Phagocytosis of Co-Developing Megakaryocytic Progenitors by Dendritic Cells in the Culture with Thrombopoietin and Tumor Necrosis Factor-α: Possible Role in Hemophagocytic Syndrome.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3082-3082
Author(s):  
Kenichi Sawada ◽  
Makoto Hirokawa ◽  
Kayo Inaba ◽  
Hiroshi Fukaya ◽  
Yoshinari Kawabata ◽  
...  

Abstract Background. Tumor necrosis factor-α (TNF-α) and thrombopoietin (TPO) have been shown to sustain differentiation and proliferation of CD34+ cells toward dendritic cells (DCs) in the presence of multi-acting cytokines. We hypothesized that co-stimulation of TPO and TNF-α generate megakaryocytic progenitors and DCs together from human CD34+ cells and that interaction of these cells may provide a physiological and/or a pathological role of DCs in megakaryopoiesis. Materials and Methods. Highly purified human CD34+ cells were cultured with TPO, with or without TNF-α, in plasma-depleted medium and induced to undergo megakaryocytic differentiation. We enumerated megakaryocytic progenitor cells using the specific markers CD41, CD42b, and CD61, and DCs using CD4, CD11c, CD80, CD83, CD86, and CD123. The character and roles of co-developing non-megakaryocytic cells in the presence of TNF-α were analyzed by fluorescence-activated cell sorter, enzyme immunohistochemistry, confocal microscopy, and autologous mixed lymphocyte reaction. Cytokine production was assessed using a cytometric bead array system. Results. When CD34+ cells were cultured for 7 days in the presence of TPO, the generated cells predominantly expressed CD41 (95±2%), CD42b (54±12%), and CD61 (96±2%), while rarely expressing CD11c (1.6±1.3%), CD80 (0.1±0.1%), CD83 (0.8±0.6%), or CD86 (3.3±1.9%). The addition of TNF-α significantly decreased the number of cells expressing CD41 (3.0±0.6%), CD42b (3.3±1.0%), or CD61 (3.2±0.9%), but did not affect the number of total cells. In the presence of TNF-α, the generated cells expressed major histocompatibility complex (MHC) class I (100%) plus MHC class II (100%). A substantial number of cells became positive for CD11c (37±1%), and even co-stimulatory molecules such as CD80 (2.4±1.9%), CD83 (8±4%), and CD86 (18±7%). Immature CD11c+ DCs were physically associated with apoptotic and CD61+ cells and capable of endocytosing CD61+ cells. Most of the CD11c+ cells co-expressed the c-mpl TPO receptor, CD4, and CD123 and about one half of CD11c+ cells co-expressed CD86. The DCs generated by TNF-α and TPO, but not those by TNF-α alone, facilitated autologous T cell proliferation in some extent, although cytokine production from activated T cells were low. We also confirmed engulfment of CD61+ cells and their fragment by CD11c+ cells in bone marrow cells from patients with hemophagocytic syndromes. Conclusions. This is the first report showing that in the presence of TNF-α, the non-megakaryocytic cells with typical feature of DCs are co-generated from human CD34+ cells during megakaryocytic differentiation by TPO. The CD4+ CD11c+ CD123+ DCs physically associates with and phagocytose developing or dying immature megakaryocytic cells. Similar phenomenon showing engulfment of CD61+ fragment by CD11c+ cells was also observed in bone marrow cells from patients with hemophagocytic syndrome. Therefore, it may be conceivable that DCs with phagocytic activity during the development in bone marrow may play a crucial role in the maintenance of tolerance for self-substances derived from hematopoietic progenitor cells.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1720-1720
Author(s):  
Yoshinobu Saito ◽  
Makoto Hirokawa ◽  
Kunie Saito ◽  
Yong-Mei Guo ◽  
Miwa Hebiguchi ◽  
...  

Abstract Tumor necrosis factor-α (TNF-α) has been shown to sustain differentiation and proliferation of CD34+ cells toward dendritic cells (DCs). Granulocyte-colony stimulating-factor (G-CSF) sustains differentiation and proliferation of CD34+ cells toward neutrophils and has been shown to have immune-modulatory effects. We hypothesized that co-stimulation of G-CSF and TNF-α generate neutrophil progenitors and DCs together from human CD34+ cells and that interaction of these cells may provide physiological and/or a pathological roles in modulating immune response. Methods. Highly purified human CD34+ cells were cultured with G-CSF, with or without TNF-α and induced to undergo differentiation toward neutrophils. We enumerated neutrophil progenitors using the specific marker CD15, and DCs using CD4, CD11c, CD80, CD83, CD86, and CD123. The character and roles of co-developing DCs in the presence of TNF-α were analyzed by fluorescence-activated cell sorter, enzyme immunohistochemistry, confocal microscopy and mixed lymphocyte reaction (MLR). Cytokine production was assessed using a cytometric bead array system. T reguratory cells (Treg) were defined as CD4+CD25+ cells and the cells expressing Fox P3. Results. When CD34+ cells were cultured for 7 days in the presence of G-CSF, the generated cells predominantly expressed CD15 (71.8±0.6%), while rarely expressing CD11c (8.0±2.2%), CD80 (1.4±1.0%), CD83 (2.9±0.5%), or CD86 (5.6±2.9%). The addition of TNF-α significantly decreased the number of cells expressing CD15 (3.5±2.1%), but did not affect the number of total cells. In the presence of TNF-α, the generated cells expressed major histocompatibility complex (MHC) class I (99.5%) plus MHC class II (90.2%). A substantial number of cells became positive for CD11c (70.9±5.3%), and even co-stimulatory molecules such as CD80 (8.0±2.7%), CD83 (15.9±3.0%), and CD86 (39.6±3.2%). Immature CD11c+ DCs were physically associated with apoptotic and CD15+ cells, and capable of endocytosing CD15+ cells. Most of the CD11c+ cells did not co-express the G-CSF receptor, but expressed CD4 and CD123. About one half of CD11c+ cells co-expressed CD86. The DCs generated by TNF-α and G-CSF facilitated alloreactive T cell proliferation in the same extent, although cytokine production from activated T cells were low. Primary MLR facilitated the proliferation of CD4+CD25+ cells and Fox P3+ Treg. The CD4+ CD25+ T cells suppressed secondary MLR, whereas CD4+ CD25− T cells enhanced secondary MLR. Conclusions. This is the first report showing that. the non-neutrophilic cells with typical feature of DCs are co-generated from human CD34+ cells during neutrophil differentiation by G-CSF in the presence of TNF-α. The CD4+ CD11c+ CD123+ DCs physically associate with and phagocytoses developing or dying immature neutrophilic cells. The generated DCs promoted the proliferation of Treg that suppressed secondary MLR. Therefore, it may be conceivable that DCs with phagocytic activity during the development in bone marrow may play a crucial role in the maintenance of tolerance for self-substances derived from hematopoietic progenitor cells.


Blood ◽  
2006 ◽  
Vol 107 (4) ◽  
pp. 1366-1374 ◽  
Author(s):  
Kunie Saito ◽  
Makoto Hirokawa ◽  
Kayo Inaba ◽  
Hiroshi Fukaya ◽  
Yoshinari Kawabata ◽  
...  

Tumor necrosis factor-α (TNF-α) and thrombopoietin (TPO) have been shown to induce the differentiation and proliferation of CD34+ cells toward dendritic cells (DCs) in the presence of multiacting cytokines. We hypothesized that the costimulation of TPO and TNF-α generates megakaryocytic progenitors and DCs together from human CD34+ cells and that the interaction of these cells may indicate a physiologic and/or a pathologic role of DCs in megakaryopoiesis. When highly purified human CD34+ cells were cultured for 7 days with TPO alone, the generated cells expressed megakaryocytic markers, such as CD41, CD42b, and CD61. The addition of TNF-α with TPO remarkably decreased the number of megakaryocytic progenitor cells without affecting the cell yield. Almost half of the cells thus generated expressed CD11c, and most of them were positive for CD4 and CD123. Furthermore, CD11c+ cells were found to capture damaged CD61+ cells and to induce autologous T-cell proliferation, although the cytokine productions were low. We also confirmed an engulfment of CD61+ cells and their fragment by CD11c+ cells in bone marrow cells from patients with hemophagocytic syndrome. These findings suggest that DCs generated under megakaryocytic and inflammatory stimuli are involved in megakaryopoiesis and the subsequent immune responses to self-antigens.


Blood ◽  
2003 ◽  
Vol 101 (1) ◽  
pp. 119-123 ◽  
Author(s):  
Heuy-Ching Wang ◽  
Jolene Dragoo ◽  
Qin Zhou ◽  
John R. Klein

Abstract Recent studies have identified a role for thyroid-stimulating hormone (TSH; ie, thyrotropin) as an inductive signal for tumor necrosis factor-α (TNF-α) secretion by bone marrow (BM) cells, although the features of that activation pathway have not been defined. Using intracellular TSH staining and enzyme-linked immunoassay for detection of secreted TSH, we demonstrate that TSH synthesis in BM cells occurs within CD45+ (leukocyte common antigen) hematopoietic cells and that the majority of that activity resides in a component of CD11b+ BM cells that are not mature T cells, B cells, or Thy-1+ cells in the BM. Conversely, TSH-responsive BM cells defined by expression of TSH receptor (TSHR) using flow cytometry were selectively associated with a nonerythroid CD11b− lymphocyte precursor population. In vitro culture of magnetic-activated cell sorted CD11b− and CD11b+ cells with titrated amounts of purified TSH resulted in significantly higher levels of TNF-α secretion from CD11b− BM cells compared to non-TSH–treated cells, with no appreciable change in TNF-α production from CD11b+cells. These findings are the first to demonstrate TSH production by BM hematopoietic cells, and they demonstrate that TSH may be involved in the regulation of TNF-α by CD11b− BM cells. They also indicate that TSH-mediated regulation of TNF-α secretion within the BM most likely operates through an intrinsic network of TSH production and use between different types of BM cells, and they suggest that local TSH may be an important homeostatic regulator of hematopoiesis mediated by TNF-α.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4132-4132
Author(s):  
Kunie Saito ◽  
Makoto Hirokawa ◽  
Hiroshi Fukaya ◽  
Yoshinari Kawabata ◽  
AtAtsushi Komatsuda ◽  
...  

Abstract Background. Thrombopoietin (TPO) and tumor necrosis factor-α (TNF-α) sustain differentiation and proliferation of CD34+ cells toward dendritic cells (DC) in the presence of multi-acting cytokines. Therefore, we hypothesized that stimulation of human CD34+ cells with TPO and TNF-α might co-develop megakaryocytic progenitors and DC, which may relate to the induction of immune tolerance and autoimmunity in megakaryopoiesis. Materials and Methods. Highly purified human CD34+ cells were cultured in liquid phase with TPO, with or without TNF-α, and induced to undergo megakaryocytic differentiation. We enumerated megakaryocytic progenitor cells using the specific markers CD41, CD42b and CD61, and DC using CD4, CD11c, CD80, CD83, CD86 and CD123. The character and roles of co-developing non-megakaryocytic cells in the presence of TNF-α were analyzed using fluorescent activating cell sorter, enzyme immunohistochemistry, confocal microscopy and autologous mixed lymphocyte reaction (AMLR). Results. When CD34+ cells were cultured for 7 days in the presence of TPO at 100 ng/ml, the generated cells predominantly expressed CD41 (95±2%), CD42b (54±12%) and CD61 (96±2%), while rarely expressed CD11c (1.6±1.3%), CD80 (0.1±0.1%), CD83 (0.8±0.6%) or CD86 (3.3±1.9%). In contrast, addition of TNF-α at 100 ng/ml significantly decreased cells expressing CD41 (3.0±0.6%), CD42b (3.3±1.0%) or CD61 (3.2±0.9%), but did not affect the number of total cells. In the presence of TNF-α, the generated cells expressed HLA class I (100%) and HLA class II (100%), and a substantial number of cells became positive for CD11c (37±1%), even costimulatory molecules, such as CD80 (2.4±1.9%), D83 (8±4%) and CD86 (18±7%). TNF-α induced apoptosis of megakaryocytic cells. Immature CD11c+ DC was physically associated with apoptotic and CD61+ cells and was capable of endocytosing CD61+ cells. All of CD11c+ cells co-expressed c-mpl, CD4 and CD123, and about a half of CD11c+ cells co-expressed CD86. Cells generated by TNF-α and TPO (DC: TPO+TNF-α) induced autologous T cell proliferation in AMLR assay, however, cells generated by TNF-α alone (DC: TNF-α) did not (Figure 1A). Immunophenotypic analysis of both populations showed the higher expression of co-stimulatory molecules such as CD80, CD83 and CD86 in cells generated by TNF-α and TPO (Figure 1B). Conclusions. Non-megakaryocytic cells co-generated from human CD34+ cells during megakaryocytic differentiation in the presence of TPO and TNF-α express DC phenotypes. The CD4+/CD11c+/CD123+ DC subset physically and selectively associates with developing immature megakaryocytic cells and then obtains and captures self-substances and are functional in AMLR. These findings suggest that DC generated from human CD34+ cells under megakaryocytic and inflammatory co-stimuli obtain a functional role and possibly leading to the antigen presentation to induce immunity or tolerance against megakaryocytic cells and/or platelets. Figure Figure


2000 ◽  
Vol 191 (2) ◽  
pp. 275-286 ◽  
Author(s):  
Kanichiro Kobayashi ◽  
Naoyuki Takahashi ◽  
Eijiro Jimi ◽  
Nobuyuki Udagawa ◽  
Masamichi Takami ◽  
...  

Osteoclast differentiation factor (ODF, also called RANKL/TRANCE/OPGL) stimulates the differentiation of osteoclast progenitors of the monocyte/macrophage lineage into osteoclasts in the presence of macrophage colony-stimulating factor (M-CSF, also called CSF-1). When mouse bone marrow cells were cultured with M-CSF, M-CSF–dependent bone marrow macrophages (M-BMMφ) appeared within 3 d. Tartrate-resistant acid phosphatase–positive osteoclasts were also formed when M-BMMφ were further cultured for 3 d with mouse tumor necrosis factor α (TNF-α) in the presence of M-CSF. Osteoclast formation induced by TNF-α was inhibited by the addition of respective antibodies against TNF receptor 1 (TNFR1) or TNFR2, but not by osteoclastogenesis inhibitory factor (OCIF, also called OPG, a decoy receptor of ODF/RANKL), nor the Fab fragment of anti–RANK (ODF/RANKL receptor) antibody. Experiments using M-BMMφ prepared from TNFR1- or TNFR2-deficient mice showed that both TNFR1- and TNFR2-induced signals were important for osteoclast formation induced by TNF-α. Osteoclasts induced by TNF-α formed resorption pits on dentine slices only in the presence of IL-1α. These results demonstrate that TNF-α stimulates osteoclast differentiation in the presence of M-CSF through a mechanism independent of the ODF/RANKL–RANK system. TNF-α together with IL-1α may play an important role in bone resorption of inflammatory bone diseases.


2019 ◽  
Vol 28 (2) ◽  
pp. 99-108 ◽  
Author(s):  
Mio Naritani ◽  
Miho Inoue ◽  
Resmi Raju ◽  
Mayu Miyagi ◽  
Masamitsu Oshima ◽  
...  

1993 ◽  
Vol 177 (6) ◽  
pp. 1531-1539 ◽  
Author(s):  
G E Tjønnfjord ◽  
O P Veiby ◽  
R Steen ◽  
T Egeland

Pluripotent lymphohematopoietic stem cells are probably confined to bone marrow cells expressing CD34 surface molecules. To investigate the capacity of adult human CD34+ bone marrow cells to differentiate along the T lymphoid lineage, we plated purified CD34+ cells from healthy adults in liquid culture on adherent thymic stromal cells prepared from HLA- or blood group-mismatched postnatal thymic tissue. We show that purified CD34+CD3-CD4-CD8- bone marrow cells contained progenitors with the ability to differentiate into CD4+ and CD8+ T lymphocytes expressing surface (s)CD3 and T cell receptor alpha/beta in vitro. These progenitors were found in the CD34+CD2+sCD3-CD4-CD8-, CD34+CD7+sCD3-CD4-CD8-, and CD34+CD2+CD7+sCD3-CD4-CD8-, as well as in the CD34+CD2-sCD3-CD4-CD8-, CD34+CD7-sCD3-CD4-CD8-, and CD34+CD2-CD7-sCD3-CD4-CD8- subsets, indicating that T lymphocyte progenitors sensitive to signals mediated by thymic stroma in vitro are not restricted to CD34+ cells already coexpressing early T lymphocyte-associated markers. Finally, we show that T lymphopoiesis was enhanced by c-kit ligand.


Blood ◽  
2002 ◽  
Vol 100 (2) ◽  
pp. 474-482 ◽  
Author(s):  
Helen A. Papadaki ◽  
Heraklis D. Kritikos ◽  
Vasilis Valatas ◽  
Dimitrios T. Boumpas ◽  
George D. Eliopoulos

Abstract Circumstantial evidence has implicated tumor necrosis factor α (TNF-α) in the pathogenesis of anemia of chronic disease (ACD) in rheumatoid arthritis (RA). We investigated the role of TNF-α in erythropoiesis of patients with active RA (n = 40) and the effect of anti–TNF-α antibody administration (cA2). Patients with RA had lower numbers of CD34+/CD71+ and CD36−/glycophorin A+ (glycoA+) bone marrow (BM) cells and increased proportions of apoptotic cells within the CD34+/CD71+ and CD36+/glycoA+ cell compartments, compared to healthy controls (n = 24). Erythroid burst-forming units (BFU-Es) obtained by BM mononuclear or purified CD34+ cells were significantly lower in RA patients compared to controls. These abnormalities were more pronounced among patients with ACD. Increased TNF-α levels in patient long-term BM culture supernatants inversely correlated with BFU-Es and hemoglobin levels and positively with the percentage of apoptotic CD34+/CD71+ and CD36+/glycoA+ cells. Following cA2 therapy, a normalization was documented in the number of CD34+/CD71+ and CD36−/glycoA+ cells, the number of BFU-Es, and the proportion of apoptotic CD34+/CD71+ and CD36+/glycoA+ cells, which was associated with a significant increase in hemoglobin levels compared to baseline. Recovery from anemia was more prominent in patients with ACD. The exogenous addition of an anti–TNF-α antibody in the cultures increased BFU-E number in patients prior to cA2 treatment but not after treatment, further substantiating the inhibitory role of TNF-α on patients' erythropoiesis. We conclude that TNF-α–mediated apoptotic depletion of BM erythroid cells may account for ACD in RA and that cA2 administration may ameliorate ACD in these patients by down-regulating the apoptotic mechanisms involved in erythropoiesis.


2006 ◽  
Vol 13 (5) ◽  
pp. 575-583 ◽  
Author(s):  
Takuya Itou ◽  
L. Vincent Collins ◽  
Fredrik B. Thorén ◽  
Claes Dahlgren ◽  
Anna Karlsson

ABSTRACT To study different activation states in polymorphonuclear leukocytes (PMN) in mice, we compared the function of murine PMN obtained from the bone marrow (BMPMN) with those of PMN obtained by intraperitoneal induction with thioglycolate (TGPMN) or uric acid (UAPMN). When stimulated with chemotactic peptides, e.g., formyl-methionyl-leucyl-phenylalanine (fMLF), WKYMVM, or WKYMVm, the TGPMN and UAPMN showed greatly enhanced generation of reactive oxygen species (ROS) compared with BMPMN, which suggests that exudation to the peritoneum per se induces a primed state in the cells. The WKYMVm peptide was the most potent stimulant of ROS generation, and it desensitized for subsequent stimulation with fMLF or WKYMVM. This desensitization was broken by the addition of cytochalasin B. The TGPMN and UAPMN appeared to be fully primed, since no increase in response was induced by pretreatment with tumor necrosis factor α (TNF-α). In contrast, the BMPMN response was increased 2.5- to 3-fold. The differences in oxidative responses were supported by degranulation studies. Preincubation with TNF-α promoted CR3 expression on BMPMN, and this level of expression was also enhanced by WKYMVm. In contrast, CR3 expression on untreated TGPMN and UAPMN was already similar to that on TNF-α-primed BMPMN and could be only slightly enhanced by TNF-α treatment. Taken together, these results indicate that BMPMN are in a resting state and have the capacity to become primed, while peritoneal exudate PMN are already fully primed upon isolation. These results have major implications for murine neutrophil research and show the importance of defining which PMN subsets to use when investigating murine models.


Sign in / Sign up

Export Citation Format

Share Document