Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Kidney Int ; 83(6): 1159-68, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23389416

RESUMO

Elevated fibroblast growth factor 23 (FGF23) is associated with cardiovascular disease in patients with chronic kidney disease. As a potential mediating mechanism, FGF23 induces left ventricular hypertrophy; however, its role in arterial calcification is less clear. In order to study this, we quantified coronary artery and thoracic aorta calcium by computed tomography in 1501 patients from the Chronic Renal Insufficiency Cohort (CRIC) study within a median of 376 days (interquartile range 331-420 days) of baseline. Baseline plasma FGF23 was not associated with the prevalence or severity of coronary artery calcium after multivariable adjustment. In contrast, higher serum phosphate levels were associated with prevalence and severity of coronary artery calcium, even after adjustment for FGF23. Neither FGF23 nor serum phosphate were consistently associated with thoracic aorta calcium. We could not detect mRNA expression of FGF23 or its coreceptor, klotho, in human or mouse vascular smooth muscle cells, or normal or calcified mouse aorta. Whereas elevated phosphate concentrations induced calcification in vitro, FGF23 had no effect on phosphate uptake or phosphate-induced calcification regardless of phosphate concentration or even in the presence of soluble klotho. Thus, in contrast to serum phosphate, FGF23 is not associated with arterial calcification and does not promote calcification experimentally. Hence, phosphate and FGF23 promote cardiovascular disease through distinct mechanisms.


Assuntos
Aorta Torácica/metabolismo , Doenças da Aorta/sangue , Cálcio/metabolismo , Doença da Artéria Coronariana/sangue , Vasos Coronários/metabolismo , Fatores de Crescimento de Fibroblastos/sangue , Insuficiência Renal Crônica/sangue , Calcificação Vascular/sangue , Adulto , Idoso , Animais , Aorta Torácica/diagnóstico por imagem , Doenças da Aorta/diagnóstico por imagem , Doenças da Aorta/epidemiologia , Aortografia/métodos , Células Cultivadas , Distribuição de Qui-Quadrado , Angiografia Coronária/métodos , Doença da Artéria Coronariana/diagnóstico por imagem , Doença da Artéria Coronariana/epidemiologia , Vasos Coronários/diagnóstico por imagem , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/genética , Glucuronidase/genética , Glucuronidase/metabolismo , Humanos , Proteínas Klotho , Modelos Logísticos , Masculino , Camundongos , Pessoa de Meia-Idade , Análise Multivariada , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Fosfatos/sangue , Prevalência , Estudos Prospectivos , RNA Mensageiro/metabolismo , Insuficiência Renal Crônica/diagnóstico por imagem , Insuficiência Renal Crônica/epidemiologia , Fatores de Risco , Índice de Gravidade de Doença , Fatores de Tempo , Tomografia Computadorizada por Raios X , Estados Unidos/epidemiologia , Regulação para Cima , Calcificação Vascular/diagnóstico por imagem , Calcificação Vascular/epidemiologia , Adulto Jovem
2.
Circ Res ; 104(6): 733-41, 2009 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-19197075

RESUMO

Vascular calcification is a major risk factor for cardiovascular morbidity and mortality. To develop appropriate prevention and/or therapeutic strategies for vascular calcification, it is important to understand the origins of the cells that participate in this process. In this report, we used the SM22-Cre recombinase and Rosa26-LacZ alleles to genetically trace cells derived from smooth muscle. We found that smooth muscle cells (SMCs) gave rise to osteochondrogenic precursor- and chondrocyte-like cells in calcified blood vessels of matrix Gla protein deficient (MGP(-/-)) mice. This lineage reprogramming of SMCs occurred before calcium deposition and was associated with an early onset of Runx2/Cbfa1 expression and the downregulation of myocardin and Msx2. There was no change in the constitutive expression of Sox9 or bone morphogenetic protein 2. Osterix, Wnt3a, and Wnt7a mRNAs were not detected in either calcified MGP(-/-) or noncalcified wild-type (MGP(+/+)) vessels. Finally, mechanistic studies in vitro suggest that Erk signaling might be required for SMC transdifferentiation under calcifying conditions. These results provide strong support for the hypothesis that adult SMCs can transdifferentiate and that SMC transdifferentiation is an important process driving vascular calcification and the appearance of skeletal elements in calcified vascular lesions.


Assuntos
Artérias/metabolismo , Calcinose/metabolismo , Condrócitos/metabolismo , Miócitos de Músculo Liso/metabolismo , Células-Tronco/metabolismo , Doenças Vasculares/metabolismo , Animais , Artérias/patologia , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/metabolismo , Calcinose/genética , Calcinose/patologia , Desdiferenciação Celular/genética , Condrócitos/patologia , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Regulação para Baixo/genética , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Camundongos Knockout , Camundongos Mutantes , Miócitos de Músculo Liso/patologia , Proteínas/genética , Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Fator de Transcrição Sp7 , Células-Tronco/patologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Doenças Vasculares/genética , Doenças Vasculares/patologia , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteína Wnt3 , Proteína Wnt3A
3.
J Exp Med ; 196(8): 1047-55, 2002 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-12391016

RESUMO

Osteopontin (OPN) is abundantly expressed in human calcified arteries. To examine the role of OPN in vascular calcification, OPN mutant mice were crossed with matrix Gla protein (MGP) mutant mice. Mice deficient in MGP alone (MGP(-/-) OPN(+/+)) showed calcification of their arteries as early as 2 weeks (wk) after birth (0.33 +/- 0.01 mmol/g dry weight), and the expression of OPN in the calcified arteries was greatly up-regulated compared with MGP wild-types. OPN accumulated adjacent to the mineral and colocalized to surrounding cells in the calcified media. Cells synthesizing OPN lacked smooth muscle (SM) lineage markers, SM alpha-actin and SM22alpha. However, most of them were not macrophages. Importantly, mice deficient in both MGP and OPN had twice as much arterial calcification as MGP(-/-) OPN(+/+) at 2 wk, and over 3 times as much at 4 wk, suggesting an inhibitory effect of OPN in vascular calcification. Moreover, these mice died significantly earlier (4.4 +/- 0.2 wk) than MGP(-/-) OPN(+/+) counterparts (6.6 +/- 1.0 wk). The cause of death in these animals was found to be vascular rupture followed by hemorrhage, most likely due to enhanced calcification. These studies are the first to demonstrate a role for OPN as an inducible inhibitor of ectopic calcification in vivo.


Assuntos
Artérias/patologia , Calcinose/genética , Proteínas de Ligação ao Cálcio/fisiologia , Regulação para Baixo , Endotélio Vascular/patologia , Proteínas da Matriz Extracelular , Sialoglicoproteínas/genética , Animais , Artérias/metabolismo , Proteínas de Ligação ao Cálcio/genética , Endotélio Vascular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteopontina , Sialoglicoproteínas/fisiologia , Proteína de Matriz Gla
4.
Genesis ; 47(12): 858-63, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19882669

RESUMO

Accelerated vascular calcification occurs in several human diseases including diabetes and chronic kidney disease (CKD). In patients with CKD, vascular calcification is highly correlated with elevated serum phosphate levels. In vitro, elevated concentrations of phosphate induced vascular smooth muscle cell matrix mineralization, and the inorganic phosphate transporter-1 (PiT-1), was shown to be required. To determine the in vivo role of PiT-1, mouse conditional and null alleles were generated. Here we show that the conditional allele, PiT-1(flox), which has loxP sites flanking exons 3 and 4, is homozygous viable. Cre-mediated recombination resulted in a null allele that is homozygous lethal. Examination of early embryonic development revealed that the PiT-1(Deltae3,4/Deltae3,4) embryos displayed anemia, a defect in yolk sac vasculature, and arrested growth. Thus, conditional and null PiT-1 mouse alleles have been successfully generated and PiT-1 has a necessary, nonredundant role in embryonic development.


Assuntos
Alelos , Regulação da Expressão Gênica no Desenvolvimento , Mutação , Fator de Transcrição Pit-1/genética , Animais , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Endogâmicos DBA , Camundongos Endogâmicos , Camundongos Knockout , Camundongos Transgênicos , Gravidez , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Fator de Transcrição Pit-1/fisiologia
5.
Kidney Int ; 75(12): 1297-1307, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19322138

RESUMO

Arterial medial calcification is a major complication in patients with chronic kidney disease and is a strong predictor of cardiovascular and all-cause mortality. We sought to determine the role of dietary phosphorus and the severity of uremia on vascular calcification in calcification-prone DBA/2 mice. Severe and moderate uremia was induced by renal ablation of varying magnitudes. Extensive arterial-medial calcification developed only when the uremic mice were placed on a high-phosphate diet. Arterial calcification in the severely uremic mice fed a high-phosphate diet was significantly associated with hyperphosphatemia. Moderately uremic mice on this diet were not hyperphosphatemic but had a significant rise in their serum levels of fibroblast growth factor 23 (FGF-23) and osteopontin that significantly correlated with arterial medial calcification. Although there was widespread arterial medial calcification, there was no histological evidence of atherosclerosis. At early stages of calcification, the osteochondrogenic markers Runx2 and osteopontin were upregulated, but the smooth muscle cell marker SM22alpha decreased in medial cells, as did the number of smooth muscle cells in extensively calcified regions. These findings suggest that phosphate loading and the severity of uremia play critical roles in controlling arterial medial calcification in mice. Further, FGF-23 and osteopontin may be markers and/or inducers of this process.


Assuntos
Artérias/patologia , Calcinose/sangue , Calcinose/etiologia , Fosfatos/administração & dosagem , Uremia/sangue , Uremia/complicações , Doenças Vasculares/sangue , Doenças Vasculares/etiologia , Animais , Artérias/metabolismo , Calcinose/metabolismo , Calcinose/patologia , Cálcio/sangue , Cálcio/metabolismo , Modelos Animais de Doenças , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/sangue , Humanos , Camundongos , Camundongos Endogâmicos DBA , Osteopontina/sangue , Osteopontina/metabolismo , Fosfatos/toxicidade , Fósforo/sangue , Uremia/metabolismo , Uremia/patologia , Doenças Vasculares/metabolismo , Doenças Vasculares/patologia
6.
Circ Res ; 98(7): 905-12, 2006 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-16527991

RESUMO

Vascular calcification is associated with cardiovascular morbidity and mortality. Hyperphosphatemia is an important contributor to vascular calcification. Our previous studies demonstrated that elevated phosphate induces calcification of smooth muscle cells (SMC) in vitro. Inhibition of phosphate transport by phosphonoformic acid blocked phosphate-induced calcification, implicating sodium-dependent phosphate cotransporters in this process. In the present study, we have investigated the role of the type III sodium-dependent phosphate cotransporter, Pit-1, in SMC calcification in vitro. Human SMC stably expressing Pit-1 small interfering double-stranded RNA (SMC-iRNA) were established using a retroviral system. SMC-iRNA had decreased Pit-1 mRNA and protein levels and sodium-dependent phosphate transport activity compared with the control transduced cells (SMC-CT) (2.9 versus 9.78 nmol/mg protein per 30 minutes, respectively). Furthermore, phosphate-induced SMC calcification was significantly inhibited in SMC-iRNA compared with SMC-CT at all time points examined. Overexpression of Pit-1 restored phosphate uptake and phosphate-induced calcification in Pit-1 deficient cells. Mechanistically, although Pit-1-mediated SMC calcification was not associated with apoptosis or cell-derived vesicles, inhibition of phosphate uptake in Pit-1 knockdown cells blocked the induction of the osteogenic markers Cbfa-1 and osteopontin. Our results indicate that phosphate uptake through Pit-1 is essential for SMC calcification and phenotypic modulation in response to elevated phosphate.


Assuntos
Calcificação Fisiológica/fisiologia , Músculo Liso Vascular/fisiologia , Fosfatos/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/metabolismo , Sódio/metabolismo , Aorta , Cálcio/metabolismo , Humanos
7.
Cardiovasc Res ; 66(2): 324-33, 2005 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15820201

RESUMO

OBJECTIVE: Vascular calcification is an actively regulated process, correlating with cardiovascular morbidity and mortality especially in patients with diabetes and chronic renal diseases. Osteopontin (OPN) is abundantly expressed in human calcified arteries and inhibits vascular calcification in vitro and in vivo. How OPN functions in vascular calcification, however, is less clear. METHODS: Smooth muscle cells (SMCs) were isolated from aortas of OPN knock-out (OPN-/-) and wild type (OPN+/+) mice. RESULTS: OPN-/- SMCs were identical to OPN+/+ SMCs in morphology and stained positively for SM lineage proteins, desmin, smooth muscle alpha-actin and SM22alpha. No spontaneous calcification was observed in OPN-/- SMCs under normal culture conditions or in medium containing 1%, 3%, or 5% fetal bovine serum. However, when cultured in medium containing elevated concentrations of inorganic phosphate, an inducer of vascular calcification, a significantly higher calcification was observed in OPN-/- SMCs compared to OPN+/+ SMCs that, in response to elevated phosphate, synthesized and secreted OPN into the culture. Finally, retroviral transduction of mouse OPN cDNA into OPN-/- SMCs rescued the calcification phenotype of the cells. CONCLUSION: These results are the first to demonstrate an inhibitory role of endogenously produced OPN on SMC calcification, suggesting a novel feedback mechanism where OPN produced locally by the SMCs may serve as an important inducible inhibitor of vascular calcification.


Assuntos
Calcinose/etiologia , Músculo Liso Vascular/metabolismo , Sialoglicoproteínas/deficiência , Animais , Aorta , Calcinose/metabolismo , Técnicas de Cultura de Células , DNA Complementar/administração & dosagem , Suscetibilidade a Doenças , Vetores Genéticos/administração & dosagem , Camundongos , Camundongos Knockout , Osteopontina , Fosfatos/farmacologia , Retroviridae/genética , Sialoglicoproteínas/genética , Transdução Genética/métodos
8.
PLoS One ; 8(12): e84465, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24386387

RESUMO

Osteoclasts are bone-resorbing cells that are critical for the normal formation and maintenance of teeth and skeleton. Osteoclast deficiency can contribute to heterotopic ossification (HO), a pathology that is particularly detrimental to the mechanical functions of joints, valves and blood vessels. On the other hand, osteoclast over-activity is a major cause of osteoporosis. A reliable method for controlled generation of osteoclasts would be useful as a potential autologous cell therapy for HO, as well as high-throughput drug screening for anti-osteoporotic drugs. In this report, we describe the development of a cell engineering approach to control monocytic precursor cell differentiation to osteoclasts. Oligomerization of receptor activator of nuclear factor κB (RANK) is known to be essential for osteoclast differentiation from monocyte/macrophage precursors. We engineered a murine monocytic cell line, RAW264.7 to express a fusion protein comprising the intracellular RANK signaling domain and FK506-derived dimerization domains that bind to a small molecule chemical inducer of dimerization (CID). Virally infected cells expressing this fusion protein were treated with CID and dose-dependent induction of tartrate-resistant acid phosphatase activity, as well as multinucleated osteoclast formation were observed. Furthermore, NF-κB signaling was upregulated in a CID-dependent fashion, demonstrating effective RANK intracellular signaling. Functionally CID-induced osteoclasts had robust mineral resorptive activity in both two-dimensional and three-dimensional in vitro resorption assays. In addition, the CID-induced osteoclasts have the same life span as native RANKL-induced osteoclasts. Most importantly and crucially, the engineered cells differentiated into osteoclasts that were resistant to the potent osteoclast inhibitor, osteoprotegerin. Taken together, these studies are the first to describe a method for inducible control of monocytic precursor differentiation to osteoclasts that may be useful for future development of an engineered autologous cell therapy as well as high-throughput drug testing systems to treat diseases of osteoclast over-activity that are independent of osteoprotegerin.


Assuntos
Diferenciação Celular , Células Progenitoras Mieloides/metabolismo , NF-kappa B/metabolismo , Osteoclastos/metabolismo , Ligante RANK/metabolismo , Animais , Linhagem Celular , Células Progenitoras Mieloides/citologia , NF-kappa B/genética , Osteoclastos/citologia , Ligante RANK/genética
9.
Cardiovasc Res ; 94(3): 545-54, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22436847

RESUMO

AIMS: Vascular cartilaginous metaplasia and calcification are common in patients with atherosclerosis. However, sources of cells contributing to the development of this complication are currently unknown. In this study, we ascertained the origin of cells that give rise to cartilaginous and bony elements in atherosclerotic vessels. METHODS AND RESULTS: We utilized genetic fate mapping strategies to trace cells of smooth muscle (SM) origin via SM22α-Cre recombinase and Rosa26-LacZ Cre reporter alleles. In animals expressing both transgenes, co-existence within a single cell of ß-galactosidase [marking cells originally derived from SM cells (SMCs)] with osteochondrogenic (Runx2/Cbfa1) or chondrocytic (Sox9, type II collagen) markers, along with simultaneous loss of SM lineage proteins, provides a strong evidence supporting reprogramming of SMCs towards osteochondrogenic or chondrocytic differentiation. Using this technique, we found that vascular SMCs accounted for ~80% of Runx2/Cbfa1-positive cells and almost all of type II collagen-positive cells (~98%) in atherosclerotic vessels of LDLr-/- and ApoE-/- mice. We also assessed contribution from bone marrow (BM)-derived cells via analysing vessels dissected from chimerical ApoE-/- mice transplanted with green fluorescence protein-expressing BM. Marrow-derived cells were found to account for ~20% of Runx2/Cbfa1-positive cells in calcified atherosclerotic vessels of ApoE-/- mice. CONCLUSION: Our results are the first to definitively identify cell sources attributable to atherosclerotic intimal calcification. SMCs were found to be a major contributor that reprogrammed its lineage towards osteochondrogenesis. Marrow-derived cells from the circulation also contributed significantly to the early osteochondrogenic differentiation in atherosclerotic vessels.


Assuntos
Células da Medula Óssea/metabolismo , Calcificação Fisiológica/genética , Diferenciação Celular/fisiologia , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Animais , Aterosclerose/genética , Aterosclerose/patologia , Células da Medula Óssea/citologia , Linhagem da Célula , Células Cultivadas , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
10.
Atherosclerosis ; 199(2): 271-7, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18179800

RESUMO

Vascular calcification is associated with increased risk of cardiovascular events that are the most common cause of death in patients with end-stage renal disease. Clinical and experimental studies indicate that hyperphosphatemia is a risk factor for vascular calcification and cardiovascular mortality in these patients. Our previous studies demonstrated that phosphate transport through the type III sodium-dependent phosphate cotransporter, Pit-1, was necessary for phosphate-induced calcification and osteochondrogenic phenotypic change in cultured human smooth muscle cells (SMC). BMP-2 is a potent osteogenic protein required for osteoblast differentiation and bone formation that has been implicated in vascular calcification. In the present study, we have examined the effects of BMP-2 on human SMC calcification in vitro. We found that treatment of SMC with BMP-2 enhanced elevated phosphate-induced calcification, but did not induce calcification under normal phosphate conditions. mRNAs for BMP receptors, including ALK2, ALK3, ALK6, BMPR-II, ActR-IIA and ActR-IIB were all detected in human SMCs. Mechanistically, BMP-2 dose-dependently stimulated phosphate uptake in SMC (200 ng/ml BMP-2 vs. vehicle: 13.94 vs. 7.09 nmol/30 min/mg protein, respectively). Real-time PCR and Western blot revealed the upregulation of Pit-1 mRNA and protein levels, respectively, by BMP-2. More importantly, inhibition of phosphate uptake by a competitive inhibitor of sodium-dependent phosphate cotransport, phosphonoformic acid, abrogated BMP-2-induced calcification. These results indicate that phosphate transport via Pit-1 is crucial in BMP-2-regulated SMC calcification. In addition, BMP-2-induced Runx2 and inhibited SM22 expression, indicating that it promotes osteogenic phenotype transition in these cells. Thus, BMP-2 may promote vascular calcification via increased phosphate uptake and induction of osteogenic phenotype modulation in SMC.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Regulação da Expressão Gênica , Músculo Liso Vascular/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Desenvolvimento Ósseo , Proteína Morfogenética Óssea 2 , Cálcio/metabolismo , Diferenciação Celular , Humanos , Modelos Biológicos , Miócitos de Músculo Liso/citologia , Osteoblastos/metabolismo , Osteogênese , Fenótipo , Fosfatos/metabolismo , RNA Mensageiro/metabolismo , Fator de Transcrição Pit-1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA