Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Int J Mol Sci ; 23(21)2022 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-36362086

RESUMO

RUNX proteins, such as RUNX2, regulate the proliferation and differentiation of chondrocytes and osteoblasts. Haploinsufficiency of RUNX2 causes cleidocranial dysplasia, but a detailed analysis of Runx2+/- mice has not been reported. Furthermore, CBFB is required for the stability and DNA binding of RUNX family proteins. CBFB has two isoforms, and CBFB2 plays a major role in skeletal development. The calvaria, femurs, vertebrae and ribs in Cbfb2-/- mice were analyzed after birth, and compared with those in Runx2+/- mice. Calvarial development was impaired in Runx2+/- mice but mildly delayed in Cbfb2-/- mice. In femurs, the cortical bone but not trabecular bone was reduced in Cbfb2-/- mice, whereas both the trabecular and cortical bone were reduced in Runx2+/- mice. The trabecular bone in vertebrae increased in Cbfb2-/- mice but not in Runx2+/- mice. Rib development was impaired in Cbfb2-/- mice but not in Runx2+/- mice. These differences were likely caused by differences in the indispensability of CBFB and RUNX2, the balance of bone formation and resorption, or the number and maturation stage of osteoblasts. Thus, different amounts of CBFB and RUNX2 were required among the bone tissues for proper bone development and maintenance.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core , Osteoblastos , Animais , Camundongos , Diferenciação Celular/genética , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Subunidades alfa de Fatores de Ligação ao Core/metabolismo , Osteoblastos/metabolismo , Osteogênese/genética , Costelas/metabolismo , Crânio/metabolismo , Coluna Vertebral/metabolismo
2.
PLoS Genet ; 16(11): e1009169, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33253203

RESUMO

Chondrocytes proliferate and mature into hypertrophic chondrocytes. Vascular invasion into the cartilage occurs in the terminal hypertrophic chondrocyte layer, and terminal hypertrophic chondrocytes die by apoptosis or transdifferentiate into osteoblasts. Runx2 is essential for osteoblast differentiation and chondrocyte maturation. Runx2-deficient mice are composed of cartilaginous skeletons and lack the vascular invasion into the cartilage. However, the requirement of Runx2 in the vascular invasion into the cartilage, mechanism of chondrocyte transdifferentiation to osteoblasts, and its significance in bone development remain to be elucidated. To investigate these points, we generated Runx2fl/flCre mice, in which Runx2 was deleted in hypertrophic chondrocytes using Col10a1 Cre. Vascular invasion into the cartilage was similarly observed in Runx2fl/fl and Runx2fl/flCre mice. Vegfa expression was reduced in the terminal hypertrophic chondrocytes in Runx2fl/flCre mice, but Vegfa was strongly expressed in osteoblasts in the bone collar, suggesting that Vegfa expression in bone collar osteoblasts is sufficient for vascular invasion into the cartilage. The apoptosis of terminal hypertrophic chondrocytes was increased and their transdifferentiation was interrupted in Runx2fl/flCre mice, leading to lack of primary spongiosa and osteoblasts in the region at E16.5. The osteoblasts appeared in this region at E17.5 in the absence of transdifferentiation, and the number of osteoblasts and the formation of primary spongiosa, but not secondary spongiosa, reached to levels similar those in Runx2fl/fl mice at birth. The bone structure and volume and all bone histomophometric parameters were similar between Runx2fl/fl and Runx2fl/flCre mice after 6 weeks of age. These findings indicate that Runx2 expression in terminal hypertrophic chondrocytes is not required for vascular invasion into the cartilage, but is for their survival and transdifferentiation into osteoblasts, and that the transdifferentiation is necessary for trabecular bone formation in embryonic and neonatal stages, but not for acquiring normal bone structure and volume in young and adult mice.


Assuntos
Transdiferenciação Celular/genética , Condrócitos/fisiologia , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Osteoblastos/fisiologia , Osteogênese/genética , Fatores Etários , Animais , Apoptose/genética , Osso Esponjoso/citologia , Osso Esponjoso/embriologia , Osso Esponjoso/crescimento & desenvolvimento , Cartilagem/irrigação sanguínea , Cartilagem/citologia , Cartilagem/metabolismo , Sobrevivência Celular/genética , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Embrião de Mamíferos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Camundongos Knockout , Modelos Animais , Periósteo/citologia , Periósteo/embriologia , Periósteo/crescimento & desenvolvimento , Fator A de Crescimento do Endotélio Vascular/metabolismo
3.
Oral Dis ; 26(7): 1424-1431, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32419185

RESUMO

OBJECTIVE: Most malignant tumors require remodeling extracellular matrices (ECMs) for invasive growth and metastasis. Cancer cells and stromal cells remodel ECM. We investigated the relationship between regional lymph node (LN) metastasis and expression of ECM-remodeling factors in oral squamous cell carcinoma (OSCC). METHODS: Using primary OSCC and cervical LNs obtained surgically, we performed immunohistochemical evaluation of the ECM-remodeling factors, lysyl oxidase (LOX), MT1-MMP, S100A8, and TIMP-1 in primary tumor and marginal sinus histiocytosis (MSH) in LNs, and determined the statistical significance of the positive rates between metastatic and metastasis-free groups. RESULTS: Marginal sinus histiocytosis was more frequently formed in the metastatic group compared to the metastasis-free group. Lymphatic metastasis correlated with the immunopositivity rates of tumor cells expressing LOX, MT1-MMP, and TIMP-1, and of stromal cells expressing TIMP-1. The case rates of MSH containing macrophages positive for LOX and MT1-MMP in the metastasis group were significantly higher than in the metastasis-free group. ECM-remodeling-associated macrophages accumulate in marginal sinus in conjunction with lymphatic metastasis. CONCLUSION: Expression of LOX, MT1-MMP, and TIMP-1 in the parenchyma, and stromal expression of TIMP-1 in primary tumor may predict lymphatic metastasis. LOX and MT1-MMP have a possibility to participate in formation of pre-metastatic niche in LNs.


Assuntos
Carcinoma de Células Escamosas , Neoplasias Bucais , Matriz Extracelular , Humanos , Imuno-Histoquímica , Linfonodos
4.
J Oral Biosci ; 62(1): 9-15, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32032750

RESUMO

BACKGROUND: Cancer arises from cumulative genetic or epigenetic aberrations, or the destabilization of central signaling pathways that regulate cell proliferation, differentiation, cell cycle, gene transcription, migration, angiogenesis and apoptosis. Investigating the cancer-specific genetic background is important to get deeper apprehension of cancer biology. In this review, we aimed to identify head and neck squamous cell carcinoma (HNSCC)-specific genes and identified DKK3 gene as a candidate. HIGHLIGHT: DKK3 belongs to the DKK family (DKK1, DKK2, DKK3 and DKK4), which codes for an evolutionally conserved secreted glycoprotein that is characterized by two distinct cysteine rich domains and functions as an antagonist of the oncogenic Wnt signaling pathway. It has been reported that DKK3 expression is decreased in many kinds of cancers, and it is thus thought to be a tumor suppressor gene. However, our investigations have demonstrated unique expression and function of DKK3 in HNSCC. DKK3 protein expression is predominantly positive in HNSCC, and DKK3-positive patients show significantly shorter disease-free survival rates, whereas DKK3-negative cases do not show metastasis. Molecular biological analyses demonstrated that DKK3 over expression significantly increased HNSCC cell proliferation, migration, and invasion via increased phosphorylation of AKT. Moreover, DKK3 knockdown in HNSCC cells significantly decreased these malignant potentials through decreased AKT phosphorylation. CONCLUSION: Our previously published data, alongside those from other reports, indicate that DKK3 may have an additional oncogenic function other than tumor suppression.


Assuntos
Neoplasias de Cabeça e Pescoço , Carcinoma de Células Escamosas de Cabeça e Pescoço , Proteínas Adaptadoras de Transdução de Sinal , Linhagem Celular Tumoral , Quimiocinas , Humanos , Peptídeos e Proteínas de Sinalização Intercelular
5.
Jpn Dent Sci Rev ; 55(1): 80-87, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31049116

RESUMO

R-spondins (Rspos) are cysteine-rich secreted glycoproteins which control a variety of cellular functions and are essential for embryonic development and tissue homeostasis. R-spondins (Rspo1 to 4) have high structural similarity and share 60% sequence homology. It has been shown that their cysteine-rich furin-like (FU) domain and the thrombospondin (TSP) type I repeat domain are essential for initiating downstream signaling cascades and therefore for their biological functions. Although numerous studies have unveiled their pivotal role as critical developmental regulators, the most important finding is that Rspos synergize Wnt signaling. Recent studies have identified novel receptors for Rspos, the Lgr receptors, closely related orphans of the leucin-rich repeat containing G protein-coupled receptors, and proposed that Rspos potentiate canonical Wnt signaling via these receptors. Given that Wnt signaling is one of the most important developmental signaling pathways that controls cell fate decisions and tissue development, growth and homeostasis, Rspos may function as key players for these processes as well as potential therapeutic targets. Here, I recapitulate the Wnt signaling and then outline the biological role of Rspos in tissue development and homeostasis and explore the possibility that Rspos may be used as therapeutic targets.

6.
Bone ; 124: 148-157, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31051317

RESUMO

Androgen deficiency is a leading cause of male osteoporosis, with bone loss driven by an inadequate level of bone formation relative to the extent of bone resorption. Abaloparatide, an osteoanabolic PTH receptor agonist used to treat women with postmenopausal osteoporosis at high risk for fracture, increases bone formation and bone strength in estrogen-deficient animals without increasing bone resorption. This study examined the effects of abaloparatide on bone formation, bone mass, and bone strength in androgen-deficient orchiectomized (ORX) rats, a male osteoporosis model. Four-month-old Sprague-Dawley rats underwent ORX or sham surgery. Eight weeks later, sham-operated rats received vehicle (saline; n = 10) while ORX rats (n = 10/group) received vehicle (Veh) or abaloparatide at 5 or 25 µg/kg (ABL5 or ABL25) by daily s.c. injection for 8 weeks, followed by sacrifice. Dynamic bone histomorphometry indicated that the tibial diaphysis of one or both abaloparatide groups had higher periosteal mineralizing surface, intracortical bone formation rate (BFR), endocortical BFR, and cortical thickness vs Veh controls. Vertebral trabecular BFR was also higher in both abaloparatide groups vs Veh, and the ABL25 group had higher trabecular osteoblast surface without increased osteoclast surface. By micro-CT, the vertebra and distal femur of both abaloparatide-groups had improved trabecular bone volume and micro-architecture, and the femur diaphysis of the ABL25 group had greater cortical thickness with no increase in porosity vs Veh. Biomechanical testing indicated that both abaloparatide-groups had stronger vertebrae and femoral necks vs Veh controls. These findings provide preclinical support for evaluating abaloparatide as an investigational treatment for male osteoporosis.


Assuntos
Osso Esponjoso/patologia , Osso Cortical/patologia , Colo do Fêmur/patologia , Colo do Fêmur/fisiopatologia , Osteoporose/patologia , Osteoporose/fisiopatologia , Proteína Relacionada ao Hormônio Paratireóideo/uso terapêutico , Coluna Vertebral/fisiopatologia , Animais , Fenômenos Biomecânicos/efeitos dos fármacos , Osso Esponjoso/diagnóstico por imagem , Osso Esponjoso/efeitos dos fármacos , Modelos Animais de Doenças , Colo do Fêmur/diagnóstico por imagem , Masculino , Tamanho do Órgão/efeitos dos fármacos , Osteoporose/diagnóstico por imagem , Proteína Relacionada ao Hormônio Paratireóideo/farmacologia , Ratos Sprague-Dawley , Coluna Vertebral/diagnóstico por imagem , Coluna Vertebral/efeitos dos fármacos , Coluna Vertebral/patologia , Microtomografia por Raio-X
7.
J Bone Miner Res ; 34(9): 1707-1720, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30998833

RESUMO

Energy metabolism and bone homeostasis share several regulatory pathways. The AP1 transcription factor ΔFosB and leptin both regulate energy metabolism and bone, yet whether their pathways intersect is not known. Transgenic mice overexpressing ΔFosB under the control of the Enolase 2 (ENO2) promoter exhibit high bone mass, high energy expenditure, low fat mass, and low circulating leptin levels. Because leptin is a regulator of bone and ΔFosB acts on leptin-responsive ventral hypothalamic (VHT) neurons to induce bone anabolism, we hypothesized that regulation of leptin may contribute to the central actions of ΔFosB in the VHT. To address this question, we used adeno-associated virus (AAV) expression of ΔFosB in the VHT of leptin-deficient ob/ob mice and genetic crossing of ENO2-ΔFosB with ob/ob mice. In both models, leptin deficiency prevented ΔFosB-triggered reduction in body weight, increase in energy expenditure, increase in glucose utilization, and reduction in pancreatic islet size. In contrast, leptin deficiency failed to prevent ΔFosB-triggered increase in bone mass. Unlike leptin deficiency, galanin deficiency blocked both the metabolic and the bone ΔFosB-induced effects. Overall, our data demonstrate that, while the catabolic energy metabolism effects of ΔFosB require intact leptin and galanin signaling, the bone mass-accruing effects of ΔFosB require galanin but are independent of leptin. © 2019 American Society for Bone and Mineral Research.


Assuntos
Osso e Ossos/anatomia & histologia , Metabolismo Energético , Galanina/metabolismo , Hipotálamo/metabolismo , Leptina/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética , Animais , Peso Corporal , Deleção de Genes , Glucose/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tamanho do Órgão , Fosfopiruvato Hidratase/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo
8.
J Bone Miner Res ; 34(6): 1129-1142, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30690791

RESUMO

Human genetic evidence demonstrates that WNT1 mutations cause osteogenesis imperfecta (OI) and early-onset osteoporosis, implicating WNT1 as a major regulator of bone metabolism. However, its main cellular source and mechanisms of action in bone remain elusive. We generated global and limb bud mesenchymal cell-targeted deletion of Wnt1 in mice. Heterozygous deletion of Wnt1 resulted in mild trabecular osteopenia due to decreased osteoblast function. Targeted deletion of Wnt1 in mesenchymal progenitors led to spontaneous fractures due to impaired osteoblast function and increased bone resorption, mimicking the severe OI phenotype in humans with homozygous WNT1 mutations. Importantly, we showed for the first time that Wnt1 signals strictly in a juxtacrine manner to induce osteoblast differentiation and to suppress osteoclastogenesis, in part via canonical Wnt signaling. In conclusion, mesenchymal cell-derived Wnt1, acting in short range, is an essential regulator of bone homeostasis and an intriguing target for therapeutic interventions for bone diseases. © 2019 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals, Inc.


Assuntos
Diferenciação Celular , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/citologia , Osteoclastos/citologia , Via de Sinalização Wnt , Proteína Wnt1/metabolismo , Animais , Doenças Ósseas Metabólicas/patologia , Linhagem Celular , Núcleo Celular/metabolismo , Fraturas Ósseas/patologia , Deleção de Genes , Heterozigoto , Camundongos Knockout , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogênese
9.
Cell ; 175(7): 1756-1768.e17, 2018 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-30550785

RESUMO

Irisin is secreted by muscle, increases with exercise, and mediates certain favorable effects of physical activity. In particular, irisin has been shown to have beneficial effects in adipose tissues, brain, and bone. However, the skeletal response to exercise is less clear, and the receptor for irisin has not been identified. Here we show that irisin binds to proteins of the αV class of integrins, and biophysical studies identify interacting surfaces between irisin and αV/ß5 integrin. Chemical inhibition of the αV integrins blocks signaling and function by irisin in osteocytes and fat cells. Irisin increases both osteocytic survival and production of sclerostin, a local modulator of bone remodeling. Genetic ablation of FNDC5 (or irisin) completely blocks osteocytic osteolysis induced by ovariectomy, preventing bone loss and supporting an important role of irisin in skeletal remodeling. Identification of the irisin receptor should greatly facilitate our understanding of irisin's function in exercise and human health.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Remodelação Óssea , Fibronectinas/metabolismo , Integrina alfaV/metabolismo , Osteócitos/metabolismo , Osteólise/metabolismo , Adipócitos/patologia , Animais , Linhagem Celular Tumoral , Feminino , Fibronectinas/genética , Células HEK293 , Humanos , Integrina alfaV/genética , Camundongos , Osteócitos/patologia , Osteólise/genética
10.
Leukemia ; 32(8): 1739-1750, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29925904

RESUMO

Myeloma bone disease is a devastating complication of multiple myeloma (MM) and is caused by dysregulation of bone remodeling processes in the bone marrow microenvironment. Previous studies showed that microRNA-138 (miR-138) is a negative regulator of osteogenic differentiation of mesenchymal stromal cells (MSCs) and that inhibiting its function enhances bone formation in vitro. In this study, we explored the role of miR-138 in myeloma bone disease and evaluated the potential of systemically delivered locked nucleic acid (LNA)-modified anti-miR-138 oligonucleotides in suppressing myeloma bone disease. We showed that expression of miR-138 was significantly increased in MSCs from MM patients (MM-MSCs) and myeloma cells compared to those from healthy subjects. Furthermore, inhibition of miR-138 resulted in enhanced osteogenic differentiation of MM-MSCs in vitro and increased the number of endosteal osteoblastic lineage cells (OBCs) and bone formation rate in mouse models of myeloma bone disease. RNA sequencing of the OBCs identified TRPS1 and SULF2 as potential miR-138 targets that were de-repressed in anti-miR-138-treated mice. In summary, these data indicate that inhibition of miR-138 enhances bone formation in MM and that pharmacological inhibition of miR-138 could represent a new therapeutic strategy for treatment of myeloma bone disease.


Assuntos
Biomarcadores Tumorais/genética , Medula Óssea/patologia , Células-Tronco Mesenquimais/patologia , MicroRNAs/antagonistas & inibidores , Mieloma Múltiplo/terapia , Osteoblastos/patologia , Osteogênese , Animais , Medula Óssea/metabolismo , Estudos de Casos e Controles , Diferenciação Celular , Células Cultivadas , Feminino , Seguimentos , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , MicroRNAs/genética , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Osteoblastos/metabolismo , Prognóstico
11.
J Cell Physiol ; 233(2): 1585-1600, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28631813

RESUMO

There is a growing and alarming prevalence of obesity and the metabolic syndrome in type I diabetic patients (T1DM), particularly in adolescence. In general, low bone mass, higher fracture risk, and increased marrow adipose tissue (MAT) are features of diabetic osteopathy in insulin-deficient subjects. On the other hand, type 2 diabetes (T2DM) is associated with normal or high bone mass, a greater risk of peripheral fractures, and no change in MAT. Therefore, we sought to determine the effect of weight gain on bone turnover in insulin-deficient mice. We evaluated the impact of a 6-week high-fat (HFD) rich in medium chain fatty acids or low-fat diet (LFD) on bone mass and MAT in a streptozotocin (STZ)-induced model using male C57BL/6J mice at 8 weeks of age. Dietary intervention was initiated after diabetes confirmation. At the endpoint, lower non-fasting glucose levels were observed in diabetic mice fed with high fat diet compared to diabetic mice fed the low fat diet (STZ-LFD). Compared to euglycemic controls, the STZ-LFD had marked polydipsia and polyphagia, as well as reduced lean mass, fat mass, and bone parameters. Interestingly, STZ-HFD mice had higher bone mass, namely less cortical bone loss and more trabecular bone than STZ-LFD. Thus, we found that a HFD, rich in medium chain fatty acids, protects against bone loss in a T1DM mouse model. Whether this may also translate to T1DM patients who are overweight or obese in respect to maintenance of bone mass remains to be determined through longitudinal studies.


Assuntos
Glicemia/metabolismo , Composição Corporal , Remodelação Óssea , Diabetes Mellitus Experimental/dietoterapia , Diabetes Mellitus Tipo 1/dietoterapia , Dieta Hiperlipídica , Ácidos Graxos/administração & dosagem , Osteoporose/prevenção & controle , Estreptozocina , Adiposidade , Animais , Biomarcadores/sangue , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/induzido quimicamente , Diabetes Mellitus Tipo 1/fisiopatologia , Insulina/sangue , Cetonas/sangue , Masculino , Camundongos Endogâmicos C57BL , Osteoporose/sangue , Osteoporose/induzido quimicamente , Osteoporose/fisiopatologia , Fatores de Tempo , Redução de Peso
12.
Endocrinology ; 158(10): 3369-3385, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28977604

RESUMO

Obesity during maturation can affect the growing skeleton directly and indirectly, although these effects and the mechanisms behind them are not fully understood. Our objective was to determine how a high-fat diet with or without metformin treatment affects skeletal development. We also sought to characterize changes that occur in white adipose tissue, circulating metabolites, lipids, and gut microbiota. A diet-induced obesity C57BL/6J mouse model was used to test the effects of obesity and metformin on bone using bone histomorphometry and microcomputed tomography. Bone marrow adipose tissue was quantified with osmium tetroxide microcomputed tomography and histology. Dual-energy x-ray absorptiometry was used to analyze body composition. Hematoxylin and eosin staining was used to assess changes in white adipose depots, mass spectrometry was used for circulating lipids and protein metabolite analysis, and ribosomal RNA sequencing was used for gut microbiome analysis. Mice fed a high fat-diet since wean displayed increased medullary areas and decreased osteoblast numbers in the long bones; this phenotype was partially normalized by metformin. Marrow and inguinal adipose expansion was also noted in obese mice, and this was partially normalized by metformin. A drug-by-diet interaction was noted for circulating lipid molecules, protein metabolites, and gut microbiome taxonomical units. Obesity was not detrimental to trabecular bone in growing mice, but bone marrow medullary expansion was observed, likely resulting from inhibition of osteoblastogenesis, and this was partially reversed by metformin treatment.


Assuntos
Adiposidade/efeitos dos fármacos , Medula Óssea/efeitos dos fármacos , Osso Cortical/efeitos dos fármacos , Metformina/farmacologia , Obesidade , Absorciometria de Fóton , Tecido Adiposo Branco/efeitos dos fármacos , Animais , Composição Corporal , Contagem de Células , Cromatografia Líquida , Osso Cortical/patologia , Dieta Hiperlipídica , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/genética , Imuno-Histoquímica , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Espectrometria de Massas , Metabolômica , Camundongos , Camundongos Endogâmicos C57BL , Tamanho do Órgão , Osteoblastos/efeitos dos fármacos , Fenótipo , RNA Bacteriano/genética , RNA Ribossômico 16S/genética , Espectrometria de Massas em Tandem , Microtomografia por Raio-X
13.
Bone ; 105: 103-114, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28821457

RESUMO

Misty mice (m/m) have a loss of function mutation in Dock7 gene, a guanine nucleotide exchange factor, resulting in low bone mineral density, uncoupled bone remodeling and reduced bone formation. Dock7 has been identified as a modulator of osteoblast number and in vitro osteogenic differentiation in calvarial osteoblast culture. In addition, m/m exhibit reduced preformed brown adipose tissue innervation and temperature as well as compensatory increase in beige adipocyte markers. While the low bone mineral density phenotype is in part due to higher sympathetic nervous system (SNS) drive in young mice, it is unclear what effect aging would have in mice homozygous for the mutation in the Dock7 gene. We hypothesized that age-related trabecular bone loss and periosteal envelope expansion would be altered in m/m. To test this hypothesis, we comprehensively characterized the skeletal phenotype of m/m at 16, 32, 52, and 78wks of age. When compared to age-matched wild-type control mice (+/+), m/m had lower areal bone mineral density (aBMD) and areal bone mineral content (aBMC). Similarly, both femoral and vertebral BV/TV, Tb.N, and Conn.D were decreased in m/m while there was also an increase in Tb.Sp. As low bone mineral density and decreased trabecular bone were already present at 16wks of age in m/m and persisted throughout life, changes in age-related trabecular bone loss were not observed highlighting the role of Dock7 in controlling trabecular bone acquisition or bone loss prior to 16wks of age. Cortical thickness was also lower in the m/m across all ages. Periosteal and endosteal circumferences were higher in m/m compared to +/+ at 16wks. However, endosteal and periosteal expansion were attenuated in m/m, resulting in m/m having lower periosteal and endosteal circumferences by 78wks of age compared to +/+, highlighting the critical role of Dock7 in appositional bone expansion. Histomorphometry revealed that osteoblasts were nearly undetectable in m/m and marrow adipocytes were elevated 3.5 fold over +/+ (p=0.014). Consistent with reduced bone formation, osteoblast gene expression of Alp, Col1a1, Runx-2, Sp7, and Bglap was significantly decreased in m/m whole bone. Furthermore, markers of osteoclasts were either unchanged or suppressed. Bone marrow stromal cell migration and motility were inhibited in culture and changes in senescence markers suggest that osteoblast function may also be inhibited with loss of Dock7 expression in m/m. Finally, increased Oil Red O staining in m/m ear mesenchymal stem cells during adipogenesis highlights a potential shift of cells from the osteogenic to adipogenic lineages. In summary, loss of Dock7 in the aging m/m resulted in an impairment of periosteal and endocortical envelope expansion, but did not alter age-related trabecular bone loss. These studies establish Dock7 as a critical regulator of both cortical and trabecular bone mass, and demonstrate for the first time a novel role of Dock7 in modulating compensatory changes in the periosteum with aging.


Assuntos
Envelhecimento/patologia , Osso Esponjoso/patologia , Fatores de Troca do Nucleotídeo Guanina/genética , Mutação/genética , Periósteo/patologia , Adipócitos/metabolismo , Adipogenia , Tecido Adiposo/metabolismo , Adiposidade , Animais , Biomarcadores/sangue , Densidade Óssea , Reabsorção Óssea/sangue , Reabsorção Óssea/patologia , Osso Esponjoso/metabolismo , Contagem de Células , Movimento Celular , Feminino , Proteínas Ativadoras de GTPase , Regulação da Expressão Gênica , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Tamanho do Órgão , Osteoblastos/metabolismo , Osteoblastos/patologia , Periósteo/crescimento & desenvolvimento , Fenótipo , Sistema Nervoso Simpático/metabolismo
14.
Bone ; 97: 153-167, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28115279

RESUMO

Collagen triple helix repeat-containing1 (Cthrc1) has previously been implicated in osteogenic differentiation and positive regulation of bone mass, however, the underlying mechanisms remain unclear. Here we characterized the bone phenotype of a novel Cthrc1 null mouse strain using bone histomorphometry, µCT analysis and functional readouts for bone strength. In male Cthrc1 null mice both trabecular bone as well as cortical bone formation was impaired, whereas in female Cthrc1 null mice only trabecular bone parameters were altered. Novel and highly specific monoclonal antibodies revealed that CTHRC1 is expressed by osteocytes and osteoblasts, but not osteoclasts. Furthermore, Cthrc1 null mice exhibited increased bone resorption with increased number of osteoclast and increased osteoclast activity together with enhanced expression of osteoclastogenic genes such as c-Fos, Rankl, Trap, and Nfatc1. Differentiation of bone marrow-derived monocytes isolated from Cthrc1 null mice differentiated into osteoclasts as effectively as those from wildtype mice. In the presence of CTHRC1 osteoclastogenic differentiation of bone marrow-derived monocytes was dramatically inhibited as was functional bone resorption by osteoclasts. This process was accompanied by downregulation of osteoclastogenic marker genes, indicating that extrinsically derived CTHRC1 is required for such activity. In vitro, CTHRC1 had no effect on osteogenic differentiation of bone marrow stromal cells, however, calvarial osteoblasts from Cthrc1 null mice exhibited reduced osteogenic differentiation compared to osteoblasts from wildtypes. In a collagen antibody-induced arthritis model Cthrc1 null mice suffered significantly more severe inflammation and joint destruction than wildtypes, suggesting that CTHRC1 expressed by the activated synoviocytes has anti-inflammatory effects. Mechanistically, we found that CTHRC1 inhibited NFκB activation by preventing IκBα degradation while also inhibiting ERK1/2 activation. Collectively our studies demonstrate that CTHRC1 secreted from osteocytes and osteoblasts functions as an inhibitor of osteoclast differentiation via inhibition of NFκB-dependent signaling. Furthermore, our data suggest that CTHRC1 has potent anti-inflammatory properties that limit arthritic joint destruction.


Assuntos
Artrite Experimental/metabolismo , Artrite Experimental/patologia , Diferenciação Celular , Proteínas da Matriz Extracelular/metabolismo , Osteoclastos/metabolismo , Osteoclastos/patologia , Animais , Anticorpos , Fenômenos Biomecânicos , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Reabsorção Óssea/patologia , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/patologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Feminino , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteoblastos/patologia , Osteoclastos/efeitos dos fármacos , Osteócitos/efeitos dos fármacos , Osteócitos/metabolismo , Osteócitos/patologia , Osteogênese/efeitos dos fármacos , Ligante RANK/farmacologia , Transdução de Sinais/efeitos dos fármacos , Crânio/patologia , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Microtomografia por Raio-X
15.
Aging (Albany NY) ; 9(2): 353-369, 2017 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-28121620

RESUMO

The ventral hypothalamus (VHT) integrates several physiological cues to maintain glucose homeostasis and energy balance. Aging is associated with increased glucose intolerance but the underlying mechanisms responsible for age-related metabolic decline, including neuronal signaling in the VHT, remain elusive. We have shown that mice with VHT-targeted overexpression of ∆FosB, a splice variant of the AP1 transcription factor FosB, exhibit increased energy expenditure, leading to decreased adiposity. Here, we show that VHT-targeted overexpression of ∆FosB also improves glucose tolerance, increases insulin sensitivity in target organs and thereby suppresses insulin secretion. These effects are also observed by the overexpression of dominant negative JunD, demonstrating that they occur via AP1 antagonism within the VHT. Furthermore, the improved glucose tolerance and insulin sensitivity persisted in aged animals overexpressing ∆FosB in the VHT. These beneficial effects on glucose metabolism were abolished by peripheral sympathectomy and α-adrenergic, but not ß-adrenergic, blockade. Taken together, our results show that antagonizing AP1 transcription activity in the VHT leads to a marked improvement in whole body glucose homeostasis via activation of the SNS, conferring protection against age-related impairment in glucose metabolism. These findings may open novel avenues for therapeutic intervention in diabetes and age-related glucose intolerance.


Assuntos
Envelhecimento/metabolismo , Glicemia/metabolismo , Hipotálamo/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Sistema Nervoso Simpático/metabolismo , Adiposidade/fisiologia , Envelhecimento/genética , Animais , Metabolismo Energético/fisiologia , Intolerância à Glucose/metabolismo , Homeostase/fisiologia , Resistência à Insulina/fisiologia , Camundongos , Proteínas Proto-Oncogênicas c-fos/genética
16.
Nat Commun ; 7: 13176, 2016 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-27759007

RESUMO

Parathyroid hormone (PTH) activates receptors on osteocytes to orchestrate bone formation and resorption. Here we show that PTH inhibition of SOST (sclerostin), a WNT antagonist, requires HDAC4 and HDAC5, whereas PTH stimulation of RANKL, a stimulator of bone resorption, requires CRTC2. Salt inducible kinases (SIKs) control subcellular localization of HDAC4/5 and CRTC2. PTH regulates both HDAC4/5 and CRTC2 localization via phosphorylation and inhibition of SIK2. Like PTH, new small molecule SIK inhibitors cause decreased phosphorylation and increased nuclear translocation of HDAC4/5 and CRTC2. SIK inhibition mimics many of the effects of PTH in osteocytes as assessed by RNA-seq in cultured osteocytes and following in vivo administration. Once daily treatment with the small molecule SIK inhibitor YKL-05-099 increases bone formation and bone mass. Therefore, a major arm of PTH signalling in osteocytes involves SIK inhibition, and small molecule SIK inhibitors may be applied therapeutically to mimic skeletal effects of PTH.


Assuntos
Osso e Ossos/efeitos dos fármacos , Osteócitos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Hormônio Paratireóideo/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/genética , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal , Animais , Osso e Ossos/citologia , Osso e Ossos/metabolismo , Regulação da Expressão Gênica , Glicoproteínas/antagonistas & inibidores , Glicoproteínas/genética , Glicoproteínas/metabolismo , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Camundongos Knockout , Osteócitos/citologia , Osteócitos/metabolismo , Osteogênese/genética , Hormônio Paratireóideo/metabolismo , Fosforilação/efeitos dos fármacos , Cultura Primária de Células , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Ligante RANK/antagonistas & inibidores , Ligante RANK/genética , Ligante RANK/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
17.
Endocrinology ; 156(11): 4129-40, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26230658

RESUMO

Previously, we reported sexually dimorphic bone mass and body composition phenotypes in Igfbp2(-/-) mice (-/-), where male mice exhibited decreased bone and increased fat mass, whereas female mice displayed increased bone but no changes in fat mass. To investigate the interaction between IGF-binding protein (IGFBP)-2 and estrogen, we subjected Igfbp2 -/- and +/+ female mice to ovariectomy (OVX) or sham surgery at 8 weeks of age. At 20 weeks of age, mice underwent metabolic cage analysis and insulin tolerance tests before killing. At harvest, femurs were collected for microcomputed tomography, serum for protein levels, brown adipose tissue (BAT) and inguinal white adipose tissue (IWAT) adipose depots for histology, gene expression, and mitochondrial respiration analysis of whole tissue. In +/+ mice, serum IGFBP-2 dropped 30% with OVX. In the absence of IGFBP-2, OVX had no effect on preformed BAT; however, there was significant "browning" of the IWAT depot coinciding with less weight gain, increased insulin sensitivity, lower intraabdominal fat, and increased bone loss due to higher resorption and lower formation. Likewise, after OVX, energy expenditure, physical activity and BAT mitochondrial respiration were decreased less in the OVX-/- compared with OVX+/+. Mitochondrial respiration of IWAT was reduced in OVX+/+ yet remained unchanged in OVX-/- mice. These changes were associated with significant increases in Fgf21 and Foxc2 expression, 2 proteins known for their insulin sensitizing and browning of WAT effects. We conclude that estrogen deficiency has a profound effect on body and bone composition in the absence of IGFBP-2 and may be related to changes in fibroblast growth factor 21.


Assuntos
Doenças Ósseas Metabólicas/metabolismo , Metabolismo Energético/genética , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Ovariectomia , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Doenças Ósseas Metabólicas/genética , Feminino , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Camundongos , Camundongos Knockout , Mitocôndrias/genética
18.
Int J Nanomedicine ; 10: 3459-73, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25999711

RESUMO

We investigated the biological activity of W9, a bone resorption inhibitor peptide, using NanoClik nanoparticles as an injectable carrier, where acryloyl group-modified cholesterol-bearing pullulan (CHPOA) nanogels were crosslinked by pentaerythritol tetra (mercaptoethyl) polyoxyethylene. Thirty 5-week-old male C57BL/6J mice were fed a low calcium diet and received once-daily subcutaneous injections of the carrier alone, W9 24 mg/kg/day alone, W9 24 mg/kg/day incorporated in cholesterol bearing pullulan (CHP) nanogels, or W9 (8 and 24 mg/kg/day) incorporated in NanoClik nanoparticles for 4 days (n=5). Mice that received a normal calcium diet with NanoClik nanoparticle injections without W9 were used as a control group. Radiological analyses showed that administration of W9 24 mg/kg/day significantly prevented low calcium-induced reduction of bone mineral density in the long bones and lumbar vertebrae, but only when the NanoClik nanoparticles were used as a carrier. Histomorphometric analyses of the proximal tibiae revealed that W9 24 mg/kg/day incorporated in NanoClik nanoparticles prevented the increase in bone resorption indices induced by a low calcium diet, which was confirmed by measurement of serum bone resorption markers. These data suggest that NanoClik nanoparticles could be a useful carrier for peptide therapeutics, and also demonstrate that daily subcutaneous injections of the W9 peptide with the nanoparticles were able to inhibit bone loss in vivo. An osteoclastogenesis inhibition assay performed in vitro confirmed a slower release profile of W9 from NanoClik nanoparticles compared with conventional CHP nanogels.


Assuntos
Reabsorção Óssea/metabolismo , Portadores de Fármacos/química , Nanopartículas/química , Peptídeos , Polietilenoglicóis/química , Polietilenoimina/química , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nanogéis , Osteogênese/efeitos dos fármacos , Peptídeos/química , Peptídeos/farmacologia
19.
Endocrinology ; 156(7): 2374-83, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25853667

RESUMO

Atypical antipsychotic (AA) drugs cause significant metabolic side effects, and clinical data are emerging that demonstrate increased fracture risk and bone loss after treatment with the AA, risperidone (RIS). The pharmacology underlying the adverse effects on bone is unknown. However, RIS action in the central nervous system could be responsible because the sympathetic nervous system (SNS) is known to uncouple bone remodeling. RIS treatment in mice significantly lowered trabecular bone volume fraction (bone volume/total volume), owing to increased osteoclast-mediated erosion and reduced osteoblast-mediated bone formation. Daytime energy expenditure was also increased and was temporally associated with the plasma concentration of RIS. Even a single dose of RIS transiently elevated expression of brown adipose tissue markers of SNS activity and thermogenesis, Pgc1a and Ucp1. Rankl, an osteoclast recruitment factor regulated by the SNS, was also increased 1 hour after a single dose of RIS. Thus, we inferred that bone loss from RIS was regulated, at least in part, by the SNS. To test this, we administered RIS or vehicle to mice that were also receiving the nonselective ß-blocker propranolol. Strikingly, RIS did not cause any changes in trabecular bone volume/total volume, erosion, or formation while propranolol was present. Furthermore, ß2-adrenergic receptor null (Adrb2(-/-)) mice were also protected from RIS-induced bone loss. This is the first report to demonstrate SNS-mediated bone loss from any AA. Because AA medications are widely prescribed, especially to young adults, clinical studies are needed to assess whether ß-blockers will prevent bone loss in this vulnerable population.


Assuntos
Antagonistas Adrenérgicos beta/farmacologia , Antipsicóticos/efeitos adversos , Doenças Ósseas Metabólicas/induzido quimicamente , Remodelação Óssea/efeitos dos fármacos , Osso e Ossos/efeitos dos fármacos , Propranolol/farmacologia , Risperidona/efeitos adversos , Sistema Nervoso Simpático/efeitos dos fármacos , Tecido Adiposo Marrom/efeitos dos fármacos , Tecido Adiposo Marrom/metabolismo , Animais , Osso e Ossos/metabolismo , Metabolismo Energético/efeitos dos fármacos , Feminino , Canais Iônicos/efeitos dos fármacos , Canais Iônicos/metabolismo , Camundongos , Camundongos Knockout , Proteínas Mitocondriais/efeitos dos fármacos , Proteínas Mitocondriais/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Ligante RANK/efeitos dos fármacos , Ligante RANK/metabolismo , Receptores Adrenérgicos beta 2/genética , Sistema Nervoso Simpático/metabolismo , Termogênese , Fatores de Transcrição/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Proteína Desacopladora 1
20.
Nat Med ; 20(11): 1279-88, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25306233

RESUMO

The WNT16 locus is a major determinant of cortical bone thickness and nonvertebral fracture risk in humans. The disability, mortality and costs caused by osteoporosis-induced nonvertebral fractures are enormous. We demonstrate here that Wnt16-deficient mice develop spontaneous fractures as a result of low cortical thickness and high cortical porosity. In contrast, trabecular bone volume is not altered in these mice. Mechanistic studies revealed that WNT16 is osteoblast derived and inhibits human and mouse osteoclastogenesis both directly by acting on osteoclast progenitors and indirectly by increasing expression of osteoprotegerin (Opg) in osteoblasts. The signaling pathway activated by WNT16 in osteoclast progenitors is noncanonical, whereas the pathway activated in osteoblasts is both canonical and noncanonical. Conditional Wnt16 inactivation revealed that osteoblast-lineage cells are the principal source of WNT16, and its targeted deletion in osteoblasts increases fracture susceptibility. Thus, osteoblast-derived WNT16 is a previously unreported key regulator of osteoclastogenesis and fracture susceptibility. These findings open new avenues for the specific prevention or treatment of nonvertebral fractures, a substantial unmet medical need.


Assuntos
Fraturas Ósseas/metabolismo , Fraturas Ósseas/prevenção & controle , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogênese , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Envelhecimento/patologia , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular , Linhagem da Célula/efeitos dos fármacos , Células Cultivadas , Suscetibilidade a Doenças , Fraturas Ósseas/genética , Fraturas Ósseas/patologia , Deleção de Genes , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos Endogâmicos C57BL , Tamanho do Órgão/efeitos dos fármacos , Tamanho do Órgão/genética , Osteoblastos/efeitos dos fármacos , Osteoblastos/patologia , Osteoclastos/efeitos dos fármacos , Osteoclastos/patologia , Osteócitos/efeitos dos fármacos , Osteócitos/metabolismo , Osteócitos/patologia , Osteogênese/efeitos dos fármacos , Osteogênese/genética , Osteoprotegerina/metabolismo , Ligante RANK/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Crânio/patologia , Proteínas Wnt/deficiência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA