Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
FASEB J ; 36(11): e22596, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36208295

RESUMEN

Cox17 is required in the assembly of mitochondrial intermembrane space (IMS) and Cu metallization of cytochrome C oxidase (CcO) in mitochondria as well as Cu homeostasis in cells. Cox deficiency is associated with hematopoietic diseases such as tubulopathy and leukodystrophy, but whether and how cox17 functions in hematopoiesis are still unknown. Here, we report the effects of zebrafish cox17 deficiency on primitive erythropoiesis, mitochondrial metabolism, and hypoxia tolerance. Cox17-/- larvae were sensitive to hypoxia stress, with reduced primitive erythropoiesis. Meanwhile, cox17-/- mutants showed a significant reduction in the expression of pivotal transcriptional regulators in erythropoiesis, such as scl, lmo2, and gata1a at 14 h post fertilization (hpf), with expression remaining downregulated for scl but upregulated for lmo2 and gata1a at 24 hpf. Mechanistically, cox17-/- mutants showed impaired mitochondrial metabolism, coupled with a significant decrease in the mitochondrial membrane potential, ATP and SAM content, and the ratio of SAM and SAH. Additionally, disrupting mitochondrial metabolism in wild type (WT) larvae treated with carbonyl cyanide 3-chlorophenylhydrazone (CCCP) could mimic the primitive erythropoiesis defects observed in cox17-/- mutants. Moreover, cox17-/- mutants exhibited significantly downregulated WNT signaling and upregulated ER stress, with a significant reduction of beta-Catenin in gata1a+ cells and of binding enrichment in both scl and lmo2 promoters of the WNT transcriptional factor TCF4. This is the first report on the novel linkage of cox17 deficiency with defective primitive erythropoiesis and reduced hypoxia tolerance. This study has shed light on the potential mechanism by which Cox deficiency, especially cox17 deficiency, induces Cu homeostasis imbalance, leading to hematopoietic diseases.


Asunto(s)
Deficiencia de Citocromo-c Oxidasa , Pez Cebra , Adenosina Trifosfato/metabolismo , Animales , Carbonil Cianuro m-Clorofenil Hidrazona , Deficiencia de Citocromo-c Oxidasa/metabolismo , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/metabolismo , Eritropoyesis , Hipoxia/metabolismo , Proteínas con Dominio LIM/metabolismo , Mitocondrias/metabolismo , Factores de Transcripción/metabolismo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , beta Catenina/metabolismo
2.
Neuropathol Appl Neurobiol ; 48(7): e12841, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35894812

RESUMEN

AIMS: Patients with dermatomyositis (DM) suffer from reduced aerobic metabolism contributing to impaired muscle function, which has been linked to cytochrome c oxidase (COX) deficiency in muscle tissue. This mitochondrial respiratory chain dysfunction is typically seen in perifascicular regions, which also show the most intense inflammatory reaction along with capillary loss and muscle fibre atrophy. The objective of this study was to investigate the pathobiology of the oxidative phosphorylation deficiency in DM. METHODS: Muscle biopsy specimens with perifascicular COX deficiency from five juveniles and seven adults with DM were investigated. We combined immunohistochemical analyses of subunits in the respiratory chain including complex I (subunit NDUFB8), complex II (succinate dehydrogenase, subunit SDHB) and complex IV (COX, subunit MTCO1) with in situ hybridisation, next generation deep sequencing and quantitative polymerase chain reaction (PCR). RESULTS: There was a profound deficiency of complexes I and IV in the perifascicular regions with enzyme histochemical COX deficiency, whereas succinate dehydrogenase activity and complex II were preserved. In situ hybridisation of mitochondrial RNA showed depletion of mitochondrial DNA (mtDNA) transcripts in the perifascicular regions. Analysis of mtDNA by next generation deep sequencing and quantitative PCR in affected muscle regions showed an overall reduction of mtDNA copy number particularly in the perifascicular regions. CONCLUSION: The respiratory chain dysfunction in DM muscle is associated with mtDNA depletion causing deficiency of complexes I and IV, which are partially encoded by mtDNA, whereas complex II, which is entirely encoded by nuclear DNA, is preserved. The depletion of mtDNA indicates a perturbed replication of mtDNA explaining the muscle pathology and the disturbed aerobic metabolism.


Asunto(s)
Deficiencia de Citocromo-c Oxidasa , Dermatomiositis , Adulto , Humanos , Deficiencia de Citocromo-c Oxidasa/metabolismo , Deficiencia de Citocromo-c Oxidasa/patología , Succinato Deshidrogenasa/análisis , Succinato Deshidrogenasa/metabolismo , Dermatomiositis/patología , Transporte de Electrón , Fibras Musculares Esqueléticas/química , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , ADN Mitocondrial/genética , Complejo IV de Transporte de Electrones/análisis , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/metabolismo , Músculo Esquelético/patología
3.
Brain Pathol ; 31(3): e12931, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33354847

RESUMEN

Muscle pathology in inclusion body myositis (IBM) typically includes inflammatory cell infiltration, muscle fibers with rimmed vacuoles and cytochrome c oxidase (COX)-deficient fibers. Previous studies have revealed clonal expansion of large mitochondrial DNA (mtDNA) deletions in the COX-deficient muscle fibers. Technical limitations have prevented complete investigations of the mtDNA deletions and other mtDNA variants. Detailed characterization by deep sequencing of mtDNA in muscle samples from 21 IBM patients and 10 age-matched controls was performed after whole genome sequencing with a mean depth of mtDNA coverage of 46,000x. Multiple large mtDNA deletions and duplications were identified in all IBM and control muscle samples. In general, the IBM muscles demonstrated a larger number of deletions and duplications with a mean heteroplasmy level of 10% (range 1%-35%) compared to controls (1%, range 0.2%-3%). There was also a small increase in the number of somatic single nucleotide variants in IBM muscle. More than 200 rearrangements were recurrent in at least two or more IBM muscles while 26 were found in both IBM and control muscles. The deletions and duplications, with a high recurrence rate, were mainly observed in three mtDNA regions, m.534-4429, m.6330-13993, and m.8636-16072, where some were flanked by repetitive sequences. The mtDNA copy number in IBM muscle was reduced to 42% of controls. Immunohistochemical and western blot analyses of IBM muscle revealed combined complex I and complex IV deficiency affecting the COX-deficient fibers. In conclusion, deep sequencing and quantitation of mtDNA variants revealed that IBM muscles had markedly increased levels of large deletions and duplications, and there were also indications of increased somatic single nucleotide variants and reduced mtDNA copy numbers compared to age-matched controls. The distribution and type of variants were similar in IBM muscle and controls indicating an accelerated aging process in IBM muscle, possibly associated with chronic inflammation.


Asunto(s)
ADN Mitocondrial/genética , Fibras Musculares Esqueléticas/patología , Miositis por Cuerpos de Inclusión/genética , Miositis por Cuerpos de Inclusión/patología , Anciano , Deficiencia de Citocromo-c Oxidasa/genética , Deficiencia de Citocromo-c Oxidasa/metabolismo , Deficiencia de Citocromo-c Oxidasa/patología , Femenino , Reordenamiento Génico/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Persona de Mediana Edad , Mitocondrias/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Miositis por Cuerpos de Inclusión/metabolismo
4.
Cells ; 9(10)2020 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-33003371

RESUMEN

Murine fibroblasts deficient in mitochondria respiratory complexes III (CIII) and IV (CIV) produced by either the ablation of Uqcrfs1 (encoding for Rieske iron sulfur protein, RISP) or Cox10 (encoding for protoheme IX farnesyltransferase, COX10) genes, respectively, showed a pleiotropic effect in complex I (CI). Exposure to 1-5% oxygen increased the levels of CI in both RISP and COX10 KO fibroblasts. De novo assembly of the respiratory complexes occurred at a faster rate and to higher levels in 1% oxygen compared to normoxia in both RISP and COX10 KO fibroblasts. Hypoxia did not affect the levels of assembly of CIII in the COX10 KO fibroblasts nor abrogated the genetic defect impairing CIV assembly. Mitochondrial signaling involving reactive oxygen species (ROS) has been implicated as necessary for HIF-1α stabilization in hypoxia. We did not observe increased ROS production in hypoxia. Exposure to low oxygen levels stabilized HIF-1α and increased CI levels in RISP and COX10 KO fibroblasts. Knockdown of HIF-1α during hypoxic conditions abrogated the beneficial effect of hypoxia on the stability/assembly of CI. These findings demonstrate that oxygen and HIF-1α regulate the assembly of respiratory complexes.


Asunto(s)
Complejo III de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Fibroblastos/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Mitocondrias/metabolismo , Transferasas Alquil y Aril/genética , Transferasas Alquil y Aril/metabolismo , Animales , Hipoxia de la Célula/genética , Línea Celular , Deficiencia de Citocromo-c Oxidasa/metabolismo , Doxiciclina/farmacología , Complejo III de Transporte de Electrones/deficiencia , Complejo III de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/genética , Técnicas de Inactivación de Genes , Silenciador del Gen , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , ARN Interferente Pequeño , Especies Reactivas de Oxígeno/metabolismo
5.
Clin Genet ; 97(2): 276-286, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31600844

RESUMEN

Autosomal dominant progressive external ophthalmoplegia (adPEO) is a late-onset, Mendelian mitochondrial disorder characterised by paresis of the extraocular muscles, ptosis, and skeletal-muscle restricted multiple mitochondrial DNA (mtDNA) deletions. Although dominantly inherited, pathogenic variants in POLG, TWNK and RRM2B are among the most common genetic defects of adPEO, identification of novel candidate genes and the underlying pathomechanisms remains challenging. We report the clinical, genetic and molecular investigations of a patient who presented in the seventh decade of life with PEO. Oxidative histochemistry revealed cytochrome c oxidase-deficient fibres and occasional ragged red fibres showing subsarcolemmal mitochondrial accumulation in skeletal muscle, while molecular studies identified the presence of multiple mtDNA deletions. Negative candidate screening of known nuclear genes associated with PEO prompted diagnostic exome sequencing, leading to the prioritisation of a novel heterozygous c.547G>C variant in GMPR (NM_006877.3) encoding guanosine monophosphate reductase, a cytosolic enzyme required for maintaining the cellular balance of adenine and guanine nucleotides. We show that the novel c.547G>C variant causes aberrant splicing, decreased GMPR protein levels in patient skeletal muscle, proliferating and quiescent cells, and is associated with subtle changes in nucleotide homeostasis protein levels and evidence of disturbed mtDNA maintenance in skeletal muscle. Despite confirmation of GMPR deficiency, demonstrating marked defects of mtDNA replication or nucleotide homeostasis in patient cells proved challenging. Our study proposes that GMPR is the 19th locus for PEO and highlights the complexities of uncovering disease mechanisms in late-onset PEO phenotypes.


Asunto(s)
ADN Mitocondrial/genética , GMP-Reductasa/genética , Enfermedades de Inicio Tardío/genética , Músculo Esquelético/enzimología , Oftalmoplejía/genética , Adenina/metabolismo , Anciano , Células Cultivadas , Deficiencia de Citocromo-c Oxidasa/metabolismo , Replicación del ADN , ADN Mitocondrial/metabolismo , Femenino , Fibroblastos/enzimología , GMP-Reductasa/deficiencia , GMP-Reductasa/metabolismo , Guanina/metabolismo , Células HEK293 , Células HeLa , Heterocigoto , Humanos , Enfermedades de Inicio Tardío/metabolismo , Enfermedades de Inicio Tardío/patología , Músculo Esquelético/patología , Oftalmoplejía/enzimología , Oftalmoplejía/fisiopatología , Fosforilación Oxidativa , Empalme del ARN , Eliminación de Secuencia , Secuenciación del Exoma
6.
Brain Dev ; 41(10): 883-887, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31324350

RESUMEN

BACKGROUND: ARX genetic defect is associated with a spectrum of neurodevelopmental disorders that exhibit a high degree of phenotypic heterogeneity. METHODS: We studied a family with a 13-year old Chinese boy and his two elder brothers presented with infantile epileptic-dyskinetic encephalopathy and clarified the unknown genetic etiology of the youngest brother by whole exome sequencing. RESULTS: The youngest brother of this family presented with developmental regression, dystonia, epilepsy, microcephaly, visual impairment and oromotor dysfunction. Hyperlactataemia, raised alanine and muscle complex IV deficiency indicated that he had mitochondrial dysfunction. Likely pathogenic hemizygous missense ARX variants (c.989G > A; p.Arg330His) located in conserved nuclear localization sequence was identified. The variant was carried by his asymptomatic mother and not found in his asymptomatic third elder brother. The intractable seizures showed complete but transient responsiveness to pyridoxal phosphate and finally controlled by valproate treatment. CONCLUSION: This is the first case of ARX-associated encephalopathy showing mitochondrial dysfunction and transient responsiveness to pyridoxal phosphate treatment.


Asunto(s)
Proteínas de Homeodominio/genética , Espasmos Infantiles/etiología , Espasmos Infantiles/genética , Factores de Transcripción/genética , Adolescente , Adulto , Pueblo Asiatico/genética , China , Deficiencia de Citocromo-c Oxidasa/metabolismo , Trastornos Distónicos/genética , Epilepsia/fisiopatología , Familia , Femenino , Humanos , Discapacidad Intelectual/genética , Masculino , Mitocondrias , Músculo Esquelético/metabolismo , Mutación , Linaje , Fenotipo , Convulsiones/fisiopatología , Espasmos Infantiles/metabolismo , Ácido Valproico/farmacología
7.
Acta Physiol (Oxf) ; 225(4): e13214, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30376218

RESUMEN

AIM: Mitochondrial DNA (mtDNA) mutations can negatively influence lifespan and organ function. More than 250 pathogenic mtDNA mutations are known, often involving neurological symptoms. Major neurodegenerative diseases share key etiopathogenetic components ie mtDNA mutations, mitochondrial dysfunction and oxidative stress. METHODS: Here, we characterized a conplastic mouse strain (C57BL/6 J-mtNOD) carrying an electron transport chain complex IV mutation that leads to an altered cytochrome c oxidase subunit III. Since this mouse also harbours adenine insertions in the mitochondrial tRNA for arginine, we chose the C57BL/6 J-mtMRL as control strain which also carries a heteroplasmic stretch of adenine repetitions in this tRNA isoform. RESULTS: Using MitoSOX fluorescence, we observed an elevated mitochondrial superoxide production and a reduced gene expression of superoxide dismutase 2 in the 24-month-old mtNOD mouse as compared to control. Together with the decreased expression of the fission-relevant gene Fis1, these data confirmed that the ageing mtNOD mouse had a mitochondrial dysfunctional phenotype. On the functional level, we could not detect significant differences in synaptic long-term potentiation, but found a markedly poor physical constitution to perform the Morris water maze task at the age of 24 months. Moreover, the median lifespan of mtNOD mice was significantly shorter than of control animals. CONCLUSION: Our findings demonstrate that a complex IV mutation leads to mitochondrial dysfunction that translates into survival.


Asunto(s)
Deficiencia de Citocromo-c Oxidasa/metabolismo , Complejo IV de Transporte de Electrones/genética , Longevidad/genética , Especies Reactivas de Oxígeno/metabolismo , Animales , Encéfalo/metabolismo , Deficiencia de Citocromo-c Oxidasa/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Técnicas In Vitro , Memoria/fisiología , Ratones Endogámicos C57BL , Dinámicas Mitocondriales/genética , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Superóxidos/metabolismo
8.
J Neurosci Res ; 96(9): 1576-1585, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30113722

RESUMEN

Spinocerebellar ataxias (SCAs) are a genetically heterogeneous group of cerebellar degenerative disorders, characterized by progressive gait unsteadiness, hand incoordination, and dysarthria. Ataxia type 1 (SCA1) is caused by the expansion of a CAG trinucleotide repeat in the SCA1 gene resulting in the atypical extension of a polyglutamine (polyQ) tract within the ataxin-1 protein. Our main objective was to investigate the mitochondrial oxidative metabolism in the cerebellum of transgenic SCA1 mice. SCA1 transgenic mice develop clinical features in the early life stages (around 5 weeks of age) presenting pathological cerebellar signs with concomitant progressive Purkinje neuron atrophy and relatively little cell loss; this evidence suggests that the SCA1 phenotype is not the result of cell death per se, but a possible effect of cellular dysfunction that occurs before neuronal demise. We studied the mitochondrial oxidative metabolism in cerebellar cells from both homozygous and heterozygous transgenic SCA1 mice, aged 2 and 6 months. Histochemical examination showed a cytochrome-c-oxidase (COX) deficiency in the Purkinje cells (PCs) of both heterozygous and homozygous mice, the oxidative defect being more prominent in older mice, in which the percentage of COX-deficient PC was up to 30%. Using a laser-microdissector, we evaluated the mitochondrial DNA (mtDNA) content on selectively isolated COX-competent and COX-deficient PC by quantitative Polymerase Chain Reaction and we found mtDNA depletion in those with oxidative dysfunction. In conclusion, the selective oxidative metabolism defect observed in neuronal PC expressing mutant ataxin occurs as early as 8 weeks of age thus representing an early step in the PC degeneration process in SCA1 disease.


Asunto(s)
Deficiencia de Citocromo-c Oxidasa/metabolismo , ADN Mitocondrial/genética , Células de Purkinje/metabolismo , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/metabolismo , Animales , Ataxina-1/genética , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones Transgénicos , Células de Purkinje/ultraestructura
9.
Hum Mol Genet ; 26(16): 3186-3201, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28575497

RESUMEN

The French-Canadian variant of Leigh Syndrome (LSFC) is an autosomal recessive oxidative phosphorylation (OXPHOS) disorder caused by a mutation in LRPPRC, coding for a protein involved in the stability of mitochondrially-encoded mRNAs. Low levels of LRPPRC are present in all patient tissues, but result in a disproportionately severe OXPHOS defect in the brain and liver, leading to unpredictable subacute metabolic crises. To investigate the impact of the OXPHOS defect in the liver, we analyzed the mitochondrial phenotype in mice harboring an hepatocyte-specific inactivation of Lrpprc. Loss of LRPPRC in the liver caused a generalized growth delay, and typical histological features of mitochondrial hepatopathy. At the molecular level, LRPPRC deficiency caused destabilization of polyadenylated mitochondrial mRNAs, altered mitochondrial ultrastructure, and a severe complex IV (CIV) and ATP synthase (CV) assembly defect. The impact of LRPPRC deficiency was not limited to OXPHOS, but also included impairment of long-chain fatty acid oxidation, a striking dysregulation of the mitochondrial permeability transition pore, and an unsuspected alteration of trans-membrane H2O2 diffusion, which was traced to the ATP synthase assembly defect, and to changes in the lipid composition of mitochondrial membranes. This study underscores the value of mitochondria phenotyping to uncover complex and unexpected mechanisms contributing to the pathophysiology of mitochondrial disorders.


Asunto(s)
Mitocondrias/metabolismo , Proteínas de Neoplasias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Permeabilidad de la Membrana Celular/genética , Deficiencia de Citocromo-c Oxidasa/genética , Deficiencia de Citocromo-c Oxidasa/metabolismo , Modelos Animales de Enfermedad , Metabolismo Energético , Femenino , Hepatocitos/metabolismo , Enfermedad de Leigh/genética , Enfermedad de Leigh/metabolismo , Hígado/metabolismo , Masculino , Ratones , Proteínas Mitocondriales/metabolismo , Proteínas de Neoplasias/deficiencia , Proteínas de Neoplasias/genética , Fosforilación Oxidativa , Poliadenilación , Estabilidad del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Mitocondrial
10.
Free Radic Biol Med ; 106: 184-195, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28189850

RESUMEN

Mitochondrial polarisation is paramount for a variety of cellular functions. Under ischemia, mitochondrial membrane potential (ΔΨm) and proton gradient (ΔpH) are maintained via a reversal of mitochondrial F1Fo ATP synthase (mATPase), which can rapidly deplete ATP and drive cells into energy crisis. We found that under normal conditions in cells with disassembled cytochrome c oxidase complex (COX-deficient HCT116), mATPase maintains ΔΨm at levels only 15-20% lower than in WT cells, and for this utilises relatively little ATP. For a small energy expenditure, mATPase enables mitochondrial ΔpH, protein import, Ca2+ turnover, and supports free radical detoxication machinery enlarged to protect the cells from oxidative damage. Whereas in COX-deficient cells the main source of ATP is glycolysis, the ΔΨm is still maintained upon inhibition of the adenine nucleotide translocators with bongkrekic acid and carboxyatractyloside, indicating that the role of ANTs is redundant, and matrix substrate level phosphorylation alone or in cooperation with ATP-Mg/Pi carriers can continuously support the mATPase activity. Intriguingly, we found that mitochondrial complex III is active, and it contributes not only to free radical production, but also to ΔΨm maintenance and energy budget of COX-deficient cells. Overall, this study demonstrates that F1Fo ATP synthase can support general mitochondrial and cellular functions, working in extremely efficient 'energy saving' reverse mode and flexibly recruiting free radical detoxication and ATP producing / transporting pathways.


Asunto(s)
Carcinoma/metabolismo , Neoplasias del Colon/metabolismo , Metabolismo Energético/genética , Mitocondrias/metabolismo , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Adenosina Trifosfato/metabolismo , Carcinoma/genética , Carcinoma/patología , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Deficiencia de Citocromo-c Oxidasa/genética , Deficiencia de Citocromo-c Oxidasa/metabolismo , Deficiencia de Citocromo-c Oxidasa/patología , Complejo IV de Transporte de Electrones/metabolismo , Células HCT116 , Humanos , Potencial de la Membrana Mitocondrial , Mitocondrias/patología , Fosforilación Oxidativa
11.
J Med Genet ; 53(12): 846-849, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27683825

RESUMEN

BACKGROUND: Assembly of cytochrome c oxidase (COX, complex IV, cIV), the terminal component of the mitochondrial respiratory chain, is assisted by several factors, most of which are conserved from yeast to humans. However, some of them, including COA7, are found in humans but not in yeast. COA7 is a 231aa-long mitochondrial protein present in animals, containing five Sel1-like tetratricopeptide repeat sequences, which are likely to interact with partner proteins. METHODS: Whole exome sequencing was carried out on a 19 year old woman, affected by early onset, progressive severe ataxia and peripheral neuropathy, mild cognitive impairment and a cavitating leukodystrophy of the brain with spinal cord hypotrophy. Biochemical analysis of the mitochondrial respiratory chain revealed the presence of isolated deficiency of cytochrome c oxidase (COX) activity in skin fibroblasts and skeletal muscle. Mitochondrial localization studies were carried out in isolated mitochondria and mitoplasts from immortalized control human fibroblasts. RESULTS: We found compound heterozygous mutations in COA7: a paternal c.410A>G, p.Y137C, and a maternal c.287+1G>T variants. Lentiviral-mediated expression of recombinant wild-type COA7 cDNA in the patient fibroblasts led to the recovery of the defect in COX activity and restoration of normal COX amount. In mitochondrial localization experiments, COA7 behaved as the soluble matrix protein Citrate Synthase. CONCLUSIONS: We report here the first patient carrying pathogenic mutations of COA7, causative of isolated COX deficiency and progressive neurological impairment. We also show that COA7 is a soluble protein localized to the matrix, rather than in the intermembrane space as previously suggested.


Asunto(s)
Deficiencia de Citocromo-c Oxidasa/metabolismo , Leucoencefalopatías/metabolismo , Proteínas Mitocondriales/genética , Mutación , Secuencia de Aminoácidos , Deficiencia de Citocromo-c Oxidasa/genética , Análisis Mutacional de ADN , Femenino , Humanos , Leucoencefalopatías/genética , Mitocondrias , Proteínas Mitocondriales/química , Alineación de Secuencia , Adulto Joven
12.
Cell Death Dis ; 7: e2309, 2016 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-27468686

RESUMEN

Mutations in the opa1 (optic atrophy 1) gene lead to autosomal dominant optic atrophy (ADOA), a hereditary eye disease. This gene encodes the Opa1 protein, a mitochondrial dynamin-related GTPase required for mitochondrial fusion and the maintenance of normal crista structure. The majority of opa1 mutations encode truncated forms of the protein, lacking a complete GTPase domain. It is unclear whether the phenotype results from haploinsufficiency or rather a deleterious effect of truncated Opa1 protein. We studied a heterozygous Opa1 mutant mouse carrying a defective allele with a stop codon in the beginning of the GTPase domain at residue 285, a mutation that mimics human pathological mutations. Using an antibody raised against an N-terminal portion of Opa1, we found that the level of wild-type protein was decreased in the mutant mice, as predicted. However, no truncated Opa1 protein was expressed. In embryonic fibroblasts isolated from the mutant mice, this partial loss of Opa1 caused mitochondrial respiratory deficiency and a selective loss of respiratory Complex IV subunits. Furthermore, partial Opa1 deficiency resulted in a substantial resistance to endoplasmic reticulum stress-induced death. On the other hand, the enforced expression of truncated Opa1 protein in cells containing normal levels of wild-type protein did not cause mitochondrial defects. Moreover, cells expressing the truncated Opa1 protein showed reduced Bax activation in response to apoptotic stimuli. Taken together, our results exclude deleterious dominant-negative or gain-of-function mechanisms for this type of Opa1 mutation and affirm haploinsufficiency as the mechanism underlying mitochondrial dysfunction in ADOA.


Asunto(s)
Deficiencia de Citocromo-c Oxidasa/genética , Complejo IV de Transporte de Electrones/genética , GTP Fosfohidrolasas/genética , Haploinsuficiencia , Mitocondrias/genética , Atrofia Óptica Autosómica Dominante/genética , Alelos , Animales , Deficiencia de Citocromo-c Oxidasa/metabolismo , Deficiencia de Citocromo-c Oxidasa/patología , Modelos Animales de Enfermedad , Complejo IV de Transporte de Electrones/metabolismo , Embrión de Mamíferos , Estrés del Retículo Endoplásmico/genética , Fibroblastos/metabolismo , Fibroblastos/patología , GTP Fosfohidrolasas/deficiencia , Regulación de la Expresión Génica , Células HeLa , Heterocigoto , Humanos , Ratones , Mitocondrias/metabolismo , Mitocondrias/patología , Mutación , Atrofia Óptica Autosómica Dominante/metabolismo , Atrofia Óptica Autosómica Dominante/patología , Cultivo Primario de Células , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
13.
Sci Rep ; 6: 25441, 2016 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-27146429

RESUMEN

Isomorphic mutation of the SBDS gene causes Shwachman-Diamond syndrome (SDS). SDS is a rare genetic bone marrow failure and cancer predisposition syndrome. SDS cells have ribosome biogenesis and their protein synthesis altered, which are two high-energy consuming cellular processes. The reported changes in reactive oxygen species production, endoplasmic reticulum stress response and reduced mitochondrial functionality suggest an energy production defect in SDS cells. In our work, we have demonstrated that SDS cells display a Complex IV activity impairment, which causes an oxidative phosphorylation metabolism defect, with a consequent decrease in ATP production. These data were confirmed by an increased glycolytic rate, which compensated for the energetic stress. Moreover, the signalling pathways involved in glycolysis activation also appeared more activated; i.e. we reported AMP-activated protein kinase hyper-phosphorylation. Notably, we also observed an increase in a mammalian target of rapamycin phosphorylation and high intracellular calcium concentration levels ([Ca(2+)]i), which probably represent new biochemical equilibrium modulation in SDS cells. Finally, the SDS cell response to leucine (Leu) was investigated, suggesting its possible use as a therapeutic adjuvant to be tested in clinical trials.


Asunto(s)
Células de la Médula Ósea/metabolismo , Enfermedades de la Médula Ósea/metabolismo , Calcio/metabolismo , Deficiencia de Citocromo-c Oxidasa/metabolismo , Insuficiencia Pancreática Exocrina/metabolismo , Lipomatosis/metabolismo , Mitocondrias/metabolismo , Proteínas/genética , Ribosomas/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Adenosina Trifosfato/deficiencia , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/patología , Enfermedades de la Médula Ósea/genética , Enfermedades de la Médula Ósea/patología , Deficiencia de Citocromo-c Oxidasa/genética , Deficiencia de Citocromo-c Oxidasa/patología , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/genética , Insuficiencia Pancreática Exocrina/genética , Insuficiencia Pancreática Exocrina/patología , Regulación de la Expresión Génica , Glucólisis/genética , Humanos , Leucina/farmacología , Lipomatosis/genética , Lipomatosis/patología , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Mutación , Fosforilación , Cultivo Primario de Células , Biosíntesis de Proteínas , Proteínas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Ribosomas/efectos de los fármacos , Ribosomas/patología , Síndrome de Shwachman-Diamond , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
14.
Cell Death Dis ; 6: e1691, 2015 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-25766330

RESUMEN

Although numerous pathogenic changes within the mitochondrial respiratory chain (RC) have been associated with an elevated occurrence of apoptosis within the affected tissues, the mechanistic insight into how mitochondrial dysfunction initiates apoptotic cell death is still unknown. In this study, we show that the specific alteration of the cytochrome c oxidase (COX), representing a common defect found in mitochondrial diseases, facilitates mitochondrial apoptosis in response to oxidative stress. Our data identified an increased ceramide synthase 6 (CerS6) activity as an important pro-apoptotic response to COX dysfunction induced either by chemical or genetic approaches. The elevated CerS6 activity resulted in accumulation of the pro-apoptotic C16 : 0 ceramide, which facilitates the mitochondrial apoptosis in response to oxidative stress. Accordingly, inhibition of CerS6 or its specific knockdown diminished the increased susceptibility of COX-deficient cells to oxidative stress. Our results provide new insights into how mitochondrial RC dysfunction mechanistically interferes with the apoptotic machinery. On the basis of its pivotal role in regulating cell death upon COX dysfunction, CerS6 might potentially represent a novel target for therapeutic intervention in mitochondrial diseases caused by COX dysfunction.


Asunto(s)
Apoptosis/genética , Deficiencia de Citocromo-c Oxidasa/metabolismo , Complejo IV de Transporte de Electrones/biosíntesis , Proteínas de la Membrana/biosíntesis , Esfingosina N-Aciltransferasa/biosíntesis , Animales , Deficiencia de Citocromo-c Oxidasa/genética , Deficiencia de Citocromo-c Oxidasa/patología , Complejo IV de Transporte de Electrones/genética , Células HeLa , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Ratones , Mitocondrias/metabolismo , Mitocondrias/patología , Estrés Oxidativo , Consumo de Oxígeno , Esfingosina N-Aciltransferasa/antagonistas & inhibidores
15.
Cell Metab ; 21(1): 109-16, 2015 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-25565209

RESUMEN

Mitochondrial calcium is an important modulator of cellular metabolism. CCDC90A was reported to be a regulator of the mitochondrial calcium uniporter (MCU) complex, a selective channel that controls mitochondrial calcium uptake, and hence was renamed MCUR1. Here we show that suppression of CCDC90A in human fibroblasts produces a specific cytochrome c oxidase (COX) assembly defect, resulting in decreased mitochondrial membrane potential and reduced mitochondrial calcium uptake capacity. Fibroblasts from patients with COX assembly defects due to mutations in TACO1 or COX10 also showed reduced mitochondrial membrane potential and impaired calcium uptake capacity, both of which were rescued by expression of the respective wild-type cDNAs. Deletion of fmp32, a homolog of CCDC90A in Saccharomyces cerevisiae, an organism that lacks an MCU, also produces a COX deficiency, demonstrating that the function of CCDC90A is evolutionarily conserved. We conclude that CCDC90A plays a role in COX assembly and does not directly regulate MCU.


Asunto(s)
Canales de Calcio/metabolismo , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Transferasas Alquil y Aril/genética , Transferasas Alquil y Aril/metabolismo , Calcio/metabolismo , Células Cultivadas , Deficiencia de Citocromo-c Oxidasa/metabolismo , Deficiencia de Citocromo-c Oxidasa/patología , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Potencial de la Membrana Mitocondrial , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/genética , Mutación , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
16.
Hum Mol Genet ; 23(11): 2901-13, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24403053

RESUMEN

Cytochrome c oxidase (CIV) deficiency is one of the most common respiratory chain defects in patients presenting with mitochondrial encephalocardiomyopathies. CIV biogenesis is complicated by the dual genetic origin of its structural subunits, and assembly of a functional holoenzyme complex requires a large number of nucleus-encoded assembly factors. In general, the functions of these assembly factors remain poorly understood, and mechanistic investigations of human CIV biogenesis have been limited by the availability of model cell lines. Here, we have used small interference RNA and transcription activator-like effector nucleases (TALENs) technology to create knockdown and knockout human cell lines, respectively, to study the function of the CIV assembly factor COX20 (FAM36A). These cell lines exhibit a severe, isolated CIV deficiency due to instability of COX2, a mitochondrion-encoded CIV subunit. Mitochondria lacking COX20 accumulate CIV subassemblies containing COX1 and COX4, similar to those detected in fibroblasts from patients carrying mutations in the COX2 copper chaperones SCO1 and SCO2. These results imply that in the absence of COX20, COX2 is inefficiently incorporated into early CIV subassemblies. Immunoprecipitation assays using a stable COX20 knockout cell line expressing functional COX20-FLAG allowed us to identify an interaction between COX20 and newly synthesized COX2. Additionally, we show that SCO1 and SCO2 act on COX20-bound COX2. We propose that COX20 acts as a chaperone in the early steps of COX2 maturation, stabilizing the newly synthesized protein and presenting COX2 to its metallochaperone module, which in turn facilitates the incorporation of mature COX2 into the CIV assembly line.


Asunto(s)
Proteínas Portadoras/metabolismo , Ciclooxigenasa 2/metabolismo , Deficiencia de Citocromo-c Oxidasa/enzimología , Complejo IV de Transporte de Electrones/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas Portadoras/genética , Línea Celular , Ciclooxigenasa 2/genética , Deficiencia de Citocromo-c Oxidasa/genética , Deficiencia de Citocromo-c Oxidasa/metabolismo , Complejo IV de Transporte de Electrones/química , Complejo IV de Transporte de Electrones/genética , Humanos , Proteínas de la Membrana/genética , Mitocondrias/enzimología , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Chaperonas Moleculares , Estabilidad Proteica
17.
Hum Mol Genet ; 23(8): 2078-93, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24293544

RESUMEN

Mitochondrial dysfunction is a significant factor in human disease, ranging from systemic disorders of childhood to cardiomyopathy, ischaemia and neurodegeneration. Cytochrome oxidase, the terminal enzyme of the mitochondrial respiratory chain, is a frequent target. Lower eukaryotes possess alternative respiratory-chain enzymes that provide non-proton-translocating bypasses for respiratory complexes I (single-subunit reduced nicotinamide adenine dinucleotide dehydrogenases, e.g. Ndi1 from yeast) or III + IV [alternative oxidase (AOX)], under conditions of respiratory stress or overload. In previous studies, it was shown that transfer of yeast Ndi1 or Ciona intestinalis AOX to Drosophila was able to overcome the lethality produced by toxins or partial knockdown of complex I or IV. Here, we show that AOX can provide a complete or substantial rescue of a range of phenotypes induced by global or tissue-specific knockdown of different cIV subunits, including integral subunits required for catalysis, as well as peripheral subunits required for multimerization and assembly. AOX was also able to overcome the pupal lethality produced by muscle-specific knockdown of subunit CoVb, although the rescued flies were short lived and had a motility defect. cIV knockdown in neurons was not lethal during development but produced a rapidly progressing locomotor and seizure-sensitivity phenotype, which was substantially alleviated by AOX. Expression of Ndi1 exacerbated the neuronal phenotype produced by cIV knockdown. Ndi1 expressed in place of essential cI subunits produced a distinct residual phenotype of delayed development, bang sensitivity and male sterility. These findings confirm the potential utility of alternative respiratory chain enzymes as tools to combat mitochondrial disease, while indicating important limitations thereof.


Asunto(s)
Animales Modificados Genéticamente/metabolismo , Deficiencia de Citocromo-c Oxidasa/complicaciones , Discapacidades del Desarrollo/prevención & control , Drosophila melanogaster/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Infertilidad Masculina/prevención & control , Proteínas Mitocondriales/metabolismo , Enfermedades Neurodegenerativas/prevención & control , Oxidorreductasas/metabolismo , Proteínas de Plantas/metabolismo , Animales , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/crecimiento & desarrollo , Western Blotting , Células Cultivadas , Deficiencia de Citocromo-c Oxidasa/genética , Deficiencia de Citocromo-c Oxidasa/metabolismo , Discapacidades del Desarrollo/etiología , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Complejo IV de Transporte de Electrones/antagonistas & inhibidores , Complejo IV de Transporte de Electrones/genética , Femenino , Humanos , Técnicas para Inmunoenzimas , Infertilidad Masculina/etiología , Masculino , Mitocondrias/metabolismo , Mitocondrias/patología , Proteínas Mitocondriales/genética , Enfermedades Neurodegenerativas/etiología , Oxidorreductasas/genética , Fenotipo , Proteínas de Plantas/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Hum Mol Genet ; 23(8): 2106-19, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24365713

RESUMEN

Mitochondrial respiratory chain (RC) disorders are the most prevalent inborn metabolic diseases and remain without effective treatment to date. Up-regulation of residual enzyme activity has been proposed as a possible therapeutic approach in this group of disorders. As resveratrol (RSV), a natural compound, was proposed to stimulate mitochondrial metabolism in rodents, we tested the effect of this compound on mitochondrial functions in control or in Complex I (CI)- or Complex IV (CIV)-deficient patients' fibroblasts. We show that RSV stimulates the expression of a panel of proteins representing structural subunits or assembly factors of the five RC complexes, in control fibroblasts. In moderate RC-deficient patients' cells, RSV treatment increases the amount of mutated proteins and stimulates residual enzyme activities. In these patients' cells, we establish that up-regulation of RC enzyme activities induced by RSV translates into increased cellular O2 consumption rates and results in the correction of RC deficiencies. Importantly, RSV also prevents the accumulation of lactate that occurred in RC-deficient fibroblasts. Different complementary approaches demonstrate that RSV induces a mitochondrial biogenesis that might underlie the increase in mitochondrial capacities. Finally, we showed that, in human fibroblasts, RSV stimulated mitochondrial functions mainly in a SIRT1- and AMPK-independent manner and that its effects rather involved the estrogen receptor (ER) and estrogen-related receptor alpha (ERRα) signaling pathways. These results represent the first demonstration that RSV could have a beneficial effect on inborn CI and CIV deficiencies from nuclear origin, in human fibroblasts and might be clinically relevant for the treatment of some RC deficiencies.


Asunto(s)
Deficiencia de Citocromo-c Oxidasa/tratamiento farmacológico , Complejo IV de Transporte de Electrones/metabolismo , Receptor alfa de Estrógeno/metabolismo , Fibroblastos/efectos de los fármacos , Receptores de Estrógenos/metabolismo , Piel/efectos de los fármacos , Estilbenos/farmacología , Anticarcinógenos/farmacología , Western Blotting , Células Cultivadas , Deficiencia de Citocromo-c Oxidasa/metabolismo , Deficiencia de Citocromo-c Oxidasa/patología , Transporte de Electrón/efectos de los fármacos , Complejo I de Transporte de Electrón/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Lactatos , Membranas Mitocondriales/metabolismo , Consumo de Oxígeno/efectos de los fármacos , Piruvatos , ARN Interferente Pequeño/genética , Resveratrol , Transducción de Señal/efectos de los fármacos , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/genética , Sirtuina 1/metabolismo , Piel/metabolismo , Piel/patología , Receptor Relacionado con Estrógeno ERRalfa
19.
Biochimie ; 100: 184-91, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24012549

RESUMEN

The five complexes of the mitochondrial respiratory chain (MRC) supply most organs and tissues with ATP produced by oxidative phosphorylation (OXPHOS). Inherited mitochondrial diseases affecting OXPHOS dysfunction are heterogeneous; symptoms may present at any age and may affect a wide range of tissues, with many diseases giving rise to devastating multisystemic disorders resulting in neonatal death. Combined respiratory chain deficiency with normal complex II accounts for a third of all respiratory deficiencies; mutations in nuclear-encoded components of the mitochondrial translation machinery account for many cases. Although mutations have been identified in over 20 such genes and our understanding of the mitochondrial translation apparatus is increasing, to date no definitive cure for these disorders exists. We evaluated the effect of seven small molecules with reported therapeutic potential in fibroblasts of four patients with combined respiratory complex disorders, each harboring a known mutation in a different nuclear-encoded component of the mitochondrial translation machinery: EFTs, GFM1, MRPS22 and TRMU. Six mitochondrial parameters were screened as follows; growth in glucose-free medium, reactive oxygen species (ROS) production, ATP content, mitochondrial content, mitochondrial membrane potential and complex IV activity. It was clearly evident that each patient displayed an individual response and there was no universally beneficial compound. AICAR increased complex IV activity in GFM1 cells and increased ATP content in MRPS22 fibroblasts but was detrimental to TRMU, who benefitted from bezafibrate. Two antioxidants, ascorbate and N-acetylcysteine (NAC), significantly improved cell growth, ATP content and mitochondrial membrane potential and decreased levels of intracellular reactive oxygen species (ROS) in EFTs fibroblasts. This study presents an expanded repertoire of assays that can be performed using the microtiter screening system with a small number of patients' fibroblasts and highlights some therapeutic options while providing additional evidence for the importance of personalized medicine in mitochondrial disorders.


Asunto(s)
Deficiencia de Citocromo-c Oxidasa/genética , Fibroblastos/efectos de los fármacos , Miopatías Mitocondriales/genética , Biosíntesis de Proteínas/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Acetilcisteína/farmacología , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Bezafibrato/farmacología , Deficiencia de Citocromo-c Oxidasa/metabolismo , Deficiencia de Citocromo-c Oxidasa/patología , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Transporte de Electrón/efectos de los fármacos , Transporte de Electrón/genética , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Regulación de la Expresión Génica , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/patología , Miopatías Mitocondriales/metabolismo , Miopatías Mitocondriales/patología , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Mutación , Fosforilación Oxidativa/efectos de los fármacos , Factor G de Elongación Peptídica/genética , Factor G de Elongación Peptídica/metabolismo , Cultivo Primario de Células , Especies Reactivas de Oxígeno/metabolismo , Ribonucleótidos/farmacología , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , ARNt Metiltransferasas/genética , ARNt Metiltransferasas/metabolismo
20.
Hum Mol Genet ; 22(15): 3138-51, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23575228

RESUMEN

It is estimated that the human mitochondrial proteome consists of 1000-1500 distinct proteins. The majority of these support the various biochemical pathways that are active in these organelles. Individuals with an oxidative phosphorylation disorder of unknown cause provide a unique opportunity to identify novel genes implicated in mitochondrial biology. We identified a homozygous deletion of CEP89 in a patient with isolated complex IV deficiency, intellectual disability and multisystemic problems. CEP89 is a ubiquitously expressed and highly conserved gene of unknown function. Immunocytochemistry and cellular fractionation experiments showed that CEP89 is present both in the cytosol and in the mitochondrial intermembrane space. Furthermore, we ascertained in vitro that downregulation of CEP89 resulted in a severe decrease in complex IV in-gel activity and altered mobility, suggesting that the complex is aberrantly formed. Two-dimensional BN-SDS gel analysis revealed that CEP89 associates with a high-molecular weight complex. Together, these data confirm a role for CEP89 in mitochondrial metabolism. In addition, we modeled CEP89 loss of function in Drosophila. Ubiquitous knockdown of fly Cep89 decreased complex IV activity and resulted in complete lethality. Furthermore, Cep89 is required for mitochondrial integrity, membrane depolarization and synaptic transmission of photoreceptor neurons, and for (sub)synaptic organization of the larval neuromuscular junction. Finally, we tested neuronal Cep89 knockdown flies in the light-off jump reflex habituation assay, which revealed its role in learning. We conclude that CEP89 proteins play an important role in mitochondrial metabolism, especially complex IV activity, and are required for neuronal and cognitive function across evolution.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Mitocondrias/metabolismo , Neuronas/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Niño , Cromosomas Humanos Par 19 , Deficiencia de Citocromo-c Oxidasa/genética , Deficiencia de Citocromo-c Oxidasa/metabolismo , Citosol , Modelos Animales de Enfermedad , Drosophila/genética , Proteínas de Drosophila/genética , Femenino , Eliminación de Gen , Expresión Génica , Técnicas de Silenciamiento del Gen , Homocigoto , Humanos , Aprendizaje , Proteínas Asociadas a Microtúbulos , Mitocondrias/genética , Mutación , Especificidad de Órganos/genética , Polimorfismo de Nucleótido Simple , Transporte de Proteínas , Sinapsis/genética , Sinapsis/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...