Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Gut ; 64(7): 1072-81, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25298539

ABSTRACT

OBJECTIVE: Under both physiological and pathological conditions, bone volume is determined by the rate of bone formation by osteoblasts and bone resorption by osteoclasts. Excessive bone loss is a common complication of human IBD whose mechanisms are not yet completely understood. Despite the role of activated CD4(+) T cells in inflammatory bone loss, the nature of the T cell subsets involved in this process in vivo remains unknown. The aim of the present study was to identify the CD4(+) T cell subsets involved in the process of osteoclastogenesis in vivo, as well as their mechanism of action. DESIGN: CD4(+) T cells were studied in IL10-/- mice and Rag1-/- mice adoptively transferred with naive CD4(+)CD45RB(high) T cells, representing two well-characterised animal models of IBD and in patients with Crohn's disease. They were phenotypically and functionally characterised by flow cytometric and gene expression analysis, as well as in in vitro cocultures with osteoclast precursors. RESULTS: In mice, we identified bone marrow (BM) CD4(+) T cells producing interleukin (IL)-17 and tumour necrosis factor (TNF)-α as an osteoclastogenic T cell subset referred to as Th17 TNF-α(+) cells. During chronic inflammation, these cells migrate to the BM where they survive in an IL-7-dependent manner and where they promote the recruitment of inflammatory monocytes, the main osteoclast progenitors. A population equivalent to the Th17 TNF-α(+) cells was also detected in patients with Crohn's disease. CONCLUSIONS: Our results highlight the osteoclastogenic function of the Th17 TNF-α(+) cells that contribute to bone loss in vivo in IBD.


Subject(s)
Bone Diseases/physiopathology , Bone Marrow Cells/physiology , Inflammatory Bowel Diseases/physiopathology , Osteoclasts/physiology , T-Lymphocyte Subsets/physiology , Th17 Cells/physiology , Adaptive Immunity/physiology , Animals , Bone Diseases/immunology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation , Crohn Disease/immunology , Crohn Disease/physiopathology , Disease Models, Animal , Humans , Immunohistochemistry , Inflammatory Bowel Diseases/immunology , Interleukin-7/physiology , Mice, Inbred BALB C , Mice, Inbred C57BL , Osteoclasts/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th17 Cells/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/physiology
2.
Arch Biochem Biophys ; 561: 29-37, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-24998177

ABSTRACT

Bone marrow is the major site of hematopoiesis in mammals. The bone marrow environment plays an essential role in the regulation of hematopoietic stem and progenitor cells by providing specialized niches in which these cells are maintained. Many cell types participate to the composition and regulation of hematopoietic stem cell (HSC) niches, integrating complex signals from the bone, immune and nervous systems. Among these cells, the bone-resorbing osteoclasts (OCLs) have been described as main regulators of HSC niches. They are not limited to carving space for HSCs, but they also provide signals that affect the molecular and cellular niche components. However, their exact role in HSC niches remains unclear because of the variety of models, signals and conditions used to address the question. The present review will discuss the importance of the implication of OCLs focusing on the formation of HSC niches, the maintenance of HSCs in these niches and the mobilization of HSCs from the bone marrow. It will underline the importance of OCLs in HSC niches.


Subject(s)
Bone Development/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Osteoclasts/cytology , Osteoclasts/physiology , Stem Cell Niche/physiology , Animals , Cell Differentiation , Cell Movement/physiology , Feedback, Physiological/physiology , Humans
3.
Elife ; 122023 02 27.
Article in English | MEDLINE | ID: mdl-36848406

ABSTRACT

Bone destruction is a hallmark of chronic inflammation, and bone-resorbing osteoclasts arising under such a condition differ from steady-state ones. However, osteoclast diversity remains poorly explored. Here, we combined transcriptomic profiling, differentiation assays and in vivo analysis in mouse to decipher specific traits for inflammatory and steady-state osteoclasts. We identified and validated the pattern-recognition receptors (PRR) Tlr2, Dectin-1, and Mincle, all involved in yeast recognition as major regulators of inflammatory osteoclasts. We showed that administration of the yeast probiotic Saccharomyces boulardii CNCM I-745 (Sb) in vivo reduced bone loss in ovariectomized but not sham mice by reducing inflammatory osteoclastogenesis. This beneficial impact of Sb is mediated by the regulation of the inflammatory environment required for the generation of inflammatory osteoclasts. We also showed that Sb derivatives as well as agonists of Tlr2, Dectin-1, and Mincle specifically inhibited directly the differentiation of inflammatory but not steady-state osteoclasts in vitro. These findings demonstrate a preferential use of the PRR-associated costimulatory differentiation pathway by inflammatory osteoclasts, thus enabling their specific inhibition, which opens new therapeutic perspectives for inflammatory bone loss.


Subject(s)
Osteoporosis , Probiotics , Animals , Mice , Osteogenesis , Osteoporosis/therapy , Toll-Like Receptor 2 , Saccharomyces/genetics , Saccharomyces/metabolism
4.
J Immunol ; 185(12): 7165-73, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21078911

ABSTRACT

Progressing tumors in humans and mice are frequently infiltrated by a highly heterogeneous population of inflammatory myeloid cells that contribute to tumor growth. Among these cells, inflammatory Gr-1(+) monocytes display a high developmental plasticity in response to specific microenvironmental signals, leading to diverse immune functions. These observations raise the question of the immune mechanisms by which inflammatory monocytes may contribute to tumor development. In this study, we found that adoptive transfer of normal inflammatory Gr-1(+) monocytes in tumor-bearing mice promotes tumor growth. In this tumoral environment, these monocytes can differentiate into tolerogenic dendritic cells (DCs) that produce IL-10 and potently induce regulatory T cell responses in vivo. Moreover, diverting the differentiation of Gr-1(+) monocytes into tolerogenic DCs by forced expression of IL-10 soluble receptor and IL-3 in tumor cells improves host immunosurveillance by reducing the regulatory T cell frequency and by inducing immunogenic DCs in the tumor. As a consequence, tumor growth is strongly reduced. Our findings indicate that Gr-1(+) monocytes represent a valuable target for innovative immunotherapeutic strategies against cancer.


Subject(s)
Adenocarcinoma/immunology , Cell Differentiation/immunology , Colonic Neoplasms/immunology , Dendritic Cells/immunology , Immune Tolerance , Immunologic Surveillance , Monocytes/immunology , Adenocarcinoma/pathology , Animals , Colonic Neoplasms/pathology , Dendritic Cells/pathology , Interleukin-10/immunology , Mice , Mice, Inbred BALB C , Monocytes/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
5.
Cell Rep ; 39(11): 110949, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35705045

ABSTRACT

Despite the ubiquitous function of macrophages across the body, the diversity, origin, and function of adrenal gland macrophages remain largely unknown. We define the heterogeneity of adrenal gland immune cells using single-cell RNA sequencing and use genetic models to explore the developmental mechanisms yielding macrophage diversity. We define populations of monocyte-derived and embryonically seeded adrenal gland macrophages and identify a female-specific subset with low major histocompatibility complex (MHC) class II expression. In adulthood, monocyte recruitment dominates adrenal gland macrophage maintenance in female mice. Adrenal gland macrophage sub-tissular distribution follows a sex-dimorphic pattern, with MHC class IIlow macrophages located at the cortico-medullary junction. Macrophage sex dimorphism depends on the presence of the cortical X-zone. Adrenal gland macrophage depletion results in altered tissue homeostasis, modulated lipid metabolism, and decreased local aldosterone production during stress exposure. Overall, these data reveal the heterogeneity of adrenal gland macrophages and point toward sex-restricted distribution and functions of these cells.


Subject(s)
Adrenal Glands , Macrophages , Monocytes , Sex Characteristics , Adrenal Glands/metabolism , Animals , Female , Histocompatibility Antigens Class II/genetics , Leukocyte Count , Macrophages/metabolism , Male , Mice
6.
Blood ; 112(13): 5074-83, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18768394

ABSTRACT

Finding that activated T cells control osteoclast (OCL) differentiation has revealed the importance of the interactions between immune and bone cells. Dendritic cells (DCs) are responsible for T-cell activation and share common precursors with OCLs. Here we show that DCs participate in bone resorption more directly than simply through T-cell activation. We show that, among the splenic DC subsets, the conventional DCs have the higher osteoclastogenic potential in vitro. We demonstrate that conventional DCs differentiate into functional OCLs in vivo when injected into osteopetrotic oc/oc mice defective in OCL resorptive function. Moreover, this differentiation involves the presence of activated CD4(+) T cells controlling a high RANK-L expression by bone marrow stromal cells. Our results open new insights in the differentiation of OCLs and DCs and offer new basis for analyzing the relations between bone and immune systems.


Subject(s)
Bone Marrow/physiology , Cell Differentiation , Dendritic Cells/cytology , Osteoclasts/cytology , Stem Cell Niche/cytology , Animals , Bone Resorption , Dendritic Cells/immunology , Lymphocyte Activation , Mice , RANK Ligand/biosynthesis , Stromal Cells/metabolism , T-Lymphocytes
7.
Elife ; 92020 05 13.
Article in English | MEDLINE | ID: mdl-32400390

ABSTRACT

Bone destruction relies on interactions between bone and immune cells. Bone-resorbing osteoclasts (OCLs) were recently identified as innate immune cells activating T cells toward tolerance or inflammation. Thus, pathological bone destruction not only relies on increased osteoclast differentiation, but also on the presence of inflammatory OCLs (i-OCLs), part of which express Cx3cr1. Here, we investigated the contribution of mouse Cx3cr1+ and Cx3cr1neg i-OCLs to bone loss. We showed that Cx3cr1+ and Cx3cr1neg i-OCLs differ considerably in transcriptional and functional aspects. Cx3cr1neg i-OCLs have a high ability to resorb bone and activate inflammatory CD4+ T cells. Although Cx3cr1+ i-OCLs are associated with inflammation, they resorb less and have in vitro an immune-suppressive effect on Cx3cr1neg i-OCLs, mediated by PD-L1. Our results provide new insights into i-OCL heterogeneity. They also reveal that different i-OCL subsets may interact to regulate inflammation. This contributes to a better understanding and prevention of inflammatory bone destruction.


Subject(s)
Bone Resorption/metabolism , CX3C Chemokine Receptor 1/metabolism , Inflammation/metabolism , Osteoclasts/metabolism , Osteogenesis , Osteoporosis/metabolism , Animals , Bone Resorption/immunology , Bone Resorption/pathology , Bone Resorption/prevention & control , CX3C Chemokine Receptor 1/genetics , Cell Communication , Cells, Cultured , Female , Inflammation/immunology , Inflammation/pathology , Inflammation/prevention & control , Mice, Inbred C57BL , Mice, Knockout , Osteoclasts/immunology , Osteoclasts/pathology , Osteoporosis/immunology , Osteoporosis/pathology , Osteoporosis/prevention & control , Ovariectomy , Phenotype , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
Joint Bone Spine ; 86(1): 43-47, 2019 01.
Article in English | MEDLINE | ID: mdl-29654948

ABSTRACT

The gut microbiome is now viewed as a tissue that interacts bidirectionally with the gastrointestinal, immune, endocrine and nervous systems, affecting the cellular responses in numerous organs. Evidence is accumulating of gut microbiome involvement in a growing number of pathophysiological processes, many of which are linked to inflammatory responses. More specifically, data acquired over the last decade point to effects of the gut microbiome on bone mass regulation and on the development of bone diseases (such as osteoporosis) and of inflammatory joint diseases characterized by bone loss. Mice lacking a gut microbiome have bone mass alteration that can be reversed by gut recolonization. Changes in the gut microbiome composition have been reported in mice with estrogen-deficiency osteoporosis and have also been found in a few studies in humans. Probiotic therapy decreases bone loss in estrogen-deficient animals. The effect of the gut microbiome on bone tissue involves complex mechanisms including modulation of CD4+T cell activation, control of osteoclastogenic cytokine production and modifications in hormone levels. This complexity may contribute to explain the discrepancies observed betwwen some studies whose results vary depending on the age, gender, genetic background and treatment duration. Further elucidation of the mechanisms involved is needed. However, the available data hold promise that gut microbiome manipulation may prove of interest in the management of bone diseases.


Subject(s)
Bone and Bones/immunology , Gastrointestinal Microbiome/immunology , Osteoclasts/immunology , Osteogenesis/immunology , Osteoporosis/immunology , Animals , Bone and Bones/microbiology , Cell Differentiation/immunology , Humans , Mice , Osteoporosis/microbiology , Osteoporosis/physiopathology
9.
Front Immunol ; 10: 1408, 2019.
Article in English | MEDLINE | ID: mdl-31275328

ABSTRACT

Osteoclasts (OCLs) are key players in controlling bone remodeling. Modifications in their differentiation or bone resorbing activity are associated with a number of pathologies ranging from osteopetrosis to osteoporosis, chronic inflammation and cancer, that are all characterized by immunological alterations. Therefore, the 2000s were marked by the emergence of osteoimmunology and by a growing number of studies focused on the control of OCL differentiation and function by the immune system. At the same time, it was discovered that OCLs are much more than bone resorbing cells. As monocytic lineage-derived cells, they belong to a family of cells that displays a wide heterogeneity and plasticity and that is involved in phagocytosis and innate immune responses. However, while OCLs have been extensively studied for their bone resorption capacity, their implication as immune cells was neglected for a long time. In recent years, new evidence pointed out that OCLs play important roles in the modulation of immune responses toward immune suppression or inflammation. They unlocked their capacity to modulate T cell activation, to efficiently process and present antigens as well as their ability to activate T cell responses in an antigen-dependent manner. Moreover, similar to other monocytic lineage cells such as macrophages, monocytes and dendritic cells, OCLs display a phenotypic and functional plasticity participating to their anti-inflammatory or pro-inflammatory effect depending on their cell origin and environment. This review will address this novel vision of the OCL, not only as a phagocyte specialized in bone resorption, but also as innate immune cell participating in the control of immune responses.


Subject(s)
Disease Susceptibility , Immunomodulation , Osteoclasts/immunology , Osteoclasts/metabolism , Animals , Antigen Presentation , Biomarkers , Bone Remodeling/immunology , Bone Resorption/immunology , Bone Resorption/metabolism , Cell Differentiation , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Lymphocyte Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Monocytes/immunology , Monocytes/metabolism , Osteoclasts/pathology , Phenotype
10.
Front Immunol ; 9: 2567, 2018.
Article in English | MEDLINE | ID: mdl-30450105

ABSTRACT

Osteoclasts (OCLs) are multinucleated phagocytes of monocytic origin responsible for physiological and pathological bone resorption including aging processes, chronic inflammation and cancer. Besides bone resorption, they are also involved in the modulation of immune responses and the regulation of hematopoietic niches. Accordingly, OCLs are the subject of an increasing number of studies. Due to their rarity and the difficulty to isolate them directly ex vivo, analyses on OCLs are usually performed on in vitro differentiated cells. In this state, however, OCLs represent a minority of differentiated cells. Since up to date a reliable purification procedure is still lacking for mature OCLs, all cells present in the culture are analyzed collectively to answer OCL-specific questions. With the development of in-depth transcriptomic and proteomic analyses, such global analyses on unsorted cells can induce severe bias effects in further results. In addition, for instance, analysis on OCL immune function requires working on purified OCLs to avoid contamination effects of monocytic precursors that may persist during the culture. This clearly highlights the need for a reliable OCL purification procedure. Here, we describe a novel and reliable method to sort OCLs based on cell multinucleation while preserving cell viability. Using this method, we successfully purified multinucleated murine cells. We showed that they expressed high levels of OCL markers and retained a high capacity of bone resorption, demonstrating that these are mature OCLs. The same approach was equally applied for the purification of human mature OCLs. Comparison of purified OCLs with mononucleated cells or unsorted cells revealed significant differences in the expression of OCL-specific markers at RNA and/or protein level. This exemplifies that substantially better outcomes for OCLs are achieved after the exclusion of mononucleated cells. Our results clearly demonstrate that the in here presented procedure for the analysis and sorting of pure OCLs represents a novel, robust and reliable method for the detailed examination of bona fide mature OCLs in a range that was previously impossible. Noteworthy, this procedure will open new perspectives into the biology of osteoclasts and osteoclast-related diseases.


Subject(s)
Aging/physiology , Bone Marrow Cells/physiology , Bone Resorption/pathology , Cell Separation/methods , Inflammation/pathology , Osteoclasts/physiology , Animals , Cell Culture Techniques , Cell Differentiation , Cells, Cultured , Hematopoiesis , Humans , Mice , Mice, Inbred C57BL , Reproducibility of Results
11.
J Am Coll Cardiol ; 71(2): 160-172, 2018 01 16.
Article in English | MEDLINE | ID: mdl-29325640

ABSTRACT

BACKGROUND: Several epidermal growth factor receptor (EGFR) inhibitors have been successfully developed for the treatment of cancer, limiting tumor growth and metastasis. EGFR is also expressed by leukocytes, but little is known about its role in the modulation of the immune response. OBJECTIVES: The aim of this study was to determine whether EGFR expressed on CD4+ T cells is functional and to address the consequences of EGFR inhibition in atherosclerosis, a T cell-mediated vascular chronic inflammatory disease. METHODS: The authors used EGFR tyrosine kinase inhibitors (AG-1478, erlotinib) and chimeric Ldlr-/-Cd4-Cre/Egfrlox/lox mouse with a specific deletion of EGFR in CD4+ T cells. RESULTS: Mouse CD4+ T cells expressed EGFR, and the EGFR tyrosine kinase inhibitor AG-1478 blocked in vitro T cell proliferation and Th1/Th2 cytokine production. In vivo, treatment of Ldlr-/- mice with the EGFR inhibitor erlotinib induced T cell anergy, reduced T cell infiltration within atherosclerotic lesions, and protected against atherosclerosis development and progression. Selective deletion of EGFR in CD4+ T cells resulted in decreased T cell proliferation and activation both in vitro and in vivo, as well as reduced interferon-γ, interleukin-4, and interleukin-2 production. Atherosclerotic lesion size was reduced by 2-fold in irradiated Ldlr-/- mice reconstituted with bone marrow from Cd4-Cre/Egfrlox/lox mice, compared to Cd4-Cre/Egfr+/+ chimeric mice, after 4, 6, and 12 weeks of high-fat diet, associated with marked reduction in T cell infiltration in atherosclerotic plaques. Human blood T cells expressed EGFR and EGFR inhibition reduced T cell proliferation both in vitro and in vivo. CONCLUSIONS: EGFR blockade induced T cell anergy in vitro and in vivo and reduced atherosclerosis development. Targeting EGFR may be a novel strategy to combat atherosclerosis.


Subject(s)
Atherosclerosis/immunology , Erlotinib Hydrochloride/pharmacology , Animals , Antineoplastic Agents/pharmacology , CD4-Positive T-Lymphocytes/immunology , Cytokines/analysis , Cytokines/classification , Cytokines/immunology , ErbB Receptors/antagonists & inhibitors , Immunity, Cellular/drug effects , Immunity, Cellular/immunology , Mice , Plaque, Atherosclerotic/drug therapy , Plaque, Atherosclerotic/immunology , Plaque, Atherosclerotic/pathology , Protein Kinase Inhibitors/pharmacology , T-Lymphocytes, Regulatory/immunology
12.
Exp Hematol ; 34(7): 851-9, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16797412

ABSTRACT

OBJECTIVE: We recently identified in the mouse bone marrow a B-lymphoid/myeloid B220+ CD11b+ progenitor population. This population is accumulated in the osteopetrotic oc/oc mouse, which suggests that it could be controlled by bone marrow factors whose expression varies in this pathologic bone environment. Among the possible factors, interleukin (IL)-7 is involved in the control of B lymphopoiesis and osteoclastogenesis. Therefore, we hypothesized that IL-7 could regulate the accumulation of the B220+ CD11b+ population in oc/oc mice. METHODS: B220+ CD11b+ cells sorted from oc/oc mice were treated with IL-7 and their phenotype was analyzed by flow cytometry and real-time reverse transcriptase polymerase chain reaction (RT-PCR). In vivo, IL-7 was injected in oc/oc mice, and B220+ CD11b+ and B cells, as well as B-cell proliferation and apoptosis, were analyzed by flow cytometry. The expression of B lymphopoiesis and myelopoiesis markers was analyzed by real-time RT-PCR. RESULTS: In vitro, IL-7 induced the differentiation of B220+ CD11b+ cells into B lymphocytes through the induction of Pax5 and the inhibition of myeloid markers. In vivo, IL-7 injections in oc/oc mice induced a decrease of the B220+ CD11b+ population and the partial restoration of B-cell population, which was reduced in oc/oc mice. In parallel, upon IL-7 injections, Pax5 expression was induced in B220+ cells and B-cell apoptosis was reduced. CONCLUSIONS: Our results demonstrate that IL-7 injection can partially rescue B lymphopoiesis in oc/oc mice through the engagement of the B220+ CD11b+ population in the B-lymphoid pathway. Therefore, IL-7 delivery could represent a new therapeutic perspective to circumvent the lymphopenia observed in infantile malignant osteopetrosis patients.


Subject(s)
B-Lymphocytes/drug effects , CD11b Antigen/immunology , Interleukin-7/therapeutic use , Leukocyte Common Antigens/immunology , Osteopetrosis/pathology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Base Sequence , Cell Lineage , DNA Primers , Flow Cytometry , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction
13.
Front Immunol ; 8: 954, 2017.
Article in English | MEDLINE | ID: mdl-28848556

ABSTRACT

Multiple myeloma (MM) is one of the most common forms of hematologic malignancy resulting from cancerous proliferation of mature malignant plasma cells (MPCs). But despite the real improvement in therapeutics in the past years, it remains largely incurable. MM is the most frequent cancer to involve bone due to the stimulation of osteoclast (OCL) differentiation and activity. OCLs have a unique capacity to resorb bone. However, recent studies reveal that they are not restrained to this sole function. They participate in the control of angiogenesis, medullary niches, and immune responses, including in MM. Therefore, therapeutic approaches targeting OCLs probably affect not only bone resorption but also many other functions, and OCLs should not be considered anymore only as targets to improve the bone phenotype but also to modulate bone microenvironment. In this review, we explore these novel contributions of OCLs to MM which reveal their strong implication in the MM physiopathology. We also underline the therapeutic interest of targeting OCLs not only to overcome bone lesions, but also to improve bone microenvironment and anti-tumoral immune responses.

14.
Front Immunol ; 8: 1991, 2017.
Article in English | MEDLINE | ID: mdl-29422893

ABSTRACT

Despite mesenchymal stromal cells (MSCs) are considered as a promising source of cells to modulate immune functions on cells from innate and adaptive immune systems, their clinical use remains restricted (few number, limited in vitro expansion, absence of a full phenotypic characterization, few insights on their in vivo fate). Standardized MSCs derived in vitro from human-induced pluripotent stem (huIPS) cells, remediating part of these issues, are considered as well as a valuable tool for therapeutic approaches, but their functions remained to be fully characterized. We generated multipotent MSCs derived from huiPS cells (huiPS-MSCs), and focusing on their immunosuppressive activity, we showed that human T-cell activation in coculture with huiPS-MSCs was significantly reduced. We also observed the generation of functional CD4+ FoxP3+ regulatory T (Treg) cells. Further tested in vivo in a model of human T-cell expansion in immune-deficient NSG mice, huiPS-MSCs immunosuppressive activity prevented the circulation and the accumulation of activated human T cells. Intracytoplasmic labeling of cytokines produced by the recovered T cells showed reduced percentages of human-differentiated T cells producing Th1 inflammatory cytokines. By contrast, T cells producing IL-10 and FoxP3+-Treg cells, absent in non-treated animals, were detected in huiPS-MSCs treated mice. For the first time, these results highlight the immunosuppressive activity of the huiPS-MSCs on human T-cell stimulation with a concomitant generation of human Treg cells in vivo. They may favor the development of new tools and strategies based on the use of huiPS cells and their derivatives for the induction of immune tolerance.

15.
J Bone Miner Res ; 31(10): 1899-1908, 2016 10.
Article in English | MEDLINE | ID: mdl-27161765

ABSTRACT

Bone destruction is a hallmark of chronic rheumatic diseases. Although the role of osteoclasts in bone loss is clearly established, their implication in the inflammatory response has not been investigated despite their monocytic origin. Moreover, specific markers are lacking to characterize osteoclasts generated in inflammatory conditions. Here, we have explored the phenotype of inflammatory osteoclasts and their effect on CD4+ T cell responses in the context of bone destruction associated with inflammatory bowel disease. We used the well-characterized model of colitis induced by transfer of naive CD4+ T cells into Rag1-/- mice, which is associated with severe bone destruction. We set up a novel procedure to sort pure osteoclasts generated in vitro to analyze their phenotype and specific immune responses by FACS and qPCR. We demonstrated that osteoclasts generated from colitic mice induced the emergence of TNFα-producing CD4+ T cells, whereas those generated from healthy mice induced CD4+ FoxP3+ regulatory T cells, in an antigen-dependent manner. This difference is related to the osteoclast origin from monocytes or dendritic cells, to their cytokine expression pattern, and their environment. We identified CX3 CR1 as a marker of inflammatory osteoclasts and we demonstrated that the differentiation of CX3 CR1+ osteoclasts is controlled by IL-17 in vitro. This work is the first demonstration that, in addition to participating to bone destruction, osteoclasts also induce immunogenic CD4+ T cell responses upon inflammation. They highlight CX3 CR1 as a novel dual target for antiresorptive and anti-inflammatory treatment in inflammatory chronic diseases. © 2016 American Society for Bone and Mineral Research.


Subject(s)
Bone Resorption/metabolism , CD4-Positive T-Lymphocytes/metabolism , CX3C Chemokine Receptor 1/biosynthesis , Gene Expression Regulation , Inflammatory Bowel Diseases/metabolism , Osteoclasts/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Bone Resorption/etiology , Bone Resorption/genetics , Bone Resorption/pathology , CD4-Positive T-Lymphocytes/pathology , CX3C Chemokine Receptor 1/genetics , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Mice , Mice, Knockout , Osteoclasts/pathology , Tumor Necrosis Factor-alpha/genetics
16.
Front Immunol ; 6: 640, 2015.
Article in English | MEDLINE | ID: mdl-26734007

ABSTRACT

Osteoimmunology is an interdisciplinary research field dedicated to the study of the crosstalk between the immune and bone systems. CD4(+) T cells are central players in this crosstalk. There is an emerging understanding that CD4(+) T cells play an important role in the bone marrow (BM) under physiological and pathological conditions and modulate the differentiation of bone-resorbing osteoclasts. However, identification of the mechanisms that maintain CD4(+) T cells in the BM is still a matter of investigation. This article describes the CD4(+) T cell populations of the BM and reviews their role as osteoclastogenic population in inflammatory bowel disease.

17.
J Bone Miner Res ; 19(7): 1137-43, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15176997

ABSTRACT

UNLABELLED: Several reports indicate that osteoclasts and B-lymphocytes share a common progenitor. This study focuses on the characterization of this bipotent progenitor from the bone marrow of the osteopetrotic oc/oc mouse, where the bipotent progenitor population is amplified, and of normal mice. INTRODUCTION: Osteoclasts have a myelomonocytic origin, but they can also arise in vitro from pro-B-cells, suggesting that a subset of normal pro-B-cells is uncommitted and may reorient into the myeloid lineage representing a B-lymphoid/osteoclastic progenitor. The aim of this study was to characterize this progenitor population. MATERIALS AND METHODS: The osteopetrotic oc/oc mouse was used as a choice model because it displays an increased number of both osteoclasts and pro-B-cells in the bone marrow. Our results have been confirmed in normal littermates. Bone marrow cells from these animals were analyzed by flow cytometry. After sorting, the cells were cultured under different conditions to assess their differentiation capacity. RESULTS: Pro-B-cells from oc/oc and normal mice include an unusual biphenotypic population expressing markers from the B-lymphoid (CD19, CD43, CD5) and the myeloid (F4/80) lineages. This population also expresses progenitor markers (CD34 and Flt3) and is uncommitted. After sorting from the oc/oc bone marrow, this population is able to differentiate in vitro into osteoclast-like cells in the presence of RANKL and macrophage colony-stimulating factor (M-CSF), into dendritic-like cells in the presence of granulocyte/macrophage colony-stimulating factor (GM-CSF), interleukin (IL)-4, and TNFalpha, and into immature B-cells when seeded onto ST2 cells in the presence of IL-7. CONCLUSION: Our results show the existence of a novel bipotent biphenotypic hematopoietic progenitor population present in the bone marrow that has retained the capacity to differentiate into myeloid and B-lymphoid cells.


Subject(s)
B-Lymphocytes/cytology , Hematopoietic Stem Cells/physiology , Osteoclasts/cytology , Osteopetrosis/etiology , Animals , Antigens, Differentiation/analysis , Antigens, Differentiation/metabolism , B-Lymphocytes/metabolism , Carrier Proteins/pharmacology , Cell Differentiation/physiology , Cytokines/pharmacology , Cytokines/physiology , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/drug effects , Macrophage Colony-Stimulating Factor/pharmacology , Membrane Glycoproteins/pharmacology , Membrane Proteins/analysis , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Mutant Strains , Osteoclasts/metabolism , Osteopetrosis/immunology , Osteopetrosis/pathology , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B
18.
J Exp Med ; 209(3): 537-49, 2012 Mar 12.
Article in English | MEDLINE | ID: mdl-22351931

ABSTRACT

Formation of the hematopoietic stem cell (HSC) niche in bone marrow (BM) is tightly associated with endochondral ossification, but little is known about the mechanisms involved. We used the oc/oc mouse, a mouse model with impaired endochondral ossification caused by a loss of osteoclast (OCL) activity, to investigate the role of osteoblasts (OBLs) and OCLs in the HSC niche formation. The absence of OCL activity resulted in a defective HSC niche associated with an increased proportion of mesenchymal progenitors but reduced osteoblastic differentiation, leading to impaired HSC homing to the BM. Restoration of OCL activity reversed the defect in HSC niche formation. Our data demonstrate that OBLs are required for establishing HSC niches and that osteoblastic development is induced by OCLs. These findings broaden our knowledge of the HSC niche formation, which is critical for understanding normal and pathological hematopoiesis.


Subject(s)
Hematopoietic Stem Cells/physiology , Osteoclasts/physiology , Stem Cell Niche/physiology , Animals , Base Sequence , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Cell Differentiation/physiology , Cell Movement/physiology , DNA Primers/genetics , Female , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Osteoblasts/cytology , Osteoblasts/physiology , Osteoclasts/cytology , Osteogenesis/physiology , Osteopetrosis/genetics , Osteopetrosis/pathology , Osteopetrosis/physiopathology , Phenotype , Pregnancy , Vacuolar Proton-Translocating ATPases/genetics , Vacuolar Proton-Translocating ATPases/physiology
19.
Diabetes ; 61(9): 2238-47, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22596049

ABSTRACT

T-cell regulation in adipose tissue provides a link between inflammation and insulin resistance. Because of alterations in adipose tissue T-cell composition in obesity, we aimed to identify the antigen-presenting cells in adipose tissue of obese mice and patients with insulin resistance. Dendritic cells (DCs) and T cells were studied in mice and in two cohorts of obese patients. In lean mice, only CD11c(+) DCs were detected in adipose tissue. Adoptive transfer of naive CD4(+) T cells in Rag1(-/-) mice led to a predominant Th1 response in adipose tissue. In contrast, during obesity DCs (human CD11c(+)CD1c(+) and mouse CD11c(high)F4/80(low)) accumulated in adipose tissue. CD11c(high)F4/80(low) DCs from obese mice induced Th17 differentiation. In patients, the presence of CD11c(+)CD1c(+) DCs correlated with the BMI and with an elevation in Th17 cells. In addition, these DCs led to ex vivo Th17 differentiation. CD1c gene expression further correlated with homeostatic model assessment-insulin resistance in the subcutaneous adipose tissue of obese patients. We show for the first time the presence and accumulation of specific DCs in adipose tissue in mouse and human obesity. These DCs were functional and could be important regulators of adipose tissue inflammation by regulating the switch toward Th17 cell responses in obesity-associated insulin resistance.


Subject(s)
Adipose Tissue/pathology , Dendritic Cells/physiology , Obesity/pathology , Adipose Tissue/physiopathology , Adult , Animals , Cell Differentiation , Female , Humans , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Middle Aged , Obesity/physiopathology , Obesity, Morbid/pathology , T-Lymphocytes, Regulatory/physiology , Th17 Cells/physiology
20.
Cell Res ; 21(7): 1102-15, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21321604

ABSTRACT

B-cell development is dependent on the interactions between B-cell precursors and bone marrow stromal cells, but the role of osteoclasts (OCLs) in this process remains unknown. B lymphocytopenia is a characteristic of osteopetrosis, suggesting a modulation of B lymphopoiesis by OCL activity. To address this question, we first rescued OCL function in osteopetrotic oc/oc mice by dendritic cell transfer, leading to a restoration of both bone phenotype and B-cell development. To further explore the link between OCL activity and B lymphopoiesis, we induced osteopetrosis in normal mice by injections of zoledronic acid (ZA), an inhibitor of bone resorption. B-cell number decreased specifically in the bone marrow of ZA-treated mice. ZA did not directly affect B-cell differentiation, proliferation and apoptosis, but induced a decrease in the expression of CXCL12 and IL-7 by stromal cells, associated with reduced osteoblastic engagement. Equivalent low osteoblastic engagement in oc/oc mice confirmed that it resulted from the reduced OCL activity rather than from a direct effect of ZA on osteoblasts. These dramatic alterations of the bone microenvironment were disadvantageous for B lymphopoiesis, leading to retention of B-cell progenitors outside of their bone marrow niches in the ZA-induced osteopetrotic model. Altogether, our data revealed that OCLs modulate B-cell development in the bone marrow by controlling the bone microenvironment and the fate of osteoblasts. They provide novel basis for the regulation of the retention of B cells in their niche by OCL activity.


Subject(s)
B-Lymphocytes/cytology , Bone Marrow Cells/cytology , Lymphopoiesis , Osteoclasts/cytology , Animals , B-Lymphocytes/drug effects , Bone Density Conservation Agents/pharmacology , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone and Bones/drug effects , Bone and Bones/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Chemokine CXCL12/metabolism , Diphosphonates/pharmacology , Female , Imidazoles/pharmacology , Interleukin-7/metabolism , Lymphopoiesis/drug effects , Mice , Mice, Inbred BALB C , Osteopetrosis/chemically induced , Stromal Cells/drug effects , Stromal Cells/metabolism , Zoledronic Acid
SELECTION OF CITATIONS
SEARCH DETAIL