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











Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 11(1): 5535, 2021 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-33692370

RESUMO

Lung cancer rates are rising globally and non-small cell lung cancer (NSCLC) has a five year survival rate of only 24%. Unfortunately, the development of drugs to treat cancer is severely hampered by the inefficiency of translating pre-clinical studies into clinical benefit. Thus, we sought to apply a tumor microenvironment system (TMES) to NSCLC. Using microvascular endothelial cells, lung cancer derived fibroblasts, and NSCLC tumor cells in the presence of in vivo tumor-derived hemodynamic flow and transport, we demonstrate that the TMES generates an in-vivo like biological state and predicts drug response to EGFR inhibitors. Transcriptomic and proteomic profiling indicate that the TMES recapitulates the in vivo and patient molecular biological state providing a mechanistic rationale for the predictive nature of the TMES. This work further validates the TMES for modeling patient tumor biology and drug response indicating utility of the TMES as a predictive tool for drug discovery and development and potential for use as a system for patient avatars.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Células Endoteliais/metabolismo , Neoplasias Pulmonares/metabolismo , Modelos Biológicos , Microambiente Tumoral , Animais , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Células Endoteliais/patologia , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Camundongos SCID
2.
Lab Chip ; 19(7): 1193-1204, 2019 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-30839006

RESUMO

The development of drugs to treat cancer is hampered by the inefficiency of translating pre-clinical in vitro monoculture and mouse studies into clinical benefit. There is a critical need to improve the accuracy of evaluating pre-clinical drug efficacy through the development of more physiologically relevant models. In this study, a human triculture 3D in vitro tumor microenvironment system (TMES) was engineered to accurately mimic the tumor microenvironment. The TMES recapitulates tumor hemodynamics and biological transport with co-cultured human microvascular endothelial cells, pancreatic ductal adenocarcinoma, and pancreatic stellate cells. We demonstrate that significant tumor cell transcriptomic changes occur in the TMES that correlate with the in vivo xenograft and patient transcriptome. Treatment with therapeutically relevant doses of chemotherapeutics yields responses paralleling the patients' clinical responses. Thus, this model provides a unique platform to rigorously evaluate novel therapies and is amenable to using patient tumor material directly, with applicability for patient avatars.


Assuntos
Biomimética/métodos , Carcinoma Ductal Pancreático/patologia , Microambiente Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Microambiente Tumoral/efeitos dos fármacos
3.
JCI Insight ; 1(20): e90954, 2016 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-27942596

RESUMO

A barrier to drug development for nonalcoholic steatohepatitis (NASH) is the absence of translational preclinical human-relevant systems. An in vitro liver model was engineered to incorporate hepatic sinusoidal flow, transport, and lipotoxic stress risk factors (glucose, insulin, free fatty acids) with cocultured primary human hepatocytes, hepatic stellate cells (HSCs), and macrophages. Transcriptomic, lipidomic, and functional endpoints were evaluated and compared with clinical data from NASH patient biopsies. The lipotoxic milieu promoted hepatocyte lipid accumulation (4-fold increase, P < 0.01) and a lipidomics signature similar to NASH biopsies. Hepatocyte glucose output increased with decreased insulin sensitivity. These changes were accompanied by increased inflammatory analyte secretion (e.g., IL-6, IL-8, alanine aminotransferase). Fibrogenic activation markers increased with lipotoxic conditions, including secreted TGF-ß (>5-fold increase, P < 0.05), extracellular matrix gene expression, and HSC activation. Significant pathway correlation existed between this in vitro model and human biopsies. Consistent with clinical trial data, 0.5 µM obeticholic acid in this model promoted a healthy lipidomic signature, reduced inflammatory and fibrotic secreted factors, but also increased ApoB secretion, suggesting a potential adverse effect on lipoprotein metabolism. Lipotoxic stress activates similar biological signatures observed in NASH patients in this system, which may be relevant for interrogating novel therapeutic approaches to treat NASH.


Assuntos
Técnicas de Cocultura , Células Estreladas do Fígado/citologia , Hepatócitos/citologia , Macrófagos/citologia , Hepatopatia Gordurosa não Alcoólica/fisiopatologia , Animais , Glucose/metabolismo , Humanos , Inflamação , Resistência à Insulina , Lipídeos/análise , Fígado , Metaboloma , Camundongos Endogâmicos C57BL , Modelos Biológicos , Transcriptoma
4.
Artigo em Inglês | MEDLINE | ID: mdl-23143566

RESUMO

Previous research has demonstrated that acoustic radiation force enhances intravascular microbubble adhesion to blood vessels in the presence of flow for moleculartargeted ultrasound imaging and drug delivery. A prototype acoustic radiation force intravascular ultrasound (ARFIVUS) catheter was designed and fabricated to displace a microbubble contrast agent in flow representative of conditions encountered in the human carotid artery. The prototype ARFIVUS transducer was designed to match the resonance frequency of 1.4- to 2.6-µm-diameter microbubbles modeled by an experimentally verified 1-D microbubble acoustic radiation force translation model. The transducer element was an elongated Navy Type I (hard) lead zirconate titanate (PZT) ceramic designed to operate at 3 MHz. Fabricated devices operated with center frequencies of 3.3 and 3.6 MHz with -6-dB fractional bandwidths of 55% and 50%, respectively. Microbubble translation velocities as high as 0.86 m/s were measured using a high-speed streak camera when insonating with the ARFIVUS transducer. Finally, the prototype was used to displace microbubbles in a flow phantom while imaging with a commercial 45-MHz imaging IVUS transducer. A sustained increase of 31 dB in average video intensity was measured following insonation with the ARFIVUS, indicating microbubble accumulation resulting from the application of acoustic radiation force.


Assuntos
Catéteres , Sistemas de Liberação de Medicamentos/instrumentação , Microbolhas , Ultrassonografia de Intervenção/instrumentação , Artérias Carótidas/diagnóstico por imagem , Meios de Contraste/química , Meios de Contraste/farmacocinética , Sistemas de Liberação de Medicamentos/métodos , Análise de Elementos Finitos , Humanos , Modelos Cardiovasculares , Imagens de Fantasmas , Transdutores , Ultrassonografia de Intervenção/métodos
5.
Am J Physiol Cell Physiol ; 302(1): C1-8, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21998140

RESUMO

Tonicity-responsive enhancer binding protein (TonEBP/nuclear factor of activated T-cells 5 [NFAT5]) is a Rel homology transcription factor classically known for its osmosensitive role in regulating cellular homeostasis during states of hypo- and hypertonic stress. A recently growing body of research indicates that TonEBP is not solely regulated by tonicity, but that it can be stimulated by various tonicity-independent mechanisms in both hypertonic and isotonic tissues. Physiological and pathophysiological stimuli such as cytokines, growth factors, receptor and integrin activation, contractile agonists, ions, and reactive oxygen species have been implicated in the positive regulation of TonEBP expression and activity in diverse cell types. These new data demonstrate that tonicity-independent stimulation of TonEBP is critical for tissue-specific functions like enhanced cell survival, migration, proliferation, vascular remodeling, carcinoma invasion, and angiogenesis. Continuing research will provide a better understanding as to how these and other alternative TonEBP stimuli regulate gene expression in both health and disease.


Assuntos
Osmose/fisiologia , Fatores de Transcrição/metabolismo , Animais , Humanos , Camundongos , Fatores de Transcrição/fisiologia
6.
Arterioscler Thromb Vasc Biol ; 31(11): 2665-75, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21903947

RESUMO

OBJECTIVE: Calcineurin (Cn) and the nuclear factor of activated T cells (NFAT) family of transcription factors are critical in vascular smooth muscle cell (SMC) development and pathology. Here, we used a genomics approach to identify and validate NFAT gene targets activated during platelet-derived growth factor-BB (PDGF-BB)-induced SMC phenotypic modulation. METHODS AND RESULTS: Genome-wide expression arrays were used to identify genes both (1) differentially activated in response to PDGF-BB and (2) whose differential expression was reduced by both the Cn inhibitor cyclosporin A and the NFAT inhibitor A-285222. The 20 most pharmacologically sensitive genes were validated by quantitative reverse transcription-polymerase chain reaction analysis of PDGF-BB-stimulated SMCs in the presence of Cn/NFAT inhibitors, including the VIVIT peptide. In all experiments, protein C receptor (PROCR) gene activation was reduced. We showed that PROCR expression was virtually absent in untreated, quiescent SMCs. PDGF-BB stimulation, however, induced significant PROCR promoter activation and downstream protein expression in a Cn/NFAT-dependent manner. Mutation of a species-conserved, NFAT binding motif significantly attenuated PDGF-BB-induced PROCR promoter activity, thereby distinguishing NFAT as the first PROCR transcriptional activator to date. Moreover, SMC PROCR expression was upregulated in the neointima as early as 7 days following acute vascular injury in rat carotid arteries. CONCLUSION: We hereby report PROCR as a novel, NFAT-dependent gene that may be implicated in vascular restenosis and consequent inward remodeling.


Assuntos
Fatores de Coagulação Sanguínea/genética , Calcineurina/genética , Genoma/genética , Músculo Liso Vascular/patologia , Fatores de Transcrição NFATC/genética , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Receptores de Superfície Celular/genética , Animais , Sequência de Bases , Becaplermina , Fatores de Coagulação Sanguínea/metabolismo , Calcineurina/metabolismo , Inibidores de Calcineurina , Lesões das Artérias Carótidas/metabolismo , Cateterismo/efeitos adversos , Células Cultivadas , Ciclosporina/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Modelos Animais , Dados de Sequência Molecular , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Fatores de Transcrição NFATC/antagonistas & inibidores , Fatores de Transcrição NFATC/metabolismo , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Proto-Oncogênicas c-sis , Pirazóis/farmacologia , Ratos , Receptores de Superfície Celular/metabolismo
7.
Arterioscler Thromb Vasc Biol ; 31(10): 2287-96, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21757659

RESUMO

OBJECTIVE: The tonicity-responsive transcription factor, nuclear factor of activated T cells 5 (NFAT5/tonicity enhancer binding protein [TonEBP]), has been well characterized in numerous cell types; however, NFAT5 function in vascular smooth muscle cells (SMCs) is unknown. Our main objective was to determine the role of NFAT5 regulation in SMCs. METHODS AND RESULTS: We showed that NFAT5 is regulated by hypertonicity in SMCs and is upregulated in atherosclerosis and neointimal hyperplasia. RNAi knockdown of NFAT5 inhibited basal expression of several SMC differentiation marker genes, including smooth muscle α actin (SMαA). Bioinformatic analysis of SMαA revealed 7 putative NFAT5 binding sites in the first intron, and chromatin immunoprecipitation analysis showed NFAT5 enrichment of intronic DNA. Overexpression of NFAT5 increased SMαA promoter-intron activity, which requires an NFAT5 cis element at +1012, whereas dominant-negative NFAT5 decreased SMαA promoter-intron activity. Because it is unlikely that SMCs experience extreme changes in tonicity, we investigated other stimuli and uncovered 2 novel NFAT5-inducing factors: angiotensin II, a contractile agonist, and platelet-derived growth factor-BB (PDGF-BB), a potent mitogen in vascular injury. Angiotensin II stimulated NFAT5 translocation and activity, and NFAT5 knockdown inhibited an angiotensin II-mediated upregulation of SMαA mRNA. PDGF-BB increased NFAT5 protein, and loss of NFAT5 inhibited PDGF-BB-induced SMC migration. CONCLUSIONS: We have identified NFAT5 as a novel regulator of SMC phenotypic modulation and have uncovered the role of NFAT5 in angiotensin II-induced SMαA expression and PDGF-BB-stimulated SMC migration.


Assuntos
Aterosclerose/metabolismo , Lesões das Artérias Carótidas/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Fatores de Transcrição NFATC/metabolismo , Túnica Íntima/metabolismo , Actinas/genética , Actinas/metabolismo , Angiotensina II/metabolismo , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/patologia , Becaplermina , Sítios de Ligação , Lesões das Artérias Carótidas/genética , Lesões das Artérias Carótidas/patologia , Movimento Celular , Proliferação de Células , Células Cultivadas , Imunoprecipitação da Cromatina , Técnicas de Cocultura , Biologia Computacional , Modelos Animais de Doenças , Regulação da Expressão Gênica , Genes Reporter , Humanos , Hiperplasia , Íntrons , Camundongos , Camundongos Knockout , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Fatores de Transcrição NFATC/genética , Fenótipo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Regiões Promotoras Genéticas , Transporte Proteico , Proteínas Proto-Oncogênicas c-sis , Interferência de RNA , RNA Mensageiro/metabolismo , Ratos , Fatores de Transcrição/metabolismo , Transfecção , Túnica Íntima/patologia
8.
J Control Release ; 154(1): 42-9, 2011 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-21549778

RESUMO

Microbubble contrast agents have been shown to enhance reagent delivery when activated by ultrasound. We hypothesized that ultrasound would enhance delivery of rapamycin, an antiproliferative agent, from the shell of microbubbles, thus reducing proliferation of vascular smooth muscle cells. Our objective was to determine optimal ultrasound parameters that maximized therapeutic efficacy, maintained cell adherence, and minimized the drug exposure time. In vitro assays determined that ultrasound (1 MHz, 0.5% duty cycle) is required to successfully deliver rapamycin from microbubbles and reduce proliferation. Co-injection of rapamycin with control microbubbles did not result in a reduction in proliferation. Successful reduction in proliferation (>50%) required pulses at least 10 cycles in length and at least 300 kPa peak negative pressure at which point 90% of cells remained adherent. The anti-proliferative effect was also localized within a 6mm wide zone by focusing the ultrasound beam.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Proliferação de Células/efeitos dos fármacos , Portadores de Fármacos/química , Microbolhas , Músculo Liso Vascular/efeitos dos fármacos , Fonoforese , Sirolimo/administração & dosagem , Animais , Antibióticos Antineoplásicos/efeitos adversos , Células Cultivadas , Composição de Medicamentos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Microscopia de Contraste de Fase , Músculo Liso Vascular/citologia , Músculo Liso Vascular/patologia , Ratos , Sirolimo/efeitos adversos , Fatores de Tempo
9.
Circ Res ; 107(6): 737-46, 2010 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-20651288

RESUMO

RATIONALE: Macrophages change their phenotype and biological functions depending on the microenvironment. In atherosclerosis, oxidative tissue damage accompanies chronic inflammation; however, macrophage phenotypic changes in response to oxidatively modified molecules are not known. OBJECTIVE: To examine macrophage phenotypic changes in response to oxidized phospholipids that are present in atherosclerotic lesions. METHODS AND RESULTS: We show that oxidized phospholipid-treated murine macrophages develop into a novel phenotype (Mox) that is strikingly different from the conventional M1 and M2 macrophage phenotypes. Compared to M1 and M2, Mox macrophages show a different gene expression pattern, as well as decreased phagocytotic and chemotactic capacity. Treatment with oxidized phospholipids induces both M1 and M2 macrophages to switch to the Mox phenotype. Whole-genome expression array analysis and subsequent gene ontology clustering revealed that the Mox phenotype was characterized by abundant overrepresentation of Nrf2-mediated expression of redox-regulatory genes. In macrophages isolated from Nrf2(-/-) mice, oxidized phospholipid-induced gene expression and regulation of redox status were compromised. Moreover, we found that Mox macrophages comprise 30% of all macrophages in advanced atherosclerotic lesions of low-density lipoprotein receptor knockout (LDLR(-/-)) mice. CONCLUSIONS: Together, we identify Nrf2 as a key regulator in the formation of a novel macrophage phenotype (Mox) that develops in response to oxidative tissue damage. The unique biological properties of Mox macrophages suggest this phenotype may play an important role in atherosclerotic lesion development as well as in other settings of chronic inflammation.


Assuntos
Aterosclerose/metabolismo , Imunofenotipagem , Macrófagos/fisiologia , Fator 2 Relacionado a NF-E2/fisiologia , Fosfolipídeos/fisiologia , Animais , Aterosclerose/genética , Células Cultivadas , Feminino , Macrófagos/classificação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 2 Relacionado a NF-E2/biossíntese , Fator 2 Relacionado a NF-E2/deficiência , Fator 2 Relacionado a NF-E2/genética , Oxirredução , Fosfolipídeos/metabolismo
10.
J Pharmacol Exp Ther ; 333(1): 34-42, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20089806

RESUMO

Cyclosporine A (CSA, calcineurin inhibitor) has been shown to block both vascular smooth muscle cell (VSMC) proliferation in cell culture and vessel neointimal formation following injury in vivo. The purpose of this study was to determine molecular and pathological effects of CSA on VSMCs. Using real-time reverse transcription-polymerase chain reaction, Western blot analysis, and immunofluorescence microscopy, we show that CSA up-regulated the expression of Krüppel-like factor-4 (KLF4) in VSMCs. KLF4 plays a key role in regulating VSMC phenotypic modulation. KLF4 antagonizes proliferation, facilitates migration, and down-regulates VSMC differentiation marker gene expression. We show that the VSMC differentiation marker genes smooth muscle alpha-actin (ACTA2), transgelin (TAGLN), smoothelin (SMTN), and myocardin (MYOCD) are all down-regulated by CSA in VSMC monoculture, whereas cyclin-dependent kinase inhibitor-1A (CDKN1A) and matrix metalloproteinase-3 (MMP3) are up-regulated. CSA did not affect the abundance of the VSMC microRNA (MIR) markers MIR143 and MIR145. Administration of CSA to rat carotid artery in vivo resulted in acute and transient suppression of ACTA2, TAGLN, SMTN, MYOCD, and smooth muscle myosin heavy chain (MYH11) mRNA levels. The tumor suppressor genes KLF4, p53, and CDKN1A, however, were up-regulated, as well as MMP3, MMP9, and collagen-VIII. CSA-treated arteries showed remarkable remodeling, including breakdown of the internal elastic lamina and reorientation of VSMCs, as well as increased KLF4 immunostaining in VSMCs and endothelial cells. Altogether, these data show that cyclosporin up-regulates KLF4 expression and promotes phenotypic modulation of VSMCs.


Assuntos
Ciclosporina/farmacologia , Imunossupressores/farmacologia , Fatores de Transcrição Kruppel-Like/biossíntese , Miócitos de Músculo Liso/efeitos dos fármacos , Animais , Antígenos de Diferenciação/metabolismo , Aorta/citologia , Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/patologia , Diferenciação Celular , Células Cultivadas , Ciclosporina/efeitos adversos , Regulação para Baixo , Imunossupressores/efeitos adversos , Fator 4 Semelhante a Kruppel , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Ratos , Ratos Sprague-Dawley , Túnica Média/efeitos dos fármacos , Túnica Média/patologia , Regulação para Cima
11.
Arterioscler Thromb Vasc Biol ; 30(2): 218-24, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19965778

RESUMO

OBJECTIVE: Hyperglycemia is a recognized risk factor for cardiovascular disease in diabetes. Recently, we reported that high glucose activates the Ca(2+)/calcineurin-dependent transcription factor nuclear factor of activated T cells (NFAT) in arteries ex vivo. Here, we sought to determine whether hyperglycemia activates NFAT in vivo and whether this leads to vascular complications. METHODS AND RESULTS: An intraperitoneal glucose-tolerance test in mice increased NFATc3 nuclear accumulation in vascular smooth muscle. Streptozotocin-induced diabetes resulted in increased NFATc3 transcriptional activity in arteries of NFAT-luciferase transgenic mice. Two NFAT-responsive sequences in the osteopontin (OPN) promoter were identified. This proinflammatory cytokine has been shown to exacerbate atherosclerosis and restenosis. Activation of NFAT resulted in increased OPN mRNA and protein in native arteries. Glucose-induced OPN expression was prevented by the ectonucleotidase apyrase, suggesting a mechanism involving the release of extracellular nucleotides. The calcineurin inhibitor cyclosporin A or the novel NFAT blocker A-285222 prevented glucose-induced OPN expression. Furthermore, diabetes resulted in higher OPN expression, which was significantly decreased by in vivo treatment with A-285222 for 4 weeks or prevented in arteries from NFATc3(-/-) mice. CONCLUSIONS: These results identify a glucose-sensitive transcription pathway in vivo, revealing a novel molecular mechanism that may underlie vascular complications of diabetes.


Assuntos
Glicemia/metabolismo , Diabetes Mellitus Experimental/metabolismo , Angiopatias Diabéticas/etiologia , Hiperglicemia/metabolismo , Músculo Liso Vascular/metabolismo , Fatores de Transcrição NFATC/metabolismo , Osteopontina/metabolismo , Animais , Apirase/farmacologia , Artérias/metabolismo , Sítios de Ligação , Calcineurina/metabolismo , Inibidores de Calcineurina , Ciclosporina/farmacologia , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/tratamento farmacológico , Angiopatias Diabéticas/metabolismo , Angiopatias Diabéticas/prevenção & controle , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Feminino , Teste de Tolerância a Glucose , Humanos , Hiperglicemia/complicações , Hiperglicemia/tratamento farmacológico , Células Jurkat , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/efeitos dos fármacos , Fatores de Transcrição NFATC/antagonistas & inibidores , Fatores de Transcrição NFATC/deficiência , Fatores de Transcrição NFATC/genética , Osteopontina/deficiência , Osteopontina/genética , Regiões Promotoras Genéticas , Pirazóis/farmacologia , RNA Mensageiro/metabolismo , Transdução de Sinais , Fatores de Tempo , Ativação Transcricional , Transfecção , Uridina Trifosfato/metabolismo
12.
Circ Res ; 100(6): 817-25, 2007 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-17322171

RESUMO

Lipoma preferred partner (LPP) is a proline rich LIM domain family protein highly expressed at plasma membrane dense bodies and focal adhesions in smooth muscle cells.(1) Using the C-terminus of LPP as bait in a yeast two hybrid system, palladin, an actin-associated protein was identified. The palladin interacting region of LPP was mapped to the first and second LIM domains. The N-terminus of palladin interacted with LPP both in vitro and in vivo, but not solely through its FPLPPP and FPPPP motifs. Like LPP, palladin, is highly expressed in differentiated smooth muscle, colocalized at focal adhesions, at isolated lamellipodia and at dense bodies in smooth muscle tissue. Both LPP and palladin enhanced cell migration and spreading. LPP and palladin expression was markedly decreased, in contrast to vinculin or paxillin, in migration defective focal adhesion kinase null cells, but was restored by expression of the paired-related homeobox gene-1 protein. We have previously shown in focal adhesion kinase null cells, that tetracycline induced expression of focal adhesion kinase upregulated expression of LPP(2) and now show upregulation of palladin, and paired-related homeobox gene-1 protein. The expression of both LPP and palladin, like smooth muscle alpha-actin, was increased by angiotensin II, regulated by actin dynamics, upregulated by myocardin and expressed in the neointima of injured aorta. Overall, the data suggest that the function of LPP and palladin is context dependent, that they play a critical role in cytoskeletal remodeling, respond to signals induced by vascular injury as well as signals that induce smooth muscle cell hypertrophy, such as angiotension II.


Assuntos
Angiotensina II/farmacologia , Proteínas do Citoesqueleto/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/fisiologia , Proteínas de Homeodomínio/fisiologia , Músculo Liso Vascular/metabolismo , Fosfoproteínas/metabolismo , Actinas/metabolismo , Animais , Aorta/lesões , Aorta/metabolismo , Western Blotting , Linhagem Celular , Movimento Celular/fisiologia , Proteínas do Citoesqueleto/genética , Modelos Animais de Doenças , Adesões Focais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Transferência de Genes , Humanos , Proteínas com Domínio LIM , Camundongos , Especificidade de Órgãos , Fosfoproteínas/genética , Estrutura Terciária de Proteína/fisiologia , Pseudópodes/metabolismo , Ratos , Ratos Sprague-Dawley , Técnicas do Sistema de Duplo-Híbrido
14.
J Appl Physiol (1985) ; 101(3): 752-62, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16763107

RESUMO

Aerobic exercise training is known to have profound cardioprotective effects in disease, yet cellular mechanisms remain largely undefined. We tested the hypothesis that increased sarcoplasmic reticulum Ca(2+) buffering and increased voltage-gated Ca(2+) channel density underlie coronary smooth muscle intracellular Ca(2+) (Ca(2+)(i)) dysregulation in diabetic dyslipidemia and that exercise training would prevent these increases. Yucatan swine were maintained in 1) control, 2) alloxan-induced hyperglycemic, 3) high fat/cholesterol fed, 4) hyperglycemic plus high fat/cholesterol fed (diabetic dyslipidemic), and 5) diabetic dyslipidemic plus exercise-trained (treadmill running) conditions. After 20 wk, the heart was removed and smooth muscle cells isolated from the right coronary artery. We utilized fura-2 imaging of Ca(2+)(i) levels to separate the functional role of the sarcoendoplasmic reticulum Ca(2+)-ATPase (SERCA) from the Na(+)-Ca(2+) exchanger and the plasmalemmal Ca(2+)-ATPase, and whole-cell patch clamp to examine voltage-gated Ca(2+) channel current density (i.e., Ca(2+) influx). Results indicated that diabetic dyslipidemia impaired plasmalemmal Ca(2+) efflux, increased basal Ca(2+)(i) levels, increased SERCA protein and sarcoplasmic reticulum Ca(2+)(i) buffering, and elicited an approximately 50% decrease in voltage-gated Ca(2+) channel current density. Exercise training concurrent with the diabetic dyslipidemic state restored plasmalemmal Ca(2+) efflux, SERCA protein, sarcoplasmic reticulum Ca(2+)(i) buffering, and voltage-gated Ca(2+) channel current density to control levels. Interestingly, basal Ca(2+)(i) levels were significantly lower in the exercise-trained group compared with control. Collectively, these results demonstrate a crucial role for exercise in the prevention of diabetic dyslipidemia-induced Ca(2+)(i) dysregulation.


Assuntos
ATPases Transportadoras de Cálcio/metabolismo , Cálcio/metabolismo , Vasos Coronários/fisiopatologia , Complicações do Diabetes/fisiopatologia , Dislipidemias/fisiopatologia , Músculo Liso Vascular/fisiopatologia , Condicionamento Físico Animal/métodos , Animais , Complicações do Diabetes/terapia , Dislipidemias/complicações , Dislipidemias/terapia , Terapia por Exercício/métodos , Masculino , Esforço Físico , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático , Suínos , Resultado do Tratamento
15.
Stem Cells ; 24(7): 1678-88, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16601077

RESUMO

The aims of this study were to develop a method for deriving purified populations of contractile smooth muscle cells (SMCs) from embryonic stem cells (ESCs) and to characterize their function. Transgenic ESC lines were generated that stably expressed a puromycin-resistance gene under the control of either a smooth muscle alpha-actin (SMalphaAlpha) or smooth muscle-myosin heavy chain (SM-MHC) promoter. Negative selection, either overnight or for 3 days, was then used to purify SMCs from embryoid bodies. Purified SMCs expressed multiple SMC markers by immunofluorescence, immunoblotting, quantitative reverse transcription-polymerase chain reaction, and flow cytometry and were designated APSCs (SMalphaAlpha-puromycin-selected cells) or MPSCs (SM-MHC-puromycin-selected cells), respectively. Both SMC lines displayed agonist-induced Ca(2+) transients, expressed functional Ca(2+) channels, and generated contractile force when aggregated within collagen gels and stimulated with vasoactive agonists, such as endothelin-1, or in response to depolarization with KCl. Importantly, subcutaneous injection of APSCs or MPSCs subjected to 18 hours of puromycin selection led to the formation of teratomas, presumably due to residual contamination by pluripotent stem cells. In contrast, APSCs or MPSCs subjected to prolonged puromycin selection for 3 days did not form teratomas in vivo. These studies describe for the first time a method for generating relatively pure populations of SMCs from ESCs which display appropriate excitation and contractile responses to vasoactive agonists. However, studies also indicate the potential for teratoma development in ESC-derived cell lines, even after prolonged differentiation, highlighting the critical requirement for efficient methods of separating differentiated cells from residual pluripotent precursors in future studies that use ESC derivatives, whether SMC or other cell types, in tissue engineering applications.


Assuntos
Embrião de Mamíferos/citologia , Indução Embrionária , Contração Muscular/fisiologia , Miócitos de Músculo Liso/fisiologia , Células-Tronco/fisiologia , Actinas/genética , Actinas/metabolismo , Animais , Cálcio/metabolismo , Células Cultivadas , Colágeno/metabolismo , Marcadores Genéticos , Camundongos , Morfogênese , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Neoplasias/prevenção & controle , Regiões Promotoras Genéticas , Seleção Genética , Transgenes , Vasoconstritores/farmacologia
16.
Circ Res ; 97(10): 1027-35, 2005 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-16210550

RESUMO

Bone marrow-derived cells (BMCs) have been implicated as a modifiers of vascular growth either directly by transdifferentiation into endothelial cells (ECs) or indirectly through growth factor release. To examine these possibilities under physiological conditions, we developed a model of hypoxia-mediated angiogenesis in the mouse spinotrapezius muscle. This allows whole-mount analysis; therefore, the morphology and location of BMCs within the vascular network may be observed along with differentiation markers. We exposed bone marrow transplant chimeric mice to hypoxia and treated a subset with granulocyte macrophage colony-stimulating factor. Exposure to hypoxia caused an 13% increase in capillary density relative to control. Hypoxia did not increase the overall number of muscle-resident BMCs, but did increase the number of rounded BMCs by 25%. There was no discernable BMC contribution to the endothelium, although some BMCs assumed a pericyte morphology around capillaries. Granulocyte macrophage colony-stimulating factor treatment further increased the number of round BMCs within the muscle and caused a 23% increase in angiogenesis. The results of this study suggest a potentially beneficial action of BMCs during hypoxia through paracrine release of growth factors but not transdifferentiation into ECs.


Assuntos
Células da Medula Óssea/fisiologia , Diferenciação Celular , Células Endoteliais/citologia , Mobilização de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/fisiologia , Hipóxia/fisiopatologia , Neovascularização Fisiológica , Animais , Células da Medula Óssea/citologia , Contagem de Células , Células-Tronco Hematopoéticas/citologia , Hipóxia/patologia , Metaloproteinase 9 da Matriz/análise , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/patologia , Músculo Liso Vascular/citologia , Fator A de Crescimento do Endotélio Vascular/análise
17.
J Clin Invest ; 115(2): 418-27, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15690088

RESUMO

CC(A/T)6GG-dependent (CArG-dependent) and serum response factor-dependent (SRF-dependent) mechanisms are required for gene expression in smooth muscle cells (SMCs). However, an unusual feature of many SMC-selective promoter CArG elements is that they contain a conserved single G or C substitution in their central A/T-rich region, which reduces binding affinity for ubiquitously expressed SRF. We hypothesized that this CArG degeneracy contributes to cell-specific expression of smooth muscle alpha-actin in vivo, since substitution of c-fos consensus CArGs for the degenerate CArGs resulted in relaxed specificity in cultured cells. Surprisingly, our present results show that these substitutions have no effect on smooth muscle-specific transgene expression during normal development and maturation in transgenic mice. However, these substitutions significantly attenuated injury-induced downregulation of the mutant transgene under conditions where SRF expression was increased but expression of myocardin, a smooth muscle-selective SRF coactivator, was decreased. Finally, chromatin immunoprecipitation analyses, together with cell culture studies, suggested that myocardin selectively enhanced SRF binding to degenerate versus consensus CArG elements. Our results indicate that reductions in myocardin expression and the degeneracy of CArG elements within smooth muscle promoters play a key role in phenotypic switching of smooth muscle cells in vivo, as well as in mediating responses of CArG-dependent smooth muscle genes and growth regulatory genes under conditions in which these 2 classes of genes are differentially expressed.


Assuntos
Actinas/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Crescimento/fisiologia , Músculo Liso/embriologia , Miócitos de Músculo Liso/metabolismo , Elemento de Resposta Sérica/fisiologia , Actinas/genética , Animais , Cromatina/metabolismo , Imunoprecipitação da Cromatina , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento/genética , Crescimento/genética , Camundongos , Camundongos Transgênicos , Músculo Liso/lesões , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Especificidade de Órgãos/genética , Especificidade de Órgãos/fisiologia , Mutação Puntual/fisiologia , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Elemento de Resposta Sérica/genética , Transativadores/genética , Transativadores/metabolismo
18.
J Clin Invest ; 113(9): 1249-51, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15124012

RESUMO

What are the true origins of the smooth muscle cells (SMCs) present in the intimal lesions of transplant arteriosclerosis? A new study in the JCI shows that Sca-1(+) cells purified from the mouse aortic root can migrate through an irradiated vein graft to the neointima of the vessel and transdifferentiate to express the early SMC differentiation marker gene SM22. Do Sca-1(+) cells transdifferentiate into SMC-like cells, or is activation of SMC marker genes a consequence of fusion of these cells with preexisting SMCs, a possibility raised by results of studies of adult stem cells in animal models of liver regeneration ? Or could this be bona fide transdifferentiation that recapitulates the pathologic processes in humans?


Assuntos
Diferenciação Celular , Músculo Liso Vascular/transplante , Veias/transplante , Animais , Aorta/citologia , Arteriosclerose/patologia , Células da Medula Óssea/citologia , Fusão Celular , Linhagem da Célula , Movimento Celular , Células Cultivadas , Modelos Animais de Doenças , Marcadores Genéticos , Células-Tronco Hematopoéticas/citologia , Humanos , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Proteínas Musculares/genética , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Doadores de Tecidos , Transplante Homólogo , Túnica Íntima/patologia , Veias/efeitos da radiação
19.
Circ Res ; 92(8): 856-64, 2003 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-12663482

RESUMO

The interactions between serum response factor (SRF) and CArG elements are critical for smooth muscle cell (SMC) marker gene transcription. However, the mechanisms whereby SRF, which is expressed ubiquitously, contributes to SMC-specific transcription are unknown. Myocardin was recently cloned as a coactivator of SRF in the heart, but its role in regulating CArG-dependent expression of SMC differentiation marker genes has not been clearly elucidated. In this study, we examined the expression and the function of myocardin in SMCs. In adult mice, myocardin mRNA was expressed in multiple smooth muscle (SM) tissues including the aorta, bladder, stomach, intestine, and colon, as well as the heart. Myocardin was also expressed in cultured rat aortic SMCs and A404 SMC precursor cells. Of particular interest, expression of myocardin was induced during differentiation of A404 cells, although it was not expressed in parental P19 cells from which A404 cells were derived. Cotransfection studies in SMCs revealed that myocardin induced the activity of multiple SMC marker gene promoters including SM alpha-actin, SM-myosin heavy chain, and SM22alpha by 9- to 60-fold in a CArG-dependent manner, whereas myocardin short interfering RNA markedly decreased activity of these promoters. Moreover, adenovirus-mediated overexpression of a dominant-negative form of myocardin significantly suppressed expression of endogenous SMC marker genes, whereas adenovirus-mediated overexpression of wild-type myocardin increased expression. Taken together, results provide compelling evidence that myocardin plays a key role as a transcriptional coactivator of SMC marker genes through CArG-dependent mechanisms.


Assuntos
Músculo Liso Vascular/metabolismo , Proteínas Nucleares/genética , Elementos de Resposta/genética , Transativadores/genética , Transcrição Gênica/genética , Células 3T3 , Animais , Biomarcadores , Linhagem Celular , Células Cultivadas , Ensaio de Desvio de Mobilidade Eletroforética , Expressão Gênica , Regulação da Expressão Gênica , Vetores Genéticos/genética , Camundongos , Músculo Liso Vascular/citologia , Mutação , Miocárdio/citologia , Miocárdio/metabolismo , Proteínas Nucleares/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/fisiologia , Ratos , Elementos de Resposta/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Resposta Sérica/metabolismo , Transativadores/fisiologia , Transfecção
20.
Am J Physiol Endocrinol Metab ; 283(5): E1040-5, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12376333

RESUMO

Muscle contractions cause numerous disturbances in intracellular homeostasis. This makes it impossible to use contracting muscle to identify which of the many signals generated by contractions are responsible for stimulating mitochondrial biogenesis. One purpose of this study was to evaluate the usefulness of L6 myotubes, which do not contract, for studying mitochondrial biogenesis. A second purpose was to evaluate further the possibility that increases in cytosolic Ca2+ can stimulate mitochondrial biogenesis. Continuous exposure to 1 microM ionomycin, a Ca2+ ionophore, for 5 days induced an increase in mitochondrial enzymes but also caused a loss of myotubes, as reflected in an approximately 40% decrease in protein per dish. However, intermittent (5 h/day) exposure to ionomycin, or to caffeine or W7, which release Ca2+ from the sarcoplasmic reticulum, did not cause a decrease in protein per dish. Raising cytosolic Ca2+ intermittently with these agents induced significant increases in mitochondrial enzymes. EGTA blocked most of this effect of ionomycin, whereas dantrolene, which blocks Ca2+ release from the sarcoplasmic reticulum, largely prevented the increases in mitochondrial enzymes induced by W7 and caffeine. These findings provide evidence that intermittently raising cytosolic Ca2+ stimulates mitochondrial biogenesis in muscle cells.


Assuntos
Aminoimidazol Carboxamida/análogos & derivados , Cálcio/metabolismo , Mitocôndrias/metabolismo , Fibras Musculares Esqueléticas/metabolismo , 5-Aminolevulinato Sintetase/metabolismo , Trifosfato de Adenosina/metabolismo , Aminoimidazol Carboxamida/farmacologia , Animais , Cafeína/farmacologia , Células Cultivadas , Grupo dos Citocromos c/metabolismo , Citosol/metabolismo , Inibidores Enzimáticos/farmacologia , Expressão Gênica/fisiologia , Concentração de Íons de Hidrogênio , Hipoglicemiantes/farmacologia , Ionomicina/farmacologia , Ionóforos/farmacologia , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/efeitos dos fármacos , Ácido Oleico/metabolismo , Oxirredução , Inibidores de Fosfodiesterase/farmacologia , Ratos , Ribonucleotídeos/farmacologia , Sulfonamidas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA