Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Curr Osteoporos Rep ; 18(1): 32-37, 2020 02.
Article in English | MEDLINE | ID: mdl-31970653

ABSTRACT

PURPOSE OF REVIEW: We reviewed recent progress on the role of sclerostin (SOST) and its effects on the immune system in order to summarize the current state of knowledge in osteoimmunology, in regard to hematopoiesis, lymphopoiesis, and inflammation. RECENT FINDINGS: Changes in sclerostin levels affect distinct niches within the bone marrow that support hematopoietic stem cells and B cell development. Sclerostin's regulation of adipogenesis could also be important for immune cell maintenance with age. Surprisingly, B cell development in the bone marrow is influenced by Sost produced by mesenchymal stem cells and osteoblasts, but not by osteocytes. Additionally, extramedullary hematopoiesis in the spleen and increased pro-inflammatory cytokine levels in the bone marrow are observed in global Sost-/- mice. In addition to changes in bone marrow density, sclerostin depletion affects B lymphopoiesis and myelopoiesis, as well as other changes within the bone marrow cavity that could affect hematopoiesis. It is therefore important to monitor for hematopoietic changes in patients receiving sclerostin-depleting therapies.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Adipogenesis/immunology , Hematopoiesis, Extramedullary/immunology , Lymphopoiesis/immunology , Animals , B-Lymphocytes , Bone Marrow/immunology , Cytokines/immunology , Hematopoiesis/immunology , Hematopoietic Stem Cells , Humans , Mesenchymal Stem Cells/immunology , Mice , Mice, Knockout , Myelopoiesis/immunology , Osteoblasts/immunology , Osteocytes/immunology
2.
Biochem Biophys Res Commun ; 501(2): 547-555, 2018 06 22.
Article in English | MEDLINE | ID: mdl-29746861

ABSTRACT

Osteolytic diseases are closely associated with osteocyte fate, indicating a more efficient and crucial role of osteocyte-targeting strategy in inhibiting osteoclastogenesis. Here, we investigated the effects of lenalidomide (Lena) on osteocyte fate in order to regulate osteoclastogenesis via effective cascade-controlling response. Our data revealed that lenalidomide treatment notably rescued IL-1ß induced loss of osteocyte viability by inhibiting osteocyte apoptosis with decreased osteoclast-related factors, RANKL and Sclerostin, as demonstrated by the restricted osteoclast formation and reduced bone resorption. Additionally, iTRAQ assay revealed that IL-1ß induced activation of NF-κB inhibitor α/ß were remarkably downregulated by lenalidomide, showing that lenalidomide impaired NF-κB signaling in osteocytes for inhibiting the expression of osteoclast specific genes in osteoclasts, which was further confirmed by KEGG pathway analysis and Western blot. More interestingly, the in vivo analysis of osteocyte apoptosis and osteoclastogenesis in osteoarthritis mice model indicated a role of lenalidomide in the regulation of osteocyte fate and the consequent inhibition of RANKL-induced osteoclastogenesis. Together, these results suggest that lenalidomide regulates osteocyte fate by attenuating IL-1ß/NF-κB signaling, thereby inhibiting RANKL expression for the attenuated osteoclastogenesis both in vitro and vivo, indicating a more efficient remedy among future anti-osteoclastogenesis approaches.


Subject(s)
Immunologic Factors/pharmacology , Interleukin-1beta/immunology , NF-kappa B/immunology , Osteocytes/drug effects , RANK Ligand/immunology , Signal Transduction/drug effects , Thalidomide/analogs & derivatives , Animals , Cell Line , Cells, Cultured , Lenalidomide , Mice, Inbred C57BL , Osteoclasts/cytology , Osteoclasts/drug effects , Osteoclasts/immunology , Osteocytes/cytology , Osteocytes/immunology , Osteogenesis/drug effects , Thalidomide/pharmacology
3.
Clin Exp Immunol ; 189(1): 1-11, 2017 07.
Article in English | MEDLINE | ID: mdl-28419440

ABSTRACT

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.


Subject(s)
Arthritis, Rheumatoid/complications , Bone Resorption/immunology , Bone and Bones/pathology , Osteocytes/immunology , Stromal Cells/cytology , Synoviocytes/cytology , Arthritis, Rheumatoid/immunology , Bone Remodeling/immunology , Bone Resorption/therapy , Bone and Bones/cytology , Cytokines/immunology , Cytokines/pharmacology , Humans , Hyperplasia , Inflammation/pathology , Stromal Cells/immunology , Synoviocytes/immunology , Wnt Signaling Pathway/immunology
4.
Amino Acids ; 49(3): 683-693, 2017 03.
Article in English | MEDLINE | ID: mdl-27357308

ABSTRACT

Osteoporosis (OP) and osteoarthritis (OA) are the most common joint diseases, with a high incidence in the elderly population. OP is characterized by trabecular bone remodeling and reabsorption, whereas articular cartilage and subchondral bone remodeling are major features of OA. Although classically considered as independent or even conflicting processes, clinical coexistence of OP and OA was recently described. Transglutaminase 2 (TG2) expression is considered a biomarker of OA, but its role in osteoporotic bone remodeling is still uncertain. We investigated TG2 and bone biological markers (Osteocalcin, Osteopontin, and Sclerostin) in osteoporotic and osteoarthritic osteocartilagineous tissue (n = 54) and human chondrocyte cultures in vitro by immunohistochemistry, immunofluorescence and RT-PCR. Histomorphometric evaluation of bone trabecular remodeling was also performed. In cartilage, TG2 expression was faint in control and OP and significantly less than in OA and OP + OA chondrocytes; the opposite was found for Osteocalcin, whereas Osteopontin and Sclerostin expression was similar. In the subchondral trabecular bone, osteocytes/osteoblasts TG2 expression was slight and similar comparing control, OP, OA, and OP + OA group, whereas Osteocalcin and Osteopontin expression was lower in OP compared to control, OA and OP + OA. Increased TG2 and reduced Osteocalcin expression were maintained in human osteoarthritic chondrocytes in vitro. Histomorphometric analysis confirmed reduced trabecular bone mass in OP and OP + OA compared with OA patients. TG2 represented a suitable biomarker of osteoarthritic chondrocyte activation, whereas osteocalcin and osteopontin characterized osteoporotic osteocyte/osteoblast changes; differences were lost in OP + OA patients, suggesting careful consideration when coexistence of the two diseases occurs.


Subject(s)
Bone Morphogenetic Proteins/immunology , GTP-Binding Proteins/immunology , Genetic Markers/immunology , Osteoarthritis/immunology , Osteocalcin/immunology , Osteopontin/immunology , Osteoporosis/immunology , Transglutaminases/immunology , Adaptor Proteins, Signal Transducing , Aged , Biomarkers/metabolism , Bone Morphogenetic Proteins/genetics , Bone and Bones/immunology , Bone and Bones/pathology , Cartilage, Articular/immunology , Cartilage, Articular/pathology , Chondrocytes/immunology , Chondrocytes/pathology , Female , GTP-Binding Proteins/genetics , Gene Expression , Genetic Markers/genetics , Humans , Male , Middle Aged , Osteoarthritis/genetics , Osteoarthritis/pathology , Osteoblasts/immunology , Osteoblasts/pathology , Osteocalcin/genetics , Osteocytes/immunology , Osteocytes/pathology , Osteopontin/genetics , Osteoporosis/genetics , Osteoporosis/pathology , Primary Cell Culture , Protein Glutamine gamma Glutamyltransferase 2 , Transglutaminases/genetics
5.
Clin Calcium ; 26(5): 699-706, 2016 May.
Article in Japanese | MEDLINE | ID: mdl-27117615

ABSTRACT

Multiple myeloma(MM)develops and expands almost exclusively in the bone marrow, and generates devastating bone destruction. MM cells produce a variety of cytokines to stimulate RANKL-mediated osteoclastogenesis and suppress osteoblastic differentiation from bone marrow stromal cells, leading to extensive bone destruction with rapid loss of bone. Furthermore, osteocyte apoptosis has been demonstrated to be induced in parallel with enhanced osteoclast recruitment and osteoclastogenesis in myeloma bone lesions. Of note, osteocytes physically interact with myeloma cells to skew their signaling pathways and thereby production of mediators responsible for exacerbated bone resorption and suppressed bone formation in myeloma. The role of osteocytes in myeloma-induced bone lesions remains to be further clarified.


Subject(s)
Bone Diseases/immunology , Bone Marrow/metabolism , Bone and Bones/metabolism , Multiple Myeloma/pathology , Osteoclasts/cytology , Osteocytes/immunology , Animals , Bone and Bones/immunology , Humans , Multiple Myeloma/immunology , Osteocytes/metabolism
6.
Calcif Tissue Int ; 94(1): 98-111, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23912951

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.


Subject(s)
Immune System/metabolism , Osteoclasts/metabolism , Osteocytes/metabolism , Osteogenesis/immunology , Animals , Bone Resorption/immunology , Humans , Immune System/immunology , Inflammation/immunology , Inflammation/metabolism , Osteoclasts/immunology , Osteocytes/immunology , Osteogenesis/physiology
7.
Bull Exp Biol Med ; 157(1): 119-24, 2014 May.
Article in English | MEDLINE | ID: mdl-24909727

ABSTRACT

We describe a method of isolation of human mesenchymal stromal cells from the umbilical cord (Wharton's jelly) and human placenta: amnion, placental villi, and trophoblast. Morphology, immunophenotypic characteristics, and differentiation potencies of isolated cells were studied. The capacity of mesenchymal stromal cells from extraembryonic tissues to osteogenic, adipogenic, and chondrogenic differentiation was demonstrated and the dynamics of this process was described. The isolated cells met the criteria for multipotent mesenchymal stem cells.


Subject(s)
Amnion/cytology , Mesenchymal Stem Cells/cytology , Multipotent Stem Cells/cytology , Umbilical Cord/cytology , Wharton Jelly/cytology , Adipocytes/cytology , Adipocytes/immunology , Amnion/immunology , Antigens, CD/immunology , Antigens, CD/metabolism , Biomarkers/metabolism , Cell Differentiation , Cell Separation , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/immunology , Chorionic Villi/immunology , Female , HLA-DR Antigens/immunology , HLA-DR Antigens/metabolism , Humans , Immunophenotyping , Mesenchymal Stem Cells/immunology , Multipotent Stem Cells/immunology , Osteocytes/cytology , Osteocytes/immunology , Pregnancy , Trophoblasts/cytology , Trophoblasts/immunology , Umbilical Cord/immunology , Wharton Jelly/immunology
8.
Clin Calcium ; 24(1): 69-73, 2014 Jan.
Article in Japanese | MEDLINE | ID: mdl-24369282

ABSTRACT

In a rare accident of nature, some families have been found to have dense and strong bones due to a recessive loss of function mutation in the SOST gene that encodes for sclerostin, a protein expressed by osteocytes that downregulates osteoblastic bone formation. Knowledge of this molecule and its actions led rather quickly to the development of anti-sclerostin antibodies that lead to marked increases in bone mass in both animals and human subjects. Blocking sclerostin action with anti-sclerostin antibodies is a promising new therapeutic approach to osteoanabolic therapy of osteoporosis.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Bone Density/physiology , Bone and Bones/metabolism , Osteoporosis/drug therapy , Animals , Bone Density/immunology , Bone and Bones/immunology , Humans , Osteocytes/immunology , Osteocytes/metabolism , Osteoporosis/immunology , Osteoporosis/metabolism , Signal Transduction
9.
Biomed Pharmacother ; 177: 117139, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39018871

ABSTRACT

The notion that obesity can be a protective factor for bone health is a topic of ongoing debate. Increased body weight may have a positive impact on bone health due to its mechanical effects and the production of estrogen by adipose tissue. However, recent studies have found a higher risk of bone fracture and delayed bone healing in elderly obese patients, which may be attributed to the heightened risk of bone immune regulation disruption associated with obesity. The balanced functions of bone cells such as osteoclasts, osteoblasts, and osteocytes, would be subverted by aberrant and prolonged immune responses under obese conditions. This review aims to explore the intricate relationship between obesity and bone health from the perspective of osteoimmunology, elucidate the impact of disturbances in bone immune regulation on the functioning of bone cells, including osteoclasts, osteoblasts, and osteocytes, highlighting the deleterious effects of obesity on various diseases development such as rheumatoid arthritis (RA), osteoarthritis (AS), bone fracture, periodontitis. On the one hand, weight loss may achieve significant therapeutic effects on the aforementioned diseases. On the other hand, for patients who have difficulty in losing weight, the osteoimmunological therapies could potentially serve as a viable approach in halting the progression of these disease. Additional research in the field of osteoimmunology is necessary to ascertain the optimal equilibrium between body weight and bone health.


Subject(s)
Bone and Bones , Obesity , Humans , Obesity/immunology , Obesity/complications , Animals , Bone and Bones/immunology , Bone and Bones/metabolism , Bone and Bones/pathology , Osteocytes/metabolism , Osteocytes/immunology , Osteoclasts/immunology , Osteoclasts/metabolism , Osteoblasts/immunology , Osteoblasts/metabolism , Bone Remodeling/immunology
10.
Bull Exp Biol Med ; 155(5): 650-4, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24288731

ABSTRACT

We studied the effect of BMP-2 added to the culture medium on osteogenic and proliferative properties of multipotent stromal cells (MSC) and on the expression of cytokine genes induced by immunization of experimental animals with bacterial antigens. It is shown that the presence of BMP-2 in the culture medium stimulates proliferation of bone marrow MSC and especially spleen MSC (which was seen from enlargement of MSC colonies); improves the efficiency of MSC cloning; increases osteogenic activity of mouse bone marrow MSC; induces osteogenic differentiation of splenic MSC (osteogenesis is normally not observed in the spleen); reduces the number of macrophages in cultures; inhibits synthesis of mRNA for proinflammatory cytokines (IL-1ß, IL-6, IL-8, TNF-α) that typically occurs in cultures of the bone marrow and spleen from animals immunized with S. typhimurium or group A streptococcus antigens. Bearing in mind that proinflammatory cytokines negatively affect osteogenic activity of the bone marrow, we can hypothesize that BMP-2 not only stimulates osteogenesis, but also provides optimal conditions for its realization by suppressing the expression of genes encoding these cytokines.


Subject(s)
Antigens, Bacterial/immunology , Bone Marrow Cells/drug effects , Bone Morphogenetic Protein 2/pharmacology , Mesenchymal Stem Cells/drug effects , RNA, Messenger/antagonists & inhibitors , Spleen/drug effects , Animals , Antigens, Bacterial/administration & dosage , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Cell Count , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Gene Expression , Immunization , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/biosynthesis , Interleukin-6/antagonists & inhibitors , Interleukin-6/biosynthesis , Interleukin-8/antagonists & inhibitors , Interleukin-8/biosynthesis , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/immunology , Mice , Mice, Inbred CBA , Osteocytes/cytology , Osteocytes/drug effects , Osteocytes/immunology , Osteogenesis/drug effects , Primary Cell Culture , RNA, Messenger/biosynthesis , Spleen/cytology , Spleen/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis
11.
Cell Immunol ; 271(1): 147-56, 2011.
Article in English | MEDLINE | ID: mdl-21757189

ABSTRACT

Mesenchymal stem cells (MSCs) have potent regulatory effects on immune and inflammatory responses. Recently the findings of functional TLR expression on MSC implicates these receptors in the function established for MSCs. Here we specially investigated the effects of TLR2, 4 ligation in mice MSC on migration, modulation of allogeneic mixed lymphocytes reaction (allo-MLR) and inducing Treg cells. We demonstrated that ligation of TLR2, but not TLR4, could significantly inhibit migration of MSC, impair MSC-mediated immunosuppression on allo-MLR, and reduce MSC-mediated expansion of CD4+CD25+Foxp3+ regulatory T cells. Compared with TLR4 activated MSCs and non-TLR activated MSC, TLR2 activation induced a relatively lower level of CXCL-10 mRNA and protein expressions which has been elucidated to act in concert with other soluble factor in MSC-mediated immunomodulation. These data indicate that TLR2 and TLR4 ligation had different effects on immunomodulatory capability of murine BMSCs, which should be considered in their use for treating inflammatory diseases.


Subject(s)
Bone Marrow Cells/immunology , Mesenchymal Stem Cells/immunology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/immunology , Adipocytes/immunology , Adipocytes/metabolism , Animals , Antigens, Ly/genetics , Antigens, Ly/metabolism , Bone Marrow Cells/metabolism , Cell Movement/immunology , Cell Proliferation , Cells, Cultured , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Chemotaxis/drug effects , Chemotaxis/immunology , Cysteine/analogs & derivatives , Cysteine/pharmacology , Dose-Response Relationship, Drug , Flow Cytometry , Lipoproteins/pharmacology , Lymphocyte Activation/immunology , Lymphocyte Culture Test, Mixed , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Osteocytes/immunology , Osteocytes/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism
12.
Cell Biol Int ; 34(7): 693-701, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20187873

ABSTRACT

Many studies have drawn attention to the emerging role of MSC (mesenchymal stem cells) as a promising population supporting new clinical concepts in cellular therapy. However, the sources from which these cells can be isolated are still under discussion. Whereas BM (bone marrow) is presented as the main source of MSC, despite the invasive procedure related to this source, the possibility of isolating sufficient numbers of these cells from UCB (umbilical cord blood) remains controversial. Here, we present the results of experiments aimed at isolating MSC from UCB, BM and UCM (umbilical cord matrix) using different methods of isolation and various culture media that summarize the main procedures and criteria reported in the literature. Whereas isolation of MSC were successful from BM (10:10) and (UCM) (8:8), only one cord blood sample (1:15) gave rise to MSC using various culture media [DMEM (Dulbecco's modified Eagle's medium) +5% platelet lysate, DMEM+10% FBS (fetal bovine serum), DMEM+10% human UCB serum, MSCGM] and different isolation methods [plastic adherence of total MNC (mononuclear cells), CD3+/CD19+/CD14+/CD38+-depleted MNC and CD133+- or LNGFR+-enriched MNC]. MSC from UCM and BM were able to differentiate into adipocytes, osteocytes and hepatocytes. The expansion potential was highest for MSC from UCM. The two cell populations had CD90+/CD73+/CD105+ phenotype with the additional expression of SSEA4 and LNGFR for BM MSC. These results clearly exclude UCB from the list of MSC sources for clinical use and propose instead UCM as a rich, non-invasive and abundant source of MSC.


Subject(s)
Fetal Blood/cytology , Mesenchymal Stem Cells/cytology , Umbilical Cord/cytology , Adipocytes/cytology , Adipocytes/immunology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Cattle , Cell Culture Techniques/methods , Cell Differentiation/physiology , Cell Separation/methods , Cells, Cultured , Flow Cytometry , Hepatocytes/cytology , Hepatocytes/immunology , Humans , Immunophenotyping , Mesenchymal Stem Cells/immunology , Osteocytes/cytology , Osteocytes/immunology , Phenotype
13.
Nat Rev Immunol ; 19(10): 626-642, 2019 10.
Article in English | MEDLINE | ID: mdl-31186549

ABSTRACT

In terrestrial vertebrates, bone tissue constitutes the 'osteoimmune' system, which functions as a locomotor organ and a mineral reservoir as well as a primary lymphoid organ where haematopoietic stem cells are maintained. Bone and mineral metabolism is maintained by the balanced action of bone cells such as osteoclasts, osteoblasts and osteocytes, yet subverted by aberrant and/or prolonged immune responses under pathological conditions. However, osteoimmune interactions are not restricted to the unidirectional effect of the immune system on bone metabolism. In recent years, we have witnessed the discovery of effects of bone cells on immune regulation, including the function of osteoprogenitor cells in haematopoietic stem cell regulation and osteoblast-mediated suppression of haematopoietic malignancies. Moreover, the dynamic reciprocal interactions between bone and malignancies in remote organs have attracted attention, extending the horizon of osteoimmunology. Here, we discuss emerging concepts in the osteoimmune dialogue in health and disease.


Subject(s)
Bone and Bones/immunology , Animals , Hematopoietic Stem Cells/physiology , Humans , Immune System/physiology , Osteoblasts/immunology , Osteoclasts/immunology , Osteocytes/immunology , Osteogenesis , Signal Transduction/physiology
14.
J Immunol Res ; 2019: 9716758, 2019.
Article in English | MEDLINE | ID: mdl-31341915

ABSTRACT

Osteocytes are abundant cells in bone, which contribute to bone maintenance. Osteocytes express receptor activator of nuclear factor kappa-B ligand (RANKL) and regulate osteoclast formation. Orthodontic tooth movement (OTM) occurs by osteoclast resorption of alveolar bone. Osteocyte-derived RANKL is critical in bone resorption during OTM. Additionally, tumor necrosis factor-α (TNF-α) is important in osteoclastogenesis during OTM. Sclerostin has been reported to enhance RANKL expression in the MLO-Y4 osteocyte-like cell line. This study investigated the effect of TNF-α on sclerostin expression in osteocytes during OTM. In vitro analysis of primary osteocytes, which were isolated from DMP1-Topaz mice by sorting the Topaz variant of GFP-positive cells, revealed that SOST mRNA expression was increased when osteocytes were cultured with TNF-α and that RANKL mRNA expression was increased when osteocytes were cultured with sclerostin. Moreover, the number of TRAP-positive cells was increased in osteocytes and osteoclast precursors cocultured with sclerostin. In vivo analysis of mouse calvariae that had been subcutaneously injected with phosphate-buffered saline (PBS) or TNF-α revealed that the number of TRAP-positive cells and the percentage of sclerostin-positive osteocytes were higher in the TNF-α group than in the PBS group. Furthermore, the level of SOST mRNA was increased by TNF-α. As an OTM model, a Ni-Ti closed-coil spring connecting the upper incisors and upper-left first molar was placed to move the first molar to the mesial direction in wild-type (WT) mice and TNF receptor 1- and 2-deficient (TNFRsKO) mice. After 6 days of OTM, the percentage of sclerostin-positive osteocytes on the compression side of the first molar in TNFRsKO mice was lower than that in WT mice. In this study, TNF-α increased sclerostin expression in osteocytes, and sclerostin enhanced RANKL expression in osteocytes. Thus, TNF-α may play an important role in sclerostin expression in osteocytes and enhance osteoclast formation during OTM.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Osteocytes/metabolism , Osteogenesis , RANK Ligand/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Adaptor Proteins, Signal Transducing , Animals , Cell Line , Cells, Cultured , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteocytes/cytology , Osteocytes/immunology , Osteogenesis/drug effects , Osteogenesis/genetics , Tooth Movement Techniques
15.
Curr Drug Targets ; 20(1): 1-15, 2019.
Article in English | MEDLINE | ID: mdl-29618305

ABSTRACT

Glucocorticoids are widely used to treat varieties of allergic and autoimmune diseases, however, long-term application results in glucocorticoid-induced osteoporosis (GIOP). Inflammatory cytokines: tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) play important regulatory roles in bone metabolism, but their roles in GIOP remain largely unknown. Osteocytes can modulate the formation and function of both osteoblasts and osteoclasts, directly via gap junctions, or indirectly by transferring molecule signaling. Apoptotic osteocytes release RANKL, HMGB1 and pro-inflammatory cytokines to stimulate osteoclastogenesis. Moreover, osteocytes can secrete FGF23 to regulate bone metabolism. Exposure to high levels of GCs can drive osteocyte apoptosis and influence gap junctions, leading to bone loss. GCs treatment is regarded to produce more FGF23 to inhibit bone mineralization. GCs also disrupt the vascular to decrease osteocyte feasibility and mineral appositional rate, resulting in a decline in bone strength. Apoptotic bodies from osteocytes induced by GCs treatment can enhance production of TNF-α and IL-6. On the other hand, TNF-α and IL-6 show synergistic effects by altering osteocytes signaling towards osteoclasts and osteoblasts. In addition, TNF-α can induce osteocyte apoptosis and attribute to a worsened bone quality in GCs. IL-6 and osteocytes may interact with each other. Therefore, we hypothesize that GCs regulate osteocyteogenesis through TNF-α and IL-6, which are highly expressed around osteocyte undergoing apoptosis. In the present review, we summarized the roles of osteocytes in regulating osteoblasts and osteoclasts. Furthermore, the mechanism of GCs altered relationship between osteocytes and osteoblasts/osteoclasts. In addition, we discussed the roles of TNF-α and IL-6 in GIOP by modulating osteocytes. Lastly, we discussed the possibility of using pro-inflammatory signaling pathway as therapeutic targets to develop drugs for GIOP.


Subject(s)
Glucocorticoids/adverse effects , Interleukin-6/antagonists & inhibitors , Osteocytes/drug effects , Osteoporosis/drug therapy , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Disease Models, Animal , Fibroblast Growth Factor-23 , Humans , Interleukin-6/immunology , Interleukin-6/metabolism , Molecular Targeted Therapy/methods , Osteoclasts/drug effects , Osteoclasts/immunology , Osteocytes/immunology , Osteocytes/metabolism , Osteogenesis/drug effects , Osteogenesis/immunology , Osteoporosis/chemically induced , Osteoporosis/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
16.
J Cell Biochem ; 103(6): 1671-80, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-17948903

ABSTRACT

The death of osteocytes, the terminally differentiated cells of the osteoblast lineage that are embedded in bone and regulate remodeling, is significant to both normal and pathological bone resorption. Apoptotic osteocytes putatively release a clarion signal that enhances the development of the bone-resorbing osteoclasts and targets their migration to the breach in the osteocyte network. This phenomenon is thought to underlie normal repair of bone microdamage and contribute to the etiologies of inflammatory bone loss. The chromatin protein high mobility group box 1 protein (HMGB1) has been identified as an "alarmin" in other tissues. An alarmin is an endogenous molecule released by dead and dying cells that alert the innate immune system to damage and the need for tissue repair. Wang and colleagues presented evidence in a landmark 1999 study showing that released HMGB1 is a lethal mediator of sepsis. Extracellular HMGB1 is a ligand for the toll-like receptors (TLRs) and for the receptor for advanced glycation end products (RAGE) all of which amplify inflammation. Recent studies by our lab and others have shown that HMGB1 is a bone-active cytokine. It is released by apoptotic osteoblasts in vitro, including the MLO-Y4 osteocyte-like cells. Extracellular HMGB1 enhances the expression of RANKL, TNFalpha, and IL6 in osteoblastogenic bone marrow stromal cell cultures, and it is chemotactic to osteoclasts. In this prospectus we will review HMGB1 activity at the immune-bone interface and propose a role for HMGB1 as an osteocyte alarmin and mediator of normal remodeling and inflammatory bone loss.


Subject(s)
HMGB1 Protein/physiology , Osteocytes/physiology , Animals , Apoptosis/physiology , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Bone Remodeling , Bone Resorption/immunology , Cells, Cultured , Chemotaxis/physiology , Cytokines/biosynthesis , Humans , Immunity, Innate , Mice , Osteoblasts/physiology , Osteoclasts/physiology , Osteocytes/immunology
17.
Mol Med Rep ; 16(6): 9539-9544, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29152655

ABSTRACT

Non-traumatic osteonecrosis of the femoral head (NONFH) is a common clinical osteoarthropathy. The present study aimed to investigate the association between transforming growth factor ß1 (TGF­ß1) and NONFH. Femoral head specimens were collected from patients with NONFH. Patients with traumatic osteonecrosis served as the control. Hematoxylin and eosin (H&E) staining was used to visualize the bone tissue architecture. Immunohistochemistry and densitometry were performed to quantify TGF­ß1 expression in tissues. Flow cytometry was used to detect cluster of differentiation (CD)3+, CD4+ and CD8+ cells, and the ratio of CD4+ to CD8+ T cells in the peripheral blood. H&E staining revealed osteonecrosis, disintegration of osteocytes with karyopyknosis and karyorrhexis, loss of osteocyte lacunae, aberrantly arranged circumferential lamellae, as well as dissolution of the lamellae and subtle osteogenesis in the experimental group, as opposed to the control group. Immunohistochemistry revealed that the expression of TGF­ß1 was significantly reduced in the experimental group (P<0.01). Further, the NONFH group had a decrease in the CD3+ and CD4+ cell populations (P<0.05 and P<0.01, respectively), an increase in the CD8+ cell population (P<0.05), as well as a reduction in the ratio of CD4+ to CD8+ cells (P<0.01). The present study indicated that TGF­ß1 expression was reduced in NONFH. This was associated with impaired repairing capacity of the femoral head and dysregulated subsets of T­lymphocytes and possible immune functions.


Subject(s)
Femur Head Necrosis/genetics , Femur Head/physiopathology , Osteonecrosis/genetics , Transforming Growth Factor beta1/genetics , Adult , Cell Differentiation/genetics , Cell Differentiation/immunology , Female , Femur Head Necrosis/physiopathology , Flow Cytometry , Gene Expression Regulation/genetics , Humans , Male , Middle Aged , Osteocytes/immunology , Osteocytes/pathology , Osteonecrosis/immunology , Osteonecrosis/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transforming Growth Factor beta1/immunology
18.
Int Immunopharmacol ; 52: 150-155, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28910745

ABSTRACT

Immunoglobulin A nephropathy (IgAN) is one of the most common glomerular diseases worldwide. Various studies have identified a host of microRNAs (miRNAs) abnormally expressed in IgAN and might affect the pathogenesis and progression of IgAN. However, miR-200bc/429 cluster in the pathopoiesis of IgAN remains poorly understood. For this study, we found that miR-200bc/429 cluster is downregulated in IgAN tissues and IgAN podocytes and HK2 cells compared with their matched controls respectively. In addition, overexpression of miR-200bc/429 cluster in IgAN podocytes and HK2 cells could attenuate the release of inflammatory cytokines MCP-1, IL-6 and RANTES. Moreover, the 3' untranslated region (UTR) of TNF-like weak inducer of apoptosis (TWEAK) was identified to be a direct target of miR-200bc/429 cluster. Furthermore, our results showed that miR-200bc/429 cluster can inhibit TWEAK mediated NF-κB pathway activation in IgAN. Overall, our findings revealed that miR-200bc/429 cluster alleviates inflammation in IgAN through TWEAK/Fn14 system and might serve as a biomarker as well as a promising therapeutic target for IgAN.


Subject(s)
Cytokine TWEAK/genetics , Glomerulonephritis, IGA/immunology , Inflammation/immunology , MicroRNAs/genetics , Osteocytes/immunology , Aminosalicylic Acids/pharmacology , Animals , Apoptosis , Benzenesulfonates/pharmacology , Butadienes/pharmacology , Cell Line , Cytokine TWEAK/metabolism , Glomerulonephritis, IGA/genetics , Glycation End Products, Advanced/metabolism , Inflammation/genetics , Interleukin-6/metabolism , MAP Kinase Signaling System , Mice , Nitriles/pharmacology , STAT3 Transcription Factor/metabolism , Vascular Endothelial Growth Factor A/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
19.
Ann N Y Acad Sci ; 1364: 11-24, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26662934

ABSTRACT

Osteoimmunology is a field of research dedicated to the study of the interactions between the immune system and bone. Among the cells of the immune system that regulate bone turnover and the responsiveness of bone cells to calciothropic hormones are bone marrow T lymphocytes. T cells secrete osteoclastogenic cytokines such as RANKL and TNF-α, as well as factors that stimulate bone formation, one of which is Wnt10b. In addition, T cells regulate the differentiation and life span of stromal cells (SCs) and their responsiveness to parathyroid hormone (PTH) via costimulatory molecules expressed on their surface. The conditioning effect of T cells on SCs is inherited by the osteoblastic and osteocytic progeny of SCs. As a result, osteoblastic cells of T cell-deficient mice have functional characteristics different from corresponding cells of T cell-replete mice. These differences include the ratio of RANKL/OPG produced in response to continuous PTH treatment, and the osteoblastogenic response to intermittent PTH treatment. This article reviews the evidence indicating that the effects of PTH are mediated not only by osteoblasts and osteocytes but also by T cells.


Subject(s)
Bone Marrow Cells/metabolism , Bone Remodeling , Models, Biological , Osteoblasts/metabolism , Osteocytes/metabolism , Parathyroid Hormone/metabolism , T-Lymphocytes/metabolism , Adaptor Proteins, Signal Transducing , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Bone Morphogenetic Proteins/metabolism , Bone and Bones/cytology , Bone and Bones/immunology , Bone and Bones/metabolism , Cell Communication , Cell Lineage , Cytokines/metabolism , Genetic Markers , Humans , Osteoblasts/cytology , Osteoblasts/immunology , Osteocytes/cytology , Osteocytes/immunology , Parathyroid Hormone/immunology , Proto-Oncogene Proteins/metabolism , Stromal Cells/cytology , Stromal Cells/immunology , Stromal Cells/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Wnt Proteins/metabolism
20.
J Bone Miner Res ; 17(5): 845-53, 2002 May.
Article in English | MEDLINE | ID: mdl-12009015

ABSTRACT

Osteocytes are the most abundant cells in bone; however, relatively little is known about their properties and functions. The development of monoclonal antibody MAb OB7.3 directed against chicken osteocytes enabled us to purify osteocytes from enzymatically isolated bone cells. Cultures of purified osteocytes were used to gain better insight into the role of osteocytes in bone metabolism. Until now, the antigen of MAb OB7.3 has not been elucidated. In this study, we examined the antigen to which this osteocyte-specific antibody is directed. Immunoprecipitation and purification of the protein, followed by amino acid sequence analysis of two isolated peptides, revealed that the antigen has high homology to human and murine PHEX/Phex protein sequences (PHosphate-regulating gene with homology to Endopeptidases on the X chromosome). The OB7.3 antigen was therefore identified as chicken Phex protein. In addition, using suppression subtractive hybridization, we obtained a complementary DNA (cDNA) sequence of 502 base pairs (bp) with high homology to the human and murine PHEX/Phex genes. This method was applied to identify genes, which are differentially expressed in osteocytes compared with osteoblasts. The results also suggest that Phex is expressed at higher levels in chicken osteocytes compared with osteoblasts. Reverse-transcription polymerase chain reaction (RT-PCR) and Northern blot analyses supported these findings. The function of Phex is not completely understood. However, it is known that the gene is preferentially expressed in bone and that mutations in PHEX/Phex lead to X-linked hypophosphatemia and bone mineralization abnormalities. Our findings suggest that osteocytes play an important role in the Phex-regulated phosphate handling in the kidney and in bone.


Subject(s)
Antibodies, Monoclonal , Osteocytes/immunology , Proteins/immunology , Amino Acid Sequence , Animals , Antibody Specificity , Base Sequence , Cells, Cultured , Chick Embryo , DNA, Complementary/genetics , Gene Expression , Humans , Mice , Molecular Sequence Data , Osteocytes/metabolism , PHEX Phosphate Regulating Neutral Endopeptidase , Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL