Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 114
Filtrar
1.
Physiol Rev ; 102(1): 379-410, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34337974

RESUMO

Osteocytes, former osteoblasts encapsulated by mineralized bone matrix, are far from being passive and metabolically inactive bone cells. Instead, osteocytes are multifunctional and dynamic cells capable of integrating hormonal and mechanical signals and transmitting them to effector cells in bone and in distant tissues. Osteocytes are a major source of molecules that regulate bone homeostasis by integrating both mechanical cues and hormonal signals that coordinate the differentiation and function of osteoclasts and osteoblasts. Osteocyte function is altered in both rare and common bone diseases, suggesting that osteocyte dysfunction is directly involved in the pathophysiology of several disorders affecting the skeleton. Advances in osteocyte biology initiated the development of novel therapeutics interfering with osteocyte-secreted molecules. Moreover, osteocytes are targets and key distributors of biological signals mediating the beneficial effects of several bone therapeutics used in the clinic. Here we review the most recent discoveries in osteocyte biology demonstrating that osteocytes regulate bone homeostasis and bone marrow fat via paracrine signaling, influence body composition and energy metabolism via endocrine signaling, and contribute to the damaging effects of diabetes mellitus and hematologic and metastatic cancers in the skeleton.


Assuntos
Remodelação Óssea/fisiologia , Osteoclastos/citologia , Osteócitos/citologia , Osteogênese/fisiologia , Animais , Reabsorção Óssea/metabolismo , Diferenciação Celular/fisiologia , Humanos
2.
FASEB J ; 36(3): e22196, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35137455

RESUMO

Parathyroid hormone (PTH) signaling downstream of the PTH 1 receptor (Pth1r) results in both bone anabolic and catabolic actions by mechanisms not yet fully understood. In this study, we show that Pth1r signaling upregulates the expression of several components of the Notch pathway and that Notch signals contribute to the catabolic actions of PTH in bone. We found that constitutive genetic activation of PTH receptor signaling in osteocytes (caPth1rOt ) or treatment with PTH daily increased the expression of several Notch ligands/receptors in bone. In contrast, sustained elevation of endogenous PTH did not change Notch components expression. Deletion of the PTH receptor or sclerostin overexpression in osteocytes abolished Notch increases by PTH. Further, deleting the canonical Notch transcription factor Rbpjk in osteocytes decreased bone mass and increased resorption and Rankl expression in caPth1rOt mice. Moreover, pharmacological bone-targeted Notch inhibition potentiated the bone mass gain induced by intermittent PTH by reducing bone resorption and preserving bone formation. Thus, Notch activation lies downstream of anabolic signaling driven by PTH actions in osteocytes, and Notch pharmacological inhibition maximizes the bone anabolic effects of PTH.


Assuntos
Reabsorção Óssea/metabolismo , Osteogênese , Hormônio Paratireóideo/metabolismo , Receptores Notch/metabolismo , Animais , Reabsorção Óssea/genética , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Osteócitos/metabolismo , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo , Receptores Notch/genética , Transdução de Sinais
3.
Transgenic Res ; 32(1-2): 135-141, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36547785

RESUMO

The Dmp1-Cre mouse, expressing Cre from an 8-kb DNA fragment of the mouse Dmp1 gene, is a common tool to study gene functions in osteocytes. Here we report that the deletion of Tsc1 (TSC complex subunit 1) by 8 kb Dmp1-Cre causes rectal prolapse in mice. Histological examination shows the presence of colon polyps in Tsc1-deficient mice in association with significantly larger colon and narrower lumen, which recapitulates the common polyps pathology in Tuberous Sclerosis, an autosomal dominant disorder caused by mutations in either TSC1 or TSC2. The intestine in Tsc1-deficient mice is also enlarged with the presence of taller villi. Using the Ai14 reporter mice that express a red fluorescence protein upon Cre recombination, we show that 8 kb Dmp1-Cre activity is evident in portion of the mesenchyme of the colon and small intestine. Lastly, our data show that Tsc1 deletion by Dmp1-Cre leads to an increased proliferation in the mesenchyme of colon, which at least partly contributes to the polyps pathology seen in this mouse model and is likely a contributing factor of the polyps in Tuberous Sclerosis.


Assuntos
Esclerose Tuberosa , Proteínas Supressoras de Tumor , Camundongos , Animais , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Proteína 1 do Complexo Esclerose Tuberosa/genética , Proteína 1 do Complexo Esclerose Tuberosa/metabolismo , Esclerose Tuberosa/genética , Esclerose Tuberosa/patologia , Integrases/genética , Proteínas da Matriz Extracelular
4.
J Periodontal Res ; 58(4): 723-732, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37128744

RESUMO

BACKGROUND AND OBJECTIVE: Mutations in low-density lipoprotein receptor-related protein 5 (LRP5) cause various bone diseases. Several mouse models were generated to study the role of LRP5 in bone development. But most of the studies were confined to the appendicular skeleton. The role of LRP5 in the axial skeleton, especially in the craniofacial skeleton, is largely unknown. The aim of this study was to investigate the craniofacial phenotype with the LRP5G171V mutation. METHODS: To understand how LRP5 affects craniofacial bone properties, we analyzed LRP5 high-bone-mass mutant mice carrying the G171V missense mutation (LRP5HBM ). Quantitative microcomputed tomographic imaging and histomorphometric analyses were used to study craniofacial phenotypes and bone density. Histology, immunohistochemistry, and in vivo fluorochrome labeling were used to study molecular mechanisms. RESULTS: LRP5HBM mice showed overall minor changes in the craniofacial bone development but with increased bone mass in the interradicular alveolar bone, edentulous ridge, palatine bone, and premaxillary suture. Elevated osteocyte density was observed in LRP5HBM mice, along with increased Runx2 expression and unmineralized bone surrounding osteocytes. Meanwhile, LRP5HBM mice exhibited increased osteoprogenitors, but no significant changes were observed in osteoclasts. This led to a high-bone-mass phenotype, and an increased osteocyte density in the alveolar bone and edentulous ridge. CONCLUSION: LRP5HBM mice display increased bone mass in the alveolar bone with minor changes in the craniofacial morphology. Collectively, these data elucidated the important role of LRP5 in axial bone development and homeostasis and provided clues into the therapeutical potential of LRP5 signaling in treating alveolar bone loss.


Assuntos
Osso e Ossos , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Camundongos , Animais , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Osso e Ossos/metabolismo , Mutação/genética , Densidade Óssea/genética , Osteoclastos/metabolismo
5.
J Clin Densitom ; 26(4): 101432, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37944445

RESUMO

The Santa Fe Bone Symposium (SFBS) held its 23rd annual event on August 5-6, 2023, in Santa Fe, New Mexico, USA. Attendees participated in-person and remotely, representing many states and countries. The program included plenary presentations, panel discussions, satellite symposia, a Project ECHO workshop, and a session on healthcare policy and reimbursement for fracture liaison programs. A broad range of topics were addressed, including transitions of osteoporosis treatments over a lifetime; controversies in vitamin D; update on Official Positions of the International Society for Clinical Densitometry; spine surgery and bone health; clinical applications of bone turnover markers; basic bone biology for clinicians; premenopausal-, pregnancy-, and lactation-associated osteoporosis; cancer treatment induced bone loss in patients with breast cancer and prostate cancer; genetic testing for skeletal diseases; and an update on nutrition and bone health. There were also sessions on rare bone diseases, including managing patients with hypophosphatasia; treatment of X-linked hypophosphatemia; and assessment and treatment of patients with hypoparathyroidism. There were oral presentations of abstracts by endocrinology fellows selected from those who participated in the Santa Fe Fellows Workshop on Metabolic Bone Diseases, held the 2 days prior to the SFBS. These proceedings of the 2023 SFBS present the clinical highlights and insights generated from many formal and informal discussions in Santa Fe.


Assuntos
Doenças Ósseas Metabólicas , Fraturas Ósseas , Osteoporose , Masculino , Feminino , Humanos , Absorciometria de Fóton , Osteoporose/tratamento farmacológico , Fraturas Ósseas/terapia , Doenças Ósseas Metabólicas/etiologia , Doenças Ósseas Metabólicas/terapia , Densidade Óssea
6.
Int J Mol Sci ; 24(5)2023 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-36902150

RESUMO

Calcium/calmodulin (CaM)-dependent protein kinase kinase 2 (CaMKK2) regulates bone remodeling through its effects on osteoblasts and osteoclasts. However, its role in osteocytes, the most abundant bone cell type and the master regulator of bone remodeling, remains unknown. Here we report that the conditional deletion of CaMKK2 from osteocytes using Dentine matrix protein 1 (Dmp1)-8kb-Cre mice led to enhanced bone mass only in female mice owing to a suppression of osteoclasts. Conditioned media isolated from female CaMKK2-deficient osteocytes inhibited osteoclast formation and function in in vitro assays, indicating a role for osteocyte-secreted factors. Proteomics analysis revealed significantly higher levels of extracellular calpastatin, a specific inhibitor of calcium-dependent cysteine proteases calpains, in female CaMKK2 null osteocyte conditioned media, compared to media from female control osteocytes. Further, exogenously added non-cell permeable recombinant calpastatin domain I elicited a marked, dose-dependent inhibition of female wild-type osteoclasts and depletion of calpastatin from female CaMKK2-deficient osteocyte conditioned media reversed the inhibition of matrix resorption by osteoclasts. Our findings reveal a novel role for extracellular calpastatin in regulating female osteoclast function and unravel a novel CaMKK2-mediated paracrine mechanism of osteoclast regulation by female osteocytes.


Assuntos
Osteoclastos , Osteócitos , Animais , Feminino , Camundongos , Cálcio/metabolismo , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/metabolismo , Meios de Cultivo Condicionados/farmacologia , Osteoclastos/metabolismo , Osteócitos/metabolismo , Caracteres Sexuais
7.
FASEB J ; 32(5): 2706-2721, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29401582

RESUMO

The functions of the paralogous transcriptional coactivators Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) in bone are controversial. Each has been observed to promote or inhibit osteogenesis in vitro, with reports of both equivalent and divergent functions. Their combinatorial roles in bone physiology are unknown. We report that combinatorial YAP/TAZ deletion from skeletal lineage cells, using Osterix-Cre, caused an osteogenesis imperfecta-like phenotype with severity dependent on allele dose and greater phenotypic expressivity with homozygous TAZ vs. YAP ablation. YAP/TAZ deletion decreased bone accrual and reduced intrinsic bone material properties through impaired collagen content and organization. These structural and material defects produced spontaneous fractures, particularly in mice with homozygous TAZ deletion and caused neonatal lethality in dual homozygous knockouts. At the cellular level in vivo, YAP/TAZ ablation reduced osteoblast activity and increased osteoclast activity, in an allele dose-dependent manner, impairing bone accrual and remodeling. Transcriptionally, YAP/TAZ deletion and small-molecule inhibition of YAP/TAZ interaction with the transcriptional coeffector TEAD reduced osteogenic and collagen-related gene expression, both in vivo and in vitro. These data demonstrate that YAP and TAZ combinatorially promote bone development through regulation of osteoblast activity, matrix quality, and osteoclastic remodeling.-Kegelman, C. D., Mason, D. E., Dawahare, J. H., Horan, D. J., Vigil, G. D., Howard, S. S., Robling, A. G., Bellido, T. M., Boerckel, J. D. Skeletal cell YAP and TAZ combinatorially promote bone development.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Desenvolvimento Ósseo , Matriz Óssea/metabolismo , Remodelação Óssea , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Fosfoproteínas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteínas de Ciclo Celular , Deleção de Genes , Camundongos , Camundongos Knockout , Osteogênese Imperfeita/genética , Osteogênese Imperfeita/metabolismo , Osteogênese Imperfeita/patologia , Fosfoproteínas/genética , Transativadores , Proteínas de Sinalização YAP
8.
FASEB J ; 32(5): 2878-2890, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29401593

RESUMO

Parathyroid hormone (PTH) affects the skeleton by acting on osteocytes (Ots) in bone through yet unclear mechanisms. We report that matrix metalloproteinase 14 (MMP14) expression/activity are increased in bones from mice with genetic constitutive activation (ca) of the PTH receptor 1 (PTH1R) in Ots (caPTH1ROt) and in bones from mice exposed to elevated PTH levels but not in mice lacking [conditional knockout (cKO)] the PTH1R in Ots (cKOPTH1ROt). Furthermore, PTH upregulates MMP14 in human bone cultures and in Ot-enriched bones from floxed control mice but not from cKOPTH1ROt mice. MMP14 activity increases soluble receptor activator of NF-κΒ ligand production, which in turn, stimulates osteoclast differentiation and resorption. Pharmacologic inhibition of MMP14 activity reduced the high bone remodeling exhibited by caPTH1ROt mice or induced by chronic PTH elevation and decreased bone resorption but allowed full stimulation of bone formation induced by PTH injections, thereby potentiating bone gain. Thus, MMP14 is a new member of the intricate gene network activated in Ots by PTH1R signaling that can be targeted to adjust the skeletal responses to PTH in favor of bone preservation.-Delgado-Calle, J., Hancock, B., Likine, E. F., Sato, A. Y., McAndrews, K., Sanudo, C., Bruzzaniti, A., Riancho, J. A., Tonra, J. R., Bellido, T. MMP14 is a novel target of PTH signaling in osteocytes that controls resorption by regulating soluble RANKL production.


Assuntos
Reabsorção Óssea/metabolismo , Metaloproteinase 14 da Matriz/metabolismo , Osteócitos/metabolismo , Hormônio Paratireóideo/metabolismo , Ligante RANK/biossíntese , Transdução de Sinais/fisiologia , Animais , Reabsorção Óssea/genética , Células Cultivadas , Redes Reguladoras de Genes/fisiologia , Metaloproteinase 14 da Matriz/genética , Camundongos , Camundongos Knockout , Osteoclastos/citologia , Osteoclastos/metabolismo , Osteócitos/citologia , Osteogênese/fisiologia , Hormônio Paratireóideo/genética , Ligante RANK/genética , Receptor Tipo 1 de Hormônio Paratireóideo/genética , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo
9.
Proc Natl Acad Sci U S A ; 112(5): E478-86, 2015 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-25605937

RESUMO

Osteocytes, >90% of the cells in bone, lie embedded within the mineralized matrix and coordinate osteoclast and osteoblast activity on bone surfaces by mechanisms still unclear. Bone anabolic stimuli activate Wnt signaling, and human mutations of components along this pathway underscore its crucial role in bone accrual and maintenance. However, the cell responsible for orchestrating Wnt anabolic actions has remained elusive. We show herein that activation of canonical Wnt signaling exclusively in osteocytes [dominant active (da)ßcat(Ot) mice] induces bone anabolism and triggers Notch signaling without affecting survival. These features contrast with those of mice expressing the same daß-catenin in osteoblasts, which exhibit decreased resorption and perinatal death from leukemia. daßcat(Ot) mice exhibit increased bone mineral density in the axial and appendicular skeleton, and marked increase in bone volume in cancellous/trabecular and cortical compartments compared with littermate controls. daßcat(Ot) mice display increased resorption and formation markers, high number of osteoclasts and osteoblasts in cancellous and cortical bone, increased bone matrix production, and markedly elevated periosteal bone formation rate. Wnt and Notch signaling target genes, osteoblast and osteocyte markers, and proosteoclastogenic and antiosteoclastogenic cytokines are elevated in bones of daßcat(Ot) mice. Further, the increase in RANKL depends on Sost/sclerostin. Thus, activation of osteocytic ß-catenin signaling increases both osteoclasts and osteoblasts, leading to bone gain, and is sufficient to activate the Notch pathway. These findings demonstrate disparate outcomes of ß-catenin activation in osteocytes versus osteoblasts and identify osteocytes as central target cells of the anabolic actions of canonical Wnt/ß-catenin signaling in bone.


Assuntos
Osso e Ossos/metabolismo , Osteócitos/fisiologia , Via de Sinalização Wnt , beta Catenina/metabolismo , Animais , Densidade Óssea , Camundongos , Camundongos Transgênicos
10.
Clin Rev Bone Miner Metab ; 16(1): 33-47, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29962904

RESUMO

Glucocorticoids (GC), produced and released by the adrenal glands, regulate numerous physiological processes in a wide range of tissues. Because of their profound immunosuppressive and anti-inflammatory actions, GC are extensively used for the treatment of immune and inflammatory conditions, the management of organ transplantation, and as a component of chemotherapy regimens for cancers. However, both pathologic endogenous elevation and long-term use of exogenous GC are associated with severe adverse effects. In particular, excess GC has devastating effects on the musculoskeletal system. GC increase bone resorption and decrease formation leading to bone loss, microarchitectural deterioration and fracture. GC also induce loss of muscle mass and strength leading to an increased incidence of falls. The combined effects on bone and muscle account for the increased fracture risk with GC. This review summarizes the advance in knowledge in the last two decades about the mechanisms of action of GC in bone and muscle and the attempts to interfere with the damaging actions of GC in these tissues with the goal of developing more effective therapeutic strategies.

11.
Kidney Int ; 92(3): 599-611, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28396120

RESUMO

Osteocytes within the mineralized bone matrix control bone remodeling by regulating osteoblast and osteoclast activity. Osteocytes express the aging suppressor Klotho, but the functional role of this protein in skeletal homeostasis is unknown. Here we identify Klotho expression in osteocytes as a potent regulator of bone formation and bone mass. Targeted deletion of Klotho from osteocytes led to a striking increase in bone formation and bone volume coupled with enhanced osteoblast activity, in sharp contrast to what is observed in Klotho hypomorphic (kl/kl) mice. Conversely, overexpression of Klotho in cultured osteoblastic cells inhibited mineralization and osteogenic activity during osteocyte differentiation. Further, the induction of chronic kidney disease with high-turnover renal osteodystrophy led to downregulation of Klotho in bone cells. This appeared to offset the skeletal impact of osteocyte-targeted Klotho deletion. Thus, our findings establish a key role of osteocyte-expressed Klotho in regulating bone metabolism and indicate a new mechanism by which osteocytes control bone formation.


Assuntos
Envelhecimento/metabolismo , Distúrbio Mineral e Ósseo na Doença Renal Crônica/patologia , Glucuronidase/metabolismo , Osteócitos/metabolismo , Osteogênese/fisiologia , Animais , Densidade Óssea , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Diferenciação Celular , Modelos Animais de Doenças , Regulação para Baixo , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/metabolismo , Glucuronidase/genética , Humanos , Imuno-Histoquímica , Proteínas Klotho , Camundongos , Camundongos Knockout , Osteoblastos/fisiologia , Osteoclastos/fisiologia , Cultura Primária de Células , Transdução de Sinais
12.
J Biol Chem ; 290(31): 18934-42, 2015 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-26085098

RESUMO

Apoptosis of osteocytes and osteoblasts precedes bone resorption and bone loss with reduced mechanical stimulation, and receptor activator of NF-κB ligand (RANKL) expression is increased with unloading in mice. Because osteocytes are major RANKL producers, we hypothesized that apoptotic osteocytes signal to neighboring osteocytes to increase RANKL expression, which, in turn, increases osteoclastogenesis and bone resorption. The traditional bisphosphonate (BP) alendronate (Aln) or IG9402, a BP analog that does not inhibit resorption, prevented the increase in osteocyte apoptosis and osteocytic RANKL expression. The BPs also inhibited osteoblast apoptosis but did not prevent the increase in osteoblastic RANKL. Unloaded mice exhibited high serum levels of the bone resorption marker C-telopeptide fragments of type I collagen (CTX), elevated osteoclastogenesis, and increased osteoclasts in bone. Aln, but not IG9402, prevented all of these effects. In addition, Aln prevented the reduction in spinal and femoral bone mineral density, spinal bone volume/tissue volume, trabecular thickness, mechanical strength, and material strength induced by unloading. Although IG9402 did not prevent the loss of bone mass, it partially prevented the loss of strength, suggesting a contribution of osteocyte viability to strength independent of bone mass. These results demonstrate that osteocyte apoptosis leads to increased osteocytic RANKL. However, blockade of these events is not sufficient to restrain osteoclast formation, inhibit resorption, or stop bone loss induced by skeletal unloading.


Assuntos
Apoptose/efeitos dos fármacos , Reabsorção Óssea/metabolismo , Colágeno Tipo I/farmacologia , Osteócitos/fisiologia , Peptídeos/farmacologia , Ligante RANK/metabolismo , Alendronato/farmacologia , Animais , Conservadores da Densidade Óssea/farmacologia , Células Cultivadas , Feminino , Elevação dos Membros Posteriores , Camundongos Endogâmicos C57BL , Osteócitos/efeitos dos fármacos
13.
J Biol Chem ; 288(12): 8168-8175, 2013 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-23362257

RESUMO

Osteocyte viability is a critical determinant of bone strength and is promoted by both mechanical stimulation and activation of the Wnt signaling pathway. Earlier studies demonstrated that both stimuli promote survival of osteocytes by activating the ERKs. Here, we show that there is interaction between the caveolin-1/ERK and Wnt/ß-catenin signaling pathways in the transduction of mechanical cues into osteocyte survival. Thus, ERK nuclear translocation and anti-apoptosis induced by mechanical stimulation are abolished by the Wnt antagonist Dkk1 and the ß-catenin degradation stimulator Axin2. Conversely, GSK3ß phosphorylation and ß-catenin accumulation induced by mechanical stimulation are abolished by either pharmacologic inhibition of ERKs or silencing caveolin-1. In contrast, the canonical Wnt signaling inhibitor dominant-negative T cell factor does not alter ERK nuclear translocation or survival induced by mechanical stimulation. These findings demonstrate that ß-catenin accumulation is an essential component of the mechanotransduction machinery in osteocytes, albeit ß-catenin/T cell factor-mediated transcription is not required. The simultaneous requirement of ß-catenin for ERK activation and of ERK activation for ß-catenin accumulation suggests a bidirectional crosstalk between the caveolin-1/ERK and Wnt/ß-catenin pathways in mechanotransduction leading to osteocyte survival.


Assuntos
Caveolina 1/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Mecanotransdução Celular , Osteócitos/metabolismo , beta Catenina/metabolismo , Animais , Caveolina 1/genética , Núcleo Celular/metabolismo , Sobrevivência Celular , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Flavonoides/farmacologia , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Camundongos , Osteócitos/fisiologia , Transporte Proteico , RNA Interferente Pequeno/genética , Receptor Cross-Talk , Fatores de Transcrição TCF/metabolismo , Via de Sinalização Wnt
14.
J Biol Chem ; 288(41): 29809-20, 2013 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-23963454

RESUMO

The contribution of remodeling-based bone formation coupled to osteoclast activity versus modeling-based bone formation that occurs independently of resorption, to the anabolic effect of PTH remains unclear. We addressed this question using transgenic mice with activated PTH receptor signaling in osteocytes that exhibit increased bone mass and remodeling, recognized skeletal effects of PTH elevation. Direct inhibition of bone formation was accomplished genetically by overexpressing the Wnt antagonist Sost/sclerostin; and resorption-dependent bone formation was inhibited pharmacologically with the bisphosphonate alendronate. We found that bone formation induced by osteocytic PTH receptor signaling on the periosteal surface depends on Wnt signaling but not on resorption. In contrast, bone formation on the endocortical surface results from a combination of Wnt-driven increased osteoblast number and resorption-dependent osteoblast activity. Moreover, elevated osteoclasts and intracortical/calvarial porosity is exacerbated by overexpressing Sost and reversed by blocking resorption. Furthermore, increased cancellous bone is abolished by Wnt inhibition but further increased by blocking resorption. Thus, resorption induced by PTH receptor signaling in osteocytes is critical for full anabolism in cortical bone, but tempers bone gain in cancellous bone. Dissecting underlying mechanisms of PTH receptor signaling would allow targeting actions in different bone compartments, enhancing the therapeutic potential of the pathway.


Assuntos
Reabsorção Óssea/metabolismo , Osso e Ossos/metabolismo , Osteócitos/metabolismo , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo , Transdução de Sinais , Absorciometria de Fóton , Proteínas Adaptadoras de Transdução de Sinal , Alendronato/administração & dosagem , Alendronato/farmacologia , Animais , Densidade Óssea/efeitos dos fármacos , Conservadores da Densidade Óssea/administração & dosagem , Conservadores da Densidade Óssea/farmacologia , Remodelação Óssea/efeitos dos fármacos , Remodelação Óssea/genética , Reabsorção Óssea/genética , Reabsorção Óssea/prevenção & controle , Osso e Ossos/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Glicoproteínas/genética , Glicoproteínas/metabolismo , Imuno-Histoquímica , Injeções Subcutâneas , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Osteócitos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Osteogênese/genética , Receptor Tipo 1 de Hormônio Paratireóideo/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Via de Sinalização Wnt/efeitos dos fármacos , Via de Sinalização Wnt/genética
15.
Blood ; 119(11): 2489-99, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22262765

RESUMO

Microenvironmental expansion of hematopoietic stem cells (HSCs) is induced by treatment with parathyroid hormone (PTH) or activation of the PTH receptor (PTH1R) in osteoblastic cells; however, the osteoblastic subset mediating this action of PTH is unknown. Osteocytes are terminally differentiated osteoblasts embedded in mineralized bone matrix but are connected with the BM. Activation of PTH1R in osteocytes increases osteoblastic number and bone mass. To establish whether osteocyte-mediated PTH1R signaling expands HSCs, we studied mice expressing a constitutively active PTH1R in osteocytes (TG mice). Osteoblasts, osteoclasts, and trabecular bone were increased in TG mice without changes in BM phenotypic HSCs or HSC function. TG mice had progressively increased trabecular bone but decreased HSC function. In severely affected TG mice, phenotypic HSCs were decreased in the BM but increased in the spleen. TG osteocytes had no increase in signals associated with microenvironmental HSC support, and the spindle-shaped osteoblastic cells that increased with PTH treatment were not present in TG bones. These findings demonstrate that activation of PTH1R signaling in osteocytes does not expand BM HSCs, which are instead decreased in TG mice. Therefore, osteocytes do not mediate the HSC expansion induced by PTH1R signaling. Further, osteoblastic expansion is not sufficient to increase HSCs.


Assuntos
Remodelação Óssea , Células-Tronco Hematopoéticas/citologia , Osteoblastos/citologia , Osteócitos/metabolismo , Receptor Tipo 1 de Hormônio Paratireóideo/fisiologia , Animais , Citometria de Fluxo , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Técnicas Imunoenzimáticas , Camundongos , Camundongos Transgênicos , Mutação/genética , Osteoblastos/metabolismo , Osteócitos/citologia , Hormônio Paratireóideo/metabolismo , Ratos , Transdução de Sinais
16.
Calcif Tissue Int ; 94(1): 25-34, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24002178

RESUMO

Osteocytes, the most abundant cells in bone, have been long postulated to detect and respond to mechanical and hormonal stimuli and to coordinate the function of osteoblasts and osteoclasts. The discovery that the inhibitor of bone formation sclerostin is primarily expressed in osteocytes in bone and downregulated by anabolic stimuli provided a mechanism by which osteocytes influence the activity of osteoblasts. Advances of the last few years provided experimental evidence demonstrating that osteocytes also participate in the recruitment of osteoclasts and the initiation of bone remodeling. Apoptotic osteocytes trigger yet-to-be-identified signals that attract osteoclast precursors to specific areas of bone, which in turn differentiate to mature, bone-resorbing osteoclasts. Osteocytes are also the source of molecules that regulate the generation and activity of osteoclasts, such as OPG and RANKL; and genetic manipulations of the mouse genome leading to loss or gain of function or to altered expression of either molecule in osteocytes markedly affect bone resorption. This review highlights these investigations and discusses how the novel concept of osteocyte-driven bone resorption and formation impacts our understanding of the mechanisms by which current therapies control bone remodeling.


Assuntos
Remodelação Óssea/fisiologia , Osso e Ossos/metabolismo , Osteócitos/metabolismo , Animais , Osso e Ossos/citologia , Diferenciação Celular/fisiologia , Humanos , Osteoclastos/metabolismo , Osteogênese/fisiologia
17.
Nat Rev Endocrinol ; 2024 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-39020007

RESUMO

Basic, translational and clinical research over the past few decades has provided new understanding on the mechanisms by which activation of the receptor of parathyroid hormone (parathyroid hormone 1 receptor (PTH1R)) regulates bone physiology and pathophysiology. A fundamental change in the field emerged upon the recognition that osteocytes, which are permanent residents of bone and the most abundant cells in bone, are targets of the actions of natural and synthetic ligands of PTH1R (parathyroid hormone and abaloparatide, respectively), and that these cells drive essential actions related to bone remodelling. Among the numerous genes regulated by PTH1R in osteocytes, SOST (which encodes sclerostin, the WNT signalling antagonist and inhibitor of bone formation) has a critical role in bone homeostasis and changes in its expression are associated with several bone pathologies. The bone fragility syndrome induced by diabetes mellitus is accompanied by increased osteocyte apoptosis and changes in the expression of osteocytic genes, including SOST. This Review will discuss advances in our knowledge of the role of osteocytes in PTH1R signalling and the new opportunities to restore bone health in diabetes mellitus by targeting the osteocytic PTH1R-sclerostin axis.

18.
Bone ; 181: 117042, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38360197

RESUMO

This study investigated the efficacy of the two FDA-approved bone anabolic ligands of the parathyroid hormone receptor 1 (PTH1R), teriparatide or human parathyroid hormone 1-34 (PTH) and abaloparatide (ABL), to restoring skeletal health using a preclinical murine model of streptozotocin-induced T1-DM. Intermittent daily subcutaneous injections of equal molar doses (12 pmoles/g/day) of PTH (50 ng/g/day), ABL (47.5 ng/g/day), or vehicle, were administered for 28 days to 5-month-old C57Bl/6 J male mice with established T1-DM or control (C) mice. ABL was superior to PTH in increasing or restoring bone mass in control or T1-MD mice, respectively, which was associated with superior stimulation of trabecular and periosteal bone formation, upregulation of osteoclastic/osteoblastic gene expression, and increased circulating bone remodeling markers. Only ABL corrected the reduction in ultimate load, which is a measure of bone strength, induced by T1-DM, and it also increased energy to ultimate load. In addition, bones from T1-DM mice treated with PTH or ABL exhibited increased ultimate stress, a material index, compared to T1-DM mice administered with vehicle. And both PTH and ABL prevented the increased expression of the Wnt antagonist Sost/sclerostin displayed by T1-DM mice. Further, PTH and ABL increased to a similar extent the circulating bone resorption marker CTX and the bone formation marker P1NP in T1-DM after 2 weeks of treatment; however, only ABL sustained these increases after 4 weeks of treatment. We conclude that at equal molar doses, ABL is more effective than PTH in increasing bone mass and restoring the cortical and trabecular bone lost with T1-DM, due to higher and longer-lasting increases in bone remodeling.


Assuntos
Diabetes Mellitus Tipo 1 , Teriparatida , Humanos , Camundongos , Masculino , Animais , Recém-Nascido , Teriparatida/farmacologia , Teriparatida/uso terapêutico , Diabetes Mellitus Tipo 1/tratamento farmacológico , Densidade Óssea/fisiologia , Proteína Relacionada ao Hormônio Paratireóideo/farmacologia , Hormônio Paratireóideo/farmacologia , Hormônio Paratireóideo/uso terapêutico
19.
J Periodontol ; 94(2): 277-289, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-35869905

RESUMO

BACKGROUND: Type 1 diabetes mellitus (T1DM) and periodontitis have long been thought to be biologically connected. Indeed, T1DM is a risk factor for periodontal disease. With the population of diabetic individuals growing, it is more important than ever to understand the negative consequences of diabetes on the periodontium and the mechanisms. The aim of this study was to find out the early effects of T1DM on the periodontium without any experimentally induced periodontitis. METHODS: We established the streptozotocin (STZ)-induced diabetic mouse model and examined the periodontium 8 weeks later by histology, molecular and cellular assays. Microcomputed tomographic (𝜇CT) imaging and in vivo fluorochrome labeling were also used to quantify bone volume and mineral apposition rates (MAR). RESULTS: The histologic appearance of epithelium tissue, connective tissue, and periodontal ligament in the diabetic condition was comparable with that of control mice. However, immune cell infiltration in the gingiva was dramatically elevated in the diabetic mice, which was accompanied by unmineralized connective tissue degeneration. Bone resorption activity was significantly increased in the diabetic mice, and quantitative 𝜇CT demonstrated the bone volume, the ratio of bone volume over tissue volume, and cemento-enamel junction to alveolar bone crest (CEJ-ABC) in the diabetic condition were equivalent to those in the control group. In vivo fluorochrome labeling revealed increased MAR and bone remodeling in the diabetic mice. Further investigation found the diabetic mice had more osteoprogenitors recruited to the periodontium, allowing more bone formation to balance the enhanced bone resorption. CONCLUSIONS: STZ-induced T1DM mice, at an early stage, have elevated gingival inflammation and soft tissue degeneration and increased bone resorption; but still the alveolar bone was preserved by recruiting more osteoprogenitor cells and increasing the rate of bone formation. We conclude that inflammation and periodontitis precede alveolar bone deterioration in diabetes.


Assuntos
Perda do Osso Alveolar , Reabsorção Óssea , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Gengivite , Periodontite , Camundongos , Animais , Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Experimental/complicações , Corantes Fluorescentes , Gengivite/complicações , Periodontite/complicações , Processo Alveolar , Inflamação , Perda do Osso Alveolar/etiologia
20.
Bioact Mater ; 21: 110-128, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36093329

RESUMO

Cell source is the key to decellularized matrix (DM) strategy. This study compared 3 cell types, osteocytes with/without dominant active Wnt/ß-catenin signaling (daCO and WTO) and bone marrow stromal cells (BMSCs) for their DMs in bone repair. Decellularization removes all organelles and >95% DNA, and retained >74% collagen and >71% GAG, maintains the integrity of cell basement membrane with dense boundaries showing oval and honeycomb structure in osteocytic DM and smooth but irregular shape in the BMSC-DM. DM produced higher cell survival rate (90%) and higher proliferative activity. In vitro, daCO-DM induces more and longer stress fibers in BMSCs, conducive to cell adhesion, spreading, and osteogenic differentiation. 8-wk after implantation of the critical-sized parietal bone defect model, daCO-DM formed tight structures, composed of a large number of densely-arranged type-I collagen under polarized light microscope, which is similar to and integrated with host bone. BV/TV (>54%) was 1.5, 2.9, and 3.5 times of WTO-DM, BMSC-DM, and none-DM groups, and N.Ob/T.Ar (3.2 × 102/mm2) was 1.7, 2.9, and 3.3 times. At 4-wk, daCO-DM induced osteoclastogenesis, 2.3 times higher than WTO-DM; but BMSC-DM or none-DM didn't. daCO-DM increased the expression of RANKL and MCSF, Vegfa and Angpt1, and Ngf in BMSCs, which contributes to osteoclastogenesis, angiogenesis, and neurogenesis, respectively. daCO-DM promoted H-type vessel formation and nerve markers ß3-tubulin and NeuN expression. Conclusion: daCO-DM produces metabolic and neurovascularized organoid bone to accelerate the repair of bone defects. These features are expected to achieve the effect of autologous bone transplantation, suitable for transformation application.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa