Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 610(7932): 555-561, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36171294

RESUMEN

CD4+ T cell differentiation requires metabolic reprogramming to fulfil the bioenergetic demands of proliferation and effector function, and enforce specific transcriptional programmes1-3. Mitochondrial membrane dynamics sustains mitochondrial processes4, including respiration and tricarboxylic acid (TCA) cycle metabolism5, but whether mitochondrial membrane remodelling orchestrates CD4+ T cell differentiation remains unclear. Here we show that unlike other CD4+ T cell subsets, T helper 17 (TH17) cells have fused mitochondria with tight cristae. T cell-specific deletion of optic atrophy 1 (OPA1), which regulates inner mitochondrial membrane fusion and cristae morphology6, revealed that TH17 cells require OPA1 for its control of the TCA cycle, rather than respiration. OPA1 deletion amplifies glutamine oxidation, leading to impaired NADH/NAD+ balance and accumulation of TCA cycle metabolites and 2-hydroxyglutarate-a metabolite that influences the epigenetic landscape5,7. Our multi-omics approach revealed that the serine/threonine kinase liver-associated kinase B1 (LKB1) couples mitochondrial function to cytokine expression in TH17 cells by regulating TCA cycle metabolism and transcriptional remodelling. Mitochondrial membrane disruption activates LKB1, which restrains IL-17 expression. LKB1 deletion restores IL-17 expression in TH17 cells with disrupted mitochondrial membranes, rectifying aberrant TCA cycle glutamine flux, balancing NADH/NAD+ and preventing 2-hydroxyglutarate production from the promiscuous activity of the serine biosynthesis enzyme phosphoglycerate dehydrogenase (PHGDH). These findings identify OPA1 as a major determinant of TH17 cell function, and uncover LKB1 as a sensor linking mitochondrial cues to effector programmes in TH17 cells.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Mitocondrias , Células Th17 , Glutamina/metabolismo , Interleucina-17/metabolismo , Mitocondrias/metabolismo , NAD/metabolismo , Fosfoglicerato-Deshidrogenasa/metabolismo , Serina/biosíntesis , Serina/metabolismo , Células Th17/citología , Células Th17/inmunología , Células Th17/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Ciclo del Ácido Cítrico , GTP Fosfohidrolasas/deficiencia , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo
2.
J Lipid Res ; 65(6): 100563, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38763493

RESUMEN

Depletion or mutations of key proteins for mitochondrial fusion, like optic atrophy 1 (OPA1) and mitofusins 1 and 2 (Mfn 1 and 2), are known to significantly impact the mitochondrial ultrastructure, suggesting alterations of their membranes' lipid profiles. In order to make an insight into this issue, we used hydrophilic interaction liquid chromatography coupled with electrospray ionization-high resolution MS to investigate the mitochondrial phospholipid (PL) profile of mouse embryonic fibroblasts knocked out for OPA1 and Mfn1/2 genes. One hundred sixty-seven different sum compositions were recognized for the four major PL classes of mitochondria, namely phosphatidylcholines (PCs, 63), phosphatidylethanolamines (55), phosphatidylinositols (21), and cardiolipins (28). A slight decrease in the cardiolipin/PC ratio was found for Mfn1/2-knockout mitochondria. Principal component analysis and hierarchical cluster analysis were subsequently used to further process hydrophilic interaction liquid chromatography-ESI-MS data. A progressive decrease in the incidence of alk(en)yl/acyl species in PC and phosphatidylethanolamine classes and a general increase in the incidence of unsaturated acyl chains across all the investigated PL classes was inferred in OPA1 and Mfn1/2 knockouts compared to WT mouse embryonic fibroblasts. These findings suggest a reshaping of the PL profile consistent with the changes observed in the mitochondrial ultrastructure when fusion proteins are absent. Based on the existing knowledge on the metabolism of mitochondrial phospholipids, we propose that fusion proteins, especially Mfns, might influence the PL transfer between the mitochondria and the endoplasmic reticulum, likely in the context of mitochondria-associated membranes.


Asunto(s)
GTP Fosfohidrolasas , Lipidómica , Mitocondrias , Fosfolípidos , Animales , GTP Fosfohidrolasas/metabolismo , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/deficiencia , Ratones , Mitocondrias/metabolismo , Fosfolípidos/metabolismo , Ratones Noqueados , Fibroblastos/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/genética
3.
EMBO J ; 36(14): 2126-2145, 2017 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-28607005

RESUMEN

Mitochondrial dynamics is a conserved process by which mitochondria undergo repeated cycles of fusion and fission, leading to exchange of mitochondrial genetic content, ions, metabolites, and proteins. Here, we examine the role of the mitochondrial fusion protein optic atrophy 1 (OPA1) in differentiated skeletal muscle by reducing OPA1 gene expression in an inducible manner. OPA1 deficiency in young mice results in non-lethal progressive mitochondrial dysfunction and loss of muscle mass. Mutant mice are resistant to age- and diet-induced weight gain and insulin resistance, by mechanisms that involve activation of ER stress and secretion of fibroblast growth factor 21 (FGF21) from skeletal muscle, resulting in increased metabolic rates and improved whole-body insulin sensitivity. OPA1-elicited mitochondrial dysfunction activates an integrated stress response that locally induces muscle atrophy, but via secretion of FGF21 acts distally to modulate whole-body metabolism.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , GTP Fosfohidrolasas/metabolismo , Resistencia a la Insulina , Músculos/metabolismo , Atrofia Muscular/patología , Obesidad/prevención & control , Animales , GTP Fosfohidrolasas/deficiencia , Técnicas de Silenciamiento del Gen , Ratones
4.
EMBO J ; 36(11): 1543-1558, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28348166

RESUMEN

Mitochondrial fusion and fission events, collectively known as mitochondrial dynamics, act as quality control mechanisms to ensure mitochondrial function and fine-tune cellular bioenergetics. Defective mitofusin 2 (Mfn2) expression and enhanced mitochondrial fission in skeletal muscle are hallmarks of insulin-resistant states. Interestingly, Mfn2 is highly expressed in brown adipose tissue (BAT), yet its role remains unexplored. Using adipose-specific Mfn2 knockout (Mfn2-adKO) mice, we demonstrate that Mfn2, but not Mfn1, deficiency in BAT leads to a profound BAT dysfunction, associated with impaired respiratory capacity and a blunted response to adrenergic stimuli. Importantly, Mfn2 directly interacts with perilipin 1, facilitating the interaction between the mitochondria and the lipid droplet in response to adrenergic stimulation. Surprisingly, Mfn2-adKO mice were protected from high-fat diet-induced insulin resistance and hepatic steatosis. Altogether, these results demonstrate that Mfn2 is a mediator of mitochondria to lipid droplet interactions, influencing lipolytic processes and whole-body energy homeostasis.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , GTP Fosfohidrolasas/metabolismo , Mitocondrias/metabolismo , Termogénesis , Animales , GTP Fosfohidrolasas/deficiencia , Ratones , Ratones Noqueados , Perilipina-1/metabolismo , Unión Proteica
5.
J Virol ; 94(21)2020 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-32796066

RESUMEN

Rabies, caused by rabies virus (RABV), is an ancient zoonosis and still a major public health problem for humans, especially in developing countries. RABV can be recognized by specific innate recognition receptors, resulting in the production of hundreds of interferon-stimulated genes (ISGs), which can inhibit viral replication at different stages. Interferon-inducible GTPase 1 (IIGP1) is a mouse-specific ISG and belongs to the immunity-related GTPases (IRGs) family. IIGP is reported to constrain intracellular parasite infection by disrupting the parasitophorous vacuole membrane. However, the role of IIGP1 in restricting viral replication has not been reported. In this present study, we found that IIGP1 was upregulated in cells and mouse brains upon RABV infection. Overexpression of IIGP1 limited RABV replication in cell lines and reduced viral pathogenicity in a mouse model. Consistently, deficiency of IIGP1 enhanced RABV replication in different parts of mouse brains. Furthermore, we found that IIGP1 could interact with RABV phosphoprotein (P protein). Mutation and immunoprecipitation analyses revealed that the Y128 site of P protein is critical for its interaction with IIGP1. Further study demonstrated that this interaction impeded the dimerization of P protein and thus suppressed RABV replication. Collectively, our findings for the first reveal a novel role of IIGP1 in restricting a typical neurotropic virus, RABV, which will provide fresh insight into the function of this mouse-specific ISG.IMPORTANCE Interferon and its downstream products, ISGs, are essential in defending against pathogen invasion. One of the ISGs, IIGP1, has been found to constrain intracellular parasite infection by disrupting their vacuole membranes. However, the role of IIGP1 in limiting viral infection is unclear. In this study, we show that infection with a typical neurotropic virus, RABV, can induce upregulation of IIGP1, which, in turn, suppresses RABV by interacting with its phosphoprotein (P protein) and thus blocking the dimerization of P protein. Our study provides the first evidence that IIGP1 functions in limiting viral infection and provides a basis for comprehensive understanding of this important ISG.


Asunto(s)
GTP Fosfohidrolasas/genética , Fosfoproteínas/genética , Virus de la Rabia/genética , Rabia/genética , Proteínas Virales/genética , Replicación Viral/genética , Animales , Línea Celular Tumoral , Femenino , GTP Fosfohidrolasas/deficiencia , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Interacciones Huésped-Patógeno/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuroglía/metabolismo , Neuroglía/virología , Neuronas/metabolismo , Neuronas/virología , Fosfoproteínas/metabolismo , Multimerización de Proteína , Rabia/mortalidad , Rabia/patología , Rabia/virología , Virus de la Rabia/crecimiento & desarrollo , Virus de la Rabia/patogenicidad , Transducción de Señal , Análisis de Supervivencia , Proteínas Virales/metabolismo
6.
J Nanobiotechnology ; 19(1): 425, 2021 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-34922554

RESUMEN

BACKGROUND: Lipoplexes are non-viral vectors based on cationic lipids used to deliver DNA into cells, also known as lipofection. The positively charge of the hydrophilic head-group provides the cationic lipids the ability to condensate the negatively charged DNA into structured complexes. The polar head can carry a large variety of chemical groups including amines as well as guanidino or imidazole groups. In particular, gemini cationic lipids consist of two positive polar heads linked by a spacer with different length. As for the hydrophobic aliphatic chains, they can be unsaturated or saturated and are connected to the polar head-groups. Many other chemical components can be included in the formulation of lipoplexes to improve their transfection efficiency, which often relies on their structural features. Varying these components can drastically change the arrangement of DNA molecules within the lamellar, hexagonal or cubic phases that are provided by the lipid matrix. Lipofection is widely used to deliver genetic material in cell culture experiments but the simpler formulations exhibit major drawbacks related to low transfection, low specificity, low circulation half-life and toxicity when scaled up to in vivo experiments. RESULTS: So far, we have explored in cell cultures the transfection ability of lipoplexes based on gemini cationic lipids that consist of two C16 alkyl chains and two imidazolium polar head-groups linked with a polyoxyethylene spacer, (C16Im)2(C4O). Here, PEGylated lipids have been introduced to the lipoplex formulation and the transgene expression of the Opa1 mitochondrial transmembrane protein in mice was assessed. The addition of PEG on the surface of the lipid mixed resulted in the formation of Ia3d bicontinuous cubic phases as determined by small angle X-ray scattering. After a single intramuscular administration, the cubic lipoplexes were accumulated in tissues with tight endothelial barriers such as brain, heart, and lungs for at least 48 h. The transgene expression of Opa1 in those organs was identified by western blotting or RNA expression analysis through quantitative polymerase chain reaction. CONCLUSIONS: The expression reported here is sufficient in magnitude, duration and toxicity to consolidate the bicontinuous cubic structures formed by (C16Im)2(C4O)-based lipoplexes as valuable therapeutic agents in the field of gene delivery.


Asunto(s)
GTP Fosfohidrolasas/genética , Imidazoles/química , Liposomas/química , Tensoactivos/química , Transfección/métodos , Animales , Encéfalo/metabolismo , Cationes/química , Línea Celular , Supervivencia Celular/efectos de los fármacos , GTP Fosfohidrolasas/deficiencia , GTP Fosfohidrolasas/metabolismo , Riñón/metabolismo , Liposomas/farmacocinética , Liposomas/farmacología , Ratones , Plásmidos/química , Plásmidos/genética , Plásmidos/metabolismo , Polietilenglicoles/química , Distribución Tisular
7.
Nucleic Acids Res ; 47(8): 4086-4110, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-30986824

RESUMEN

Ataxia with oculomotor apraxia type 1 (AOA1) is an early onset progressive spinocerebellar ataxia caused by mutation in aprataxin (APTX). APTX removes 5'-AMP groups from DNA, a product of abortive ligation during DNA repair and replication. APTX deficiency has been suggested to compromise mitochondrial function; however, a detailed characterization of mitochondrial homeostasis in APTX-deficient cells is not available. Here, we show that cells lacking APTX undergo mitochondrial stress and display significant changes in the expression of the mitochondrial inner membrane fusion protein optic atrophy type 1, and components of the oxidative phosphorylation complexes. At the cellular level, APTX deficiency impairs mitochondrial morphology and network formation, and autophagic removal of damaged mitochondria by mitophagy. Thus, our results show that aberrant mitochondrial function is a key component of AOA1 pathology. This work corroborates the emerging evidence that impaired mitochondrial function is a characteristic of an increasing number of genetically diverse neurodegenerative disorders.


Asunto(s)
Proteínas de Unión al ADN/genética , GTP Fosfohidrolasas/genética , Mitocondrias/genética , Mitofagia/genética , Proteínas Nucleares/genética , Ataxias Espinocerebelosas/congénito , Línea Celular Transformada , Línea Celular Tumoral , Proteínas de Unión al ADN/deficiencia , Proteínas del Complejo de Cadena de Transporte de Electrón/genética , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , GTP Fosfohidrolasas/deficiencia , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Homeostasis/genética , Humanos , Linfocitos/metabolismo , Linfocitos/patología , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Proteínas Nucleares/deficiencia , Análisis de Secuencia por Matrices de Oligonucleótidos , Osteoblastos/metabolismo , Osteoblastos/patología , Fosforilación Oxidativa , Transducción de Señal , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/metabolismo , Ataxias Espinocerebelosas/patología
8.
BMC Med Genet ; 21(1): 236, 2020 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-33243194

RESUMEN

BACKGROUND: Dominant optic atrophy (DOA) is an inherited optic neuropathy that mainly affects visual acuity, central visual fields and color vision due to a progressive loss of retinal ganglion cells and their axons that form the optic nerve. Approximately 45-90% of affected individuals with DOA harbor pathogenic variants in the OPA1 gene. The mutation spectrum of OPA1 comprises nonsense, canonical and non-canonical splice site, frameshift and missense as well as copy number variants, but intragenic inversions have not been reported so far. CASE PRESENTATION: We report a 33-year-old male with characteristic clinical features of DOA. Whole-genome sequencing identified a structural variant of 2.4 kb comprising an inversion of 937 bp at the OPA1 locus. Fine mapping of the breakpoints to single nucleotide level revealed that the structural variation was an inversion flanked by two deletions. As this rearrangement inverts the entire first exon of OPA1, it was classified as likely pathogenic. CONCLUSIONS: We report the first DOA case harboring an inversion in the OPA1 gene. Our study demonstrates that copy-neutral genomic rearrangements have to be considered as a possible cause of disease in DOA cases.


Asunto(s)
GTP Fosfohidrolasas/genética , Atrofia Óptica Autosómica Dominante/genética , Inversión de Secuencia , Adulto , Axones , Secuencia de Bases , GTP Fosfohidrolasas/deficiencia , Expresión Génica , Humanos , Masculino , Atrofia Óptica Autosómica Dominante/diagnóstico , Atrofia Óptica Autosómica Dominante/patología , Tomografía de Coherencia Óptica , Secuenciación Completa del Genoma
9.
Proc Natl Acad Sci U S A ; 114(5): 980-985, 2017 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-28096346

RESUMEN

The physiological role of LepA, a paralog of EF-G found in all bacteria, has been a mystery for decades. Here, we show that LepA functions in ribosome biogenesis. In cells lacking LepA, immature 30S particles accumulate. Four proteins are specifically underrepresented in these particles-S3, S10, S14, and S21-all of which bind late in the assembly process and contribute to the folding of the 3' domain of 16S rRNA. Processing of 16S rRNA is also delayed in the mutant strain, as indicated by increased levels of precursor 17S rRNA in assembly intermediates. Mutation ΔlepA confers a synthetic growth phenotype in absence of RsgA, another GTPase, well known to act in 30S subunit assembly. Analysis of the ΔrsgA strain reveals accumulation of intermediates that resemble those seen in the absence of LepA. These data suggest that RsgA and LepA play partially redundant roles to ensure efficient 30S assembly.


Asunto(s)
Proteínas de Escherichia coli/fisiología , Escherichia coli/metabolismo , Biogénesis de Organelos , Factores de Iniciación de Péptidos/fisiología , Subunidades Ribosómicas Pequeñas Bacterianas/metabolismo , Ribosomas/metabolismo , Proteínas Bacterianas/metabolismo , Escherichia coli/genética , Escherichia coli/crecimiento & desarrollo , Proteínas de Escherichia coli/genética , GTP Fosfohidrolasas/deficiencia , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/fisiología , Modelos Moleculares , Factores de Iniciación de Péptidos/deficiencia , Factores de Iniciación de Péptidos/genética , Conformación Proteica , Precursores del ARN/metabolismo , ARN Bacteriano/metabolismo , ARN Ribosómico 16S/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Ribosómicas/metabolismo
10.
Proc Natl Acad Sci U S A ; 114(5): 1183-1188, 2017 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-28096359

RESUMEN

The abnormal aggregation of fibrillar α-synuclein in Lewy bodies plays a critical role in the pathogenesis of Parkinson's disease. However, the molecular mechanisms regulating α-synuclein pathological effects are incompletely understood. Here we show that α-synuclein binds phosphoinositide-3 kinase enhancer L (PIKE-L) in a phosphorylation-dependent manner and sequesters it in Lewy bodies, leading to dopaminergic cell death via AMP-activated protein kinase (AMPK) hyperactivation. α-Synuclein interacts with PIKE-L, an AMPK inhibitory binding partner, and this action is increased by S129 phosphorylation through AMPK and is decreased by Y125 phosphorylation via Src family kinase Fyn. A pleckstrin homology (PH) domain in PIKE-L directly binds α-synuclein and antagonizes its aggregation. Accordingly, PIKE-L overexpression decreases dopaminergic cell death elicited by 1-methyl-4-phenylpyridinium (MPP+), whereas PIKE-L knockdown elevates α-synuclein oligomerization and cell death. The overexpression of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or α-synuclein induces greater dopaminergic cell loss and more severe motor defects in PIKE-KO and Fyn-KO mice than in wild-type mice, and these effects are attenuated by the expression of dominant-negative AMPK. Hence, our findings demonstrate that α-synuclein neutralizes PIKE-L's neuroprotective actions in synucleinopathies, triggering dopaminergic neuronal death by hyperactivating AMPK.


Asunto(s)
Adenilato Quinasa/metabolismo , Neuronas Dopaminérgicas/metabolismo , Proteínas de Unión al GTP/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Cuerpos de Lewy/metabolismo , alfa-Sinucleína/metabolismo , 1-Metil-4-fenilpiridinio/toxicidad , Anciano , Anciano de 80 o más Años , Animales , Muerte Celular , Neuronas Dopaminérgicas/ultraestructura , Activación Enzimática , GTP Fosfohidrolasas/deficiencia , Proteínas de Unión al GTP/química , Proteínas Activadoras de GTPasa/química , Humanos , Intoxicación por MPTP/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/deficiencia , Fosforilación , Dominios Homólogos a Pleckstrina , Agregación Patológica de Proteínas , Unión Proteica , Mapeo de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-fyn/deficiencia , Proteínas Proto-Oncogénicas c-fyn/metabolismo
11.
J Allergy Clin Immunol ; 143(1): 245-257.e6, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30616774

RESUMEN

BACKGROUND: GTPase of immunity-associated protein 5 (GIMAP5) is essential for lymphocyte homeostasis and survival. Recently, human GIMAP5 single nucleotide polymorphisms have been linked to an increased risk for asthma, whereas loss of Gimap5 in mice has been associated with severe CD4+ T cell-driven immune pathology. OBJECTIVE: We sought to identify the molecular and cellular mechanisms by which Gimap5 deficiency predisposes to allergic airway disease. METHODS: CD4+ T-cell polarization and development of pathogenic CD4+ T cells were assessed in Gimap5-deficient mice and a human patient with a GIMAP5 loss-of-function (LOF) mutation. House dust mite-induced airway inflammation was assessed by using a complete Gimap5 LOF (Gimap5sph/sph) and conditional Gimap5fl/flCd4Cre/ert2 mice. RESULTS: GIMAP5 LOF mutations in both mice and human subjects are associated with spontaneous polarization toward pathogenic TH17 and TH2 cells in vivo. Mechanistic studies in vitro reveal that impairment of Gimap5-deficient TH cell differentiation is associated with increased DNA damage, particularly during TH1-polarizing conditions. DNA damage in Gimap5-deficient CD4+ T cells could be controlled by TGF-ß, thereby promoting TH17 polarization. When challenged with house dust mite in vivo, Gimap5-deficient mice displayed an exacerbated asthma phenotype (inflammation and airway hyperresponsiveness), with increased development of TH2, TH17, and pathogenic TH17/TH2 cells. CONCLUSION: Activation of Gimap5-deficient CD4+ T cells is associated with increased DNA damage and reduced survival that can be overcome by TGF-ß. This leads to selective survival of pathogenic TH17 cells but also TH2 cells in human subjects and mice, ultimately promoting allergic airway disease.


Asunto(s)
Asma/inmunología , GTP Fosfohidrolasas/deficiencia , Mutación con Pérdida de Función , Células Th17/inmunología , Células Th2/inmunología , Animales , Asma/genética , Asma/patología , GTP Fosfohidrolasas/inmunología , Proteínas de Unión al GTP , Humanos , Ratones , Ratones Transgénicos , Células Th17/patología , Células Th2/patología , Factor de Crecimiento Transformador beta/genética
12.
EMBO Rep ; 18(7): 1123-1138, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28539390

RESUMEN

BAT-controlled thermogenic activity is thought to be required for its capacity to prevent the development of insulin resistance. This hypothesis predicts that mediators of thermogenesis may help prevent diet-induced insulin resistance. We report that the mitochondrial fusion protein Mitofusin 2 (Mfn2) in BAT is essential for cold-stimulated thermogenesis, but promotes insulin resistance in obese mice. Mfn2 deletion in mice through Ucp1-cre (BAT-Mfn2-KO) causes BAT lipohypertrophy and cold intolerance. Surprisingly however, deletion of Mfn2 in mice fed a high fat diet (HFD) results in improved insulin sensitivity and resistance to obesity, while impaired cold-stimulated thermogenesis is maintained. Improvement in insulin sensitivity is associated with a gender-specific remodeling of BAT mitochondrial function. In females, BAT mitochondria increase their efficiency for ATP-synthesizing fat oxidation, whereas in BAT from males, complex I-driven respiration is decreased and glycolytic capacity is increased. Thus, BAT adaptation to obesity is regulated by Mfn2 and with BAT-Mfn2 absent, BAT contribution to prevention of insulin resistance is independent and inversely correlated to whole-body cold-stimulated thermogenesis.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , GTP Fosfohidrolasas/deficiencia , GTP Fosfohidrolasas/genética , Resistencia a la Insulina , Termogénesis/genética , Animales , Dieta Alta en Grasa , Metabolismo Energético , Femenino , Glucólisis , Masculino , Ratones , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Obesidad
13.
Cell Mol Life Sci ; 74(7): 1347-1363, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27858084

RESUMEN

As an organellar network, mitochondria dynamically regulate their organization via opposing fusion and fission pathways to maintain bioenergetic homeostasis and contribute to key cellular pathways. This dynamic balance is directly linked to bioenergetic function: loss of transmembrane potential across the inner membrane (Δψ m) disrupts mitochondrial fission/fusion balance, causing fragmentation of the network. However, the level of Δψ m required for mitochondrial dynamic balance, as well as the relative contributions of fission and fusion pathways, have remained unclear. To explore this, mitochondrial morphology and Δψ m were examined via confocal imaging and tetramethyl rhodamine ester (TMRE) flow cytometry, respectively, in cultured 143B osteosarcoma cells. When normalized to the TMRE value of untreated 143B cells as 100%, both genetic (mtDNA-depleted ρ0) and pharmacological [carbonyl cyanide m-chlorophenyl hydrazone (CCCP)-treated] cell models below 34% TMRE fluorescence were unable to maintain mitochondrial interconnection, correlating with loss of fusion-active long OPA1 isoforms (L-OPA1). Mechanistically, this threshold is maintained by mechanistic coordination of DRP1-mediated fission and OPA1-mediated fusion: cells lacking either DRP1 or the OMA1 metalloprotease were insensitive to loss of Δψ m, instead maintaining an obligately fused morphology. Collectively, these findings demonstrate a mitochondrial 'tipping point' threshold mediated by the interaction of Δψ m with both DRP1 and OMA1; moreover, DRP1 appears to be required for effective OPA1 maintenance and processing, consistent with growing evidence for direct interaction of fission and fusion pathways. These results suggest that Δψ m below threshold coordinately activates both DRP1-mediated fission and OMA1 cleavage of OPA1, collapsing mitochondrial dynamic balance, with major implications for a range of signaling pathways and cellular life/death events.


Asunto(s)
GTP Fosfohidrolasas/metabolismo , Metaloproteasas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias/fisiología , Dinámicas Mitocondriales , Proteínas Mitocondriales/metabolismo , Animales , Carbonil Cianuro m-Clorofenil Hidrazona/farmacología , Línea Celular Tumoral , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Dinaminas , GTP Fosfohidrolasas/deficiencia , GTP Fosfohidrolasas/genética , Células HCT116 , Humanos , Potenciales de la Membrana/efectos de los fármacos , Metaloproteasas/deficiencia , Metaloproteasas/genética , Ratones Noqueados , Microscopía Fluorescente , Proteínas Asociadas a Microtúbulos/deficiencia , Proteínas Asociadas a Microtúbulos/genética , Mitocondrias/química , Mitocondrias/genética , Dinámicas Mitocondriales/efectos de los fármacos , Proteínas Mitocondriales/deficiencia , Proteínas Mitocondriales/genética , Reacción en Cadena de la Polimerasa
14.
Biochim Biophys Acta ; 1857(8): 1277-1283, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27060252

RESUMEN

Mitochondria are bioenergetic hotspots, producing the bulk of ATP by the oxidative phosphorylation process. Mitochondria are also structurally dynamic and undergo coordinated fusion and fission to maintain their function. Recent studies of the mitochondrial fusion machinery have provided new evidence in detailing their role in mitochondrial metabolism. Remarkably, mitofusin 2, in addition to its role in fusion, is important for maintaining coenzyme Q levels and may be an integral player in the mevalonate synthesis pathway. Here, we review the bioenergetic roles of mitochondrial dynamics and emphasize the importance of the in vitro growth conditions when evaluating mitochondrial respiration. This article is part of a Special Issue entitled 'EBEC 2016: 19th European Bioenergetics Conference, Riva del Garda, Italy, July 2-6, 2016,' edited by Prof. Paolo Bernardi.


Asunto(s)
GTP Fosfohidrolasas/genética , Ácido Mevalónico/metabolismo , Mitocondrias/metabolismo , Dinámicas Mitocondriales/genética , Fosforilación Oxidativa , Ubiquinona/metabolismo , Animales , Línea Celular Transformada , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , GTP Fosfohidrolasas/deficiencia , GTP Fosfohidrolasas/metabolismo , Expresión Génica , Genoma Mitocondrial , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Noqueados , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
15.
Biochim Biophys Acta ; 1857(8): 1307-1312, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26992930

RESUMEN

Parkin is familiar to many because of its link to Parkinson's disease, and to others because of its well-characterized role as a central factor mediating selective mitophagy of damaged mitochondria for mitochondrial quality control. The genetic connection between Parkin and Parkinson's disease derives from clinical gene-association studies, whereas our mechanistic understanding of Parkin functioning in mitophagy is based almost entirely on work performed in cultured cells. Surprisingly, experimental evidence linking the disease and the presumed mechanism derives almost entirely from fruit flies; germline Parkin deficient mice do not develop Parkinson's disease phenotypes. Moreover, genetic manipulation of Parkin signaling in mouse hearts does not support a central role for Parkin in homeostatic mitochondrial quality control in this mitochondria-rich and -dependent organ. Here, I provide an overview of data suggesting that (in mouse hearts at least) Parkin functions more as a stress-induced and developmentally-programmed facilitator of cardiomyocyte mitochondrial turnover. This article is part of a Special Issue entitled 'EBEC 2016: 19th European Bioenergetics Conference, Riva del Garda, Italy, July 2-6, 2016.


Asunto(s)
Dinaminas/genética , Mitocondrias Cardíacas/metabolismo , Infarto del Miocardio/genética , Miocardio/metabolismo , Proteínas Quinasas/genética , Ubiquitina-Proteína Ligasas/genética , Animales , Modelos Animales de Enfermedad , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Dinaminas/deficiencia , GTP Fosfohidrolasas/deficiencia , GTP Fosfohidrolasas/genética , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Noqueados , Mitocondrias Cardíacas/patología , Proteínas Mitocondriales/deficiencia , Proteínas Mitocondriales/genética , Mitofagia/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Proteínas Quinasas/deficiencia , Transducción de Señal , Ubiquitina-Proteína Ligasas/deficiencia
16.
J Pediatr ; 171: 313-6.e1-2, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26825290

RESUMEN

We describe two infants with hypotonia, absent respiratory effort, and giant mitochondria in neurons due to compound heterozygosity for 2 nonsense mutations of DNM1L. DNM1L has a critical role in regulating mitochondrial morphology and function. This observation confirms the central role of mitochondrial fission to normal human development.


Asunto(s)
GTP Fosfohidrolasas/genética , Proteínas Asociadas a Microtúbulos/genética , Enfermedades Mitocondriales/genética , Dinámicas Mitocondriales , Proteínas Mitocondriales/genética , Mutación , Codón sin Sentido , Análisis Mutacional de ADN , Dinaminas , Exoma , Salud de la Familia , Resultado Fatal , Femenino , Forminas , GTP Fosfohidrolasas/deficiencia , Heterocigoto , Humanos , Recién Nacido , Masculino , Proteínas de Microfilamentos/genética , Microscopía Fluorescente , Proteínas Asociadas a Microtúbulos/deficiencia , Proteínas Mitocondriales/deficiencia , Linaje
17.
Tumour Biol ; 37(7): 8643-55, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26733181

RESUMEN

Mitofusin-2 (MFN2) was initially identified as a hyperplasia suppressor in hyper-proliferative vascular smooth muscle cells (VSMCs) of hypertensive rat arteries, which has also been implicated in various cancers. There exists a controversy in whether it is an oncogene or exerting anti-proliferative effect on tumor cells. Our previous cell cycle analysis and MTT assay showed that cell proliferation was inhibited in MFN2 deficient A549 human lung adenocarcinoma cells, without investigating the changes in regulatory network or addressing the underlying mechanisms. Here, we performed expression profiling in MFN2 knockdown A549 cells and found that cancer-related pathways were among the most susceptible pathways to MFN2 deficiency. Through comparison with expression profiling of a cohort consisting of 61 pairs of tumor-normal matched samples from The Cancer Genome Atlas (TCGA), we teased out the specific pathways to address the impact that MFN2 ablation had on A549 cells, as well as identified a few genes whose expression level associated with clinicopathologic parameters. In addition, transcriptional factor target enrichment analysis identified E2F as a potential transcription factor that was deregulated in response to MFN2 deficiency. Although bioinformatics analysis usually entail further verification, our study provided considerable information for future scientific inquiries in related areas as well as a paradigm for characterizing perturbation in regulatory network.


Asunto(s)
Biomarcadores de Tumor/genética , Proliferación Celular/genética , GTP Fosfohidrolasas/deficiencia , Perfilación de la Expresión Génica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Proteínas Mitocondriales/deficiencia , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Apoptosis , Estudios de Casos y Controles , Biología Computacional , GTP Fosfohidrolasas/genética , Redes Reguladoras de Genes , Humanos , Pulmón/metabolismo , Pulmón/patología , Proteínas Mitocondriales/genética , Ratas , Factores de Riesgo , Células Tumorales Cultivadas
18.
Biochim Biophys Acta ; 1843(7): 1295-307, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24709290

RESUMEN

Omi/HtrA2 is a nuclear encoded mitochondrial serine protease with dual and opposite functions that depend entirely on its subcellular localization. During apoptosis, Omi/HtrA2 is released into the cytoplasm where it participates in cell death. While confined in the inter-membrane space of the mitochondria, Omi/HtrA2 has a pro-survival function that may involve the regulation of protein quality control (PQC) and mitochondrial homeostasis. Loss of Omi/HtrA2's protease activity causes the neuromuscular disorder of the mnd2 (motor neuron degeneration 2) mutant mice. These mice develop multiple defects including neurodegeneration with parkinsonian features. Loss of Omi/HtrA2 in non-neuronal tissues has also been shown to cause premature aging. The normal function of Omi/HtrA2 in the mitochondria and how its deregulation causes neurodegeneration or premature aging are unknown. Here we report that the mitochondrial Mulan E3 ubiquitin ligase is a specific substrate of Omi/HtrA2. During exposure to H(2)O(2), Omi/HtrA2 degrades Mulan, and this regulation is lost in cells that carry the inactive protease. Furthermore, we show accumulation of Mulan protein in various tissues of mnd2 mice as well as in Omi/HtrA2(-/-) mouse embryonic fibroblasts (MEFs). This causes a significant decrease of mitofusin 2 (Mfn2) protein, and increased mitophagy. Our work describes a new stress-signaling pathway that is initiated in the mitochondria and involves the regulation of Mulan by Omi/HtrA2 protease. Deregulation of this pathway, as it occurs in mnd2 mutant mice, causes mitochondrial dysfunction and mitophagy, and could be responsible for the motor neuron disease and the premature aging phenotype observed in these animals.


Asunto(s)
Fibroblastos/metabolismo , GTP Fosfohidrolasas/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Mitofagia/genética , Serina Endopeptidasas/genética , Ubiquitina-Proteína Ligasas/genética , Envejecimiento Prematuro/genética , Envejecimiento Prematuro/metabolismo , Animales , Apoptosis , Línea Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Fibroblastos/patología , GTP Fosfohidrolasas/deficiencia , Regulación de la Expresión Génica , Células HEK293 , Serina Peptidasa A2 que Requiere Temperaturas Altas , Humanos , Ratones , Ratones Noqueados , Mitocondrias/patología , Proteínas Mitocondriales/deficiencia , Enfermedad de la Neurona Motora/genética , Enfermedad de la Neurona Motora/metabolismo , Enfermedad de la Neurona Motora/patología , Estrés Oxidativo , Transporte de Proteínas , Serina Endopeptidasas/deficiencia , Transducción de Señal , Ubiquitina-Proteína Ligasas/metabolismo
19.
J Neurochem ; 132(6): 687-702, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25359615

RESUMEN

Mitochondrial dysfunction is implicated in age-related degenerative disorders such as Alzheimer's disease (AD). Maintenance of mitochondrial dynamics is essential for regulating mitochondrial function. Aß oligomers (AßOs), the typical cause of AD, lead to mitochondrial dysfunction and neuronal loss. AßOs have been shown to induce mitochondrial fragmentation, and their inhibition suppresses mitochondrial dysfunction and neuronal cell death. Oxidative stress is one of the earliest hallmarks of AD. Cyclin-dependent kinase 5 (Cdk5) may cause oxidative stress by disrupting the antioxidant system, including Prx2. Cdk5 is also regarded as a modulator of mitochondrial fission; however, a precise mechanistic link between Cdk5 and mitochondrial dynamics is lacking. We estimated mitochondrial morphology and alterations in mitochondrial morphology-related proteins in Neuro-2a (N2a) cells stably expressing the Swedish mutation of amyloid precursor protein (APP), which is known to increase AßO production. We demonstrated that mitochondrial fragmentation by AßOs accompanies reduced mitofusin 1 and 2 (Mfn1/2) levels. Interestingly, the Cdk5 pathway, including phosphorylation of the Prx2-related oxidative stress, has been shown to regulate Mfn1 and Mfn2 levels. Furthermore, Mfn2, but not Mfn1, over-expression significantly inhibits the AßO-mediated cell death pathway. Therefore, these results indicate that AßO-mediated oxidative stress triggers mitochondrial fragmentation via decreased Mfn2 expression by activating Cdk5-induced Prx2 phosphorylation. Mitochondrial fragmentation induced by amyloid-beta oligomer (AßOs) which is generated from the Swedish mutation of amyloid precursor protein (APP) accompanies reduced Mfn1/2 levels. Interestingly, the Cdk5 pathway, including phosphorylation of the Prx2-related oxidative stress, has been shown to regulate Mfn1/2. Furthermore, Mfn2 over-expression significantly inhibits the AßO-mediated neuronal cells death pathway, but not Mfn1 over-expression. Therefore, these results indicate that AßO-mediated oxidative stress triggers mitochondrial fragmentation via decreased Mfn2 expression by activating Cdk5-induced Prx2 phosphorylation. ATP, adenosine triphosphate; Bax, Bcl-2-associated X protein; Bcl-2, B-cell lymphoma 2; Cdk5, Cyclin-dependent kinase; Cyt C, cytochrome C; Mfn2, mitofusin 2; Prx2, peroxiredoxin 2; ROS, reactive oxygen species.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Quinasa 5 Dependiente de la Ciclina/metabolismo , GTP Fosfohidrolasas/biosíntesis , Mitocondrias/metabolismo , Neuronas/metabolismo , Estrés Oxidativo/fisiología , Péptidos beta-Amiloides/metabolismo , Animales , Línea Celular Tumoral , GTP Fosfohidrolasas/deficiencia , Ratones , Mitocondrias/efectos de los fármacos , Neuronas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos
20.
Eur J Immunol ; 44(2): 561-72, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24510501

RESUMEN

Gimap3 (IAN4) and Gimap5 (IAN5) are highly homologous GTP-binding proteins of the Gimap family. Gimap3 and Gimap5, whose transcripts are abundant in mature lymphocytes, can associate with antiapoptotic Bcl-2 family proteins. While it is established that Gimap5 regulates T-cell survival, the in vivo role of Gimap3 is unclear. Here we report the preparation and characteristics of mouse strains lacking Gimap3 and/or Gimap5. We found that the number of T cells was markedly reduced in mice deficient in both Gimap3 and Gimap5. The defects in T-cell cellularity were more severe in mice lacking both Gimap3 and Gimap5 than in mice lacking only Gimap5. No defects in the cellularity of T cells were detected in mice lacking only Gimap3, whereas bone marrow cells from Gimap3-deficient mice showed reduced T-cell production in a competitive hematopoietic environment. Moreover, retroviral overexpression and short hairpin RNAs-mediated silencing of Gimap3 in bone marrow cells elevated and reduced, respectively, the number of T cells produced in irradiated mice. These results suggest that Gimap3 is a regulator of T-cell numbers in the mouse and that multiple Gimap family proteins cooperate to maintain T-cell survival.


Asunto(s)
GTP Fosfohidrolasas/metabolismo , Proteínas de Unión al GTP/metabolismo , Proteínas de la Membrana/metabolismo , Linfocitos T/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Recuento de Células , Supervivencia Celular/fisiología , Células Cultivadas , GTP Fosfohidrolasas/deficiencia , Proteínas de Unión al GTP/deficiencia , Proteínas de la Membrana/deficiencia , Ratones , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA