Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 377
Filtrar
1.
Int Immunopharmacol ; 132: 112000, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38583238

RESUMO

Various studies have been investigated the phenotypic and functional distinctions of craniofacial and long bone cells involved in bone regeneration. However, the process of bone tissue regeneration after bone grafting involves complicated interactions between different cell types at the donor-recipient site. Additionally, differences in alterations of the immune microenvironment at the recipient site remained to be explored. Osteoblasts (OBs) and macrophages (MØ) play essential roles in the bone restoration and regeneration processes in the bone and immune systems, respectively. The modulation of MØ on OBs has been extensively explored in the literature, whereas limited research has been conducted on the influence of OBs on the MØ phenotype and function. In the present study, OBs from the mandible and femur (MOBs and FOBs, respectively) promoted cranial defect regeneration in rats, with better outcomes noted in the MOBs-treated group. After MOBs transplantation, a significant inflammatory response was induced, accompanied by an early increase in IL-10 secretion. And then, there was an upregulation in M2-MØ-related cell markers and inflammatory factor expression. Condition media (CM) of OBs mildly inhibited apoptosis in MØ, enhanced their migration and phagocytic functions, and concurrently increased iNOS and Arg1 expression, with MOB-CM demonstrating more pronounced effects compared to FOB-CM. In conclusion, our investigation showed that MOBs and FOBs have the ability to modulate MØ phenotype and function, with MOBs exhibiting a stronger regulatory potential. These findings provide a new direction for improving therapeutic strategies for bone regeneration in autologous bone grafts from the perspective of the immune microenvironment.


Assuntos
Regeneração Óssea , Fêmur , Macrófagos , Mandíbula , Osteoblastos , Fenótipo , Animais , Osteoblastos/metabolismo , Osteoblastos/imunologia , Macrófagos/imunologia , Mandíbula/cirurgia , Masculino , Ratos , Células Cultivadas , Ratos Sprague-Dawley , Apoptose , Fagocitose , Transplante Ósseo
2.
Front Immunol ; 15: 1383113, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38646530

RESUMO

It is well established that inflammatory processes in the vicinity of bone often induce osteoclast formation and bone resorption. Effects of inflammatory processes on bone formation are less studied. Therefore, we investigated the effect of locally induced inflammation on bone formation. Toll-like receptor (TLR) 2 agonists LPS from Porphyromonas gingivalis and PAM2 were injected once subcutaneously above mouse calvarial bones. After five days, both agonists induced bone formation mainly at endocranial surfaces. The injection resulted in progressively increased calvarial thickness during 21 days. Excessive new bone formation was mainly observed separated from bone resorption cavities. Anti-RANKL did not affect the increase of bone formation. Inflammation caused increased bone formation rate due to increased mineralizing surfaces as assessed by dynamic histomorphometry. In areas close to new bone formation, an abundance of proliferating cells was observed as well as cells robustly stained for Runx2 and alkaline phosphatase. PAM2 increased the mRNA expression of Lrp5, Lrp6 and Wnt7b, and decreased the expression of Sost and Dkk1. In situ hybridization demonstrated decreased Sost mRNA expression in osteocytes present in old bone. An abundance of cells expressed Wnt7b in Runx2-positive osteoblasts and ß-catenin in areas with new bone formation. These data demonstrate that inflammation, not only induces osteoclastogenesis, but also locally activates canonical WNT signaling and stimulates new bone formation independent on bone resorption.


Assuntos
Inflamação , Osteogênese , Receptor 2 Toll-Like , Via de Sinalização Wnt , Animais , Masculino , Camundongos , Proteínas Adaptadoras de Transdução de Sinal , Inflamação/induzido quimicamente , Inflamação/metabolismo , Lipopolissacarídeos , Camundongos Endogâmicos C57BL , Osteoblastos/metabolismo , Osteoblastos/imunologia , Osteócitos/efeitos dos fármacos , Osteócitos/metabolismo , Osteogênese/efeitos dos fármacos , Crânio , Receptor 2 Toll-Like/agonistas , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo , Proteínas Wnt/metabolismo
3.
Nat Commun ; 13(1): 160, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-35013289

RESUMO

Immune response and new tissue formation are important aspects of tissue repair. However, only a single aspect is generally considered in previous biomedical interventions, and the synergistic effect is unclear. Here, a dual-effect coating with immobilized immunomodulatory metal ions (e.g., Zn2+) and osteoinductive growth factors (e.g., BMP-2 peptide) is designed via mussel adhesion-mediated ion coordination and molecular clicking strategy. Compared to the bare TiO2 group, Zn2+ can increase M2 macrophage recruitment by up to 92.5% in vivo and upregulate the expression of M2 cytokine IL-10 by 84.5%; while the dual-effect of Zn2+ and BMP-2 peptide can increase M2 macrophages recruitment by up to 124.7% in vivo and upregulate the expression of M2 cytokine IL-10 by 171%. These benefits eventually significantly enhance bone-implant mechanical fixation (203.3 N) and new bone ingrowth (82.1%) compared to the bare TiO2 (98.6 N and 45.1%, respectively). Taken together, the dual-effect coating can be utilized to synergistically modulate the osteoimmune microenvironment at the bone-implant interface, enhancing bone regeneration for successful implantation.


Assuntos
Proteína Morfogenética Óssea 2/farmacologia , Interface Osso-Implante/crescimento & desenvolvimento , Macrófagos/efeitos dos fármacos , Titânio/farmacologia , Zinco/farmacologia , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Biomarcadores/metabolismo , Bivalves/química , Diferenciação Celular/efeitos dos fármacos , Fêmur/citologia , Fêmur/imunologia , Interleucina-10/genética , Interleucina-10/imunologia , Lipopolissacarídeos/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/citologia , Macrófagos/imunologia , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/imunologia , Camundongos , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/imunologia , Osteogênese/efeitos dos fármacos , Próteses e Implantes , Precursores de Proteínas/farmacologia , Células RAW 264.7 , Ratos , Ratos Sprague-Dawley
4.
Actual. osteol ; 18(1): 40-52, 2022. ilus, tab
Artigo em Espanhol | LILACS, UNISALUD, BINACIS | ID: biblio-1396075

RESUMO

El "microbioma" no solo está constituido por los microbios, sino por todos los componen-tes que viven en el mismo hábitat conforman-do un nicho ecológico. Es decir, está conformado por los microorganismos (bacterias, hongos, protozoos, etc.), todo el espectro de moléculas producidas por ellos tales como sus componentes estructurales (ácidos nucleicos, proteínas, lípidos y glúcidos), meta-bolitos, toxinas, etc., y las moléculas producidas por el huésped. El microbioma intestinal (MI) ha emergido como un factor que tiene un gran efecto sobre la cantidad, calidad y fuerza del hueso. Las investigaciones revelan que la homeostasis ósea está ligada al micro-bioma saludable, mientras que la disbiosis (alteración en la biodiversidad microbiana) puede exacerbar la actividad osteoclástica y promover la osteoporosis. Los mecanismos potenciales involucrados en la interacción del microbioma intestinal y el hueso son la influencia del metabolismo del huésped, el mantenimiento de la integridad intestinal y regulación de la absorción de nutrientes, la regulación del eje intestino-sistema inmune y la modulación del sistema endocrino. Es decir que hay múltiples vías por las cuales el MI influye sobre el hueso, pero estos y otros mecanismos deben profundizarse más aún. También es necesario que se identifiquen y caractericen mejor los microorganismos que están asociados a las enfermedades óseas. El conocimiento de estos aspectos podría ser útil para el desarrollo de herramientas terapéuticas basadas en el MI que puedan mejorar la eficacia de los distintos tratamientos existentes. (AU)


The microbiome is not only constituted by microbes, but by all the components that live in the same habitat forming an ecological niche. It is conformed by the microorganisms ( bacteria, fungi, protozoa, etc), the entire spectrum of molecules produced by them (nucleic acids, proteins, lipid and carbohydrates, metabolites, toxins, etc) and the molecules produced by the host. The intestinal microbiome (IM) has emerged as a factor with great effects on the quantity, quality and strength of bone. The investigations reveal that bone homeostasis is linked to the healthy microbiome, while the dysbiosis (alteration in the microbial biodiversity) can exacerbate the osteoclastic activity and promote osteoporosis. The potential mechanisms involved in the interaction between IM and bone are the influence of the host metabolism, the maintenance of the intestinal integrity and regulation of the nutrient absorption, the regulation of the intestine/ immune system axis and the modulation of the endocrine system. That is, there are multiple ways through which IM influences on bone, but these and other mechanisms need to be further studied. It is also necessary to identify and characterize the microorganisms associated with the bone diseases. Knowledge of these aspects could be useful to develop therapeutical tools based on the IM that could improve the efficacy of the current treatments. (AU)


Assuntos
Humanos , Osteoblastos/imunologia , Osteoclastos/imunologia , Osso e Ossos/imunologia , Disbiose/complicações , Microbioma Gastrointestinal/imunologia , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osso e Ossos/metabolismo , Intestinos/imunologia , Intestinos/microbiologia
5.
Int Immunopharmacol ; 101(Pt A): 108179, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34601329

RESUMO

The term "osteoimmunology" was coined to denote the bridge between the immune system and the skeletal system. Osteoimmunology is interdisciplinary, and a full understanding and development of this "bridge" will provide an in-depth understanding of the switch between body health and disease development. B lymphocytes can promote the maturation and differentiation of osteoclasts, and osteoclasts have a negative feedback effect on B lymphocytes. Different subtypes of T lymphocytes regulate osteoclasts in different directions. T lymphocytes have a two-way regulatory effect on osteoblasts, while B lymphocytes have minimal regulatory effects on osteoblasts. In contrast, osteoblasts can promote the differentiation and maturation of T lymphocytes and B lymphocytes. Different immune cells have different effects on chondrocytes; some cooperate with each other, while some antagonize each other. In a healthy adult body, bone resorption and bone formation are in a dynamic balance under the action of multiple mechanisms. In this review, we summarize the interactions and key signaling molecular mechanisms between each type of cell in the immune system and the skeletal system.


Assuntos
Comunicação Celular/imunologia , Osteoartrite/imunologia , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Diferenciação Celular/imunologia , Condrócitos/imunologia , Condrócitos/patologia , Modelos Animais de Doenças , Humanos , Células-Tronco Mesenquimais/fisiologia , Osteoartrite/patologia , Osteoblastos/imunologia , Osteoblastos/metabolismo , Osteoclastos/imunologia , Osteoclastos/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo
6.
J Cell Mol Med ; 25(14): 6634-6651, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34075722

RESUMO

Available therapies aimed at treating age-related osteoporosis are still insufficient. Therefore, designing reliable in vitro model for the analysis of molecular mechanisms underlying senile osteoporosis is highly required. We have isolated and characterized progenitor cells isolated from bone marrow (BMSCs) of osteoporotic mice strain SAM/P6 (BMSCSAM/P6 ). The cytophysiology of BMSCSAM/P6 was for the first time compared with BMSCs isolated from healthy BALB/c mice (BMSCBALB/c ). Characterization of the cells included evaluation of their multipotency, morphology and determination of specific phenotype. Viability of BMSCs cultures was determined in reference to apoptosis profile, metabolic activity, oxidative stress, mitochondrial membrane potential and caspase activation. Additionally, expression of relevant biomarkers was determined with RT-qPCR. Obtained results indicated that BMSCSAM/P6 and BMSCBALB/c show the typical phenotype of mesenchymal stromal cells (CD44+, CD73+, CD90+) and do not express CD45. Further, BMSCSAM/P6 were characterized by deteriorated multipotency, decreased metabolic activity and increased apoptosis occurrence, accompanied by elevated oxidative stress and mitochondria depolarisation. The transcriptome analyses showed that BMSCSAM/P6 are distinguished by lowered expression of molecules crucial for proper osteogenesis, including Coll-1, Opg and Opn. However, the expression of Trap, DANCR1 and miR-124-3p was significantly up-regulated. Obtained results show that BMSCSAM/P6 present features of progenitor cells with disturbed metabolism and could serve as appropriate model for in vitro investigation of age-dependent osteoporosis.


Assuntos
Diferenciação Celular/genética , Células-Tronco Mesenquimais/imunologia , Osteogênese/genética , Osteoporose/genética , 5'-Nucleotidase/genética , 5'-Nucleotidase/imunologia , Animais , Diferenciação Celular/imunologia , Humanos , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/imunologia , Antígenos Comuns de Leucócito/genética , Antígenos Comuns de Leucócito/imunologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Osteoblastos/imunologia , Osteoblastos/metabolismo , Osteogênese/imunologia , Osteoporose/imunologia , Osteoporose/patologia , Células-Tronco/imunologia , Células-Tronco/metabolismo , Antígenos Thy-1/genética , Antígenos Thy-1/imunologia
7.
Cell Reprogram ; 23(2): 108-116, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33861637

RESUMO

In our previous study, we constructed Schwann cells (SCs) that stably express Simian virus 40 T antigen (SV40T-SCs). SV40T-SCs functions and markers are similar to those of neural crest cells. There we used bone morphogenetic protein 9 (BMP9) to induce SV40T-SCs differentiation in vitro and in vivo and study possible related mechanism. SV40T-SCs differentiation was induced by BMP9 conditioned medium. The lipogenic differentiation of SV40T-SCs was assessed by Oil Red O staining. Alizarin red and Alcian blue staining, and alkaline phosphatase (ALP) assays were used to evaluate the SV40T-SCs osteogenic differentiation. The expression of adipocyte differentiation (c/EBPα and c/EBPß) and osteoblast differentiation markers (OSX and RUNX2) were detected by quantitative polymerase chain reaction (qPCR). To study possible mechanism related to SV40T-SCs differentiation, the P53 and E2F1 activity were assessed by luciferase reporter plasmid, and Slug and E-cadherin expression by qPCR. In vivo, SV40T-SCs infected by Ad-BMP9 or Ad-GFP were injected under the skin of nude mice. After 4-6 W, the mice were euthanized and subcutaneously mass formed at injecting sites was collected for pathological analysis. After SV40T-SCs were cultured in BMP9 conditioned medium, lipid droplets were formed in the cytoplasm of these cells. Alizarin red and Alcian blue staining were positive, and ALP activity of SV40T-SCs increased significantly. The expression of adipocyte differentiation (c/EBPα and c/EBPß) and osteoblast differentiation markers (OSX and RUNX2) in SV40T-SCs was upregulated by BMP9. SV40T significantly increased Slug expression and decreased E-cadherin expression. SV40T-SCs infected with Ad-BMP9 were able to differentiate into adipose tissue and form a small bone matrix under the nude mice skin. SV40T-SCs have the ability to differentiate into adipocytes and osteoblasts in vivo and in vitro. SV40T can upregulate the Slug expression and downregulate the E-cadherin expression to produce endothelial-to-mesenchymal transition (EMT). The multidirectional differentiation ability of SV40T-SCs may be related to EMT.


Assuntos
Adipócitos/citologia , Antígenos Virais de Tumores/imunologia , Fator 2 de Diferenciação de Crescimento/metabolismo , Osteoblastos/citologia , Osteogênese , Células de Schwann/citologia , Vírus 40 dos Símios/imunologia , Adipócitos/imunologia , Adipócitos/metabolismo , Animais , Antígenos Virais de Tumores/metabolismo , Técnicas In Vitro , Masculino , Camundongos , Camundongos Nus , Osteoblastos/imunologia , Osteoblastos/metabolismo , Células de Schwann/imunologia , Células de Schwann/metabolismo , Vírus 40 dos Símios/metabolismo
8.
Int J Mol Sci ; 22(6)2021 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-33805598

RESUMO

Metastasis to the bone is a common feature of many cancers including those of the breast, prostate, lung, thyroid and kidney. Once tumors metastasize to the bone, they are essentially incurable. Bone metastasis is a complex process involving not only intravasation of tumor cells from the primary tumor into circulation, but extravasation from circulation into the bone where they meet an environment that is generally suppressive of their growth. The bone microenvironment can inhibit the growth of disseminated tumor cells (DTC) by inducing dormancy of the DTC directly and later on following formation of a micrometastatic tumour mass by inhibiting metastatic processes including angiogenesis, bone remodeling and immunosuppressive cell functions. In this review we will highlight some of the mechanisms mediating DTC dormancy and the complex relationships which occur between tumor cells and bone resident cells in the bone metastatic microenvironment. These inter-cellular interactions may be important targets to consider for development of novel effective therapies for the prevention or treatment of bone metastases.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Ósseas/prevenção & controle , Regulação Neoplásica da Expressão Gênica , Neovascularização Patológica/prevenção & controle , Evasão Tumoral/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos , Antineoplásicos/uso terapêutico , Neoplasias Ósseas/genética , Neoplasias Ósseas/imunologia , Neoplasias Ósseas/patologia , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/imunologia , Osso e Ossos/patologia , Comunicação Celular , Citocinas/genética , Citocinas/metabolismo , Humanos , Metástase Linfática , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/patologia , Células Supressoras Mieloides/efeitos dos fármacos , Células Supressoras Mieloides/imunologia , Células Supressoras Mieloides/patologia , Neovascularização Patológica/genética , Neovascularização Patológica/imunologia , Neovascularização Patológica/patologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/imunologia , Osteoblastos/patologia , Osteoclastos/efeitos dos fármacos , Osteoclastos/imunologia , Osteoclastos/patologia , Transdução de Sinais , Evasão Tumoral/genética , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia , Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fatores de Crescimento do Endotélio Vascular/genética , Fatores de Crescimento do Endotélio Vascular/metabolismo
9.
Exp Cell Res ; 400(2): 112505, 2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-33516666

RESUMO

Inflammation and alveolar bone destruction constitute the main pathological process of periodontitis. However, the molecular mechanisms of bone destruction under the inflammation environment remain unclear. This study aims to explore the role of Ephrin-B2/EphB4 signaling in osteogenic differentiation under the inflammation environment. Mouse pre-osteoblasts MC3T3-E1 were pretreated with lipopolysaccharide of Porphyromonas gingivalis (Pg-LPS). The Ephrin-B2/EphB4 signaling was activated, and the osteogenic differentiation of cells was examined. The results showed that activation of Ephrin-B2/EphB4 signaling promoted the expression levels of osteogenic differentiation-related genes, and also relieved the inhibitory effect of Pg-LPS on osteogenesis. Noticeably, the effect of Ephrin-B2/EphB4 signaling might be related to the mitogen-activated protein kinase (MAPK) pathway. While applying Ephrin-B2-Fc and EphB4-Fc to periodontitis mice, we observed the reduction of alveolar crest destruction. The current study revealed the possible role of Ephrin-B2/EphB4 signaling in reducing bone destruction in periodontitis and suggested its potential values for further research.


Assuntos
Efrina-B2/metabolismo , Fragmentos Fc das Imunoglobulinas/imunologia , Inflamação/prevenção & controle , Osteoblastos/citologia , Osteogênese , Periodontite/prevenção & controle , Receptor EphB4/metabolismo , Animais , Diferenciação Celular , Efrina-B2/genética , Efrina-B2/imunologia , Inflamação/imunologia , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos/imunologia , Osteoblastos/metabolismo , Periodontite/imunologia , Periodontite/metabolismo , Receptor EphB4/genética , Receptor EphB4/imunologia , Transdução de Sinais
10.
Cytokine ; 137: 155342, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33130337

RESUMO

BACKGROUND: The developing field of osteoimmunology supports importance of an interferon (IFN) response pathway in osteoblasts. Clarifying osteoblast-IFN interactions is important because IFN is used as salvage anti-tumor therapy but systemic toxicity is high with variable clinical results. In addition, osteoblast response to systemic bursts and disruptions of IFN pathways induced by viral infection may influence bone remodeling. ZIKA virus (ZIKV) infection impacts bone development in humans and IFN response in vitro. Consistently, initial evidence of permissivity to ZIKV has been reported in human osteoblasts. HYPOTHESIS: Osteoblast-like Saos-2 cells are permissive to ZIKV and responsive to IFN. METHODS: Multiple approaches were used to assess whether Saos-2 cells are permissive to ZIKV infection and exhibit IFN-mediated ZIKV suppression. Proteomic methods were used to evaluate impact of ZIKV and IFN on Saos-2 cells. RESULTS: Evidence is presented confirming Saos-2 cells are permissive to ZIKV and support IFN-mediated suppression of ZIKV. ZIKV and IFN differentially impact the Saos-2 proteome, exemplified by HELZ2 protein which is upregulated by IFN but non responsive to ZIKV. Both ZIKV and IFN suppress proteins associated with microcephaly/pseudo-TORCH syndrome (BI1, KI20A and UBP18), and ZIKV induces potential entry factor PLVAP. CONCLUSIONS: Transient ZIKV infection influences osteoimmune state, and IFN and ZIKV activate distinct proteomes in Saos-2 cells, which could inform therapeutic, engineered, disruptions.


Assuntos
Antivirais/imunologia , Interferon Tipo I/imunologia , Osteoblastos/imunologia , Infecção por Zika virus/imunologia , Zika virus/imunologia , Animais , Antivirais/farmacologia , Linhagem Celular Tumoral , Chlorocebus aethiops , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Interações Hospedeiro-Patógeno/imunologia , Humanos , Interferon Tipo I/farmacologia , Camundongos Knockout , Osteoblastos/metabolismo , Osteoblastos/virologia , Proteoma/imunologia , Proteoma/metabolismo , Proteômica/métodos , Células Vero , Replicação Viral/efeitos dos fármacos , Replicação Viral/imunologia , Zika virus/fisiologia , Infecção por Zika virus/metabolismo , Infecção por Zika virus/virologia
11.
Acta Histochem ; 122(8): 151636, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33132168

RESUMO

INTRODUCTION: Mesenchymal stem cells (MSCs) are characterized by the potential to differentiate into multiple cell lineages, high proliferation rates, and self-renewal capacity, in addition to the ability to maintain their undifferentiated state. These cells have been identified in physiological oral tissues such as pulp tissue, dental follicle, apical papilla and periodontal ligament, as well as in pathological situations such as chronic periapical lesions (CPLs). The criteria used for the identification of MSCs include the positive expression of specific surface antigens, with CD73, CD90, CD105, CD44, CD146, STRO-1, CD166, NANOG and OCT4 being the most specific for these cells. AIM: The aim of this review was to explore the literature on markers able to identify MSCs as well as the presence of these cells in the healthy periodontal ligament and CPLs, highlighting their role in regenerative medicine and implications in the progression of these lesions. METHODS: Narrative literature review searching the PubMed and Medline databases. Articles published in English between 1974 and 2020 were retrieved. CONCLUSION: The included studies confirmed the presence of MSCs in the healthy periodontal ligament and in CPLs. Several surface markers are used for the characterization of these cells which, although not specific, are effective in cell recognition. Mesenchymal stem cells participate in tissue repair, exerting anti- inflammatory, immunosuppressive and proangiogenic effects, and are therefore involved in the progression and attenuation of CPLs or even in the persistence of these lesions.


Assuntos
Células-Tronco Mesenquimais/citologia , Doenças Periapicais/patologia , Ligamento Periodontal/citologia , Endodontia Regenerativa/métodos , Adipócitos/citologia , Adipócitos/imunologia , Antígenos CD/genética , Antígenos CD/imunologia , Antígenos de Superfície/genética , Antígenos de Superfície/imunologia , Biomarcadores/metabolismo , Diferenciação Celular , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Condrócitos/citologia , Condrócitos/imunologia , Polpa Dentária/citologia , Polpa Dentária/imunologia , Expressão Gênica , Humanos , Células-Tronco Mesenquimais/imunologia , Proteína Homeobox Nanog/genética , Proteína Homeobox Nanog/imunologia , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/imunologia , Osteoblastos/citologia , Osteoblastos/imunologia , Osteogênese/genética , Osteogênese/imunologia , Doenças Periapicais/genética , Doenças Periapicais/imunologia , Doenças Periapicais/terapia , Ligamento Periodontal/imunologia
12.
Biosci Trends ; 14(5): 342-348, 2020 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-32908076

RESUMO

The human immune system has evolved to recognize and eradicate pathogens, a process that is known as "host defense". If, however, the immune system does not work properly, it can mistakenly attack the body's own tissues and induce autoimmune diseases. Rheumatoid arthritis (RA) is such an autoimmune disease in which the synovial joints are predominately attacked by the immune system. Moreover, RA is associated with bone destruction and joint deformity. Although biologic agents have propelled RA treatment forward dramatically over the past 30 years, a considerable number of patients with RA still experience progressive bone damage and joint disability. That is to be expected since current RA therapies are all intended to halt inflammation but not to alleviate bone destruction. A better understanding of bone erosions is crucial to developing a novel strategy to treat RA-associated erosions. This review provides insights into RA-associated bone destruction and perspectives for future clinical interventions.


Assuntos
Artrite Reumatoide/complicações , Fatores Biológicos/farmacologia , Conservadores da Densidade Óssea/farmacologia , Osteoporose/imunologia , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/imunologia , Artrite Reumatoide/patologia , Fatores Biológicos/uso terapêutico , Conservadores da Densidade Óssea/uso terapêutico , Caderinas/farmacologia , Caderinas/uso terapêutico , Humanos , Cápsula Articular/efeitos dos fármacos , Cápsula Articular/imunologia , Cápsula Articular/patologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/imunologia , Osteoclastos/efeitos dos fármacos , Osteoclastos/imunologia , Osteogênese/efeitos dos fármacos , Osteogênese/imunologia , Osteoporose/tratamento farmacológico , Osteoporose/patologia , Proteínas/antagonistas & inibidores , Proteínas/metabolismo , Ligante RANK/antagonistas & inibidores , Ligante RANK/imunologia , Ligante RANK/metabolismo , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Líquido Sinovial/efeitos dos fármacos , Líquido Sinovial/imunologia
13.
J Mol Histol ; 51(3): 221-232, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32356234

RESUMO

Periodontitis is characterized by alveolar bone destruction and is one of the most common chronic oral diseases. Inflammatory cytokines released by pyroptosis, which can be triggered by oxidative stress, are critical in the development of periodontitis. This study aims to clarify whether oxidative stress causes osteoblast dysfunction by inducing pyroptosis in the process of periodontitis. We found that treatment with lipopolysaccharide (LPS) led to NLRP3 inflammasome-mediated pyroptosis of MG63 cells as well as decreased cell migration. Of note, LPS stimulation increased LDH release in a time- and dose-dependent manner. However, inhibition of reactive oxygen species with N-acetyl-L-cysteine attenuated oxidative stress-mediated pyroptosis and improved migration injury in osteoblasts treated with LPS. Further, inhibition of the NLRP3 inflammasome with MCC950 improved osteoblast migration and restored the expression of osteogenic differentiation-related proteins such as COL 1, RUNX 2 and ALP. In conclusion, oxidative stress caused by LPS induces pyroptosis in osteoblasts, leading to osteogenic dysfunction.


Assuntos
Inflamassomos/metabolismo , Osteoblastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Periodontite/metabolismo , Piroptose/efeitos dos fármacos , Antioxidantes/metabolismo , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Humanos , Lipopolissacarídeos/farmacologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Osteoblastos/imunologia , Osteoblastos/metabolismo , Osteoblastos/patologia , Osteogênese/imunologia , Estresse Oxidativo/imunologia , Periodontite/imunologia , Periodontite/patologia , Piroptose/imunologia , Espécies Reativas de Oxigênio/metabolismo
14.
Mater Sci Eng C Mater Biol Appl ; 110: 110722, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32204034

RESUMO

It is known that introducing a porous ceramic coating on titanium (Ti) implant surface fabricated by micro-arc oxidation (MAO) could enhance the differentiation of osteoblasts. However, the osteogenic capacity of MAO-fabricated coating still remains unknown when immune cells especially macrophages are involved. The influence of the inflammatory microenvironment and the co-influence of the inflammatory microenvironment and surface characteristics of MAO-fabricated coating on osteoblast response need to be explored. In this study, a new in vitro cell culture strategy is proposed by mimicking the biological events happened after implantation based on the recruitment of osteoblasts to biomaterial surfaces to investigate biological performances of MAO-modified Ti surface. It is found that macrophages grown on MAO-modified Ti surface were switched to M1-like phenotype, evidenced by the promoted expressions of inflammatory genes (tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and IL-1ß) and production of pro-inflammatory cytokine TNF-α. Moreover, the inflammatory microenvironment created by macrophage/MAO-modified Ti surface interactions could promote the collagen syntheses and matrix mineralization of osteoblast-like cells grown tissue culture plate. When osteoblasts were cultured on MAO-modified Ti surface and cultured by macrophage/MAO-modified Ti surface conditioned medium (CM), the alkaline phosphatase (ALP) activity and collagen synthesis of osteoblast-like cells were promoted. This study suggests that MAO-modified Ti surface is beneficial for osteogenesis at both stages after implantation (before and after osteoblast recruitment to biomaterial surfaces).


Assuntos
Técnicas de Cultura de Células , Materiais Revestidos Biocompatíveis , Macrófagos/imunologia , Osteoblastos/imunologia , Osteogênese , Titânio , Animais , Linhagem Celular Tumoral , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/farmacologia , Citocinas/imunologia , Avaliação de Medicamentos , Humanos , Macrófagos/citologia , Camundongos , Osteoblastos/citologia , Osteogênese/efeitos dos fármacos , Osteogênese/imunologia , Oxirredução , Células RAW 264.7 , Propriedades de Superfície , Titânio/química , Titânio/farmacologia
15.
J Clin Invest ; 130(6): 2888-2902, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32078586

RESUMO

Given the numerous health benefits of exercise, understanding how exercise capacity is regulated is a question of paramount importance. Circulating interleukin 6 (IL-6) levels surge during exercise and IL-6 favors exercise capacity. However, neither the cellular origin of circulating IL-6 during exercise nor the means by which this cytokine enhances exercise capacity has been formally established yet. Here we show through genetic means that the majority of circulating IL-6 detectable during exercise originates from muscle and that to increase exercise capacity, IL-6 must signal in osteoblasts to favor osteoclast differentiation and the release of bioactive osteocalcin in the general circulation. This explains why mice lacking the IL-6 receptor only in osteoblasts exhibit a deficit in exercise capacity of similar severity to the one seen in mice lacking muscle-derived IL-6 (mIL-6), and why this deficit is correctable by osteocalcin but not by IL-6. Furthermore, in agreement with the notion that IL-6 acts through osteocalcin, we demonstrate that mIL-6 promotes nutrient uptake and catabolism into myofibers during exercise in an osteocalcin-dependent manner. Finally, we show that the crosstalk between osteocalcin and IL-6 is conserved between rodents and humans. This study provides evidence that a muscle-bone-muscle endocrine axis is necessary to increase muscle function during exercise in rodents and humans.


Assuntos
Interleucina-6/imunologia , Músculo Esquelético/imunologia , Osteoblastos/imunologia , Transdução de Sinais/imunologia , Animais , Feminino , Interleucina-6/genética , Macaca mulatta , Camundongos , Camundongos Knockout , Transdução de Sinais/genética
16.
Curr Osteoporos Rep ; 18(1): 23-31, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32002770

RESUMO

PURPOSE OF REVIEW: Diabetes has a detrimental effect on bone, increasing the risk of fracture and formation of osteolytic lesions such as those seen in periodontitis. Several diabetic complications are caused by diabetes-enhanced inflammation. This review examines mechanisms by which IL-17 contributes to diabetes-enhanced periodontitis and other effects of IL-17 on bone. RECENT FINDINGS: IL-17 upregulates anti-bacterial defenses, yet its expression is also linked to a destructive host response in the periodontium. Periodontal disease is caused by bacteria that stimulate an inflammatory response. Diabetes-enhanced IL-17 increases gingival inflammation, which alters the composition of the oral microbiota to increase its pathogenicity. In addition, IL-17 can induce osteoclastogenesis by upregulation of TNF and RANKL in a number of cell types, and IL-17 has differential effects on osteoblasts and their progenitors. Increased IL-17 production caused by diabetes alters the pathogenicity of the oral microbiota and can promote periodontal bone resorption.


Assuntos
Perda do Osso Alveolar/imunologia , Diabetes Mellitus/imunologia , Interleucina-17/imunologia , Microbiota/imunologia , Periodontite/imunologia , Reabsorção Óssea/imunologia , Humanos , Inflamação/imunologia , Boca/microbiologia , Osteoblastos/imunologia , Osteoclastos/imunologia
17.
Cancer Res ; 80(5): 1036-1048, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31911552

RESUMO

Multiple myeloma is a plasma cell malignancy that thrives in the bone marrow (BM), with frequent progression to new local and distant bone sites. Our previous studies demonstrated that multiple myeloma cells at primary sites secrete soluble factors and suppress osteoblastogenesis via the inhibition of Runt-related transcription factor 2 (Runx2) in pre- and immature osteoblasts (OB) in new bone sites, prior to the arrival of metastatic tumor cells. However, it is unknown whether OB-Runx2 suppression in new bone sites feeds back to promote multiple myeloma dissemination to and progression in these areas. Hence, we developed a syngeneic mouse model of multiple myeloma in which Runx2 is specifically deleted in the immature OBs of C57BL6/KaLwRij mice (OB-Runx2-/- mice) to study the effect of OB-Runx2 deficiency on multiple myeloma progression in new bone sites. In vivo studies with this model demonstrated that OB-Runx2 deficiency attracts multiple myeloma cells and promotes multiple myeloma tumor growth in bone. Mechanistic studies further revealed that OB-Runx2 deficiency induces an immunosuppressive microenvironment in BM that is marked by an increase in the concentration and activation of myeloid-derived suppressor cells (MDSC) and the suppression and exhaustion of cytotoxic CD8+ T cells. In contrast, MDSC depletion by either gemcitabine or 5-fluorouracil treatment in OB-Runx2-/- mice prevented these effects and inhibited multiple myeloma tumor growth in BM. These novel discoveries demonstrate that OB-Runx2 deficiency in new bone sites promotes multiple myeloma dissemination and progression by increasing metastatic cytokines and MDSCs in BM and inhibiting BM immunity. Importantly, MDSC depletion can block multiple myeloma progression promoted by OB-Runx2 deficiency.Significance: This study demonstrates that Runx2 deficiency in immature osteoblasts at distant bone sites attracts myeloma cells and allows myeloma progression in new bone sites via OB-secreted metastatic cytokines and MDSC-mediated suppression of bone marrow immunity.


Assuntos
Neoplasias Ósseas/secundário , Subunidade alfa 1 de Fator de Ligação ao Core/deficiência , Mieloma Múltiplo/patologia , Osteoblastos/patologia , Microambiente Tumoral/imunologia , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/imunologia , Medula Óssea/patologia , Neoplasias Ósseas/imunologia , Osso e Ossos/citologia , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/imunologia , Osso e Ossos/patologia , Linhagem Celular Tumoral/transplante , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Modelos Animais de Doenças , Feminino , Fluoruracila , Humanos , Masculino , Camundongos , Camundongos Knockout , Mieloma Múltiplo/imunologia , Células Supressoras Mieloides/efeitos dos fármacos , Células Supressoras Mieloides/imunologia , Células Supressoras Mieloides/patologia , Osteoblastos/imunologia , Gencitabina
18.
Curr Osteoporos Rep ; 18(1): 32-37, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31970653

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Adipogenia/imunologia , Hematopoese Extramedular/imunologia , Linfopoese/imunologia , Animais , Linfócitos B , Medula Óssea/imunologia , Citocinas/imunologia , Hematopoese/imunologia , Células-Tronco Hematopoéticas , Humanos , Células-Tronco Mesenquimais/imunologia , Camundongos , Camundongos Knockout , Mielopoese/imunologia , Osteoblastos/imunologia , Osteócitos/imunologia
19.
Int J Mol Sci ; 22(1)2020 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-33383915

RESUMO

The interaction of hematopoietic cells and the bone microenvironment to maintain bone homeostasis is increasingly appreciated. We hypothesized that the transfer of allogeneic T lymphocytes has extensive effects on bone biology and investigated trabecular and cortical bone structures, the osteoblast reconstitution, and the bone vasculature in experimental hematopoietic stem cell transplantations (HSCT). Allogeneic or syngeneic hematopoietic stem cells (HSC) and allogeneic T lymphocytes were isolated and transferred in a murine model. After 20, 40, and 60 days, bone structures were visualized using microCT and histology. Immune cells were monitored using flow cytometry and bone vessels, bone cells and immune cells were fluorescently stained and visualized. Remodeling of the bone substance, the bone vasculature and bone cell subsets were found to occur as early as day +20 after allogeneic HSCT (including allogeneic T lymphocytes) but not after syngeneic HSCT. We discovered that allogeneic HSCT (including allogeneic T lymphocytes) results in a transient increase of trabecular bone number and bone vessel density. This was paralleled by a cortical thinning as well as disruptive osteoblast lining and loss of B lymphocytes. In summary, our data demonstrate that the adoptive transfer of allogeneic HSCs and allogeneic T lymphocytes can induce profound structural and spatial changes of bone tissue homeostasis as well as bone marrow cell composition, underlining the importance of the adaptive immune system for maintaining a balanced bone biology.


Assuntos
Células da Medula Óssea/metabolismo , Remodelação Óssea , Animais , Biomarcadores , Medula Óssea/metabolismo , Medula Óssea/patologia , Diáfises , Doença Enxerto-Hospedeiro/etiologia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Transplante de Células-Tronco Hematopoéticas/métodos , Imunofenotipagem , Camundongos , Osteoblastos/imunologia , Osteoblastos/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Quimeras de Transplante , Transplante Homólogo
20.
Curr Drug Targets ; 21(3): 213-227, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31433756

RESUMO

Osteoimmunology is a new subject which focuses on the communication between the immune and the skeletal systems. Both the immune system and bone communicate with each other. Proinflammatory cytokines, such as tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) play important roles in immune responses and bone metabolism. TNF-α and IL-6 enhance macrophage activation and antigen presentation, as well as regulating immunity through different mechanisms. A variety of groups have reported that TNF-α suppresses osteoblasts activity at some stages of differentiation and stimulates osteoclast proliferation and differentiation. In contrast, IL-6 mediates the actions of osteoblasts and osteoclasts through sophisticated mechanisms, which reflect dual effects. Both TNF-α and IL-6 can mediate the activity of osteocytes. Furthermore, both TNF-α and IL-6 are important pathogenic factors related to immune-mediated bone diseases including rheumatoid arthritis and postmenopausal osteoporosis. This review will discuss the contradictory findings concerning TNF-α and IL-6 in osteoimmunology and their potential for clinical application.


Assuntos
Osso e Ossos/imunologia , Osso e Ossos/metabolismo , Interleucina-6/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Artrite Reumatoide/imunologia , Artrite Reumatoide/metabolismo , Humanos , Osteoblastos/imunologia , Osteoblastos/metabolismo , Osteoclastos/imunologia , Osteoclastos/metabolismo , Osteoporose Pós-Menopausa/imunologia , Osteoporose Pós-Menopausa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA