Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Nature ; 583(7814): 122-126, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32461692

RESUMO

The cellular NADH/NAD+ ratio is fundamental to biochemistry, but the extent to which it reflects versus drives metabolic physiology in vivo is poorly understood. Here we report the in vivo application of Lactobacillus brevis (Lb)NOX1, a bacterial water-forming NADH oxidase, to assess the metabolic consequences of directly lowering the hepatic cytosolic NADH/NAD+ ratio in mice. By combining this genetic tool with metabolomics, we identify circulating α-hydroxybutyrate levels as a robust marker of an elevated hepatic cytosolic NADH/NAD+ ratio, also known as reductive stress. In humans, elevations in circulating α-hydroxybutyrate levels have previously been associated with impaired glucose tolerance2, insulin resistance3 and mitochondrial disease4, and are associated with a common genetic variant in GCKR5, which has previously been associated with many seemingly disparate metabolic traits. Using LbNOX, we demonstrate that NADH reductive stress mediates the effects of GCKR variation on many metabolic traits, including circulating triglyceride levels, glucose tolerance and FGF21 levels. Our work identifies an elevated hepatic NADH/NAD+ ratio as a latent metabolic parameter that is shaped by human genetic variation and contributes causally to key metabolic traits and diseases. Moreover, it underscores the utility of genetic tools such as LbNOX to empower studies of 'causal metabolism'.


Assuntos
Fígado/metabolismo , NAD/metabolismo , Estresse Fisiológico , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Citosol/metabolismo , Modelos Animais de Doenças , Fatores de Crescimento de Fibroblastos/sangue , Variação Genética , Teste de Tolerância a Glucose , Humanos , Resistência à Insulina , Levilactobacillus brevis/enzimologia , Levilactobacillus brevis/genética , Masculino , Camundongos , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo , NADH NADPH Oxirredutases/genética , NADH NADPH Oxirredutases/metabolismo , Oxirredução , Triglicerídeos/sangue
2.
Mol Genet Metab ; 133(1): 83-93, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33752971

RESUMO

Leigh syndrome is a severe mitochondrial neurodegenerative disease with no effective treatment. In the Ndufs4-/- mouse model of Leigh syndrome, continuously breathing 11% O2 (hypoxia) prevents neurodegeneration and leads to a dramatic extension (~5-fold) in lifespan. We investigated the effect of hypoxia on the brain metabolism of Ndufs4-/- mice by studying blood gas tensions and metabolite levels in simultaneously sampled arterial and cerebral internal jugular venous (IJV) blood. Relatively healthy Ndufs4-/- and wildtype (WT) mice breathing air until postnatal age ~38 d were compared to Ndufs4-/- and WT mice breathing air until ~38 days old followed by 4-weeks of breathing 11% O2. Compared to WT control mice, Ndufs4-/- mice breathing air have reduced brain O2 consumption as evidenced by an elevated partial pressure of O2 in IJV blood (PijvO2) despite a normal PO2 in arterial blood, and higher lactate/pyruvate (L/P) ratios in IJV plasma revealed by metabolic profiling. In Ndufs4-/- mice, hypoxia treatment normalized the cerebral venous PijvO2 and L/P ratios, and decreased levels of nicotinate in IJV plasma. Brain concentrations of nicotinamide adenine dinucleotide (NAD+) were lower in Ndufs4-/- mice breathing air than in WT mice, but preserved at WT levels with hypoxia treatment. Although mild hypoxia (17% O2) has been shown to be an ineffective therapy for Ndufs4-/- mice, we find that when combined with nicotinic acid supplementation it provides a modest improvement in neurodegeneration and lifespan. Therapies targeting both brain hyperoxia and NAD+ deficiency may hold promise for treating Leigh syndrome.


Assuntos
Encéfalo/metabolismo , Complexo I de Transporte de Elétrons/genética , Doença de Leigh/metabolismo , NAD/genética , Oxigênio/metabolismo , Animais , Encéfalo/patologia , Hipóxia Celular/fisiologia , Modelos Animais de Doenças , Complexo I de Transporte de Elétrons/metabolismo , Humanos , Doença de Leigh/genética , Doença de Leigh/terapia , Metabolômica , Camundongos , Mitocôndrias , NAD/deficiência , Doenças Neurodegenerativas , Respiração/genética
3.
Proc Natl Acad Sci U S A ; 114(43): E9096-E9104, 2017 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-29073106

RESUMO

Comparative analyses of transcriptional profiles from humans and mice with cardiovascular pathologies revealed consistently elevated expression of MICU2, a regulatory subunit of the mitochondrial calcium uniporter complex. To determine if MICU2 expression was cardioprotective, we produced and characterized Micu2-/- mice. Mutant mice had left atrial enlargement and Micu2-/- cardiomyocytes had delayed sarcomere relaxation and cytosolic calcium reuptake kinetics, indicating diastolic dysfunction. RNA sequencing (RNA-seq) of Micu2-/- ventricular tissues revealed markedly reduced transcripts encoding the apelin receptor (Micu2-/- vs. wild type, P = 7.8 × 10-40), which suppresses angiotensin II receptor signaling via allosteric transinhibition. We found that Micu2-/- and wild-type mice had comparable basal blood pressures and elevated responses to angiotensin II infusion, but that Micu2-/- mice exhibited systolic dysfunction and 30% lethality from abdominal aortic rupture. Aneurysms and rupture did not occur with norepinephrine-induced hypertension. Aortic tissue from Micu2-/- mice had increased expression of extracellular matrix remodeling genes, while single-cell RNA-seq analyses showed increased expression of genes related to reactive oxygen species, inflammation, and proliferation in fibroblast and smooth muscle cells. We concluded that Micu2-/- mice recapitulate features of diastolic heart disease and define previously unappreciated roles for Micu2 in regulating angiotensin II-mediated hypertensive responses that are critical in protecting the abdominal aorta from injury.


Assuntos
Canais de Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Cálcio/metabolismo , Cardiomiopatia Hipertrófica Familiar/genética , Angiotensina Amida/genética , Angiotensina II/farmacologia , Animais , Aorta Abdominal/patologia , Canais de Cálcio/genética , Proteínas de Ligação ao Cálcio/genética , Cardiomiopatia Hipertrófica Familiar/patologia , Eletrocardiografia , Regulação da Expressão Gênica , Homeostase/efeitos dos fármacos , Homeostase/fisiologia , Humanos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mitocôndrias Hepáticas/fisiologia , Miócitos Cardíacos/patologia , Miócitos Cardíacos/fisiologia
4.
Proc Natl Acad Sci U S A ; 114(21): E4241-E4250, 2017 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-28483998

RESUMO

The most common pediatric mitochondrial disease is Leigh syndrome, an episodic, subacute neurodegeneration that can lead to death within the first few years of life, for which there are no proven general therapies. Mice lacking the complex I subunit, Ndufs4, develop a fatal progressive encephalopathy resembling Leigh syndrome and die at ≈60 d of age. We previously reported that continuously breathing normobaric 11% O2 from an early age prevents neurological disease and dramatically improves survival in these mice. Here, we report three advances. First, we report updated survival curves and organ pathology in Ndufs4 KO mice exposed to hypoxia or hyperoxia. Whereas normoxia-treated KO mice die from neurodegeneration at about 60 d, hypoxia-treated mice eventually die at about 270 d, likely from cardiac disease, and hyperoxia-treated mice die within days from acute pulmonary edema. Second, we report that more conservative hypoxia regimens, such as continuous normobaric 17% O2 or intermittent hypoxia, are ineffective in preventing neuropathology. Finally, we show that breathing normobaric 11% O2 in mice with late-stage encephalopathy reverses their established neurological disease, evidenced by improved behavior, circulating disease biomarkers, and survival rates. Importantly, the pathognomonic MRI brain lesions and neurohistopathologic findings are reversed after 4 wk of hypoxia. Upon return to normoxia, Ndufs4 KO mice die within days. Future work is required to determine if hypoxia can be used to prevent and reverse neurodegeneration in other animal models, and to determine if it can be provided in a safe and practical manner to allow in-hospital human therapeutic trials.


Assuntos
Complexo I de Transporte de Elétrons/genética , Hipóxia/metabolismo , Doença de Leigh/patologia , Doença de Leigh/terapia , Mitocôndrias/patologia , Doenças Neurodegenerativas/terapia , Animais , Modelos Animais de Doenças , Doença de Leigh/mortalidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Doenças Neurodegenerativas/patologia , Oxigênio/uso terapêutico , Respiração
5.
Am J Physiol Lung Cell Mol Physiol ; 316(2): L391-L399, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30520688

RESUMO

Hypoxic pulmonary vasoconstriction (HPV) is a physiological vasomotor response that maintains systemic oxygenation by matching perfusion to ventilation during alveolar hypoxia. Although mitochondria appear to play an essential role in HPV, the impact of mitochondrial dysfunction on HPV remains incompletely defined. Mice lacking the mitochondrial complex I (CI) subunit Ndufs4 ( Ndufs4-/-) develop a fatal progressive encephalopathy and serve as a model for Leigh syndrome, the most common mitochondrial disease in children. Breathing normobaric 11% O2 prevents neurological disease and improves survival in Ndufs4-/- mice. In this study, we found that either genetic Ndufs4 deficiency or pharmacological inhibition of CI using piericidin A impaired the ability of left mainstem bronchus occlusion (LMBO) to induce HPV. In mice breathing air, the partial pressure of arterial oxygen during LMBO was lower in Ndufs4-/- and in piericidin A-treated Ndufs4+/+ mice than in respective controls. Impairment of HPV in Ndufs4-/- mice was not a result of nonspecific dysfunction of the pulmonary vascular contractile apparatus or pulmonary inflammation. In Ndufs4-deficient mice, 3 wk of breathing 11% O2 restored HPV in response to LMBO. When compared with Ndufs4-/- mice breathing air, chronic hypoxia improved systemic oxygenation during LMBO. The results of this study show that, when breathing air, mice with a congenital Ndufs4 deficiency or chemically inhibited CI function have impaired HPV. Our study raises the possibility that patients with inborn errors of mitochondrial function may also have defects in HPV.


Assuntos
Complexo I de Transporte de Elétrons/deficiência , Hipóxia/fisiopatologia , Doença de Leigh/fisiopatologia , Vasoconstrição/fisiologia , Animais , Brônquios/metabolismo , Modelos Animais de Doenças , Hipóxia/metabolismo , Pulmão/metabolismo , Pulmão/fisiopatologia , Camundongos Transgênicos , Mitocôndrias/metabolismo , Artéria Pulmonar/metabolismo , Circulação Pulmonar/fisiologia
6.
Nature ; 476(7360): 341-5, 2011 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-21685886

RESUMO

Mitochondria from diverse organisms are capable of transporting large amounts of Ca(2+) via a ruthenium-red-sensitive, membrane-potential-dependent mechanism called the uniporter. Although the uniporter's biophysical properties have been studied extensively, its molecular composition remains elusive. We recently used comparative proteomics to identify MICU1 (also known as CBARA1), an EF-hand-containing protein that serves as a putative regulator of the uniporter. Here, we use whole-genome phylogenetic profiling, genome-wide RNA co-expression analysis and organelle-wide protein coexpression analysis to predict proteins functionally related to MICU1. All three methods converge on a novel predicted transmembrane protein, CCDC109A, that we now call 'mitochondrial calcium uniporter' (MCU). MCU forms oligomers in the mitochondrial inner membrane, physically interacts with MICU1, and resides within a large molecular weight complex. Silencing MCU in cultured cells or in vivo in mouse liver severely abrogates mitochondrial Ca(2+) uptake, whereas mitochondrial respiration and membrane potential remain fully intact. MCU has two predicted transmembrane helices, which are separated by a highly conserved linker facing the intermembrane space. Acidic residues in this linker are required for its full activity. However, an S259A point mutation retains function but confers resistance to Ru360, the most potent inhibitor of the uniporter. Our genomic, physiological, biochemical and pharmacological data firmly establish MCU as an essential component of the mitochondrial Ca(2+) uniporter.


Assuntos
Canais de Cálcio/química , Canais de Cálcio/metabolismo , Genômica , Sequência de Aminoácidos , Animais , Cálcio/metabolismo , Canais de Cálcio/genética , Células HEK293 , Células HeLa , Humanos , Transporte de Íons , Camundongos , Mitocôndrias Hepáticas/metabolismo , Membranas Mitocondriais/química , Membranas Mitocondriais/metabolismo , Dados de Sequência Molecular , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Filogenia , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína
7.
Nat Genet ; 38(5): 576-82, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16582907

RESUMO

The majority of inherited mitochondrial disorders are due to mutations not in the mitochondrial genome (mtDNA) but rather in the nuclear genes encoding proteins targeted to this organelle. Elucidation of the molecular basis for these disorders is limited because only half of the estimated 1,500 mitochondrial proteins have been identified. To systematically expand this catalog, we experimentally and computationally generated eight genome-scale data sets, each designed to provide clues as to mitochondrial localization: targeting sequence prediction, protein domain enrichment, presence of cis-regulatory motifs, yeast homology, ancestry, tandem-mass spectrometry, coexpression and transcriptional induction during mitochondrial biogenesis. Through an integrated analysis we expand the collection to 1,080 genes, which includes 368 novel predictions with a 10% estimated false prediction rate. By combining this expanded inventory with genetic intervals linked to disease, we have identified candidate genes for eight mitochondrial disorders, leading to the discovery of mutations in MPV17 that result in hepatic mtDNA depletion syndrome. The integrative approach promises to better define the role of mitochondria in both rare and common human diseases.


Assuntos
Genômica , Doenças Mitocondriais/genética , Sequência de Bases , Primers do DNA , Ligação Genética , Humanos , Espectrometria de Massas/métodos
8.
Cell Metab ; 36(1): 144-158.e7, 2024 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-38101397

RESUMO

Common genetic variants in glucokinase regulator (GCKR), which encodes GKRP, a regulator of hepatic glucokinase (GCK), influence multiple metabolic traits in genome-wide association studies (GWASs), making GCKR one of the most pleiotropic GWAS loci in the genome. It is unclear why. Prior work has demonstrated that GCKR influences the hepatic cytosolic NADH/NAD+ ratio, also referred to as reductive stress. Here, we demonstrate that reductive stress is sufficient to activate the transcription factor ChREBP and necessary for its activation by the GKRP-GCK interaction, glucose, and ethanol. We show that hepatic reductive stress induces GCKR GWAS traits such as increased hepatic fat, circulating FGF21, and circulating acylglycerol species, which are also influenced by ChREBP. We define the transcriptional signature of hepatic reductive stress and show its upregulation in fatty liver disease and downregulation after bariatric surgery in humans. These findings highlight how a GCKR-reductive stress-ChREBP axis influences multiple human metabolic traits.


Assuntos
Estudo de Associação Genômica Ampla , Glucoquinase , Humanos , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Glucoquinase/genética , Glucoquinase/metabolismo , Glucose/metabolismo , Fígado/metabolismo , Fatores de Transcrição/metabolismo
9.
Proc Natl Acad Sci U S A ; 107(4): 1571-5, 2010 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-20080599

RESUMO

Mutations in either the mitochondrial or nuclear genomes can give rise to respiratory chain disease (RCD), a large class of devastating metabolic disorders. Their clinical management is challenging, in part because we lack facile and accurate biomarkers to aid in diagnosis and in the monitoring of disease progression. Here we introduce a sequential strategy that combines biochemical analysis of spent media from cell culture with analysis of patient plasma to identify disease biomarkers. First, we applied global metabolic profiling to spotlight 32 metabolites whose uptake or secretion kinetics were altered by chemical inhibition of the respiratory chain in cultured muscle . These metabolites span a wide range of pathways and include lactate and alanine, which are used clinically as biomarkers of RCD. We next measured the cell culture-defined metabolites in human plasma to discover that creatine is reproducibly elevated in two independent cohorts of RCD patients, exceeding lactate and alanine in magnitude of elevation and statistical significance. In cell culture extracellular creatine was inversely related to the intracellular phosphocreatine:creatine ratio suggesting that the elevation of plasma creatine in RCD patients signals a low energetic state of tissues using the phosphocreatine shuttle. Our study identifies plasma creatine as a potential biomarker of human mitochondrial dysfunction that could be clinically useful. More generally, we illustrate how spent media from cellular models of disease may provide a window into the biochemical derangements in human plasma, an approach that could, in principle, be extended to a range of complex diseases.


Assuntos
Doenças Mitocondriais/sangue , Células Musculares/química , Adulto , Animais , Biomarcadores , Linhagem Celular , Creatina/sangue , Creatina/metabolismo , Meios de Cultura , Transporte de Elétrons , Feminino , Humanos , Masculino , Metabolômica , Camundongos , Pessoa de Meia-Idade , Células Musculares/metabolismo , Adulto Jovem
10.
Nat Commun ; 13(1): 2483, 2022 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-35513392

RESUMO

The SLC25 carrier family consists of 53 transporters that shuttle nutrients and co-factors across mitochondrial membranes. The family is highly redundant and their transport activities coupled to metabolic state. Here, we use a pooled, dual CRISPR screening strategy that knocks out pairs of transporters in four metabolic states - glucose, galactose, OXPHOS inhibition, and absence of pyruvate - designed to unmask the inter-dependence of these genes. In total, we screen 63 genes in four metabolic states, corresponding to 2016 single and pair-wise genetic perturbations. We recover 19 gene-by-environment (GxE) interactions and 9 gene-by-gene (GxG) interactions. One GxE interaction hit illustrates that the fitness defect in the mitochondrial folate carrier (SLC25A32) KO cells is genetically buffered in galactose due to a lack of substrate in de novo purine biosynthesis. GxG analysis highlights a buffering interaction between the iron transporter SLC25A37 (A37) and the poorly characterized SLC25A39 (A39). Mitochondrial metabolite profiling, organelle transport assays, and structure-guided mutagenesis identify A39 as critical for mitochondrial glutathione (GSH) import. Functional studies reveal that A39-mediated glutathione homeostasis and A37-mediated mitochondrial iron uptake operate jointly to support mitochondrial OXPHOS. Our work underscores the value of studying family-wide genetic interactions across different metabolic environments.


Assuntos
Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Galactose , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Glutationa , Homeostase , Ferro , Proteínas de Membrana Transportadoras/genética
11.
Life Sci Alliance ; 3(10)2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32769116

RESUMO

The mitochondrial calcium uniporter (MCU) is a calcium-activated calcium channel critical for signaling and bioenergetics. MCU, the pore-forming subunit of the uniporter, contains two transmembrane domains and is found in all major eukaryotic taxa. In amoeba and fungi, MCU homologs are sufficient to form a functional calcium channel, whereas human MCU exhibits a strict requirement for the metazoan protein essential MCU regulator (EMRE) for conductance. Here, we exploit this evolutionary divergence to decipher the molecular basis of human MCU's dependence on EMRE. By systematically generating chimeric proteins that consist of EMRE-independent Dictyostelium discoideum MCU and Homo sapiens MCU (HsMCU), we converged on a stretch of 10 amino acids in D. discoideum MCU that can be transplanted to HsMCU to render it EMRE independent. We call this region in human MCU the EMRE dependence domain (EDD). Crosslinking experiments show that EMRE directly interacts with HsMCU at its transmembrane domains as well as the EDD. Our results suggest that EMRE stabilizes the EDD of MCU, permitting both channel opening and calcium conductance, consistent with recently published structures of MCU-EMRE.


Assuntos
Canais de Cálcio/metabolismo , Evolução Biológica , Cálcio/metabolismo , Canais de Cálcio/fisiologia , Dictyostelium/genética , Dictyostelium/metabolismo , Evolução Molecular , Células HEK293 , Humanos , Transporte de Íons/genética , Transporte de Íons/fisiologia , Mitocôndrias/metabolismo , Domínios Proteicos
12.
Cell Rep ; 27(5): 1364-1375.e5, 2019 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-31042465

RESUMO

The mitochondrial calcium uniporter has been proposed to coordinate the organelle's energetics with calcium signaling. Uniporter current has previously been reported to be extremely high in brown adipose tissue (BAT), yet it remains unknown how the uniporter contributes to BAT physiology. Here, we report the generation and characterization of a mouse model lacking Mcu, the pore forming subunit of the uniporter, specifically in BAT (BAT-Mcu-KO). BAT-Mcu-KO mice lack uniporter-based calcium uptake in BAT mitochondria but exhibit unaffected cold tolerance, diet-induced obesity, and transcriptional response to cold in BAT. Unexpectedly, we found in wild-type animals that cold powerfully activates the ATF4-dependent integrated stress response (ISR) in BAT and upregulates circulating FGF21 and GDF15, raising the hypothesis that the ISR partly underlies the pleiotropic effects of BAT on systemic metabolism. Our study demonstrates that the uniporter is largely dispensable for BAT thermogenesis and demonstrates activation of the ISR in BAT in response to cold.


Assuntos
Tecido Adiposo Marrom/metabolismo , Canais de Cálcio/genética , Resposta ao Choque Frio , Mitocôndrias/metabolismo , Proteínas Mitocondriais/genética , Termogênese , Fator 4 Ativador da Transcrição/metabolismo , Animais , Cálcio/metabolismo , Linhagem Celular , Dieta Hiperlipídica/efeitos adversos , Metabolismo Energético , Feminino , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Fator 15 de Diferenciação de Crescimento/genética , Fator 15 de Diferenciação de Crescimento/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/etiologia , Obesidade/genética
13.
Cell Metab ; 30(4): 824-832.e3, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31402314

RESUMO

Leigh syndrome is a devastating mitochondrial disease for which there are no proven therapies. We previously showed that breathing chronic, continuous hypoxia can prevent and even reverse neurological disease in the Ndufs4 knockout (KO) mouse model of complex I (CI) deficiency and Leigh syndrome. Here, we show that genetic activation of the hypoxia-inducible factor transcriptional program via any of four different strategies is insufficient to rescue disease. Rather, we observe an age-dependent decline in whole-body oxygen consumption. These mice exhibit brain tissue hyperoxia, which is normalized by hypoxic breathing. Alternative experimental strategies to reduce oxygen delivery, including breathing carbon monoxide (600 ppm in air) or severe anemia, can reverse neurological disease. Therefore, unused oxygen is the most likely culprit in the pathology of this disease. While pharmacologic activation of the hypoxia response is unlikely to alleviate disease in vivo, interventions that safely normalize brain tissue hyperoxia may hold therapeutic potential.


Assuntos
Encéfalo/metabolismo , Monóxido de Carbono/uso terapêutico , Hiperóxia/terapia , Doença de Leigh/terapia , Oxigênio/metabolismo , Anemia/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Hiperóxia/metabolismo , Hipóxia/metabolismo , Fator 1 Induzível por Hipóxia/genética , Fator 1 Induzível por Hipóxia/metabolismo , Doença de Leigh/metabolismo , Camundongos
14.
Science ; 352(6281): 54-61, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26917594

RESUMO

Defects in the mitochondrial respiratory chain (RC) underlie a spectrum of human conditions, ranging from devastating inborn errors of metabolism to aging. We performed a genome-wide Cas9-mediated screen to identify factors that are protective during RC inhibition. Our results highlight the hypoxia response, an endogenous program evolved to adapt to limited oxygen availability. Genetic or small-molecule activation of the hypoxia response is protective against mitochondrial toxicity in cultured cells and zebrafish models. Chronic hypoxia leads to a marked improvement in survival, body weight, body temperature, behavior, neuropathology, and disease biomarkers in a genetic mouse model of Leigh syndrome, the most common pediatric manifestation of mitochondrial disease. Further preclinical studies are required to assess whether hypoxic exposure can be developed into a safe and effective treatment for human diseases associated with mitochondrial dysfunction.


Assuntos
Doença de Leigh/genética , Doença de Leigh/terapia , Mitocôndrias/metabolismo , Oxigênio/metabolismo , Proteína Supressora de Tumor Von Hippel-Lindau/genética , Anaerobiose , Animais , Antimicina A/análogos & derivados , Antimicina A/farmacologia , Proteínas de Bactérias , Biomarcadores/sangue , Temperatura Corporal , Peso Corporal , Proteína 9 Associada à CRISPR , Modelos Animais de Doenças , Transporte de Elétrons/efeitos dos fármacos , Complexo I de Transporte de Elétrons/genética , Endonucleases , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/genética , Técnicas de Inativação de Genes , Estudo de Associação Genômica Ampla , Glicina/análogos & derivados , Glicina/farmacologia , Glicina/uso terapêutico , Humanos , Fator 1 Induzível por Hipóxia/metabolismo , Isoquinolinas/farmacologia , Isoquinolinas/uso terapêutico , Células K562 , Doença de Leigh/patologia , Camundongos , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , Respiração , Supressão Genética , Proteína Supressora de Tumor Von Hippel-Lindau/antagonistas & inibidores , Peixe-Zebra
15.
Elife ; 52016 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-27307216

RESUMO

Mitochondrial dysfunction is associated with a spectrum of human disorders, ranging from rare, inborn errors of metabolism to common, age-associated diseases such as neurodegeneration. How these lesions give rise to diverse pathology is not well understood, partly because their proximal consequences have not been well-studied in mammalian cells. Here we provide two lines of evidence that mitochondrial respiratory chain dysfunction leads to alterations in one-carbon metabolism pathways. First, using hypothesis-generating metabolic, proteomic, and transcriptional profiling, followed by confirmatory experiments, we report that mitochondrial DNA depletion leads to an ATF4-mediated increase in serine biosynthesis and transsulfuration. Second, we show that lesioning the respiratory chain impairs mitochondrial production of formate from serine, and that in some cells, respiratory chain inhibition leads to growth defects upon serine withdrawal that are rescuable with purine or formate supplementation. Our work underscores the connection between the respiratory chain and one-carbon metabolism with implications for understanding mitochondrial pathogenesis.


Assuntos
Carbono/metabolismo , Fosforilação Oxidativa , Linhagem Celular , Perfilação da Expressão Gênica , Humanos , Metaboloma , Proteoma/análise
16.
Science ; 342(6164): 1379-82, 2013 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-24231807

RESUMO

The mitochondrial uniporter is a highly selective calcium channel in the organelle's inner membrane. Its molecular components include the EF-hand-containing calcium-binding proteins mitochondrial calcium uptake 1 (MICU1) and MICU2 and the pore-forming subunit mitochondrial calcium uniporter (MCU). We sought to achieve a full molecular characterization of the uniporter holocomplex (uniplex). Quantitative mass spectrometry of affinity-purified uniplex recovered MICU1 and MICU2, MCU and its paralog MCUb, and essential MCU regulator (EMRE), a previously uncharacterized protein. EMRE is a 10-kilodalton, metazoan-specific protein with a single transmembrane domain. In its absence, uniporter channel activity was lost despite intact MCU expression and oligomerization. EMRE was required for the interaction of MCU with MICU1 and MICU2. Hence, EMRE is essential for in vivo uniporter current and additionally bridges the calcium-sensing role of MICU1 and MICU2 with the calcium-conducting role of MCU.


Assuntos
Canais de Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Membrana Celular/metabolismo , Mitocôndrias/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Sequência de Aminoácidos , Canais de Cálcio/química , Canais de Cálcio/genética , Proteínas de Ligação ao Cálcio/genética , Proteínas de Transporte de Cátions/genética , Motivos EF Hand , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Proteínas de Transporte da Membrana Mitocondrial/genética , Dados de Sequência Molecular , Filogenia , Estrutura Terciária de Proteína , Proteômica
17.
Cell Metab ; 14(3): 428-34, 2011 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-21907147

RESUMO

The metazoan mitochondrial translation machinery is unusual in having a single tRNA(Met) that fulfills the dual role of the initiator and elongator tRNA(Met). A portion of the Met-tRNA(Met) pool is formylated by mitochondrial methionyl-tRNA formyltransferase (MTFMT) to generate N-formylmethionine-tRNA(Met) (fMet-tRNA(met)), which is used for translation initiation; however, the requirement of formylation for initiation in human mitochondria is still under debate. Using targeted sequencing of the mtDNA and nuclear exons encoding the mitochondrial proteome (MitoExome), we identified compound heterozygous mutations in MTFMT in two unrelated children presenting with Leigh syndrome and combined OXPHOS deficiency. Patient fibroblasts exhibit severe defects in mitochondrial translation that can be rescued by exogenous expression of MTFMT. Furthermore, patient fibroblasts have dramatically reduced fMet-tRNA(Met) levels and an abnormal formylation profile of mitochondrially translated COX1. Our findings demonstrate that MTFMT is critical for efficient human mitochondrial translation and reveal a human disorder of Met-tRNA(Met) formylation.


Assuntos
Ciclo-Oxigenase 1/metabolismo , DNA Mitocondrial/química , Fibroblastos/metabolismo , Doença de Leigh/genética , Mitocôndrias/genética , Proteínas Mitocondriais/genética , Biossíntese de Proteínas , RNA de Transferência de Metionina/metabolismo , Células Cultivadas , Criança , Ciclo-Oxigenase 1/genética , DNA Mitocondrial/genética , Fibroblastos/patologia , Heterozigoto , Humanos , Hidroximetil e Formil Transferases , Immunoblotting , Doença de Leigh/metabolismo , Doença de Leigh/patologia , Lentivirus , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Mutação , Biossíntese de Proteínas/genética , Análise de Sequência de DNA , Transdução Genética , Vírion
18.
Nat Genet ; 42(10): 851-8, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20818383

RESUMO

Discovering the molecular basis of mitochondrial respiratory chain disease is challenging given the large number of both mitochondrial and nuclear genes that are involved. We report a strategy of focused candidate gene prediction, high-throughput sequencing and experimental validation to uncover the molecular basis of mitochondrial complex I disorders. We created seven pools of DNA from a cohort of 103 cases and 42 healthy controls and then performed deep sequencing of 103 candidate genes to identify 151 rare variants that were predicted to affect protein function. We established genetic diagnoses in 13 of 60 previously unsolved cases using confirmatory experiments, including cDNA complementation to show that mutations in NUBPL and FOXRED1 can cause complex I deficiency. Our study illustrates how large-scale sequencing, coupled with functional prediction and experimental validation, can be used to identify causal mutations in individual cases.


Assuntos
Complexo I de Transporte de Elétrons/genética , Estudos de Associação Genética , Doenças Mitocondriais/genética , Proteínas Mitocondriais/genética , Mutação/genética , Western Blotting , Estudos de Casos e Controles , Dosagem de Genes , Humanos , Proteínas Mitocondriais/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA
19.
J Biol Chem ; 281(34): 24365-74, 2006 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-16807246

RESUMO

Syndecans comprise a major family of cell surface heparan sulfate proteoglycans (HSPGs). Syndecans bind and modulate a wide variety of biological molecules through their heparan sulfate (HS) moiety. Although all syndecans contain the ligand binding HS chains, they likely perform specific functions in vivo because their temporal and spatial expression patterns are different. However, how syndecan expression is regulated has yet to be clearly defined. In this study, we examined how syndecan-1 expression is regulated in epithelial cells. Our results showed that among several bioactive agents tested, only forskolin and three isoforms of TGFbeta (TGFbeta1-TGFbeta3) significantly induced syndecan-1, but not syndecan-4, expression on various epithelial cells. Steady-state syndecan-1 mRNA was not increased by TGFbeta treatment and cycloheximide did not inhibit syndecan-1 induction by TGFbeta, indicating that TGFbeta induces syndecan-1 in a post-translational manner. However, TGFbeta induction of syndecan-1 was inhibited by transient expression of a dominant-negative construct of protein kinase A (PKA) and by specific inhibitors of PKA. Further (i) syndecan-1 cytoplasmic domains were Ser-phosphorylated when cells were treated with TGFbeta and this was inhibited by a PKA inhibitor, (ii) PKA was co-immunoprecipitated from cell lysates by anti-syndecan-1 antibodies, (iii) PKA phosphorylated recombinant syndecan-1 cytoplasmic domains in vitro, and (iv) expression of a syndecan-1 construct with its invariant Ser(286) replaced with a Gly was not induced by TGFbeta. Together, these findings define a regulatory mechanism where TGFbeta signals through PKA to phosphorylate the syndecan-1 cytoplasmic domain and increases syndecan-1 expression on epithelial cells.


Assuntos
Glicoproteínas de Membrana/biossíntese , Proteoglicanas/biossíntese , Fator de Crescimento Transformador beta/metabolismo , Animais , Linhagem Celular , Colforsina/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Células Epiteliais/metabolismo , Humanos , Glicoproteínas de Membrana/genética , Camundongos , Fosforilação , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Estrutura Terciária de Proteína , Proteoglicanas/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serina , Transdução de Sinais/efeitos dos fármacos , Sindecana-1 , Sindecanas , Fator de Crescimento Transformador beta/farmacologia
20.
Biochemistry ; 45(18): 5703-11, 2006 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-16669614

RESUMO

Syndecans are a family of four transmembrane heparan sulfate proteoglycans that act as coreceptors for a variety of cell-surface ligands and receptors. Receptor activation in several cell types leads to shedding of syndecan-1 and syndecan-4 ectodomains into the extracellular space by metalloproteinase-mediated cleavage of the syndecan core protein. We have found that 3T3-L1 adipocytes express syndecan-1 and syndecan-4 and that their ectodomains are shed in response to insulin in a dose-, time-, and metalloproteinase-dependent manner. Insulin responsive shedding is not seen in 3T3-L1 fibroblasts. This shedding involves both Ras-MAP kinase and phosphatidylinositol 3-kinase pathways. In response to insulin, adipocytes are known to secrete active lipoprotein lipase, an enzyme that binds to heparan sulfate on the luminal surface of capillary endothelia. Lipoprotein lipase is transported as a stable enzyme from its site of synthesis to its site of action, but the transport mechanism is unknown. Our studies indicate that shed adipocyte syndecans associate with lipoprotein lipase. The shed syndecan ectodomain can stabilize active lipoprotein lipase. These data suggest that syndecan ectodomains, shed by adipocytes in response to insulin, are physiological extracellular chaperones for lipoprotein lipase as it translocates from its site of synthesis to its site of action.


Assuntos
Insulina/farmacologia , Lipase Lipoproteica/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteoglicanas/metabolismo , Células 3T3-L1 , Animais , Bovinos , Estabilidade Enzimática , Imunoprecipitação , Sistema de Sinalização das MAP Quinases , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Sindecana-1 , Sindecanas
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa