Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Cell Biochem ; 118(12): 4240-4253, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28419519

RESUMO

Receptor activator of NF-κB ligand (RANKL) is a TNF-like cytokine which mediates diverse physiological functions including bone remodeling and immune regulation. RANKL has been identified in atherosclerotic lesions; however, its role in atherosclerotic plaque development remains elusive. An enhancer located 75 kb upstream of the murine Rankl gene's transcription start site designated D5 is important for its calciotropic hormone- and cytokine-mediated expression. Here, we determined the impact of RANKL levels in atherosclerotic plaque development in the D5 enhancer-null (D5-/- ) mice in an atherogenic Apoe-/- background fed a high-fat diet (HFD). Rankl mRNA transcripts were increased in aortic arches and thoracic aortae of Apoe-/- mice; however, this increase was blunted in Apoe-/- ;D5-/- mice. Similarly, higher Rankl transcripts were identified in splenic T lymphocytes in Apoe-/- mice, and their levels were reduced in Apoe-/- ;D5-/- mice. When analyzed by micro-computed tomography (µCT), atherosclerotic plaque calcification was identified in six out of eight Apoe-/- mice, whereas only one out of eight Apoe-/- ;D5-/- mice developed plaque calcification after 12 weeks of HFD. However, following 18 weeks of HFD challenge, all of Apoe-/- and Apoe-/- ;D5-/- animals developed atherosclerotic plaque calcification. Likewise, atherosclerotic lesion sizes were site-specifically reduced in the aortic arch of Apoe-/- ;D5-/- mice at initial stage of atherosclerosis and this effect was diminished as atherosclerosis proceeded to a more advanced stage. Our data suggest that deletion of the RANKL D5 enhancer delays the progression of atherosclerotic plaque development and plaque calcification in hypercholesterolemic mice. This work provides important insight into RANKL's regulatory role in atherosclerosis. J. Cell. Biochem. 118: 4240-4253, 2017. © 2017 Wiley Periodicals, Inc.


Assuntos
Calcinose/metabolismo , Elementos Facilitadores Genéticos , Hipercolesterolemia/complicações , Placa Aterosclerótica/metabolismo , Ligante RANK/genética , Deleção de Sequência , Animais , Sequência de Bases , Calcinose/etiologia , Calcinose/genética , Dieta Hiperlipídica , Modelos Animais de Doenças , Progressão da Doença , Feminino , Camundongos , Camundongos Knockout , Placa Aterosclerótica/etiologia , Placa Aterosclerótica/genética
2.
J Cell Biochem ; 118(5): 1050-1064, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27567005

RESUMO

Epidemiological and clinical data suggest adverse cardiovascular outcomes with respect to vitamin D deficiency. Here, we explored the effects of vitamin D in atherosclerotic plaque calcification in vivo by utilizing vitamin D receptor (Vdr)-deficient mice in an Apoe-/- background. Animals were fed a high-fat diet (HFD) for either 12 or 18 weeks and then examined for atherosclerotic plaque development. In order to prevent calcium deficiency, Vdr-/- and Apoe-/- ;Vdr-/- animals were fed a high-calcium rescue diet prior to initiation of the HFD feeding and supplemented with high-calcium water during HFD feeding. Although calcium supplementation improved bone mass in Vdr-/- and Apoe-/- ;Vdr-/- mice, neither strain was fully rescued. Systemic inflammatory responses observed in the absence of VDR were exaggerated in Apoe-/- mice. Whereas, hyperlipidemic profiles seen in Apoe-/- mice were ameliorated in the absence of VDR. Micro-computed tomography (µCT) analysis revealed that six out of eight Apoe-/- animals developed atherosclerotic plaque calcification following 12 weeks of HFD feeding and 100% of the mice developed plaque calcification after 18 weeks. In contrast, although atherosclerotic lesions were evident in Apoe-/- ;Vdr-/- mice at 12 and 18 weeks of HFD challenge, none of these animals developed plaque calcification at either time point. The active vitamin D hormone, 1,25(OH)2 D3 likely increased calcification in aortic smooth muscle cells perhaps by directly modulating expression of Alpl, Rankl, and Opg. Our data suggest that the absence of VDR inhibits atherosclerotic plaque calcification in hypercholesterolemic Apoe-/- mice, providing additional insight into the role of vitamin D in atherosclerotic plaque calcification. J. Cell. Biochem. 118: 1050-1064, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Apolipoproteínas E/deficiência , Cálcio/administração & dosagem , Hipercolesterolemia/complicações , Placa Aterosclerótica/prevenção & controle , Receptores de Calcitriol/deficiência , Animais , Apolipoproteínas E/genética , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Feminino , Hidroxicolecalciferóis/metabolismo , Hipercolesterolemia/induzido quimicamente , Camundongos , Camundongos Knockout , Placa Aterosclerótica/diagnóstico por imagem , Placa Aterosclerótica/genética , Receptores de Calcitriol/genética , Microtomografia por Raio-X
3.
J Steroid Biochem Mol Biol ; 164: 277-280, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-26361013

RESUMO

Transcribed from the SOST gene, sclerostin is an osteocyte-derived negative regulator of bone formation that inhibits osteoblastogenesis via antagonism of the Wnt pathway. Sclerostin is a promising therapeutic target for low bone mass diseases and neutralizing antibody therapies that target sclerostin are in development. Diverse stimuli regulate SOST including the vitamin D hormone, forskolin (Fsk), bone morphogenic protein 2 (BMP-2), oncostatin M (OSM), dexamethasone (Dex), and transforming growth factor (TGFß1). To explore the mechanisms by which these compounds regulate SOST expression, we examined their ability to regulate a SOST reporter minigene containing the entire SOST locus including the downstream regionor mutant minigenes containing a deletion of the -1kb to -2kb promoter proximal region (-1kb), ECR2, ECR5, or two point mutations in the MEF2 binding site of ECR5 (ECR5/MEF2). Previous reports suggest that both the PTH and TGFß1 effects on SOST are mediated through ECR5 and that the action of PTH is mediated specifically via the MEF2 binding site at ECR5. Consistent with these reports, the suppressive effects of Fsk were abrogated following both ECR5 deletion and ECR5/MEF2 mutation. In contrast, we found that TGFß1 negatively regulated SOST and that neither ECR5 nor ECR5/MEF2 was involved. Surprisingly, none of these four deletions/mutations abrogated the suppressive effects of the vitamin D hormone, OSM, Dex, or TGFß1, or the positive effects of BMP-2. These data suggest that we need to move beyond ECR5 to understand SOST regulation.


Assuntos
Colforsina/farmacologia , Elementos Facilitadores Genéticos , Glicoproteínas/genética , Fator de Crescimento Transformador beta1/genética , Proteínas Adaptadoras de Transdução de Sinal , Animais , Sequência de Bases , Sítios de Ligação , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/metabolismo , Calcitriol/farmacologia , Cromossomos Artificiais Bacterianos/genética , Cromossomos Artificiais Bacterianos/metabolismo , Sequência Conservada , Dexametasona/farmacologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Glicoproteínas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/metabolismo , Camundongos , Oncostatina M/farmacologia , Ligação Proteica , Fator de Crescimento Transformador beta1/metabolismo
4.
J Bone Miner Res ; 31(2): 416-29, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26332516

RESUMO

Receptor activator of nuclear factor-κB ligand (RANKL) is a tumor necrosis factor (TNF)-like cytokine that is necessary for osteoclast formation and survival. Elevated RANKL synthesis is associated with both increased osteoclast number and bone resorption. Earlier studies identified an enhancer 76 kb upstream of the Tnfsf11 transcriptional start site (TSS) termed RL-D5 or the distal control region (DCR) that modulates RANKL expression in response to PTH, 1,25(OH)2D3,, and an array of cytokines. Mice lacking RL-D5 exhibit high bone mass associated with decreased RANKL expression in bone, spleen, and thymus. In addition to RL-D5, genome-wide studies have identified 9 additional Tnfsf11 enhancers residing upstream of the gene's TSS, which provide RANKL cell type-specificity and responsiveness to local and systemic factors. ChIP-chip analyses has revealed inducible vitamin D receptor (VDR) and cAMP response element-binding protein (CREB) binding at an enhancer termed RL-D2 23 kb upstream of the Tnfsf11 TSS in osteoblastic ST2 cells. Herein, we use ChIP-seq analyses to confirm this finding and then delete this enhancer from the mouse genome to determine its physiological role in vivo. RL-D2(-/-) primary stromal cells showed decreased RANKL-induction by both forskolin and 1,25(OH)2D3 ex vivo. Consistent with this, the parathyroid hormone (PTH) induction of RANKL expression was significantly blunted in RL-D2(-/-) mice in vivo. In contrast, lack of RL-D2 had no effect on 1,25(OH)2D3 induction of RANKL in vivo. Similar to the results found in RL-D5(-/-) mice, lack of RL-D2 led to decreased skeletal RANKL expression, resulting in decreased osteoclast numbers and a progressive increase in bone mineral density. Lack of RL-D2 increased cancellous bone mass in femur and spine but did not alter femoral cortical bone thickness. These results highlight the role of distal enhancers in the regulation of RANKL expression by PTH and perhaps 1,25(OH)2D3 and suggest that the RL-D2 and RL-D5 enhancers contribute in either an additive or synergistic manner to regulate bone remodeling.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Osteoblastos/metabolismo , Hormônio Paratireóideo/farmacologia , Fenótipo , Ligante RANK/biossíntese , Elementos de Resposta , Animais , Calcitriol/farmacologia , Colforsina/farmacologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Camundongos , Camundongos Knockout , Tamanho do Órgão/efeitos dos fármacos , Tamanho do Órgão/genética , Osteoblastos/citologia , Ligante RANK/genética , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo
5.
Bone ; 81: 757-764, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25819039

RESUMO

Genomic annotation of unique and combinatorial epigenetic modifications along with transcription factor occupancy is having a profound impact on our understanding of the genome. These studies have led to a better appreciation of the dynamic nature of the epigenetic and transcription factor binding components that reveal overarching principles of the genome as well as tissue specificity. In this minireview, we discuss the presence and potential functions of several of these features across the genome in osteoblast lineage cells. We examine how these features are modulated during cellular maturation, affect transcriptional output and phenotype, and how they alter the ability of cells to respond to systemic signals directed by calcemic hormones such as 1,25-dihydroxyvitamin D3 and PTH. In particular, we describe recent experiments which indicate that progressive stages of bone cell differentiation affect RUNX2 binding to the genome, modify and restrict patterns of gene expression, and dramatically alter cellular response to the vitamin D hormone. These studies expand our understanding of mechanisms that govern steroid hormone regulation of gene expression, while highlighting the increasing complexity that is evident relative to these basic cellular processes. The results also have profound implications with respect to the impact of skeletal diseases on transcriptional outcomes as well. This article is part of a Special Issue entitled Epigenetics and Bone.


Assuntos
Osso e Ossos/citologia , Osso e Ossos/metabolismo , Epigênese Genética , Código das Histonas/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Osso e Ossos/efeitos dos fármacos , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Calcitriol/farmacologia , Diferenciação Celular/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Humanos , Modelos Biológicos , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Hormônio Paratireóideo/farmacologia , Receptores de Calcitriol/metabolismo
6.
Bone ; 72: 81-91, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25460572

RESUMO

Although localized to the mineralized matrix of bone, osteocytes are able to respond to systemic factors such as the calciotropic hormones 1,25(OH)2D3 and PTH. In the present studies, we examined the transcriptomic response to PTH in an osteocyte cell model and found that this hormone regulated an extensive panel of genes. Surprisingly, PTH uniquely modulated two cohorts of genes, one that was expressed and associated with the osteoblast to osteocyte transition and the other a cohort that was expressed only in the mature osteocyte. Interestingly, PTH's effects were largely to oppose the expression of differentiation-related genes in the former cohort, while potentiating the expression of osteocyte-specific genes in the latter cohort. A comparison of the transcriptional effects of PTH with those obtained previously with 1,25(OH)2D3 revealed a subset of genes that was strongly overlapping. While 1,25(OH)2D3 potentiated the expression of osteocyte-specific genes similar to that seen with PTH, the overlap between the two hormones was more limited. Additional experiments identified the PKA-activated phospho-CREB (pCREB) cistrome, revealing that while many of the differentiation-related PTH regulated genes were apparent targets of a PKA-mediated signaling pathway, a reduction in pCREB binding at sites associated with osteocyte-specific PTH targets appeared to involve alternative PTH activation pathways. That pCREB binding activities positioned near important hormone-regulated gene cohorts were localized to control regions of genes was reinforced by the presence of epigenetic enhancer signatures exemplified by unique modifications at histones H3 and H4. These studies suggest that both PTH and 1,25(OH)2D3 may play important and perhaps cooperative roles in limiting osteocyte differentiation from its precursors while simultaneously exerting distinct roles in regulating mature osteocyte function. Our results provide new insight into transcription factor-associated mechanisms through which PTH and 1,25(OH)2D3 regulate a plethora of genes important to the osteoblast/osteocyte lineage.


Assuntos
Calcitriol/química , Regulação da Expressão Gênica , Osteócitos/citologia , Hormônio Paratireóideo/metabolismo , Transcriptoma , Células 3T3 , Animais , Osso e Ossos/metabolismo , Diferenciação Celular , Imunoprecipitação da Cromatina , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Epigênese Genética , Perfilação da Expressão Gênica , Humanos , Camundongos , Osteoblastos/citologia , Análise de Sequência de RNA , Transdução de Sinais
7.
J Bone Miner Res ; 30(5): 855-68, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25431114

RESUMO

Receptor activator of NF-κB ligand (RANKL) is a TNFα-like cytokine that is produced by a diverse set of lineage-specific cells and is involved in a wide variety of physiological processes that include skeletal remodeling, lymph node organogenesis, mammary gland development, and thermal regulation. Consistent with these diverse functions, control of RANKL expression is accomplished in a cell-specific fashion via a set of at least 10 regulatory enhancers that are located up to 170 kb upstream of the gene's transcriptional start site. Here we examined the in vivo consequence of introducing a contiguous DNA segment containing these components into a genetically deleted RANKL null mouse strain. In contrast to RANKL null littermates, null mice containing the transgene exhibited normalized body size, skeletal development, and bone mass as well as normal bone marrow cavities, normalized spleen weights, and the presence of developed lymph nodes. These mice also manifested normalized reproductive capacity, including the ability to lactate and to produce normal healthy litters. Consistent with this, the transgene restored endogenous-like RANKL transcript levels in several RANKL-expressing tissues. Most importantly, restoration of RANKL expression from this segment of DNA was fully capable of rescuing the complex aberrant skeletal and immune phenotype of the RANKL null mouse. RANKL also restored appropriate levels of B220+ IgM+ and B220+ IgD+ B cells in spleen. Finally, we found that RANKL expression from this transgene was regulated by exogenously administered 1,25(OH)2 D3 , parathyroid hormone (PTH), and lipopolysaccharide (LPS), thus recapitulating the ability of these same factors to regulate the endogenous gene. These findings fully highlight the properties of the Tnfsf11 gene locus predicted through previous in vitro dissection. We conclude that the mouse Tnfsf11 gene locus identified originally through unbiased chromatin immunoprecipitation with DNA microarray (ChIP-chip) analysis contains the necessary genetic information to direct appropriate tissue-specific and factor-regulated RANKL expression in vivo.


Assuntos
DNA/genética , Ligante RANK/deficiência , Ligante RANK/genética , Sequências Reguladoras de Ácido Nucleico/genética , Transcrição Gênica , Animais , Linfócitos B/citologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Colecalciferol/farmacologia , Lâmina de Crescimento/efeitos dos fármacos , Lâmina de Crescimento/metabolismo , Humanos , Lipopolissacarídeos/farmacologia , Tecido Linfoide/metabolismo , Camundongos , Camundongos Transgênicos , Especificidade de Órgãos/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteogênese/efeitos dos fármacos , Hormônio Paratireóideo/farmacologia , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Transcrição Gênica/efeitos dos fármacos , Transgenes
8.
Mol Endocrinol ; 28(7): 1150-65, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24877565

RESUMO

Osteocytes are derived from osteoblast lineage cells that become progressively embedded in mineralized bone. Development of the osteocytogenic cell line IDG-SW3 has enabled a temporal and mechanistic investigation of this process. Through RNA-sequencing analyses, we show that although substantial changes in gene expression occur during the osteoblast to osteocyte transition, the majority of the transcriptome remains qualitatively osteoblast like. Genes either up-regulated or expressed uniquely in the osteocyte include local and systemic factors such as Sost and Fgf23 as well as genes implicated in neuronal, muscle, vascular, or regulatory function. As assessed by chromatin immunoprecipitation coupled to high-throughput sequencing, numerous changes in epigenetic histone modifications also occur during osteocytogenesis; these are largely qualitative rather than quantitative. Specific epigenetic changes correlate with altered gene expression patterns that are observed during the transition. These genomic changes likely influence the highly restricted transcriptomic response to 1,25(OH)(2)D(3) that occurs during differentiation. VDR binding in osteocytes revealed an extensive cistrome co-occupied by retinoid X receptor and located predominantly at sites distal to regulated genes. Although sites of VDR binding were apparent near many 1,25(OH)(2)D(3)-regulated genes, the expression of others adjacent to VDR-binding sites were unaffected; lack of VDR binding was particularly prevalent at down-regulated genes. Interestingly, 1,25(OH)(2)D(3) was found to induce the Boc and Cdon coreceptors that are active in hedgehog signaling in osteocytes. We conclude that osteocytogenesis is accompanied by changes in gene expression that may be driven by both genetic and epigenetic components. These changes are likely responsible for the osteocyte phenotype and may contribute to reduced sensitivity to 1,25(OH)(2)D(3).


Assuntos
Conservadores da Densidade Óssea/farmacologia , Calcificação Fisiológica/efeitos dos fármacos , Calcitriol/farmacologia , Osteoblastos/citologia , Osteócitos/citologia , Células 3T3 , Proteínas Adaptadoras de Transdução de Sinal , Animais , Moléculas de Adesão Celular/biossíntese , Moléculas de Adesão Celular/genética , Diferenciação Celular , Linhagem Celular , Imunoprecipitação da Cromatina , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/biossíntese , Fatores de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica , Glicoproteínas/biossíntese , Glicoproteínas/genética , Imunoglobulina G/biossíntese , Imunoglobulina G/genética , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Calcitriol/metabolismo , Receptores de Superfície Celular/biossíntese , Receptores de Superfície Celular/genética , Receptores X de Retinoides/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...