Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cell Death Discov ; 9(1): 221, 2023 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-37400464

RESUMO

Understanding of human brain development, dysfunction and neurological diseases has remained limited and challenging due to inability to recapitulate human brain-specific features in animal models. Though the anatomy and physiology of the human brain has been understood in a remarkable way using post-mortem, pathological samples of human and animal models, however, modeling of human brain development and neurological diseases remains a challenge owing to distinct complexity of human brain. In this perspective, three-dimensional (3D) brain organoids have shown a beam of light. Tremendous growth in stem cell technologies has permitted the differentiation of pluripotent stem cells under 3D culture conditions into brain organoids, which recapitulate the unique features of human brain in many ways and also offer the detailed investigation of brain development, dysfunction and neurological diseases. Their translational value has also emerged and will benefit the society once the protocols for the upscaling of brain organoids are in place. Here, we summarize new advancements in methods for generation of more complex brain organoids including vascularized and mixed lineage tissue from PSCs. How synthetic biomaterials and microfluidic technology is boosting brain organoid development, has also been highlighted. We discuss the applications of brain organoids in studying preterm birth associated brain dysfunction; viral infections mediated neuroinflammation, neurodevelopmental and neurodegenerative diseases. We also highlight the translational value of brain organoids and current challenges that the field is experiencing.

2.
Hum Mol Genet ; 31(13): 2137-2154, 2022 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-35075486

RESUMO

Retinal diseases exhibit extensive genetic heterogeneity and complex etiology with varying onset and severity. Mutations in over 200 genes can lead to photoreceptor dysfunction and/or cell death in retinal neurodegeneration. To deduce molecular pathways that initiate and/or drive cell death, we adopted a temporal multiomics approach and examined molecular and cellular events in newborn and developing photoreceptors before the onset of degeneration in a widely-used Pde6brd1/rd1 (rd1) mouse, a model of autosomal recessive retinitis pigmentosa caused by PDE6B mutations. Transcriptome profiling of neonatal and developing rods from the rd1 retina revealed early downregulation of genes associated with anabolic pathways and energy metabolism. Quantitative proteomics of rd1 retina showed early changes in calcium signaling and oxidative phosphorylation, with specific partial bypass of complex I electron transfer, which precede the onset of cell death. Concurrently, we detected alterations in central carbon metabolism, including dysregulation of components associated with glycolysis, pentose phosphate and purine biosynthesis. Ex vivo assays of oxygen consumption and transmission electron microscopy validated early and progressive mitochondrial stress and abnormalities in mitochondrial structure and function of rd1 rods. These data uncover mitochondrial overactivation and related metabolic alterations as determinants of early pathology and implicate aberrant calcium signaling as an initiator of higher mitochondrial stress. Our studies thus provide a mechanistic framework with mitochondrial damage and metabolic disruptions as early drivers of photoreceptor cell death in retinal degeneration.


Assuntos
Degeneração Retiniana , Retinose Pigmentar , Animais , Morte Celular/genética , Modelos Animais de Doenças , Camundongos , Células Fotorreceptoras de Vertebrados/metabolismo , Retina/metabolismo , Degeneração Retiniana/patologia , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Retinose Pigmentar/patologia
3.
Stem Cells ; 38(5): 683-697, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32012382

RESUMO

The role of miRNAs in determining human neural stem cell (NSC) fate remains elusive despite their high expression in the developing nervous system. In this study, we investigate the role of miR-137, a brain-enriched miRNA, in determining the fate of human induced pluripotent stem cells-derived NSCs (hiNSCs). We show that ectopic expression of miR-137 in hiNSCs reduces proliferation and accelerates neuronal differentiation and migration. TargetScan and MicroT-CDS predict myocyte enhancer factor-2A (MEF2A), a transcription factor that regulates peroxisome proliferator-activated receptor-gamma coactivator (PGC1α) transcription, as a target of miR-137. Using a reporter assay, we validate MEF2A as a downstream target of miR-137. Our results indicate that reduced levels of MEF2A reduce the transcription of PGC1α, which in turn impacts mitochondrial dynamics. Notably, miR-137 accelerates mitochondrial biogenesis in a PGC1α independent manner by upregulating nuclear factor erythroid 2 (NFE2)-related factor 2 (NRF2) and transcription factor A of mitochondria (TFAM). In addition, miR-137 modulates mitochondrial dynamics by inducing mitochondrial fusion and fission events, resulting in increased mitochondrial content and activation of oxidative phosphorylation (OXPHOS) and oxygen consumption rate. Pluripotency transcription factors OCT4 and SOX2 are known to have binding sites in the promoter region of miR-137 gene. Ectopic expression of miR-137 elevates the expression levels of OCT4 and SOX2 in hiNSCs which establishes a feed-forward self-regulatory loop between miR-137 and OCT4/SOX2. Our study provides novel molecular insights into NSC fate determination by miR-137.


Assuntos
MicroRNAs/metabolismo , Dinâmica Mitocondrial/fisiologia , Células-Tronco Neurais/metabolismo , Diferenciação Celular/fisiologia , Regulação para Baixo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/metabolismo , MicroRNAs/genética , Células-Tronco Neurais/citologia , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Biogênese de Organelas , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo
4.
Mol Vis ; 25: 663-678, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31814692

RESUMO

PURPOSE: Retinal organoids generated from human pluripotent stem cells exhibit considerable variability during differentiation. Our goals are to assess developmental maturity of the neural retina in vitro and design improved protocols based on objective criteria. METHODS: We performed transcriptome analyses of developing retinal organoids from human embryonic and induced pluripotent stem cell lines and utilized multiple bioinformatic tools for comparative analysis. Immunohistochemistry, immunoblotting and electron microscopy were employed for validation. RESULTS: We show that the developmental variability in organoids was reflected in gene expression profiles and could be evaluated by molecular staging with the human fetal and adult retinal transcriptome data. We also demonstrate that the addition of 9-cis retinal, instead of the widely used all-trans retinoic acid, accelerated rod photoreceptor differentiation in organoid cultures, with higher rhodopsin expression and more mature mitochondrial morphology evident by day 120. CONCLUSION: Our studies provide an objective transcriptome-based modality for determining the differentiation state of retinal organoids and for comparisons across different stem cell lines and platforms, which should facilitate disease modeling and evaluation of therapies in vitro.


Assuntos
Diferenciação Celular , Diterpenos/farmacologia , Células-Tronco Embrionárias Humanas/citologia , Organoides/citologia , Retina/citologia , Células Fotorreceptoras Retinianas Bastonetes/citologia , Retinaldeído/farmacologia , Transcriptoma/genética , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Forma Celular/efeitos dos fármacos , Perfilação da Expressão Gênica , Células-Tronco Embrionárias Humanas/efeitos dos fármacos , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Organoides/efeitos dos fármacos , Organoides/ultraestrutura , Células Fotorreceptoras Retinianas Bastonetes/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos
5.
J Neuroinflammation ; 15(1): 317, 2018 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-30442172

RESUMO

Upon publication of the original article [1], it was noticed that there is an error in Fig. 10, the dialog box in panel (b) was missing. The correct Fig. 10 is shown below.

6.
Cell Death Differ ; 25(10): 1837-1854, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30050059

RESUMO

Zika virus (ZV) infects neural stem cells (NSCs) and causes quiescence in NSCs, reducing the pool of brain cells, leading to microcephaly. Despite conscientious efforts, the molecular mechanisms for ZV-mediated effects on NSCs lack clarity. This study aimed to explore the underlying mechanisms for ZV-mediated induction of quiescence in the primary cultures of human fetal neural stem cells (fNSCs). We demonstrate that expression of ZV envelope (E) protein displays maximum quiescence in human fNSCs by accumulating cells in the G0/G1 phase of the cell cycle as compared to other non-structural proteins, viz. NS2A, NS4A and NS4B. E protein induces immature differentiation by induction of pro-neuronal genes in proliferating fNSCs, induces apoptosis in differentiating fNSCs 3 days post differentiation, and disrupts migration of cells from differentiating neurospheres. In utero electroporation of mouse brain with E protein shows drastic downregulation of proliferating cells in ventricular and subventricular zone regions. Global microRNA sequencing suggests that E protein modulates miRNA circuitry. Among differentially expressed miRNAs, we found 14 upregulated and 11 downregulated miRNAs. Mir-204-3p and mir-1273g-3p directly regulate NOTCH2 and PAX3 expression, respectively, by binding to their 3'UTR. Bioinformatic analysis using GO analysis for the targets of differentially expressed miRNAs revealed enrichment of cell cycle and developmental processes. Furthermore, WNT, CCKR, PDGF, EGF, p53, and NOTCH signaling pathways were among the top enriched pathways. Thus, our study provides evidence for the involvement of ZV E protein and novel insights into the molecular mechanism through identification of miRNA circuitry. Art work depicting the effect of Zika virus E protein on human fetal neural stem cells.


Assuntos
Redes Reguladoras de Genes , MicroRNAs/metabolismo , Proteínas do Envelope Viral/metabolismo , Zika virus/metabolismo , Regiões 3' não Traduzidas , Antagomirs/metabolismo , Apoptose , Diferenciação Celular , Sobrevivência Celular , Regulação para Baixo , Feto/citologia , Pontos de Checagem da Fase G1 do Ciclo Celular , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Humanos , MicroRNAs/antagonistas & inibidores , MicroRNAs/química , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/virologia , Receptor Notch2/química , Receptor Notch2/genética , Receptor Notch2/metabolismo , Transdução de Sinais , Regulação para Cima , Proteínas do Envelope Viral/genética , Proteínas não Estruturais Virais/metabolismo
7.
J Neuroinflammation ; 15(1): 177, 2018 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-29885667

RESUMO

BACKGROUND: Interleukin-1ß (IL-1ß) is one of the most important cytokine secreted by activated microglia as it orchestrates the vicious cycle of inflammation by inducing the expression of various other pro-inflammatory cytokines along with its own production. Microglia-mediated IL-1ß production is a tightly regulated mechanism which involves the activation of nucleotide-binding oligomerization domain leucine-rich repeat and pyrin domain-containing 3 (NLRP3) inflammasome pathway. Our previous study suggests the critical role of heat shock protein 60 (HSP60) in IL-1ß-induced inflammation in microglia through TLR4-p38 MAPK axis. However, whether HSP60 regulates endogenous IL-1ß production is not known. Therefore, to probe the underlying mechanism, we elucidate the role of HSP60 in endogenous IL-1ß production. METHODS: We used in vitro (N9 murine microglial cells) and in vivo (BALB/c mouse) models for our study. HSP60 overexpression and knockdown experiment was done to elucidate the role of HSP60 in endogenous IL-1ß production by microglia. Western blotting and quantitative real-time PCR was performed using N9 cells and BALB/c mice brain, to analyze various proteins and transcript levels. Reactive oxygen species levels and mitochondrial membrane depolarization in N9 cells were analyzed by flow cytometry. We also performed caspase-1 activity assay and enzyme-linked immunosorbent assay to assess caspase-1 activity and IL-1ß production, respectively. RESULTS: HSP60 induces the phosphorylation and nuclear localization of NF-κB both in vitro and in vivo. It also induces perturbation in mitochondrial membrane potential and enhances reactive oxygen species (ROS) generation in microglia. HSP60 further activates NLRP3 inflammasome by elevating NLRP3 expression both at RNA and protein levels. Furthermore, HSP60 enhances caspase-1 activity and increases IL-1ß secretion by microglia. Knockdown of HSP60 reduces the IL-1ß-induced production of IL-1ß both in vitro and in vivo. Also, we have shown for the first time that knockdown of HSP60 leads to decreased IL-1ß production during Japanese encephalitis virus (JEV) infection, which eventually leads to decreased inflammation and increased survival of JEV-infected mice. CONCLUSION: HSP60 mediates microglial IL-1ß production by regulating NLRP3 inflammasome pathway and reduction of HSP60 leads to reduction of inflammation in JEV infection.


Assuntos
Chaperonina 60/farmacologia , Regulação da Expressão Gênica/fisiologia , Interleucina-1beta/metabolismo , Microglia/efeitos dos fármacos , Proteínas Mitocondriais/farmacologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Encéfalo/citologia , Encéfalo/metabolismo , Chaperonina 60/genética , Chaperonina 60/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Vírus da Encefalite Japonesa (Subgrupo)/fisiologia , Encefalite Japonesa/metabolismo , Encefalite Japonesa/patologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-1beta/genética , Interleucina-1beta/farmacologia , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Morfolinos/farmacologia , Óxido Nítrico Sintase Tipo II/metabolismo , RNA Interferente Pequeno/farmacologia
8.
Methods Mol Biol ; 1753: 167-177, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29564788

RESUMO

Mitochondrial dysfunction, and consequently altered aerobic energy metabolism, is associated with numerous retinal diseases, including photoreceptor degeneration and diabetic retinopathy. Here, we describe a detailed protocol to directly measure oxygen consumption in the intact retina ex vivo using microplate-based fluorescence technology. We have used this method to assess preferred energy substrate for retinal tissue and suggested its application for investigating mechanisms of retinal disease.


Assuntos
Bioensaio/métodos , Mitocôndrias/metabolismo , Oxigênio/análise , Retina/metabolismo , Doenças Retinianas/patologia , Animais , Fatores de Transcrição de Zíper de Leucina Básica/genética , Bioensaio/instrumentação , Respiração Celular , Modelos Animais de Doenças , Proteínas do Olho/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oxigênio/metabolismo , Consumo de Oxigênio , Retina/citologia , Doenças Retinianas/genética , Espectrometria de Fluorescência/instrumentação , Espectrometria de Fluorescência/métodos
9.
Prog Neurobiol ; 148: 21-39, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27979736

RESUMO

Research over the last few years in cellular reprogramming has enlightened the magical potential of microRNAs (miRNAs) in changing the cell fate from somatic to pluripotent. Recent investigations on exploring the role(s) of miRNAs in somatic cell reprogramming revealed that they target a wide range of molecules and refine their protein output. This leads to fine tuning of distinct cellular processes including cell cycle, signalling pathways, transcriptional activation/silencing and epigenetic modelling. The concerted actions of miRNA on different pathways simultaneously strengthen the transition from a differentiated to de-differentiated state. Despite the well characterized transcriptional and epigenetic machinery underlying somatic cell reprogramming, the molecular circuitry for miRNA mediated cellular reprogramming is rather fragmented. This review summarizes recent findings addressing the role of miRNAs in inducing or suppressing reprogramming thus uncovering novel potentials of miRNAs as regulators of induced pluripotency maintenance, establishment and associated signalling pathways. Our bioinformatic analysis sheds light on various unexplored biological processes and pathways associated with reprogramming inducing miRNAs, thus helps in identifying roadblocks to full reprogramming. Specifically, the biological significance of highly conserved and most studied miRNA cluster, i.e. miR-302-367, in reprogramming is also highlighted. Further, roles of miRNAs in the differentiation of neurons from iPSCs are discussed. A recent approach of direct conversion or transdifferentiation of differentiated cells into neurons by miRNAs is also elaborated. This approach is now widely gaining impetus for the generation of neurological patient's brain cells directly from his/her somatic cells in an efficient and safe manner. Thus, decoding the intricate circuitry between miRNAs and other gene regulatory networks will not only uncover novel pathways in the direct reprogramming of somatic cells but will also open new avenues in stem cell biology.


Assuntos
Reprogramação Celular/fisiologia , MicroRNAs/fisiologia , Animais , Humanos
10.
Tumour Biol ; 37(7): 8529-54, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27105614

RESUMO

Resistance to apoptosis leads to tumorigenesis and failure of anti-cancer therapy. Recent studies also highlight abrogated lipid/cholesterol metabolism as one of the root causes of cancer that can lead to metastatic transformations. Cancer cells are dependent on tremendous supply of cellular cholesterol for the formation of new membranes and continuation of cell signaling. Cholesterol homeostasis network tightly regulates this metabolic need of cancer cells on cholesterol and other lipids. Genetic landscape is also shared between apoptosis and cholesterol metabolism. MicroRNAs (miRNAs) are the new fine tuners of signaling pathways and cellular processes and are known for their ability to post-transcriptionally repress gene expression in a targeted manner. This review summarizes the current knowledge about the cross talk between apoptosis and cholesterol metabolism via miRNAs. In addition, we also emphasize herein recent therapeutic modulations of specific miRNAs and their promising potential for the treatment of deadly diseases including cancer and cholesterol related pathologies. Understanding of the impact of miRNA-based regulation of apoptosis and metabolic processes is still at its dawn and needs further research for the development of future miRNA-based therapies. As both these physiological processes affect cellular homeostasis, we believe that this comprehensive summary of miRNAs modulating both apoptosis and cholesterol metabolism will open uncharted territory for scientific exploration and will provide the foundation for discovering novel drug targets for cancer and metabolic diseases.


Assuntos
Apoptose , Colesterol/metabolismo , Doenças Metabólicas/genética , Doenças Metabólicas/patologia , MicroRNAs/genética , Neoplasias/genética , Neoplasias/patologia , Animais , Humanos
11.
J Neuroinflammation ; 13: 27, 2016 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-26838598

RESUMO

BACKGROUND: IL-1ß, also known as "the master regulator of inflammation", is a potent pro-inflammatory cytokine secreted by activated microglia in response to pathogenic invasions or neurodegeneration. It initiates a vicious cycle of inflammation and orchestrates various molecular mechanisms involved in neuroinflammation. The role of IL-1ß has been extensively studied in neurodegenerative disorders; however, molecular mechanisms underlying inflammation induced by IL-1ß are still poorly understood. The objective of our study is the comprehensive identification of molecular circuitry involved in IL-1ß-induced inflammation in microglia through protein profiling. METHODS: To achieve our aim, we performed the proteomic analysis of N9 microglial cells with and without IL-1ß treatment at different time points. Expression of HSP60 in response to IL-1ß administration was checked by quantitative real-time PCR, immunoblotting, and immunofluorescence. Interaction of HSP60 with TLR4 was determined by co-immunoprecipitation. Inhibition of TLR4 was done using TLR4 inhibitor to reveal its effect on IL-1ß-induced inflammation. Further, effect of HSP60 knockdown and overexpression were assessed on the inflammation in microglia. Specific MAPK inhibitors were used to reveal the downstream MAPK exclusively involved in HSP60-induced inflammation in microglia. RESULTS: Total 21 proteins were found to be differentially expressed in response to IL-1ß treatment in N9 microglial cells. In silico analysis of these proteins revealed unfolded protein response as one of the most significant molecular functions, and HSP60 turned out to be a key hub molecule. IL-1ß induced the expression as well as secretion of HSP60 in extracellular milieu during inflammation of N9 cells. Secreted HSP60 binds to TLR4 and inhibition of TLR4 suppressed IL-1ß-induced inflammation to a significant extent. Our knockdown and overexpression studies demonstrated that HSP60 increases the phosphorylation of ERK, JNK, and p38 MAPKs in N9 cells during inflammation. Specific inhibition of p38 by inhibitors suppressed HSP60-induced inflammation, thus pointed towards the major role of p38 MAPK rather than ERK1/2 and JNK in HSP60-induced inflammation. Furthermore, silencing of upstream modulator of p38, i.e., MEK3/6 also reduced HSP60-induced inflammation. CONCLUSIONS: IL-1ß induces expression of HSP60 in N9 microglial cells that further augments inflammation via TLR4-p38 MAPK axis.


Assuntos
Chaperonina 60/metabolismo , Inflamação , Interleucina-1beta/toxicidade , Microglia/patologia , Proteínas Mitocondriais/metabolismo , Proteína Quinase 13 Ativada por Mitógeno/metabolismo , Transdução de Sinais/fisiologia , Receptor 4 Toll-Like/metabolismo , Animais , Animais Recém-Nascidos , Células Cultivadas , Bases de Dados Bibliográficas/estatística & dados numéricos , Inibidores Enzimáticos/farmacologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Inflamação/induzido quimicamente , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Microglia/efeitos dos fármacos , Proteômica , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/fisiologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
12.
Mol Cancer ; 13: 33, 2014 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-24555688

RESUMO

MicroRNAs, the non-coding single-stranded RNA of 19-25 nucleotides are emerging as robust players of gene regulation. Plethora of evidences support that the ability of microRNAs to regulate several genes of a pathway or even multiple cross talking pathways have significant impact on a complex regulatory network and ultimately the physiological processes and diseases. Brain being a complex organ with several cell types, expresses more distinct miRNAs than any other tissues. This review aims to discuss about the microRNAs in brain development, function and their dysfunction in brain tumors. We also provide a comprehensive summary of targets of brain specific and brain enriched miRNAs that contribute to the diversity and plasticity of the brain. In particular, we uncover recent findings on miRNA-128, a brain-enriched microRNA that is induced during neuronal differentiation and whose aberrant expression has been reported in several cancers. This review describes the wide spectrum of targets of miRNA-128 that have been identified till date with potential roles in apoptosis, angiogenesis, proliferation, cholesterol metabolism, self renewal, invasion and cancer progression and how this knowledge might be exploited for the development of future miRNA-128 based therapies for the treatment of cancer as well as metabolic diseases.


Assuntos
Encéfalo/metabolismo , Regulação da Expressão Gênica/genética , MicroRNAs/genética , Animais , Apoptose/genética , Humanos , MicroRNAs/metabolismo , Neoplasias/genética , Neoplasias/metabolismo
13.
Cell Mol Life Sci ; 68(8): 1415-28, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20924637

RESUMO

MicroRNAs (miRNAs) are short ~21-nt non-coding RNA molecules that have been shown to regulate a number of biological processes. Previous reports have shown that overexpression of miR-128 in glioma cells inhibited cell proliferation. Literature also suggests that miR-128 negatively regulates prostate cancer cell invasion. Here, we show that overexpression of hsa-miR-128, a brain-enriched microRNA, induces apoptosis in HEK293T cells as elucidated by apoptosis assay, cell cycle changes, loss of mitochondrial membrane potential and multicaspase assay. By in silico analysis, we identified a putative target site within the 3' untranslated region (UTR) of Bax, a proapoptotic member of the apoptosis pathway. We found that ectopic expression of hsa-miR-128 suppressed a luciferase reporter containing the Bax-3' UTR and reduced the levels of Bax in HEK293T cells. Taken together, our study demonstrates that overexpression of hsa-miR-128 not only induces apoptosis in HEK293T cells but also is an endogenous regulator of Bax protein.


Assuntos
Apoptose/fisiologia , Regulação para Baixo , MicroRNAs/metabolismo , MicroRNAs/farmacologia , Proteína X Associada a bcl-2/metabolismo , Western Blotting , Células HEK293 , Humanos , Rim/citologia , MicroRNAs/genética , Espécies Reativas de Oxigênio , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína X Associada a bcl-2/genética
14.
PLoS One ; 4(6): e5848, 2009 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-19513126

RESUMO

miRNAs have emerged as important players in the regulation of gene expression and their deregulation is a common feature in a variety of diseases, especially cancer. Currently, many efforts are focused on studying miRNA expression patterns, as well as miRNA target validation. Here, we show that the over expression of miR-23a approximately 27a approximately 24-2 cluster in HEK293T cells induces apoptosis by caspase-dependent as well as caspase-independent pathway as proved by the annexin assay, caspase activation, release of cytochrome-c and AIF (apoptosis inducing factor) from mitochondria. Furthermore, the over expressed cluster modulates the expression of a number of genes involved in apoptosis including FADD (Fas Associated protein with Death Domain). Bioinformatically, FADD is predicted to be the target of hsa-miR-27a and interestingly, FADD protein was found to be up regulated consistent with very less expression of hsa-miR-27a in HEK293T cells. This effect was direct, as hsa-miR-27a negatively regulated the expression of FADD 3'UTR based reporter construct. Moreover, we also showed that over expression of miR-23a approximately 27a approximately 24-2 sensitized HEK293T cells to TNF-alpha cytotoxicity. Taken together, our study demonstrates that enhanced TNF-alpha induced apoptosis in HEK293T cells by over expression of miR-23a approximately 27a approximately 24-2 cluster provides new insights in the development of novel therapeutics for cancer.


Assuntos
Apoptose , Caspases/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Rim/embriologia , MicroRNAs/biossíntese , MicroRNAs/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Citocromos c/metabolismo , Humanos , Rim/citologia , Potenciais da Membrana , Ratos , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA