Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Cell ; 158(6): 1415-1430, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25215496

ABSTRACT

The manner by which genotype and environment affect complex phenotypes is one of the fundamental questions in biology. In this study, we quantified the transcriptome--a subset of the metabolome--and, using targeted proteomics, quantified a subset of the liver proteome from 40 strains of the BXD mouse genetic reference population on two diverse diets. We discovered dozens of transcript, protein, and metabolite QTLs, several of which linked to metabolic phenotypes. Most prominently, Dhtkd1 was identified as a primary regulator of 2-aminoadipate, explaining variance in fasted glucose and diabetes status in both mice and humans. These integrated molecular profiles also allowed further characterization of complex pathways, particularly the mitochondrial unfolded protein response (UPR(mt)). UPR(mt) shows strikingly variant responses at the transcript and protein level that are remarkably conserved among C. elegans, mice, and humans. Overall, these examples demonstrate the value of an integrated multilayered omics approach to characterize complex metabolic phenotypes.


Subject(s)
Gene Expression Profiling , Liver/chemistry , Mice/metabolism , Mitochondria/chemistry , Proteome/analysis , Serum/chemistry , Animals , Glucose/metabolism , Humans , Ketone Oxidoreductases/metabolism , Liver/cytology , Liver/metabolism , Mice/classification , Mice/genetics , Mice, Inbred C57BL , Mice, Inbred DBA , Mitochondria/metabolism , Quantitative Trait Loci , Serum/metabolism , Unfolded Protein Response
2.
Nat Rev Mol Cell Biol ; 17(4): 213-26, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26956194

ABSTRACT

Mitochondria participate in crucial cellular processes such as energy harvesting and intermediate metabolism. Although mitochondria possess their own genome--a vestige of their bacterial origins and endosymbiotic evolution--most mitochondrial proteins are encoded in the nucleus. The expression of the mitochondrial proteome hence requires tight coordination between the two genomes to adapt mitochondrial function to the ever-changing cellular milieu. In this Review, we focus on the pathways that coordinate the communication between mitochondria and the nucleus during homeostasis and mitochondrial stress. These pathways include nucleus-to-mitochondria (anterograde) and mitochondria-to-nucleus (retrograde) communication, mitonuclear feedback signalling and proteostasis regulation, the integrated stress response and non-cell-autonomous communication. We discuss how mitonuclear communication safeguards cellular and organismal fitness and regulates lifespan.


Subject(s)
Cell Nucleus/physiology , Homeostasis , Mitochondria/physiology , Stress, Physiological , Animals , Cell Communication , Mitochondrial Proteins/metabolism , Models, Biological , Signal Transduction
3.
Cell ; 154(2): 430-41, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-23870130

ABSTRACT

NAD(+) is an important cofactor regulating metabolic homeostasis and a rate-limiting substrate for sirtuin deacylases. We show that NAD(+) levels are reduced in aged mice and Caenorhabditis elegans and that decreasing NAD(+) levels results in a further reduction in worm lifespan. Conversely, genetic or pharmacological restoration of NAD(+) prevents age-associated metabolic decline and promotes longevity in worms. These effects are dependent upon the protein deacetylase sir-2.1 and involve the induction of mitonuclear protein imbalance as well as activation of stress signaling via the mitochondrial unfolded protein response (UPR(mt)) and the nuclear translocation and activation of FOXO transcription factor DAF-16. Our data suggest that augmenting mitochondrial stress signaling through the modulation of NAD(+) levels may be a target to improve mitochondrial function and prevent or treat age-associated decline.


Subject(s)
Forkhead Transcription Factors/metabolism , Longevity , Mitochondria/metabolism , NAD/metabolism , Signal Transduction , Unfolded Protein Response , Aging , Animals , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Hepatocytes/metabolism , Mice , Poly(ADP-ribose) Polymerase Inhibitors , Reactive Oxygen Species/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism , Sirtuins/genetics , Sirtuins/metabolism , Transcription Factors/metabolism
4.
Mol Cell ; 73(4): 775-787.e10, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30642763

ABSTRACT

Little information is available about how post-transcriptional mechanisms regulate the aging process. Here, we show that the RNA-binding protein Pumilio2 (PUM2), which is a translation repressor, is induced upon aging and acts as a negative regulator of lifespan and mitochondrial homeostasis. Multi-omics and cross-species analyses of PUM2 function show that it inhibits the translation of the mRNA encoding for the mitochondrial fission factor (Mff), thereby impairing mitochondrial fission and mitophagy. This mechanism is conserved in C. elegans by the PUM2 ortholog PUF-8. puf-8 knock-down in old nematodes and Pum2 CRISPR/Cas9-mediated knockout in the muscles of elderly mice enhances mitochondrial fission and mitophagy in both models, hence improving mitochondrial quality control and tissue homeostasis. Our data reveal how a PUM2-mediated layer of post-transcriptional regulation links altered Mff translation to mitochondrial dynamics and mitophagy, thereby mediating age-related mitochondrial dysfunctions.


Subject(s)
Aging/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Mitochondria/metabolism , Mitochondrial Dynamics , Mitophagy , RNA-Binding Proteins/metabolism , Age Factors , Aging/genetics , Aging/pathology , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Female , HEK293 Cells , HeLa Cells , Humans , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mitochondria/genetics , Mitochondria/pathology , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/pathology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , RNA-Binding Proteins/genetics , Signal Transduction , Up-Regulation
5.
Nature ; 563(7731): 354-359, 2018 11.
Article in English | MEDLINE | ID: mdl-30356218

ABSTRACT

Nicotinamide adenine dinucleotide (NAD+) is a co-substrate for several enzymes, including the sirtuin family of NAD+-dependent protein deacylases. Beneficial effects of increased NAD+ levels and sirtuin activation on mitochondrial homeostasis, organismal metabolism and lifespan have been established across species. Here we show that α-amino-ß-carboxymuconate-ε-semialdehyde decarboxylase (ACMSD), the enzyme that limits spontaneous cyclization of α-amino-ß-carboxymuconate-ε-semialdehyde in the de novo NAD+ synthesis pathway, controls cellular NAD+ levels via an evolutionarily conserved mechanism in Caenorhabditis elegans and mouse. Genetic and pharmacological inhibition of ACMSD boosts de novo NAD+ synthesis and sirtuin 1 activity, ultimately enhancing mitochondrial function. We also characterize two potent and selective inhibitors of ACMSD. Because expression of ACMSD is largely restricted to kidney and liver, these inhibitors may have therapeutic potential for protection of these tissues from injury. In summary, we identify ACMSD as a key modulator of cellular NAD+ levels, sirtuin activity and mitochondrial homeostasis in kidney and liver.


Subject(s)
Carboxy-Lyases/metabolism , Conserved Sequence , Evolution, Molecular , Health , Mitochondria/physiology , NAD/biosynthesis , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/enzymology , Caenorhabditis elegans/metabolism , Carboxy-Lyases/antagonists & inhibitors , Carboxy-Lyases/chemistry , Carboxy-Lyases/deficiency , Cell Line , Choline , Disease Models, Animal , Female , Gene Knockdown Techniques , Hepatocytes/cytology , Hepatocytes/drug effects , Homeostasis/drug effects , Humans , Kidney/cytology , Kidney/drug effects , Liver/cytology , Liver/drug effects , Longevity/drug effects , Male , Methionine/deficiency , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/physiopathology , Non-alcoholic Fatty Liver Disease/prevention & control , Rats , Sirtuins/metabolism
6.
Nephrol Dial Transplant ; 36(1): 60-68, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33099633

ABSTRACT

BACKGROUND: Nicotinamide adenine dinucleotide (NAD+) is a ubiquitous coenzyme involved in electron transport and a co-substrate for sirtuin function. NAD+ deficiency has been demonstrated in the context of acute kidney injury (AKI). METHODS: We studied the expression of key NAD+ biosynthesis enzymes in kidney biopsies from human allograft patients and patients with chronic kidney disease (CKD) at different stages. We used ischaemia-reperfusion injury (IRI) and cisplatin injection to model AKI, urinary tract obstruction [unilateral ureteral obstruction (UUO)] and tubulointerstitial fibrosis induced by proteinuria to investigate CKD in mice. We assessed the effect of nicotinamide riboside (NR) supplementation on AKI and CKD in animal models. RESULTS: RNA sequencing analysis of human kidney allograft biopsies during the reperfusion phase showed that the NAD+de novo synthesis is impaired in the immediate post-transplantation period, whereas the salvage pathway is stimulated. This decrease in de novo NAD+ synthesis was confirmed in two mouse models of IRI where NR supplementation prevented plasma urea and creatinine elevation and tubular injury. In human biopsies from CKD patients, the NAD+de novo synthesis pathway was impaired according to CKD stage, with better preservation of the salvage pathway. Similar alterations in gene expression were observed in mice with UUO or chronic proteinuric glomerular disease. NR supplementation did not prevent CKD progression, in contrast to its efficacy in AKI. CONCLUSION: Impairment of NAD+ synthesis is a hallmark of AKI and CKD. NR supplementation is beneficial in ischaemic AKI but not in CKD models.


Subject(s)
Acute Kidney Injury/pathology , Disease Models, Animal , Niacinamide/analogs & derivatives , Renal Insufficiency, Chronic/pathology , Reperfusion Injury/pathology , Acute Kidney Injury/chemically induced , Acute Kidney Injury/drug therapy , Acute Kidney Injury/metabolism , Animals , Antineoplastic Agents/toxicity , Cisplatin/toxicity , Disease Progression , Humans , Male , Mice , Mice, Inbred C57BL , Niacinamide/administration & dosage , Niacinamide/deficiency , Pyridinium Compounds , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/metabolism , Reperfusion Injury/chemically induced , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism
7.
Genes Dev ; 27(8): 819-35, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23630073

ABSTRACT

Epigenetic regulation of gene expression is strongly influenced by the accessibility of nucleosomal DNA or the state of chromatin compaction. In this context, coregulators, including both coactivators and corepressors, are pivotal intermediates that bridge chromatin-modifying enzymes and transcription factors. NCoR1 (nuclear receptor corepressor) and SMRT (silencing mediator of retinoic acid and thyroid hormone receptor) are among the best-characterized corepressors from a molecular point of view. These coregulators have conserved orthologs in lower organisms, which underscores their functional importance. Here we summarize the results from recent in vivo studies that reveal the wide-ranging roles of NCoR1 and SMRT in developmental as well as homeostatic processes, including metabolism, inflammation, and circadian rhythms. We also discuss the potential implications of NCoR1 and SMRT regulation of pathways ranging from genomic stability and carcinogenesis to metabolic diseases such as type 2 diabetes.


Subject(s)
Homeostasis/physiology , Nuclear Receptor Co-Repressor 1/metabolism , Nuclear Receptor Co-Repressor 2/metabolism , Animals , Gene Expression Regulation , Homeostasis/genetics , Humans , Nuclear Receptor Co-Repressor 1/genetics , Nuclear Receptor Co-Repressor 2/genetics
8.
Mamm Genome ; 25(9-10): 424-33, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24898297

ABSTRACT

Mitochondria, the main site of cellular energy harvesting, are derived from proteobacteria that evolved within our cells in endosymbiosis. Mitochondria retained vestiges of their proteobacterial genome, the circular mitochondrial DNA, which encodes 13 subunits of the oxidative phosphorylation multiprotein complexes in the electron transport chain (ETC), while the remaining ~80 ETC components are encoded in the nuclear DNA (nDNA). A further ~1,400 proteins, which are essential for mitochondrial function are also encoded in nDNA. Thus, a majority of mitochondrial proteins are translated in the cytoplasm, then imported, processed, and assembled in the mitochondria. An intricate protein quality control (PQC) network, constituted of chaperones and proteases that refold or degrade defective proteins, maintains mitochondrial proteostasis and ensures the cell and organism health. The mitochondrial unfolded protein response is a relatively recently discovered PQC pathway, which senses the proteostatic disturbances specifically in the mitochondria and resolves the stress by retrograde signaling to the nucleus and consequent transcriptional activation of protective genes. This PQC system does not only transiently resolve the local stress but also can have long-lasting effects on whole body metabolism, fitness, and longevity. A delicate tuning of its activation levels might constitute a treatment of various diseases, such as metabolic diseases, cancer, and neurodegenerative disorders.


Subject(s)
Mammals/physiology , Mitochondria/metabolism , Unfolded Protein Response , Animals , Cell Nucleus/metabolism , Gene Expression Regulation , Humans , Metabolic Diseases/genetics , Metabolic Diseases/metabolism , Mitochondria/genetics , Neoplasms/genetics , Neoplasms/metabolism , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Signal Transduction
9.
Cell Discov ; 9(1): 92, 2023 Sep 07.
Article in English | MEDLINE | ID: mdl-37679337

ABSTRACT

Lysosomes are central platforms for not only the degradation of macromolecules but also the integration of multiple signaling pathways. However, whether and how lysosomes mediate the mitochondrial stress response (MSR) remain largely unknown. Here, we demonstrate that lysosomal acidification via the vacuolar H+-ATPase (v-ATPase) is essential for the transcriptional activation of the mitochondrial unfolded protein response (UPRmt). Mitochondrial stress stimulates v-ATPase-mediated lysosomal activation of the mechanistic target of rapamycin complex 1 (mTORC1), which then directly phosphorylates the MSR transcription factor, activating transcription factor 4 (ATF4). Disruption of mTORC1-dependent ATF4 phosphorylation blocks the UPRmt, but not other similar stress responses, such as the UPRER. Finally, ATF4 phosphorylation downstream of the v-ATPase/mTORC1 signaling is indispensable for sustaining mitochondrial redox homeostasis and protecting cells from ROS-associated cell death upon mitochondrial stress. Thus, v-ATPase/mTORC1-mediated ATF4 phosphorylation via lysosomes links mitochondrial stress to UPRmt activation and mitochondrial function resilience.

10.
JHEP Rep ; 5(9): 100815, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37600955

ABSTRACT

Background & Aims: Non-alcoholic fatty liver disease (NAFLD) and steatohepatitis (NASH) have become the world's most common liver diseases, placing a growing strain on healthcare systems worldwide. Nonetheless, no effective pharmacological treatment has been approved. The naturally occurring compound cyclo histidine-proline (His-Pro) (CHP) is an interesting candidate for NAFLD management, given its safety profile and anti-inflammatory effects. Methods: Two different mouse models of liver disease were used to evaluate protective effects of CHP on disease progression towards fibrosis: a model of dietary NAFLD/NASH, achieved by thermoneutral housing (TN) in combination with feeding a western diet (WD), and liver fibrosis caused by repeated injections with carbon tetrachloride (CCl4). Results: Treatment with CHP limited overall lipid accumulation, lowered systemic inflammation, and prevented hyperglycaemia. Histopathology and liver transcriptomics highlighted reduced steatosis and demonstrated remarkable protection from the development of inflammation and fibrosis, features which herald the progression of NAFLD. We identified the extracellular signal-regulated kinase (ERK) pathway as an early mediator of the cellular response to CHP. Conclusions: CHP was active in both the preventive and therapeutic setting, reducing liver steatosis, fibrosis, and inflammation and improving several markers of liver disease. Impact and implications: Considering the incidence and the lack of approved treatments, it is urgent to identify new strategies that prevent and manage NAFLD. CHP was effective in attenuating NAFLD progression in two animal models of the disease. Overall, our work points to CHP as a novel and effective strategy for the management of NAFLD, fuelling optimism for potential clinical studies.

11.
Nat Aging ; 2(3): 199-213, 2022 03.
Article in English | MEDLINE | ID: mdl-37118378

ABSTRACT

Aging is typified by a progressive decline in mitochondrial activity and stress resilience. Here, we review how mitochondrial stress pathways have pleiotropic effects on cellular and systemic homeostasis, which can comprise protective or detrimental responses during aging. We describe recent evidence arguing that defects in these conserved adaptive pathways contribute to aging and age-related diseases. Signaling pathways regulating the mitochondrial unfolded protein response, mitochondrial membrane dynamics, and mitophagy are discussed, emphasizing how their failure contributes to heteroplasmy and de-regulation of key metabolites. Our current understanding of how these processes are controlled and interconnected explains how mitochondria can widely impact fundamental aspects of aging.


Subject(s)
Mitochondria , Mitophagy , Mitochondria/genetics , Mitochondrial Dynamics , Signal Transduction
12.
J Clin Invest ; 132(17)2022 09 01.
Article in English | MEDLINE | ID: mdl-35787521

ABSTRACT

Mitohormesis defines the increase in fitness mediated by adaptive responses to mild mitochondrial stress. Tetracyclines inhibit not only bacterial but also mitochondrial translation, thus imposing a low level of mitochondrial stress on eukaryotic cells. We demonstrate in cell and germ-free mouse models that tetracyclines induce a mild adaptive mitochondrial stress response (MSR), involving both the ATF4-mediated integrative stress response and type I interferon (IFN) signaling. To overcome the interferences of tetracyclines with the host microbiome, we identify tetracycline derivatives that have minimal antimicrobial activity, yet retain full capacity to induce the MSR, such as the lead compound, 9-tert-butyl doxycycline (9-TB). The MSR induced by doxycycline (Dox) and 9-TB improves survival and disease tolerance against lethal influenza virus (IFV) infection when given preventively. 9-TB, unlike Dox, did not affect the gut microbiome and also showed encouraging results against IFV when given in a therapeutic setting. Tolerance to IFV infection is associated with the induction of genes involved in lung epithelial cell and cilia function, and with downregulation of inflammatory and immune gene sets in lungs, liver, and kidneys. Mitohormesis induced by non-antimicrobial tetracyclines and the ensuing IFN response may dampen excessive inflammation and tissue damage during viral infections, opening innovative therapeutic avenues.


Subject(s)
Influenza, Human , Orthomyxoviridae Infections , Animals , Anti-Bacterial Agents , Doxycycline/pharmacology , Humans , Influenza, Human/drug therapy , Mice , Tetracycline , Tetracyclines/pharmacology
13.
Nat Aging ; 1(2): 165-178, 2021 02.
Article in English | MEDLINE | ID: mdl-33718883

ABSTRACT

Organisms respond to mitochondrial stress by activating multiple defense pathways including the mitochondrial unfolded protein response (UPRmt). However, how UPRmt regulators are orchestrated to transcriptionally activate stress responses remains largely unknown. Here we identified CBP-1, the worm ortholog of the mammalian acetyltransferases CBP/p300, as an essential regulator of the UPRmt, as well as mitochondrial stress-induced immune response, reduction of amyloid-ß aggregation and lifespan extension in Caenorhabditis elegans. Mechanistically, CBP-1 acts downstream of histone demethylases, JMJD-1.2/JMJD-3.1, and upstream of UPRmt transcription factors including ATFS-1, to systematically induce a broad spectrum of UPRmt genes and execute multiple beneficial functions. In mouse and human populations, transcript levels of CBP/p300 positively correlate with UPRmt transcripts and longevity. Furthermore, CBP/p300 inhibition disrupts, while forced expression of p300 is sufficient to activate, the UPRmt in mammalian cells. These results highlight an evolutionarily conserved mechanism that determines mitochondrial stress response, and promotes health and longevity through CBP/p300.


Subject(s)
CREB-Binding Protein , Caenorhabditis elegans Proteins , Longevity , Animals , Humans , Mice , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Histone Acetyltransferases/metabolism , Histone Demethylases/metabolism , Longevity/genetics , Mammals/metabolism , Transcription Factors/metabolism
14.
Science ; 366(6467): 827-832, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31727828

ABSTRACT

Throughout the animal kingdom, mitochondria are the only organelles that retain their own genome and the transcription and translation machineries that are all essential for energy harvesting. Mitochondria have developed a complex communication network, allowing them to stay in tune with cellular needs and nuclear transcriptional programs and to alleviate mitochondrial dysfunction. Here, we review recent findings on the wide array of mechanisms that contribute to these mitocellular communication networks, spanning from well-studied messenger molecules to mitonuclear genetic interactions. Based on these observations and developments, we advocate a broad and inclusive view on mitocellular interactions, which can have profound impacts on physiological, pathological, and evolutionary processes.


Subject(s)
Disease , Mitochondria/metabolism , Stress, Physiological , Animals , Cell Communication , Humans
15.
Free Radic Biol Med ; 143: 203-208, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31408725

ABSTRACT

The ability to respond to fluctuations of reactive oxygen species (ROS) within the cell is a central aspect of mammalian physiology. This dynamic process depends on the coordinated action of transcriptional factors to promote the expression of genes encoding for antioxidant enzymes. Here, we demonstrate that the transcriptional coregulators, PGC-1α and NCoR1, are essential mediators of mitochondrial redox homeostasis in skeletal muscle cells. Our findings reveal an antagonistic role of these coregulators in modulating mitochondrial antioxidant induction through Sod2 transcriptional control. Importantly, the activation of this mechanism by either PGC-1α overexpression or NCoR1 knockdown attenuates mitochondrial ROS levels and prevents cell death caused by lipid overload in skeletal muscle cells. The opposing actions of coactivators and corepressors, therefore, exert a commanding role over cellular antioxidant capacity.


Subject(s)
Gene Expression Regulation , Mitochondria/metabolism , Nuclear Receptor Co-Repressor 1/metabolism , Oxidation-Reduction/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Animals , Antioxidants/metabolism , Caenorhabditis elegans , Cell Survival , Green Fluorescent Proteins/metabolism , Homeostasis , Lipids/chemistry , Mice , Muscle, Skeletal/metabolism , Palmitates/pharmacology , Propidium/pharmacology , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Trans-Activators/metabolism , Transcription, Genetic
16.
Cell Rep ; 10(10): 1681-1691, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25772356

ABSTRACT

In recent years, tetracyclines, such as doxycycline, have become broadly used to control gene expression by virtue of the Tet-on/Tet-off systems. However, the wide range of direct effects of tetracycline use has not been fully appreciated. We show here that these antibiotics induce a mitonuclear protein imbalance through their effects on mitochondrial translation, an effect that likely reflects the evolutionary relationship between mitochondria and proteobacteria. Even at low concentrations, tetracyclines induce mitochondrial proteotoxic stress, leading to changes in nuclear gene expression and altered mitochondrial dynamics and function in commonly used cell types, as well as worms, flies, mice, and plants. Given that tetracyclines are so widely applied in research, scientists should be aware of their potentially confounding effects on experimental results. Furthermore, these results caution against extensive use of tetracyclines in livestock due to potential downstream impacts on the environment and human health.

17.
PLoS One ; 9(7): e103573, 2014.
Article in English | MEDLINE | ID: mdl-25072851

ABSTRACT

BACKGROUND: SIRT2 belongs to a highly conserved family of NAD+-dependent deacylases, consisting of seven members (SIRT1-SIRT7), which vary in subcellular localizations and have substrates ranging from histones to transcription factors and enzymes. Recently SIRT2 was revealed to play an important role in inflammation, directly binding, deacetylating, and inhibiting the p65 subunit of NF-κB. METHODS: A Sirt2 deficient mouse line (Sirt2-/-) was generated by deleting exons 5-7, encoding part of the SIRT2 deacetylase domain, by homologous recombination. Age- and sex-matched Sirt2-/- and Sirt2+/+ littermate mice were subjected to dextran sulfate sodium (DSS)-induced colitis and analyzed for colitis susceptibility. RESULTS: Sirt2-/- mice displayed more severe clinical and histological manifestations after DSS colitis compared to wild type littermates. Notably, under basal condition, Sirt2 deficiency does not affect the basal phenotype and intestinal morphology Sirt2 deficiency, however, affects macrophage polarization, creating a pro-inflammatory milieu in the immune cells compartment. CONCLUSION: These data confirm a protective role for SIRT2 against the development of inflammatory processes, pointing out a potential role for this sirtuin as a suppressor of colitis. In fact, SIRT2 deletion promotes inflammatory responses by increasing NF-κB acetylation and by reducing the M2-associated anti-inflammatory pathway. Finally, we speculate that the activation of SIRT2 may be a potential approach for the treatment of inflammatory bowel disease.


Subject(s)
Colitis/pathology , Macrophages/metabolism , Sirtuin 2/metabolism , Animals , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Bone Marrow Cells/cytology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Polarity , Cells, Cultured , Colitis/chemically induced , Colitis/metabolism , Colon/metabolism , Colon/pathology , Cytokines/blood , Cytokines/genetics , Dextran Sulfate/toxicity , Disease Models, Animal , Disease Susceptibility , Female , Lectins, C-Type/metabolism , Lymph Nodes/cytology , Lymph Nodes/immunology , Macrophages/cytology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Severity of Illness Index , Sirtuin 2/deficiency , Sirtuin 2/genetics
18.
Sci Rep ; 4: 5285, 2014 Jun 13.
Article in English | MEDLINE | ID: mdl-24923838

ABSTRACT

Mitochondria are semi-autonomous organelles regulated by a complex network of proteins that are vital for many cellular functions. Because mitochondrial modulators can impact many aspects of cellular homeostasis, their identification and validation has proven challenging. It requires the measurement of multiple parameters in parallel to understand the exact nature of the changes induced by such compounds. We developed a platform of assays scoring for mitochondrial function in two complementary models systems, mammalian cells and C. elegans. We first optimized cell culture conditions and established the mitochondrial signature of 1,200 FDA-approved drugs in liver cells. Using cell-based and C. elegans assays, we further defined the metabolic effects of two pharmacological classes that emerged from our hit list, i.e. imidazoles and statins. We found that these two drug classes affect respiration through different and cholesterol-independent mechanisms in both models. Our screening strategy enabled us to unequivocally identify compounds that have toxic or beneficial effects on mitochondrial activity. Furthermore, the cross-species approach provided novel mechanistic insight and allowed early validation of hits that act on mitochondrial function.


Subject(s)
Caenorhabditis elegans/drug effects , Mitochondria/drug effects , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , Pharmaceutical Preparations/administration & dosage , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line , Cell Line, Tumor , Cluster Analysis , Drug Approval , Drug Evaluation, Preclinical/methods , Fatty Acids, Monounsaturated/pharmacology , Fluvastatin , Gene Expression/drug effects , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Imidazoles/pharmacology , Indoles/pharmacology , Lovastatin/pharmacology , MCF-7 Cells , Mice , Mitochondria/metabolism , Pharmaceutical Preparations/classification , Reproducibility of Results , Simvastatin/pharmacology , United States , United States Food and Drug Administration
SELECTION OF CITATIONS
SEARCH DETAIL