Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Biochim Biophys Acta Mol Cell Res ; 1865(9): 1260-1276, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29932989

RESUMEN

The role of mitochondria is emerging in regulation of innate immunity, inflammation and cell death beyond its primary role in energy metabolism. Mitochondria act as molecular platform for immune adaptor protein complexes, which participate in innate immune signaling. The mitochondrial localized immune adaptors are widely expressed in non-immune cells, however their role in regulation of mitochondrial function and metabolic adaption is not well understood. NLRX1, a member of NOD family receptor proteins, localizes to mitochondria and is a negative regulator of anti-viral signaling. However, the submitochondrial localization of NLRX1 and its implication in regulation of mitochondrial functions remains elusive. Here, we confirm that NLRX1 translocates to mitochondrial matrix and associates with mitochondrial FASTKD5 (Fas-activated serine-threonine kinase family protein-5), a bonafide component of mitochondrial RNA granules (MRGs). The association of NLRX1 with FASTKD5 negatively regulates the processing of mitochondrial genome encoded transcripts for key components of complex-I and complex-IV, to modulate its activity and supercomplexes formation. The evidences, here, suggest an important role of NLRX1 in regulating the post-transcriptional processing of mitochondrial RNA, which may have an important implication in bioenergetic adaptation during metabolic stress, oncogenic transformation and innate immunity.


Asunto(s)
Mitocondrias/genética , Proteínas Mitocondriales/metabolismo , ARN Mitocondrial/metabolismo , Proteínas de Unión al ARN/metabolismo , Metabolismo Energético , Regulación de la Expresión Génica , Células HEK293 , Células HeLa , Humanos , Células MCF-7 , Mitocondrias/metabolismo , Transporte de Proteínas , ARN Mitocondrial/genética
2.
Mol Cell Biochem ; 461(1-2): 23-36, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31309409

RESUMEN

Antibiotics are the front-line treatment against many bacterial infectious diseases in human. The excessive and long-term use of antibiotics in human cause several side effects. It is important to understand the underlying molecular mechanisms of action of antibiotics in the host cell to avoid the side effects due to the prevalent uses. In the current study, we investigated the crosstalk between mitochondria and lysosomes in the presence of widely used antibiotics: erythromycin (ERM) and clindamycin (CLDM), which target the 50S subunit of bacterial ribosomes. We report here that both ERM and CLDM induced caspase activation and cell death in several different human cell lines. The activity of the mitochondrial respiratory chain was compromised in the presence of ERM and CLDM leading to bioenergetic crisis and generation of reactive oxygen species. Antibiotics treatment impaired autophagy flux and lysosome numbers, resulting in decreased removal of damaged mitochondria through mitophagy, hence accumulation of defective mitochondria. We further show that over-expression of transcription factor EB (TFEB) increased the lysosome number, restored mitochondrial function and rescued ERM- and CLDM-induced cell death. These studies indicate that antibiotics alter mitochondria and lysosome interactions leading to apoptotsis and may develop a novel approach for targeting inter-organelle crosstalk to limit deleterious antibiotic-induced side effects.


Asunto(s)
Apoptosis/efectos de los fármacos , Clindamicina/farmacología , Eritromicina/farmacología , Lisosomas/metabolismo , Mitocondrias/metabolismo , Biogénesis de Organelos , Antibacterianos/farmacología , Autofagosomas/efectos de los fármacos , Autofagosomas/metabolismo , Autofagia/efectos de los fármacos , Línea Celular , Humanos , Lisosomas/efectos de los fármacos , Fusión de Membrana/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitofagia/efectos de los fármacos , Modelos Biológicos , Especies Reactivas de Oxígeno/metabolismo , Subunidades Ribosómicas Grandes Bacterianas/metabolismo
3.
Biochim Biophys Acta ; 1852(3): 451-61, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25481834

RESUMEN

Parkinson's disease (PD) is a complex neurological disorder of the elderly population and majorly shows the selective loss of dopaminergic (DAergic) neurons in the substantia nigra pars compacta (SNpc) region of the brain. The mechanisms leading to increased cell death of DAergic neurons are not well understood. Tumor necrosis factor-alpha (TNF-α), a pro-inflammatory cytokine is elevated in blood, CSF and striatum region of the brain in PD patients. The increased level of TNF-α and its role in pathogenesis of PD are not well understood. In the current study, we investigated the role of TNF-α in the regulation of cell death and miRNA mediated mitochondrial functions using, DAergic cell line, SH-SY5Y (model of dopaminergic neuron degeneration akin to PD). The cells treated with low dose of TNF-α for prolonged period induce cell death which was rescued in the presence of zVAD.fmk, a caspase inhibitor and N-acetyl-cysteine (NAC), an antioxidant. TNF-α alters mitochondrial complex-I activity, decreases adenosine triphosphate (ATP) levels, increases reactive oxygen species levels and mitochondrial turnover through autophagy. TNF-α differentially regulates miRNA expression involved in pathogenesis of PD. Bioinformatics analysis revealed that the putative targets of altered miRNA included both pro/anti apoptotic genes and subunits of mitochondrial complex. The cells treated with TNF-α showed decreased level of nuclear encoded transcript of mitochondrial complexes, the target of miRNA. To our knowledge, the evidences in the current study demonstrated that TNF-α is a potential regulator of miRNAs which may regulate mitochondrial functions and neuronal cell death, having important implication in pathogenesis of PD.


Asunto(s)
Neuronas Dopaminérgicas/enzimología , Complejo I de Transporte de Electrón/metabolismo , MicroARNs/metabolismo , Mitocondrias/enzimología , Enfermedad de Parkinson/enzimología , Factor de Necrosis Tumoral alfa/farmacología , Acetilcisteína/farmacología , Adenosina Trifosfato/metabolismo , Clorometilcetonas de Aminoácidos/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular , Neuronas Dopaminérgicas/patología , Depuradores de Radicales Libres/farmacología , Humanos , Mitocondrias/patología , Enfermedad de Parkinson/patología , Inhibidores de Proteasas/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
4.
Biochim Biophys Acta ; 1853(5): 1073-86, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25639646

RESUMEN

Chronic inflammation in tumor microenvironment plays an important role at different stages of tumor development. The specific mechanisms of the association and its role in providing a survival advantage to the tumor cells are not well understood. Mitochondria are emerging as a central platform for the assembly of signaling complexes regulating inflammatory pathways, including the activation of type-I IFN and NF-κB. These complexes in turn may affect metabolic functions of mitochondria and promote tumorigenesis. NLRX1, a mitochondrial NOD-like receptor protein, regulate inflammatory pathways, however its role in regulation of cross talk of cell death and metabolism and its implication in tumorigenesis is not well understood. Here we demonstrate that NLRX1 sensitizes cells to TNF-α induced cell death by activating Caspase-8. In the presence of TNF-α, NLRX1 and active subunits of Caspase-8 are preferentially localized to mitochondria and regulate the mitochondrial ROS generation. NLRX1 regulates mitochondrial Complex I and Complex III activities to maintain ATP levels in the presence of TNF-α. The expression of NLRX1 compromises clonogenicity, anchorage-independent growth, migration of cancer cells in vitro and suppresses tumorigenicity in vivo in nude mice. We conclude that NLRX1 acts as a potential tumor suppressor by regulating the TNF-α induced cell death and metabolism.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas Mitocondriales/metabolismo , Neoplasias/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Proteínas Supresoras de Tumor/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Carcinogénesis/metabolismo , Carcinogénesis/patología , Caspasa 8/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Clonales , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Activación Enzimática/efectos de los fármacos , Humanos , Ratones Desnudos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Neoplasias/patología , Unión Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Rotenona/farmacología
5.
Biochim Biophys Acta ; 1842(2): 144-53, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24239807

RESUMEN

Emerging evidences suggest that chronic inflammation is one of the major causes of tumorigenesis. The role of inflammation in regulation of breast cancer progression is not well established. Recently Mediator of IRF3 Activation (MITA) protein has been identified that regulates NF-κB and IFN pathways. Role of MITA in the context of inflammation and cancer progression has not been investigated. In the current report, we studied the role of MITA in the regulation of cross talk between cell death and inflammation in breast cancer cells. The expression of MITA was significantly lower on in estrogen receptor (ER) positive breast cancer cells than ER negative cells. Similarly, it was significantly down regulated in tumor tissue as compared to the normal tissue. The overexpression of MITA in MCF-7 and T47D decreases the cell proliferation and increases the cell death by activation of caspases. MITA positively regulates NF-κB transcription factor, which is essential for MITA induced cell death. The activation of NF-κB induces TNF-α production which further sensitizes MITA induced cell death by activation of death receptor pathway through capsase-8. MITA expression decreases the colony forming units and migration ability of MCF-7 cells. Thus, our finding suggests that MITA acts as a tumor suppressor which is down regulated during tumorigenesis providing survival advantage to tumor cell.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas de la Membrana/metabolismo , FN-kappa B/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Caspasas/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Supervivencia Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Células MCF-7 , Proteínas de la Membrana/genética , FN-kappa B/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Supresoras de Tumor/genética
6.
Mol Neurobiol ; 55(6): 4689-4701, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28710704

RESUMEN

Parkinson's disease (PD) is complex neurological disorder and is prevalent in the elderly population. This is primarily due to loss of dopaminergic neurons in the substantia nigra pars compacta (SNc) region of the brain. The modulators of the selective loss of dopaminergic neurons in PD are still not well understood. The small non-coding RNAs specifically miRNAs fine-tune the protein levels by post-transcriptional gene regulation. The role of miRNAs in PD pathogenesis is still not well characterized. In the current study, we identified the miRNA expression pattern in 6-OHDA-induced PD stress condition in SH-SY5Y, dopaminergic neuronal cell line. The targets of top 5 miRNAs both up- and down regulated were analyzed by using StarBase. The putative pathways of identified miRNAs included neurotrophin signaling, neuronal processes, mTOR, and cell death. The level of miR-5701 was significantly downregulated in the presence of 6-OHDA. The putative targets of miR-5701 miRNA include genes involved in lysosomal biogenesis and mitochondrial quality control. The transfection of miR-5701 mimic decreased the transcript level of VCP, LAPTM4A, and ATP6V0D1. The expression of miR-5701 mimic induces mitochondrial dysfunction, defect in autophagy flux, and further sensitizes SH-SY5Y cells to 6-OHDA-induced cell death. To our knowledge, the evidence in the current study demonstrated the dysregulation of specific pattern of miRNAs in PD stress conditions. We further characterized the role of miR-5701, a novel miRNA, as a potential regulator of the mitochondrial and lysosomal function determining the fate of neurons which has important implication in the pathogenesis of PD.


Asunto(s)
Lisosomas/metabolismo , MicroARNs/metabolismo , Mitocondrias/metabolismo , Enfermedad de Parkinson/genética , Estrés Fisiológico/genética , Apoptosis/genética , Autofagosomas/metabolismo , Línea Celular Tumoral , Humanos , Fusión de Membrana , MicroARNs/genética , Modelos Biológicos , Neuronas/metabolismo , Neuronas/patología , Oxidopamina , Enfermedad de Parkinson/patología
7.
Cell Signal ; 48: 1-12, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29678622

RESUMEN

In cancer patients, treatment modalities like chemotherapy and radiation exert their anticancer effects by inducing DNA damage. The cancer cells can survive under genotoxic stress by inducing DNA damage response (DDR) or can undergo cell death. The process of autophagy is emerging as crucial regulator of cell survival during different stress conditions. Post translational modification through ubiquitin plays an essential role in DDR during genotoxic stress conditions. Ubiquitin ligases regulate autophagy and cell death pathways however their role during genotoxic stress conditions is not understood. In the current study we identified TRIM8, RING E3 Ligase, as a novel regulator of autophagy during DDR. TRIM8 regulates lysosomal biogenesis and autophagy flux. The turnover of TRIM8 is high and is stabilized during genotoxic stress conditions. TRIM8 regulated autophagy is essential for its cytoprotective role during genotoxic stress induced cell death. TRIM8 stabilizes the turnover of XIAP during genotoxic stress and forms complex with XIAP and caspase-3 to inhibit its activation in presence of etoposide. TRIM8 mediated autophagy promotes degradation of cleaved caspase-3 subunits. This study described TRIM8, as a novel regulator of DDR-autophagy crosstalk, which may play role in survival of cancer cells in presence of genotoxic agents.


Asunto(s)
Autofagia , Proteínas Portadoras/fisiología , Caspasa 3/metabolismo , Daño del ADN , Proteínas del Tejido Nervioso/fisiología , Supervivencia Celular , Células HEK293 , Células HeLa , Humanos , Lisosomas/metabolismo , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo
8.
Cell Signal ; 35: 73-83, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28366813

RESUMEN

The crosstalk between inflammation and autophagy is an emerging phenomenon observed during tumorigenesis. Activation of NF-κB and IRF3 plays a key role in the regulation of cytokines that are involved in tumor growth and progression. The genes of innate immunity are known to regulate the master transcription factors like NF-κB and IRF3. Innate immunity pathways at the same time regulate the genes of the autophagy pathway which are essential for tumor cell metabolism. In the current study, we studied the role of MITA (Mediator of IRF3 Activation), a regulator of innate immunity, in the regulation of autophagy and its implication in cell death of breast cancer cells. Here, we report that MITA inhibits the fusion of autophagosome with lysosome as evident from different autophagy flux assays. The expression of MITA induces the translocation of p62 and NDP52 to mitochondria which further recruits LC3 for autophagosome formation. The expression of MITA decreased mitochondrial number and enhances mitochondrial ROS by increasing complex-I activity. The enhancement of autophagy flux with rapamycin or TFEB expression normalized MITA induced cell death. The evidences clearly show that MITA regulates autophagy flux and modulates mitochondrial turnover through mitophagy.


Asunto(s)
Neoplasias de la Mama/genética , Carcinogénesis/genética , Inflamación/genética , Factor 3 Regulador del Interferón/genética , Proteínas de la Membrana/genética , Autofagosomas/metabolismo , Autofagia/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Inmunidad Innata/genética , Inflamación/metabolismo , Inflamación/patología , Factor 3 Regulador del Interferón/metabolismo , Lisosomas/metabolismo , Proteínas de la Membrana/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Mitofagia/genética , FN-kappa B/genética , Transducción de Señal/genética , Sirolimus/administración & dosificación
9.
J Mol Med (Berl) ; 95(6): 641-651, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28220193

RESUMEN

The modulation of mitochondrial functions is important for maintaining cellular homeostasis. Mitochondria essentially depend on the import of RNAs and proteins encoded by the nuclear genome. MicroRNAs encoded in the nucleus can translocate to mitochondria and target the genome, affecting mitochondrial function. Here, we analyzed the role of miR-4485 in the regulation of mitochondrial functions. We showed that miR-4485 translocated to mitochondria where its levels varied in response to different stress conditions. A direct binding of miR-4485 to mitochondrial 16S rRNA was demonstrated. MiR-4485 regulated the processing of pre-rRNA at the 16S rRNA-ND1 junction and the translation of downstream transcripts. MiR-4485 modulated mitochondrial complex I activity, the production of ATP, ROS levels, caspase-3/7 activation, and apoptosis. Transfection of a miR-4485 mimic downregulated the expression of regulatory glycolytic pathway genes and reduced the clonogenic ability of breast cancer cells. Ectopic expression of miR-4485 in MDA-MB-231 breast carcinoma cells decreased the tumorigenicity in a nude mouse xenograft model. Furthermore, levels of both precursor and mature miR-4485 are decreased in tumor tissue of breast cancer patients. We conclude that the mitochondria-targeted miR-4485 may act as a tumor suppressor in breast carcinoma cells by negatively regulating mitochondrial RNA processing and mitochondrial functions.


Asunto(s)
Neoplasias de la Mama/metabolismo , MicroARNs/metabolismo , Mitocondrias/metabolismo , ARN Ribosómico 16S/metabolismo , Animales , Línea Celular Tumoral , Núcleo Celular/metabolismo , Supervivencia Celular , Humanos , Ratones Desnudos , Transcitosis
10.
Free Radic Biol Med ; 89: 1036-48, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26524401

RESUMEN

The emerging evidences suggest that posttranslational modification of target protein by ubiquitin (Ub) not only regulate its turnover through ubiquitin proteasome system (UPS) but is a critical regulator of various signaling pathways. During ubiquitination, E3 ligase recognizes the target protein and determines the topology of ubiquitin chains. In current study, we studied the role of TRIM4, a member of the TRIM/RBCC protein family of RING E3 ligase, in regulation of hydrogen peroxide (H2O2) induced cell death. TRIM4 is expressed differentially in human tissues and expressed in most of the analyzed human cancer cell lines. The subcellular localization studies showed that TRIM4 forms distinct cytoplasmic speckle like structures which transiently interacts with mitochondria. The expression of TRIM4 induces mitochondrial aggregation and increased level of mitochondrial ROS in the presence of H2O2. It sensitizes the cells to H2O2 induced death whereas knockdown reversed the effect. TRIM4 potentiates the loss of mitochondrial transmembrane potential and cytochrome c release in the presence of H2O2. The analysis of TRIM4 interacting proteins showed its interaction with peroxiredoxin 1 (PRX1), including other proteins involved in regulation of mitochondrial and redox homeostasis. TRIM4 interaction with PRX1 is critical for the regulation of H2O2 induced cell death. Collectively, the evidences in the current study suggest the role of TRIM4 in regulation of oxidative stress induced cell death.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Oxidantes/farmacología , Estrés Oxidativo/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/genética , Western Blotting , Proliferación Celular/efectos de los fármacos , Citocromos c/metabolismo , Técnica del Anticuerpo Fluorescente , Células HEK293 , Humanos , Inmunoprecipitación , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Proteínas de la Membrana/genética , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Proteómica , ARN Mensajero/genética , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Ubiquitina/metabolismo , Ubiquitinación/efectos de los fármacos
11.
Cell Signal ; 26(11): 2350-7, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25093807

RESUMEN

Breast cancer is one of the leading causes of mortality in the females. Intensive efforts have been made to understand the molecular mechanisms of pathogenesis of breast cancer. The physiological conditions that lead to tumorigenesis including breast cancer are not well understood. Toll like receptors (TLRs) are essential components of innate immune system that protect the host against bacterial and viral infection. The emerging evidences suggest that TLRs are activated through pathogen associated molecular patterns (PAMPs) as well as endogenous molecules, which lead to the activation of inflammatory pathways. This leads to increased levels of several pro-inflammatory cytokines and chemokines mounting inflammation. Several evidences support the view that chronic inflammation can lead to cancerous condition. Inflammation aids in tumor progression and metastasis. Association of inflammation with breast cancer is emerging. TLR mediated activation of NF-κB and IRF is an essential link connecting inflammation to cancer. The recent reports provide several evidences, which suggest the important role of TLRs in breast cancer pathogenesis and recurrence. The current review focuses on emerging studies suggesting the strong linkages of TLR mediated regulation of inflammation during breast cancer and its metastasis emphasizing the initiation of the systematic study.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores Toll-Like/metabolismo , Animales , Neoplasias de la Mama/etiología , Neoplasias de la Mama/genética , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Quimiocinas/genética , Quimiocinas/metabolismo , Enfermedad Crónica , Femenino , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/mortalidad , Inflamación/patología , Ratones , FN-kappa B/genética , FN-kappa B/metabolismo , Proteínas de Neoplasias/genética , Receptores Toll-Like/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA