Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Curr Osteoporos Rep ; 17(1): 26-35, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30725321

RESUMO

PURPOSE OF REVIEW: We reviewed recent literature on oxygen sensing in osteogenic cells and its contribution to development of a skeletal phenotype, the coupling of osteogenesis with angiogenesis and integration of hypoxia into canonical Wnt signaling, and opportunities to manipulate oxygen sensing to promote skeletal repair. RECENT FINDINGS: Oxygen sensing in osteocytes can confer a high bone mass phenotype in murine models; common and unique targets of HIF-1α and HIF-2α and lineage-specific deletion of oxygen sensing machinery suggest differentia utilization and requirement of HIF-α proteins in the differentiation from mesenchymal stem cell to osteoblast to osteocyte; oxygen-dependent but HIF-α-independent signaling may contribute to observed skeletal phenotypes. Manipulating oxygen sensing machinery in osteogenic cells influences skeletal phenotype through angiogenesis-dependent and angiogenesis-independent pathways and involves HIF-1α, HIF-2α, or both proteins. Clinically, an FDA-approved iron chelator promotes angiogenesis and osteogenesis, thereby enhancing the rate of fracture repair.


Assuntos
Osso e Ossos/metabolismo , Hipóxia/metabolismo , Osteócitos/metabolismo , Osteogênese/fisiologia , Oxigênio/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Densidade Óssea/fisiologia , Regeneração Óssea/fisiologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Neovascularização Fisiológica/fisiologia , Via de Sinalização Wnt/fisiologia
2.
BMC Vet Res ; 10: 173, 2014 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-25088159

RESUMO

BACKGROUND: The intermediate filament protein nestin is used as a marker for neural stem cells, and its expression is inversely correlated with cellular differentiation. More recently, nestin expression has also been described in other cell types including multipotential mesenchymal stromal cells (MSCs). In this study, we examined the expression of nestin in equine, canine and human bone marrow-derived MSCs undergoing osteogenic differentiation, to determine whether nestin levels were attenuated as the cells acquired a more mature phenotype. In addition, the expression of nestin may be under the influence of cellular hypoxia, as nestin expression is known to increase in areas of ischemic tissue damage. Therefore, we also examined the effects of hypoxia on expression of nestin in human MSCs and examined a role for hypoxia inducible factor 1-alpha (HIF-1α) and vascular endothelial growth factor (VEGF) in the response. Additionally, we quantified the temporal expression of nestin in the fracture callus during bone regeneration, a site that has been characterized as hypoxic. RESULTS: There were no significant changes in nestin expression in MSCs during osteogenic differentiation. There was a significant increase in expression of nestin mRNA and protein in human MSCs in response to hypoxia (1% O2) or the chemical hypoxia mimetic desferroxamine. This may be due to upregulation of VEGF under hypoxia, as treatment of cells with the VEGF receptor antagonist CPO-P11 attenuated hypoxia-induced nestin expression. A significant increase in nestin mRNA expression was observed in the fracture callus of mice three and seven days post fracture. CONCLUSIONS: Nestin was not a selective marker for MSCs, as its expression was maintained during osteogenic differentiation, in all species examined. Furthermore our data suggest that nestin expression can be induced by hypoxia, and that this increase in nestin is partially regulated by HIF-1α and VEGF. Interestingly, nestin levels were significantly upregulated at the fracture site. Further studies are required to understand the role of nestin in bone cell biology and ultimately bone regeneration.


Assuntos
Células da Medula Óssea/metabolismo , Regulação da Expressão Gênica/fisiologia , Células-Tronco Mesenquimais/metabolismo , Nestina/metabolismo , Osteogênese/fisiologia , Oxigênio/farmacologia , Animais , Células Cultivadas , Consolidação da Fratura/fisiologia , Fraturas Ósseas/metabolismo , Humanos , Nestina/genética , Especificidade da Espécie
3.
J Orthop Res ; 42(5): 1033-1044, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38044472

RESUMO

High fat diets overwhelm the physiological mechanisms for absorption, storage, and utilization of triglycerides (TG); consequently TG, TG-rich lipoproteins (TGRL), and TGRL remnants accumulate, circulate systemically, producing dyslipidemia. This associates with, or is causative for increased atherosclerotic cardiovascular risk, ischemic stroke, fatty liver disease, and pancreatitis. TGRL hydrolysis by endothelial surface-bound lipoprotein lipase (LPL) generates metabolites like free fatty acids which have proinflammatory properties. While osteoblasts utilize fatty acids as an energy source, dyslipidemia is associated with negative effects on the skeleton. In this study we investigated the effects of TGRL lipolysis products (TGRL-LP) on expression of a stress responsive transcription factor, termed activating transcription factor 3 (ATF3), reactive oxygen species (ROS), ATF3 target genes, and angiopoietin-like 4 (Angptl4) in osteoblasts. As ATF3 negatively associates with osteoblast differentiation, we also investigated the skeletal effects of global ATF3 deletion in mice. TGRL-LP increased expression of Atf3, proinflammatory proteins Ptgs2 and IL-6, and induced ROS in MC3T3-E1 osteoblastic cells. Angptl4 is an endogenous inhibitor of LPL which was transcriptionally induced by TGRL-LP, while recombinant Angptl4 prevented TG-driven Atf3 induction. Atf3 global knockout male mice demonstrated increased trabecular and cortical microarchitectural parameters. In summary, we find that TGRL-LP induce osteoblastic cell stress as evidenced by expression of ATF3, which may contribute to the negative impact of dyslipidemia in the skeleton. Further, concomitant induction of Angptl4 in osteoblasts might play a protective role by reducing local lipolysis.


Assuntos
Dislipidemias , Lipólise , Masculino , Animais , Camundongos , Lipólise/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Fator 3 Ativador da Transcrição/genética , Fator 3 Ativador da Transcrição/metabolismo , Proteínas de Choque Térmico/metabolismo , Triglicerídeos/metabolismo , Lipoproteínas/genética , Lipoproteínas/metabolismo , Osteoblastos/metabolismo
4.
Cytotherapy ; 15(9): 1136-47, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23831362

RESUMO

BACKGROUND AIMS: Delivery of bone marrow-derived stem and progenitor cells to the site of injury is an effective strategy to enhance bone healing. An alternate approach is to mobilize endogenous, heterogeneous stem cells that will home to the site of injury. AMD3100 is an antagonist of the chemokine receptor 4 (CXCR4) that rapidly mobilizes stem cell populations into peripheral blood. Our hypothesis was that increasing circulating numbers of stem and progenitor cells using AMD3100 will improve bone fracture healing. METHODS: A transverse femoral fracture was induced in C57BL/6 mice, after which they were subcutaneously injected for 3 d with AMD3100 or saline control. Mesenchymal stromal cells, hematopoietic stem and progenitor cells and endothelial progenitor cells in the peripheral blood and bone marrow were evaluated by means of flow cytometry, automated hematology analysis and cell culture 24 h after injection and/or fracture. Healing was assessed up to 84 d after fracture by histomorphometry and micro-computed tomography. RESULTS: AMD3100 injection resulted in higher numbers of circulating mesenchymal stromal cells, hematopoietic stem cells and endothelial progenitor cells. Micro-computed tomography data demonstrated that the fracture callus was significantly larger compared with the saline controls at day 21 and significantly smaller (remodeled) at day 84. AMD3100-treated mice have a significantly higher bone mineral density than do saline-treated counterparts at day 84. CONCLUSIONS: Our data demonstrate that early cell mobilization had significant positive effects on healing throughout the regenerative process. Rapid mobilization of endogenous stem cells could provide an effective alternative strategy to cell transplantation for enhancing tissue regeneration.


Assuntos
Técnicas de Cultura de Células/métodos , Fraturas do Fêmur/terapia , Consolidação da Fratura/fisiologia , Mobilização de Células-Tronco Hematopoéticas/métodos , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/fisiologia , Células-Tronco/fisiologia , Animais , Benzilaminas , Densidade Óssea/efeitos dos fármacos , Densidade Óssea/fisiologia , Medula Óssea/efeitos dos fármacos , Medula Óssea/fisiologia , Células Cultivadas , Ciclamos , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Fraturas do Fêmur/tratamento farmacológico , Fraturas do Fêmur/fisiopatologia , Consolidação da Fratura/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Compostos Heterocíclicos/uso terapêutico , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco/efeitos dos fármacos
5.
Vet Surg ; 42(7): 814-8, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24033641

RESUMO

OBJECTIVE: To determine presence of bacterial DNA in canine stifles with cranial cruciate ligament rupture (CCLR) and medial patellar luxation (MPL) compared to normal canine stifles (control). STUDY DESIGN: Prospective clinical study. ANIMALS: Dogs (n = 44). METHODS: Dogs of varying age, breed, sex, and weight residing in California were assessed for stifle pathology (CCLR, MPL, or normal control). Synovial fluid of all stifles was assessed for the presence of bacterial DNA using broad-ranging 16S rRNA primers and PCR. RESULTS: Bacterial DNA was detected in normal control stifles and those with CCLR and MPL. There were no statistical differences in the copy numbers of bacterial DNA in the stifle synovial fluid among groups (P > .05); however, synovial fluid specimens from dogs with stifle pathology (CCLR and MPL combined) tended to have higher copy numbers of bacterial DNA than those from controls (P = .06). There was no significant difference in the number of bacterial DNA between the CCLR and MPL groups (P = .57). The copy numbers of bacterial DNA had a weak positive significant correlation with the duration of lameness in CCLR group (P < .05). CONCLUSIONS: Increased detection of bacterial DNA in the stifle synovial fluid may indicate joint pathology but not be directly linked to a specific joint disease.


Assuntos
Infecções Bacterianas/diagnóstico , DNA Bacteriano/isolamento & purificação , Doenças do Cão/diagnóstico , Artropatias/veterinária , Reação em Cadeia da Polimerase/veterinária , Joelho de Quadrúpedes/patologia , Líquido Sinovial/microbiologia , Animais , Ligamento Cruzado Anterior/patologia , Doenças do Cão/microbiologia , Cães , Feminino , Artropatias/diagnóstico , Artropatias/microbiologia , Masculino , Luxação Patelar/patologia , Luxação Patelar/veterinária , Reação em Cadeia da Polimerase/métodos
6.
JBMR Plus ; 7(4): e10733, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37065626

RESUMO

Hypoxia-inducible factors (HIFs) are oxygen-dependent heterodimeric transcription factors that mediate molecular responses to reductions in cellular oxygen (hypoxia). HIF signaling involves stable HIF-ß subunits and labile, oxygen-sensitive HIF-α subunits. Under hypoxic conditions, the HIF-α subunit is stabilized, complexes with nucleus-confined HIF-ß subunit, and transcriptionally regulates hypoxia-adaptive genes. Transcriptional responses to hypoxia include altered energy metabolism, angiogenesis, erythropoiesis, and cell fate. Three isoforms of HIF-α-HIF-1α, HIF-2α, and HIF-3α-are found in diverse cell types. HIF-1α and HIF-2α serve as transcriptional activators, whereas HIF-3α restricts HIF-1α and HIF-2α. The structure and isoform-specific functions of HIF-1α in mediating molecular responses to hypoxia are well established across a wide range of cell and tissue types. The contributions of HIF-2α to hypoxic adaptation are often unconsidered if not outrightly attributed to HIF-1α. This review establishes what is currently known about the diverse roles of HIF-2α in mediating the hypoxic response in skeletal tissues, with specific focus on development and maintenance of skeletal fitness. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

7.
JBMR Plus ; 7(4): e10724, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37065633

RESUMO

Molecular oxygen levels vary during development and disease. Adaptations to decreased oxygen bioavailability (hypoxia) are mediated by hypoxia-inducible factor (HIF) transcription factors. HIFs are composed of an oxygen-dependent α subunit (HIF-α), of which there are two transcriptionally active isoforms (HIF-1α and HIF-2α), and a constitutively expressed ß subunit (HIFß). Under normoxic conditions, HIF-α is hydroxylated via prolyl hydroxylase domain (PHD) proteins and targeted for degradation via Von Hippel-Lindau (VHL). Under hypoxic conditions, hydroxylation via PHD is inhibited, allowing for HIF-α stabilization and induction of target transcriptional changes. Our previous studies showed that Vhl deletion in osteocytes (Dmp1-cre; Vhl f/f ) resulted in HIF-α stabilization and generation of a high bone mass (HBM) phenotype. The skeletal impact of HIF-1α accumulation has been well characterized; however, the unique skeletal impacts of HIF-2α remain understudied. Because osteocytes orchestrate skeletal development and homeostasis, we investigated the role of osteocytic HIF-α isoforms in driving HBM phenotypes via osteocyte-specific loss-of-function and gain-of-function HIF-1α and HIF-2α mutations in C57BL/6 female mice. Deletion of Hif1a or Hif2a in osteocytes showed no effect on skeletal microarchitecture. Constitutively stable, degradation-resistant HIF-2α (HIF-2α cDR), but not HIF-1α cDR, generated dramatic increases in bone mass, enhanced osteoclast activity, and expansion of metaphyseal marrow stromal tissue at the expense of hematopoietic tissue. Our studies reveal a novel influence of osteocytic HIF-2α in driving HBM phenotypes that can potentially be harnessed pharmacologically to improve bone mass and reduce fracture risk. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

8.
Vet Sci ; 9(10)2022 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-36288137

RESUMO

Sex steroids regulate bone metabolism directly and indirectly through receptors on bone. Estrogen receptors (ER-∝, ER-ß), progesterone receptor (PR), and androgen receptor (AR), have been previously identified on human osteosarcoma (OSA) cells, and are considered to influence tumor growth, but their expression and role in canine OSA is unknown. The aim of this study was to characterize sex hormone receptor expression levels in naturally occurring OSA tissue and in three canine OSA cell lines. The expression of ER-α, ER-ß, PR, and AR was investigated using RT-PCR. PR expression levels were also quantified in OSA cells cultured under hypoxic conditions or in the presence of estradiol. The effects of progesterone on cell proliferation were quantified. Results demonstrated varying expression levels of these receptors in five OSA subtypes. OSA cell lines demonstrated high gene expression levels of PR and low gene expression levels of ER-α and ER-ß and no gene expression of AR. PR expression was increased in OSA cells cultured under hypoxic conditions in a HIF-∝ independent manner. Interestingly, one cell line expressed very high levels of PR, expression of which decreased in response to estradiol. In addition, progesterone decreased OSA cell proliferation in this particular cell line. Further investigation of the role of sex steroids, particularly PR and its ligands, in regulation of canine OSA is recommended.

9.
J Cell Biochem ; 112(11): 3354-63, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21751239

RESUMO

Tenascins regulate cell interaction with the surrounding pericellular matrix. Within bone, tenascins C and W influence osteoblast adhesion and differentiation, although little is known about the regulation of tenascin expression. In this study we examined the effect of osteogenic differentiation, bone morphogenetic protein (BMP) and Wnt growth factors, and mechanical loading on tenascin expression in osteogenic cells. Osteogenic differentiation increased tenascin C (TnC), and decreased tenascin W (TnW), expression. Both growth factors and mechanical loading increased both TnC and TnW expression, albeit via distinct signaling mechanisms. Both BMP-2 and Wnt5a induction of tenascin expression were mediated by MAP kinases. These data establish a role for BMP, Wnts, and mechanical loading in the regulation of tenascin expression in osteoblasts.


Assuntos
Osso e Ossos/metabolismo , Tenascina/metabolismo , Animais , Western Blotting , Proteínas Morfogenéticas Ósseas/fisiologia , Osso e Ossos/citologia , Diferenciação Celular , Linhagem Celular , Mecanotransdução Celular , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fatores de Transcrição NFATC/metabolismo , Reação em Cadeia da Polimerase , RNA Mensageiro/genética , Transdução de Sinais , Tenascina/genética , Fosfolipases Tipo C/metabolismo , Proteínas Wnt/fisiologia
10.
Cell Biol Int ; 35(10): 981-9, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21574962

RESUMO

A variety of pathologies such as skeletal fracture, neoplasia and inflammation compromise tissue perfusion and thereby decrease tissue oxygen tension. We and others have demonstrated that hypoxia is a potent stimulant for MSC (mesenchymal stem cell) recruitment and differentiation, yet to date little research has focused on the effects of oxygen tension on MSC migration. In the present study, we examined the effects of hypoxia and the potential role of the GTPase RhoA and HIF-1α (hypoxia-inducible factor 1α) on MSC migration. Our results demonstrate that hypoxia decreases MSC migration through an HIF-1α and RhoA-mediated pathway. The active GTP-bound form of RhoA was reduced in 1% oxygen, whereas activation of RhoA under hypoxic conditions rescued migration. Furthermore, stabilization of HIF-1α under normoxic conditions attenuated cell migration similar to that of hypoxia. These results suggest that hypoxia negatively affects MSC migration by regulating activation of GTPases. These results highlight the importance of oxygen in regulating the recruitment of progenitor cells to areas of ischaemic tissue damage.


Assuntos
Hipóxia Celular , Movimento Celular , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Células-Tronco Mesenquimais/citologia , Proteína rhoA de Ligação ao GTP/metabolismo , Citoesqueleto de Actina/metabolismo , Adolescente , Células da Medula Óssea/citologia , Humanos , Masculino , Transdução de Sinais
11.
J Cell Biochem ; 110(1): 87-96, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20213746

RESUMO

Skeletal defects commonly suffer from poor oxygen microenvironments resulting from compromised vascularization associated with injury or disease. Adipose stem cells (ASCs) represent a promising cell population for stimulating skeletal repair by differentiating toward the osteogenic lineage or by secreting trophic factors. However, the osteogenic or trophic response of ASCs to reduced oxygen microenvironments is poorly understood. Moreover, a direct comparison between 2D and 3D response of ASCs to hypoxia is lacking. Thus, we characterized the osteogenic and angiogenic potential of human ASCs under hypoxic (1%), normoxic (5%), and atmospheric (21%) oxygen tensions in both 2D and 3D over 4 weeks in culture. We detected greatest alkaline phosphatase activity and extracellular calcium deposition in cells cultured in both 2D and 3D under 21% oxygen, and reductions in enzyme activity corresponded to reductions in oxygen tension. ASCs cultured in 1% oxygen secreted more vascular endothelial growth factor (VEGF) over the 4-week period than cells cultured in other conditions, with cells cultured in 2D secreting VEGF in a more sustained manner than those in 3D. Expression of osteogenic markers revealed temporal changes under different oxygen conditions with peak expression occurring earlier in 3D. In addition, the increase of most osteogenic markers was significantly higher in 2D compared to 3D cultures at 1% and 5% oxygen. These results suggest that oxygen, in conjunction with dimensionality, affects the timing of the differentiation program in ASCs. These findings offer new insights for the use of ASCs in bone repair while emphasizing the importance of the culture microenvironment.


Assuntos
Tecido Adiposo/citologia , Técnicas de Cultura de Células/métodos , Diferenciação Celular/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Oxigênio/farmacologia , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Adulto , Fosfatase Alcalina/metabolismo , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , DNA/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Pessoa de Meia-Idade , Neovascularização Fisiológica/efeitos dos fármacos , Osteogênese/genética , Células-Tronco/enzimologia , Células-Tronco/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
12.
J Cell Biochem ; 110(2): 457-67, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20336693

RESUMO

Mutations in sclerostin function or expression cause sclerosing bone dysplasias, involving decreased antagonism of Wnt/Lrp5 signaling. Conversely, deletion of the VHL tumor suppressor in osteoblasts, which stabilize HIF-alpha isoforms and thereby enables HIF-alpha/beta-driven gene transcription, increases bone mineral content and cross-sectional area compared to wild-type controls. We examined the influence of cellular hypoxia (1% oxygen) upon sclerostin expression and canonical Wnt signaling. Osteoblasts and osteocytes cultured under hypoxia revealed decreased sclerostin transcript and protein, and increased expression and nuclear localization of activated beta-catenin. Similarly, both hypoxia and the hypoxia mimetic DFO increased beta-catenin gene reporter activity. Hypoxia and its mimetics increased expression of the BMP antagonists gremlin and noggin and decreased Smad-1/5/8 phosphorylation. As a partial explanation for the mechanism of regulation of sclerostin by oxygen, MEF2 reporter assays revealed decreased activity. Modulation of VEGF signaling under normoxia or hypoxia revealed no influence upon Sost transcription. These data suggest that hypoxia inhibits sclerostin expression, through enhanced antagonism of BMP signaling independent of VEGF.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Hipóxia Celular , Osteoblastos/metabolismo , Transdução de Sinais , Proteínas Wnt/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Western Blotting , Linhagem Celular , Marcadores Genéticos , Humanos , Transcrição Gênica , Fator A de Crescimento do Endotélio Vascular/metabolismo , beta Catenina/metabolismo
13.
Cells Tissues Organs ; 191(3): 175-84, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19690399

RESUMO

The use of mesenchymal stem cells in tissue engineering to augment the repair of a variety of tissues including bone is a rapidly growing and exciting field. Although oxygen tension is a powerful stimulus for cells both in vitro and in vivo, the oxygen environment in which such cells would undergo differentiation is commonly overlooked. We examined the effect of long-term (21-days) low oxygen tension (1, 2 and 5%) on the osteogenic differentiation and matrix metalloproteinase (MMP)/tissue inhibitor of MMP (TIMP) expression of human mesenchymal stem cells (MSCs). Our data suggest that MSCs undergo osteoblastic differentiation most rapidly under 21% oxygen while oxygen tensions below 5% have an inhibitory effect. Interestingly, there was not a statistically significant difference in osteogenic markers between 5 and 21% oxygen. In addition, our data suggest that oxygen tension affects the expression of individual MMP and TIMPs differently. Low oxygen tension has an inhibitory effect on MMP-13 and TIMP-1 expression, which are involved in extracellular matrix remodeling and potentially vascular invasion. In contrast, MMP-2, a metalloproteinase involved in cell migration was not affected by oxygen tension. This data suggests that 21% oxygen may be beneficial for rapid osteogenic differentiation as would be required for the production of individual patient ex vivo constructs. In addition, this has important in vivo implications relating to the importance of early vascularization of sites of orthopedic injury. By augmenting the neovascularization process, it may be possible to facilitate more rapid differentiation of progenitors and thus the repair process.


Assuntos
Diferenciação Celular , Metaloproteinases da Matriz/biossíntese , Células-Tronco Mesenquimais/citologia , Osteogênese , Oxigênio/metabolismo , Inibidores Teciduais de Metaloproteinases/biossíntese , Fosfatase Alcalina/metabolismo , Cálcio/metabolismo , Células Cultivadas , Regulação da Expressão Gênica , Humanos , Metaloproteinase 13 da Matriz/biossíntese , Metaloproteinase 2 da Matriz/biossíntese , Metaloproteinase 9 da Matriz/biossíntese , Células-Tronco Mesenquimais/fisiologia , Osteocalcina/biossíntese , Osteopontina/biossíntese , Oxigênio/química , Fatores de Tempo , Engenharia Tecidual
14.
J Bone Miner Metab ; 28(1): 8-16, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19471853

RESUMO

Conditions such as fracture and unloading have been shown to be associated with tissue and cellular hypoxia in bone. The effects of hypoxia on bone cell physiology and ultimately its impact on bone tissue repair and remodeling are not well understood. In this study, we investigated the role of hypoxia on prostaglandin release from osteoblastic cells cultured in 2% (hypoxia), 5% (potentially cellular normoxia), and 21% (normoxia for standard cell culture conditions) oxygen for up to 24 h. We quantified the effects of reduced oxygen tension on the release of prostaglandin (PG)E(2), PGF(2alpha), PGD(2), and PGI(2). The mechanism by which hypoxia increases PG production was investigated by examining the various regulatory components of the PG biosynthetic pathway. Our data show that PGE(2) levels alone are significantly elevated under hypoxic conditions. Also, we show that cyclooxygenase (COX)-1 and COX-2 play an important role in hypoxia-induced PGE(2) production, possibly via a mechanism involving changes in their respective activity levels under low oxygen conditions. The effect of hypoxia on PGE(2) levels was mimicked by dimethyloxaloglycine, a known activator of the HIF pathway. In addition, we confirmed that HIF-1alpha was stabilized in osteoblastic cells under hypoxia. Taken together these data suggest a role for the HIF pathway in regulation of PGE(2) levels under hypoxic conditions. Previous studies have detected release of prostaglandins from areas of damaged bone, such as a fracture site, and our data may contribute to an understanding of how this release is regulated.


Assuntos
Hipóxia Celular , Dinoprostona/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Osteoblastos/fisiologia , Oxigênio/fisiologia , Animais , Remodelação Óssea/fisiologia , Linhagem Celular , Meios de Cultivo Condicionados/química , Inibidores de Ciclo-Oxigenase/farmacologia , Dinoprosta/análise , Dinoprosta/metabolismo , Dinoprostona/análise , Epoprostenol/análise , Epoprostenol/metabolismo , Hidroxiprostaglandina Desidrogenases/genética , Hidroxiprostaglandina Desidrogenases/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/agonistas , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Camundongos , Osteoblastos/metabolismo , Fosfolipases A2/genética , Fosfolipases A2/metabolismo , Reação em Cadeia da Polimerase , Prostaglandina D2/análise , Prostaglandina D2/metabolismo , RNA Mensageiro , Fatores de Tempo
15.
Am J Vet Res ; 71(10): 1237-45, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20919913

RESUMO

OBJECTIVE: To determine the optimal osteogenic source of equine mesenchymal stem cells (eMSCs) and optimize collection of and expansion conditions for those cells. ANIMALS: 10 adult Quarter Horses and 8 newborn Thoroughbred foals. PROCEDURES: eMSCs were isolated from bone marrow (BM), adipose tissue, and umbilical cord blood and tissue, and the osteogenic potential of each type was assessed. Effects of anatomic site, aspiration volume, and serum type on eMSC yield from BM were investigated. RESULTS: BM-eMSCs had the highest overall expression of the osteogenic genes Cbfa1, Osx, and Omd and staining for ALP activity and calcium deposition. There was no significant difference in BM-eMSC yield from the tuber coxae or sternum, but yield was significantly greater from the first 60-mL aspirate than from subsequent aspirates. The BM-eMSC expansion rate was significantly higher when cells were cultured in fetal bovine serum instead of autologous serum (AS). CONCLUSIONS AND CLINICAL RELEVANCE: eMSCs from BM possessed the highest in vitro osteogenic potential; eMSCs from adipose tissue also had robust osteogenic potential. The tuber coxae and the sternum were viable sources of BM-eMSCs in yearlings, and 60 mL of BM aspirate was sufficient for culture and expansion. Expanding BM-eMSCs in AS to avoid potential immunologic reactions decreased the total yield because BM-eMSCs grew significantly slower in AS than in fetal bovine serum. Additional studies are needed to determine optimal ex vivo eMSC culture and expansion conditions, including the timing and use of growth factor­supplemented AS.


Assuntos
Tecido Adiposo/citologia , Células da Medula Óssea/fisiologia , Sangue Fetal/citologia , Células-Tronco Mesenquimais/citologia , Osteogênese/fisiologia , Cordão Umbilical/citologia , Animais , Diferenciação Celular , Cavalos , Células-Tronco Mesenquimais/fisiologia
16.
J Cell Biochem ; 107(2): 233-9, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19277991

RESUMO

Changes in regional oxygen tension that occur during skeletal development and fracture stimulate local bone cell activity to regulate bone formation, maintenance, and repair. The adaptive responses of bone cells to hypoxia are only beginning to be understood. The transcription factor hypoxia-inducible factor-1alpha (HIF-1alpha) is activated under hypoxia and promotes expression of genes required for adaptation and cell survival, and also regulates both bone development and fracture repair. We have previously demonstrated that hypoxic osteoblasts increase PGE(2) release and expression of the PGE(2) receptor EP1. In the present studies, we investigated the impact of altered HIF-1alpha activity and expression on EP1 expression in osteoblasts. HIF-1alpha stabilization was induced in cells cultured in 21% oxygen by treatment with dimethyloxaloglycine (DMOG) or siRNA targeted against PHD2. To implicate HIF-1alpha in hypoxia-induced EP1 expression, osteoblastic cells were treated with siRNA targeted against HIF-1alpha prior to exposure to hypoxia. EP1 expression was significantly increased in cells cultured in 21% oxygen with DMOG or PHD2 siRNA treatment compared to controls. Hypoxia responsive element (HRE) activation in hypoxia was attenuated in cells treated with HIF-1alpha siRNA compared to controls, indicating HIF-1alpha as the functional HIF-alpha isoform in this system. Furthermore, hypoxic cells treated with HIF-1alpha siRNA demonstrated reduced EP1 expression in hypoxia compared to controls. Inhibition of SAPK/JNK activity significantly reduced hypoxia-induced EP1 expression but had no impact on HIF-1alpha expression or activity. These data strongly implicate a role for HIF-1alpha in hypoxia-induced EP1 expression and may provide important insight into the mechanisms by which HIF-1alpha regulates bone development and fracture repair.


Assuntos
Hipóxia Celular/fisiologia , Regulação da Expressão Gênica/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Osteoblastos/metabolismo , Receptores de Prostaglandina E/biossíntese , Transdução de Sinais/fisiologia , Animais , Western Blotting , Linhagem Celular , Inativação Gênica , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Prolina Dioxigenases do Fator Induzível por Hipóxia , MAP Quinase Quinase 4/metabolismo , Camundongos , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Pró-Colágeno-Prolina Dioxigenase/genética , RNA Interferente Pequeno , Receptores de Prostaglandina E Subtipo EP1 , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
17.
PLoS One ; 14(9): e0222462, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31513647

RESUMO

Migration of cells from both local and systemic sources is essential for the inflammatory and regenerative processes that occur during normal wound healing. CXCL12 is considered a critical regulator of CXCR4-positive cell migration during tissue regeneration. In this study, we investigated the expression of Cxcl12 and Cxcr4 during healing of a murine full thickness ear wound. We also investigated the expression of angiopoietin-like 4, which has been shown to participate in wound angiogenesis and reepithelialization. At time points up to 48hrs, complete blood counts were performed using automated hematology analysis, and the numbers of circulating stem and progenitor cells quantified using flow cytometry. Expression of both Cxcr4 and Angptl4 was significantly elevated within 3 days of wounding, and both were strongly expressed in cells of the epidermis. ANGPTL4 protein expression remained elevated in the epithelium through day 14. Cxcl12 expression was increased significantly at day 3, and remained elevated through day 21. Faint Cxcl12 staining was detectable in the epithelium at day 1, and thereafter staining was faint and more generalized. There were significantly fewer circulating total white blood cells and lymphocytes 1hr following ear punching. Similarly, there was a significant early (1hr) reduction in the number of circulating endothelial progenitor cells. Further studies are warranted to investigate whether ANGPTL4 and CXCL12/CXCR4 interact or synergize to facilitate cell recruitment and migration, and to potentiate reepithelialization and wound healing.


Assuntos
Quimiocina CXCL12/genética , Receptores CXCR4/genética , Cicatrização/fisiologia , Proteína 4 Semelhante a Angiopoietina/genética , Proteína 4 Semelhante a Angiopoietina/metabolismo , Animais , Movimento Celular , Quimiocina CXCL12/metabolismo , Orelha/patologia , Células Progenitoras Endoteliais/metabolismo , Células Epidérmicas/metabolismo , Epiderme/metabolismo , Regulação da Expressão Gênica/genética , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/metabolismo , Receptores CXCR4/metabolismo , Transdução de Sinais , Células-Tronco/metabolismo , Transcriptoma/genética
18.
J Bone Miner Res ; 34(1): 157-170, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30189111

RESUMO

The most reliable predictor of future fracture risk is a previous fracture of any kind. The etiology of this increased fracture risk is not fully known, but it is possible that fracture initiates systemic bone loss, leading to greater fracture risk at all skeletal sites. In this study, we investigated systemic bone loss and recovery after femoral fracture in young (3-month-old) and middle-aged (12-month-old) mice. Transverse femur fractures were created using a controlled impact, and whole-body bone mineral density (BMD), trabecular and cortical microstructure, bone mechanical properties, bone formation and resorption rates, mouse voluntary movement, and systemic inflammation were quantified at multiple time points post-fracture. We found that fracture led to decreased whole-body BMD in both young and middle-aged mice 2 weeks post-fracture; this bone loss was recovered by 6 weeks in young but not middle-aged mice. Similarly, trabecular bone volume fraction (BV/TV) of the L5 vertebral body was significantly reduced in fractured mice relative to control mice 2 weeks post-fracture (-11% for young mice, -18% for middle-aged mice); no significant differences were observed 6 weeks post-fracture. At 3 days post-fracture, we observed significant increases in serum levels of interleukin-6 and significant decreases in voluntary movement in fractured mice compared with control mice, with considerably greater changes in middle-aged mice than in young mice. At this time point, we also observed increased osteoclast number on L5 vertebral body trabecular bone of fractured mice compared with control mice. These data show that systemic bone loss occurs after fracture in both young and middle-aged mice, and recovery from this bone loss may vary with age. This systemic response could contribute to increased future fracture risk after fracture; these data may inform clinical treatment of fractures with respect to improving long-term skeletal health. © 2018 American Society for Bone and Mineral Research.


Assuntos
Envelhecimento , Densidade Óssea , Doenças Ósseas Metabólicas , Fraturas do Fêmur , Osteogênese , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Doenças Ósseas Metabólicas/metabolismo , Doenças Ósseas Metabólicas/patologia , Doenças Ósseas Metabólicas/fisiopatologia , Feminino , Fraturas do Fêmur/metabolismo , Fraturas do Fêmur/patologia , Fraturas do Fêmur/fisiopatologia , Camundongos
19.
Biochem Biophys Res Commun ; 366(4): 1061-6, 2008 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-18155656

RESUMO

Little is known about the role or identity of signaling molecules released by osteocytes to recruit MSCs to areas of matrix damage. Vascular disruption at fracture sites results in hypoxia which is known to up-regulate genes involved in cell migration including osteopontin (OPN). We examined the effect of conditioned media from hypoxic osteocytes on MSC migration. Hypoxic osteocyte media significantly increased MSC migration and expression of OPN was significantly increased in hypoxic osteocytes. OPN and CD44 neutralizing antibodies significantly reduced MSC migration. Further, recombinant OPN significantly increased MSC migration in a dose-dependent manner. Our data support the hypothesis that hypoxia at a fracture site stimulates the release of chemotactic factors, such as OPN, from osteocytes, that induce MSC migration to aid in fracture repair. To our knowledge, these are the first data to suggest a role for osteocytes and OPN in the recruitment of MSCs to aid in fracture repair.


Assuntos
Movimento Celular , Receptores de Hialuronatos/metabolismo , Células-Tronco Mesenquimais/citologia , Osteócitos/citologia , Osteopontina/metabolismo , Hipóxia Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Meios de Cultivo Condicionados , Relação Dose-Resposta a Droga , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Testes de Neutralização , Osteócitos/efeitos dos fármacos , Osteopontina/farmacologia , Proteínas Recombinantes/farmacologia
20.
Bone ; 116: 307-314, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30172741

RESUMO

Tissue oxygen (O2) levels vary during development and disease; adaptations to decreased O2 (hypoxia) are mediated by hypoxia-inducible factor (HIF) transcription factors. HIFs are active in the skeleton, and stabilizing HIF-α isoforms cause high bone mass (HBM) phenotypes. A fundamental limitation of previous studies examining the obligate role for HIF-α isoforms in the skeleton involves the persistence of gene deletion as osteolineage cells differentiate into osteocytes. Because osteocytes orchestrate skeletal development and homeostasis, we evaluated the influence of Vhl or Hif1a disruption in osteocytes. Osteocytic Vhl deletion caused HBM phenotype, but Hif1a was dispensable in osteocytes. Vhl cKO mice revealed enhanced canonical Wnt signaling. B cell development was reduced while myelopoiesis increased in osteocytic Vhl cKO, revealing a novel influence of Vhl/HIF-α function in osteocytes on maintenance of bone microarchitecture via canonical Wnt signaling and effects on hematopoiesis.


Assuntos
Osso e Ossos/metabolismo , Hematopoese , Osteócitos/metabolismo , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo , Animais , Osso Esponjoso/patologia , Osso Cortical/patologia , Deleção de Genes , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Linfopoese , Camundongos Endogâmicos C57BL , Tamanho do Órgão , Via de Sinalização Wnt
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa