Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 347
Filtrar
1.
Dis Model Mech ; 17(6)2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38836374

RESUMEN

TANGO2 deficiency disease (TDD) is a rare genetic disorder estimated to affect ∼8000 individuals worldwide. It causes neurodegeneration often accompanied by potentially lethal metabolic crises that are triggered by diet or illness. Recent work has demonstrated distinct lipid imbalances in multiple model systems either depleted for or devoid of the TANGO2 protein, including human cells, fruit flies and zebrafish. Importantly, vitamin B5 supplementation has been shown to rescue TANGO2 deficiency-associated defects in flies and human cells. The notion that vitamin B5 is needed for synthesis of the lipid precursor coenzyme A (CoA) corroborates the hypothesis that key aspects of TDD pathology may be caused by lipid imbalance. A natural history study of 73 individuals with TDD reported that either multivitamin or vitamin B complex supplementation prevented the metabolic crises, suggesting this as a potentially life-saving treatment. Although recently published work supports this notion, much remains unknown about TANGO2 function, the pathological mechanism of TDD and the possible downsides of sustained vitamin supplementation in children and young adults. In this Perspective, we discuss these recent findings and highlight areas for immediate scientific attention.


Asunto(s)
Metabolismo de los Lípidos , Proteínas Mitocondriales , Proteínas de Transporte Vesicular , Animales , Humanos , Suplementos Dietéticos , Metabolismo de los Lípidos/genética , Lípidos , Proteínas Mitocondriales/deficiencia , Proteínas de Transporte Vesicular/deficiencia
2.
FEBS Lett ; 598(14): 1740-1752, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38639871

RESUMEN

Reactive oxygen species (ROS) are associated with oocyte maturation inhibition, and N-acetyl-l-cysteine (NAC) partially reduces their harmful effects. Mitochondrial E3 ubiquitin ligase 1 (Mul1) localizes to the mitochondrial outer membrane. We found that female Mul1-deficient mice are infertile, and their oocytes contain high ROS concentrations. After fertilization, Mul1-deficient embryos showed a DNA damage response (DDR) and abnormal preimplantation embryogenesis, which was rescued by NAC addition and ROS depletion. These observations clearly demonstrate that loss of Mul1 in oocytes increases ROS concentrations and triggers DDR, resulting in abnormal preimplantation embryogenesis. We conclude that manipulating the mitochondrial ROS levels in oocytes may be a potential therapeutic approach to target infertility.


Asunto(s)
Desarrollo Embrionario , Oocitos , Especies Reactivas de Oxígeno , Ubiquitina-Proteína Ligasas , Animales , Femenino , Ratones , Acetilcisteína/farmacología , Blastocisto/metabolismo , Daño del ADN , Desarrollo Embrionario/genética , Ratones Noqueados , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/deficiencia , Oocitos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/deficiencia
3.
Nat Commun ; 13(1): 374, 2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-35042858

RESUMEN

Age-related macular degeneration (AMD) is the leading cause of blindness among the elderly. Dry AMD has unclear etiology and no treatment. Lipid-rich drusen are the hallmark of dry AMD. An AMD mouse model and insights into drusenogenesis are keys to better understanding of this disease. Chloride intracellular channel 4 (CLIC4) is a pleomorphic protein regulating diverse biological functions. Here we show that retinal pigment epithelium (RPE)-specific Clic4 knockout mice exhibit a full spectrum of functional and pathological hallmarks of dry AMD. Multidisciplinary longitudinal studies of disease progression in these mice support a mechanistic model that links RPE cell-autonomous aberrant lipid metabolism and transport to drusen formation.


Asunto(s)
Canales de Cloruro/genética , Degeneración Macular/genética , Proteínas Mitocondriales/genética , Mutación/genética , Epitelio Pigmentado de la Retina/metabolismo , Animales , Muerte Celular , Canales de Cloruro/deficiencia , Modelos Animales de Enfermedad , Fondo de Ojo , Homeostasis , Metabolismo de los Lípidos , Degeneración Macular/diagnóstico por imagen , Degeneración Macular/fisiopatología , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Mitocondriales/deficiencia , Especificidad de Órganos/genética , Drusas Retinianas/complicaciones , Drusas Retinianas/diagnóstico por imagen , Drusas Retinianas/patología , Epitelio Pigmentado de la Retina/diagnóstico por imagen , Epitelio Pigmentado de la Retina/fisiopatología , Epitelio Pigmentado de la Retina/ultraestructura , Factores de Riesgo , Transcripción Genética , Visión Ocular/fisiología
4.
Redox Biol ; 50: 102248, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35091324

RESUMEN

Genetic mitochondrial dysfunction is frequently associated with various embryonic developmental defects. However, how mitochondria contribute to early development and cell fate determination is poorly studied, especially in humans. Using human pluripotent stem cells (hPSCs), we established a Dox-induced knockout model with mitochondrial dysfunction and evaluated the effect of mitochondrial dysfunction on human pluripotency maintenance and lineage differentiation. The nucleus-encoded gene TFAM (transcription factor A, mitochondrial), essential for mitochondrial gene transcription and mitochondrial DNA replication, is targeted to construct the mitochondrial dysfunction model. The hPSCs with TFAM depletion exhibit the decrease of mtDNA level and oxidative respiration efficiency, representing a typical mitochondrial dysfunction phenotype. Mitochondrial dysfunction leads to impaired self-renewal in hPSCs due to proliferation arrest. Although the mitochondrial dysfunction does not affect pluripotent gene expression, it results in a severe defect in lineage differentiation. Further study in mesoderm differentiation reveals that mitochondrial dysfunction causes proliferation disability and YAP nuclear translocalization and thus together blocks mesoderm lineage differentiation. These findings provide new insights into understanding the mitochondrial function in human pluripotency maintenance and mesoderm differentiation.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Mitocondrias , Proteínas Mitocondriales , Células Madre Pluripotentes , Factores de Transcripción , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular/fisiología , Proliferación Celular , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/deficiencia , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Factores de Transcripción/metabolismo
5.
PLoS One ; 16(12): e0247261, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34928942

RESUMEN

POLDIP2 is a multifunctional protein whose roles are only partially understood. Our laboratory previously reported physiological studies performed using a mouse gene trap model, which suffered from three limitations: perinatal lethality in homozygotes, constitutive Poldip2 inactivation and inadvertent downregulation of the adjacent Tmem199 gene. To overcome these limitations, we developed a new conditional floxed Poldip2 model. The first part of the present study shows that our initial floxed mice were affected by an unexpected mutation, which was not readily detected by Southern blotting and traditional PCR. It consisted of a 305 kb duplication around Poldip2 with retention of the wild type allele and could be traced back to the original targeted ES cell clone. We offer simple suggestions to rapidly detect similar accidents, which may affect genome editing using both traditional and CRISPR-based methods. In the second part of the present study, correctly targeted floxed Poldip2 mice were generated and used to produce a new constitutive knockout line by crossing with a Cre deleter. In contrast to the gene trap model, many homozygous knockout mice were viable, in spite of having no POLDIP2 expression. To further characterize the effects of Poldip2 ablation in the vasculature, RNA-seq and RT-qPCR experiments were performed in constitutive knockout arteries. Results show that POLDIP2 inactivation affects multiple cellular processes and provide new opportunities for future in-depth study of its functions.


Asunto(s)
Sistemas CRISPR-Cas , Marcación de Gen , Proteínas de la Membrana/genética , Proteínas Mitocondriales/deficiencia , Células Madre Embrionarias de Ratones/metabolismo , Proteínas Nucleares/deficiencia , RNA-Seq , Animales , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Proteínas Mitocondriales/metabolismo , Proteínas Nucleares/metabolismo
6.
Nucleic Acids Res ; 49(22): 13108-13121, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34878141

RESUMEN

Mutations in genes encoding mitochondrial aminoacyl-tRNA synthetases are linked to diverse diseases. However, the precise mechanisms by which these mutations affect mitochondrial function and disease development are not fully understood. Here, we develop a Drosophila model to study the function of dFARS2, the Drosophila homologue of the mitochondrial phenylalanyl-tRNA synthetase, and further characterize human disease-associated FARS2 variants. Inactivation of dFARS2 in Drosophila leads to developmental delay and seizure. Biochemical studies reveal that dFARS2 is required for mitochondrial tRNA aminoacylation, mitochondrial protein stability, and assembly and enzyme activities of OXPHOS complexes. Interestingly, by modeling FARS2 mutations associated with human disease in Drosophila, we provide evidence that expression of two human FARS2 variants, p.G309S and p.D142Y, induces seizure behaviors and locomotion defects, respectively. Together, our results not only show the relationship between dysfunction of mitochondrial aminoacylation system and pathologies, but also illustrate the application of Drosophila model for functional analysis of human disease-causing variants.


Asunto(s)
Discapacidades del Desarrollo/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Proteínas Mitocondriales/genética , Mutación , Fenilalanina-ARNt Ligasa/genética , ARN de Transferencia/genética , Convulsiones/genética , Animales , Línea Celular , Discapacidades del Desarrollo/enzimología , Modelos Animales de Enfermedad , Proteínas de Drosophila/deficiencia , Drosophila melanogaster/enzimología , Técnicas de Silenciamiento del Gen , Humanos , Microscopía Electrónica de Transmisión , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Proteínas Mitocondriales/deficiencia , Fosforilación Oxidativa , Fenilalanina-ARNt Ligasa/deficiencia , ARN de Transferencia/metabolismo , Convulsiones/enzimología , Aminoacilación de ARN de Transferencia
7.
EMBO J ; 40(21): e108648, 2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34542926

RESUMEN

So-called ρ0 cells lack mitochondrial DNA and are therefore incapable of aerobic ATP synthesis. How cells adapt to survive ablation of oxidative phosphorylation remains poorly understood. Complexome profiling analysis of ρ0 cells covered 1,002 mitochondrial proteins and revealed changes in abundance and organization of numerous multiprotein complexes including previously not described assemblies. Beyond multiple subassemblies of complexes that would normally contain components encoded by mitochondrial DNA, we observed widespread reorganization of the complexome. This included distinct changes in the expression pattern of adenine nucleotide carrier isoforms, other mitochondrial transporters, and components of the protein import machinery. Remarkably, ablation of mitochondrial DNA hardly affected the complexes organizing cristae junctions indicating that the altered cristae morphology in ρ0 mitochondria predominantly resulted from the loss of complex V dimers required to impose narrow curvatures to the inner membrane. Our data provide a comprehensive resource for in-depth analysis of remodeling of the mitochondrial complexome in response to respiratory deficiency.


Asunto(s)
Adaptación Fisiológica , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Complejos Multiproteicos/genética , Adenosina Trifosfato/metabolismo , Línea Celular Tumoral , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Expresión Génica , Humanos , Mitocondrias/patología , Membranas Mitocondriales/química , Membranas Mitocondriales/metabolismo , Proteínas Mitocondriales/deficiencia , Complejos Multiproteicos/deficiencia , Osteoblastos/metabolismo , Osteoblastos/patología , Fosforilación Oxidativa
8.
Front Immunol ; 12: 723683, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34456930

RESUMEN

Mitofusin 2 (MFN2) is a mitochondrial outer membrane GTPase, which modulates mitochondrial fusion and affects the interaction between endoplasmic reticulum and mitochondria. Here, we explored how MFN2 influences mitochondrial functions and inflammatory responses towards zymosan in primary human macrophages. A knockdown of MFN2 by small interfering RNA decreased mitochondrial respiration without attenuating mitochondrial membrane potential and reduced interactions between endoplasmic reticulum and mitochondria. A MFN2 deficiency potentiated zymosan-elicited inflammatory responses of human primary macrophages, such as expression and secretion of pro-inflammatory cytokines interleukin-1ß, -6, -8 and tumor necrosis factor α, as well as induction of cyclooxygenase 2 and prostaglandin E2 synthesis. MFN2 silencing also increased zymosan-induced nuclear factor kappa-light-chain-enhancer of activated B cells and mitogen-activated protein kinases inflammatory signal transduction, without affecting mitochondrial reactive oxygen species production. Mechanistic studies revealed that MFN2 deficiency enhanced the toll-like receptor 2-dependent branch of zymosan-triggered responses upstream of inhibitor of κB kinase. This was associated with elevated, cytosolic expression of interleukin-1 receptor-associated kinase 4 in MFN2-deficient cells. Our data suggest pro-inflammatory effects of MFN2 deficiency in human macrophages.


Asunto(s)
Estrés del Retículo Endoplásmico/fisiología , GTP Fosfohidrolasas/metabolismo , Inflamación/metabolismo , Macrófagos/metabolismo , Proteínas Mitocondriales/metabolismo , Transducción de Señal/fisiología , Citocinas/metabolismo , Retículo Endoplásmico/metabolismo , GTP Fosfohidrolasas/deficiencia , Humanos , Mitocondrias/metabolismo , Proteínas Mitocondriales/deficiencia , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Especies Reactivas de Oxígeno/metabolismo
9.
J Cell Biol ; 220(9)2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34259806

RESUMEN

ER-plasma membrane (PM) contacts are proposed to be held together by distinct families of tethering proteins, which in yeast include the VAP homologues Scs2/22, the extended-synaptotagmin homologues Tcb1/2/3, and the TMEM16 homologue Ist2. It is unclear whether these tethers act redundantly or whether individual tethers have specific functions at contacts. Here, we show that Ist2 directly recruits the phosphatidylserine (PS) transport proteins and ORP family members Osh6 and Osh7 to ER-PM contacts through a binding site located in Ist2's disordered C-terminal tethering region. This interaction is required for phosphatidylethanolamine (PE) production by the PS decarboxylase Psd2, whereby PS transported from the ER to the PM by Osh6/7 is endocytosed to the site of Psd2 in endosomes/Golgi/vacuoles. This role for Ist2 and Osh6/7 in nonvesicular PS transport is specific, as other tethers/transport proteins do not compensate. Thus, we identify a molecular link between the ORP and TMEM16 families and a role for endocytosis of PS in PE synthesis.


Asunto(s)
Proteínas de Unión a Ácidos Grasos/metabolismo , Metabolismo de los Lípidos/genética , Fosfolípidos/metabolismo , Receptores de Esteroides/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Sitios de Unión , Transporte Biológico , Carboxiliasas/deficiencia , Carboxiliasas/genética , Membrana Celular/metabolismo , Retículo Endoplásmico/metabolismo , Endosomas/metabolismo , Proteínas de Unión a Ácidos Grasos/genética , Regulación Fúngica de la Expresión Génica , Ingeniería Genética , Aparato de Golgi/metabolismo , Proteínas de Microfilamentos/deficiencia , Proteínas de Microfilamentos/genética , Proteínas Mitocondriales/deficiencia , Proteínas Mitocondriales/genética , Modelos Moleculares , Fosfatidiletanolaminas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Fosfatidilserinas/metabolismo , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Unión Proteica , Conformación Proteica , Mapeo de Interacción de Proteínas , Receptores de Esteroides/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Transducción de Señal
10.
Am J Physiol Cell Physiol ; 321(3): C519-C534, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34319827

RESUMEN

Mitochondria are recognized as signaling organelles, because under stress, mitochondria can trigger various signaling pathways to coordinate the cell's response. The specific pathway(s) engaged by mitochondria in response to mitochondrial energy defects in vivo and in high-energy tissues like the heart are not fully understood. Here, we investigated cardiac pathways activated in response to mitochondrial energy dysfunction by studying mice with cardiomyocyte-specific loss of the mitochondrial phosphate carrier (SLC25A3), an established model that develops cardiomyopathy as a result of defective mitochondrial ATP synthesis. Mitochondrial energy dysfunction induced a striking pattern of acylome remodeling, with significantly increased posttranslational acetylation and malonylation. Mass spectrometry-based proteomics further revealed that energy dysfunction-induced remodeling of the acetylome and malonylome preferentially impacts mitochondrial proteins. Acetylation and malonylation modified a highly interconnected interactome of mitochondrial proteins, and both modifications were present on the enzyme isocitrate dehydrogenase 2 (IDH2). Intriguingly, IDH2 activity was enhanced in SLC25A3-deleted mitochondria, and further study of IDH2 sites targeted by both acetylation and malonylation revealed that these modifications can have site-specific and distinct functional effects. Finally, we uncovered a novel cross talk between the two modifications, whereby mitochondrial energy dysfunction-induced acetylation of sirtuin 5 (SIRT5), inhibited its function. Because SIRT5 is a mitochondrial deacylase with demalonylase activity, this finding suggests that acetylation can modulate the malonylome. Together, our results position acylations as an arm of the mitochondrial response to energy dysfunction and suggest a mechanism by which focal disruption to the energy production machinery can have an expanded impact on global mitochondrial function.


Asunto(s)
Cardiomiopatías/genética , Proteínas de Transporte de Catión/genética , Isocitrato Deshidrogenasa/genética , Mitocondrias Cardíacas/metabolismo , Proteínas Mitocondriales/genética , Miocitos Cardíacos/metabolismo , Proteínas de Transporte de Fosfato/genética , Procesamiento Proteico-Postraduccional , Proteínas Transportadoras de Solutos/genética , Acetilación , Animales , Transporte Biológico , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Proteínas de Transporte de Catión/deficiencia , Metabolismo Energético , Femenino , Redes Reguladoras de Genes , Isocitrato Deshidrogenasa/metabolismo , Masculino , Malonatos/metabolismo , Ratones , Ratones Noqueados , Mitocondrias Cardíacas/genética , Mitocondrias Cardíacas/patología , Proteínas Mitocondriales/deficiencia , Modelos Moleculares , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/patología , Proteínas de Transporte de Fosfato/deficiencia , Fosfatos , Conformación Proteica , Mapeo de Interacción de Proteínas , Transducción de Señal , Sirtuinas/genética , Sirtuinas/metabolismo , Proteínas Transportadoras de Solutos/deficiencia
11.
PLoS One ; 16(7): e0255355, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34320035

RESUMEN

Mitochondrial dysfunction is significantly associated with neurological deficits and age-related neurological diseases. While mitochondria are dynamically regulated and properly maintained during neurogenesis, the manner in which mitochondrial activities are controlled and contribute to these processes is not fully understood. Mitochondrial transcription factor A (TFAM) contributes to mitochondrial function by maintaining mitochondrial DNA (mtDNA). To clarify how mitochondrial dysfunction affects neurogenesis, we induced mitochondrial dysfunction specifically in murine neural stem cells (NSCs) by inactivating Tfam. Tfam inactivation in NSCs resulted in mitochondrial dysfunction by reducing respiratory chain activities and causing a severe deficit in neural differentiation and maturation both in vivo and in vitro. Brain tissue from Tfam-deficient mice exhibited neuronal cell death primarily at layer V and microglia were activated prior to cell death. Cultured Tfam-deficient NSCs showed a reduction in reactive oxygen species produced by the mitochondria. Tfam inactivation during neurogenesis resulted in the accumulation of ATF4 and activation of target gene expression. Therefore, we propose that the integrated stress response (ISR) induced by mitochondrial dysfunction in neurogenesis is activated to protect the progression of neurodegenerative diseases.


Asunto(s)
Encéfalo/patología , Proteínas de Unión al ADN/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Estrés Oxidativo , Factores de Transcripción/genética , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Diferenciación Celular , Células Cultivadas , ADN Mitocondrial/metabolismo , Proteínas de Unión al ADN/deficiencia , Regulación hacia Abajo , Proteínas del Complejo de Cadena de Transporte de Electrón/genética , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/citología , Microglía/metabolismo , Proteínas Mitocondriales/deficiencia , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis , Especies Reactivas de Oxígeno/metabolismo , Factores de Transcripción/deficiencia
12.
Mol Metab ; 53: 101276, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34153520

RESUMEN

OBJECTIVE: Insulin regulates mitochondrial function, thereby propagating an efficient metabolism. Conversely, diabetes and insulin resistance are linked to mitochondrial dysfunction with a decreased expression of the mitochondrial chaperone HSP60. The aim of this investigation was to determine the effect of a reduced HSP60 expression on the development of obesity and insulin resistance. METHODS: Control and heterozygous whole-body HSP60 knockout (Hsp60+/-) mice were fed a high-fat diet (HFD, 60% calories from fat) for 16 weeks and subjected to extensive metabolic phenotyping. To understand the effect of HSP60 on white adipose tissue, microarray analysis of gonadal WAT was performed, ex vivo experiments were performed, and a lentiviral knockdown of HSP60 in 3T3-L1 cells was conducted to gain detailed insights into the effect of reduced HSP60 levels on adipocyte homeostasis. RESULTS: Male Hsp60+/- mice exhibited lower body weight with lower fat mass. These mice exhibited improved insulin sensitivity compared to control, as assessed by Matsuda Index and HOMA-IR. Accordingly, insulin levels were significantly reduced in Hsp60+/- mice in a glucose tolerance test. However, Hsp60+/- mice exhibited an altered adipose tissue metabolism with elevated insulin-independent glucose uptake, adipocyte hyperplasia in the presence of mitochondrial dysfunction, altered autophagy, and local insulin resistance. CONCLUSIONS: We discovered that the reduction of HSP60 in mice predominantly affects adipose tissue homeostasis, leading to beneficial alterations in body weight, body composition, and adipocyte morphology, albeit exhibiting local insulin resistance.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Chaperonina 60/metabolismo , Proteínas Mitocondriales/metabolismo , Obesidad/metabolismo , Células 3T3-L1 , Animales , Células Cultivadas , Chaperonina 60/deficiencia , Dieta Alta en Grasa/efectos adversos , Metabolismo Energético , Homeostasis , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Mitocondriales/deficiencia
13.
Commun Biol ; 4(1): 666, 2021 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-34079053

RESUMEN

Calcium dynamics control synaptic transmission. Calcium triggers synaptic vesicle fusion, determines release probability, modulates vesicle recycling, participates in long-term plasticity and regulates cellular metabolism. Mitochondria, the main source of cellular energy, serve as calcium signaling hubs. Mitochondrial calcium transients are primarily determined by the balance between calcium influx, mediated by the mitochondrial calcium uniporter (MCU), and calcium efflux through the sodium/lithium/calcium exchanger (NCLX). We identified a human recessive missense SLC8B1 variant that impairs NCLX activity and is associated with severe mental retardation. On this basis, we examined the effect of deleting NCLX in mice on mitochondrial and synaptic calcium homeostasis, synaptic activity, and plasticity. Neuronal mitochondria exhibited basal calcium overload, membrane depolarization, and a reduction in the amplitude and rate of calcium influx and efflux. We observed smaller cytoplasmic calcium transients in the presynaptic terminals of NCLX-KO neurons, leading to a lower probability of release and weaker transmission. In agreement, synaptic facilitation in NCLX-KO hippocampal slices was enhanced. Importantly, deletion of NCLX abolished long term potentiation of Schaffer collateral synapses. Our results show that NCLX controls presynaptic calcium transients that are crucial for defining synaptic strength as well as short- and long-term plasticity, key elements of learning and memory processes.


Asunto(s)
Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Intercambiador de Sodio-Calcio/genética , Intercambiador de Sodio-Calcio/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Señalización del Calcio , Cardiomiopatías/genética , Cardiomiopatías/metabolismo , Femenino , Hipocampo/metabolismo , Humanos , Técnicas In Vitro , Potenciación a Largo Plazo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Mitocondriales/química , Proteínas Mitocondriales/deficiencia , Plasticidad Neuronal , Neuronas/metabolismo , Linaje , Mutación Puntual , Terminales Presinápticos/metabolismo , Intercambiador de Sodio-Calcio/química , Transmisión Sináptica/genética , Transmisión Sináptica/fisiología
14.
J Trace Elem Med Biol ; 66: 126759, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33872833

RESUMEN

BACKGROUND: Acquisition and distribution of zinc supports a number of biological processes. Various molecular factors are involved in zinc metabolism but not fully explored. BASIC PROCEDURES: Spontaneous mutants were generated in yeast with excess zinc culture followed by whole genome DNA sequencing to discover zinc metabolism related genes by bioinformatics. An identified mutant was characterized through metallomic and molecular biology methods. MAIN FINDINGS: Here we reported that MTM1 knockout cells displayed much stronger zinc tolerance than wild type cells on SC medium when exposed to excess zinc. Zn accumulation of mtm1Δ cells was dramatically decreased compared to wild type cells under excessive zinc condition due to MTM1 deletion reduced zinc uptake. ZRC1 mRNA level of mtm1Δ cells was significantly higher than that in the wild-type strain leading to increased vacuolar zinc accumulations in mtm1Δ cells. The mRNA levels of ZRT1 and ZAP1 decreased in mtm1Δ cells contributing to less Zn uptake. The zrc1Δmtm1Δ double knockout strain exhibited Zn sensitivity. MTM1 knockout did not afford resistance to excess zinc through an effect mediated through an influence on levels of ROS. Superoxide dismutase 2 (Sod2p) activity in mtm1Δ cells was severely impaired and not restored through Zn supplementation. Meanwhile, additional Zn showed no significant effect on the localization and expression of Mtm1p. PRINCIPAL CONCLUSIONS: Our study reveals the MTM1 gene plays an important role in the regulation of zinc homeostasis in yeast cells via changing zinc uptake and distribution. This discovery provides new insights for better understanding biochemical communication between vacuole and mitochondrial in relation to zinc-metabolism.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Zinc/metabolismo , Proteínas Portadoras/genética , Proteínas Mitocondriales/deficiencia , Proteínas Mitocondriales/genética , Mutación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Saccharomyces cerevisiae/genética
15.
J Biol Chem ; 296: 100539, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33722607

RESUMEN

Phosphatidylethanolamine (PE) is essential for mitochondrial respiration in yeast, Saccharomyces cerevisiae, whereas the most abundant mitochondrial phospholipid, phosphatidylcholine (PC), is largely dispensable. Surprisingly, choline (Cho), which is a biosynthetic precursor of PC, has been shown to rescue the respiratory growth of mitochondrial PE-deficient yeast; however, the mechanism underlying this rescue has remained unknown. Using a combination of yeast genetics, lipid biochemistry, and cell biological approaches, we uncover the mechanism by showing that Cho rescues mitochondrial respiration by partially replenishing mitochondrial PE levels in yeast cells lacking the mitochondrial PE-biosynthetic enzyme Psd1. This rescue is dependent on the conversion of Cho to PC via the Kennedy pathway as well as on Psd2, an enzyme catalyzing PE biosynthesis in the endosome. Metabolic labeling experiments reveal that in the absence of exogenously supplied Cho, PE biosynthesized via Psd2 is mostly directed to the methylation pathway for PC biosynthesis and is unavailable for replenishing mitochondrial PE in Psd1-deleted cells. In this setting, stimulating the Kennedy pathway for PC biosynthesis by Cho spares Psd2-synthesized PE from the methylation pathway and redirects it to the mitochondria. Cho-mediated elevation in mitochondrial PE is dependent on Vps39, which has been recently implicated in PE trafficking to the mitochondria. Accordingly, epistasis experiments placed Vps39 downstream of Psd2 in Cho-based rescue. Our work, thus, provides a mechanism of Cho-based rescue of mitochondrial PE deficiency and uncovers an intricate interorganelle phospholipid regulatory network that maintains mitochondrial PE homeostasis.


Asunto(s)
Carboxiliasas/deficiencia , Colina/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/deficiencia , Fosfatidilcolinas/metabolismo , Fosfatidiletanolaminas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/crecimiento & desarrollo
16.
Biochem Biophys Res Commun ; 549: 67-74, 2021 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-33667711

RESUMEN

Amyloid-ß (Aß) plaques are strongly associated with the development of Alzheimer's disease (AD). However, it remains unclear how morphological differences in Aß plaques determine the pathogenesis of Aß. Here, we categorized Aß plaques into four types based on the macroscopic features of the dense core, and found that the Aß-plaque subtype containing a larger dense core showed the strongest association with neuritic dystrophy. Astrocytes dominantly accumulated toward these expanded/dense-core-containing Aß plaques. Previously, we indicated that deletion of the mitochondrial ubiquitin ligase MITOL/MARCH5 triggers mitochondrial impairments and exacerbates cognitive decline in a mouse model with AD-related Aß pathology. In this study, MITOL deficiency accelerated the formation of expanded/dense-core-containing Aß plaques, which showed reduced contacts with astrocytes, but not microglia. Our findings suggest that expanded/dense-core-containing Aß-plaque formation enhanced by the alteration of mitochondrial function robustly contributes to the exacerbation of Aß neuropathology, at least in part, through the reduced contacts between Aß plaques and astrocytes.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Astrocitos/patología , Neurotoxinas/toxicidad , Placa Amiloide/patología , Animales , Astrocitos/efectos de los fármacos , Eliminación de Gen , Ratones Transgénicos , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/patología , Proteínas Mitocondriales/deficiencia , Proteínas Mitocondriales/genética , Neuritas/efectos de los fármacos , Neuritas/metabolismo , Neuritas/patología , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/genética
17.
EMBO J ; 40(8): e105268, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33528041

RESUMEN

Mitochondrial translation dysfunction is associated with neurodegenerative and cardiovascular diseases. Cells eliminate defective mitochondria by the lysosomal machinery via autophagy. The relationship between mitochondrial translation and lysosomal function is unknown. In this study, mitochondrial translation-deficient hearts from p32-knockout mice were found to exhibit enlarged lysosomes containing lipofuscin, suggesting impaired lysosome and autolysosome function. These mice also displayed autophagic abnormalities, such as p62 accumulation and LC3 localization around broken mitochondria. The expression of genes encoding for nicotinamide adenine dinucleotide (NAD+ ) biosynthetic enzymes-Nmnat3 and Nampt-and NAD+ levels were decreased, suggesting that NAD+ is essential for maintaining lysosomal acidification. Conversely, nicotinamide mononucleotide (NMN) administration or Nmnat3 overexpression rescued lysosomal acidification. Nmnat3 gene expression is suppressed by HIF1α, a transcription factor that is stabilized by mitochondrial translation dysfunction, suggesting that HIF1α-Nmnat3-mediated NAD+ production is important for lysosomal function. The glycolytic enzymes GAPDH and PGK1 were found associated with lysosomal vesicles, and NAD+ was required for ATP production around lysosomal vesicles. Thus, we conclude that NAD+ content affected by mitochondrial dysfunction is essential for lysosomal maintenance.


Asunto(s)
Lisosomas/metabolismo , Mitocondrias Cardíacas/metabolismo , Proteínas Mitocondriales/genética , NAD/metabolismo , Animales , Células Cultivadas , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Mitocondriales/deficiencia , Nicotinamida-Nucleótido Adenililtransferasa/metabolismo , Fosfoglicerato Quinasa/metabolismo
18.
Mol Genet Metab ; 132(2): 146-153, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33485800

RESUMEN

TRMU is a nuclear gene crucial for mitochondrial DNA translation by encoding tRNA 5-methylaminomethyl-2-thiouridylate methyltransferase, which thiolates mitochondrial tRNA. Biallelic pathogenic variants in TRMU are associated with transient infantile liver failure. Other less common presentations such as Leigh syndrome, myopathy, and cardiomyopathy have been reported. Recent studies suggested that provision of exogenous L-cysteine or N-acetylcysteine may ameliorate the effects of disease-causing variants and improve the natural history of the disease. Here, we report six infants with biallelic TRMU variants, including four previously unpublished patients, all treated with exogenous cysteine. We highlight the first report of an affected patient undergoing orthotopic liver transplantation, the long-term effects of cysteine supplementation, and the ability of the initial presentation to mimic multiple inborn errors of metabolism. We propose that TRMU deficiency should be suspected in all children presenting with persistent lactic acidosis and hypoglycemia, and that combined N-acetylcysteine and L-cysteine supplementation should be considered prior to molecular diagnosis, as this is a low-risk approach that may increase survival and mitigate the severity of the disease course.


Asunto(s)
Enfermedad de Leigh/terapia , Fallo Hepático/terapia , Proteínas Mitocondriales/genética , Biosíntesis de Proteínas , ARNt Metiltransferasas/genética , Acetilcisteína/administración & dosificación , Acetilcisteína/metabolismo , Acidosis/genética , Acidosis/metabolismo , Cisteína/administración & dosificación , Cisteína/metabolismo , ADN Mitocondrial/genética , Femenino , Humanos , Lactante , Enfermedad de Leigh/genética , Enfermedad de Leigh/metabolismo , Enfermedad de Leigh/patología , Fallo Hepático/genética , Fallo Hepático/metabolismo , Fallo Hepático/patología , Trasplante de Hígado/métodos , Masculino , Mitocondrias/enzimología , Proteínas Mitocondriales/deficiencia , ARN de Transferencia/genética , ARNt Metiltransferasas/deficiencia
20.
Autophagy ; 17(11): 3530-3546, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33459136

RESUMEN

Mitophagy formed the basis of the original description of autophagy by Christian de Duve when he demonstrated that GCG (glucagon) induced macroautophagic/autophagic turnover of mitochondria in the liver. However, the molecular basis of liver-specific activation of mitophagy by GCG, or its significance for metabolic stress responses in the liver is not understood. Here we show that BNIP3 is required for GCG-induced mitophagy in the liver through interaction with processed LC3B; an interaction that is also necessary to localize LC3B out of the nucleus to cytosolic mitophagosomes in response to nutrient deprivation. Loss of BNIP3-dependent mitophagy caused excess mitochondria to accumulate in the liver, disrupting metabolic zonation within the liver parenchyma, with expansion of zone 1 metabolism at the expense of zone 3 metabolism. These results identify BNIP3 as a regulator of metabolic homeostasis in the liver through its effect on mitophagy and mitochondrial mass distribution.Abbreviations: ASS1, arginosuccinate synthetase; BNIP3, BCL2/adenovirus E1B interacting protein 3; CV, central vein; GCG - glucagon; GLUL, glutamate- ammonia ligase (glutamine synthetase); HCQ, hydroxychloroquine; LIR, LC3-interacting region; MAP1LC3B/LC3B, microtubule-associated protein 1 light chain 3 beta; mtDNA:nucDNA, ratio of mitochondrial DNA to nuclear DNA; PV, periportal vein; TOMM20, translocase of outer mitochondrial membrane protein 20.


Asunto(s)
Hígado/citología , Hígado/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Mitocondriales/metabolismo , Mitofagia/fisiología , Animales , Células Cultivadas , Citosol/metabolismo , Glucagón/metabolismo , Glucagón/farmacología , Homeostasis , Humanos , Hígado/efectos de los fármacos , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/genética , Mitocondrias Hepáticas/metabolismo , Proteínas Mitocondriales/deficiencia , Proteínas Mitocondriales/genética , Mitofagia/efectos de los fármacos , Mitofagia/genética , Proteínas Proto-Oncogénicas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA