Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Free Radic Biol Med ; 165: 100-110, 2021 03.
Article in English | MEDLINE | ID: mdl-33497798

ABSTRACT

Fragile X-associated tremor/ataxia syndrome (FXTAS) is a progressive neurodegenerative disorder caused by an expansion of 55 to 200 CGG repeats located within 5'UTR of FMR1.These CGG repeats are transcribed into RNAs, which sequester several RNA binding proteins and alter the processing of miRNAs. CGG repeats are also translated into a toxic polyglycine-containing protein, FMRpolyG, that affects mitochondrial and nuclear functions reported in cell and animal models and patient studies. Nuclear-encoded small non-coding RNAs, including miRNAs, are transported to mitochondria; however, the role of mitochondrial miRNAs in FXTAS pathogenesis is not understood. Here, we analyzed mitochondrial miRNAs from HEK293 cells expressing expanded CGG repeats and their implication in the regulation of mitochondrial functions. The analysis of next generation sequencing (NGS) data of small RNAs from HEK293 cells expressing CGG premutation showed decreased level of cellular miRNAs and an altered pattern of association of miRNAs with mitochondria (mito-miRs). Among such mito-miRs, miR-320a was highly enriched in mitoplast and RNA immunoprecipitation of Ago2 (Argonaute-2) followed by Droplet digital PCR (ddPCR)suggested that miR-320a may form a complex with Ago2 and mitotranscripts. Finally, transfection of miR-320a mimic in cells expressing CGG permutation recovers mitochondrial functions and rescues cell death. Overall, this work reveals an altered translocation of miRNAs to mitochondria and the role of miR-320a in FXTAS pathology.


Subject(s)
MicroRNAs , Tremor , Animals , Ataxia , Cell Death , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome , HEK293 Cells , Humans , MicroRNAs/genetics , Mitochondria/genetics
2.
Mol Cell Biochem ; 461(1-2): 23-36, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31309409

ABSTRACT

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.


Subject(s)
Apoptosis/drug effects , Clindamycin/pharmacology , Erythromycin/pharmacology , Lysosomes/metabolism , Mitochondria/metabolism , Organelle Biogenesis , Anti-Bacterial Agents/pharmacology , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagy/drug effects , Cell Line , Humans , Lysosomes/drug effects , Membrane Fusion/drug effects , Mitochondria/drug effects , Mitophagy/drug effects , Models, Biological , Reactive Oxygen Species/metabolism , Ribosome Subunits, Large, Bacterial/metabolism
3.
Endocr Relat Cancer ; 26(6): 551-563, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30999276

ABSTRACT

ATC is an aggressive disease with limited therapeutic options due to drug resistance. TRAIL is an attractive anti-cancer therapy that can trigger apoptosis in a cancer cell-selective manner. However, TRAIL resistance is a major clinical obstacle for its use as a therapeutic drug. Previously, we demonstrated that MADD is a cancer cell pro-survival factor that can modulate TRAIL resistance. However, its role, if any, in overcoming TRAIL resistance in ATC is unknown. First, we characterized ATC cell lines as either TRAIL resistant, TRAIL sensitive or moderately TRAIL sensitive and evaluated MADD expression/cellular localization. We determined the effect of MADD siRNA on cellular growth and investigated its effect on TRAIL treatment. We assessed the effect of combination treatment (MADD siRNA and TRAIL) on mitochondrial membrane potential (MMP) and reactive oxygen species (ROS) levels. The effect of combination treatment on tumor growth was assessed in vivo. We found increased levels of MADD in ATC cells relative to Nthy-ori 3-1. MADD protein localizes in the cytosol (endoplasmic reticulum and Golgi body) and membrane. MADD knockdown resulted in spontaneous cell death that was synergistically enhanced when combined with TRAIL treatment in otherwise resistant ATC cells. Combination treatment resulted in a significant reduction in MMP and enhanced generation of ROS indicating the putative mechanism of action. In an orthotopic mouse model of TRAIL-resistant ATC, treatment with MADD siRNA alone reduced tumor growth that, when combined with TRAIL, resulted in significant tumor regressions. We demonstrated the potential clinical utility of MADD knockdown in sensitizing cells to TRAIL-induced apoptosis in ATC.


Subject(s)
Death Domain Receptor Signaling Adaptor Proteins/genetics , Guanine Nucleotide Exchange Factors/genetics , RNA, Small Interfering/administration & dosage , TNF-Related Apoptosis-Inducing Ligand/therapeutic use , Thyroid Carcinoma, Anaplastic/drug therapy , Thyroid Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Cell Line, Tumor , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Gene Silencing , Guanine Nucleotide Exchange Factors/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Mice, Nude , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Thyroid Carcinoma, Anaplastic/genetics , Thyroid Carcinoma, Anaplastic/metabolism , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism
4.
Cell Death Dis ; 10(2): 145, 2019 02 13.
Article in English | MEDLINE | ID: mdl-30760700

ABSTRACT

Anaplastic Thyroid Cancer (ATC) is an aggressive malignancy with limited therapeutic options and dismal patient survival. We have previously shown MADD to be differentially overexpressed in multiple cancer histologies and to contribute to tumor cell growth and survival. Therefore, we targeted MADD by gene silencing, explored its effect on cellular proliferation and metastases and examined its therapeutic potential in an orthotopic ATC model in athymic nude mice. When compared to untreated control and scramble siRNA, MADD siRNA treatment inhibited the proliferative capacity of 8505C, C643 and HTH7 cells in vitro and 8505C-derived-orthotopic tumor growth in vivo. MADD ablation caused a significant reduction in cellular migration and invasion potential; clonogenic capacity; as well as, mitochondrial length and potential in vitro. This MADD siRNA-induced anti-migratory/invasive effect corresponded with inhibition of epithelial-mesenchymal transition (EMT) and Wnt signaling. Mechanistically, MADD siRNA inhibited TNFα induced activation of pERK, pGSK3ß and ß-catenin, suggesting that MADD knockdown might exert its anti-migratory/invasive effects, by blocking TNFα/ERK/GSK3ß axis. MADD siRNA can inhibit ß-catenin nuclear translocation and consequently, the expression of its target genes in ATC cells. In in vivo experiments, along with tumor regression, MADD siRNA treatment also decreased evidence of lung metastases. Immunohistochemically, MADD siRNA-treated tumor tissues exhibited a reduction in Ki67 and N-Cadherin expression, and an increase in E-Cadherin expression. In conclusion, we show the crucial role of MADD in ATC tumorigenesis and metastasis and its potential implications as a molecular target for ATC therapy.


Subject(s)
Death Domain Receptor Signaling Adaptor Proteins/biosynthesis , Guanine Nucleotide Exchange Factors/biosynthesis , Thyroid Carcinoma, Anaplastic/metabolism , Thyroid Neoplasms/metabolism , Animals , Cell Cycle Checkpoints/physiology , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Death Domain Receptor Signaling Adaptor Proteins/deficiency , Death Domain Receptor Signaling Adaptor Proteins/genetics , Gene Knockdown Techniques , Guanine Nucleotide Exchange Factors/deficiency , Guanine Nucleotide Exchange Factors/genetics , Heterografts , Humans , Mice , Mice, Nude , Thyroid Carcinoma, Anaplastic/genetics , Thyroid Carcinoma, Anaplastic/pathology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Transfection
5.
Biochim Biophys Acta Mol Basis Dis ; 1865(6): 1379-1388, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30771487

ABSTRACT

Fragile X-associated tremor/ataxia syndrome (FXTAS) is an inherited neurodegenerative disorder caused by an expansion of 55 to 200 CGG repeats (premutation) in FMR1. These CGG repeats are Repeat Associated non-ATG (RAN) translated into a small and pathogenic protein, FMRpolyG. The cellular and molecular mechanisms of FMRpolyG toxicity are unclear. Various mitochondrial dysfunctions have been observed in FXTAS patients and animal models. However, the causes of these mitochondrial alterations are not well understood. In the current study, we investigated interaction of FMRpolyG with mitochondria and its role in modulating mitochondrial functions. Beside nuclear inclusions, FMRpolyG also formed small cytosolic aggregates that interact with mitochondria both in cell and mouse model of FXTAS. Importantly, expression of FMRpolyG reduces ATP levels, mitochondrial transmembrane potential, mitochondrial supercomplexes assemblies and activities and expression of mitochondrial DNA encoded transcripts in cell and animal model of FXTAS, as well as in FXTAS patient brain tissues. Overall, these results suggest that FMRpolyG alters mitochondrial functions, bioenergetics and initiates cell death. The further study in this direction will help to establish the role of mitochondria in FXTAS conditions.


Subject(s)
Ataxia/genetics , Electron Transport Chain Complex Proteins/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Mitochondria/genetics , RNA, Messenger/genetics , Tremor/genetics , Trinucleotide Repeat Expansion , Adenosine Triphosphate/biosynthesis , Aged , Aged, 80 and over , Animals , Ataxia/metabolism , Ataxia/pathology , Cell Line, Tumor , Cerebellum/metabolism , Cerebellum/pathology , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Disease Models, Animal , Electron Transport Chain Complex Proteins/metabolism , Energy Metabolism/genetics , Fragile X Mental Retardation Protein/chemistry , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Fragile X Syndrome/pathology , Gene Expression , HEK293 Cells , Humans , Membrane Potential, Mitochondrial/genetics , Mice , Mice, Transgenic , Mitochondria/metabolism , Mitochondria/pathology , Neurons/metabolism , Neurons/pathology , Protein Aggregates/genetics , RNA, Messenger/metabolism , Tremor/metabolism , Tremor/pathology
6.
Biochim Biophys Acta Mol Basis Dis ; 1865(6): 1460-1476, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30802640

ABSTRACT

An increased level of proinflammatory cytokines, including TNF-α in tumor microenvironment regulates the bioenergetic capacity, immune evasion and survival of cancer cells. Emerging evidences suggest that mitochondrial immune signaling proteins modulates mitochondrial bioenergetic capacity, in addition to the regulation of innate immune response. The optimal oxidative phosphorylation (OxPhos) capacity is required for the maintenance of functional lysosomes and autophagy flux. NLRX1, a mitochondrial NOD family receptor protein, regulates mitochondrial function during apoptosis and tissue injury. However, its role in regulation of mitochondrial and lysosomal function to modulate autophagy flux during inflammatory conditions is not understood. In the current study, we investigated the role of NLRX1 in modulating TNF-α induced autophagy flux and mitochondrial turnover and its implication in regulating the invasive and metastatic capability of breast cancer cells. Expression analyses of clinical breast cancer samples and meta-analysis of multiple public databases revealed that NLRX1 expression is significantly increased in basal-like and metastatic breast carcinoma as compared to non-basal-like and primary breast cancer. Depletion of NLRX1 expression in triple-negative breast cancer cells, altered the organization and activity of OxPhos complexes in presence of TNF-α. NLRX1 depletion further impaired lysosomal function and hence the turnover of damaged mitochondria through mitophagy in presence of TNF-α. Importantly, loss of NLRX1 decreased OxPhos-dependent cell proliferation and migration ability of triple-negative breast cancer cells in presence of TNF-α. These evidences suggest an essential role of NLRX1 in maintaining the crosstalk of mitochondrial metabolism and lysosomal function to regulate invasion and metastasis capability of breast cancer cells.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Lysosomes/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Tumor Necrosis Factor-alpha/genetics , Autophagy/drug effects , Autophagy/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , HEK293 Cells , Humans , Lymphatic Metastasis , Lysosomes/drug effects , MCF-7 Cells , Mitochondria/drug effects , Mitochondria/pathology , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/metabolism , Mitophagy/drug effects , Mitophagy/genetics , Neoplasm Invasiveness , Oxidative Phosphorylation/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Tumor Microenvironment/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
7.
Biochim Biophys Acta Mol Cell Res ; 1865(9): 1260-1276, 2018 09.
Article in English | MEDLINE | ID: mdl-29932989

ABSTRACT

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.


Subject(s)
Mitochondria/genetics , Mitochondrial Proteins/metabolism , RNA, Mitochondrial/metabolism , RNA-Binding Proteins/metabolism , Energy Metabolism , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , MCF-7 Cells , Mitochondria/metabolism , Protein Transport , RNA, Mitochondrial/genetics
8.
Mol Neurobiol ; 55(6): 4689-4701, 2018 Jun.
Article in English | MEDLINE | ID: mdl-28710704

ABSTRACT

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.


Subject(s)
Lysosomes/metabolism , MicroRNAs/metabolism , Mitochondria/metabolism , Parkinson Disease/genetics , Stress, Physiological/genetics , Apoptosis/genetics , Autophagosomes/metabolism , Cell Line, Tumor , Humans , Membrane Fusion , MicroRNAs/genetics , Models, Biological , Neurons/metabolism , Neurons/pathology , Oxidopamine , Parkinson Disease/pathology
9.
Cell Signal ; 35: 73-83, 2017 07.
Article in English | MEDLINE | ID: mdl-28366813

ABSTRACT

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.


Subject(s)
Breast Neoplasms/genetics , Carcinogenesis/genetics , Inflammation/genetics , Interferon Regulatory Factor-3/genetics , Membrane Proteins/genetics , Autophagosomes/metabolism , Autophagy/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Immunity, Innate/genetics , Inflammation/metabolism , Inflammation/pathology , Interferon Regulatory Factor-3/metabolism , Lysosomes/metabolism , Membrane Proteins/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitophagy/genetics , NF-kappa B/genetics , Signal Transduction/genetics , Sirolimus/administration & dosage
10.
J Mol Med (Berl) ; 95(6): 641-651, 2017 06.
Article in English | MEDLINE | ID: mdl-28220193

ABSTRACT

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.


Subject(s)
Breast Neoplasms/metabolism , MicroRNAs/metabolism , Mitochondria/metabolism , RNA, Ribosomal, 16S/metabolism , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Survival , Humans , Mice, Nude , Transcytosis
11.
Biochim Biophys Acta ; 1853(5): 1073-86, 2015 May.
Article in English | MEDLINE | ID: mdl-25639646

ABSTRACT

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.


Subject(s)
Apoptosis/drug effects , Mitochondrial Proteins/metabolism , Neoplasms/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Tumor Suppressor Proteins/metabolism , Adenosine Triphosphate/metabolism , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Caspase 8/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Clone Cells , Electron Transport Chain Complex Proteins/metabolism , Enzyme Activation/drug effects , Humans , Mice, Nude , Mitochondria/drug effects , Mitochondria/metabolism , Neoplasms/pathology , Protein Binding/drug effects , Protein Transport/drug effects , Reactive Oxygen Species/metabolism , Rotenone/pharmacology
12.
Biochim Biophys Acta ; 1852(3): 451-61, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25481834

ABSTRACT

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.


Subject(s)
Dopaminergic Neurons/enzymology , Electron Transport Complex I/metabolism , MicroRNAs/metabolism , Mitochondria/enzymology , Parkinson Disease/enzymology , Tumor Necrosis Factor-alpha/pharmacology , Acetylcysteine/pharmacology , Adenosine Triphosphate/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Cell Death/drug effects , Cell Line , Dopaminergic Neurons/pathology , Free Radical Scavengers/pharmacology , Humans , Mitochondria/pathology , Parkinson Disease/pathology , Protease Inhibitors/pharmacology , Tumor Necrosis Factor-alpha/metabolism
13.
Mitochondrion ; 16: 50-4, 2014 May.
Article in English | MEDLINE | ID: mdl-24246912

ABSTRACT

Antibiotics are frontline therapy against microbial infectious diseases. Many antibiotics are known to cause several side effects in humans. Ribosomal RNA (rRNA) is the main target of antibiotics that inhibit protein synthesis. According to the endosymbiont theory, mitochondrion is of bacterial origin and their molecular and structural components of the protein expression system are almost similar. It has been observed that the rate of mutations in mitochondrial rRNA is higher as compared to that of nuclear rRNA. The presence of these mutations may mimic prokaryotic rRNA structure and bind to antibiotics targeted to ribosomes of bacteria. Mitochondrial functions are compromised hence may be one of the major causes of side effects observed during antibiotic therapy. The current review had summarized the studies on the role of antibiotics on mitochondrial functions and its relevance to the observed side effects in physiological and pathological conditions.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/adverse effects , Bacterial Infections/drug therapy , Mitochondria/drug effects , Humans , Metabolic Networks and Pathways/drug effects
14.
Biochim Biophys Acta ; 1842(2): 144-53, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24239807

ABSTRACT

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.


Subject(s)
Breast Neoplasms/metabolism , Membrane Proteins/metabolism , NF-kappa B/metabolism , Tumor Suppressor Proteins/metabolism , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Caspases/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Cell Survival/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , MCF-7 Cells , Membrane Proteins/genetics , NF-kappa B/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Proteins/genetics
15.
Biochim Biophys Acta ; 1833(12): 3134-3144, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24021263

ABSTRACT

The emerging evidences suggest that endoplasmic (ER) stress is involved in onset of many pathological conditions like cancer and neurodegeneration. The persistent ER stress results in misfolded protein aggregates, which are degraded through the process of autophagy or lead to cell death through activation of caspases. The regulation of crosstalk of autophagy and cell death during ER stress is emerging. Ubiquitination plays regulatory role in crosstalk of autophagy and cell death. In the current study, we describe the role of TRIM13, RING E3 ubiquitin ligase, in regulation of ER stress induced cell death. The expression of TRIM13 sensitizes cells to ER stress induced death. TRIM13 induced autophagy is essential for ER stress induced caspase activation and cell death. TRIM13 induces K63 linked poly-ubiquitination of caspase-8, which results in its stabilization and activation during ER stress. TRIM13 regulates translocation of caspase-8 to autophagosome and its fusion with lysosome during ER stress. This study first time demonstrated the role of TRIM13 as novel regulator of caspase-8 activation and cell death during ER stress.


Subject(s)
Caspase 8/metabolism , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum Stress , Phagosomes/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitination , Adaptor Proteins, Signal Transducing/metabolism , Autophagy/drug effects , Endoplasmic Reticulum Stress/drug effects , Enzyme Activation/drug effects , HEK293 Cells , Humans , Lysosomes/drug effects , Lysosomes/metabolism , MCF-7 Cells , Phagosomes/drug effects , Protein Transport/drug effects , Regulatory Factor X Transcription Factors , Sequestosome-1 Protein , Signal Transduction/drug effects , Transcription Factors/metabolism , Tunicamycin/pharmacology , Ubiquitination/drug effects , Unfolded Protein Response/drug effects
16.
PLoS One ; 7(11): e48662, 2012.
Article in English | MEDLINE | ID: mdl-23152791

ABSTRACT

TNF induced nuclear factor kappa B (NF-κB) is one of the central signaling pathways that plays a critical role in carcinogenesis and inflammatory diseases. Post-translational modification through ubiquitin plays important role in the regulation of this pathway. In the current study, we investigated the role of TRIM8, member of RING family ubiquitin ligase in regulation of NF-κB pathway. We observed that TRIM8 positively regulates TNF induced NF-κB pathway. Different domains of TRIM8 showed discrete functions at the different steps in regulation of TNF induced NF-κB pathway. Ubiquitin ligase activity of TRIM8 is essential for regulation of NF-κB activation in both cytoplasm as well as nucleus. TRIM8 negates PIAS3 mediated negative repression of NF-κB at p65 by inducing translocation of PIAS3 from nucleus to cytoplasm as well as its turnover. TNF induces translocation of TRIM8 from nucleus to cytoplasm, which positively regulates NF-κB. The cytoplasmic translocation of TRIM8 is essential for TNF induced NF-κB but not for p65 mediated NF-κB regulation. TRIM8 also enhanced the clonogenic and migration ability of cells by modulating NF-κB. The further study will help to understand the role of TRIM8 in inflammation and cancer.


Subject(s)
Carrier Proteins/metabolism , I-kappa B Kinase/metabolism , Nerve Tissue Proteins/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Carrier Proteins/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Nucleus/metabolism , Cytoplasm/metabolism , Enzyme Activation , Gene Silencing , Humans , MCF-7 Cells , Molecular Chaperones/metabolism , Nerve Tissue Proteins/genetics , Protein Inhibitors of Activated STAT/metabolism , Protein Interaction Domains and Motifs , Protein Transport
17.
Mitochondrion ; 12(6): 593-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23085198

ABSTRACT

The cellular processes are controlled by a narrow range of mRNA and proteins levels, where small RNAs (sRNAs) known as miRNAs play a critical role. The spatial and temporal regulation of miRNA processing components and mature miRNA is emerging. The recent studies suggest that mitochondria are one of the destinations of pre as well as mature miRNAs. The role of mitochondria extends beyond energy metabolism to many other cellular processes like metabolism, cell death and inflammation. The new found destination of miRNAs suggest the role of mitochondria in monitoring site specific regulations of proteins as well as the function of mitochondria. The studies in this direction will decipher the novel role of mitochondria-associated miRNAs in different cellular processes. This review is focussed on the recent studies demonstrating the presence of miRNAs in mitochondria and its possible significance in different cellular and physiological conditions.


Subject(s)
Gene Expression Regulation , MicroRNAs/genetics , MicroRNAs/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Animals , Humans
18.
PLoS One ; 7(9): e44873, 2012.
Article in English | MEDLINE | ID: mdl-22984580

ABSTRACT

Mitochondria are one of the central regulators of many cellular processes beyond its well established role in energy metabolism. The inter-organellar crosstalk is critical for the optimal function of mitochondria. Many nuclear encoded proteins and RNA are imported to mitochondria. The translocation of small RNA (sRNA) including miRNA to mitochondria and other sub-cellular organelle is still not clear. We characterized here sRNA including miRNA associated with human mitochondria by cellular fractionation and deep sequencing approach. Mitochondria were purified from HEK293 and HeLa cells for RNA isolation. The sRNA library was generated and sequenced using Illumina system. The analysis showed the presence of unique population of sRNA associated with mitochondria including miRNA. Putative novel miRNAs were characterized from unannotated sRNA sequences. The study showed the association of 428 known, 196 putative novel miRNAs to mitochondria of HEK293 and 327 known, 13 putative novel miRNAs to mitochondria of HeLa cells. The alignment of sRNA to mitochondrial genome was also studied. The targets were analyzed using DAVID to classify them in unique networks using GO and KEGG tools. Analysis of identified targets showed that miRNA associated with mitochondria regulates critical cellular processes like RNA turnover, apoptosis, cell cycle and nucleotide metabolism. The six miRNAs (counts >1000) associated with mitochondria of both HEK293 and HeLa were validated by RT-qPCR. To our knowledge, this is the first systematic study demonstrating the associations of sRNA including miRNA with mitochondria that may regulate site-specific turnover of target mRNA important for mitochondrial related functions.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , MicroRNAs/metabolism , Mitochondria/metabolism , Conserved Sequence/genetics , DNA, Mitochondrial/metabolism , Genome , HEK293 Cells , HeLa Cells , Humans , RNA, Messenger/metabolism , Ribonuclease, Pancreatic/metabolism , Sequence Analysis, RNA/methods , Subcellular Fractions/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...