1
|
Xu H, Li Y, Gao Y. The role of immune cells settled in the bone marrow on adult hematopoietic stem cells. Cell Mol Life Sci 2024; 81:420. [PMID: 39367881 PMCID: PMC11456083 DOI: 10.1007/s00018-024-05445-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 09/09/2024] [Accepted: 09/09/2024] [Indexed: 10/07/2024]
Abstract
Certain immune cells, including neutrophils, macrophages, dendritic cells, B cells, Breg cells, CD4+ T cells, CD8+ T cells, and Treg cells, establish enduring residency within the bone marrow. Their distinctive interactions with hematopoiesis and the bone marrow microenvironment are becoming increasingly recognized alongside their multifaceted immune functions. These cells play a dual role in shaping hematopoiesis. They directly influence the quiescence, self-renewal, and multi-lineage differentiation of hematopoietic stem and progenitor cells through either direct cell-to-cell interactions or the secretion of various factors known for their immunological functions. Additionally, they actively engage with the cellular constituents of the bone marrow niche, particularly mesenchymal stem cells, endothelial cells, osteoblasts, and osteoclasts, to promote their survival and contribute to tissue repair, thereby fostering a supportive environment for hematopoietic stem and progenitor cells. Importantly, these bone marrow immune cells function synergistically, both locally and functionally, rather than in isolation. In summary, immune cells residing in the bone marrow are pivotal components of a sophisticated network of regulating hematopoiesis.
Collapse
Affiliation(s)
- Hui Xu
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, National Clinical Research Center for Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Yinghui Li
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, National Clinical Research Center for Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
| | - Yingdai Gao
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, National Clinical Research Center for Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
| |
Collapse
|
2
|
Brown B, Ojha V, Fricke I, Al-Sheboul SA, Imarogbe C, Gravier T, Green M, Peterson L, Koutsaroff IP, Demir A, Andrieu J, Leow CY, Leow CH. Innate and Adaptive Immunity during SARS-CoV-2 Infection: Biomolecular Cellular Markers and Mechanisms. Vaccines (Basel) 2023; 11:408. [PMID: 36851285 PMCID: PMC9962967 DOI: 10.3390/vaccines11020408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/01/2023] [Accepted: 02/04/2023] [Indexed: 02/16/2023] Open
Abstract
The coronavirus 2019 (COVID-19) pandemic was caused by a positive sense single-stranded RNA (ssRNA) severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, other human coronaviruses (hCoVs) exist. Historical pandemics include smallpox and influenza, with efficacious therapeutics utilized to reduce overall disease burden through effectively targeting a competent host immune system response. The immune system is composed of primary/secondary lymphoid structures with initially eight types of immune cell types, and many other subtypes, traversing cell membranes utilizing cell signaling cascades that contribute towards clearance of pathogenic proteins. Other proteins discussed include cluster of differentiation (CD) markers, major histocompatibility complexes (MHC), pleiotropic interleukins (IL), and chemokines (CXC). The historical concepts of host immunity are the innate and adaptive immune systems. The adaptive immune system is represented by T cells, B cells, and antibodies. The innate immune system is represented by macrophages, neutrophils, dendritic cells, and the complement system. Other viruses can affect and regulate cell cycle progression for example, in cancers that include human papillomavirus (HPV: cervical carcinoma), Epstein-Barr virus (EBV: lymphoma), Hepatitis B and C (HB/HC: hepatocellular carcinoma) and human T cell Leukemia Virus-1 (T cell leukemia). Bacterial infections also increase the risk of developing cancer (e.g., Helicobacter pylori). Viral and bacterial factors can cause both morbidity and mortality alongside being transmitted within clinical and community settings through affecting a host immune response. Therefore, it is appropriate to contextualize advances in single cell sequencing in conjunction with other laboratory techniques allowing insights into immune cell characterization. These developments offer improved clarity and understanding that overlap with autoimmune conditions that could be affected by innate B cells (B1+ or marginal zone cells) or adaptive T cell responses to SARS-CoV-2 infection and other pathologies. Thus, this review starts with an introduction into host respiratory infection before examining invaluable cellular messenger proteins and then individual immune cell markers.
Collapse
Affiliation(s)
| | | | - Ingo Fricke
- Independent Immunologist and Researcher, 311995 Lamspringe, Germany
| | - Suhaila A Al-Sheboul
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid 22110, Jordan
- Department of Medical Microbiology, International School of Medicine, Medipol University-Istanbul, Istanbul 34810, Turkey
| | | | - Tanya Gravier
- Independent Researcher, MPH, San Francisco, CA 94131, USA
| | | | | | | | - Ayça Demir
- Faculty of Medicine, Afyonkarahisar University, Istanbul 03030, Turkey
| | - Jonatane Andrieu
- Faculté de Médecine, Aix–Marseille University, 13005 Marseille, France
| | - Chiuan Yee Leow
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, USM, Penang 11800, Malaysia
| | - Chiuan Herng Leow
- Institute for Research in Molecular Medicine, (INFORMM), Universiti Sains Malaysia, USM, Penang 11800, Malaysia
| |
Collapse
|
3
|
Pastille E, Didovic S, Brauckmann D, Rani M, Agrawal H, Schade FU, Zhang Y, Flohé SB. Modulation of dendritic cell differentiation in the bone marrow mediates sustained immunosuppression after polymicrobial sepsis. THE JOURNAL OF IMMUNOLOGY 2010; 186:977-86. [PMID: 21160046 DOI: 10.4049/jimmunol.1001147] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Murine polymicrobial sepsis is associated with a sustained reduction of dendritic cell (DC) numbers in lymphoid organs and with a dysfunction of DC that is considered to mediate the chronic susceptibility of post-septic mice to secondary infections. We investigated whether polymicrobial sepsis triggered an altered de novo formation and/or differentiation of DC in the bone marrow. BrdU labeling experiments indicated that polymicrobial sepsis did not affect the formation of splenic DC. DC that differentiated from bone marrow (bone marrow-derived DC [BMDC]) of post-septic mice released enhanced levels of IL-10 but did not show an altered phenotype in comparison with BMDC from sham mice. Adoptive transfer experiments of BMDC into naive mice revealed that BMDC from post-septic mice impaired Th1 priming but not Th cell expansion and suppressed the innate immune defense mechanisms against Pseudomonas bacteria in the lung. Accordingly, BMDC from post-septic mice inhibited the release of IFN-γ from NK cells that are critical for the protection against Pseudomonas. Additionally, sepsis was associated with a loss of resident DC in the bone marrow. Depletion of resident DC from bone marrow of sham mice led to the differentiation of BMDC that were impaired in Th1 priming similar to BMDC from post-septic mice. Thus, in response to polymicrobial sepsis, DC precursor cells in the bone marrow developed into regulatory DC that impaired Th1 priming and NK cell activity and mediated immunosuppression. The absence of resident DC in the bone marrow after sepsis might have contributed to the modulation of DC differentiation.
Collapse
Affiliation(s)
- Eva Pastille
- Surgical Research, Department of Trauma Surgery, University Hospital Essen, University Duisburg-Essen, D-45147 Essen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
4
|
Jung ID, Lee JS, Lee CM, Noh KT, Jeong YI, Park WS, Chun SH, Jeong SK, Park JW, Son KH, Heo DR, Lee MG, Shin YK, Kim HW, Yun CH, Park YM. Induction of indoleamine 2,3-dioxygenase expression via heme oxygenase-1-dependant pathway during murine dendritic cell maturation. Biochem Pharmacol 2010; 80:491-505. [PMID: 20430013 DOI: 10.1016/j.bcp.2010.04.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2010] [Revised: 04/19/2010] [Accepted: 04/20/2010] [Indexed: 12/17/2022]
Abstract
Heme oxygenase (HO)-1 is expressed in a variety of conditions involved in the regulation of immune responses. In this study, we examined the role of HO-1 in dendritic cell (DC) maturation and expression of indoleamine 2,3-dioxygenase (IDO), a key enzyme that catalyzes the initial, rate-limiting step in tryptophan degradation. IDO deficiency led to diminished phenotypic and functional maturation of DCs in vitro and in vivo. IDO expression and DC maturation was abrogated by the HO inhibitor zinc protoporphrin, but increased by hemin, a potent inducer of HO-1. Moreover, LPS-induced HO-1 expression was mediated by an NF-kappaB-dependent pathway. Our findings provide additional insight into the immunological functions of IDO and HO-1, and suggest possible therapeutic adjuvants for the treatment of DC-related acute and chronic diseases.
Collapse
Affiliation(s)
- In Duk Jung
- Department of Microbiology, Pusan National University, Beom-eo Ri, Mulgum Eop, Yangsan, Gyeongsangnam-do, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Jung ID, Lee MG, Chang JH, Lee JS, Jeong YI, Lee CM, Park WS, Han J, Seo SK, Lee SY, Park YM. Blockade of Indoleamine 2,3-Dioxygenase Protects Mice against Lipopolysaccharide-Induced Endotoxin Shock. THE JOURNAL OF IMMUNOLOGY 2009; 182:3146-54. [DOI: 10.4049/jimmunol.0803104] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
6
|
Factors determining the formation and release of bioactive IL-12: Regulatory mechanisms for IL-12p70 synthesis and inhibition. Biochem Biophys Res Commun 2008; 372:509-12. [DOI: 10.1016/j.bbrc.2008.05.081] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2008] [Accepted: 05/16/2008] [Indexed: 11/18/2022]
|
7
|
Dendritic cells reach out in different directions. Blood 2007. [DOI: 10.1182/blood-2007-07-099770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
8
|
Li G, Abediankenari S, Kim YJ, Campbell TB, Ito S, Graham-Evans B, Cooper S, Broxmeyer HE. TGF-beta combined with M-CSF and IL-4 induces generation of immune inhibitory cord blood dendritic cells capable of enhancing cytokine-induced ex vivo expansion of myeloid progenitors. Blood 2007; 110:2872-9. [PMID: 17585053 PMCID: PMC2018669 DOI: 10.1182/blood-2006-10-050583] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Tolerogenic dendritic cells (DCs) may be valuable in transplantation for silencing immune reaction. Macrophage colony-stimulating factor (M-CSF)/IL-4 induces differentiation of cord blood (CB) monocytes into DCs (M-DCs) with tolerogenic phenotype/function. We assessed whether factors produced by tolerogenic DCs could modulate hematopoiesis. TGF-beta1 added to CB M-DC cultures induced bona fide DC morphology (TGF-M-DCs), similar to that of DCs generated with TGF-beta and granulocyte-macrophage colony-stimulating factor (GM-CSF)/IL-4 (TGF-GM-DCs). Of conditioned media (CM) produced from TGF-M-DCs, TGF-GM-DCs, M-DCs, and GM-DCs, TGF-M-DC CM was the only one that enhanced SCF, Flt3 ligand, and TPO expansion of myeloid progenitor cells ex vivo. This effect was blocked by neutralizing anti-M-CSF Ab, but protein analysis of CM suggested that M-CSF alone was not manifesting enhanced expansion of myeloid progenitors. LPS-stimulated TGF-M-DCs induced T-cell tolerance/anergy as effectively as M-DCs. TGF-M-DCs secreted significantly lower concentrations of progenitor cell inhibitory cytokines and were less potent in activating T cells than TGF-GM-DCs. Functional differences between TGF-M-DCs and TGF-GM-DCs included enhanced responses to LPS-induced ERK, JNK, and P38 activation in TGF-M-DCs and their immune suppressive-skewed cytokine release profiles. TGF-M-DCs appear unique among culture-generated DCs in their capability for silencing immunity while promoting expansion of myeloid progenitors, events that may be of therapeutic value.
Collapse
Affiliation(s)
- Geling Li
- Department of Microbiology and Immunology, Walther Oncology Center, Indiana University School of Medicine, Indianapolis, IN 46202-5181, USA
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Jung ID, Lee CM, Jeong YI, Lee JS, Park WS, Han J, Park YM. Differential regulation of indoleamine 2,3-dioxygenase by lipopolysaccharide and interferon gamma in murine bone marrow derived dendritic cells. FEBS Lett 2007; 581:1449-56. [PMID: 17367785 DOI: 10.1016/j.febslet.2007.02.073] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2006] [Revised: 02/26/2007] [Accepted: 02/27/2007] [Indexed: 11/17/2022]
Abstract
Indoleamine 2,3-dioxygenase (IDO) is a rate-limiting enzyme in the L-tryptophan-kynurenine pathway, which converts an essential amino acid, L-tryptophan, to N-formylkynurenine. The expression of IDO increases when inflammation is induced by wounding, infection or tumor growth. Although recent studies have suggested that IDO expression is up-regulated by IFN-gamma in various cell types and that the induction of IDO can also be mediated through an IFN-gamma-independent mechanism, these mechanisms still remain unknown. In this study, we investigated whether lipopolysaccharide (LPS) induces the expression of IDO through an IFN-gamma-mediated signaling pathway or not. IFN-gamma-induced expression of IDO expression was inhibited only by JAK inhibitor I. However, LPS-induced expression of IDO was inhibited by LY294002 and SP600125 but not by JAK inhibitor I, SB203580, or U0126. These findings clearly indicate that LPS can induce the IDO expression via an IFN-gamma-independent mechanism and PI3 kinase and JNK in the LPS-induced pathway leading to IDO expression.
Collapse
Affiliation(s)
- In Duk Jung
- Department of Microbiology and Immunology and National Research Laboratory of Dendritic, Cell Differentiation and Regulation, Medical Research Institute, Pusan National University, College of Medicine, Ami-dong 1-10, Seo-gu, Busan 602-739, South Korea
| | | | | | | | | | | | | |
Collapse
|
10
|
Wang Q, Liu Y, Wang J, Ding G, Zhang W, Chen G, Zhang M, Zheng S, Cao X. Induction of allospecific tolerance by immature dendritic cells genetically modified to express soluble TNF receptor. THE JOURNAL OF IMMUNOLOGY 2006; 177:2175-85. [PMID: 16887977 DOI: 10.4049/jimmunol.177.4.2175] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The ability of dendritic cells (DC) to initiate immune responses or induce immune tolerance is strictly dependent on their maturation state. TNF-alpha plays a pivotal role in the differentiation and maturation of DC. Blockade of TNF-alpha action may arrest DC in an immature state, prolonging their window of tolerogenic opportunity. Immature DC (imDC) were transfected with recombinant adenovirus to express soluble TNF-alpha receptor type I (sTNFRI), a specific inhibitor of TNF-alpha. The capacity of sTNFRI gene-modified imDC (DC-sTNFRI) to induce immune tolerance was analyzed. sTNFRI expression renders imDC resistant to maturation induction and impairs their capacity to migrate or present Ag. This process leads to induction of allogeneic T cell hyporesponsiveness and the generation of IL-10-producing T regulatory-like cells. In vivo pretreatment of transplant recipients with DC-sTNFRI induces long-term survival of cardiac allografts in 50% of cases, and leads to a substantial increase in the generation of microchimerism and T regulatory cell numbers. Thus, blockade of TNF-alpha action by sTNFRI genetic modification can inhibit the maturation of DC and potentiate the in vivo capacity of imDC to induce donor-specific immune tolerance and prolong allograft survival.
Collapse
MESH Headings
- Animals
- Antigen Presentation/genetics
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Cells, Cultured
- Dendritic Cells/cytology
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Heart Transplantation/immunology
- Immunity, Innate/genetics
- Lymphocyte Activation/genetics
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Receptors, Tumor Necrosis Factor, Type I/biosynthesis
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/physiology
- Skin Transplantation/immunology
- Solubility
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Transfection/methods
- Transplantation Tolerance/genetics
- Tumor Necrosis Factor-alpha/antagonists & inhibitors
- Tumor Necrosis Factor-alpha/physiology
Collapse
Affiliation(s)
- Quanxing Wang
- Institute of Immunology, Second Military Medical University, 800 Yiangyin Road, Shanghai 200433, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Osuji N, Pettengell R. Growth factors in haematological cancers. Expert Opin Emerg Drugs 2005; 7:175-88. [PMID: 15989543 DOI: 10.1517/14728214.7.1.175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Since their discovery just under a century ago, growth factors (GFs) have been used almost ubiquitously in haematology. Many haematological cancers are associated with bone marrow failure, either as a direct consequence of the disease or its treatment. Colony stimulating factors (CSFs) have been used to address the problems associated with the resulting cytopenias, however, concerns about the potential leukaemogenic effects of some of these CSFs led to a degree of initial hesitancy in usage, particularly in the management of acute myeloid leukaemia (AML). This has now been largely overcome. Other limitations have included cost and side effect profiles (the latter particularly with the multilineage factors). There has been wide variation locally, nationally and internationally in the usage of GFs. The American Society of Clinical Oncologists (ASCO) attempted to rationalise the usage of GFs by producing a consensus document enumerating the evidence-based indications for use of GFs. There is little information on cost effectiveness, this remains an important issue for the future. Peripheral blood stem cell transplantation (PBSCT) has revolutionised the management of many malignant conditions and has contributed to the increased use of growth factors. Many other indications are emerging for GFs used singly or in combination. Current clinical applications of GFs include: i) amelioration of cytopenias following chemotherapy and stem cell transplantation, ii) chemotherapy dose maintenance and escalation, iii) chemosensitisation and modification of disease states, iv) optimisation of methods for mobilisation of progenitor stem cells, v) immunotherapy, and vi) as therapeutic targets for treatment of haematolgical malignancies.
Collapse
Affiliation(s)
- Nnenna Osuji
- Department of Haematology, St. George's Hospital, Cranmer Terrace, London, SW17 0RE, UK.
| | | |
Collapse
|
12
|
Pan J, Zhang M, Wang J, Wang Q, Xia D, Sun W, Zhang L, Yu H, Liu Y, Cao X. Interferon-gamma is an autocrine mediator for dendritic cell maturation. Immunol Lett 2004; 94:141-51. [PMID: 15234546 DOI: 10.1016/j.imlet.2004.05.003] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2004] [Revised: 04/28/2004] [Accepted: 05/01/2004] [Indexed: 11/21/2022]
Abstract
Maturation of dendritic cells (DC) is critical for efficient antigen presentation and initiation of an immune response. Interferon-gamma (IFN-gamma) is an important Th1 cytokine. In this study, we investigated the role of IFN-gamma in DC maturation using either IFN-gamma receptor deficient- or IFN-gamma overexpression-models. We showed that immature DC generated in vitro from bone marrow (BM) progenitor cells produced low level of IFN-gamma. After LPS stimulation, DC produced more IFN-gamma, and IFN-gamma productions were at comparable levels among C57BL/6 mice-derived DC (C57BL/6 DC), wild-type 129 mice-derived DC (129 DC) and IFN-gamma receptor deficient 129 mice-derived DC (IFN-gammaR-/-DC). We found that IFN-gammaR-/-DC exhibited decreased expression of CD54, CD86, reduced capacity to secrete IL-1beta and IL-12p70, and impaired capacity to stimulate alloreactive T cells and to drive Th1 differentiation. Transfection of IFN-gamma gene into DC promoted DC to express higher CD40, CD54, CD80, CD86, CCR7 and I-Ab, secrete more IL-1beta and IL-12p70, and more potently activate both CD4 and CD8 T cells. These data suggest that IFN-gamma signaling pathway is important for the maturation of DC in an autocrine fashion.
Collapse
Affiliation(s)
- Jianping Pan
- Institute of Immunology, Zhejiang University, Hangzhou 310031, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Alves-Rosa F, Vermeulen M, Cabrera J, Stanganelli C, Capozzo A, Narbaitz M, van Rooijen N, Palermo M, Isturiz MA. Macrophage depletion following liposomal-encapsulated clodronate (LIP-CLOD) injection enhances megakaryocytopoietic and thrombopoietic activities in mice. Br J Haematol 2003; 121:130-8. [PMID: 12670343 DOI: 10.1046/j.1365-2141.2003.04259.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Megakaryocytopoiesis is the cellular process by which stem cells progress through commitment, proliferation and differentiation, leading to the production of platelets. In the mouse, this process is accomplished within the bone marrow (BM) and spleen microenvironment and is carried out by regulatory molecules and accessory cells, including macrophages, fibroblasts and endothelial-like cells. Previously, we demonstrated that specific macrophage depletion, using liposomal-encapsulated clodronate (LIP-CLOD), induced a rapid recovery of the platelet count in a mouse model of immune thrombocytopenia. We now show that LIP-CLOD treatment also provoked enhancement of both megakaryocytopoiesis and thrombocytopoiesis. In fact, a dose-dependent increase in the number of BM and spleen megakaryocytes was detected after treatment and this pattern correlated inversely to the macrophage count detected in these organs. Furthermore, the mice treated with the higher dose of LIP-CLOD showed signs of enhanced thrombopoiesis as they had an increased frequency of reticulated platelets and an improvement in the total platelet count 2 d later. In addition, the in vitro cytokine-induced megakaryocytopoiesis in BM and spleen cell cultures was significantly augmented in the presence of LIP-CLOD. Taken together, these results suggest that BM and spleen microenvironmental macrophages could be involved in the regulation of megakaryocyte and platelet production.
Collapse
Affiliation(s)
- Fernanda Alves-Rosa
- División Inmunología, Instituto de Investigaciones Hematológicas, Academia Nacional de Medicina, Pacheco de Melo 3081, 1425 Buenos Aires, Argentina.
| | | | | | | | | | | | | | | | | |
Collapse
|