Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 615(7953): 712-719, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36922590

RESUMEN

Mitochondria are critical to the governance of metabolism and bioenergetics in cancer cells1. The mitochondria form highly organized networks, in which their outer and inner membrane structures define their bioenergetic capacity2,3. However, in vivo studies delineating the relationship between the structural organization of mitochondrial networks and their bioenergetic activity have been limited. Here we present an in vivo structural and functional analysis of mitochondrial networks and bioenergetic phenotypes in non-small cell lung cancer (NSCLC) using an integrated platform consisting of positron emission tomography imaging, respirometry and three-dimensional scanning block-face electron microscopy. The diverse bioenergetic phenotypes and metabolic dependencies we identified in NSCLC tumours align with distinct structural organization of mitochondrial networks present. Further, we discovered that mitochondrial networks are organized into distinct compartments within tumour cells. In tumours with high rates of oxidative phosphorylation (OXPHOSHI) and fatty acid oxidation, we identified peri-droplet mitochondrial networks wherein mitochondria contact and surround lipid droplets. By contrast, we discovered that in tumours with low rates of OXPHOS (OXPHOSLO), high glucose flux regulated perinuclear localization of mitochondria, structural remodelling of cristae and mitochondrial respiratory capacity. Our findings suggest that in NSCLC, mitochondrial networks are compartmentalized into distinct subpopulations that govern the bioenergetic capacity of tumours.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Metabolismo Energético , Neoplasias Pulmonares , Mitocondrias , Humanos , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/ultraestructura , Ácidos Grasos/metabolismo , Glucosa/metabolismo , Gotas Lipídicas/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/ultraestructura , Microscopía Electrónica , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Fosforilación Oxidativa , Fenotipo , Tomografía de Emisión de Positrones
3.
Cell ; 142(3): 456-67, 2010 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-20691904

RESUMEN

RNA import into mammalian mitochondria is considered essential for replication, transcription, and translation of the mitochondrial genome but the pathway(s) and factors that control this import are poorly understood. Previously, we localized polynucleotide phosphorylase (PNPASE), a 3' --> 5' exoribonuclease and poly-A polymerase, in the mitochondrial intermembrane space, a location lacking resident RNAs. Here, we show a new role for PNPASE in regulating the import of nuclear-encoded RNAs into the mitochondrial matrix. PNPASE reduction impaired mitochondrial RNA processing and polycistronic transcripts accumulated. Augmented import of RNase P, 5S rRNA, and MRP RNAs depended on PNPASE expression and PNPASE-imported RNA interactions were identified. PNPASE RNA processing and import activities were separable and a mitochondrial RNA targeting signal was isolated that enabled RNA import in a PNPASE-dependent manner. Combined, these data strongly support an unanticipated role for PNPASE in mediating the translocation of RNAs into mitochondria.


Asunto(s)
Mitocondrias/metabolismo , Polirribonucleótido Nucleotidiltransferasa/metabolismo , ARN/metabolismo , Animales , Línea Celular , Técnicas de Inactivación de Genes , Humanos , Ratones , Ratones Endogámicos C57BL , Polirribonucleótido Nucleotidiltransferasa/genética , Procesamiento Postranscripcional del ARN , Ribonucleasa P/metabolismo , Saccharomyces cerevisiae/metabolismo
4.
Nature ; 575(7782): 380-384, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31666695

RESUMEN

Mitochondria are essential regulators of cellular energy and metabolism, and have a crucial role in sustaining the growth and survival of cancer cells. A central function of mitochondria is the synthesis of ATP by oxidative phosphorylation, known as mitochondrial bioenergetics. Mitochondria maintain oxidative phosphorylation by creating a membrane potential gradient that is generated by the electron transport chain to drive the synthesis of ATP1. Mitochondria are essential for tumour initiation and maintaining tumour cell growth in cell culture and xenografts2,3. However, our understanding of oxidative mitochondrial metabolism in cancer is limited because most studies have been performed in vitro in cell culture models. This highlights a need for in vivo studies to better understand how oxidative metabolism supports tumour growth. Here we measure mitochondrial membrane potential in non-small-cell lung cancer in vivo using a voltage-sensitive, positron emission tomography (PET) radiotracer known as 4-[18F]fluorobenzyl-triphenylphosphonium (18F-BnTP)4. By using PET imaging of 18F-BnTP, we profile mitochondrial membrane potential in autochthonous mouse models of lung cancer, and find distinct functional mitochondrial heterogeneity within subtypes of lung tumours. The use of 18F-BnTP PET imaging enabled us to functionally profile mitochondrial membrane potential in live tumours.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/fisiopatología , Neoplasias Pulmonares/fisiopatología , Potencial de la Membrana Mitocondrial , Células A549 , Animales , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico por imagen , Humanos , Neoplasias Pulmonares/diagnóstico por imagen , Ratones , Ratones Transgénicos , Compuestos Organofosforados , Tomografía de Emisión de Positrones
5.
Cell ; 138(4): 628-44, 2009 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-19703392

RESUMEN

Most mitochondrial proteins are synthesized on cytosolic ribosomes and must be imported across one or both mitochondrial membranes. There is an amazingly versatile set of machineries and mechanisms, and at least four different pathways, for the importing and sorting of mitochondrial precursor proteins. The translocases that catalyze these processes are highly dynamic machines driven by the membrane potential, ATP, or redox reactions, and they cooperate with molecular chaperones and assembly complexes to direct mitochondrial proteins to their correct destinations. Here, we discuss recent insights into the importing and sorting of mitochondrial proteins and their contributions to mitochondrial biogenesis.


Asunto(s)
Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Humanos , Proteínas Mitocondriales/química , Señales de Clasificación de Proteína , Transporte de Proteínas
6.
Cell ; 134(3): 382-3, 2008 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-18692459

RESUMEN

Most mitochondrial proteins are synthesized in the cytosol and imported into the mitochondrion via molecular machines called translocons on the outer and inner mitochondrial membranes. Alder et al. (2008b) examine protein translocation into intact mitochondria by adapting fluorescent techniques first used to study translocation in the endoplasmic reticulum.


Asunto(s)
Mitocondrias/metabolismo , Proteínas Mitocondriales/química , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/metabolismo , Mitocondrias/química , Proteínas Mitocondriales/metabolismo , Transporte de Proteínas , Proteínas de Saccharomyces cerevisiae/metabolismo , Espectrometría de Fluorescencia
7.
J Biol Chem ; 297(4): 101135, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34461091

RESUMEN

Yeast is a facultative anaerobe and uses diverse electron acceptors to maintain redox-regulated import of cysteine-rich precursors via the mitochondrial intermembrane space assembly (MIA) pathway. With the growing diversity of substrates utilizing the MIA pathway, understanding the capacity of the intermembrane space (IMS) to handle different types of stress is crucial. We used MS to identify additional proteins that interacted with the sulfhydryl oxidase Erv1 of the MIA pathway. Altered inheritance of mitochondria 32 (Aim32), a thioredoxin-like [2Fe-2S] ferredoxin protein, was identified as an Erv1-binding protein. Detailed localization studies showed that Aim32 resided in both the mitochondrial matrix and IMS. Aim32 interacted with additional proteins including redox protein Osm1 and protein import components Tim17, Tim23, and Tim22. Deletion of Aim32 or mutation of conserved cysteine residues that coordinate the Fe-S center in Aim32 resulted in an increased accumulation of proteins with aberrant disulfide linkages. In addition, the steady-state level of assembled TIM22, TIM23, and Oxa1 protein import complexes was decreased. Aim32 also bound to several mitochondrial proteins under nonreducing conditions, suggesting a function in maintaining the redox status of proteins by potentially targeting cysteine residues that may be sensitive to oxidation. Finally, Aim32 was essential for growth in conditions of stress such as elevated temperature and hydroxyurea, and under anaerobic conditions. These studies suggest that the Fe-S protein Aim32 has a potential role in general redox homeostasis in the matrix and IMS. Thus, Aim32 may be poised as a sensor or regulator in quality control for a broad range of mitochondrial proteins.


Asunto(s)
Ferredoxinas/metabolismo , Saccharomyces cerevisiae/metabolismo , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/metabolismo , Ferredoxinas/genética , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oxidación-Reducción , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/genética , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
8.
Mol Cell ; 56(5): 613-4, 2014 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-25479635

RESUMEN

By using a combination of biochemistry and genetics, in this issue of Molecular CellIeva et al. (2014) uncover an unexpected role for Mgr2 of the mitochondrial TIM23 translocon as a gatekeeper in the release of membrane proteins from the translocon.


Asunto(s)
Proteínas de la Membrana/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Membranas Mitocondriales/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales
10.
Proc Natl Acad Sci U S A ; 114(33): 8770-8775, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28760994

RESUMEN

Fibrils and oligomers are the aggregated protein agents of neuronal dysfunction in ALS diseases. Whereas we now know much about fibril architecture, atomic structures of disease-related oligomers have eluded determination. Here, we determine the corkscrew-like structure of a cytotoxic segment of superoxide dismutase 1 (SOD1) in its oligomeric state. Mutations that prevent formation of this structure eliminate cytotoxicity of the segment in isolation as well as cytotoxicity of the ALS-linked mutants of SOD1 in primary motor neurons and in a Danio rerio (zebrafish) model of ALS. Cytotoxicity assays suggest that toxicity is a property of soluble oligomers, and not large insoluble aggregates. Our work adds to evidence that the toxic oligomeric entities in protein aggregation diseases contain antiparallel, out-of-register ß-sheet structures and identifies a target for structure-based therapeutics in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Superóxido Dismutasa-1/metabolismo , Esclerosis Amiotrófica Lateral/genética , Animales , Cristalografía por Rayos X/métodos , Ratones , Neuronas Motoras/metabolismo , Mutación/genética , Conformación Proteica en Lámina beta , Superóxido Dismutasa-1/genética
11.
J Biol Chem ; 293(51): 19797-19811, 2018 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-30366982

RESUMEN

Erythropoietin (EPO) signaling is critical to many processes essential to terminal erythropoiesis. Despite the centrality of iron metabolism to erythropoiesis, the mechanisms by which EPO regulates iron status are not well-understood. To this end, here we profiled gene expression in EPO-treated 32D pro-B cells and developing fetal liver erythroid cells to identify additional iron regulatory genes. We determined that FAM210B, a mitochondrial inner-membrane protein, is essential for hemoglobinization, proliferation, and enucleation during terminal erythroid maturation. Fam210b deficiency led to defects in mitochondrial iron uptake, heme synthesis, and iron-sulfur cluster formation. These defects were corrected with a lipid-soluble, small-molecule iron transporter, hinokitiol, in Fam210b-deficient murine erythroid cells and zebrafish morphants. Genetic complementation experiments revealed that FAM210B is not a mitochondrial iron transporter but is required for adequate mitochondrial iron import to sustain heme synthesis and iron-sulfur cluster formation during erythroid differentiation. FAM210B was also required for maximal ferrochelatase activity in differentiating erythroid cells. We propose that FAM210B functions as an adaptor protein that facilitates the formation of an oligomeric mitochondrial iron transport complex, required for the increase in iron acquisition for heme synthesis during terminal erythropoiesis. Collectively, our results reveal a critical mechanism by which EPO signaling regulates terminal erythropoiesis and iron metabolism.


Asunto(s)
Células Eritroides/metabolismo , Eritropoyetina/metabolismo , Ferroquelatasa/metabolismo , Hemo/biosíntesis , Hierro/metabolismo , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Animales , Células Eritroides/citología , Eritropoyesis , Células HEK293 , Humanos , Proteínas de la Membrana/química , Ratones , Membranas Mitocondriales/metabolismo , Proteínas Mitocondriales/química , Transporte de Proteínas
12.
Nat Chem Biol ; 13(12): 1239-1244, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28991239

RESUMEN

Tim17 and Tim23 are the main subunits of the TIM23 complex, one of the two major essential mitochondrial inner-membrane protein translocon machineries (TIMs). No chemical probes that specifically inhibit TIM23-dependent protein import were known to exist. Here we show that the natural product stendomycin, produced by Streptomyces hygroscopicus, is a potent and specific inhibitor of the TIM23 complex in yeast and mammalian cells. Furthermore, stendomycin-mediated blockage of the TIM23 complex does not alter normal processing of the major regulatory mitophagy kinase PINK1, but TIM23 is required to stabilize PINK1 on the outside of mitochondria to initiate mitophagy upon membrane depolarization.


Asunto(s)
Proteínas Mitocondriales/metabolismo , Péptidos/farmacología , Proteínas de Saccharomyces cerevisiae/antagonistas & inhibidores , Péptidos Catiónicos Antimicrobianos , Relación Dosis-Respuesta a Droga , Células HeLa , Humanos , Proteínas de Transporte de Membrana/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Estructura Molecular , Péptidos/química , Transporte de Proteínas/efectos de los fármacos , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Relación Estructura-Actividad , Células Tumorales Cultivadas
13.
J Biol Chem ; 292(13): 5429-5442, 2017 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-28167535

RESUMEN

Diverse protein import pathways into mitochondria use translocons on the outer membrane (TOM) and inner membrane (TIM). We adapted a genetic screen, based on Ura3 mistargeting from mitochondria to the cytosol, to identify small molecules that attenuated protein import. Small molecule mitochondrial import blockers of the Carla Koehler laboratory (MB)-10 inhibited import of substrates that require the TIM23 translocon. Mutational analysis coupled with molecular docking and molecular dynamics modeling revealed that MB-10 binds to a specific pocket in the C-terminal domain of Tim44 of the protein-associated motor (PAM) complex. This region was proposed to anchor Tim44 to the membrane, but biochemical studies with MB-10 show that this region is required for binding to the translocating precursor and binding to mtHsp70 in low ATP conditions. This study also supports a direct role for the PAM complex in the import of substrates that are laterally sorted to the inner membrane, as well as the mitochondrial matrix. Thus, MB-10 is the first small molecule modulator to attenuate PAM complex activity, likely through binding to the C-terminal region of Tim44.


Asunto(s)
Proteínas de Transporte de Membrana Mitocondrial/antagonistas & inhibidores , Proteínas de Saccharomyces cerevisiae/antagonistas & inhibidores , Saccharomyces cerevisiae/efectos de los fármacos , Animales , Sitios de Unión , Pruebas Genéticas , Células HeLa , Humanos , Proteínas de Transporte de Membrana Mitocondrial/química , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Modelos Moleculares , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Neurospora crassa , Transporte de Proteínas/efectos de los fármacos , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Pez Cebra
14.
Hum Mol Genet ; 25(9): 1754-70, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26908608

RESUMEN

The X-linked disease Barth syndrome (BTHS) is caused by mutations in TAZ; TAZ is the main determinant of the final acyl chain composition of the mitochondrial-specific phospholipid, cardiolipin. To date, a detailed characterization of endogenous TAZ has only been performed in yeast. Further, why a given BTHS-associated missense mutation impairs TAZ function has only been determined in a yeast model of this human disease. Presently, the detailed characterization of yeast tafazzin harboring individual BTHS mutations at evolutionarily conserved residues has identified seven distinct loss-of-function mechanisms caused by patient-associated missense alleles. However, whether the biochemical consequences associated with individual mutations also occur in the context of human TAZ in a validated mammalian model has not been demonstrated. Here, utilizing newly established monoclonal antibodies capable of detecting endogenous TAZ, we demonstrate that mammalian TAZ, like its yeast counterpart, is localized to the mitochondrion where it adopts an extremely protease-resistant fold, associates non-integrally with intermembrane space-facing membranes and assembles in a range of complexes. Even though multiple isoforms are expressed at the mRNA level, only a single polypeptide that co-migrates with the human isoform lacking exon 5 is expressed in human skin fibroblasts, HEK293 cells, and murine heart and liver mitochondria. Finally, using a new genome-edited mammalian BTHS cell culture model, we demonstrate that the loss-of-function mechanisms for two BTHS alleles that represent two of the seven functional classes of BTHS mutation as originally defined in yeast, are the same when modeled in human TAZ.


Asunto(s)
Síndrome de Barth/genética , Fibroblastos/metabolismo , Mitocondrias Cardíacas/metabolismo , Mitocondrias Hepáticas/metabolismo , Mutación/genética , Piel/metabolismo , Factores de Transcripción/metabolismo , Aciltransferasas , Animales , Síndrome de Barth/metabolismo , Síndrome de Barth/patología , Células Cultivadas , Fibroblastos/citología , Células HEK293 , Humanos , Ratones , Mitocondrias Cardíacas/patología , Mitocondrias Hepáticas/patología , Isoformas de Proteínas , Piel/citología , Factores de Transcripción/clasificación , Factores de Transcripción/genética
15.
Brain ; 139(11): 2877-2890, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27543974

RESUMEN

Disturbed mitochondrial fusion and fission have been linked to various neurodegenerative disorders. In siblings from two unrelated families who died soon after birth with a profound neurodevelopmental disorder characterized by pontocerebellar hypoplasia and apnoea, we discovered a missense mutation and an exonic deletion in the SLC25A46 gene encoding a mitochondrial protein recently implicated in optic atrophy spectrum disorder. We performed functional studies that confirmed the mitochondrial localization and pro-fission properties of SLC25A46. Knockdown of slc24a46 expression in zebrafish embryos caused brain malformation, spinal motor neuron loss, and poor motility. At the cellular level, we observed abnormally elongated mitochondria, which was rescued by co-injection of the wild-type but not the mutant slc25a46 mRNA. Conversely, overexpression of the wild-type protein led to mitochondrial fragmentation and disruption of the mitochondrial network. In contrast to mutations causing non-lethal optic atrophy, missense mutations causing lethal congenital pontocerebellar hypoplasia markedly destabilize the protein. Indeed, the clinical severity appears inversely correlated with the relative stability of the mutant protein. This genotype-phenotype correlation underscores the importance of SLC25A46 and fine tuning of mitochondrial fission and fusion in pontocerebellar hypoplasia and central neurodevelopment in addition to optic and peripheral neuropathy across the life span.


Asunto(s)
Enfermedades Cerebelosas/genética , Predisposición Genética a la Enfermedad/genética , Proteínas Mitocondriales/genética , Mutación/genética , Proteínas de Transporte de Fosfato/genética , Polimorfismo de Nucleótido Simple/genética , Aminoácidos/genética , Animales , Animales Modificados Genéticamente , Encéfalo/anomalías , Línea Celular Transformada , Células Cultivadas , Enfermedades Cerebelosas/diagnóstico por imagen , Estudios de Cohortes , Embrión no Mamífero , Femenino , Humanos , Lactante , Imagen por Resonancia Magnética , Masculino , Mitocondrias/metabolismo , Mitocondrias/patología , Dinámicas Mitocondriales/genética , Modelos Moleculares , Pez Cebra
16.
Proc Natl Acad Sci U S A ; 111(40): 14406-11, 2014 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-25237136

RESUMEN

Primary hyperoxaluria 1 (PH1; Online Mendelian Inheritance in Man no. 259900), a typically lethal biochemical disorder, may be caused by the AGT(P11LG170R) allele in which the alanine:glyoxylate aminotransferase (AGT) enzyme is mistargeted from peroxisomes to mitochondria. AGT contains a C-terminal peroxisomal targeting sequence, but mutations generate an N-terminal mitochondrial targeting sequence that directs AGT from peroxisomes to mitochondria. Although AGT(P11LG170R) is functional, the enzyme must be in the peroxisome to detoxify glyoxylate by conversion to alanine; in disease, amassed glyoxylate in the peroxisome is transported to the cytosol and converted to oxalate by lactate dehydrogenase, leading to kidney failure. From a chemical genetic screen, we have identified small molecules that inhibit mitochondrial protein import. We tested whether one promising candidate, Food and Drug Administration (FDA)-approved dequalinium chloride (DECA), could restore proper peroxisomal trafficking of AGT(P11LG170R). Indeed, treatment with DECA inhibited AGT(P11LG170R) translocation into mitochondria and subsequently restored trafficking to peroxisomes. Previous studies have suggested that a mitochondrial uncoupler might work in a similar manner. Although the uncoupler carbonyl cyanide m-chlorophenyl hydrazone inhibited AGT(P11LG170R) import into mitochondria, AGT(P11LG170R) aggregated in the cytosol, and cells subsequently died. In a cellular model system that recapitulated oxalate accumulation, exposure to DECA reduced oxalate accumulation, similar to pyridoxine treatment that works in a small subset of PH1 patients. Moreover, treatment with both DECA and pyridoxine was additive in reducing oxalate levels. Thus, repurposing the FDA-approved DECA may be a pharmacologic strategy to treat PH1 patients with mutations in AGT because an additional 75 missense mutations in AGT may also result in mistrafficking.


Asunto(s)
Decualinio/farmacología , Hiperoxaluria Primaria/metabolismo , Transaminasas/metabolismo , Animales , Antiinfecciosos Locales/farmacología , Células CHO , Cricetinae , Cricetulus , Evaluación Preclínica de Medicamentos/métodos , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/embriología , Humanos , Hiperoxaluria Primaria/genética , Hiperoxaluria Primaria/prevención & control , Immunoblotting , Microscopía Fluorescente , Mitocondrias/metabolismo , Mutación , Oxalatos/metabolismo , Peroxisomas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/genética , Piridoxina/farmacología , Transaminasas/genética , Pez Cebra/embriología
17.
Trends Biochem Sci ; 37(1): 32-41, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22014644

RESUMEN

Cardiolipin, the signature phospholipid of mitochondria, is a lipid dimer that is important for a diverse range of mitochondrial activities beyond the process of ATP production. Thus not surprisingly, derangements in cardiolipin metabolism are now appreciated to contribute to an assortment of pathological conditions. A comprehensive inventory of enzymes involved in cardiolipin biosynthesis and remodeling was just recently obtained. Post-biosynthesis, the acyl chain composition of cardiolipin is modified by up to three distinct remodeling enzymes that produce either a homogeneous tissue-specific mature form of cardiolipin or alternatively 'bad' cardiolipin that has been linked to mitochondrial dysfunction. In this review, we initially focus on the newly identified players in cardiolipin metabolism and then shift our attention to how changes in cardiolipin metabolism contribute to human disease.


Asunto(s)
Cardiolipinas/metabolismo , Enfermedad , Estado de Salud , Cardiolipinas/biosíntesis , Enzimas/metabolismo , Humanos
18.
J Biol Chem ; 290(34): 20804-20814, 2015 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-26085103

RESUMEN

A redox-regulated import pathway consisting of Mia40 and Erv1 mediates the import of cysteine-rich proteins into the mitochondrial intermembrane space. Mia40 is the oxidoreductase that inserts two disulfide bonds into the substrate simultaneously. However, Mia40 has one redox-active cysteine pair, resulting in ambiguity about how Mia40 accepts numerous electrons during substrate oxidation. In this study, we have addressed the oxidation of Tim13 in vitro and in organello. Reductants such as glutathione and ascorbate inhibited both the oxidation of the substrate Tim13 in vitro and the import of Tim13 and Cmc1 into isolated mitochondria. In addition, a ternary complex consisting of Erv1, Mia40, and substrate, linked by disulfide bonds, was not detected in vitro. Instead, Mia40 accepted six electrons from substrates, and this fully reduced Mia40 was sensitive to protease, indicative of conformational changes in the structure. Mia40 in mitochondria from the erv1-101 mutant was also trapped in a completely reduced state, demonstrating that Mia40 can accept up to six electrons as substrates are imported. Therefore, these studies support that Mia40 functions as an electron sink to facilitate the insertion of two disulfide bonds into substrates.


Asunto(s)
Electrones , Regulación Fúngica de la Expresión Génica , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Proteínas Mitocondriales/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Ácido Ascórbico/farmacología , Disulfuros/química , Disulfuros/metabolismo , Glutatión/farmacología , Metalochaperonas/genética , Metalochaperonas/metabolismo , Mitocondrias/efectos de los fármacos , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Proteínas Mitocondriales/genética , Mutación , Oxidación-Reducción , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/genética , Plásmidos/química , Plásmidos/metabolismo , Transporte de Proteínas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sustancias Reductoras/farmacología , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Transducción de Señal
19.
Am J Hum Genet ; 93(5): 906-14, 2013 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-24119684

RESUMEN

We used exome sequencing to identify mutations in sideroflexin 4 (SFXN4) in two children with mitochondrial disease (the more severe case also presented with macrocytic anemia). SFXN4 is an uncharacterized mitochondrial protein that localizes to the mitochondrial inner membrane. sfxn4 knockdown in zebrafish recapitulated the mitochondrial respiratory defect observed in both individuals and the macrocytic anemia with megaloblastic features of the more severe case. In vitro and in vivo complementation studies with fibroblasts from the affected individuals and zebrafish demonstrated the requirement of SFXN4 for mitochondrial respiratory homeostasis and erythropoiesis. Our findings establish mutations in SFXN4 as a cause of mitochondriopathy and macrocytic anemia.


Asunto(s)
Anemia Macrocítica/genética , Proteínas de la Membrana/genética , Enfermedades Mitocondriales/genética , Adolescente , Animales , Niño , Eritropoyesis/genética , Exoma , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Proteínas Mitocondriales/genética , Mutación , Pez Cebra/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA