Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
JCI Insight ; 8(17)2023 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-37681409

RESUMO

The identity and origin of the stem/progenitor cells for adult joint cartilage repair remain unknown, impeding therapeutic development. Simulating the common therapeutic modality for cartilage repair in humans, i.e., full-thickness microfracture joint surgery, we combined the mouse full-thickness injury model with lineage tracing and identified a distinct skeletal progenitor cell type enabling long-term (beyond 7 days after injury) articular cartilage repair in vivo. Deriving from a population with active Prg4 expression in adulthood while lacking aggrecan expression, these progenitors proliferate, differentiate to express aggrecan and type II collagen, and predominate in long-term articular cartilage wounds, where they represent the principal repair progenitors in situ under native repair conditions without cellular transplantation. They originate outside the adult bone marrow or superficial zone articular cartilage. These findings have implications for skeletal biology and regenerative medicine for joint injury repair.


Assuntos
Cartilagem Articular , Adulto , Humanos , Animais , Camundongos , Agrecanas , Colágeno Tipo II , Modelos Animais de Doenças , Células-Tronco , Proteoglicanas
2.
J Bone Miner Res ; 36(4): 757-767, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33400836

RESUMO

Blocking the Wnt inhibitor, sclerostin, increases the rate of bone formation in rodents and in humans. On a cellular level, the antibody against sclerostin acts by increasing osteoblast numbers partly by activating the quiescent bone-lining cells in vivo. No evidence currently exists, to determine whether blocking sclerostin affects early cells of the osteoblast lineage. Here we use a lineage-tracing strategy that uses a tamoxifen-dependent cre recombinase, driven by the Sox9 promoter to mark early cells of the osteoblast lineage. We show that, when adult mice are treated with the rat-13C7, an antibody that blocks sclerostin action in rodents, it increases the numbers of osteoblast precursors and their differentiation into mature osteoblasts in vivo. We also show that rat-13C7 administration suppresses adipogenesis by suppressing the differentiation of Sox9creER+ skeletal precursors into bone marrow adipocytes in vivo. Using floxed alleles of the CTNNB1 gene encoding ß-catenin, we show that these precursor cells express the canonical Wnt signaling mediator, ß-catenin, and that the actions of the rat-13C7 antibody to increase the number of early precursors is dependent on direct stimulation of Wnt signaling. The increase in osteoblast precursors and their progeny after the administration of the antibody leads to a robust suppression of apoptosis without affecting the rate of their proliferation. Thus, neutralizing the Wnt-inhibitor sclerostin increases the numbers of early cells of the osteoblast lineage osteoblasts and suppresses their differentiation into adipocytes in vivo. © 2021 American Society for Bone and Mineral Research (ASBMR).


Assuntos
Osteoblastos , Osteócitos , Adipogenia , Animais , Camundongos , Osteoblastos/metabolismo , Osteócitos/metabolismo , Osteogênese , Ratos , Via de Sinalização Wnt , beta Catenina/metabolismo
3.
Curr Top Dev Biol ; 133: 1-24, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30902249

RESUMO

Accumulating evidence supports the idea that stem and progenitor cells play important roles in skeletal development. Over the last decade, the definition of skeletal stem and progenitor cells has evolved from cells simply defined by their in vitro behaviors to cells fully defined by a combination of sophisticated approaches, including serial transplantation assays and in vivo lineage-tracing experiments. These approaches have led to better identification of the characteristics of skeletal stem cells residing in multiple sites, including the perichondrium of the fetal bone, the resting zone of the postnatal growth plate, the bone marrow space and the periosteum in adulthood. These diverse groups of skeletal stem cells appear to closely collaborate and achieve a number of important biological functions of bones, including not only bone development and growth, but also bone maintenance and repair. Although these are important findings, we are only beginning to understand the diversity and the nature of skeletal stem and progenitor cells, and how they actually behave in vivo.


Assuntos
Desenvolvimento Ósseo , Células-Tronco/citologia , Animais , Linhagem da Célula , Ensaio de Unidades Formadoras de Colônias , Lâmina de Crescimento/embriologia , Humanos , Osteogênese , Células-Tronco/metabolismo
4.
Bone ; 118: 16-19, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29800694

RESUMO

Intermittent PTH-like drugs are the only approved so-called anabolic agent that increases bone mass in both mice and humans. It is well documented that PTH targets mature cells of the osteoblast lineage, with only indirect evidence of its actions on early cells of the osteoblast lineage. Using a triple transgenic mouse model that allowed labeling of very early cells of the osteoblast lineage, we traced the progeny of these into osteoblast lineage in adult mice. These early cells expressed PTH1R and multiplied when PTH (1-34) was administered daily. We also showed that the early mesenchymal cells showed accelerated differentiation into mature osteocalcin-positive osteoblasts and osteocytes. Rather surprisingly, when teriparatide administration was stopped, these early mesenchymal precursors differentiated into adipocytes. We showed that the adipogenic differentiation is accompanied by a decrease in wnt signaling in osteoblast precursors. In this review, we discuss the possible clinical relevance of this finding and the possible molecular mechanisms that contribute to this phenotype in vivo.


Assuntos
Adipogenia/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Hormônio Paratireóideo/administração & dosagem , Hormônio Paratireóideo/farmacologia , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Animais , Linhagem da Célula/efeitos dos fármacos , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos
5.
J Clin Invest ; 127(9): 3327-3338, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28758904

RESUMO

Teriparatide, a recombinant form of parathyroid hormone (PTH), is the only approved treatment for osteoporosis that increases the rate of bone formation. Teriparatide increases osteoblast numbers by suppressing osteoblast apoptosis and activating bone-lining cells. No direct evidence for teriparatide's actions on early cells of the osteoblast lineage has been demonstrated. Here, we have employed a lineage-tracing strategy that uses a tamoxifen-dependent, promoter-driven cre to mark early cells of the osteoblast lineage in adult mice. We show that teriparatide increases the numbers of osteoblast precursors and drives their differentiation into mature osteoblasts. Unexpectedly, following withdrawal of teriparatide therapy, bone marrow adipocytes increased dramatically in number. Some of these adipocytes derived from cells marked by Sox9-cre expression weeks earlier. Continued therapy with teriparatide prevented the appearance of adipocytes. Selective, inducible deletion of the PTH receptor in Sox9-cre cells demonstrated that PTH receptor expression is required for teriparatide-mediated increases in early osteoblast precursors. The increase in early precursors after teriparatide administration was associated with robust suppression of precursor apoptosis without affecting their rate of proliferation. Thus, teriparatide increases the numbers of early cells of the osteoblast lineage, hastens their differentiation into osteoblasts, and suppresses their differentiation into adipocytes in vivo.


Assuntos
Linhagem da Célula , Osteoblastos/citologia , Hormônio Paratireóideo/metabolismo , Adipócitos/citologia , Adipócitos/metabolismo , Animais , Apoptose , Diferenciação Celular , Proliferação de Células , Genes Reporter , Proteínas de Fluorescência Verde/genética , Camundongos , Camundongos Transgênicos , Nestina/genética , Osteoblastos/metabolismo , Osteócitos/citologia , Osteócitos/metabolismo , Osteogênese , Osteoporose/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição SOX9/genética , Teriparatida/uso terapêutico
6.
Sci Transl Med ; 9(387)2017 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-28446689

RESUMO

Bone tissue harbors unique and essential physiological processes, such as hematopoiesis, bone growth, and bone remodeling. To enable visualization of these processes at the cellular level in an intact environment, we developed "Bone CLARITY," a bone tissue clearing method. We used Bone CLARITY and a custom-built light-sheet fluorescence microscope to detect the endogenous fluorescence of Sox9-tdTomato+ osteoprogenitor cells in the tibia, femur, and vertebral column of adult transgenic mice. To obtain a complete distribution map of these osteoprogenitor cells, we developed a computational pipeline that semiautomatically detects individual Sox9-tdTomato+ cells in their native three-dimensional environment. Our computational method counted all labeled osteoprogenitor cells without relying on sampling techniques and displayed increased precision when compared with traditional stereology techniques for estimating the total number of these rare cells. We demonstrate the value of the clearing-imaging pipeline by quantifying changes in the population of Sox9-tdTomato-labeled osteoprogenitor cells after sclerostin antibody treatment. Bone tissue clearing is able to provide fast and comprehensive visualization of biological processes in intact bone tissue.


Assuntos
Células da Medula Óssea/citologia , Células-Tronco/citologia , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Camundongos , Camundongos Transgênicos , Osteoblastos/citologia , Osteogênese/fisiologia , Fatores de Transcrição SOX9/metabolismo , Células-Tronco/metabolismo
7.
Bone ; 97: 267-277, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28161590

RESUMO

Levels of circulating cytokines are elevated in inflammatory diseases. Previously, it was shown that interleukin (IL-)17A, in synergism with 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] and tumor necrosis factor α (TNFα), induces the release of granulocyte-macrophage colony-stimulating factor (GM-CSF) by murine osteoblasts in vitro. In this study, we further analyzed the effects of GM-CSF on osteoclast development in vitro. The effects of IL-17A, TNFα, and 1,25(OH)2D3 on the regulation of osteoclast development were investigated in cocultures of bone marrow-derived osteoclast progenitor cells (OPC) and mouse calvarial osteoblasts. Additionally, OPC were grown for 3days in media containing macrophage colony-stimulating factor (M-CSF), GM-CSF, or M-CSF/GM-CSF. Subsequently, the osteoclastogenic potential and the capacity to dissolve amorphous calcium phosphate were assessed in each of the three populations of OPC. IL-17A, in synergism with TNFα and 1,25(OH)2D3, inhibited the development of osteoclasts in cocultures by stimulating the osteoblast lineage cells to release GM-CSF. GM-CSF-treated OPC expressed traits characteristic of dendritic cells. Upon removal of GM-CSF and supplementation of the culture media with M-CSF/RANKL, the cells lost their dendritic cell characteristics and differentiated into osteoclasts. OPC pretreated with GM-CSF and M-CSF/GM-CSF exhibited delayed development to osteoclasts and an extended proliferation phase. Elevated levels of GM-CSF in systemic inflammatory diseases may cause an expansion of the OPC pools in the bone, bone marrow, and blood. Upon homing to the bone, this may lead to an increase in the number of osteoclasts and in bone resorption.


Assuntos
Antígeno CD11c/metabolismo , Diferenciação Celular/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Osteoclastos/citologia , Osteoclastos/metabolismo , Animais , Fosfatos de Cálcio/farmacologia , Linhagem da Célula/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Regulação da Expressão Gênica/efeitos dos fármacos , Interleucina-17/metabolismo , Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/citologia , Camundongos Endogâmicos C57BL , Monócitos/citologia , Osteoclastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Fenótipo , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
8.
J Bone Miner Res ; 32(5): 892-901, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-27862326

RESUMO

Sclerostin antibody (Scl-Ab) increases osteoblast activity, in part through increasing modeling-based bone formation on previously quiescent surfaces. Histomorphometric studies have suggested that this might occur through conversion of bone lining cells into active osteoblasts. However, direct data demonstrating Scl-Ab-induced conversion of lining cells into active osteoblasts are lacking. Here, we used in vivo lineage tracing to determine if Scl-Ab promotes the conversion of lining cells into osteoblasts on periosteal and endocortical bone surfaces in mice. Two independent, tamoxifen-inducible lineage-tracing strategies were used to label mature osteoblasts and their progeny using the DMP1 and osteocalcin promoters. After a prolonged "chase" period, the majority of labeled cells on bone surfaces assumed a thin, quiescent morphology. Then, mice were treated with either vehicle or Scl-Ab (25 mg/kg) twice over the course of the subsequent week. After euthanization, marked cells were enumerated, their thickness quantified, and proliferation and apoptosis examined. Scl-Ab led to a significant increase in the average thickness of labeled cells on periosteal and endocortical bone surfaces, consistent with osteoblast activation. Scl-Ab did not induce proliferation of labeled cells, and Scl-Ab did not regulate apoptosis of labeled cells. Therefore, direct reactivation of quiescent bone lining cells contributes to the acute increase in osteoblast numbers after Scl-Ab treatment in mice. © 2016 American Society for Bone and Mineral Research.


Assuntos
Anticorpos/farmacologia , Osso Cortical/metabolismo , Glicoproteínas/antagonistas & inibidores , Osteoblastos/metabolismo , Periósteo/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Osso Cortical/citologia , Glicoproteínas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Camundongos Transgênicos , Osteoblastos/citologia , Periósteo/citologia
9.
RMD Open ; 2(1): e000079, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26819749

RESUMO

OBJECTIVE: To investigate the regulatory effect of tumour necrosis factor (TNF) blockade with infliximab on the distribution of peripheral blood monocyte subpopulations in patients with rheumatoid arthritis (RA) and ankylosing spondylitis (AS). METHODS: Purified CD11b+CD14+ monocytes from 5 patients with RA and 5 AS were analysed ex vivo before and after infliximab treatment by flow cytometry for CD16, CD163, CD11b, C-C chemokine receptor type 2 (CCR2) and CXC chemokine receptor 4 (CXCR4) at baseline and at days 2, 14, 84 and 168 after the first infliximab administration. Serum levels of the stromal cell-derived factor (SDF)-1 and monocyte chemotactic peptide (MCP)-1 at different time points were measured in either patient group before and on infliximab treatment. RESULTS: Anti-TNF treatment with infliximab led to a significant increase of circulating CD11b+ non-classical and a concomitantly decrease of CD11b+ classical monocytes, to a decline in SDF-1 levels and reduced expression of CCR2 and CXCR4 on non-classical monocyte subpopulation. CONCLUSIONS: Our study shows, that TNFα blockade by infliximab resulted in a dichotomy of the regulation of classical and non-classical monocytes that might have substantial impact on inhibition of osteoclastogenesis and of subsequent juxta-articular bone destruction and systemic bone loss in RA and AS.

10.
Arthritis Rheum ; 65(2): 436-46, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23124514

RESUMO

OBJECTIVE: To investigate the effects of interleukin-17A (IL-17A) on osteoclastogenesis in vitro. METHODS: Bone marrow cells (BMCs) were isolated from the excised tibia and femora of wild-type C57BL/6J mice, and osteoblasts were obtained by sequential digestion of the calvariae of ddY, C57BL/6J, and granulocyte-macrophage colony-stimulating factor-knockout (GM-CSF(-/-)) mice. Monocultures of BMCs or cocultures of BMCs and osteoblasts were supplemented with or without 1,25-dihydroxyvitamin D(3)(1,25[OH](2)D(3)), recombinant human macrophage colony-stimulating factor (M-CSF), RANKL, and IL-17A. After 5-6 days, the cultures were fixed with 4% paraformaldehyde and subsequently stained for the osteoclast marker enzyme tartrate-resistant acid phosphatase (TRAP). Osteoprotegerin (OPG) and GM-CSF expression were measured by enzyme-linked immunosorbent assay, and transcripts for RANK and RANKL were detected by real-time polymerase chain reaction. RESULTS: In both culture systems, IL-17A alone did not affect the development of osteoclasts. However, the addition of IL-17A plus 1,25(OH)(2)D(3) to cocultures inhibited early osteoclast development within the first 3 days of culture and induced release of GM-CSF into the culture supernatants. Furthermore, in cocultures of GM-CSF(-/-) mouse osteoblasts and wild-type mouse BMCs, IL-17A did not affect osteoclast development, corroborating the role of GM-CSF as the mediator of the observed inhibition of osteoclastogenesis by IL-17A. CONCLUSION: These findings suggest that IL-17A interferes with the differentiation of osteoclast precursors by inducing the release of GM-CSF from osteoblasts.


Assuntos
Calcitriol/farmacologia , Diferenciação Celular/fisiologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Interleucina-17/farmacologia , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Animais , Contagem de Células , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoclastos/citologia , Osteoclastos/efeitos dos fármacos , Osteoprotegerina/metabolismo
11.
Clin Oral Implants Res ; 22(6): 578-86, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21054552

RESUMO

OBJECTIVE: Central to the process of osseointegration is the recruitment of mesenchymal progenitor cells to the healing site, their proliferation and differentiation to bone synthesising osteoblasts. The process is under the control of pro-inflammatory cytokines and growth factors. The aim of this study was to monitor these key stages of osseointegration and the signalling milieu during bone healing around implants placed in healthy and diabetic bone. METHODS: Implants were placed into the sockets of incisors extracted from the mandibles of normal Wistar and diabetic Goto-Kakizaki rats. Mandibles 1-12 weeks post-insertion of the implant were examined by histochemistry and immunocytochemistry to localise the presence of Stro-1- positive mesenchymal progenitor cells, proliferating cellular nuclear antigen proliferative cells, osteopontin and osteocalcin, macrophages, pro-inflammatory cytokines interleukin (IL)-1ß, IL-6, tumour necrosis factor (TNF)-α and tumour growth factor (TGF)-ß1. Image analysis provided a semi-quantification of positively expressing cells. RESULTS: Histological staining identified a delay in the formation of mineralised bone around implants placed in diabetic animals. Within the diabetic bone, the migration of Stro-1 mesenchymal cells in the healing tissue appeared to be unaffected. However, in the diabetic healing bone, the onset of cell proliferation and osteoblast differentiation were delayed and subsequently prolonged compared with normal bone. Similar patterns of change were observed in diabetic bone for the presence of IL-1ß, TNF-α, macrophages and TGF-ß1. CONCLUSION: The observed alterations in the extracellular presence of pro-inflammatory cytokines, macrophages and growth factors within diabetic tissues that correlate to changes in the signalling milieu, may affect the proliferation and differentiation of mesenchymal progenitor cells in the osseointegration process.


Assuntos
Implantes Dentários , Diabetes Mellitus Tipo 2/patologia , Incisivo , Mediadores da Inflamação/fisiologia , Osteoblastos/patologia , Alvéolo Dental/cirurgia , Animais , Antígenos de Superfície/análise , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Proliferação de Células , Processamento de Imagem Assistida por Computador/métodos , Interleucina-1beta/análise , Macrófagos/patologia , Masculino , Mandíbula/cirurgia , Células-Tronco Mesenquimais/patologia , Osseointegração/fisiologia , Osteocalcina/análise , Osteogênese/fisiologia , Osteopontina/análise , Antígeno Nuclear de Célula em Proliferação/análise , Ratos , Ratos Wistar , Alvéolo Dental/patologia , Fator de Crescimento Transformador beta1/análise , Fator de Necrose Tumoral alfa/análise , Cicatrização/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...