Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Aging Cell ; 20(4): e13329, 2021 04.
Article in English | MEDLINE | ID: mdl-33734555

ABSTRACT

Senescence phenotypes and mitochondrial dysfunction are implicated in aging and in premature aging diseases, including ataxia telangiectasia (A-T). Loss of mitochondrial function can drive age-related decline in the brain, but little is known about whether improving mitochondrial homeostasis alleviates senescence phenotypes. We demonstrate here that mitochondrial dysfunction and cellular senescence with a senescence-associated secretory phenotype (SASP) occur in A-T patient fibroblasts, and in ATM-deficient cells and mice. Senescence is mediated by stimulator of interferon genes (STING) and involves ectopic cytoplasmic DNA. We further show that boosting intracellular NAD+ levels with nicotinamide riboside (NR) prevents senescence and SASP by promoting mitophagy in a PINK1-dependent manner. NR treatment also prevents neurodegeneration, suppresses senescence and neuroinflammation, and improves motor function in Atm-/- mice. Our findings suggest a central role for mitochondrial dysfunction-induced senescence in A-T pathogenesis, and that enhancing mitophagy as a potential therapeutic intervention.


Subject(s)
Ataxia Telangiectasia/diet therapy , Ataxia Telangiectasia/metabolism , Dietary Supplements , Membrane Proteins/metabolism , Mitophagy/drug effects , NAD/metabolism , Niacinamide/analogs & derivatives , Pyridinium Compounds/administration & dosage , Senescence-Associated Secretory Phenotype/genetics , Signal Transduction/drug effects , Animals , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Case-Control Studies , Cell Line, Tumor , Disease Models, Animal , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , Mitochondria/metabolism , Mitophagy/genetics , Neurons/drug effects , Neurons/metabolism , Niacinamide/administration & dosage , Rats , Rats, Sprague-Dawley , Signal Transduction/genetics , Transfection , Treatment Outcome
2.
Nucleic Acids Res ; 48(12): 6611-6623, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32453416

ABSTRACT

Mitochondria are vital for cellular energy supply and intracellular signaling after stress. Here, we aimed to investigate how mitochondria respond to acute DNA damage with respect to mitophagy, which is an important mitochondrial quality control process. Our results show that mitophagy increases after DNA damage in primary fibroblasts, murine neurons and Caenorhabditis elegans neurons. Our results indicate that modulation of mitophagy after DNA damage is independent of the type of DNA damage stimuli used and that the protein Spata18 is an important player in this process. Knockdown of Spata18 suppresses mitophagy, disturbs mitochondrial Ca2+ homeostasis, affects ATP production, and attenuates DNA repair. Importantly, mitophagy after DNA damage is a vital cellular response to maintain mitochondrial functions and DNA repair.


Subject(s)
Calcium/metabolism , Mitochondrial Proteins/genetics , Mitophagy/genetics , Neurons/metabolism , Animals , Caenorhabditis elegans/genetics , Cell Line , Cell Proliferation/genetics , DNA Damage/genetics , DNA Repair/genetics , Fibroblasts/metabolism , Humans , Mice , Mitochondria/genetics
3.
Acta Neuropathol ; 140(1): 25-47, 2020 07.
Article in English | MEDLINE | ID: mdl-32333098

ABSTRACT

Alzheimer's disease (AD) is an incurable neurodegenerative disease that is more prevalent in women. The increased risk of AD in women is not well understood. It is well established that there are sex differences in metabolism and that metabolic alterations are an early component of AD. We utilized a cross-species approach to evaluate conserved metabolic alterations in the serum and brain of human AD subjects, two AD mouse models, a human cell line, and two Caenorhabditis elegans AD strains. We found a mitochondrial complex I-specific impairment in cortical synaptic brain mitochondria in female, but not male, AD mice. In the hippocampus, Polß haploinsufficiency caused synaptic complex I impairment in male and female mice, demonstrating the critical role of DNA repair in mitochondrial function. In non-synaptic, glial-enriched, mitochondria from the cortex and hippocampus, complex II-dependent respiration increased in female, but not male, AD mice. These results suggested a glial upregulation of fatty acid metabolism to compensate for neuronal glucose hypometabolism in AD. Using an unbiased metabolomics approach, we consistently observed evidence of systemic and brain metabolic remodeling with a shift from glucose to lipid metabolism in humans with AD, and in AD mice. We determined that this metabolic shift is necessary for cellular and organismal survival in C. elegans, and human cell culture AD models. We observed sex-specific, systemic, and brain metabolic alterations in humans with AD, and that these metabolite changes significantly correlate with amyloid and tau pathology. Among the most significant metabolite changes was the accumulation of glucose-6-phosphate in AD, an inhibitor of hexokinase and rate-limiting metabolite for the pentose phosphate pathway (PPP). Overall, we identified novel mechanisms of glycolysis inhibition, PPP, and tricarboxylic acid cycle impairment, and a neuroprotective augmentation of lipid metabolism in AD. These findings support a sex-targeted metabolism-modifying strategy to prevent and treat AD.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , DNA Repair-Deficiency Disorders/metabolism , Mitochondria/metabolism , Sex Characteristics , Alzheimer Disease/pathology , Animals , Brain/pathology , Caenorhabditis elegans , DNA Repair-Deficiency Disorders/pathology , Energy Metabolism/physiology , Female , Glucose/metabolism , Humans , Lipid Metabolism/physiology , Male , Mice , Mitochondria/pathology
4.
Mech Ageing Dev ; 186: 111207, 2020 03.
Article in English | MEDLINE | ID: mdl-31923475

ABSTRACT

Aging is associated with multiple human pathologies. In the past few years mitochondrial homeostasis has been well correlated with age-related disorders and longevity. Mitochondrial homeostasis involves generation, biogenesis and removal of dysfunctional mitochondria via mitophagy. Mitophagy is regulated by various mitochondrial and extra-mitochondrial factors including morphology, oxidative stress and DNA damage. For decades, DNA damage and inefficient DNA repair have been considered as major determinants for age-related disorders. Although defects in DNA damage recognition and repair and mitophagy are well documented to be major factors in age-associated diseases, interactivity between these is poorly understood. Mitophagy efficiency decreases with age leading to accumulation of dysfunctional mitochondria enhancing the severity of age-related disorders including neurodegenerative diseases, inflammatory diseases, cancer, diabetes and many more. Therefore, mitophagy is being targeted for intervention in age-associated disorders. NAD+ supplementation has emerged as one intervention to target both defective DNA repair and mitophagy. In this review, we discuss the molecular signaling pathways involved in regulation of DNA damage and repair and of mitophagy, and we highlight the opportunities for clinical interventions targeting these processes to improve the quality of life during aging.


Subject(s)
Aging/physiology , DNA Damage , Mitophagy/physiology , Aging/genetics , DNA Repair , Humans , Signal Transduction
5.
Nat Rev Neurol ; 15(10): 565-581, 2019 10.
Article in English | MEDLINE | ID: mdl-31501588

ABSTRACT

Ageing is the primary risk factor for most neurodegenerative diseases, including Alzheimer disease (AD) and Parkinson disease (PD). One in ten individuals aged ≥65 years has AD and its prevalence continues to increase with increasing age. Few or no effective treatments are available for ageing-related neurodegenerative diseases, which tend to progress in an irreversible manner and are associated with large socioeconomic and personal costs. This Review discusses the pathogenesis of AD, PD and other neurodegenerative diseases, and describes their associations with the nine biological hallmarks of ageing: genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, mitochondrial dysfunction, cellular senescence, deregulated nutrient sensing, stem cell exhaustion and altered intercellular communication. The central biological mechanisms of ageing and their potential as targets of novel therapies for neurodegenerative diseases are also discussed, with potential therapies including NAD+ precursors, mitophagy inducers and inhibitors of cellular senescence.


Subject(s)
Aging/genetics , Aging/metabolism , Brain/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Aging/pathology , Brain/pathology , DNA Damage/physiology , Epigenesis, Genetic/physiology , Humans , Mitophagy/physiology , Neurodegenerative Diseases/pathology , Risk Factors
6.
Nucleic Acids Res ; 47(16): 8548-8562, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31276581

ABSTRACT

Cockayne syndrome is an accelerated aging disorder, caused by mutations in the CSA or CSB genes. In CSB-deficient cells, poly (ADP ribose) polymerase (PARP) is persistently activated by unrepaired DNA damage and consumes and depletes cellular nicotinamide adenine dinucleotide, which leads to mitochondrial dysfunction. Here, the distribution of poly (ADP ribose) (PAR) was determined in CSB-deficient cells using ADPr-ChAP (ADP ribose-chromatin affinity purification), and the results show striking enrichment of PAR at transcription start sites, depletion of heterochromatin and downregulation of H3K9me3-specific methyltransferases SUV39H1 and SETDB1. Induced-expression of SETDB1 in CSB-deficient cells downregulated PAR and normalized mitochondrial function. The results suggest that defects in CSB are strongly associated with loss of heterochromatin, downregulation of SETDB1, increased PAR in highly-transcribed regions, and mitochondrial dysfunction.


Subject(s)
Cellular Senescence/genetics , Cockayne Syndrome/genetics , DNA Helicases/genetics , DNA Repair Enzymes/genetics , Histones/genetics , Mitochondria/metabolism , Poly-ADP-Ribose Binding Proteins/genetics , Protein Methyltransferases/genetics , Transcription Factors/genetics , Cell Line, Transformed , Chromatin/chemistry , Chromatin/metabolism , Cockayne Syndrome/metabolism , Cockayne Syndrome/pathology , DNA/genetics , DNA/metabolism , DNA Damage , DNA Helicases/metabolism , DNA Repair Enzymes/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation , Histone-Lysine N-Methyltransferase , Histones/metabolism , Humans , Methyltransferases/genetics , Methyltransferases/metabolism , Mitochondria/pathology , Mutation , NAD/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , Protein Methyltransferases/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Transcription Initiation Site , Transcription, Genetic
7.
J Exp Clin Cancer Res ; 38(1): 271, 2019 Jun 20.
Article in English | MEDLINE | ID: mdl-31221176

ABSTRACT

BACKGROUND: Recently, we have reported the characterization of a novel protein named Coiled-coil Helix Tumor and Metabolism 1 (CHTM1). CHTM1 localizes to both cytosol and mitochondria. Sequence corresponding to CHTM1 is also annotated in the database as CHCHD5. CHTM1 is deregulated in human breast and colon cancers and its deficiency in human cancer cells leads to defective lipid metabolism and poor growth under glucose/glutamine starvation. METHODS: Human cancer cell lines and tissue specimens were used. CHTM1 knockdown was done via lentiviral approach. CHTM1-expresssion constructs were developed and mutants were generated via site-directed mutagenesis approach. Western blotting, immunostaining, immunohistochemistry, cell fractionation and luciferase assays were performed. Reactive oxygen species and reactive nitrogen species were also measured. RESULTS: Here we report that CHTM1 deficiency sensitizes human lung cancer cells to metabolic stress-induced cell death mediated by glucose/glutamine deprivation and metformin treatment. CHTM1 interacts with Apoptosis Inducing Factor 1 (AIF1) that is one of the important death inducing molecules. CHTM1 appears to negatively regulate AIF1 by preventing AIF1 translocation to cytosol/nucleus and thereby inhibit AIF1-mediated caspase-independent cell death. Our results also indicate that p38, a stress kinase, plays a critical role in metabolic stress-induced cell death in CHTM1-deficient cells. Furthermore, p38 appears to enhance AIF1 translocation from mitochondria to cytosol particularly in metabolically stressed CHTM1-deficient cells and CHTM1 negatively regulates p38 kinase activity. The expression status of CHTM1 in lung cancer patient samples is also investigated and our results indicate that CHTM1 levels are increased in the majority of lung tumors when compared to their matching normal tissues. CONCLUSION: Thus, CHTM1 appears to be an important metabolic marker that regulates cancer cell survival under metabolic stress conditions, and has the potential to be developed as a predictive tumor marker.


Subject(s)
Apoptosis Inducing Factor/metabolism , Lung Neoplasms/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Stress, Physiological , p38 Mitogen-Activated Protein Kinases/metabolism , A549 Cells , Cell Line, Tumor , Cell Survival , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HeLa Cells , Humans , Lipid Metabolism , Lung Neoplasms/genetics , MCF-7 Cells , Metformin/pharmacology , Protein Transport , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Up-Regulation
8.
Free Radic Biol Med ; 141: 47-58, 2019 09.
Article in English | MEDLINE | ID: mdl-31175982

ABSTRACT

Cellular exposure to ionizing radiation leads to oxidatively generated DNA damage, which has been implicated in neurodegenerative diseases. DNA damage is repaired by the evolutionarily conserved base excision repair (BER) system. Exposure of mice to ionizing radiation affects neurogenesis and neuroinflammation. However, the consequences of deficient DNA repair on adult neurogenesis and neuroinflammation are poorly understood despite their potential relevance for homeostasis. We previously reported that loss of NEIL1, an important DNA glycosylase involved in BER, is associated with deficiencies in spatial memory, olfaction, and protection against ischemic stroke in mice. Here, we show that Neil1-/- mice display an anxiety-mediated behavior in the open field test, a deficient recognitive memory in novel object recognition and increased neuroinflammatory response under basal conditions. Further, mice lacking NEIL1 have decreased neurogenesis and deficient resolution of neuroinflammation following gamma irradiation (IR)-induced stress compared to WT mice. Neil1-/- IR-exposed mice also exhibit increased DNA damage and apoptosis in the hippocampus. Interestingly, behavioral tests two weeks after IR showed impaired stress response in the Neil1-/- mice. Our data indicate that NEIL1 plays an important role in adult neurogenesis and in the resolution of neuroinflammation.


Subject(s)
Behavior, Animal , DNA Glycosylases/metabolism , Inflammation , Neurodegenerative Diseases/metabolism , Stress, Psychological/metabolism , Aging , Animals , Apoptosis , Cell Proliferation , Central Nervous System/metabolism , DNA Damage , DNA Glycosylases/genetics , DNA Repair , Fear , Gamma Rays , Gene Expression Profiling , Hippocampus/metabolism , Male , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/cytology , Oligonucleotide Array Sequence Analysis , RNA/analysis
9.
Oncogene ; 37(15): 2052-2066, 2018 04.
Article in English | MEDLINE | ID: mdl-29371680

ABSTRACT

A better understanding of the link between cellular metabolism and tumorigenesis is needed. Here, we report characterization of a novel protein named coiled-coil helix tumor and metabolism 1 (CHTM1). We have found that CHTM1 is associated with cancer and cellular metabolism. CHTM1 localizes to mitochondria and cytosol, and its deficiency in cancer cells results in decreased mitochondrial oxygen consumption and ATP levels as well as oxidative stress indicating mitochondrial dysfunction. CHTM1-deficient cancer cells display poor growth under glucose/glutamine-deprived conditions, whereas cells expressing increased levels of exogenous CHTM1 exhibit enhanced proliferation and survival under similar conditions. CHTM1 deficiency also leads to defects in lipid metabolism resulting in fatty acid accumulation, which explains poor growth of CHTM1-deficient cells under glucose/glutamine deprivation since nutrient deprivation increases dependency on lipids for energy generation. We also demonstrate that CHTM1 mediates its effect via the PKC, CREB, and PGC-1alpha signaling axis, and cytosolic accumulation of CHTM1 during nutrient deprivation appears to be important for its effect on cellular signaling events. Furthermore, analyses of tissue specimens from 71 breast and 97 colon cancer patients show CHTM1 expression to be upregulated in the majority of tumor specimens representing these malignancies. Collectively, our findings are highly significant because CHTM1 is a novel metabolic marker that is important for the growth of tumorigenic cells under limiting nutrient supplies and thus, links cellular metabolism and tumorigenesis.


Subject(s)
Biomarkers, Tumor/physiology , Energy Metabolism/genetics , Membrane Proteins/physiology , Mitochondrial Proteins/physiology , Neoplasms/genetics , Neoplasms/metabolism , Base Sequence , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cytosol/metabolism , Glucose/deficiency , Glucose/metabolism , Glutamine/deficiency , Glutamine/metabolism , HEK293 Cells , Humans , Lipid Metabolism/genetics , MCF-7 Cells , Membrane Proteins/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Neoplasms/pathology , Nutrients , Signal Transduction/genetics , Tumor Cells, Cultured
10.
Mol Cell Pharmacol ; 5(3): 109-133, 2013.
Article in English | MEDLINE | ID: mdl-24533171

ABSTRACT

Mitochondrial morphology and metabolism play an important role in cellular homeostasis. Recent studies have shown that the fidelity of mitochondrial morphology is important in maintaining mitochondrial shape, number, size, membrane potential, ATP synthesis, mtDNA, motility, signaling, quality control, response to cellular stress, mitophagy and apoptosis. This article provides an overview of the current state of knowledge of the fission and fusion machinery with a focus on the mechanisms underlying the regulation of the mitochondrial morphology and cellular energy state. Several lines of evidence indicate that dysregulation of mitochondrial fission or fusion is associated with mitochondrial dysfunction, which in turn impacts mitophagy and apoptosis. Metabolic disorders are also associated with dysregulation of fission or fusion and the available lines of evidence point to a bidirectional interplay between the mitochondrial fission or fusion reactions and bioenergetics. Clearly, more in-depth studies are needed to fully elucidate the mechanisms that control mitochondrial fission and fusion. It is envisioned that the outcome of such studies will improve the understanding of the molecular basis of related metabolic disorders and also facilitate the development of better therapeutics.

SELECTION OF CITATIONS
SEARCH DETAIL
...