Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
J Neuroinflammation ; 17(1): 194, 2020 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-32560730

RESUMO

BACKGROUND: Ischemic stroke is a devastating disease without a cure. The available treatments for ischemic stroke, thrombolysis by tissue plasminogen activator, and thrombectomy are suitable only to a fraction of patients and thus novel therapeutic approaches are urgently needed. The neuroinflammatory responses elicited secondary to the ischemic attack further aggravate the stroke-induced neuronal damage. It has been demonstrated that these responses are regulated at the level of non-coding RNAs, especially miRNAs. METHODS: We utilized lentiviral vectors to overexpress miR-669c in BV2 microglial cells in order to modulate their polarization. To detect whether the modulation of microglial activation by miR-669c provides protection in a mouse model of transient focal ischemic stroke, miR-669c overexpression was driven by a lentiviral vector injected into the striatum prior to induction of ischemic stroke. RESULTS: Here, we demonstrate that miR-669c-3p, a member of chromosome 2 miRNA cluster (C2MC), is induced upon hypoxic and excitotoxic conditions in vitro and in two different in vivo models of stroke. Rather than directly regulating the neuronal survival in vitro, miR-669c is capable of attenuating the microglial proinflammatory activation in vitro and inducing the expression of microglial alternative activation markers arginase 1 (Arg1), chitinase-like 3 (Ym1), and peroxisome proliferator-activated receptor gamma (PPAR-γ). Intracerebral overexpression of miR-669c significantly decreased the ischemia-induced cell death and ameliorated the stroke-induced neurological deficits both at 1 and 3 days post injury (dpi). Albeit miR-669c overexpression failed to alter the overall Iba1 protein immunoreactivity, it significantly elevated Arg1 levels in the ischemic brain and increased colocalization of Arg1 and Iba1. Moreover, miR-669c overexpression under cerebral ischemia influenced several morphological characteristics of Iba1 positive cells. We further demonstrate the myeloid differentiation primary response gene 88 (MyD88) transcript as a direct target for miR-669c-3p in vitro and show reduced levels of MyD88 in miR-669c overexpressing ischemic brains in vivo. CONCLUSIONS: Collectively, our data provide the evidence that miR-669c-3p is protective in a mouse model of ischemic stroke through enhancement of the alternative microglial/macrophage activation and inhibition of MyD88 signaling. Our results accentuate the importance of controlling miRNA-regulated responses for the therapeutic benefit in conditions of stroke and neuroinflammation.


Assuntos
Ventrículos Cerebrais/metabolismo , AVC Isquêmico/metabolismo , Ativação de Macrófagos/fisiologia , Macrófagos/metabolismo , MicroRNAs/metabolismo , Microglia/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , AVC Isquêmico/genética , Camundongos , MicroRNAs/genética , Neurônios/metabolismo , Transdução de Sinais/fisiologia
2.
Hum Mol Genet ; 25(18): 3960-3974, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27466195

RESUMO

MyomiRs are muscle-specific microRNAs (miRNAs) that regulate myoblast proliferation and differentiation. Extracellular myomiRs (ex-myomiRs) are highly enriched in the serum of Duchenne Muscular Dystrophy (DMD) patients and dystrophic mouse models and consequently have potential as disease biomarkers. The biological significance of miRNAs present in the extracellular space is not currently well understood. Here we demonstrate that ex-myomiR levels are elevated in perinatal muscle development, during the regenerative phase that follows exercise-induced myoinjury, and concomitant with myoblast differentiation in culture. Whereas ex-myomiRs are progressively and specifically released by differentiating human primary myoblasts and C2C12 cultures, chemical induction of apoptosis in C2C12 cells results in indiscriminate miRNA release. The selective release of myomiRs as a consequence of cellular differentiation argues against the idea that they are solely waste products of muscle breakdown, and suggests they may serve a biological function in specific physiological contexts. Ex-myomiRs in culture supernatant and serum are predominantly non-vesicular, and their release is independent of ceramide-mediated vesicle secretion. Furthermore, ex-myomiRs levels are reduced in aged dystrophic mice, likely as a consequence of chronic muscle wasting. In conclusion, we show that myomiR release accompanies periods of myogenic differentiation in cell culture and in vivo. Serum myomiR abundance is therefore a function of the regenerative/degenerative status of the muscle, overall muscle mass, and tissue expression levels. These findings have implications for the use of ex-myomiRs as biomarkers for DMD disease progression and monitoring response to therapy.


Assuntos
MicroRNAs/genética , Desenvolvimento Muscular/genética , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Animais , Diferenciação Celular/genética , Proliferação de Células/genética , Espaço Extracelular/genética , Humanos , Camundongos , MicroRNAs/sangue , Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/sangue , Distrofia Muscular de Duchenne/patologia , Mioblastos/metabolismo , Mioblastos/patologia , Especificidade de Órgãos , Cultura Primária de Células
3.
Adv Exp Med Biol ; 983: 195-205, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28639201

RESUMO

The prevalence of cardiovascular diseases is steadily increasing, and it is the leading cause of death worldwide. Therefore, new treatments, such as gene therapy are needed. During the last decade, the role of small noncoding RNAs (ncRNAs) in the regulation of gene expression at the transcriptional level has been shown. Promoter-targeted small RNAs recruit histone-modifying enzymes and can either repress or induce target gene expression. As an example, we have targeted mouse VEGF-A promoter with small hairpin RNAs (shRNAs) and identified two shRNAs which either repressed or induced VEGF-A expression on messenger RNA and protein level in vitro, depending on the targeted location. The changes in expression levels correlate with changes in the levels of epigenetic markers, such as histone modifications associated with repressed or active state of chromatin. In ischemic mouse hindlimbs, upregulation of VEGF-A expression increased vascularity and blood flow. When VEGF-A was upregulated in mouse myocardial infarction model, the blood vessel formation in the risk zone was observed and infarct size was significantly decreased already 2 weeks after treatment. We suggest that epigenetic upregulation of VEGF-A by ncRNAs can be transferred to clinical use for the treatment of ischemic diseases in the near future.


Assuntos
Indutores da Angiogênese/farmacologia , Neovascularização Fisiológica , RNA Interferente Pequeno/genética , Fator A de Crescimento do Endotélio Vascular/fisiologia , Animais , Camundongos , Regiões Promotoras Genéticas , Regulação para Cima
4.
Curr Opin Lipidol ; 24(5): 438-43, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23872638

RESUMO

PURPOSE OF REVIEW: This review summarizes recent findings in the epigenetics of vascular cells and discusses the new challenges for therapeutic strategies of cardiovascular diseases. RECENT FINDINGS: There is emerging optimism that epigenetic mechanisms can provide the missing link to connect (epi)genomes with the cause of complex diseases. Environmental factors like intrauterine conditions during fetal development appear to preprogram humans for complex diseases. The purpose of this review is to summarize the newest results about the inheritable epigenetic features of cardiovascular diseases. Also, the recently discovered role of small RNAs in epigenetic gene regulation is discussed. SUMMARY: Epigenetic mechanisms of gene regulation will likely become major determinants in the pathogenesis of complex diseases and may offer new opportunities for the treatment of these diseases.


Assuntos
Doenças Cardiovasculares , Epigênese Genética , Animais , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/patologia , Humanos
5.
Genes (Basel) ; 14(2)2023 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-36833188

RESUMO

Extracellular vesicles (EVs) naturally carry cargo from producer cells, such as RNA and protein, and can transfer these messengers to other cells and tissue. This ability provides an interesting opportunity for using EVs as delivery vehicles for therapeutic agents, such as for gene therapy. However, endogenous loading of cargo, such as microRNAs (miRNAs), is not very efficient as the copy number of miRNAs per EV is quite low. Therefore, new methods and tools to enhance the loading of small RNAs is required. In the current study, we developed fusion protein of EV membrane protein CD9 and RNA-binding protein AGO2 (hCD9.hAGO2). We show that the EVs engineered with hCD9.hAGO2 contain significantly higher levels of miRNA or shRNA (miR-466c or shRNA-451, respectively) compared to EVs that are isolated from cells that only overexpress the desired miRNA or shRNA. These hCD9.hAGO2 engineered EVs also transfer their RNA cargo to recipient cells more efficiently. We were not able to detect changes in gene expression levels in recipient cells after the EV treatments, but we show that the cell viability of HUVECs was increased after hCD9.hAGO2 EV treatments. This technical study characterizes the hCD9.hAGO2 fusion protein for the future development of enhanced RNA loading to EVs.


Assuntos
Vesículas Extracelulares , MicroRNAs , MicroRNAs/genética , Vesículas Extracelulares/metabolismo , RNA Interferente Pequeno/metabolismo , Comunicação Celular , Proteínas/metabolismo
6.
PLoS One ; 17(3): e0265948, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35358280

RESUMO

MicroRNAs are well characterized in their role in silencing gene expression by targeting 3´-UTR of mRNAs in cytoplasm. However, recent studies have shown that miRNAs have a role in the regulation of genes in the nucleus, where they are abundantly located. We show here that in mouse endothelial cell line (C166), nuclear microRNA miR-466c participates in the regulation of vascular endothelial growth factor a (Vegfa) gene expression in hypoxia. Upregulation of Vegfa expression in response to hypoxia was significantly compromised after removal of miR-466c with CRISPR-Cas9 genomic deletion. We identified a promoter-associated long non-coding RNA on mouse Vegfa promoter and show that miR-466c directly binds to this transcript to modulate Vegfa expression. Collectively, these observations suggest that miR-466c regulates Vegfa gene transcription in the nucleus by targeting the promoter, and expands on our understanding of the role of miRNAs well beyond their canonical role.


Assuntos
MicroRNAs , RNA Longo não Codificante , Fator A de Crescimento do Endotélio Vascular , Animais , Hipóxia/genética , Camundongos , MicroRNAs/genética , RNA Longo não Codificante/genética , RNA Mensageiro , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
Biochim Biophys Acta ; 1803(11): 1276-86, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20691220

RESUMO

The biologically most active vitamin D compound, 1α,25-dihydroxyvitamin D3 (1α,25(OH)2D3), influences the status of inflammation by modulating the expression of several cytokine genes. In this study, we have examined the mechanism of transcriptional regulation of interleukin 10 (IL-10) by 1α,25(OH)2D3 in lipopolysaccharide (LPS)-treated human monocytes (THP-1). Quantitative PCR showed that IL-10 mRNA expression was significantly down-regulated (2.8-fold) during the first 8h of 1α,25(OH)2D3 treatment, while after 48 h it was up-regulated (3-fold). Gel shift and quantitative chromatin immunoprecipitation (ChIP) assays showed that the vitamin D receptor (VDR) binds in a cyclical fashion to a promoter region 1500-1700 bp upstream of the IL-10 transcription start site (TSS) containing two conserved VDR binding sites. Targeting of VDR binding sites by enhancer specific duplex RNAs revealed that only the more distal element is functional and chromosome conformation capture analysis suggested that this region loops 1α,25(OH)2D3-dependently to the TSS. Quantitative ChIP and micrococcal nuclease assays also revealed 1α,25(OH)2D3-dependent cyclical epigenetic changes and nucleosome remodeling at this promoter region. In conclusion, in LPS-treated THP-1 cells the primary effect of 1α,25(OH)2D3 on IL-10 expression is down-regulation, which is achieved via a cyclical recruitment of VDR to the promoter.


Assuntos
Regulação para Baixo/efeitos dos fármacos , Interleucina-10/genética , Monócitos/efeitos dos fármacos , Vitamina D/análogos & derivados , Sítios de Ligação/genética , Linhagem Celular Tumoral , Células Cultivadas , Montagem e Desmontagem da Cromatina/efeitos dos fármacos , Proteínas Correpressoras/genética , Proteínas Correpressoras/metabolismo , Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-10/metabolismo , Luciferases/genética , Luciferases/metabolismo , Monócitos/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica/efeitos dos fármacos , Interferência de RNA , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Elementos de Resposta/genética , Receptores X de Retinoides/genética , Receptores X de Retinoides/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Sítio de Iniciação de Transcrição , Vitamina D/farmacologia , Vitaminas/farmacologia
8.
Circ Res ; 105(6): 604-9, 2009 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-19696410

RESUMO

RATIONALE: We studied a possibility that shRNAs can lead to transcriptional gene activation at the promoter level via epigenetic mechanism. OBJECTIVE: The purpose of this study was to test the effects on vascular endothelial growth factor (VEGF-A) expression by promoter targeted small hairpin RNAs (shRNAs) in vitro and in experimental animals in vivo using stable local lentiviral gene transfer. METHODS AND RESULTS: One shRNA was identified which strongly increased VEGF-A expression in C166 endothelial cells at mRNA and protein level whereas another shRNA decreased VEGF-A expression. Quantitative chromatin immunoprecipitation analysis revealed that the repressing shRNA caused epigenetic changes, which increased nucleosome density within the promoter and transcription start site and led to repression of VEGF-A expression. Epigenetic changes caused by the activating shRNA were opposite to those caused by the repressing shRNA. These results were confirmed in vivo in an ischemic mouse hindlimb model after local gene transfer where VEGF-A upregulation achieved by promoter-targeted shRNA increased vascularity and blood flow. CONCLUSIONS: We show that lentivirus-mediated delivery of shRNA molecules targeted to specific regions in the mVEGF-A promoter either induce or repress VEGF-A expression via epigenetic modulation. Thus, we describe a new approach of gene therapy, epigenetherapy, based on an epigenetic mechanism at the promoter level. Controlling transcription through manipulation of specific epigenetic marks provides a novel approach for the treatment of several diseases.


Assuntos
Epigênese Genética , Terapia Genética/métodos , Membro Posterior/irrigação sanguínea , Isquemia/terapia , Lentivirus , Regiões Promotoras Genéticas , RNA/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Linhagem Celular , Células Endoteliais/metabolismo , Isquemia/genética , Camundongos , RNA/genética , Transcrição Gênica , Fator A de Crescimento do Endotélio Vascular/metabolismo
9.
Aging Cell ; 20(1): e13287, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33369048

RESUMO

Ischemic stroke, the third leading cause of death in the Western world, affects mainly the elderly and is strongly associated with comorbid conditions such as atherosclerosis or diabetes, which are pathologically characterized by increased inflammation and are known to influence the outcome of stroke. Stroke incidence peaks during influenza seasons, and patients suffering from infections such as pneumonia prior to stroke exhibit a worse stroke outcome. Earlier studies have shown that comorbidities aggravate the outcome of stroke, yet the mediators of this phenomenon remain obscure. Here, we show that acute peripheral inflammation aggravates stroke-induced neuronal damage and motor deficits specifically in aged mice. This is associated with increased levels of plasma proinflammatory cytokines, rather than with an increase of inflammatory mediators in the affected brain parenchyma. Nascent transcriptomics data with mature microRNA sequencing were used to identify the neuron-specific miRNome, in order to decipher dysregulated miRNAs in the brains of aged animals with stroke and co-existing inflammation. We pinpoint a previously uninvestigated miRNA in the brain, miR-127, that is highly neuronal, to be associated with increased cell death in the aged, LPS-injected ischemic mice. Target prediction tools indicate that miR-127 interacts with several basally expressed neuronal genes, and of these we verify miR-127 binding to Psmd3. Finally, we report reduced expression of miR-127 in human stroke brains. Our results underline the impact of peripheral inflammation on the outcome of stroke in aged subjects and pinpoint molecular targets for restoring endogenous neuronal capacity to combat ischemic stroke.


Assuntos
Isquemia Encefálica/genética , Inflamação/genética , MicroRNAs/metabolismo , Envelhecimento , Animais , Isquemia Encefálica/mortalidade , Modelos Animais de Doenças , Humanos , Masculino , Camundongos
10.
Biochim Biophys Acta ; 1790(9): 886-91, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19233248

RESUMO

The contribution of epigenetic mechanisms to cardiovascular diseases remains poorly understood. Hypomethylation of genomic DNA is present in human atherosclerotic lesions and methylation changes also occur at the promoter level of several genes involved in the pathogenesis of atherosclerosis, such as extracellular superoxide dismutase, estrogen receptor-alpha, endothelial nitric oxide synthase and 15-lipoxygenase. So far, no clear data is available about histone modification marks in atherosclerotic lesions. It remains unclear whether epigenetic changes are causally related to the pathogenetic features, such as clonal proliferation of lesion smooth muscle cells, lipid accumulation and modulation of immune responses in the lesions, or whether they merely represent a consequence of the ongoing pathological process. However, epigenetic changes could at least partly explain poorly understood environmental and dietary effects on atherogenesis and the rapid increases and decreases in the incidence of coronary heart disease observed in various populations. RNAi mechanisms may also contribute to the epigenetic regulation of vascular cells. Therapies directed towards modification of the epigenetic status of vascular cells might provide new tools to control atherosclerosis-related cardiovascular diseases.


Assuntos
Aterosclerose/genética , Epigênese Genética , Animais , Metilação de DNA , Histonas/metabolismo , Humanos , Hiper-Homocisteinemia/genética
11.
Cardiovasc Res ; 78(1): 175-84, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18192240

RESUMO

AIMS: Statins have beneficial vascular effects beyond their cholesterol-lowering action. Since macrophages play a central role in atherogenesis, we characterized the effects of simvastatin on gene expression profile of human peripheral blood monocyte (HPBM)-macrophages. METHODS AND RESULTS: Gene expression profile was studied using Affymetrix gene chip analysis. Lentiviral gene transfer of Kruppel-like factor 2 (KLF-2) was used to further study its role in macrophages. Simvastatin treatment lead to downregulation of many pro-inflammatory genes including several chemokines [e.g. monocyte chemotactic protein-1 (MCP-1), macrophage inflammatory proteins-1alpha and beta, interleukin-2 receptor-beta], members of the tumour necrosis factor family (e.g. lymphotoxin beta), vascular cell adhesion molecule-1, and tissue factor (TF). Simvastatin also modulated the expression of several transcription factors essential for inflammation: NF-kappaB relA/p65 subunit and ets-1 were downregulated, and an atheroprotective transcription factor KLF-2 was upregulated. The effects of simvastatin on MCP-1 and TF could be mimicked by KLF-2 overexpression using lentiviral gene transfer. CONCLUSION: Simvastatin has a strong anti-inflammatory effect on HPBM cells including upregulation of the atheroprotective factor KLF-2. This may partly explain the beneficial effects of statins on cardiovascular diseases.


Assuntos
Anti-Inflamatórios/farmacologia , Citocinas/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Fatores de Transcrição Kruppel-Like/metabolismo , Macrófagos/efeitos dos fármacos , Sinvastatina/farmacologia , Linhagem Celular , Células Cultivadas , Quimiocina CCL2/metabolismo , Citocinas/genética , Perfilação da Expressão Gênica/métodos , Técnicas de Transferência de Genes , Humanos , Fatores de Transcrição Kruppel-Like/genética , Macrófagos/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Prenilação de Proteína , Proteínas Proto-Oncogênicas c-ets/metabolismo , RNA Mensageiro/metabolismo , Tromboplastina/metabolismo , Fatores de Tempo , Fator de Transcrição RelA/metabolismo , Fatores de Necrose Tumoral/metabolismo , Regulação para Cima
12.
Sci Rep ; 9(1): 10332, 2019 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-31316122

RESUMO

MicroRNAs (miRNAs) are small non-coding RNAs that have well-characterized roles in cytoplasmic gene regulation, where they act by binding to mRNA transcripts and inhibiting their translation (i.e. post-transcriptional gene silencing, PTGS). However, miRNAs have also been implicated in transcriptional gene regulation and alternative splicing, events that are restricted to the cell nucleus. Here we performed nuclear-cytoplasmic fractionation in a mouse endothelial cell line and characterized the localization of miRNAs in response to hypoxia using small RNA sequencing. A highly diverse population of abundant miRNA species was detected in the nucleus, of which the majority (56%) was found to be preferentially localized in one compartment or the other. Induction of hypoxia resulted in changes in miRNA levels in both nuclear and cytoplasmic compartments, with the majority of changes being restricted to one location and not the other. Notably, the classical hypoxamiR (miR-210-3p) was highly up-regulated in the nuclear compartment after hypoxic stimulus. These findings reveal a previously unappreciated level of molecular complexity in the physiological response occurring in ischemic tissue. Furthermore, widespread differential miRNA expression in the nucleus strongly suggests that these small RNAs are likely to perform extensive nuclear regulatory functions in the general case.


Assuntos
Hipóxia Celular/genética , Hipóxia Celular/fisiologia , MicroRNAs/genética , MicroRNAs/metabolismo , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Células Endoteliais/metabolismo , Regulação da Expressão Gênica , Camundongos , Estresse Fisiológico/genética
13.
PLoS One ; 13(1): e0190981, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29351307

RESUMO

Vascular endothelial growth factor-A (VEGF-A) is the master regulator of angiogenesis, vascular permeability and growth. However, its role in mature blood vessels is still not well understood. To better understand the role of VEGF-A in the adult vasculature, we generated a VEGF-A knockdown mouse model carrying a doxycycline (dox)-regulatable short hairpin RNA (shRNA) transgene, which silences VEGF-A. The aim was to find the critical level of VEGF-A reduction for vascular well-being in vivo. In vitro, the dox-inducible lentiviral shRNA vector decreased VEGF-A expression efficiently and dose-dependently in mouse endothelial cells and cardiomyocytes. In the generated transgenic mice plasma VEGF-A levels decreased shortly after the dox treatment but returned back to normal after two weeks. VEGF-A expression decreased shortly after the dox treatment only in some tissues. Surprisingly, increasing the dox exposure time and dose led to elevated VEGF-A expression in some tissues of both wildtype and knockdown mice, suggesting that dox itself has an effect on VEGF-A expression. When the effect of dox on VEGF-A levels was further tested in naïve/non-transduced cells, the dox administration led to a decreased VEGF-A expression in endothelial cells but to an increased expression in cardiomyocytes. In conclusion, the VEGF-A knockdown was achieved in a dox-regulatable fashion with a VEGF-A shRNA vector in vitro, but not in the knockdown mouse model in vivo. Dox itself was found to regulate VEGF-A expression explaining the unexpected results in mice. The effect of dox on VEGF-A levels might at least partly explain its previously reported beneficial effects on myocardial and brain ischemia. Also, this effect on VEGF-A should be taken into account in all studies using dox-regulated vectors.


Assuntos
Doxiciclina/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Lentivirus/genética , RNA Interferente Pequeno/genética , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Vetores Genéticos , Camundongos , Camundongos Transgênicos
14.
Circulation ; 109(9): 1140-6, 2004 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-14769706

RESUMO

BACKGROUND: Platelet-derived growth factor (PDGF) antagonists have demonstrated beneficial effects on neointima formation, but in studies using PDGF inhibitors and extended follow-up, the lesions reoccur. These findings implicate a need to combine targeting of PDGF with other strategies. Stimulation of reendothelialization by treatment with endothelial cell mitogens of the vascular endothelial growth factor (VEGF) family counteracts restenosis, but there are also concerns regarding the durability of the effect with this approach. METHODS AND RESULTS: To explore whether a combined use of PDGF antagonist and stimulation of reendothelialization confers better results than each therapy alone, we combined systemic administration of imatinib mesylate (STI571/Gleevec, 10 mg/kg(-1) per d(-1)), a tyrosine kinase inhibitor with activity against PDGF receptors, with local intravascular adenovirus-mediated VEGF-C gene transfer (1.15x10(10) pfu) in cholesterol-fed, balloon-injured rabbits. Throughout the course of the STI571 therapy, the circulating concentrations were able to suppress PDGF receptor phosphorylation. At 3 weeks, the treatment with STI571 led to a transient decrease in intralesion macrophages and to an increase in intimal smooth muscle cell apoptosis. VEGF-C application reduced neointima formation and accelerated reendothelialization. However, none of the therapies alone reduced intimal thickening at a 6-week time point, whereas the combined treatment led to a persistent reduction (55% versus control) in lesion size at this time point. CONCLUSIONS: Our study provides one of the first successful examples of gene therapy combined with a pharmacological treatment to modulate 2 distinct ligand-receptor signaling systems and suggests combination of local VEGF-C gene therapy with systemic inhibition of PDGF signaling as a novel principle to prevent intimal hyperplasia after vascular manipulations.


Assuntos
Inibidores Enzimáticos/uso terapêutico , Hipercolesterolemia/terapia , Piperazinas/uso terapêutico , Pirimidinas/uso terapêutico , Receptores do Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Túnica Íntima/patologia , Fator C de Crescimento do Endotélio Vascular/genética , Adenoviridae/genética , Administração Oral , Animais , Artérias , Benzamidas , Divisão Celular/efeitos dos fármacos , Movimento Celular , Terapia Combinada , Endotélio Vascular/patologia , Inibidores Enzimáticos/administração & dosagem , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Hipercolesterolemia/tratamento farmacológico , Hipercolesterolemia/patologia , Mesilato de Imatinib , Macrófagos/imunologia , Músculo Liso Vascular/patologia , Piperazinas/administração & dosagem , Pirimidinas/administração & dosagem , Coelhos , Túnica Íntima/citologia , Túnica Íntima/efeitos dos fármacos , Túnica Média/patologia
15.
Methods Enzymol ; 346: 311-20, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-11883076

RESUMO

Local gene transfer into the vascular wall offers a promising alternative to treat atherosclerosis-related diseases. Blood vessels are among the easiest targets for gene therapy because of percutaneous, catheter-based treatment methods. On the other hand, gene transfer to the artery wall can also be accomplished from adventitia either by ex vivo gene transfer and implantation of transfected cells or by direct in vivo gene transfer methods. In the future, as the pathological processes in arteries are better understood, several therapeutic genes could be combined and these "gene cocktails" are expected to produce enhanced therapeutic effects in vascular gene therapy. We have developed a new, efficient technique for performing ex vivo gene transfer to rabbit arterial wall using autologous SMC. The cells were harvested from rabbit ear artery, transfected in vitro with VSV-G pseudotyped lacZ retrovirus, and returned back to the adventitial surface of the carotid artery using a silicone collar or collagen sheet placed around the artery. The transduced SMCs implanted with a high efficiency and expressed beta-galactosidase marker gene at a very high level 7 days and 14 days after the operation. The level of lacZ expression decreased thereafter, but was still easily detectable for at least 6 months and was exclusively localized to the site of cell implantation inside the collar. Development of new vectors, such as baculovirus, for gene transfer will provide targeted, efficient, and safer methods for gene delivery. Plasmids and viruses coding for more than one protein, and bearing regulatory elements, would be useful for future gene therapy applications. Also, constructing second-generation viruses that contain fewer endogenous genes in their genome may reduce immunological reactions caused by the first-generation adenoviruses. In conditions where stable expression of therapeutic proteins is needed, it is necessary to develop better ex vivo and in vivo gene transfer strategies. Also, production of viruses that can efficiently transfect nondividing cells will be important for future applications of vascular gene therapy. However, current knowledge from vascular gene transfer experiments strongly suggests that vascular gene transfer is a promising new alternative for the treatment of cardiovascular diseases.


Assuntos
Doenças Cardiovasculares/terapia , Terapia Genética , Animais , Vasos Sanguíneos/metabolismo , Técnicas de Transferência de Genes , Coelhos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Mol Ther Nucleic Acids ; 3: e197, 2014 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-25405466

RESUMO

This review discusses recent developments in the use of non-coding RNAs (ncRNAs) for the regulation of therapeutically relevant genes, with special focus on applications for the treatment of cardiovascular diseases. The interest in using ncRNAs as therapeutics has steadily increased since the discovery of RNA interference. During the last decade it has become evident that these RNAs, delivered either as oligos or expressed as small hairpin RNAs (shRNAs) from vectors, can either upregulate (transcriptional gene activation, TGA) or downregulate (transcriptional gene silencing, TGS) gene expression, typically inducing epigenetic changes in their target sites in the chromatin. Also, the important role of naturally occurring long non-coding RNAs (lncRNAs) has been recently discovered and will likely provide new insights into cardiovascular pathology and provide new treatment strategies based on the manipulation of their expression. In this review, we discuss the possibility of using ncRNAs for activating or silencing therapeutically relevant genes, such as VEGF-A, for the treatment of cardiovascular disease.

17.
PLoS One ; 9(2): e89979, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24587164

RESUMO

"Epigenetherapy" alters epigenetic status of the targeted chromatin and modifies expression of the endogenous therapeutic gene. In this study we used lentiviral in vivo delivery of small hairpin RNA (shRNA) into hearts in a murine infarction model. shRNA complementary to the promoter of vascular endothelial growth factor (VEGF-A) was able to upregulate endogenous VEGF-A expression. Histological and multiphoton microscope analysis confirmed the therapeutic effect in the transduced hearts. Magnetic resonance imaging (MRI) showed in vivo that the infarct size was significantly reduced in the treatment group 14 days after the epigenetherapy. Importantly, we show that promoter-targeted shRNA upregulates all isoforms of endogenous VEGF-A and that an intact hairpin structure is required for the shRNA activity. In conclusion, regulation of gene expression at the promoter level is a promising new treatment strategy for myocardial infarction and also potentially useful for the upregulation of other endogenous genes.


Assuntos
Epigênese Genética , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Regulação para Cima/genética , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Sequência de Bases , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Metilação de DNA , Inativação Gênica , Sequências Repetidas Invertidas/genética , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/metabolismo , Fosfoproteínas/metabolismo , Regiões Promotoras Genéticas/genética , Isoformas de Proteínas/genética , RNA Interferente Pequeno/genética , Transcrição Gênica/genética , Ativação Transcricional
18.
Cardiovasc Res ; 90(3): 441-6, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21498421

RESUMO

The role of small RNAs in epigenetic regulation is an emerging field. This research may also open novel treatment strategies based on manipulation of the epigenetic status of the target tissues. Our objective is to review epigenetic regulation of key vascular genes and growth factors. Vascular endothelial growth factor A (VEGF-A) is one of the key players in regulating and maintaining cardiovascular functions and pathology. Although its epigenetic regulation is still not completely understood, expression of the VEGF gene can be manipulated by epigenetic mechanisms using small RNAs that are targeted to the gene promoter which results in the alteration of histone code. VEGF exerts its effects mostly through two receptors, VEGFR1 and VEGFR2, and their expression is also regulated by promoter DNA methylation in various cancer cells. These findings suggest the importance of epigenetic mechanisms in the regulation of vascular functions.


Assuntos
Proteínas Angiogênicas/genética , Vasos Sanguíneos/metabolismo , Epigênese Genética , Animais , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/terapia , Humanos , Metaloproteinases da Matriz/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética
19.
Biomol Concepts ; 2(3): 127-34, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25962024

RESUMO

Small RNAs have been shown to regulate gene transcription by interacting with the promoter region and modifying the histone code. The exact mechanism of function is still unclear but the feasibility to activate or repress endogenous gene expression with small RNA molecules has already been demonstrated in vitro and in vivo. In traditional gene therapy non-mutated or otherwise useful genes are inserted into patient's cells to treat a disease. In epigenetherapy the action of small RNAs is utilized by delivering only the small RNAs to patient's cells where they then regulate gene expression by epigenetic mechanisms. This method could be widely useful not only for basic research but also for clinical applications of small RNAs.

20.
Cardiovasc Res ; 88(3): 530-8, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-20634212

RESUMO

AIMS: Macrophage scavenger receptor A (SR-A) and class B scavenger receptor CD36 (CD36) contribute to foam cell formation and atherogenesis via uptake of modified lipoproteins. So far, the role of these scavenger receptors has been studied mainly using knockout models totally lacking these receptors. We studied the role of SR-A and CD36 in foam cell formation and atherogenesis by RNA interference (RNAi)-mediated silencing, which is a clinically feasible method to down-regulate the expression of these receptors. METHODS AND RESULTS: We constructed lentivirus vectors encoding short hairpin RNAs (shRNAs) against mouse SR-A and CD36. Decreased SR-A but not CD36 expression led to reduced foam cell formation caused by acetylated low-density lipoprotein (LDL) in mouse macrophages, whereas the uptake of oxidized LDL was not altered. More importantly, silencing of SR-A upregulates CD36 and vice versa. In LDL receptor-deficient apolipoprotein B100 (LDLR(-/-)ApoB(100/100)) mice kept on a western diet, silencing of either SR-A or CD36 in bone marrow cells led to a marked decrease (37.4 and 34.2%, respectively) in cross-sectional lesion area, whereas simultaneous silencing of both receptors was not effective. CONCLUSION: Our results suggest that silencing of either SR-A or CD36 alone reduces atherogenesis in mice. However, due to reciprocal upregulation, silencing of both SR-A and CD36 is not effective.


Assuntos
Aterosclerose/prevenção & controle , Antígenos CD36/genética , Hiperlipidemias/complicações , Interferência de RNA , Receptores Depuradores Classe A/genética , Regulação para Cima/genética , Animais , Apolipoproteínas B/genética , Apolipoproteínas B/metabolismo , Aterosclerose/genética , Aterosclerose/metabolismo , Antígenos CD36/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Feminino , Células Espumosas/metabolismo , Células Espumosas/patologia , Humanos , Hiperlipidemias/metabolismo , Hiperlipidemias/patologia , Lentivirus/genética , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Knockout , Receptores de LDL/genética , Receptores de LDL/metabolismo , Receptores Depuradores Classe A/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa