Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 123
Filtrar
1.
Nature ; 563(7730): 254-258, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30401834

RESUMO

Skeletal stem cells regulate bone growth and homeostasis by generating diverse cell types, including chondrocytes, osteoblasts and marrow stromal cells. The emerging concept postulates that there exists a distinct type of skeletal stem cell that is closely associated with the growth plate1-4, which is a type of cartilaginous tissue that has critical roles in bone elongation5. The resting zone maintains the growth plate by expressing parathyroid hormone-related protein (PTHrP), which interacts with Indian hedgehog (Ihh) that is released from the hypertrophic zone6-10, and provides a source of other chondrocytes11. However, the identity of skeletal stem cells and how they are maintained in the growth plate are unknown. Here we show, in a mouse model, that skeletal stem cells are formed among PTHrP-positive chondrocytes within the resting zone of the postnatal growth plate. PTHrP-positive chondrocytes expressed a panel of markers for skeletal stem and progenitor cells, and uniquely possessed the properties of skeletal stem cells in cultured conditions. Cell-lineage analysis revealed that PTHrP-positive chondrocytes in the resting zone continued to form columnar chondrocytes in the long term; these chondrocytes underwent hypertrophy, and became osteoblasts and marrow stromal cells beneath the growth plate. Transit-amplifying chondrocytes in the proliferating zone-which was concertedly maintained by a forward signal from undifferentiated cells (PTHrP) and a reverse signal from hypertrophic cells (Ihh)-provided instructive cues to maintain the cell fates of PTHrP-positive chondrocytes in the resting zone. Our findings unravel a type of somatic stem cell that is initially unipotent and acquires multipotency at the post-mitotic stage, underscoring the malleable nature of the skeletal cell lineage. This system provides a model in which functionally dedicated stem cells and their niches are specified postnatally, and maintained throughout tissue growth by a tight feedback regulation system.


Assuntos
Lâmina de Crescimento/citologia , Células-Tronco/citologia , Animais , Linhagem da Célula , Condrócitos/citologia , Condrócitos/metabolismo , Lâmina de Crescimento/metabolismo , Técnicas In Vitro , Camundongos , Osteoblastos/citologia , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Nicho de Células-Tronco , Células-Tronco/metabolismo , Células Estromais/citologia
2.
Annu Rev Cell Dev Biol ; 25: 629-48, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19575648

RESUMO

In the past few years, our molecular understanding of bone formation has continued to increase. This review aims to present a comprehensive view of the current state of knowledge in the field. Thus, it will cover our current knowledge of chondrogenesis and osteoblastogenesis. It will also cover the most salient aspects of osteoblast function.


Assuntos
Condrogênese , Regulação da Expressão Gênica no Desenvolvimento , Osteogênese , Animais , Humanos , Osteoblastos/metabolismo , Fatores de Transcrição/metabolismo
3.
Circ Res ; 126(10): 1363-1378, 2020 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-32160132

RESUMO

RATIONALE: The PTH1R (PTH [parathyroid hormone]/PTHrP [PTH-related protein] receptor) is expressed in vascular smooth muscle (VSM) and increased VSM PTH1R signaling mitigates diet-induced arteriosclerosis in LDLR-/- mice. OBJECTIVE: To study the impact of VSM PTH1R deficiency, we generated mice SM22-Cre:PTH1R(fl/fl);LDLR-/- mice (PTH1R-VKO) and Cre-negative controls. METHODS AND RESULTS: Immunofluorescence and Western blot confirmed PTH1R expression in arterial VSM that was reduced by Cre-mediated knockout. PTH1R-VKO cohorts exhibited increased aortic collagen accumulation in vivo, and VSM cultures from PTH1R-VKO mice elaborated more collagen (2.5-fold; P=0.01) with elevated Col3a1 and Col1a1 expression. To better understand these profibrotic responses, we performed mass spectrometry on nuclear proteins extracted from Cre-negative controls and PTH1R-VKO VSM. PTH1R deficiency reduced Gata6 but upregulated the MADS (MCM1, Agamous, Deficiens, and Srf DNA-binding domain)-box transcriptional co-regulator, Mkl-1 (megakaryoblastic leukemia [translocation] 1). Co-transfection assays (Col3a1 promoter-luciferase reporter) confirmed PTH1R-mediated inhibition and Mkl-1-mediated activation of Col3a1 transcription. Regulation mapped to a conserved hybrid CT(A/T)6GG MADS-box cognate in the Col3a1 promoter. Mutations of C/G in this motif markedly reduced Col3a1 transcriptional regulation by PTH1R and Mkl-1. Upregulation of Col3a1 and Col1a1 in PTH1R-VKO VSM was inhibited by small interfering RNA targeting Mkl1 and by treatment with the Mkl-1 antagonist CCG1423 or the Rock (Rho-associated coiled-coil containing protein kinase)-2 inhibitor KD025. Chromatin precipitation demonstrated that VSM PTH1R deficiency increased Mkl-1 binding to Col3a1 and Col1a1, but not TNF, promoters. Proteomic studies of plasma extracellular vesicles and VSM from PTH1R-VKO mice identified C1r (complement component 1, r) and C1s (complement component 1, s), complement proteins involved in vascular collagen metabolism, as potential biomarkers. VSM C1r protein and C1r message were increased with PTH1R deficiency, mediated by Mkl-1-dependent transcription and inhibited by CCG1423 or KD025. CONCLUSIONS: PTH1R signaling restricts collagen production in the VSM lineage, in part, via Mkl-1 regulatory circuits that control collagen gene transcription. Strategies that maintain homeostatic VSM PTH1R signaling, as reflected in extracellular vesicle biomarkers of VSM PTH1R/Mkl-1 action, may help mitigate arteriosclerosis and vascular fibrosis.


Assuntos
Aterosclerose/metabolismo , Colágeno Tipo I/metabolismo , Diabetes Mellitus/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Hormônio Paratireóideo/metabolismo , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo , Transativadores/metabolismo , Animais , Aorta/metabolismo , Aorta/patologia , Aterosclerose/genética , Aterosclerose/patologia , Células Cultivadas , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I , Colágeno Tipo III/genética , Colágeno Tipo III/metabolismo , Diabetes Mellitus/genética , Diabetes Mellitus/patologia , Dieta Hiperlipídica , Modelos Animais de Doenças , Fibrose , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Ratos , Receptor Tipo 1 de Hormônio Paratireóideo/deficiência , Receptor Tipo 1 de Hormônio Paratireóideo/genética , Receptores de LDL/deficiência , Receptores de LDL/genética , Transdução de Sinais , Transativadores/genética , Transcrição Gênica , Remodelação Vascular
4.
Proc Natl Acad Sci U S A ; 116(2): 575-580, 2019 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-30509999

RESUMO

Formation of functional skeletal tissues requires highly organized steps of mesenchymal progenitor cell differentiation. The dental follicle (DF) surrounding the developing tooth harbors mesenchymal progenitor cells for various differentiated cells constituting the tooth root-bone interface and coordinates tooth eruption in a manner dependent on signaling by parathyroid hormone-related peptide (PTHrP) and the PTH/PTHrP receptor (PPR). However, the identity of mesenchymal progenitor cells in the DF and how they are regulated by PTHrP-PPR signaling remain unknown. Here, we show that the PTHrP-PPR autocrine signal maintains physiological cell fates of DF mesenchymal progenitor cells to establish the functional periodontal attachment apparatus and orchestrates tooth eruption. A single-cell RNA-seq analysis revealed cellular heterogeneity of PTHrP+ cells, wherein PTHrP+ DF subpopulations abundantly express PPR. Cell lineage analysis using tamoxifen-inducible PTHrP-creER mice revealed that PTHrP+ DF cells differentiate into cementoblasts on the acellular cementum, periodontal ligament cells, and alveolar cryptal bone osteoblasts during tooth root formation. PPR deficiency induced a cell fate shift of PTHrP+ DF mesenchymal progenitor cells to nonphysiological cementoblast-like cells precociously forming the cellular cementum on the root surface associated with up-regulation of Mef2c and matrix proteins, resulting in loss of the proper periodontal attachment apparatus and primary failure of tooth eruption, closely resembling human genetic conditions caused by PPR mutations. These findings reveal a unique mechanism whereby proper cell fates of mesenchymal progenitor cells are tightly maintained by an autocrine system mediated by PTHrP-PPR signaling to achieve functional formation of skeletal tissues.


Assuntos
Comunicação Autócrina/fisiologia , Células-Tronco Mesenquimais/metabolismo , Proteína Relacionada ao Hormônio Paratireóideo/genética , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo , Transdução de Sinais/fisiologia , Erupção Dentária/fisiologia , Animais , Diferenciação Celular/fisiologia , Saco Dentário/citologia , Saco Dentário/metabolismo , Humanos , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Transgênicos , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Receptor Tipo 1 de Hormônio Paratireóideo/genética
6.
Zhonghua Yi Xue Za Zhi ; 98(10): 781-787, 2018 Mar 13.
Artigo em Chinês | MEDLINE | ID: mdl-29562406

RESUMO

Objective: To investigate the distinct effects of intermittent and continuous administration of parathyroid hormone (PTH) on bone and mesenchymal stem cell (MSC). Methods: Six weeks old mice with C57/BL6J background and SOX9-creERT/Td-tomato/Osteocalcin-GFP genotype were divided into 6 groups: intermittent administration and withdraw group (subcutaneous injection with PTH 500 µg·kg-1·d-1), continuous administration and withdraw group (subcutaneous implantation of PTH pump, 80 µg·kg-1·d-1, with a rate of 0.25 µl/h), control administration and withdraw group, with 8 mice in each group. Serum calcium level and bone mineral density (BMD) were measured after two weeks' treatment and two weeks after drug withdraw. Histopathology and immunofluorescence analyses were performed to assess the effects of PTH on bone and mesenchymal stem cell. Results: Serum calcium level increased transiently in intermittent group[(1.36±0.03) mmol/L]and increased gradually in continuous group[up to (2.33±0.03) mmol/L], but reduced to normal level (1.12-1.27 mmol/L) 14 days after drug withdraw. BMD of both intermittent[(0.047±0.002) g/cm2]and continuous[(0.046±0.001) g/cm2]PTH administration groups increased compared with control group[(0.044±0.001) g/cm2], but there was no significant difference among three groups 2 weeks after drug withdraw. Femoral histopathology showed that bone mass, trabecular number and little fibrous tissue hyperplasia in intermittent PTH group increased. Osteoblasts number increased, but lining cells decreased. There was no significant difference in osteocyte and osteoclast numbers. After withdrawing of intermittent PTH, osteocyte and osteoblast number declined significantly, but there was an increased number of lining cells. Continuous PTH caused very high amount of fibrosis, and osteoclast number increased significantly, while osteoblast and osteocyte number increased slightly. After withdrawing of continuous PTH, fibrosis disappeared significantly, and lining cell number increased. Immunofluorescence staining in the epiphyseal-metaphyseal regions in fibula showed intermittent PTH increased undifferentiated Td-Tomato MSC, but declined significantly after withdrawing. Undifferentiated Td-Tomato MSC in continuous PTH increased slightly and decreased after drug withdraw. Conclusions: Intermittent PTH increased undifferentiated Td-Tomato MSC and osteoblast number, and might transform lining cell into osteocytes and osteoblasts, and thus lead to bone formation. Continuous PTH increased undifferentiated Td/Tomato MSC, osteoblast and osteocyte number slightly, but high amount of fibrosis and osteoclasts can be seen, leading to metabolic bone disease. However, lining cell ascended after drug withdraw, which may be beneficial to bone remodeling.


Assuntos
Células-Tronco Mesenquimais , Animais , Densidade Óssea , Osso e Ossos , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos , Osteócitos , Hormônio Paratireóideo
7.
J Biol Chem ; 291(4): 1631-1642, 2016 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-26598522

RESUMO

Parathyroid hormone (PTH) is an important regulator of osteoblast function and is the only anabolic therapy currently approved for treatment of osteoporosis. The PTH receptor (PTH1R) is a G protein-coupled receptor that signals via multiple G proteins including Gsα. Mice expressing a constitutively active mutant PTH1R exhibited a dramatic increase in trabecular bone that was dependent upon expression of Gsα in the osteoblast lineage. Postnatal removal of Gsα in the osteoblast lineage (P-Gsα(OsxKO) mice) yielded markedly reduced trabecular and cortical bone mass. Treatment with anabolic PTH(1-34) (80 µg/kg/day) for 4 weeks failed to increase trabecular bone volume or cortical thickness in male and female P-Gsα(OsxKO) mice. Surprisingly, in both male and female mice, PTH administration significantly increased osteoblast numbers and bone formation rate in both control and P-Gsα(OsxKO) mice. In mice that express a mutated PTH1R that activates adenylyl cyclase and protein kinase A (PKA) via Gsα but not phospholipase C via Gq/11 (D/D mice), PTH significantly enhanced bone formation, indicating that phospholipase C activation is not required for increased bone turnover in response to PTH. Therefore, although the anabolic effect of intermittent PTH treatment on trabecular bone volume is blunted by deletion of Gsα in osteoblasts, PTH can stimulate osteoblast differentiation and bone formation. Together these findings suggest that alternative signaling pathways beyond Gsα and Gq/11 act downstream of PTH on osteoblast differentiation.


Assuntos
Anabolizantes/administração & dosagem , Desenvolvimento Ósseo/efeitos dos fármacos , Subunidades alfa Gs de Proteínas de Ligação ao GTP/deficiência , Terapia de Reposição Hormonal , Osteoporose/tratamento farmacológico , Osteoporose/enzimologia , Hormônio Paratireóideo/administração & dosagem , Animais , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Feminino , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Humanos , Masculino , Camundongos , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/enzimologia , Osteoblastos/metabolismo , Osteoporose/metabolismo , Osteoporose/fisiopatologia , Receptor Tipo 1 de Hormônio Paratireóideo/genética , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo
8.
J Biol Chem ; 290(27): 16744-58, 2015 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-25953900

RESUMO

Although bone responds to its mechanical environment, the cellular and molecular mechanisms underlying the response of the skeleton to mechanical unloading are not completely understood. Osteocytes are the most abundant but least understood cells in bones and are thought to be responsible for sensing stresses and strains in bone. Sclerostin, a product of the SOST gene, is produced postnatally primarily by osteocytes and is a negative regulator of bone formation. Recent studies show that SOST is mechanically regulated at both the mRNA and protein levels. During prolonged bed rest and immobilization, circulating sclerostin increases both in humans and in animal models, and its increase is associated with a decrease in parathyroid hormone. To investigate whether SOST/sclerostin up-regulation in mechanical unloading is a cell-autonomous response or a hormonal response to decreased parathyroid hormone levels, we subjected osteocytes to an in vitro unloading environment achieved by the NASA rotating wall vessel system. To perform these studies, we generated a novel osteocytic cell line (Ocy454) that produces high levels of SOST/sclerostin at early time points and in the absence of differentiation factors. Importantly, these osteocytes recapitulated the in vivo response to mechanical unloading with increased expression of SOST (3.4 ± 1.9-fold, p < 0.001), sclerostin (4.7 ± 0.1-fold, p < 0.001), and the receptor activator of nuclear factor κΒ ligand (RANKL)/osteoprotegerin (OPG) (2.5 ± 0.7-fold, p < 0.001) ratio. These data demonstrate for the first time a cell-autonomous increase in SOST/sclerostin and RANKL/OPG ratio in the setting of unloading. Thus, targeted osteocyte therapies could hold promise as novel osteoporosis and disuse-induced bone loss treatments by directly modulating the mechanosensing cells in bone.


Assuntos
Glicoproteínas/genética , Osteócitos/metabolismo , Regulação para Cima , Proteínas Wnt/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Fenômenos Biomecânicos , Linhagem Celular , Glicoproteínas/metabolismo , Gravitação , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Osteócitos/química , Ligante RANK/genética , Ligante RANK/metabolismo , Proteínas Wnt/genética
9.
Blood ; 121(6): 930-9, 2013 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-23160461

RESUMO

Hematopoietic progenitors are regulated in their respective niches by cells of the bone marrow microenvironment. The bone marrow microenvironment is composed of a variety of cell types, and the relative contribution of each of these cells for hematopoietic lineage maintenance has remained largely unclear. Osteocytes, the most abundant yet least understood cells in bone, are thought to initiate adaptive bone remodeling responses via osteoblasts and osteoclasts. Here we report that these cells regulate hematopoiesis, constraining myelopoiesis through a Gsα-mediated mechanism that affects G-CSF production. Mice lacking Gsα in osteocytes showed a dramatic increase in myeloid cells in bone marrow, spleen, and peripheral blood. This hematopoietic phenomenon was neither intrinsic to the hematopoietic cells nor dependent on osteoblasts but was a consequence of an altered bone marrow microenvironment imposed by Gsα deficiency in osteocytes. Conditioned media from osteocyte-enriched bone explants significantly increased myeloid colony formation in vitro, which was blocked by G-CSF­neutralizing antibody, indicating a critical role of osteocyte-derived G-CSF in the myeloid expansion.


Assuntos
Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Mielopoese , Osteócitos/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal , Animais , Doenças Ósseas Metabólicas/genética , Doenças Ósseas Metabólicas/metabolismo , Células da Medula Óssea/metabolismo , Proliferação de Células , Células Cultivadas , Microambiente Celular/genética , Feminino , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Expressão Gênica , Glicoproteínas/genética , Glicoproteínas/metabolismo , Fator Estimulador de Colônias de Granulócitos/genética , Fator Estimulador de Colônias de Granulócitos/metabolismo , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intercelular , Masculino , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Varredura , Células Mieloides/metabolismo , Osteócitos/citologia , Osteócitos/ultraestrutura , Receptor Tipo 1 de Hormônio Paratireóideo/genética , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/citologia , Baço/metabolismo
10.
Nature ; 459(7243): 103-7, 2009 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-19322176

RESUMO

Haematopoietic stem and progenitor cells (HSPCs) change location during development and circulate in mammals throughout life, moving into and out of the bloodstream to engage bone marrow niches in sequential steps of homing, engraftment and retention. Here we show that HSPC engraftment of bone marrow in fetal development is dependent on the guanine-nucleotide-binding protein stimulatory alpha subunit (Galpha(s)). HSPCs from adult mice deficient in Galpha(s) (Galpha(s)(-/-)) differentiate and undergo chemotaxis, but also do not home to or engraft in the bone marrow in adult mice and demonstrate a marked inability to engage the marrow microvasculature. If deleted after engraftment, Galpha(s) deficiency did not lead to lack of retention in the marrow, rather cytokine-induced mobilization into the blood was impaired. Testing whether activation of Galpha(s) affects HSPCs, pharmacological activators enhanced homing and engraftment in vivo. Galpha(s) governs specific aspects of HSPC localization under physiological conditions in vivo and may be pharmacologically targeted to improve transplantation efficiency.


Assuntos
Medula Óssea/fisiologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Transdução de Sinais/fisiologia , Adjuvantes Imunológicos/farmacologia , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/embriologia , Transplante de Medula Óssea/fisiologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Toxina da Cólera/farmacologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Fator Estimulador de Colônias de Granulócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
11.
Proc Natl Acad Sci U S A ; 109(19): 7433-8, 2012 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-22538810

RESUMO

Parathyroid hormone (PTH), the major calcium-regulating hormone, and norepinephrine (NE), the principal neurotransmitter of sympathetic nerves, regulate bone remodeling by activating distinct cell-surface G protein-coupled receptors in osteoblasts: the parathyroid hormone type 1 receptor (PTHR) and the ß(2)-adrenergic receptor (ß(2)AR), respectively. These receptors activate a common cAMP/PKA signal transduction pathway mediated through the stimulatory heterotrimeric G protein. Activation of ß(2)AR via the sympathetic nervous system decreases bone formation and increases bone resorption. Conversely, daily injection of PTH (1-34), a regimen known as intermittent (i)PTH treatment, increases bone mass through the stimulation of trabecular and cortical bone formation and decreases fracture incidences in severe cases of osteoporosis. Here, we show that iPTH has no osteoanabolic activity in mice lacking the ß(2)AR. ß(2)AR deficiency suppressed both iPTH-induced increase in bone formation and resorption. We showed that the lack of ß(2)AR blocks expression of iPTH-target genes involved in bone formation and resorption that are regulated by the cAMP/PKA pathway. These data implicate an unexpected functional interaction between PTHR and ß(2)AR, two G protein-coupled receptors from distinct families, which control bone formation and PTH anabolism.


Assuntos
Osso e Ossos/efeitos dos fármacos , Hormônio Paratireóideo/farmacologia , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Absorciometria de Fóton , Anabolizantes/metabolismo , Anabolizantes/farmacologia , Animais , Densidade Óssea/efeitos dos fármacos , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/metabolismo , Feminino , Fêmur/efeitos dos fármacos , Fêmur/metabolismo , Fluoresceínas , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Osteogênese/efeitos dos fármacos , Osteogênese/genética , Hormônio Paratireóideo/metabolismo , Receptor Tipo 1 de Hormônio Paratireóideo/genética , Receptores Adrenérgicos beta 2/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Microtomografia por Raio-X
12.
Proc Natl Acad Sci U S A ; 108(1): 191-6, 2011 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-21173257

RESUMO

Parathyroid hormone (PTH)-related protein (PTHrP), regulated by Indian hedgehog and acting through the PTH/PTHrP receptor (PPR), is crucial for normal cartilage development. These observations suggest a possible role of PPR signaling in the postnatal growth plate; however, the role of PPR signaling in postnatal chondrocytes is unknown. In this study, we have generated tamoxifen-inducible and cartilage-specific PPR KO mice to evaluate the physiological role of PPR signaling in postnatal chondrocytes. We found that inactivation of the PPR in chondrocytes postnatally leads to accelerated differentiation of chondrocytes, followed by disappearance of the growth plate. We also observed an increase of TUNEL-positive cells and activities of caspase-3 and caspase-9 in the growth plate, along with a decrease in phosphorylation of Bad at Ser155 in postnatal PPR KO mice. Administration of a low-phosphate diet, which prevents apoptosis of chondrocytes, prevented the disappearance of the growth plate. Taken together, these observations suggest that the major consequences of PPR activation are similar in both the fetal and postnatal growth plates. Moreover, chondrocyte apoptosis through the activation of a mitochondrial pathway may be involved in the process of premature disappearance of the growth plate by postnatal inactivation of the PPR in chondrocytes.


Assuntos
Lâmina de Crescimento/crescimento & desenvolvimento , Hormônio Paratireóideo/metabolismo , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo , Transdução de Sinais/fisiologia , Animais , Western Blotting , Bromodesoxiuridina , Caspase 3/metabolismo , Caspase 9/metabolismo , Condrócitos/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Knockout , Receptor Tipo 1 de Hormônio Paratireóideo/genética
13.
Am J Pathol ; 180(2): 811-8, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22155108

RESUMO

Bone marrow (BM) fibrosis is a feature of severe hyperparathyroidism. Consistent with this observation, mice expressing constitutively active parathyroid hormone (PTH)/PTH-related peptide receptors (PPR) in osteoblasts (PPR*Tg) display BM fibrosis. To obtain insight into the nature of BM fibrosis in such a model, a double-mutant mouse expressing constitutively active PPR and green fluorescent protein (GFP) under the control of the type I collagen promoter (PPR*Tg/GFP) was generated. Confocal microscopy and flow cytometry revealed the presence of a cell population expressing GFP (GFP(+)) that was also positive for the hematopoietic marker CD45 in the BM of both PPR*Tg/GFP and control animals. This cell population was expanded in PPR*Tg/GFP. The existence of cells expressing both type I collagen and CD45 in the adult BM was confirmed by IHC and fluorescence-activated cell sorting. An analysis of total RNA extracted from sorted GFP(+)CD45(+) cells showed that these cells produced type I collagen and PTH/PTH-related peptide receptor and receptor activator for NF-κB mRNAs, further supporting their features of being both mesenchymal and hematopoietic lineages. Similar cells, known as fibrocytes, are also present in pathological fibroses. Our findings, thus, indicate that the BM is a permissive microenvironment for the differentiation of fibrocyte-like cells and raise the possibility that these cells could contribute to the pathogenesis of BM fibrosis.


Assuntos
Biomarcadores/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Mielofibrose Primária/patologia , Animais , Medula Óssea/metabolismo , Diferenciação Celular , Colágeno Tipo I , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Transgênicos , Osteoblastos/metabolismo , Hormônio Paratireóideo/farmacologia , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Mielofibrose Primária/metabolismo , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo
14.
Cell Genom ; 3(5): 100299, 2023 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-37228756

RESUMO

Alterations in the growth and maturation of chondrocytes can lead to variation in human height, including monogenic disorders of skeletal growth. We aimed to identify genes and pathways relevant to human growth by pairing human height genome-wide association studies (GWASs) with genome-wide knockout (KO) screens of growth-plate chondrocyte proliferation and maturation in vitro. We identified 145 genes that alter chondrocyte proliferation and maturation at early and/or late time points in culture, with 90% of genes validating in secondary screening. These genes are enriched in monogenic growth disorder genes and in KEGG pathways critical for skeletal growth and endochondral ossification. Further, common variants near these genes capture height heritability independent of genes computationally prioritized from GWASs. Our study emphasizes the value of functional studies in biologically relevant tissues as orthogonal datasets to refine likely causal genes from GWASs and implicates new genetic regulators of chondrocyte proliferation and maturation.

15.
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
16.
J Cell Physiol ; 227(2): 408-15, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21866553

RESUMO

Bone is maintained by continuous bone formation by osteoblasts provided by proliferation and differentiation of osteoprogenitors. Parathyroid hormone (PTH) activates bone formation, but because of the complexity of cells in the osteoblast lineage, how these osteoprogenitors are regulated by PTH in vivo is incompletely understood. To elucidate how signals by PTH in differentiated osteoblasts regulate osteoprogenitors in vivo, we conducted bone marrow ablation using Col1a1-constitutively active PTH/PTHrP receptor (caPPR) transgenic mice. These mice express caPPR specifically in osteoblasts by using 2.3 kb Col1a1 promoter and showed higher trabecular bone volume under steady-state conditions. In contrast, after bone marrow ablation, stromal cells recruited from bone surface extensively proliferated in the marrow cavity in transgenic mice, compared to limited proliferation in wild-type mice. Whereas de novo bone formation was restricted to the ablated area in wild-type mice, the entire marrow cavity, including not only ablated area but also outside the ablated area, was filled with newly formed bone in transgenic mice. Bone mineral density was significantly increased after ablation in transgenic mice. Bone marrow cell culture in osteogenic medium revealed that alkaline phosphatase-positive area was markedly increased in the cells obtained from transgenic mice. Furthermore, mRNA expression of Wnt-signaling molecules such as LRP5, Wnt7b, and Wnt10b were upregulated after marrow ablation in bone marrow cells of transgenic mice. These results indicate that constitutive activation of PTH/PTHrP receptor in differentiated osteoblasts enhances bone marrow ablation-induced recruitment, proliferation, and differentiation of osteoprogenitors.


Assuntos
Medula Óssea/fisiologia , Regulação da Expressão Gênica/fisiologia , Osteoblastos/metabolismo , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo , Animais , Desenvolvimento Ósseo/fisiologia , Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Proliferação de Células , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Camundongos , Camundongos Transgênicos , Receptor Tipo 1 de Hormônio Paratireóideo/classificação , Receptor Tipo 1 de Hormônio Paratireóideo/genética , Células Estromais/citologia , Células Estromais/fisiologia
17.
J Cell Biol ; 177(3): 451-64, 2007 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-17470636

RESUMO

Recent evidence suggests that low oxygen tension (hypoxia) may control fetal development and differentiation. A crucial mediator of the adaptive response of cells to hypoxia is the transcription factor Hif-1alpha. In this study, we provide evidence that mesenchymal condensations that give origin to endochondral bones are hypoxic during fetal development, and we demonstrate that Hif-1alpha is expressed and transcriptionally active in limb bud mesenchyme and in mesenchymal condensations. To investigate the role of Hif-1alpha in mesenchymal condensations and in early chondrogenesis, we conditionally inactivated Hif-1alpha in limb bud mesenchyme using a Prx1 promoter-driven Cre transgenic mouse. Conditional knockout of Hif-1alpha in limb bud mesenchyme does not impair mesenchyme condensation, but alters the formation of the cartilaginous primordia. Late hypertrophic differentiation is also affected as a result of the delay in early chondrogenesis. In addition, mutant mice show a striking impairment of joint development. Our study demonstrates a crucial, and previously unrecognized, role of Hif-1alpha in early chondrogenesis and joint formation.


Assuntos
Condrogênese/fisiologia , Membro Posterior/embriologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Articulações/embriologia , Mesoderma/metabolismo , Animais , Hipóxia Celular/fisiologia , Feminino , Membro Posterior/citologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/deficiência , Articulações/citologia , Mesoderma/citologia , Camundongos , Camundongos Knockout , Gravidez
18.
Endocrinology ; 163(7)2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35460406

RESUMO

Rodent models are commonly used to evaluate parathyroid hormone (PTH) and PTH-related protein (PTHrP) ligands and analogues for their pharmacologic activities and potential therapeutic utility toward diseases of bone and mineral ion metabolism. Divergence, however, in the amino acid sequences of rodent and human PTH receptors (rat and mouse PTH1Rs are 91% identical to the human PTH1R) can lead to differences in receptor-binding and signaling potencies for such ligands when assessed on rodent vs human PTH1Rs, as shown by cell-based assays in vitro. This introduces an element of uncertainty in the accuracy of rodent models for performing such preclinical evaluations. To overcome this potential uncertainty, we used a homologous recombination-based knockin (KI) approach to generate a mouse (in-host strain C57Bl/6N) in which complementary DNA encoding the human PTH1R replaces a segment (exon 4) of the murine PTH1R gene so that the human and not the mouse PTH1R protein is expressed. Expression is directed by the endogenous mouse promoter and hence occurs in all biologically relevant cells and tissues and at appropriate levels. The resulting homozygous hPTH1R-KI (humanized) mice were healthy over at least 10 generations and showed functional responses to injected PTH analog peptides that are consistent with a fully functional human PTH1R in target bone and kidney cells. The initial evaluation of these mice and their potential utility for predicting behavior of PTH analogues in humans is reported here.


Assuntos
Proteína Relacionada ao Hormônio Paratireóideo , Hormônio Paratireóideo , Receptor Tipo 1 de Hormônio Paratireóideo , Sequência de Aminoácidos , Animais , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Hormônio Paratireóideo/genética , Hormônio Paratireóideo/metabolismo , Proteína Relacionada ao Hormônio Paratireóideo/genética , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Ratos , Receptor Tipo 1 de Hormônio Paratireóideo/genética , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo , Receptores de Hormônios Paratireóideos/genética , Receptores de Hormônios Paratireóideos/metabolismo , Transdução de Sinais
19.
J Clin Endocrinol Metab ; 108(1): 191-197, 2022 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-36056816

RESUMO

CONTEXT: Fragility fractures increase risks for future fractures, morbidity, and mortality. Available pharmacotherapy for underlying osteoporosis is safe and effective but underused. OBJECTIVE: To improve pharmacotherapy rate representing secondary prevention of osteoporotic fractures. METHODS: This single-center, observational, follow-up study included patients with fragility fractures admitted to the Massachusetts General Hospital between February 2016 and December 2019. For patients admitted to the orthopedics service with fragility fracture, the Massachusetts General Hospital Fracture Liaison Service (FLS) was systematically consulted. Initial outpatient follow-up with FLS was established in conjunction with the orthopedic postoperative follow-up visit. Patients at risk for failing timely outpatient follow-up were administered zoledronic acid (ZA) during the index fracture hospitalization. The main outcome measures were percentage of patients with fragility fracture(s) started on pharmacotherapy for osteoporosis and average length of stay and 30-day readmission rate of patients treated with ZA. RESULTS: Compared with baseline (8-11%) and reference (5-20%) rates, integration of FLS to the orthopedics service, along with appropriate inpatient administration of ZA, increased the pharmacotherapy rate to 70% (412/589) among eligible patients with verified treatment status. Inpatient ZA administration neither affected the average length of stay nor 30-day readmission rate. Treatment status of 37.9% (471/1240) of the study patients remained unknown due to lack of or unknown follow-up. CONCLUSION: Integration of a FLS and orthopedics services along with inpatient ZA administration improved the osteoporosis pharmacotherapy rate among patients with fragility fracture(s) who often had obstacles for outpatient follow-up.


Assuntos
Conservadores da Densidade Óssea , Ortopedia , Osteoporose , Fraturas por Osteoporose , Humanos , Ácido Zoledrônico/uso terapêutico , Conservadores da Densidade Óssea/uso terapêutico , Pacientes Internados , Seguimentos , Fraturas por Osteoporose/prevenção & controle , Fraturas por Osteoporose/tratamento farmacológico , Osteoporose/complicações , Osteoporose/tratamento farmacológico , Prevenção Secundária
20.
Front Neurol ; 13: 855157, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35370900

RESUMO

Patients with fibrous dysplasia (FD) often present with craniofacial lesions that affect the trigeminal nerve system. Debilitating pain, headache, and migraine are frequently experienced by FD patients with poor prognosis, while some individuals with similar bone lesions are asymptomatic. The clinical and biological factors that contribute to the etiopathogenesis of pain in craniofacial FD are largely unknown. We present two adult females with comparable craniofacial FD lesion size and location, as measured by 18F-sodium fluoride positron emission tomography/computed tomography (PET/CT), yet their respective pain phenotypes differed significantly. Over 4 weeks, the average pain reported by Patient A was 0.4/0-10 scale. Patient B reported average pain of 7.8/0-10 scale distributed across the entire skull and left facial region. Patient B did not experience pain relief from analgesics or more aggressive treatments (denosumab). In both patients, evaluation of trigeminal nerve divisions (V1, V2, and V3) with CT and magnetic resonance imaging (MRI) revealed nerve compression and displacement with more involvement of the left trigeminal branches relative to the right. First-time employment of diffusion MRI and tractography suggested reduced apparent fiber density within the cisternal segment of the trigeminal nerve, particularly for Patient B and in the left hemisphere. These cases highlight heterogeneous clinical presentation and neurobiological properties in craniofacial FD and also, the disconnect between peripheral pathology and pain severity. We hypothesize that a detailed phenotypic characterization of patients that incorporates an advanced imaging approach probing the trigeminal system may provide enhanced insights into the variable experiences with pain in craniofacial FD.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa