Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Int J Mol Sci ; 25(5)2024 Feb 24.
Article in English | MEDLINE | ID: mdl-38473893

ABSTRACT

Neurological diseases and neurotrauma manifest significant sex differences in prevalence, progression, outcome, and therapeutic responses. Genetic predisposition, sex hormones, inflammation, and environmental exposures are among many physiological and pathological factors that impact the sex disparity in neurological diseases. MicroRNAs (miRNAs) are a powerful class of gene expression regulator that are extensively involved in mediating biological pathways. Emerging evidence demonstrates that miRNAs play a crucial role in the sex dimorphism observed in various human diseases, including neurological diseases. Understanding the sex differences in miRNA expression and response is believed to have important implications for assessing the risk of neurological disease, defining therapeutic intervention strategies, and advancing both basic research and clinical investigations. However, there is limited research exploring the extent to which miRNAs contribute to the sex disparities observed in various neurological diseases. Here, we review the current state of knowledge related to the sexual dimorphism in miRNAs in neurological diseases and neurotrauma research. We also discuss how sex chromosomes may contribute to the miRNA sexual dimorphism phenomenon. We attempt to emphasize the significance of sexual dimorphism in miRNA biology in human diseases and to advocate a gender/sex-balanced science.


Subject(s)
MicroRNAs , Nervous System Diseases , Humans , Female , Male , MicroRNAs/genetics , Gonadal Steroid Hormones
2.
J Neurotrauma ; 40(21-22): 2396-2409, 2023 11.
Article in English | MEDLINE | ID: mdl-37476976

ABSTRACT

Mild traumatic brain injury (mTBI) results in impairment of brain metabolism, which is propagated by mitochondrial dysfunction in the brain. Mitochondrial dysfunction has been identified as a pathobiological therapeutic target to quell cellular dyshomeostasis. Further, therapeutic approaches targeting mitochondrial impairments, such as mild mitochondrial uncoupling, have been shown to alleviate behavioral alterations after TBI. To examine how mild mitochondrial uncoupling modulates acute mitochondrial outcomes in a military-relevant model of mTBI, we utilized repeated blast overpressure of 11 psi peak overpressure to model repeated mild blast traumatic brain injury (rmbTBI) in rats followed by assessment of mitochondrial respiration and mitochondrial-related oxidative damage at 2 days post-rmbTBI. Treatment groups were administered 8 or 80 mg/kg MP201, a prodrug of 2,4 dinitrophenol (DNP) that displays improved pharmacokinetics compared with its metabolized form. Synaptic and glia-enriched mitochondria were isolated using fractionated a mitochondrial magnetic separation technique. There was a consistent physiological response, decreased heart rate, following mbTBI among experimental groups. Although there was a lack of injury effect in mitochondrial respiration of glia-enriched mitochondria, there were impairments in mitochondrial respiration in synaptic mitochondria isolated from the prefrontal cortex (PFC) and the amygdala/entorhinal/piriform cortex (AEP) region. Impairments in synaptic mitochondrial respiration were rescued by oral 80 mg/kg MP201 treatment after rmbTBI, which may be facilitated by increases in complex II and complex IV activity. Mitochondrial oxidative damage in glia-enriched mitochondria was increased in the PFC and hippocampus after rmbTBI. MP201 treatment alleviated elevated glia-enriched mitochondrial oxidative damage following rmbTBI. However, there was a lack of injury-associated differences in oxidative damage in synaptic mitochondria. Overall, our report demonstrates that rmbTBI results in mitochondrial impairment diffusely throughout the brain and mild mitochondrial uncoupling can restore mitochondrial bioenergetics and oxidative balance.


Subject(s)
Blast Injuries , Brain Concussion , Brain Injuries, Traumatic , Prodrugs , Rats , Animals , Prodrugs/pharmacology , Mitochondria , Brain , Oxidative Stress
3.
Cells ; 12(10)2023 05 22.
Article in English | MEDLINE | ID: mdl-37408279

ABSTRACT

The brain undergoes oxidative stress and mitochondrial dysfunction following physiological insults such as Traumatic brain injury (TBI), ischemia-reperfusion, and stroke. Pharmacotherapeutics targeting mitochondria (mitoceuticals) against oxidative stress include antioxidants, mild uncouplers, and enhancers of mitochondrial biogenesis, which have been shown to improve pathophysiological outcomes after TBI. However, to date, there is no effective treatment for TBI. Studies have suggested that the deletion of LDL receptor-related protein 1 (LRP1) in adult neurons or glial cells could be beneficial and promote neuronal health. In this study, we used WT and LRP1 knockout (LKO) mouse embryonic fibroblast cells to examine mitochondrial outcomes following exogenous oxidative stress. Furthermore, we developed a novel technique to measure mitochondrial morphometric dynamics using transgenic mitochondrial reporter mice mtD2g (mitochondrial-specific Dendra2 green) in a TBI model. We found that oxidative stress increased the quantity of fragmented and spherical-shaped mitochondria in the injury core of the ipsilateral cortex following TBI, whereas rod-like elongated mitochondria were seen in the corresponding contralateral cortex. Critically, LRP1 deficiency significantly decreased mitochondrial fragmentation, preserving mitochondrial function and cell growth following exogenous oxidative stress. Collectively, our results show that targeting LRP1 to improve mitochondrial function is a potential pharmacotherapeutic strategy against oxidative damage in TBI and other neurodegenerative diseases.


Subject(s)
Brain Injuries, Traumatic , Fibroblasts , Low Density Lipoprotein Receptor-Related Protein-1 , Oxidative Stress , Animals , Mice , Brain/metabolism , Brain Injuries, Traumatic/metabolism , Fibroblasts/metabolism , Mice, Transgenic , Mitochondria/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/genetics
4.
Cell Mol Neurobiol ; 43(1): 423-429, 2023 Jan.
Article in English | MEDLINE | ID: mdl-34761332

ABSTRACT

Sex is a key biological variable in traumatic brain injury (TBI) and plays a significant role in neuroinflammatory responses. However, the molecular mechanisms contributing to this sexually dimorphic neuroinflammatory response remain elusive. Here we describe a significant and previously unreported tissue enrichment and sex-specific alteration of a set of inflammatory microRNAs (miRNAs) in CD11b+ cells of brain and bone marrow isolated from naïve mice as well as mice subjected to TBI. Our data from naïve mice demonstrated that expression levels of miR-146a-5p and miR-150-5p were relatively higher in brain CD11b+ cells, and that miR-155-5p and miR-223-3p were highly enriched in bone marrow CD11b+ cells. Furthermore, while miR-150-5p and miR-155-5p levels were higher in male brain CD11b+ cells, no significant sexual difference was observed for miR-146a-5p and miR-223-3p. However, TBI resulted in sex-specific differential responses of these miRNAs in brain CD11b+ cells. Specifically, miR-223-3p levels in brain CD11b+ cells were markedly elevated in both sexes in response to TBI at 3 and 24 h, with levels in females being significantly higher than males at 24 h. We then focused on analyzing several miR-223-3p targets and inflammation-related marker genes following injury. Corresponding to the greater elevation of miR-223-3p in females, the miR-223-3p targets, TRAF6 and FBXW7 were significantly reduced in females compared to males. Interestingly, anti-inflammatory genes ARG1 and IL4 were higher in females after TBI than in males. These observations suggest miR-223-3p and other inflammatory responsive miRNAs may play a key role in sex-specific neuroinflammatory response following TBI.


Subject(s)
Brain Injuries, Traumatic , MicroRNAs , Animals , Female , Male , Mice , Bone Marrow/metabolism , Brain/metabolism , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/metabolism , Inflammation/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism
5.
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
6.
Neural Regen Res ; 16(3): 514-522, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32985480

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNA molecules that regulate post-transcriptional gene expression and contribute to all aspects of cellular function. We previously reported that the activities of several mitochondria-enriched miRNAs regulating inflammation (i.e., miR-142-3p, miR-142-5p, and miR-146a) are altered in the hippocampus at 3-12 hours following a severe traumatic brain injury. In the present study, we investigated the temporal expression profile of these inflammatory miRNAs in mitochondria and cytosol fractions at more chronic post-injury times following severe controlled cortical impact injury in rats. In addition, several inflammatory genes were analyzed in the cytosol fractions. The analysis showed that while elevated levels were observed in cytoplasm, the mitochondria-enriched miRNAs, miR-142-3p and miR-142-5p continued to be significantly reduced in mitochondria from injured hippocampi for at least 3 days and returned to near normal levels at 7 days post-injury. Although not statistically significant, miR-146a also remained at reduced levels for up to 3 days following controlled cortical impact injury, and recovered by 7 days. In contrast, miRNAs that are not enriched in mitochondria, including miR-124a, miR-150, miR-19b, miR-155, and miR-223 were either increased or demonstrated no change in their levels in mitochondrial fractions for 7 days. The one exception was that miR-223 levels were reduced in mitochondria at 1 day following injury. No major alterations were observed in sham operated animals. This temporal pattern was unique to mitochondria-enriched miRNAs and correlated with injury-induced changes in mitochondrial bioenergetics as well as expression levels of several inflammatory markers. These observations suggested a potential compartmental re-distribution of the mitochondria-enriched inflammatory miRNAs and may reflect an intracellular mechanism by which specific miRNAs regulate injury-induced inflammatory signaling. To test this, we utilized a novel peptide-based nanoparticle strategy for in vitro and in vivo delivery of a miR-146a mimic as a potential therapeutic strategy for targeting nuclear factor-kappaB inflammatory modulators in the injured brain. Nanoparticle delivery of miR-146a to BV-2 or SH-SY5Y cells significantly reduced expression of TNF receptor-associated factor 6 (TRAF6) and interleukin-1 receptor-associated kinase 1 (IRAK1), two important modulators of the nuclear factor-kappaB (NF-κB) pro-inflammatory pathway. Moreover, injections of miR-146a containing nanoparticles into the brain immediately following controlled cortical impact injury significantly reduced hippocampal TNF receptor-associated factor 6 and interleukin-1 receptor-associated kinase 1 levels. Taken together, our studies demonstrate the subcellular alteration of inflammatory miRNAs after traumatic brain injury and establish proof of principle that nanoparticle delivery of miR-146a has therapeutic potential for modulating pro-inflammatory effectors in the injured brain. All of the studies performed were approved by the University of Kentucky Institutional Animal Care and Usage Committee (IACUC protocol # 2014-1300) on August 17, 2017.

7.
Cell Signal ; 76: 109777, 2020 12.
Article in English | MEDLINE | ID: mdl-32918979

ABSTRACT

Emerging evidence suggests that ubiquitin mediated post translational modification is a critical regulatory process involved in diverse cellular pathways including cell death. During ubiquitination, E3 ligases recognize target proteins and determine the topology of ubiquitin chains. Recruitment of E3 ligases to targets proteins under stress conditions including oxidative stress and their implication in cell death have not been systemically explored. In the present study, we characterized the role of TRIM32 as an E3 ligase in regulation of oxidative stress induced cell death. TRIM32 is ubiquitously expressed in cell lines of different origin and form cytoplasmic speckle like structures that transiently interact with mitochondria under oxidative stress conditions. The ectopic expression of TRIM32 sensitizes cell death induced by oxidative stress whereas TRIM32 knockdown shows a protective effect. The turnover of TRIM32 is enhanced during oxidative stress and its expression induces ROS generation, loss of mitochondrial transmembrane potential and decrease in complex-I activity. The pro-apoptotic effect was rescued by pan-caspase inhibitor or antioxidant treatment. E3 ligase activity of TRIM32 is essential for oxidative stress induced apoptotic cell death. Furthermore, TRIM32 decreases X-linked inhibitor of apoptosis (XIAP) level and overexpression of XIAP rescued cells from TRIM32 mediated oxidative stress and cell death. Overall, the results of this study provide the first evidence supporting the role of TRIM32 in regulating oxidative stress induced cell death, which has implications in numerous pathological conditions including cancer and neurodegeneration.


Subject(s)
Cell Death , Mitochondria/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Transcription Factors/physiology , Tripartite Motif Proteins/physiology , Ubiquitin-Protein Ligases/physiology , X-Linked Inhibitor of Apoptosis Protein/metabolism , HEK293 Cells , Humans , Membrane Potential, Mitochondrial
8.
Mol Neurobiol ; 57(7): 2996-3013, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32451872

ABSTRACT

The mitochondria-associated endoplasmic reticulum (ER) membranes (MAMs) are specific ER domains that contact the mitochondria and function to facilitate communication between ER and mitochondria. Disruption of contact between the mitochondria and ER is associated with a variety of pathophysiological conditions including neurodegenerative diseases. Considering the many cellular functions of MAMs, we hypothesized that MAMs play an important role in regulating microRNA (miRNA) activity linked to its unique location between mitochondria and ER. Here we present new findings from human and rat brains indicating that the MAMs are subcellular sites enriched for specific miRNAs. We employed subcellular fractionation and TaqMan® RT-qPCR miRNA analysis to quantify miRNA levels in subcellular fractions isolated from male rat brains and six human brain samples. We found that MAMs contain a substantial number of miRNAs and the profile differs significantly from that of cytosolic, mitochondria, or ER. Interestingly, MAMs are particularly enriched in inflammatory-responsive miRNAs, including miR-146a, miR-142-3p, and miR-142-5p in both human and rat brains; miR-223 MAM enrichment was observed only in human brain samples. Further, mitochondrial uncoupling or traumatic brain injury in male rats resulted in the alteration of inflammatory miRNA enrichment in the isolated subcellular fractions. These observations demonstrate that miRNAs are distributed differentially in organelles and may re-distribute between organelles and the cytosol in response to cellular stress and metabolic demands.


Subject(s)
Brain/metabolism , Endoplasmic Reticulum/metabolism , Inflammation/metabolism , Intracellular Membranes/metabolism , MicroRNAs/metabolism , Mitochondria/metabolism , Aged , Aged, 80 and over , Animals , Cognitive Dysfunction/metabolism , Cytosol/metabolism , Dementia/metabolism , Female , Humans , Male , Rats , Rats, Sprague-Dawley , Subcellular Fractions/metabolism
9.
Methods Mol Biol ; 2063: 139-154, 2020.
Article in English | MEDLINE | ID: mdl-31667768

ABSTRACT

Eukaryotic cell organelles exert unique functions individually but also interact with each other for essential cellular functions. This physical interface between the organelles serves as an important platform for biomolecule trafficking and signaling. Mitochondria are membrane-bound organelles and form a dynamic contact with other organelles. The interactions and communication between mitochondria and endoplasmic reticulum (ER) are facilitated by an ER specific domain, named mitochondria associated ER membrane (MAM). Due to its unique location, the MAM is a "hotspot" for important cell signaling and biochemical processes including calcium homeostasis, lipid synthesis/exchange, inflammasome and autophagosome formation, and mitochondria fission/fusion. Although techniques are available for isolation of organelle fractions including MAM, most utilize animal tissues and cell lines. Here we describe a protocol that is tailored to the isolation of highly purified MAM, mitochondria, ER, and cytosol from human brain. In addition, we include a protocol for the isolation of total RNA and subsequent analysis of microRNAs from these highly purified organelle fractions. Finally, we include a panel of protein markers that are useful for validating the enrichment and purity of each subcellular fraction.


Subject(s)
Brain/pathology , Endoplasmic Reticulum/metabolism , MicroRNAs/isolation & purification , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Subcellular Fractions/metabolism , Animals , Cytosol , Humans , MicroRNAs/genetics , Mitochondria/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Polymerase Chain Reaction , Rats , Signal Transduction
10.
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
11.
Sci Rep ; 9(1): 9656, 2019 07 04.
Article in English | MEDLINE | ID: mdl-31273236

ABSTRACT

While mitochondria maintain essential cellular functions, such as energy production, calcium homeostasis, and regulating programmed cellular death, they also play a major role in pathophysiology of many neurological disorders. Furthermore, several neurodegenerative diseases are closely linked with synaptic damage and synaptic mitochondrial dysfunction. Unfortunately, the ability to assess mitochondrial dysfunction and the efficacy of mitochondrial-targeted therapies in experimental models of neurodegenerative disease and CNS injury is limited by current mitochondrial isolation techniques. Density gradient ultracentrifugation (UC) is currently the only technique that can separate synaptic and non-synaptic mitochondrial sub-populations, though small brain regions cannot be assayed due to low mitochondrial yield. To address this limitation, we used fractionated mitochondrial magnetic separation (FMMS), employing magnetic anti-Tom22 antibodies, to develop a novel strategy for isolation of functional synaptic and non-synaptic mitochondria from mouse cortex and hippocampus without the usage of UC. We compared the yield and functionality of mitochondria derived using FMMS to those derived by UC. FMMS produced 3x more synaptic mitochondrial protein yield compared to UC from the same amount of tissue, a mouse hippocampus. FMMS also has increased sensitivity, compared to UC separation, to measure decreased mitochondrial respiration, demonstrated in a paradigm of mild closed head injury. Taken together, FMMS enables improved brain-derived mitochondrial yield for mitochondrial assessments and better detection of mitochondrial impairment in CNS injury and neurodegenerative disease.


Subject(s)
Brain Injuries, Traumatic/physiopathology , Brain/physiology , Cell Fractionation/methods , Magnets , Mitochondria/metabolism , Synapses/physiology , Animals , Male , Mice , Mice, Inbred C57BL , Synaptic Transmission
12.
Int J Inj Contr Saf Promot ; 26(4): 336-342, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31184258

ABSTRACT

Injury has been increasing exponentially, especially in children, and it has become a public health concern. The present study was conducted with the objective of determining the prevalence and profile of unintentional injury among children of 1-5-year age group in a rural community of India and also to find out the predictors. Primary caregiver was interviewed by using a structured interview schedule. Parent supervisory behaviour was assessed using Parent Supervision Attributes Profile Questionnaire (PSAPQ), and child injury risk-taking behaviour was assessed by using injury behaviour checklist (IBC). Children encountered any unintentional injury event during last three-month period were 261 (62.7%). PSAPQ score was significantly higher in those parents where children had no episode of injury compared to others. Among four components of the PSAPQ, protectiveness (p = 0.049) and risk tolerance (p = 0.001) score had significant positive association with the incidence of unintentional injury. Binary multivariable logistic regression technique had found that age of the child, gender, primary care giver, birth order of the baby, the number of siblings, social class and IBC score has significant association with history of unintentional injury. There is utmost need for the development of effective programmes and training strategies to prevent unintentional injury among under-five children in future.


Subject(s)
Developing Countries/statistics & numerical data , Parenting , Rural Population/statistics & numerical data , Wounds and Injuries/epidemiology , Adult , Age Factors , Birth Order , Child, Preschool , Cross-Sectional Studies , Family Characteristics , Female , Humans , Incidence , India/epidemiology , Infant , Male , Prevalence , Risk Factors , Risk-Taking , Sex Factors , Social Class , Surveys and Questionnaires
13.
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
14.
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
15.
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
16.
Cell Signal ; 48: 1-12, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29678622

ABSTRACT

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.


Subject(s)
Autophagy , Carrier Proteins/physiology , Caspase 3/metabolism , DNA Damage , Nerve Tissue Proteins/physiology , Cell Survival , HEK293 Cells , HeLa Cells , Humans , Lysosomes/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism
17.
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
18.
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
19.
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
20.
Free Radic Biol Med ; 89: 1036-48, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26524401

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis/drug effects , Hydrogen Peroxide/pharmacology , Membrane Proteins/metabolism , Mitochondria/metabolism , Oxidants/pharmacology , Oxidative Stress/drug effects , Adaptor Proteins, Signal Transducing/genetics , Blotting, Western , Cell Proliferation/drug effects , Cytochromes c/metabolism , Fluorescent Antibody Technique , HEK293 Cells , Humans , Immunoprecipitation , Membrane Potential, Mitochondrial/drug effects , Membrane Proteins/genetics , Mitochondria/drug effects , Mitochondria/pathology , Proteomics , RNA, Messenger/genetics , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Ubiquitin/metabolism , Ubiquitination/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...