1
|
Capo V, Abinun M, Villa A. Osteoclast rich osteopetrosis due to defects in the TCIRG1 gene. Bone 2022; 165:116519. [PMID: 35981697 DOI: 10.1016/j.bone.2022.116519] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 11/28/2022]
Abstract
Discovery that mutations in TCIRG1 (also known as Atp6i) gene are responsible for most instances of autosomal recessive osteopetrosis (ARO) heralded a new era for comprehension and treatment of this phenotypically heterogeneous rare bone disease. TCIRG1 encodes the a3 subunit, an essential isoform of the vacuolar ATPase proton pump involved in acidification of the osteoclast resorption lacuna and in secretory lysosome trafficking. TCIRG1 defects lead to inefficient bone resorption by nonfunctional osteoclasts seen in abundance on bone marrow biopsy, delineating this ARO as 'osteoclast-rich'. Presentation is usually in early childhood and features of extramedullary haematopoiesis (hepatosplenomegaly, anaemia, thrombocytopenia) due to bone marrow fibrosis, and cranial nerve impingement (blindness in particular). Impaired dietary calcium uptake due to high pH causes the co-occurrence of rickets, described as "osteopetrorickets". Osteoclast dysfunction leads to early death if untreated, and allogeneic haematopoietic stem cell transplantation is currently the treatment of choice. Studies of patients as well as of mouse models carrying spontaneous (the oc/oc mouse) or targeted disruption of Atp6i (TCIRG1) gene have been instrumental providing insight into disease pathogenesis and development of novel cellular therapies that exploit gene correction.
Collapse
Affiliation(s)
- Valentina Capo
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy; Institute of Genetic and Biomedical Research, Milan Unit, National Research Council, Milan, Italy
| | - Mario Abinun
- Children's Haematopoietic Stem Cell Transplantation Unit, Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK; Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Anna Villa
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy; Institute of Genetic and Biomedical Research, Milan Unit, National Research Council, Milan, Italy.
| |
Collapse
|
2
|
Association between testosterone levels and bone mineral density in females aged 40-60 years from NHANES 2011-2016. Sci Rep 2022; 12:16426. [PMID: 36180560 PMCID: PMC9525583 DOI: 10.1038/s41598-022-21008-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/21/2022] [Indexed: 11/22/2022] Open
Abstract
Growing evidence indicates that testosterone is a conspicuous marker for assessing male bone mineral density (BMD). However, research regarding testosterone levels and BMD is sparse and controversial for females. Hence, we aimed to investigate the association between testosterone levels and BMD among adult females aged 40–60 years in the United States. In this cross-sectional study, all participants were part of the National Health and Nutrition Examination Survey (2011–2016). A weighted general linear model was used to estimate the association between testosterone levels and lumbar BMD. Age, race, income level, education level, body mass index (BMI), blood urea nitrogen (BUN) level, serum uric acid (UA) level, serum calcium (Ca) level, serum phosphorus (P) level, the use of oral contraceptive pills, the use of hormone replacement therapy (HRT), smoking status, drinking status, and the use of corticosteroids were adjusted using a weighted multiple regression model. Subgroup analyses were performed using the same regression model. We included 2198 female participants in the study, and testosterone levels were positively associated with lumbar BMD after adjusting for all the covariates (β = 1.12, 95% CI 0.31, 1.93). In subgroup analyses, the associations in the fourth quartile of testosterone levels were stronger for the participants aged 40–50 years old (quartile 4, β = 42.92, 95% CI 7.53, 78.30 vs. quartile 1) and 50 to 60-year-old (quartile 4, β = 32.41, 95% CI 0.14, 64.69 vs. quartile 1). Similar results were found in other subgroups, including subgroups for race (Non-Hispanic Black, Other), income level (income ≤ 1.3, income > 3.5), education level (college or higher), BMI > 25 kg/m2, BUN levels ≤ 20 mg/dL, UA levels ≤ 6 mg/dL, Ca levels ≤ 10.1 mg/dL, P levels ≤ 5 mg/dL, drinking status, never smoker, never taking birth control pills, and HRT user. There was no interaction among the covariates in the association between lumbar BMD and testosterone levels (P for interaction > 0.05). In US adult females aged 40–60 years, the testosterone level was a positive predictor of the lumbar BMD after adjusting for covariates.
Collapse
|
3
|
Lagerquist MK, Gupta P, Sehic E, Horkeby KL, Scheffler JM, Nordqvist J, Lawenius L, Islander U, Corciulo C, Henning P, Carlsten H, Engdahl C. Reduction of mature B cells and immunoglobulins results in increased trabecular bone. JBMR Plus 2022; 6:e10670. [PMID: 36111205 PMCID: PMC9465004 DOI: 10.1002/jbm4.10670] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/07/2022] [Accepted: 07/22/2022] [Indexed: 11/10/2022] Open
Abstract
Inflammation has a significant effect on bone remodeling and can result in bone loss via increased stimulation of osteoclasts. Activated immunoglobulins, especially autoantibodies, can increase osteoclastogenesis and are associated with pathological bone loss. Whether immunoglobulins and mature B lymphocytes are important for general bone architecture has not been completely determined. Here we demonstrate, using a transgenic mouse model, that reduction of mature B cells and immunoglobulins leads to increased trabecular bone mass compared to wild‐type (WT) littermate controls. This bone effect is associated with a decrease in the number of osteoclasts and reduced bone resorption, despite decreased expression of osteoprotegerin. We also demonstrate that the reduction of mature B cells and immunoglobulins do not prevent bone loss caused by estrogen deficiency or arthritis compared to WT littermate controls. In conclusion, the reduction of mature B cells and immunoglobulins results in disturbed regulation of trabecular bone turnover in healthy conditions but is dispensable for pathological bone loss. © 2022 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Marie K. Lagerquist
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| | - Priti Gupta
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| | - Edina Sehic
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| | | | - Julia M. Scheffler
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| | - Jauquline Nordqvist
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| | - Lina Lawenius
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| | - Ulrika Islander
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| | - Carmen Corciulo
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| | - Petra Henning
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| | - Hans Carlsten
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| | - Cecilia Engdahl
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| |
Collapse
|
4
|
Zhang Z, Huang X, Wang E, Huang Y, Yang R. Identification and characterization of B220 +/B220 - subpopulations in murine Gr1 +CD11b + cells during tumorigenesis. Oncoimmunology 2021; 10:1912472. [PMID: 33948392 PMCID: PMC8057082 DOI: 10.1080/2162402x.2021.1912472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Although all murine MDSCs are defined as Gr1+CD11b+, their true immunophenotype remains elusive. In this study, we found murine Gr1+CD11b+ cells can be divided into two subsets: Gr1+CD11b+B220- and Gr1+CD11b+B220+, especially in the spleen tissues. Unlike the dominant B220- subset, the B220+ subpopulation was not induced by tumor in vivo. Moreover, Gr1+CD11b+B220+ cells from tumor-bearing mice spleens were unable to induce arginase 1 and inducible nitric oxide synthase expression, inhibit T cell proliferation, or promote tumor growth in primary tumor site. Nevertheless, these cells suppressed tumor metastasis in vivo and reduced cancer cell motility in vitro, while Gr1+CD11b+B220- cells from tumor-bearing mice spleens promoted tumor metastasis and enhanced cancer cell motility. Furthermore, both the polymorphonuclear (PMN-MDSCs) and monocytic MDSCs (Mo-MDSCs) could be further divided into B220- and B220+ subsets; interestingly, tumor only induced the expansion of B220- PMN-MDSCs and B220- Mo-MDSCs, but not the B220+ counterparts. Compared with B220- PMN-MDSCs and B220- Mo-MDSCs, the Ly6G+Ly6C-CD11b+B220+ and Ly6G-Ly6C+CD11b+B220+ cells from tumor-bearing mice spleens exhibited a more mature phenotype without immunosuppressive activity. Additionally, IL-6 deficiency attenuated the tumor-induced accumulation of MDSCs, B220- MDSCs and B220- PMN-MDSCs but increased the percentages of Gr1+CD11b+B220+, Ly6G+Ly6C-CD11b+B220+, and Ly6G-Ly6C+CD11b+B220+ cells, indicating the opposing roles of the IL-6 signaling pathway in the expansion of B220- MDSCs and their B220+ counterparts. Taken together, our findings indicate that the B220+ subset is a distinct subset of Gr1+CD11b+ cells functionally different from the B220- subpopulation during tumorigenesis and induction of MDSCs to B220+ cells may be helpful for cancer therapy.
Collapse
Affiliation(s)
- Zhiqian Zhang
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China.,School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Xu Huang
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Enlin Wang
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Yugang Huang
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Rongcun Yang
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China.,Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, China.,Department of Immunology, School of Medicine, Nankai University, Tianjin, China
| |
Collapse
|
5
|
Kolomansky A, Kaye I, Ben-Califa N, Gorodov A, Awida Z, Sadovnic O, Ibrahim M, Liron T, Hiram-Bab S, Oster HS, Sarid N, Perry C, Gabet Y, Mittelman M, Neumann D. Anti-CD20-Mediated B Cell Depletion Is Associated With Bone Preservation in Lymphoma Patients and Bone Mass Increase in Mice. Front Immunol 2020; 11:561294. [PMID: 33193330 PMCID: PMC7604358 DOI: 10.3389/fimmu.2020.561294] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/21/2020] [Indexed: 12/14/2022] Open
Abstract
Immunotherapy with anti-CD20-specific antibodies (rituximab), has become the standard of care for B cell lymphoproliferative disorders and many autoimmune diseases. In rheumatological patients the effect of rituximab on bone mass yielded conflicting results, while in lymphoma patients it has not yet been described. Here, we used cross-sectional X-ray imaging (CT/PET-CT) to serially assess bone density in patients with follicular lymphoma receiving rituximab maintenance therapy. Remarkably, this treatment prevented the decline in bone mass observed in the control group of patients who did not receive active maintenance therapy. In accordance with these data, anti-CD20-mediated B cell depletion in normal C57BL/6J female mice led to a significant increase in bone mass, as reflected by a 7.7% increase in bone mineral density (whole femur), and a ~5% increase in cortical as well as trabecular tissue mineral density. Administration of anti-CD20 antibodies resulted in a significant decrease in osteoclastogenic signals, including RANKL, which correlated with a reduction in osteoclastogenic potential of bone marrow cells derived from B-cell-depleted animals. Taken together, our data suggest that in addition to its anti-tumor activity, anti-CD20 treatment has a favorable effect on bone mass. Our murine studies indicate that B cell depletion has a direct effect on bone remodeling.
Collapse
Affiliation(s)
- Albert Kolomansky
- Department of Cell and Developmental Biology, Tel Aviv University, Tel Aviv, Israel.,Department of Medicine A, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Irit Kaye
- Department of Medicine A, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nathalie Ben-Califa
- Department of Cell and Developmental Biology, Tel Aviv University, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Anton Gorodov
- Department of Cell and Developmental Biology, Tel Aviv University, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Open University of Israel, Ra'anana, Israel
| | - Zamzam Awida
- Department of Cell and Developmental Biology, Tel Aviv University, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ofer Sadovnic
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Radiology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Maria Ibrahim
- Department of Cell and Developmental Biology, Tel Aviv University, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tamar Liron
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Anatomy and Anthropology, Tel Aviv University, Tel Aviv, Israel
| | - Sahar Hiram-Bab
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Anatomy and Anthropology, Tel Aviv University, Tel Aviv, Israel
| | - Howard S Oster
- Department of Medicine A, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nadav Sarid
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Tel Aviv Sourasky Medical Center, Institute of Hematology, Tel Aviv, Israel
| | - Chava Perry
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Tel Aviv Sourasky Medical Center, Institute of Hematology, Tel Aviv, Israel
| | - Yankel Gabet
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Anatomy and Anthropology, Tel Aviv University, Tel Aviv, Israel
| | - Moshe Mittelman
- Department of Medicine A, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Drorit Neumann
- Department of Cell and Developmental Biology, Tel Aviv University, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
6
|
Madel MB, Ibáñez L, Rouleau M, Wakkach A, Blin-Wakkach C. A Novel Reliable and Efficient Procedure for Purification of Mature Osteoclasts Allowing Functional Assays in Mouse Cells. Front Immunol 2018; 9:2567. [PMID: 30450105 PMCID: PMC6224441 DOI: 10.3389/fimmu.2018.02567] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 10/17/2018] [Indexed: 01/07/2023] Open
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.
Collapse
Affiliation(s)
- Maria-Bernadette Madel
- CNRS UMR7370, Laboratoire de PhysioMédecine Moléculaire, Faculty of Medicine, Nice, France.,University Nice Sophia Antipolis, Nice, France
| | - Lidia Ibáñez
- CNRS UMR7370, Laboratoire de PhysioMédecine Moléculaire, Faculty of Medicine, Nice, France.,University Nice Sophia Antipolis, Nice, France
| | - Matthieu Rouleau
- CNRS UMR7370, Laboratoire de PhysioMédecine Moléculaire, Faculty of Medicine, Nice, France.,University Nice Sophia Antipolis, Nice, France
| | - Abdelilah Wakkach
- CNRS UMR7370, Laboratoire de PhysioMédecine Moléculaire, Faculty of Medicine, Nice, France.,University Nice Sophia Antipolis, Nice, France
| | - Claudine Blin-Wakkach
- CNRS UMR7370, Laboratoire de PhysioMédecine Moléculaire, Faculty of Medicine, Nice, France.,University Nice Sophia Antipolis, Nice, France
| |
Collapse
|
7
|
Almeida M, Laurent MR, Dubois V, Claessens F, O'Brien CA, Bouillon R, Vanderschueren D, Manolagas SC. Estrogens and Androgens in Skeletal Physiology and Pathophysiology. Physiol Rev 2017; 97:135-187. [PMID: 27807202 PMCID: PMC5539371 DOI: 10.1152/physrev.00033.2015] [Citation(s) in RCA: 547] [Impact Index Per Article: 68.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Estrogens and androgens influence the growth and maintenance of the mammalian skeleton and are responsible for its sexual dimorphism. Estrogen deficiency at menopause or loss of both estrogens and androgens in elderly men contribute to the development of osteoporosis, one of the most common and impactful metabolic diseases of old age. In the last 20 years, basic and clinical research advances, genetic insights from humans and rodents, and newer imaging technologies have changed considerably the landscape of our understanding of bone biology as well as the relationship between sex steroids and the physiology and pathophysiology of bone metabolism. Together with the appreciation of the side effects of estrogen-related therapies on breast cancer and cardiovascular diseases, these advances have also drastically altered the treatment of osteoporosis. In this article, we provide a comprehensive review of the molecular and cellular mechanisms of action of estrogens and androgens on bone, their influences on skeletal homeostasis during growth and adulthood, the pathogenetic mechanisms of the adverse effects of their deficiency on the female and male skeleton, as well as the role of natural and synthetic estrogenic or androgenic compounds in the pharmacotherapy of osteoporosis. We highlight latest advances on the crosstalk between hormonal and mechanical signals, the relevance of the antioxidant properties of estrogens and androgens, the difference of their cellular targets in different bone envelopes, the role of estrogen deficiency in male osteoporosis, and the contribution of estrogen or androgen deficiency to the monomorphic effects of aging on skeletal involution.
Collapse
Affiliation(s)
- Maria Almeida
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Michaël R Laurent
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Vanessa Dubois
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Frank Claessens
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Charles A O'Brien
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Roger Bouillon
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Dirk Vanderschueren
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Stavros C Manolagas
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| |
Collapse
|
8
|
Fujiwara Y, Piemontese M, Liu Y, Thostenson JD, Xiong J, O'Brien CA. RANKL (Receptor Activator of NFκB Ligand) Produced by Osteocytes Is Required for the Increase in B Cells and Bone Loss Caused by Estrogen Deficiency in Mice. J Biol Chem 2016; 291:24838-24850. [PMID: 27733688 DOI: 10.1074/jbc.m116.742452] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 09/25/2016] [Indexed: 12/20/2022] Open
Abstract
The cytokine receptor activator of NFκB ligand (RANKL) produced by osteocytes is essential for osteoclast formation in cancellous bone under physiological conditions, and RANKL production by B lymphocytes is required for the bone loss caused by estrogen deficiency. Here, we examined whether RANKL produced by osteocytes is also required for the bone loss caused by estrogen deficiency. Mice lacking RANKL in osteocytes were protected from the increase in osteoclast number and the bone loss caused by ovariectomy. Moreover, these mice did not exhibit the increase in bone marrow B lymphocytes caused by ovariectomy that occurred in control littermates. Deletion of estrogen receptor α from B cells did not alter B cell number or bone mass and did not alter the response to ovariectomy. In addition, lineage-tracing studies demonstrated that B cells do not act as osteoclast progenitors in estrogen-replete or estrogen-deficient mice. Taken together, these results demonstrate that RANKL expressed by osteocytes is required for the bone loss as well as the increase in B cell number caused by estrogen deficiency. Moreover, they suggest that estrogen control of B cell number is indirect via osteocytes and that the increase in bone marrow B cells may be a necessary component of the cascade of events that lead to cancellous bone loss during estrogen deficiency. However, the role of B cells is not to act as osteoclast progenitors but may be to act as osteoclast support cells.
Collapse
Affiliation(s)
- Yuko Fujiwara
- From the Center for Osteoporosis and Metabolic Bone Diseases and.,Central Arkansas Veterans Healthcare System, Little Rock, Arkansas 72205
| | - Marilina Piemontese
- From the Center for Osteoporosis and Metabolic Bone Diseases and.,Central Arkansas Veterans Healthcare System, Little Rock, Arkansas 72205
| | - Yu Liu
- From the Center for Osteoporosis and Metabolic Bone Diseases and.,Central Arkansas Veterans Healthcare System, Little Rock, Arkansas 72205
| | - Jeff D Thostenson
- Department of Biostatistics, University of Arkansas for Medical Sciences and
| | - Jinhu Xiong
- From the Center for Osteoporosis and Metabolic Bone Diseases and.,Central Arkansas Veterans Healthcare System, Little Rock, Arkansas 72205
| | - Charles A O'Brien
- From the Center for Osteoporosis and Metabolic Bone Diseases and .,Central Arkansas Veterans Healthcare System, Little Rock, Arkansas 72205
| |
Collapse
|
9
|
Ibáñez L, Abou-Ezzi G, Ciucci T, Amiot V, Belaïd N, Obino D, Mansour A, Rouleau M, Wakkach A, Blin-Wakkach C. Inflammatory Osteoclasts Prime TNFα-Producing CD4 + T Cells and Express CX 3 CR1. J Bone Miner Res 2016; 31:1899-1908. [PMID: 27161765 DOI: 10.1002/jbmr.2868] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 04/19/2016] [Accepted: 05/06/2016] [Indexed: 11/07/2022]
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.
Collapse
Affiliation(s)
- Lidia Ibáñez
- CNRS, LP2M, UMR7370, Faculté de Médecine, Nice, France.,Université Nice Sophia Antipolis, Nice, France
| | - Grazia Abou-Ezzi
- CNRS, LP2M, UMR7370, Faculté de Médecine, Nice, France.,Université Nice Sophia Antipolis, Nice, France
| | - Thomas Ciucci
- CNRS, LP2M, UMR7370, Faculté de Médecine, Nice, France.,Université Nice Sophia Antipolis, Nice, France
| | - Vanessa Amiot
- CNRS, LP2M, UMR7370, Faculté de Médecine, Nice, France.,Université Nice Sophia Antipolis, Nice, France
| | - Nourhène Belaïd
- CNRS, LP2M, UMR7370, Faculté de Médecine, Nice, France.,Université Nice Sophia Antipolis, Nice, France
| | - Dorian Obino
- INSERM-U932 Immunité et Cancer, Institut Curie, Paris Sciences et Lettres Research University, Paris, France
| | - Anna Mansour
- CNRS, LP2M, UMR7370, Faculté de Médecine, Nice, France.,Université Nice Sophia Antipolis, Nice, France
| | - Matthieu Rouleau
- CNRS, LP2M, UMR7370, Faculté de Médecine, Nice, France.,Université Nice Sophia Antipolis, Nice, France
| | - Abdelilah Wakkach
- CNRS, LP2M, UMR7370, Faculté de Médecine, Nice, France.,Université Nice Sophia Antipolis, Nice, France
| | - Claudine Blin-Wakkach
- CNRS, LP2M, UMR7370, Faculté de Médecine, Nice, France. .,Université Nice Sophia Antipolis, Nice, France.
| |
Collapse
|
10
|
Blin-Wakkach C, Rouleau M, Wakkach A. Roles of osteoclasts in the control of medullary hematopoietic niches. Arch Biochem Biophys 2014; 561:29-37. [PMID: 24998177 DOI: 10.1016/j.abb.2014.06.032] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Revised: 06/23/2014] [Accepted: 06/25/2014] [Indexed: 12/23/2022]
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.
Collapse
Affiliation(s)
- Claudine Blin-Wakkach
- CNRS UMR7370, LP2M, Faculty of Medicine, 28 Av de Valombrose, 06107 Nice, France; University Nice Sophia Antipolis, Faculty of Sciences, Parc Valrose, 06100 Nice, France.
| | - Matthieu Rouleau
- CNRS UMR7370, LP2M, Faculty of Medicine, 28 Av de Valombrose, 06107 Nice, France; University Nice Sophia Antipolis, Faculty of Sciences, Parc Valrose, 06100 Nice, France
| | - Abdelilah Wakkach
- CNRS UMR7370, LP2M, Faculty of Medicine, 28 Av de Valombrose, 06107 Nice, France; University Nice Sophia Antipolis, Faculty of Sciences, Parc Valrose, 06100 Nice, France
| |
Collapse
|
11
|
Panaroni C, Tzeng YS, Saeed H, Wu JY. Mesenchymal progenitors and the osteoblast lineage in bone marrow hematopoietic niches. Curr Osteoporos Rep 2014; 12:22-32. [PMID: 24477415 PMCID: PMC4077781 DOI: 10.1007/s11914-014-0190-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The bone marrow cavity is essential for the proper development of the hematopoietic system. In the last few decades, it has become clear that mesenchymal stem/progenitor cells as well as cells of the osteoblast lineage, besides maintaining bone homeostasis, are also fundamental regulators of bone marrow hematopoiesis. Several studies have demonstrated the direct involvement of mesenchymal and osteoblast lineage cells in the maintenance and regulation of supportive microenvironments necessary for quiescence, self-renewal and differentiation of hematopoietic stem cells. In addition, specific niches have also been identified within the bone marrow for maturing hematopoietic cells. Here we will review recent findings that have highlighted the roles of mesenchymal progenitors and cells of the osteoblast lineage in regulating distinct stages of hematopoiesis.
Collapse
Affiliation(s)
- Cristina Panaroni
- Division of Endocrinology, Stanford University School of Medicine, 300 Pasteur Dr., S-025, Stanford, CA, 94305, USA
| | | | | | | |
Collapse
|
12
|
Kimura M, Nagai T, Matsushita R, Hashimoto A, Miyashita T, Hirohata S. Role of FK506 binding protein 5 (FKBP5) in osteoclast differentiation. Mod Rheumatol 2014. [DOI: 10.3109/s10165-012-0809-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
13
|
Wythe SE, Nicolaidou V, Horwood NJ. Cells of the immune system orchestrate changes in bone cell function. Calcif Tissue Int 2014; 94:98-111. [PMID: 23912951 DOI: 10.1007/s00223-013-9764-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2013] [Accepted: 06/30/2013] [Indexed: 12/17/2022]
Abstract
There is a complex interplay between the cells of the immune system and bone. Immune cells, such as T and NK cells, are able to enhance osteoclast formation via the production of RANKL. Yet there is increasing evidence to show that during the resolution of inflammation or as a consequence of increased osteoclastogenesis there is an anabolic response via the formation of more osteoblasts. Furthermore, osteoblasts themselves are involved in the control of immune cell function, thus promoting the resolution of inflammation. Hence, the concept of "coupling"-how bone formation is linked to resorption-needs to be more inclusive rather than restricting our focus to osteoblast-osteoclast interactions as in a whole organism these cells are never in isolation. This review will investigate the role of immune cells in normal bone homeostasis and in inflammatory diseases where the balance between resorption and formation is lost.
Collapse
Affiliation(s)
- Sarah E Wythe
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Roosevelt Drive, Oxford, OX3 7FY, UK
| | | | | |
Collapse
|
14
|
Panaroni C, Wu JY. Interactions between B lymphocytes and the osteoblast lineage in bone marrow. Calcif Tissue Int 2013; 93:261-8. [PMID: 23839529 PMCID: PMC3762579 DOI: 10.1007/s00223-013-9753-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 05/22/2013] [Indexed: 10/26/2022]
Abstract
The regulatory effects of the immune system on the skeleton during homeostasis and activation have been appreciated for years. In the past decade it has become evident that bone tissue can also regulate immune cell development. In the bone marrow, the differentiation of hematopoietic progenitors requires specific microenvironments, called "niches," provided by various subsets of stromal cells, many of which are of mesenchymal origin. Among these stromal cell populations, cells of the osteoblast lineage serve a supportive function in the maintenance of normal hematopoiesis, and B lymphopoiesis in particular. Within the osteoblast lineage, distinct differentiation stages exert differential regulatory effects on hematopoietic development. In this review we will highlight the critical role of osteoblast progenitors in the perivascular B lymphocyte niche.
Collapse
Affiliation(s)
- Cristina Panaroni
- Division of Endocrinology, Gerontology and Metabolism, Stanford University School of Medicine, CA 94305, USA
| | | |
Collapse
|
15
|
Kimura M, Nagai T, Matsushita R, Hashimoto A, Miyashita T, Hirohata S. Role of FK506 binding protein 5 (FKBP5) in osteoclast differentiation. Mod Rheumatol 2012; 23:1133-9. [PMID: 23263277 DOI: 10.1007/s10165-012-0809-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Accepted: 11/29/2012] [Indexed: 10/27/2022]
Abstract
OBJECTIVES We previously disclosed the enhanced expression of FK506 binding protein 5 (FKBP5) messenger RNA (mRNA) in bone marrow (BM) CD34(+) cells in rheumatoid arthritis (RA), in which systemic osteoporosis takes place. Since BM CD34(+) cells are precursors of osteoclasts, it is possible that FKBP5 overexpression might lead to osteoporosis by affecting osteoclastogenesis. We therefore explore the influences of FKBP5 in osteoclast differentiation. METHODS Stable transfectants of RAW264.7 overexpressing murine FKBP5 gene were established. Osteoclast differentiation was induced by receptor activator of nuclear factor kappa B (NF-κB) ligand and was evaluated by tartrate-resistant acid phosphatase (TRAP) staining and pit formation assay. RESULTS FKBP5 transfectants of RAW264.7 generated higher numbers of TRAP-positive multinucleated cells with increased activity of pit formation on calcium phosphate-coated culture slides than mock transfectants. The enhancement of osteoclast differentiation of FKBP5 transfectants was only partially inhibited by N-acetyl L-cysteine. Finally, glucocorticoid enhanced FKBP5 mRNA expression as well as osteoclast differentiation of RAW264.7 cells in a dose-dependent manner. CONCLUSIONS These results indicate that FKBP5 promotes osteoclast differentiation by a mechanism distinct from NF-κB activation. Moreover, the data suggest that FKBP5 might play a role in bone destruction and development of osteoporosis in RA as well as in glucocorticoid-induced osteoporosis.
Collapse
Affiliation(s)
- Miho Kimura
- Department of Rheumatology and Infectious Diseases, Kitasato University School of Medicine, 1-15-1 Kitasato, Sagamihara, Kanagawa, 228-8555, Japan
| | | | | | | | | | | |
Collapse
|
16
|
Bertola A, Ciucci T, Rousseau D, Bourlier V, Duffaut C, Bonnafous S, Blin-Wakkach C, Anty R, Iannelli A, Gugenheim J, Tran A, Bouloumié A, Gual P, Wakkach A. Identification of adipose tissue dendritic cells correlated with obesity-associated insulin-resistance and inducing Th17 responses in mice and patients. Diabetes 2012; 61:2238-47. [PMID: 22596049 PMCID: PMC3425417 DOI: 10.2337/db11-1274] [Citation(s) in RCA: 291] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
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.
Collapse
Affiliation(s)
- Adeline Bertola
- Université de Nice-Sophia Antipolis, Faculté de Médecine, Nice, France
- INSERM, U1065, Equipe Complications hépatiques de l’obésité, Nice, France
| | - Thomas Ciucci
- Université de Nice-Sophia Antipolis, Faculté de Médecine, Nice, France
- LP2M-CNRS-FRE3472, Hôpital de l'Archet, Nice, France
| | - Déborah Rousseau
- Université de Nice-Sophia Antipolis, Faculté de Médecine, Nice, France
- INSERM, U1065, Equipe Complications hépatiques de l’obésité, Nice, France
| | - Virginie Bourlier
- INSERM, U1048, Institut des maladies métaboliques et cardiovasculaires, Toulouse, France
- Université Paul Sabatier Toulouse-III, Toulouse, France
| | - Carine Duffaut
- INSERM, U1048, Institut des maladies métaboliques et cardiovasculaires, Toulouse, France
- Université Paul Sabatier Toulouse-III, Toulouse, France
| | - Stéphanie Bonnafous
- Université de Nice-Sophia Antipolis, Faculté de Médecine, Nice, France
- INSERM, U1065, Equipe Complications hépatiques de l’obésité, Nice, France
- Centre Hospitalier Universitaire de Nice, Hôpital de l’Archet, Département Digestif, Nice, France
| | - Claudine Blin-Wakkach
- Université de Nice-Sophia Antipolis, Faculté de Médecine, Nice, France
- LP2M-CNRS-FRE3472, Hôpital de l'Archet, Nice, France
| | - Rodolphe Anty
- Université de Nice-Sophia Antipolis, Faculté de Médecine, Nice, France
- INSERM, U1065, Equipe Complications hépatiques de l’obésité, Nice, France
- Centre Hospitalier Universitaire de Nice, Hôpital de l’Archet, Département Digestif, Nice, France
| | - Antonio Iannelli
- Université de Nice-Sophia Antipolis, Faculté de Médecine, Nice, France
- INSERM, U1065, Equipe Complications hépatiques de l’obésité, Nice, France
- Centre Hospitalier Universitaire de Nice, Hôpital de l’Archet, Département Digestif, Nice, France
| | - Jean Gugenheim
- Université de Nice-Sophia Antipolis, Faculté de Médecine, Nice, France
- INSERM, U1065, Equipe Complications hépatiques de l’obésité, Nice, France
- Centre Hospitalier Universitaire de Nice, Hôpital de l’Archet, Département Digestif, Nice, France
| | - Albert Tran
- Université de Nice-Sophia Antipolis, Faculté de Médecine, Nice, France
- INSERM, U1065, Equipe Complications hépatiques de l’obésité, Nice, France
- Centre Hospitalier Universitaire de Nice, Hôpital de l’Archet, Département Digestif, Nice, France
| | - Anne Bouloumié
- INSERM, U1048, Institut des maladies métaboliques et cardiovasculaires, Toulouse, France
- Université Paul Sabatier Toulouse-III, Toulouse, France
| | - Philippe Gual
- Université de Nice-Sophia Antipolis, Faculté de Médecine, Nice, France
- INSERM, U1065, Equipe Complications hépatiques de l’obésité, Nice, France
- Centre Hospitalier Universitaire de Nice, Hôpital de l’Archet, Département Digestif, Nice, France
| | - Abdelilah Wakkach
- Université de Nice-Sophia Antipolis, Faculté de Médecine, Nice, France
- LP2M-CNRS-FRE3472, Hôpital de l'Archet, Nice, France
- Corresponding author: Abdelilah Wakkach, , or Philippe Gual,
| |
Collapse
|
17
|
Onal M, Xiong J, Chen X, Thostenson JD, Almeida M, Manolagas SC, O'Brien CA. Receptor activator of nuclear factor κB ligand (RANKL) protein expression by B lymphocytes contributes to ovariectomy-induced bone loss. J Biol Chem 2012; 287:29851-60. [PMID: 22782898 DOI: 10.1074/jbc.m112.377945] [Citation(s) in RCA: 182] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Production of the cytokine receptor activator of NFκB ligand (RANKL) by lymphocytes has been proposed as a mechanism by which sex steroid deficiency causes bone loss. However, there have been no studies that functionally link RANKL expression in lymphocytes with bone loss in this condition. Herein, we examined whether RANKL expression in either B or T lymphocytes contributes to ovariectomy-induced bone loss in mice. Mice harboring a conditional RANKL allele were crossed with CD19-Cre or Lck-Cre mice to delete RANKL in B or T lymphocytes, respectively. Deletion of RANKL from either cell type had no impact on bone mass in estrogen-replete mice up to 7 months of age. However, mice lacking RANKL in B lymphocytes were partially protected from the bone loss caused by ovariectomy. This protection occurred in cancellous, but not cortical, bone and was associated with a failure to increase osteoclast numbers in the conditional knock-out mice. Deletion of RANKL from T lymphocytes had no impact on ovariectomy-induced bone loss. These results demonstrate that lymphocyte RANKL is not involved in basal bone remodeling, but B cell RANKL does contribute to the increase in osteoclasts and cancellous bone loss that occurs after loss of estrogen.
Collapse
Affiliation(s)
- Melda Onal
- Central Arkansas Veterans Healthcare System, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Mansour A, Abou-Ezzi G, Sitnicka E, Jacobsen SEW, Wakkach A, Blin-Wakkach C. Osteoclasts promote the formation of hematopoietic stem cell niches in the bone marrow. ACTA ACUST UNITED AC 2012; 209:537-49. [PMID: 22351931 PMCID: PMC3302238 DOI: 10.1084/jem.20110994] [Citation(s) in RCA: 151] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Osteoclasts promote the formation of the HSC niche by inducing the differentiation of osteoblastic cells from mesenchymal stem cells. 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.
Collapse
Affiliation(s)
- Anna Mansour
- Université de Nice Sophia Antipolis, 06000 Nice, France
| | | | | | | | | | | |
Collapse
|
19
|
B-1 lymphocytes differentiate into functional osteoclast-like cells. Immunobiology 2011; 217:336-44. [PMID: 21855167 DOI: 10.1016/j.imbio.2011.07.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2011] [Accepted: 07/10/2011] [Indexed: 11/22/2022]
Abstract
The existence of murine peritoneal osteoclast precursors has been already described. Also, recent reports evidenced an interplay between B lymphocytes and osteoclasts development. B-1 cells comprise a B-lymphocyte subset that resides mostly in pleural and peritoneal cavities. It has been demonstrated that B-1 cells can differentiate into mononuclear phagocytes and form multinucleated giant cells. Based on these findings, we investigated the role of B-1 lymphocytes in bone resorption and osteoclastogenesis. In vivo experimental periodontitis induced in B-1 deficient Xid mice demonstrated that bone resorption is impaired in these animals. However, reconstitution of Xid mice with B-1 cells increased bone resorption to near Balb/c values. B-1 cell derived phagocytes express the receptor activator of nuclear factor-κB (RANK) and the macrophage colony-stimulating factor receptor (M-CSFR). When cultured with RANK-ligand (RANKL) and M-CSF, B-1 cells became tartrate resistant acid phosphatase (TRAP) positive multinucleated cells, a typical osteoclast phenotype. Lacunae formation was observed when cells were cultivated onto a calcium phosphate analog, indicating functional differentiation of B1 cells into osteoclast-like cells. The dynamics of their IgM expression showed that this lymphoid marker was downregulated along the differentiation of B-1 lymphocytes into osteoclasts. Our results unveiled the first evidence that B-1 cells have a role in osteoclastogenesis and bone resorption and offer new insights in the relationship between bone and lymphoid cells.
Collapse
|
20
|
Mansour A, Anginot A, Mancini SJC, Schiff C, Carle GF, Wakkach A, Blin-Wakkach C. Osteoclast activity modulates B-cell development in the bone marrow. Cell Res 2011; 21:1102-15. [PMID: 21321604 DOI: 10.1038/cr.2011.21] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
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.
Collapse
Affiliation(s)
- Anna Mansour
- CNRS, GEPITOS, UMR 6235, Faculté de Médecine, 06100 Nice, France
| | | | | | | | | | | | | |
Collapse
|
21
|
Horowitz MC, Fretz JA, Lorenzo JA. How B cells influence bone biology in health and disease. Bone 2010; 47:472-9. [PMID: 20601290 PMCID: PMC2941392 DOI: 10.1016/j.bone.2010.06.011] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Revised: 06/13/2010] [Accepted: 06/14/2010] [Indexed: 12/27/2022]
Abstract
It is now well established that important regulatory interactions occur between the cells in the hematopoietic, immune and skeletal systems (osteoimmunology). B lymphocytes (B cells) are responsible for the generation and production of antibodies or immunoglobulins in the body. Together with T cells these lymphocytes comprise the adaptive immune system, which allows an individual to develop specific responses to an infection and retain memory of that infection, allowing for a faster and more robust response if that same infection occurs again. In addition to this immune function, B cells have a close and multifaceted relationship with bone cells. B cells differentiate from hematopoietic stem cells (HSCs) in supportive niches found on endosteal bone surfaces. Cells in the osteoblast lineage support HSC and B cell differentiation in these niches. B cell differentiation is regulated, at least in part, by a series of transcription factors that function in a temporal manner. While these transcription factors are required for B cell differentiation, their loss causes profound changes in the bone phenotype. This is due, in part, to the close relationship between macrophage/osteoclast and B cell differentiation. Cross talk between B cells and bone cells is reciprocal with defects in the RANKL-RANK, OPG signaling axis resulting in altered bone phenotypes. While the role of B cells during normal bone remodeling appears minimal, activated B cells play an important role in many inflammatory diseases with associated bony changes. This review examines the relationship between B cells and bone cells and how that relationship affects the skeleton and hematopoiesis during health and disease.
Collapse
Affiliation(s)
- Mark C Horowitz
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT 06510, USA.
| | | | | |
Collapse
|
22
|
Breuil V, Ticchioni M, Testa J, Roux CH, Ferrari P, Breittmayer JP, Albert-Sabonnadière C, Durant J, De Perreti F, Bernard A, Euller-Ziegler L, Carle GF. Immune changes in post-menopausal osteoporosis: the Immunos study. Osteoporos Int 2010; 21:805-14. [PMID: 19876583 DOI: 10.1007/s00198-009-1018-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2009] [Accepted: 06/24/2009] [Indexed: 10/20/2022]
Abstract
UNLABELLED The phenotypic and functional characteristics of immune cells of osteoporotic women compared to healthy controls similar for age and estrogen level showed for the first time significant changes in several B lymphocytes populations in postmenopausal osteoporosis, related to bone mineral density (BMD) and fractures, and a significant lower basal secretion of interferon-gamma (IFN-gamma) by CD4(+). INTRODUCTION To investigate the interactions between bone and immune system, we studied the phenotypic and functional characteristics of immune cells of 26 postmenopausal women with osteoporotic (OP) fractures compared to 24 healthy controls. METHODS We analyzed surface markers of peripheral B, CD4(+) and CD8(+) lymphocytes and cytokine secretion in supernatants of these cells cultured with or without stimulation. Body composition was assessed by dual energy X-ray absorptiometry. RESULTS The two groups were similar for age and estrogen level. OP women had a significantly lower body mass index, fat mass, and lean mass. The number of CD19(+), CD19(+)/CD27(+), CD19(+)/CD27(+)/CD5(-)/CD38(+) and CD19(+)/CD27(+)/RANK(+), CD4(+)/CD27(+)/CD45RA(-)/RANK(+), and CD4(+)/CD27(+)/CD45RA(-)/CD28(+) was lower in OP women and positively correlated to BMD. In OP women, under basal conditions, CD4(+) secreted less IFN-gamma and B lymphocytes more granulocyte macrophage colony-stimulating factor (GM-CSF). GM-CSF was positively correlated to fracture rate and negatively to BMD. CONCLUSIONS Our results suggest that, regardless of age and estrogen status, postmenopausal OP is associated with immune changes, highlighting a possible role of IFN-gamma in the pathophysiology of OP and reporting, for the first time, changes in several B lymphocyte populations. These alterations may reflect the frailty observed after fracture, providing new insight into the mechanisms of morbidity and mortality associated with OP fractures.
Collapse
Affiliation(s)
- V Breuil
- Service de Rhumatologie, CHU de Nice, Hôpital l'Archet, Nice, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Liu YCG, Teng YTA. Dendritic Cell-Associated Osteoclastogenesis and Bone Loss. Clin Rev Bone Miner Metab 2009. [DOI: 10.1007/s12018-009-9059-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
24
|
Lorenzo J, Horowitz M, Choi Y. Osteoimmunology: interactions of the bone and immune system. Endocr Rev 2008; 29:403-40. [PMID: 18451259 PMCID: PMC2528852 DOI: 10.1210/er.2007-0038] [Citation(s) in RCA: 392] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2007] [Accepted: 04/01/2008] [Indexed: 12/20/2022]
Abstract
Bone and the immune system are both complex tissues that respectively regulate the skeleton and the body's response to invading pathogens. It has now become clear that these organ systems often interact in their function. This is particularly true for the development of immune cells in the bone marrow and for the function of bone cells in health and disease. Because these two disciplines developed independently, investigators in each don't always fully appreciate the significance that the other system has on the function of the tissue they are studying. This review is meant to provide a broad overview of the many ways that bone and immune cells interact so that a better understanding of the role that each plays in the development and function of the other can develop. It is hoped that an appreciation of the interactions of these two organ systems will lead to better therapeutics for diseases that affect either or both.
Collapse
Affiliation(s)
- Joseph Lorenzo
- Department of Medicine, The University of Connecticut Health Center, N4054, MC5456, 263 Farmington Avenue, Farmington, Connecticut 06030-5456, USA.
| | | | | |
Collapse
|
25
|
Li Y, Li A, Yang X, Weitzmann MN. Ovariectomy-induced bone loss occurs independently of B cells. J Cell Biochem 2007; 100:1370-5. [PMID: 17131364 DOI: 10.1002/jcb.21121] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Estrogen withdrawal is associated with a significant expansion in B cell precursor and mature B cell populations. However, despite significant circumstantial evidence the role of B lineage cells in ovariectomy-induced bone loss in vivo is unclear. In vitro studies have demonstrated that mature B cells have the potential to both positively and negatively impact osteoclastogenesis by virtue of their capacity to secrete pro-osteoclastogenic cytokines including receptor activator of NFkappaB ligand (RANKL), as well as anti-osteoclastogenic cytokines such as osteoprotegerin (OPG) and transforming growth factor beta (TGFbeta). Although several studies have suggested that expansion of the B lineage following ovariectomy may play a key role in the etiology of ovariectomy-induced bone loss, in vivo studies to directly test this notion have yet to be conducted. In this study, we performed ovariectomy on microMT(-/-) mice which are specifically deficient in mature B cells. Analysis of bone mineral density (BMD) by dual-energy X-ray absorptiometry (DXA) and micro-computed tomography (CT) demonstrate that mature B cell-deficient mice undergo an identical loss of bone mass relative to wild-type (WT) control mice. Our data demonstrate that mature B cells are not central mediators of ovariectomy-induced bone loss in vivo.
Collapse
Affiliation(s)
- Yan Li
- Division of Endocrinology and Metabolism, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | | | | |
Collapse
|
26
|
Andersen TL, Boissy P, Sondergaard TE, Kupisiewicz K, Plesner T, Rasmussen T, Haaber J, Kølvraa S, Delaissé JM. Osteoclast nuclei of myeloma patients show chromosome translocations specific for the myeloma cell clone: a new type of cancer-host partnership? J Pathol 2007; 211:10-7. [PMID: 17083146 DOI: 10.1002/path.2078] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A major clinical manifestation of bone cancers is bone destruction. It is widely accepted that this destruction is not caused by the malignant cells themselves, but by osteoclasts, multinucleated cells of monocytic origin that are considered to be the only cells able to degrade bone. The present study demonstrates that bone-resorbing osteoclasts from myeloma patients contain nuclei with translocated chromosomes of myeloma B-cell clone origin, in addition to nuclei without these translocations, by using combined FISH and immunohistochemistry on bone sections. These nuclei of malignant origin are transcriptionally active and appear fully integrated amongst the other nuclei. The contribution of malignant nuclei to the osteoclast population analysed in this study was greater than 30%. Osteoclast-myeloma clone hybrids contained more nuclei than normal osteoclasts and their occurrence correlated with the proximity of myeloma cells. Similar hybrid cells were generated in myeloma cell-osteoclast co-cultures, as revealed by tracing myeloma nuclei using translocations, bromo-deoxyuridine, or the Y chromosome of male myeloma cells in female osteoclasts. These observations indicate that hybrid cells can originate through fusion between myeloma cells and osteoclasts. In conclusion, malignant cells contribute significantly to the formation of bone-resorbing osteoclasts in multiple myeloma. Osteoclast-myeloma clone hybrids reflect a previously unrecognized mechanism of bone destruction in which malignant cells participate directly. The possibility that malignant cells corrupt host cells by the transfer of malignant DNA may have been underestimated to date in cancer research.
Collapse
Affiliation(s)
- T L Andersen
- Department of Clinical Cell Biology (KCB), Southern Denmark University, CeSFUVA, Vejle Hospital, 7100 Vejle, Denmark.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Aiello FB, Keller JR, Klarmann KD, Dranoff G, Mazzucchelli R, Durum SK. IL-7 induces myelopoiesis and erythropoiesis. THE JOURNAL OF IMMUNOLOGY 2007; 178:1553-63. [PMID: 17237404 DOI: 10.4049/jimmunol.178.3.1553] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IL-7 administration to mice was previously reported to increase the mobilization of progenitor cells from marrow to peripheral sites. We now report that IL-7 increases the number of mature myeloid and monocytic cells in spleen and peripheral blood. This effect required T cells, and we show that IL-7 treatment in vivo induced GM-CSF and IL-3 production by T cells with memory phenotype. However, additional myelopoietic cytokines were shown to be involved because mice deficient in both GM-CSF and IL-3 also responded to IL-7 with increased myelopoiesis. Candidate cytokines included IFN-gamma and Flt3 ligand, which were also produced in response to IL-7. Because IFN-gamma-deficient mice also increased myelopoiesis, it was suggested that IL-7 induced production of redundant myelopoietic cytokines. In support of this hypothesis, we found that the supernatant from IL-7-treated, purified T cells contained myelopoietic activity that required a combination of Abs against GM-CSF, IL-3, and anti-Flt3 ligand to achieve maximum neutralization. IL-7 administration increased the number of splenic erythroid cells in either normal, Rag1 or GM-CSF-IL-3-deficient mice, suggesting that IL-7 might directly act on erythroid progenitors. In support of this theory, we detected a percentage of TER-119(+) erythroid cells that expressed the IL-7Ralpha-chain and common gamma-chain. Bone marrow cells expressing IL-7R and B220 generated erythroid colonies in vitro in response to IL-7, erythropoietin, and stem cell factor. This study demonstrates that IL-7 can promote nonlymphoid hemopoiesis and production of cytokines active in the host defense system in vivo, supporting its possible clinical utility.
Collapse
Affiliation(s)
- Francesca B Aiello
- Laboratory of Molecular Immunoregulation, National Cancer Institute, Frederick, MD 21702, USA
| | | | | | | | | | | |
Collapse
|
28
|
Sato T, Watanabe K, Masuhara M, Hada N, Hakeda Y. Production of IL-7 is increased in ovariectomized mice, but not RANKL mRNA expression by osteoblasts/stromal cells in bone, and IL-7 enhances generation of osteoclast precursors in vitro. J Bone Miner Metab 2007; 25:19-27. [PMID: 17187190 DOI: 10.1007/s00774-006-0723-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2006] [Accepted: 06/30/2006] [Indexed: 12/15/2022]
Abstract
Osteoclastogenic cytokines produced by T and B lineage cells and interleukin (IL)-7-induced expansion of the pool size of osteoclast precursors have been suggested to play an important role in acceleration of osteoclastogenesis induced by estrogen deficiency. However, the contribution of increased RANKL produced by osteoblasts/stromal cells to increase osteoclastogenesis in a mouse model of estrogen-deficient osteoporosis and in vitro effects of IL-7 on osteoclast precursor generation remain controversial. Thus, we investigated the effect of ovariectomy (OVX) of mice on production of RANKL, osteoprotegerin (OPG), and IL-7 in bone and the effect of IL-7 on osteoclast precursor generation in vitro. OVX did not significantly stimulate mRNA expressions of RANKL and OPG in whole femurs. Because the epiphysis, but not the femoral shaft (diaphysis) or bone marrow, is the main site of osteoclastogenesis, it is important to specifically analyze mRNA expression by osteoblasts/stromal cells at these parts of the femur. Therefore, we isolated RNA from bone marrow cell-free epiphysis, diaphysis, and flushed-out bone marrow and examined mRNA expression. The results showed no significant changes of RANKL and OPG mRNA expression in any part of the femur. In addition, OVX did not significantly affect RANKL and OPG mRNA expression by the adherent stromal cells isolated from flushed-out bone marrow cells but did stimulate RANKL mRNA expression by B220(+) cells in the nonadherent cell fraction. On the other hand, OVX increased IL-7 mRNA expression in the femur as well as IL-7 concentrations in bone fluid. In cultures of unfractionated bone cells isolated by vigorous agitation of minced whole long bones to release the cells tightly attached to the bone surfaces, but not in cocultures of clonal osteoblasts/stromal cells and flushed-out bone marrow cells, IL-7 stimulated generations of osteoclasts as well as osteoclast precursors. These data suggest that increased RANKL production by osteoblasts/stromal cells is unlikely to play a central role in acceleration of osteoclastogenesis in estrogen deficiency of mice and that IL-7 stimulates osteoclast precursor generation, presumably through an action of IL-7 on the cells attached to bone rather than on cells contained in the bone marrow cell population.
Collapse
Affiliation(s)
- Takuya Sato
- Division of Oral Anatomy, Department of Human Development and Fostering, Meikai University School of Dentistry, 1-1 Keyaki-dai, Sakado, 350-0283, Japan.
| | | | | | | | | |
Collapse
|
29
|
|
30
|
Boyce BF, Schwarz EM, Xing L. Osteoclast precursors: cytokine-stimulated immunomodulators of inflammatory bone disease. Curr Opin Rheumatol 2006; 18:427-32. [PMID: 16763465 DOI: 10.1097/01.bor.0000231913.32364.32] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE OF REVIEW Proinflammatory cytokines produced in inflamed joints promote the generation and release from the bone marrow of osteoclast precursors (OCPs). These OCPs are recruited to inflamed joints where, in response to proinflammatory cytokines, they differentiate into osteoclasts and behave as immunomodulatory cells. We review the regulation of OCP formation and differentiation, and propose that they are not only sources of bone-resorbing osteoclasts, but also of immune cells that influence the progression of inflammatory bone diseases. RECENT FINDINGS OCP numbers are increased in the blood of humans and animals with tumor necrosis factor (TNF)-mediated arthritis and they correlate with serum TNF levels, which can be lowered by anti-TNF therapy. TNF stimulates bone marrow OCP generation in part by increasing their expression of c-Fms. These precursors then leave the bone marrow and circulate in the blood stream. Receptor activator NF-kappaB ligand (RANKL) and TNF, which are elevated in inflamed joints, mediate OCP differentiation into osteoclasts and other cell types, including dendritic cells. They also stimulate OCPs to produce proinflammatory cytokines and chemokines. RANKL inhibition prevents joint inflammation in TNF-mediated arthritis. SUMMARY OCPs are effector immunomodulatory cells in inflammatory bone diseases. They respond to and secrete cytokines, and are therefore targets for anti-inflammatory therapy.
Collapse
Affiliation(s)
- Brendan F Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA.
| | | | | |
Collapse
|
31
|
Atkins GJ, Kostakis P, Vincent C, Farrugia AN, Houchins JP, Findlay DM, Evdokiou A, Zannettino ACW. RANK Expression as a cell surface marker of human osteoclast precursors in peripheral blood, bone marrow, and giant cell tumors of bone. J Bone Miner Res 2006; 21:1339-49. [PMID: 16939392 DOI: 10.1359/jbmr.060604] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
UNLABELLED RANK expression in vivo on hematopoietic subsets including pre-osteoclasts, identified by monoclonal antibodies, has not been described. We describe the lineages that express RANK in bone marrow, peripheral blood, and GCTs. We show that CD14(+)RANK(high) cells constitute a circulating pre-osteoclast pool. INTRODUCTION The expression of RANK by subsets of hematopoietic cells has not been adequately studied in humans. While attributed to the monocytoid lineage, the phenotype of the pre-osteoclast (pre-OC) with respect to RANK expression in vivo remains unclear. We tested monoclonal antibodies (MAbs) raised against the extracellular domain of recombinant human RANK for reactivity with normal peripheral blood (PB) and bone marrow (BM) mononuclear cells (PBMNCs and BMMNCs, respectively). We also tested reactivity with giant cell tumor cells (GCT), a confirmed source of pre-OC and mature OCs. MATERIALS AND METHODS Human PBMNCs, BMMNCs, and GCT cells were analyzed for reactivity with anti-RANK MAbs by flow cytometry in combination with hematopoietic lineage restricted markers. GCTs were also analyzed by immunofluorescence. CD14+ monocytoid cells were sorted by fluorescence-activated cell sorting (FACS) based on their relative RANK expression and cultured under OC-forming conditions. RESULTS RANK+ cells were detected similarly by three independent anti-RANK MAbs. One MAb (80736) immunoprecipitated RANK-RANKL complexes from surface-biotinylated GCT lysates. Using dual-color flow cytometry, RANK was detected on CD14+ (monocytoid), CD19+ (B-lymphoid), CD56+ (NK cell), and glycophorin A+ erythroid progenitors. Minor populations of both CD3+ T lymphocytes and BM CD34+ hematopoietic progenitors also expressed cell surface RANK. In GCTs, RANK expression was identified on mononuclear CD45(+)CD14(+)alphaVbeta3(+)c-Fms+ cells, likely to be committed pre-OC, and on multinucleated CD45(+)alphaVbeta3(+)TRACP(+) OCs. Importantly, sorted CD14(+)RANK(high) PBMNCs treated with recombinant RANKL and macrophage-colony stimulating factor (M-CSF) gave rise to approximately twice the number of osteoclasts than RANK(mid) or RANK(low) cells. CONCLUSIONS These results suggest that committed monocytoid RANK+ pre-OCs are represented in the marrow and circulate in the periphery, forming a pool of cells capable of responding rapidly to RANKL. The ability to reliably detect committed pre-OC in peripheral blood could have important clinical applications in the management of diseases characterized by abnormal osteoclastic activity.
Collapse
Affiliation(s)
- Gerald J Atkins
- Department of Orthopaedics and Trauma, University of Adelaide, Adelaide, South Australia, Australia
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Blin-Wakkach C, Wakkach A, Quincey D, Carle GF. Interleukin-7 partially rescues B-lymphopoiesis in osteopetrotic oc/oc mice through the engagement of B220+ CD11b+ progenitors. Exp Hematol 2006; 34:851-9. [PMID: 16797412 DOI: 10.1016/j.exphem.2006.04.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2005] [Revised: 03/31/2006] [Accepted: 04/03/2006] [Indexed: 12/14/2022]
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.
Collapse
|
33
|
Blin-Wakkach C, Breuil V, Quincey D, Bagnis C, Carle GF. Establishment and characterization of new osteoclast progenitor cell lines derived from osteopetrotic and wild type mice. Bone 2006; 39:53-60. [PMID: 16503212 DOI: 10.1016/j.bone.2005.12.078] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2005] [Accepted: 12/16/2005] [Indexed: 11/30/2022]
Abstract
Malignant infantile osteopetrosis is a rare and lethal disease characterized by the absence of bone resorption due to inactive osteoclasts (OCLs). Among the murine models of osteopetrosis, the Tcirg1oc/oc mouse is the most resembling to the human pathology. In the majority of patients as in Tcirg1oc/oc mouse, the gene involved is the Tcirg1 gene, encoding the a3 subunit of the vacuolar proton pump. However, to date, no osteoclastic cell lines from osteopetrotic mice are available to facilitate the study of either OCL differentiation in osteopetrosis or the factors involved in the control of Tcirg1 gene expression. Heterozygotes Tcirg1+/oc mice were crossed with p53+/- mice to obtain homozygotes p53-/-Tcirg1oc/oc and p53-/-Tcirg1+/+ animals. The p53-/-Tcirg1oc/oc mice display the same bone and hematological phenotype as the original Tcirg1oc/oc mice. From the bone marrow of these mice, we have derived cell lines named POC-MGoc/oc and POC-MG+/+. These cell lines express standard osteoclastogenic markers and differentiate into OCLs in the presence of RANK-L and M-CSF. Furthermore, both cell lines can be transduced by a lentiviral vector with a high efficiency and without alteration of their OCL differentiation potential. Therefore, these cell lines provide valuable new tools to study the differentiation and function of osteoclasts in normal and resorption defective conditions.
Collapse
Affiliation(s)
- Claudine Blin-Wakkach
- CNRS/UNSA Unité K2943, IFR50, Faculté de Médecine, and Service de Rhumatologie, Hôpital L'Archet 1, Nice, France
| | | | | | | | | |
Collapse
|
34
|
Jacquin C, Gran DE, Lee SK, Lorenzo JA, Aguila HL. Identification of multiple osteoclast precursor populations in murine bone marrow. J Bone Miner Res 2006; 21:67-77. [PMID: 16355275 DOI: 10.1359/jbmr.051007] [Citation(s) in RCA: 156] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2005] [Revised: 08/26/2005] [Accepted: 10/11/2005] [Indexed: 11/18/2022]
Abstract
UNLABELLED Murine BM was fractionated using a series of hematopoietic markers to characterize its osteoclast progenitor populations. We found that the early osteoclastogenic activity in total BM was recapitulated by a population of cells contained within the CD11b(-/low) CD45R- CD3- CD115high fraction. INTRODUCTION Osteoclasts are of hematopoietic origin and they have been shown to share the same lineage as macrophages. We further characterized the phenotype of osteoclast progenitor populations in murine bone marrow (BM) by analyzing their cell surface markers. MATERIALS AND METHODS We used fluorescence-activated cell sorting (FACS) to identify the subsets of BM cells that contained osteoclast progenitors. We fractionated BM according to several markers and cultured the sorted populations for a period of 2-6 days with macrophage-colony stimulating factor (M-CSF) and RANKL. The numbers of multinucleated osteoclast-like cells (OCLs) that formed in the cultures were counted. RESULTS We found that the CD45R- CD11b(-/low) population recapitulated the early osteoclastogenic activity of total BM. In addition, although previous experiments indicated that osteoclastogenic activity was enriched within the CD45R+ population, we found that highly purified CD45R+ BM was incapable of differentiating into osteoclasts in vitro. We also found that CD45R- CD11b(high) BM cells were an inefficient source of osteoclast progenitors. However, CD11b was transiently upregulated by cells of the CD45R- CD11b(-/low) fraction early (within 24 h) during culture with M-CSF. Finally, further fractionation of BM using CD115 and CD117 showed that, as osteoclast precursor cells matured, they downregulate CD117 but remain CD115+. Curiously, pure populations of CD117- (CD115high) cells isolated fresh from BM have low osteoclastogenic activity in vitro. CONCLUSIONS We provided a refined analysis of the precise subpopulations of murine BM that are capable of differentiating into OCLs in vitro when treated with M-CSF and RANKL.
Collapse
Affiliation(s)
- Claire Jacquin
- Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut 06030, USA
| | | | | | | | | |
Collapse
|
35
|
Abstract
Osteoporosis is increasingly recognized as a complication of chronic childhood illnesses, particularly when glucocorticoids (GCs) are necessary for treatment. Elucidation of the mechanisms leading to bone fragility in these settings requires disentanglement of the relative contributions of myriad risk factors, including disease activity, muscle weakness, immobilization, delayed growth and puberty, compromised nutrition, and osteotoxic medications. Over the years, bone mass and density evaluations by dual energy X-ray absorptiometry (DXA) have become popular for assessing bone health in children; however, such measurements are difficult to interpret because of the confounding effect of bone size and the lack of DXA-based densitometric criteria for defining osteoporosis in childhood. Recently, a new diagnostic approach for evaluation of densitometric data in children has been suggested, driven by Frost's mechanostat theory. A diagnostic algorithm based on the mechanostat theory of bone-muscle development is proposed for the characterization of bone disease in children with chronic illness. In addition to DXA-based assessments, techniques such as peripheral quantitative computerized tomography and ilial histomorphometry, for which there are pediatric reference data, are gaining ground in the characterization of skeletal changes due to chronic illness. Although these diagnostic techniques expand our understanding of osteoporosis in children, they do not replace clinical assessment. Concrete clinical evidence for GC-induced bone fragility can be seen in spinal changes due to vertebral compression, with spinal morphometry emerging as an essential, but frequently overlooked, tool in the evaluation of children's bone health. Presently, osteoporosis treatment in the chronic illness setting remains experimental and should be restricted to clinical studies. Following an understanding of the natural history of GC-induced osteoporosis in children, randomized, placebo-controlled prevention and intervention trials will be the next step toward the development of clinical practice guidelines.
Collapse
Affiliation(s)
- Leanne M Ward
- Department of Pediatrics, University of Ottawa and Division of Endocrinology, Children's Hospital of Eastern Ontario, Ottawa, Canada.
| |
Collapse
|