLoS One
.
2015
;
10
(
10
):
e0138623
.
164.
Schneidawind
D
,
Pierini
A
,
Alvarez
M
, et al
.
CD4+ invariant natural killer T cells protect from murine GVHD lethality through expansion of donor CD4+CD25+FoxP3+ regulatory T cells
.
Blood
.
2014
;
124
(
22
):
3320
-
3328
.
165.
Munneke
JM
,
Björklund
AT
,
Mjösberg
JM
, et al
.
Activated innate lymphoid cells are associated with a reduced susceptibility to graft-versus-host disease
.
Blood
.
2014
;
124
(
5
):
812
-
821
.
166.
van Hoeven
V
,
Munneke
JM
,
Cornelissen
AS
, et al
.
Mesenchymal stromal cells stimulate the proliferation and IL-22 production of group 3 innate lymphoid cells
.
J Immunol
.
2018
;
201
(
4
):
1165
-
1173
.
167.
Tikhonova
AN
,
Dolgalev
I
,
Hu
H
, et al
.
The bone marrow microenvironment at single-cell resolution
.
Nature
.
2019
;
569
(
7755
):
222
-
228
.
168.
Schürch
CM
,
Bhate
SS
,
Barlow
GL
, et al
.
Coordinated cellular neighborhoods orchestrate antitumoral immunity at the colorectal cancer invasive front
.
Cell
.
2020
;
182
(
5
):
1341
-
1359.e19
.
169.
Baccin
C
,
Al-Sabah
J
,
Velten
L
, et al
.
Combined single-cell and spatial transcriptomics reveal the molecular, cellular and spatial bone marrow niche organization
.
Nat Cell Biol
.
2020
;
22
(
1
):
38
-
48
.

Supplemental data

Sign in via your Institution