RESUMO
Bone functions not only as a critical element of the musculoskeletal system but also serves as the primary lymphoid organ harboring hematopoietic stem cells (HSCs) and immune progenitor cells. The interdisciplinary field of osteoimmunology has illuminated the dynamic interactions between the skeletal and immune systems, vital for the maintenance of skeletal tissue homeostasis and the pathogenesis of immune and skeletal diseases. Aberrant immune activation stimulates bone cells such as osteoclasts and osteoblasts, disturbing the bone remodeling and leading to skeletal disorders as seen in autoimmune diseases like rheumatoid arthritis. On the other hand, intricate multicellular network within the bone marrow creates a specialized microenvironment essential for the maintenance and differentiation of HSCs and the progeny. Dysregulation of immune-bone crosstalk in the bone marrow environment can trigger tumorigenesis and exacerbated inflammation. A comprehensive deciphering of the complex "immune-bone crosstalk" leads to a deeper understanding of the pathogenesis of immune diseases as well as skeletal diseases, and might provide insight into potential therapeutic approaches.
Assuntos
Osso e Ossos , Sistema Imunitário , Humanos , Osso e Ossos/imunologia , Animais , Sistema Imunitário/imunologia , Osteoclastos/imunologia , Osteoclastos/metabolismo , Células-Tronco Hematopoéticas/imunologia , Doenças Ósseas/imunologia , Doenças Ósseas/patologia , Osteoblastos/imunologia , Osteoblastos/metabolismo , Remodelação Óssea/imunologia , Remodelação Óssea/fisiologiaRESUMO
PURPOSE OF REVIEW: The purpose of this review is to summarize what is known in the literature about the role inflammation plays during bone fracture healing. Bone fracture healing progresses through four distinct yet overlapping phases: formation of the hematoma, development of the cartilaginous callus, development of the bony callus, and finally remodeling of the fracture callus. Throughout this process, inflammation plays a critical role in robust bone fracture healing. RECENT FINDINGS: At the onset of injury, vessel and matrix disruption lead to the generation of an inflammatory response: inflammatory cells are recruited to the injury site where they differentiate, activate, and/or polarize to secrete cytokines for the purposes of cell signaling and cell recruitment. This process is altered by age and by sex. Bone fracture healing is heavily influenced by the presence of inflammatory cells and cytokines within the healing tissue.
Assuntos
Calo Ósseo , Citocinas , Consolidação da Fratura , Inflamação , Consolidação da Fratura/imunologia , Consolidação da Fratura/fisiologia , Humanos , Calo Ósseo/imunologia , Citocinas/imunologia , Citocinas/metabolismo , Inflamação/imunologia , Remodelação Óssea/imunologia , Animais , Hematoma/imunologia , Fraturas Ósseas/imunologiaRESUMO
Bone remodeling and bone regeneration are essential for preserving skeletal integrity and maintaining mineral homeostasis. T cells, as key members of adaptive immunity, play a pivotal role in bone remodeling and bone regeneration by producing a range of cytokines and growth factors. In the physiological state, T cells are involved in the maintenance of bone homeostasis through interactions with mesenchymal stem cells, osteoblasts, and osteoclasts. In pathological states, T cells participate in the pathological process of different types of osteoporosis through interaction with estrogen, glucocorticoids, and parathyroid hormone. During fracture healing for post-injury repair, T cells play different roles during the inflammatory hematoma phase, the bone callus formation phase and the bone remodeling phase. Targeting T cells thus emerges as a potential strategy for regulating bone homeostasis. This article reviews the research progress on related mechanisms of T cells immunity involved in bone remodeling and bone regeneration, with a view to providing a scientific basis for targeting T cells to regulate bone remodeling and bone regeneration.
Assuntos
Regeneração Óssea , Remodelação Óssea , Linfócitos T , Remodelação Óssea/imunologia , Remodelação Óssea/fisiologia , Humanos , Regeneração Óssea/imunologia , Linfócitos T/imunologia , AnimaisRESUMO
PURPOSE OF REVIEW: The purpose of this review is to recognize clinical features of Paget's disease of bone and to describe how the osteoclast, a myeloid-derived cell responsible for bone resorption, contributes to the disease. RECENT FINDINGS: Recent studies have identified several variants in SQSTM1, OPTN, and other genes that may predispose individuals to Paget's disease of bone; studies of these genes and their protein products have elucidated new roles for these proteins in bone physiology. Understanding the pathologic mechanisms in the Pagetic osteoclast may lead to the identification of future treatment targets for other inflammatory and autoimmune diseases characterized by abnormal bone erosion and/or osteoclast activation.
Assuntos
Remodelação Óssea , Osteíte Deformante , Osteoclastos , Algoritmos , Remodelação Óssea/efeitos dos fármacos , Remodelação Óssea/genética , Remodelação Óssea/imunologia , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/imunologia , Osso e Ossos/patologia , Humanos , Osteíte Deformante/diagnóstico , Osteíte Deformante/etiologia , Osteíte Deformante/fisiopatologia , Osteíte Deformante/terapia , Osteoclastos/efeitos dos fármacos , Osteoclastos/imunologia , Osteoclastos/patologiaRESUMO
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells with immunosuppressive functions; these cells play a key role in infection, immunization, chronic inflammation, and cancer. Recent studies have reported that immunosuppression plays an important role in the healing process of tissues and that Treg play an important role in fracture healing. MDSCs suppress active T cell proliferation and reduce the severity of arthritis in mice and humans. Together, these findings suggest that MDSCs play a role in bone biotransformation. In the present study, we examined the role of MDSCs in the bone healing process by creating a bone injury at the tibial epiphysis in mice. MDSCs were identified by CD11b and GR1 immunohistochemistry and their role in new bone formation was observed by detection of Runx2 and osteocalcin expression. Significant numbers of MDSCs were observed in transitional areas from the reactionary to repair stages. Interestingly, MDSCs exhibited Runx2 and osteocalcin expression in the transitional area but not in the reactionary area. And at the same area, cllagene-1 and ALP expression level increased in osteoblast progenitor cells. These data is suggesting that MDSCs emerge to suppress inflammation and support new bone formation. Here, we report, for the first time (to our knowledge), the role of MDSCs in the initiation of bone formation. MDSC appeared at the transition from inflammation to bone making and regulates bone healing by suppressing inflammation.
Assuntos
Remodelação Óssea/imunologia , Fraturas Ósseas/imunologia , Células Supressoras Mieloides/imunologia , Osteogênese/imunologia , Animais , Proliferação de Células , Modelos Animais de Doenças , Feminino , Fraturas Ósseas/patologia , Humanos , Inflamação/imunologia , Inflamação/patologia , Camundongos , Tíbia/imunologia , Tíbia/lesões , Tíbia/patologiaRESUMO
Osteoporosis is characterized by low bone mass and damage to the bone tissue's microarchitecture, leading to increased fracture risk. Several studies have provided evidence for associations between psychosocial stress and osteoporosis through various pathways, including the hypothalamic-pituitary-adrenocortical axis, the sympathetic nervous system, and other endocrine factors. As psychosocial stress provokes oxidative cellular stress with consequences for mitochondrial function and cell signaling (e.g., gene expression, inflammation), it is of interest whether extracellular vesicles (EVs) may be a relevant biomarker in this context or act by transporting substances. EVs are intercellular communicators, transfer substances encapsulated in them, modify the phenotype and function of target cells, mediate cell-cell communication, and, therefore, have critical applications in disease progression and clinical diagnosis and therapy. This review summarizes the characteristics of EVs, their role in stress and osteoporosis, and their benefit as biological markers. We demonstrate that EVs are potential mediators of psychosocial stress and osteoporosis and may be beneficial in innovative research settings.
Assuntos
Suscetibilidade a Doenças , Vesículas Extracelulares/metabolismo , Osteoporose/etiologia , Osteoporose/metabolismo , Estresse Psicológico , Animais , Biomarcadores , Remodelação Óssea/genética , Remodelação Óssea/imunologia , Regulação da Expressão Gênica , Humanos , MicroRNAs/genética , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogênese/genética , Osteoporose/patologiaRESUMO
Global data correlate severe vitamin D deficiency with COVID-19-associated coagulopathy, further suggesting the presence of a hypercoagulable state in severe COVID-19 patients, which could promote thrombosis in the lungs and in other organs. The feedback loop between COVID-19-associated coagulopathy and vitamin D also involves platelets (PLTs), since vitamin D deficiency stimulates PLT activation and aggregation and increases fibrinolysis and thrombosis. Vitamin D and PLTs share and play specific roles not only in coagulation and thrombosis but also during inflammation, endothelial dysfunction, and immune response. Additionally, another 'fil rouge' between vitamin D and PLTs is represented by their role in mineral metabolism and bone health, since vitamin D deficiency, low PLT count, and altered PLT-related parameters are linked to abnormal bone remodeling in certain pathological conditions, such as osteoporosis (OP). Hence, it is possible to speculate that severe COVID-19 patients are characterized by the presence of several predisposing factors to bone fragility and OP that may be monitored to avoid potential complications. Here, we hypothesize different pervasive actions of vitamin D and PLT association in COVID-19, also allowing for potential preliminary information on bone health status during COVID-19 infection.
Assuntos
Plaquetas/imunologia , COVID-19/complicações , Osteoporose/imunologia , Trombose/imunologia , Deficiência de Vitamina D/imunologia , Vitamina D/metabolismo , Plaquetas/metabolismo , Remodelação Óssea/imunologia , COVID-19/sangue , COVID-19/diagnóstico , COVID-19/imunologia , Retroalimentação Fisiológica , Humanos , Osteoporose/sangue , Ativação Plaquetária/imunologia , Contagem de Plaquetas , SARS-CoV-2/imunologia , Índice de Gravidade de Doença , Trombose/sangue , Vitamina D/sangue , Deficiência de Vitamina D/sangue , Deficiência de Vitamina D/complicaçõesRESUMO
Osteoarthritis (OA) is a family of degenerative diseases affecting multiple joint tissues. Despite the diverse etiology and pathogenesis of OA, increasing evidence suggests that macrophages can play a significant role in modulating joint inflammation, and thus OA severity, via various secreted mediators. Recent advances in next-generation sequencing technologies coupled with proteomic and epigenetic tools have greatly facilitated research to elucidate the embryonic origin of macrophages in various tissues including joint synovium. Furthermore, scientists have now begun to appreciate that macrophage polarization can span beyond the conventionally recognized binary states (i.e., pro-inflammatory M1-like vs anti-inflammatory M2-like) and may encompass a broad spectrum of phenotypes. Although the presence of these cells has been shown in multiple joint tissues, additional mechanistic studies are required to provide a comprehensive understanding of the precise role of these diverse macrophage populations in OA onset and progression. New approaches that can modulate macrophages into desired functional phenotypes may provide novel therapeutic strategies for preventing OA or enhancing cartilage repair and regeneration.
Assuntos
Cartilagem Articular/imunologia , Inflamação/imunologia , Macrófagos/imunologia , Osteoartrite/imunologia , Regeneração/imunologia , Tecido Adiposo/imunologia , Remodelação Óssea/imunologia , Cartilagem Articular/fisiologia , Humanos , Membrana Sinovial/imunologiaRESUMO
Bone healing is a complex process with closely linked phases of inflammation, regeneration, and remodeling. IL-6 may crucially regulate this process; however, the underlying mechanisms are unclear. IL-6 signals are transmitted via the transmembrane glycoprotein 130 by two distinct mechanisms: classic signaling using the membrane-anchored IL-6 receptor and trans-signaling using its soluble form. Herein, we investigated the hypothesis that IL-6 classic and trans-signaling have different functions during bone healing. To investigate fracture healing, 12-week-old C57BL/6J mice underwent a femur osteotomy. To study the function of IL-6 during the inflammatory phase, either an anti-IL-6 antibody, which inhibits IL-6 classic and trans-signaling, or soluble glycoprotein 130 fusion protein, which selectively blocks trans-signaling, was injected after 30 minutes and 48 hours. To analyze IL-6 effects in the repair phase, compounds were injected from day 7 onwards. Global IL-6 inhibition in the early phase after fracture reduced systemic inflammation, the recruitment of immune cells, and bone regeneration, resulting in delayed fracture healing. Global IL-6 inhibition during the repair phase disturbed bone formation and remodeling. In contrast, inhibition of IL-6 trans-signaling exerted minor effects on the immune response and did not influence bone repair, suggesting that the classic pathway accounts for most of the effects observed after global IL-6 inhibition. Our results reveal that IL-6 classic signaling, but not IL-6 trans-signaling, is essential for bone repair.
Assuntos
Consolidação da Fratura/imunologia , Interleucina-6/imunologia , Animais , Remodelação Óssea/imunologia , Calo Ósseo/imunologia , Quimiocinas/sangue , Citocinas/sangue , Fêmur/fisiologia , Fêmur/cirurgia , Inflamação/imunologia , Mediadores da Inflamação/imunologia , Masculino , Camundongos Endogâmicos C57BL , Osteogênese/imunologia , Osteotomia , Receptores de Interleucina-6/imunologia , Transdução de Sinais/imunologia , Microtomografia por Raio-XRESUMO
We propose a mathematical model describing the dynamics of osteoblasts and osteoclasts in bone remodeling. The goal of this work is to develop an integrated modeling framework for bone remodeling and bone cell signaling dynamics that could be used to explore qualitatively combination treatments for osteoporosis in humans. The model has been calibrated using 57 checks from the literature. Specific global optimization methods based on qualitative objectives have been developed to perform the model calibration. We also added pharmacokinetics representations of three drugs to the model, which are teriparatide (PTH(1-34)), denosumab (a RANKL antibody) and romosozumab (a sclerostin antibody), achieving excellent goodness-of-fit of human clinical data. The model reproduces the paradoxical effects of PTH on the bone mass, where continuous administration of PTH results in bone loss but intermittent administration of PTH leads to bone gain, thus proposing an explanation of this phenomenon. We used the model to simulate different categories of osteoporosis. The main attributes of each disease are qualitatively well captured by the model, for example changes in bone turnover in the disease states. We explored dosing regimens for each disease based on the combination of denosumab and romosozumab, identifying adequate ratios and doses of both drugs for subpopulations of patients in function of categories of osteoporosis and the degree of severity of the disease.
Assuntos
Remodelação Óssea/efeitos dos fármacos , Remodelação Óssea/fisiologia , Modelos Biológicos , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacologia , Conservadores da Densidade Óssea/administração & dosagem , Conservadores da Densidade Óssea/farmacologia , Remodelação Óssea/imunologia , Simulação por Computador , Denosumab/administração & dosagem , Denosumab/farmacologia , Humanos , Conceitos Matemáticos , Osteoblastos/efeitos dos fármacos , Osteoblastos/imunologia , Osteoblastos/fisiologia , Osteoclastos/efeitos dos fármacos , Osteoclastos/imunologia , Osteoclastos/fisiologia , Osteoporose/tratamento farmacológico , Osteoprotegerina/fisiologia , Hormônio Paratireóideo/fisiologia , Ligante RANK/antagonistas & inibidores , Ligante RANK/fisiologia , Receptor Ativador de Fator Nuclear kappa-B/fisiologia , Transdução de Sinais , Teriparatida/administração & dosagem , Teriparatida/farmacologia , Via de Sinalização WntRESUMO
Bone infection and inflammation leads to the infiltration of immune cells at the site of infection, where they modulate the differentiation and function of osteoclasts and osteoblasts by the secretion of various cytokines and signal mediators. In recent years, there has been a tremendous effort to understand the cells involved in these interactions and the complex pathways of signal transduction and their ultimate effect on bone metabolism. These crosstalk mechanisms between the bone and immune system finally emerged, forming a new field of research called osteoimmunology. Diseases falling into the category of osteoimmunology, such as osteoporosis, periodontitis, and bone infections are considered to have a significant implication in mortality and morbidity of patients, along with affecting their quality of life. There is a much-needed research focus in this new field, as the reported data on the immunomodulation of immune cells and their signaling pathways seems to have promising therapeutic benefits for patients.
Assuntos
Doenças Ósseas/microbiologia , Sistema Imunitário/fisiologia , Remodelação Óssea/imunologia , Diferenciação Celular , Humanos , Imunomodulação , Modelos Imunológicos , Osteoblastos/citologia , Osteoblastos/imunologia , Osteoclastos/citologia , Osteoclastos/imunologia , Linfócitos T/imunologiaRESUMO
PURPOSE OF REVIEW: Bone is constantly being remodeled throughout adult life through constant anabolic and catabolic actions that maintain tissue homeostasis. A number of hormones, cytokines growth factors, and the proximity of various cells to bone surfaces influence this process. Inflammatory changes at the bone microenvironment result in alterations leading to both excessive bone loss and bone formation. Detailed understanding of the physiological and pathological mechanisms that dictate these changes will allow us to harness inflammatory signals in bone regeneration. RECENT FINDINGS: Recent reports have suggested that inflammatory signals are able to stimulate transcription factors that regulate osteoblast differentiation from their precursors. SUMMARY: In this review, we summarized current understanding of the roles of inflammation in bone resorption and bone formation, which give rise to different disorders and discuss the huge potential of harnessing these inflammatory signals to achieve bone regeneration.
Assuntos
Remodelação Óssea/imunologia , Reabsorção Óssea/imunologia , Osso e Ossos/imunologia , Diferenciação Celular/imunologia , Citocinas/imunologia , Inflamação/imunologia , Osteogênese/imunologia , Animais , Humanos , Osteoblastos/citologia , Osteoblastos/imunologia , Osteoclastos/citologia , Osteoclastos/imunologia , Fatores de Transcrição/metabolismoRESUMO
Psoriatic arthritis (PsA) is an inflammatory rheumatic disorder that occurs in patients with psoriasis and predominantly affects musculoskeletal structures, skin, and nails. The etiology of PsA is not well understood but evidence supports an interplay of genetic, immunologic, and environmental factors which promote pathological bone remodeling and joint damage in PsA. Localized and systemic bone loss due to increased activity of osteoclasts is well established in PsA based on animal models and translational studies. In contrast, the mechanisms responsible for pathological bone remodeling in PsA remain enigmatic although new candidate molecules and pathways have been identified. Recent reports have revealed novel findings related to bone erosion and pathologic bone formation in PsA. Many associated risk factors and contributing molecular mechanisms have also been identified. In this review, we discuss new developments in the field, point out unresolved questions regarding the pathogenetic origins of the wide array of bone phenotypes in PsA, and discuss new directions for investigation.
Assuntos
Artrite Psoriásica/tratamento farmacológico , Remodelação Óssea/efeitos dos fármacos , Reabsorção Óssea/tratamento farmacológico , Osso e Ossos/efeitos dos fármacos , Animais , Artrite Psoriásica/imunologia , Remodelação Óssea/imunologia , Reabsorção Óssea/imunologia , Osso e Ossos/imunologia , Humanos , Osteoclastos/efeitos dos fármacos , Osteoclastos/imunologia , Osteoprotegerina/imunologiaRESUMO
The signaling network involved in the pathogenesis of periodontal disease is not yet fully understood. This review aims to describe possible mechanisms through which the bacterial modulators may be linked directly or indirectly to the process of alveolar bone loss in periodontitis. From the late 1970s to present, new paradigm shifts have been developed regarding our understanding of pathological bone remodeling in periodontal disease. Upcoming evidence suggests that in periodontal disease the local immune response is exacerbated and involves the existence of signaling pathways that have been shown to modulate bone-cell function leading to alveolar bone loss. Those complex signaling pathways have been observed not only between bacteria but also between bacteria and the gingival surface of the host. More specifically, it has been shown that bacteria, through their secretion molecules, may interact indirectly and directly with immune-type cells of the host, resulting in the production of osteolytic agents that enhance bone resorption. Further research is required to provide a clear understanding of the role of these molecules in the pathogenesis of periodontal disease, and the availability of new technologies, such as next-generation sequencing and metagenomic analysis, may be useful tools in achieving this.
Assuntos
Perda do Osso Alveolar/imunologia , Bactérias/imunologia , Fenômenos Fisiológicos Bacterianos/imunologia , Remodelação Óssea/imunologia , Bolsa Periodontal/imunologia , Perda do Osso Alveolar/microbiologia , Perda do Osso Alveolar/patologia , Antígenos de Bactérias/imunologia , Autoimunidade , Bactérias/metabolismo , Bactérias/patogenicidade , Citocinas/metabolismo , Humanos , Lipopeptídeos , Lipopolissacarídeos/imunologia , Lipoproteínas , Osteoclastos , Osteólise/imunologia , Doenças Periodontais/imunologia , Doenças Periodontais/microbiologia , Doenças Periodontais/patologia , Bolsa Periodontal/microbiologia , Bolsa Periodontal/patologia , Periodontite/imunologia , Periodontite/microbiologia , Periodontite/patologia , Ácidos TeicoicosRESUMO
PURPOSE OF REVIEW: Osteonecrosis of the jaw (ONJ) is a rare and severe necrotic bone disease reflecting a compromise in the body's osseous healing mechanisms and unique to the craniofacial region. Antiresorptive and antiangiogenic medications have been suggested to be associated with the occurrence of ONJ; yet, the pathophysiology of this disease has not been fully elucidated. This article raises the current theories underlying the pathophysiology of ONJ. RECENT FINDINGS: The proposed mechanisms highlight the unique localization of ONJ. The evidence-based mechanisms of ONJ pathogenesis include disturbed bone remodeling, inflammation or infection, altered immunity, soft tissue toxicity, and angiogenesis inhibition. The role of dental infections and the oral microbiome is central to ONJ, and systemic conditions such as rheumatoid arthritis and diabetes mellitus contribute through their impact on immune resiliency. Current experimental studies on mechanisms of ONJ are summarized. The definitive pathophysiology is as yet unclear. Recent studies are beginning to clarify the relative importance of the proposed mechanisms. A better understanding of osteoimmunology and the relationship of angiogenesis to the development of ONJ is needed along with detailed studies of the impact of drug holidays on the clinical condition of ONJ.
Assuntos
Remodelação Óssea/imunologia , Infecções/imunologia , Inflamação/imunologia , Doenças Maxilomandibulares/imunologia , Osteonecrose/imunologia , Inibidores da Angiogênese/efeitos adversos , Osteonecrose da Arcada Osseodentária Associada a Difosfonatos/etiologia , Osteonecrose da Arcada Osseodentária Associada a Difosfonatos/imunologia , Osteonecrose da Arcada Osseodentária Associada a Difosfonatos/metabolismo , Conservadores da Densidade Óssea/efeitos adversos , Remodelação Óssea/fisiologia , Colágeno/metabolismo , Difosfonatos/efeitos adversos , Humanos , Infecções/metabolismo , Inflamação/metabolismo , Doenças Maxilomandibulares/induzido quimicamente , Doenças Maxilomandibulares/metabolismo , Células Matadoras Naturais/imunologia , Mucosa Bucal/imunologia , Mucosa Bucal/lesões , Mucosa Bucal/metabolismo , Neutrófilos/imunologia , Osteonecrose/induzido quimicamente , Osteonecrose/metabolismo , Linfócitos T/imunologia , CicatrizaçãoRESUMO
Osteoimmunology is an interdisciplinary research field focused on the molecular understanding of the interplay between the immune and skeletal systems. Although osteoimmunology started with the study of the immune regulation of osteoclasts, its scope has been extended to encompass a wide range of molecular and cellular interactions, including those between osteoblasts and osteoclasts, lymphocytes and osteoclasts, and osteoblasts and haematopoietic cells. Therefore, the two systems should be understood to be integrated and operating in the context of the 'osteoimmune' system, a heuristic concept that provides not only a framework for obtaining new insights by basic research, but also a scientific basis for the discovery of novel treatments for diseases related to both systems.
Assuntos
Osso e Ossos/imunologia , Sistema Imunitário/fisiologia , Osteoblastos/imunologia , Osteoclastos/imunologia , Transdução de Sinais/imunologia , Animais , Desenvolvimento Ósseo/imunologia , Remodelação Óssea/imunologia , Reabsorção Óssea/imunologia , Comunicação Celular/imunologia , Diferenciação Celular/imunologia , Humanos , Linfócitos/imunologia , Osteoblastos/citologia , Osteoclastos/citologiaRESUMO
Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic inflammation, local and systemic bone loss and a lack of compensatory bone repair. Fibroblast-like synoviocytes (FLS) are the most abundant cells of the stroma and a key population in autoimmune diseases such as RA. An increasing body of evidence suggests that these cells play not only an important role in chronic inflammation and synovial hyperplasia, but also impact bone remodelling. Under inflammatory conditions FLS release inflammatory cytokines, regulate bone destruction and formation and communicate with immune cells to control bone homeostasis. Other stromal cells, such as osteoblasts and terminally differentiated osteoblasts, termed osteocytes, are also involved in the regulation of bone homeostasis and are dysregulated during inflammation. This review highlights our current understanding of how stromal cells influence the balance between bone formation and bone destruction. Increasing our understanding of these processes is critical to enable the development of novel therapeutic strategies with which to treat bone loss in RA.
Assuntos
Artrite Reumatoide/complicações , Reabsorção Óssea/imunologia , Osso e Ossos/patologia , Osteócitos/imunologia , Células Estromais/citologia , Sinoviócitos/citologia , Artrite Reumatoide/imunologia , Remodelação Óssea/imunologia , Reabsorção Óssea/terapia , Osso e Ossos/citologia , Citocinas/imunologia , Citocinas/farmacologia , Humanos , Hiperplasia , Inflamação/patologia , Células Estromais/imunologia , Sinoviócitos/imunologia , Via de Sinalização Wnt/imunologiaRESUMO
Osteoporosis increases fracture risk, a cause of crippling morbidity and mortality. The immunoskeletal interface (ISI) is a centralization of cell and cytokine effectors shared between skeletal and immune systems. Consequently, the immune system mediates powerful effects on bone turnover. Physiologically, B cells secrete osteoprotegerin (OPG), a potent anti-osteoclastogenic factor that preserves bone mass. However, activated T cells and B cells secrete pro-osteoclastogenic factors including receptor activator of Nuclear factor-kappaB (NF-kB) ligand (RANKL), Interleukin (IL)-17A, and tumor necrosis factor (TNF)-α promoting bone loss in inflammatory states such as rheumatoid arthritis. Recently, ISI disruption has been linked to osteoporosis in human immunodeficiency virus (HIV) infection/acquired immunodeficiency syndrome (AIDS), where elevated B cell RANKL and diminished OPG drive bone resorption. HIV-antiretroviral therapy paradoxically intensifies bone loss during disease reversal, as immune reconstitution produces osteoclastogenic cytokines. Interestingly, in estrogen deficiency, activated T cells secrete RANKL, TNF, and IL-17A that amplify bone resorption and contribute to postmenopausal osteoporosis. T cell-produced TNF and IL-17A further contribute to bone loss in hyperparathyroidism, while T cell production of the anabolic Wingless integration site (Wnt) ligand, Wnt10b, promotes bone formation in response to anabolic parathyroid hormone and the immunomodulatory costimulation inhibitor cytotoxic T lymphocyte-associated protein-4-IgG (abatacept). These findings provide a window into the workings of the ISI and suggest novel targets for future therapeutic interventions to reduce fracture risk.
Assuntos
Remodelação Óssea/imunologia , Reabsorção Óssea/imunologia , Osso e Ossos/imunologia , Sistema Imunitário , Animais , Terapia Antirretroviral de Alta Atividade/efeitos adversos , Remodelação Óssea/efeitos dos fármacos , Reabsorção Óssea/tratamento farmacológico , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/fisiologia , Citocinas/metabolismo , Estrogênios/sangue , Estrogênios/deficiência , Infecções por HIV/complicações , Infecções por HIV/tratamento farmacológico , Humanos , Linfócitos/imunologia , NF-kappa B/metabolismo , Osteoclastos/metabolismo , Osteoprotegerina/metabolismo , Hormônio Paratireóideo/fisiologia , Ligante RANK/metabolismo , Fator de Necrose Tumoral alfa/metabolismoRESUMO
PURPOSE OF THE REVIEW: In this review, we present the role of regulatory T (Treg) cells in bone remodelling and bone-related disease such as osteoporosis or inflammatory bone loss. We also discuss the cellular and molecular mechanism how Treg cells regulate osteoclastogenesis. RECENT FINDINGS: Treg cells could regulate osteoclastogenesis by secreting TGF-ß and IL-10 as well as IL-4 cytokines. Moreover, Treg cells can additionally regulate osteoclast differentiation, in a cell-to-cell contact via the cytotoxic T lymphocyte antigen (CTLA-4). The latter induces the apoptosis of osteoclasts dependent on CD80/86 in vitro and in vivo. Treg cells mediate immunosuppressive function that controls undesired immune reactions, such as autoimmunity. Recently, Treg cells have been shown to influence non-immunological processes, such as bone homeostasis. Accumulated evidences have demonstrated that Treg cells can suppress osteoclast differentiation in vitro and in vivo.
Assuntos
Remodelação Óssea/imunologia , Osteoporose/imunologia , Linfócitos T Reguladores/imunologia , Apoptose/imunologia , Antígeno CTLA-4/imunologia , Humanos , Interleucina-10/imunologia , Interleucina-4/imunologia , Osteoclastos , Osteogênese/imunologia , Fator de Crescimento Transformador beta/imunologiaRESUMO
PURPOSE OF REVIEW: In the process of bone fracture healing, inflammation is thought to be an essential process that precedes bone formation and remodeling. We review recent studies on bone fracture healing from an osteoimmunological point of view. RECENT FINDINGS: Based on previous observations that many types of immune cells infiltrate into the bone injury site and release a variety of molecules, recent studies have addressed the roles of specific immune cell subsets. Macrophages and interleukin (IL)-17-producing γδ T cells enhance bone healing, whereas CD8+ T cells impair bone repair. Additionally, IL-10-producing B cells may contribute to bone healing by suppressing excessive and/or prolonged inflammation. Although the involvement of other cells and molecules has been suggested, the precise underlying mechanisms remain elusive. Accumulating evidence has begun to reveal the deeper picture of bone fracture healing. Further studies are required for the development of novel therapeutic strategies for bone fracture.