Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Semin Cell Dev Biol ; 98: 26-33, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31175937

RESUMEN

The thriving field that constitutes cancer metabolism has unveiled some groundbreaking facts over the past two decades, at the heart of which is the TCA cycle and its intermediates. As such and besides its metabolic role, α-ketoglutarate was shown to withstand a wide range of physiological reactions from protection against oxidative stress, collagen and bone maintenance to development and immunity. Most importantly, it constitutes the rate-limiting substrate of 2-oxoglutarate-dependent dioxygenases family enzymes, which are involved in hypoxia sensing and in the shaping of cellular epigenetic landscape, two major drivers of oncogenic transformation. Based on literature reports, we hereby review the benefits of this metabolite as a possible novel adjuvant therapeutic opportunity to target tumor progression. This article is part of the special issue "Mitochondrial metabolic alterations in cancer cells and related therapeutic targets".


Asunto(s)
Ácidos Cetoglutáricos/metabolismo , Neoplasias/metabolismo , Animales , Progresión de la Enfermedad , Humanos , Mitocondrias/metabolismo , Neoplasias/patología
2.
Molecules ; 27(4)2022 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-35209128

RESUMEN

The finding that the most common mitochondrial DNA mutation m.11778G>A/MT-ND4 (p.R340H) associated with Leber's hereditary optic neuropathy (LHON) induces rotenone resistance has produced a long-standing debate, because it contrasts structural evidence showing that the ND4 subunit is far away from the quinone-reaction site in complex I, where rotenone acts. However, recent cryo-electron microscopy data revealed that rotenone also binds to the ND4 subunit. We investigated the possible structural modifications induced by the LHON mutation and found that its amino acid replacement would disrupt a possible hydrogen bond between native R340 and Q139 in ND4, thereby destabilizing rotenone binding. Our analysis thus explains rotenone resistance in LHON patients as a biochemical signature of its pathogenic effect on complex I.


Asunto(s)
Alelos , Sustitución de Aminoácidos , Resistencia a Medicamentos/genética , Complejo I de Transporte de Electrón/genética , Mutación , Atrofia Óptica Hereditaria de Leber/genética , Rotenona/farmacología , Secuencia de Aminoácidos , Sitios de Unión , Secuencia Conservada , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo , Modelos Moleculares , Atrofia Óptica Hereditaria de Leber/metabolismo , Unión Proteica , Conformación Proteica , Rotenona/química , Relación Estructura-Actividad , Desacopladores/farmacología
3.
PLoS Genet ; 14(2): e1007210, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29444077

RESUMEN

We here report on the existence of Leber's hereditary optic neuropathy (LHON) associated with peculiar combinations of individually non-pathogenic missense mitochondrial DNA (mtDNA) variants, affecting the MT-ND4, MT-ND4L and MT-ND6 subunit genes of Complex I. The pathogenic potential of these mtDNA haplotypes is supported by multiple evidences: first, the LHON phenotype is strictly inherited along the maternal line in one very large family; second, the combinations of mtDNA variants are unique to the two maternal lineages that are characterized by recurrence of LHON; third, the Complex I-dependent respiratory and oxidative phosphorylation defect is co-transferred from the proband's fibroblasts into the cybrid cell model. Finally, all but one of these missense mtDNA variants cluster along the same predicted fourth E-channel deputed to proton translocation within the transmembrane domain of Complex I, involving the ND1, ND4L and ND6 subunits. Hence, the definition of the pathogenic role of a specific mtDNA mutation becomes blurrier than ever and only an accurate evaluation of mitogenome sequence variation data from the general population, combined with functional analyses using the cybrid cell model, may lead to final validation. Our study conclusively shows that even in the absence of a clearly established LHON primary mutation, unprecedented combinations of missense mtDNA variants, individually known as polymorphisms, may lead to reduced OXPHOS efficiency sufficient to trigger LHON. In this context, we introduce a new diagnostic perspective that implies the complete sequence analysis of mitogenomes in LHON as mandatory gold standard diagnostic approach.


Asunto(s)
ADN Mitocondrial/genética , Herencia Multifactorial , Mutación Missense , Atrofia Óptica Hereditaria de Leber/genética , Penetrancia , Adulto , Secuencia de Aminoácidos , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/genética , Epistasis Genética , Familia , Femenino , Genes Mitocondriales , Humanos , Masculino , Modelos Moleculares , NADH Deshidrogenasa/química , NADH Deshidrogenasa/genética , Linaje , Adulto Joven
4.
Proc Natl Acad Sci U S A ; 114(43): E9066-E9075, 2017 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-29073103

RESUMEN

The horizontal transfer of mtDNA and its role in mediating resistance to therapy and an exit from dormancy have never been investigated. Here we identified the full mitochondrial genome in circulating extracellular vesicles (EVs) from patients with hormonal therapy-resistant (HTR) metastatic breast cancer. We generated xenograft models of HTR metastatic disease characterized by EVs in the peripheral circulation containing mtDNA. Moreover, these human HTR cells had acquired host-derived (murine) mtDNA promoting estrogen receptor-independent oxidative phosphorylation (OXPHOS). Functional studies identified cancer-associated fibroblast (CAF)-derived EVs (from patients and xenograft models) laden with whole genomic mtDNA as a mediator of this phenotype. Specifically, the treatment of hormone therapy (HT)-naive cells or HT-treated metabolically dormant populations with CAF-derived mtDNAhi EVs promoted an escape from metabolic quiescence and HTR disease both in vitro and in vivo. Moreover, this phenotype was associated with the acquisition of EV mtDNA, especially in cancer stem-like cells, expression of EV mtRNA, and restoration of OXPHOS. In summary, we have demonstrated that the horizontal transfer of mtDNA from EVs acts as an oncogenic signal promoting an exit from dormancy of therapy-induced cancer stem-like cells and leading to endocrine therapy resistance in OXPHOS-dependent breast cancer.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , ADN Mitocondrial/metabolismo , Resistencia a Antineoplásicos/genética , Exosomas/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , ADN Mitocondrial/genética , Femenino , Fibroblastos/patología , Transferencia de Gen Horizontal , Genoma Mitocondrial/genética , Humanos , Células MCF-7 , NADH Deshidrogenasa/genética , Fosforilación Oxidativa , Receptores de Estrógenos/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Neurobiol Dis ; 124: 14-28, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30389403

RESUMEN

Spinocerebellar ataxia 28 is an autosomal dominant neurodegenerative disorder caused by missense mutations affecting the proteolytic domain of AFG3L2, a major component of the mitochondrial m-AAA protease. However, little is known of the underlying pathogenetic mechanisms or how to treat patients with SCA28. Currently available Afg3l2 mutant mice harbour deletions that lead to severe, early-onset neurological phenotypes that do not faithfully reproduce the late-onset and slowly progressing SCA28 phenotype. Here we describe production and detailed analysis of a new knock-in murine model harbouring an Afg3l2 allele carrying the p.Met665Arg patient-derived mutation. Heterozygous mutant mice developed normally but adult mice showed signs of cerebellar ataxia detectable by beam test. Although cerebellar pathology was negative, electrophysiological analysis showed a trend towards increased spontaneous firing in Purkinje cells from heterozygous mutants with respect to wild-type controls. As homozygous mutants died perinatally with evidence of cardiac atrophy, for each genotype we generated mouse embryonic fibroblasts (MEFs) to investigate mitochondrial function. MEFs from mutant mice showed altered mitochondrial bioenergetics, with decreased basal oxygen consumption rate, ATP synthesis and mitochondrial membrane potential. Mitochondrial network formation and morphology was altered, with greatly reduced expression of fusogenic Opa1 isoforms. Mitochondrial alterations were also detected in cerebella of 18-month-old heterozygous mutants and may be a hallmark of disease. Pharmacological inhibition of de novo mitochondrial protein translation with chloramphenicol caused reversal of mitochondrial morphology in homozygous mutant MEFs, supporting the relevance of mitochondrial proteotoxicity for SCA28 pathogenesis and therapy development.


Asunto(s)
Proteasas ATP-Dependientes/genética , ATPasas Asociadas con Actividades Celulares Diversas/genética , Modelos Animales de Enfermedad , Mitocondrias/metabolismo , Ataxias Espinocerebelosas/congénito , Animales , Femenino , Técnicas de Sustitución del Gen , Potencial de la Membrana Mitocondrial , Ratones Endogámicos C57BL , Proteínas Mitocondriales/metabolismo , Mutación Missense , Células de Purkinje/fisiología , Células de Purkinje/ultraestructura , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/metabolismo , Ataxias Espinocerebelosas/patología
6.
Hum Mol Genet ; 26(15): 2961-2974, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28486623

RESUMEN

Development of chemoresistance is a cogent clinical issue in oncology, whereby combination of anticancer drugs is usually preferred also to enhance efficacy. Paclitaxel (PTX), combined with carboplatin, represents the standard first-line chemotherapy for different types of cancers. We here depict a double-edge role of mitochondrial DNA (mtDNA) mutations induced in cancer cells after treatment with platinum. MtDNA mutations were positively selected by PTX, and they determined a decrease in the mitochondrial respiratory function, as well as in proliferative and tumorigenic potential, in terms of migratory and invasive capacity. Moreover, cells bearing mtDNA mutations lacked filamentous tubulin, the main target of PTX, and failed to reorient the Golgi body upon appropriate stimuli. We also show that the bioenergetic and cytoskeletal phenotype were transferred along with mtDNA mutations in transmitochondrial hybrids, and that this also conferred PTX resistance to recipient cells. Overall, our data show that platinum-induced deleterious mtDNA mutations confer resistance to PTX, and confirm what we previously reported in an ovarian cancer patient treated with carboplatin and PTX who developed a quiescent yet resistant tumor mass harboring mtDNA mutations.


Asunto(s)
ADN Mitocondrial/efectos de los fármacos , ADN Mitocondrial/metabolismo , Paclitaxel/metabolismo , Antineoplásicos/farmacología , Carboplatino/metabolismo , Línea Celular Tumoral , Citoesqueleto/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Femenino , Humanos , Mutación/efectos de los fármacos , Neoplasias Ováricas/genética , Platino (Metal) , Tubulina (Proteína)/efectos de los fármacos , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
7.
Int J Mol Sci ; 20(12)2019 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-31242642

RESUMEN

The ketogenic diet (KD), a high-fat/low-carbohydrate/adequate-protein diet, has been proposed as a treatment for a variety of diseases, including cancer. KD leads to generation of ketone bodies (KBs), predominantly acetoacetate (AcAc) and 3-hydroxy-butyrate, as a result of fatty acid oxidation. Several studies investigated the antiproliferative effects of lithium acetoacetate (LiAcAc) and sodium 3-hydroxybutyrate on cancer cells in vitro. However, a critical point missed in some studies using LiAcAc is that Li ions have pleiotropic effects on cell growth and cell signaling. Thus, we tested whether Li ions per se contribute to the antiproliferative effects of LiAcAc in vitro. Cell proliferation was analyzed on neuroblastoma, renal cell carcinoma, and human embryonic kidney cell lines. Cells were treated for 5 days with 2.5, 5, and 10 mM LiAcAc and with equimolar concentrations of lithium chloride (LiCl) or sodium chloride (NaCl). LiAcAc affected the growth of all cell lines, either negatively or positively. However, the effects of LiAcAc were always similar to those of LiCl. In contrast, NaCl showed no effects, indicating that the Li ion impacts cell proliferation. As Li ions have significant effects on cell growth, it is important for future studies to include sources of Li ions as a control.


Asunto(s)
Acetoacetatos/farmacología , Litio/farmacología , Caspasa 3/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Cloruros/farmacología , Expresión Génica , Humanos , Cloruro de Litio/farmacología
8.
Hum Mutat ; 39(1): 92-102, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28967163

RESUMEN

Respiratory complex III (CIII) is the first enzymatic bottleneck of the mitochondrial respiratory chain both in its native dimeric form and in supercomplexes. The mammalian CIII comprises 11 subunits among which cytochrome b is central in the catalytic core, where oxidation of ubiquinol occurs at the Qo site. The Qo- or PEWY-motif of cytochrome b is the most conserved through species. Importantly, the highly conserved glutamate at position 271 (Glu271) has never been studied in higher eukaryotes so far and its role in the Q-cycle remains debated. Here, we showed that the homoplasmic m.15557G > A/MT-CYB, which causes the p.Glu271Lys amino acid substitution predicted to dramatically affect CIII, induces a mild mitochondrial dysfunction in human transmitochondrial cybrids. Indeed, we found that the severity of such mutation is mitigated by the proper assembly of CIII into supercomplexes, which may favor an optimal substrate channeling and buffer superoxide production in vitro.


Asunto(s)
Alelos , Citocromos b/genética , Estudios de Asociación Genética , Mutación , Fenotipo , Adenosina Trifosfato , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Línea Celular , Supervivencia Celular/genética , Secuencia Conservada , Complejo III de Transporte de Electrones/genética , Complejo III de Transporte de Electrones/metabolismo , Metabolismo Energético , Humanos , Potencial de la Membrana Mitocondrial , Especies Reactivas de Oxígeno/metabolismo
9.
Neurobiol Dis ; 114: 129-139, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29486301

RESUMEN

There is growing evidence that the sequence variation of mitochondrial DNA (mtDNA), which clusters in population- and/or geographic-specific haplogroups, may result in functional effects that, in turn, become relevant in disease predisposition or protection, interaction with environmental factors and ultimately in modulating longevity. To unravel functional differences between mtDNA haplogroups we here employed transmitochondrial cytoplasmic hybrid cells (cybrids) grown in galactose medium, a culture condition that forces oxidative phosphorylation, and in the presence of rotenone, the classic inhibitor of respiratory Complex I. Under this experimental paradigm we assessed functional parameters such as cell viability and respiration, ATP synthesis, reactive oxygen species production and mtDNA copy number. Our analyses show that haplogroup J1, which is common in western Eurasian populations, is the most sensitive to rotenone, whereas K1 mitogenomes orchestrate the best compensation, possibly because of the haplogroup-specific missense variants impinging on Complex I function. Remarkably, haplogroups J1 and K1 fit the genetic associations previously established with Leber's hereditary optic neuropathy (LHON) for J1, as a penetrance enhancer, and with Parkinson's disease (PD) for K1, as a protective background. Our findings provide functional evidences supporting previous well-established genetic associations of specific haplogroups with two neurodegenerative pathologies, LHON and PD. Our experimental paradigm is instrumental to highlighting the subtle functional differences characterizing mtDNA haplogroups, which will be increasingly needed to dissect the role of mtDNA genetic variation in health, disease and longevity.


Asunto(s)
ADN Mitocondrial/genética , Genoma Mitocondrial/genética , Haplotipos/genética , Enfermedad de Parkinson Secundaria/genética , Plaguicidas/toxicidad , Rotenona/toxicidad , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , ADN Mitocondrial/química , Fibroblastos/efectos de los fármacos , Fibroblastos/fisiología , Genoma Mitocondrial/efectos de los fármacos , Haplotipos/efectos de los fármacos , Humanos , Consumo de Oxígeno/efectos de los fármacos , Consumo de Oxígeno/fisiología , Enfermedad de Parkinson Secundaria/inducido químicamente , Filogenia , Estructura Secundaria de Proteína
10.
Int J Cancer ; 143(7): 1706-1719, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29672841

RESUMEN

Familial aggregation is a significant risk factor for the development of thyroid cancer and familial non-medullary thyroid cancer (FNMTC) accounts for 5-7% of all NMTC. Whole exome sequencing analysis in the family affected by FNMTC with oncocytic features where our group previously identified a predisposing locus on chromosome 19p13.2, revealed a novel heterozygous mutation (c.400G > A, NM_012335; p.Gly134Ser) in exon 5 of MYO1F, mapping to the linkage locus. In the thyroid FRTL-5 cell model stably expressing the mutant MYO1F p.Gly134Ser protein, we observed an altered mitochondrial network, with increased mitochondrial mass and a significant increase in both intracellular and extracellular reactive oxygen species, compared to cells expressing the wild-type (wt) protein or carrying the empty vector. The mutation conferred a significant advantage in colony formation, invasion and anchorage-independent growth. These data were corroborated by in vivo studies in zebrafish, since we demonstrated that the mutant MYO1F p.Gly134Ser, when overexpressed, can induce proliferation in whole vertebrate embryos, compared to the wt one. MYO1F screening in additional 192 FNMTC families identified another variant in exon 7, which leads to exon skipping, and is predicted to alter the ATP-binding domain in MYO1F. Our study identified for the first time a role for MYO1F in NMTC.


Asunto(s)
Proliferación Celular , Embrión no Mamífero/patología , Mitocondrias/patología , Mutación , Miosina Tipo I/genética , Cáncer Papilar Tiroideo/patología , Neoplasias de la Tiroides/patología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Apoptosis , Células Cultivadas , Niño , Cromosomas Humanos Par 19 , Embrión no Mamífero/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Mitocondrias/genética , Mitocondrias/metabolismo , Miosina Tipo I/química , Miosina Tipo I/metabolismo , Consumo de Oxígeno , Linaje , Conformación Proteica , Cáncer Papilar Tiroideo/genética , Cáncer Papilar Tiroideo/metabolismo , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/metabolismo , Adulto Joven , Pez Cebra
11.
Int J Mol Sci ; 19(3)2018 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-29518970

RESUMEN

Mammalian respiratory complex I (CI) biogenesis requires both nuclear and mitochondria-encoded proteins and is mostly organized in respiratory supercomplexes. Among the CI proteins encoded by the mitochondrial DNA, NADH-ubiquinone oxidoreductase chain 1 (ND1) is a core subunit, evolutionary conserved from bacteria to mammals. Recently, ND1 has been recognized as a pivotal subunit in maintaining the structural and functional interaction among the hydrophilic and hydrophobic CI arms. A critical role of human ND1 both in CI biogenesis and in the dynamic organization of supercomplexes has been depicted, although the proof of concept is still missing and the critical amount of ND1 protein necessary for a proper assembly of both CI and supercomplexes is not defined. By exploiting a unique model in which human ND1 is allotopically re-expressed in cells lacking the endogenous protein, we demonstrated that the lack of this protein induces a stall in the multi-step process of CI biogenesis, as well as the alteration of supramolecular organization of respiratory complexes. We also defined a mutation threshold for the m.3571insC truncative mutation in mitochondrially encoded NADH:ubiquinone oxidoreductase core subunit 1 (MT-ND1), below which CI and its supramolecular organization is recovered, strengthening the notion that a certain amount of human ND1 is required for CI and supercomplexes biogenesis.


Asunto(s)
Alelos , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/genética , Mutación , NADH Deshidrogenasa/química , NADH Deshidrogenasa/genética , Respiración de la Célula , ADN Mitocondrial/genética , Complejo I de Transporte de Electrón/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , NADH Deshidrogenasa/metabolismo , Consumo de Oxígeno , Unión Proteica , Relación Estructura-Actividad
12.
Biochim Biophys Acta Bioenerg ; 1858(8): 582-590, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28213331

RESUMEN

Energy homeostasis is pivotal for cell fate since metabolic regulation, cell proliferation and death are strongly dependent on the balance between catabolic and anabolic pathways. In particular, metabolic and energetic changes have been observed in cancer cells even before the discovery of oncogenes and tumor suppressors, but have been neglected for a long time. Instead, during the past 20years a renaissance of the study of tumor metabolism has led to a revised and more accurate sight of the metabolic landscape of cancer cells. In this scenario, genetic, biochemical and clinical evidences place mitochondria as key actors in cancer metabolic restructuring, not only because there are energy and biosynthetic intermediates manufacturers, but also because occurrence of mutations in metabolic enzymes encoded by both nuclear and mitochondrial DNA has been associated to different types of cancer. Here we provide an overview of the possible mechanisms modulating mitochondrial energy production and homeostasis in the intriguing scenario of neoplastic cells, focusing on the double-edged role of 5'-AMP activated protein kinase in cancer metabolism. This article is part of a Special Issue entitled Mitochondria in Cancer, edited by Giuseppe Gasparre, Rodrigue Rossignol and Pierre Sonveaux.


Asunto(s)
Metabolismo Energético , Mitocondrias/metabolismo , Neoplasias/metabolismo , Proteínas Quinasas Activadas por AMP/fisiología , Adenosina Trifosfato/metabolismo , Animales , Progresión de la Enfermedad , Genes Mitocondriales , Homeostasis , Humanos , Ratones , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/fisiología , Modelos Biológicos , Proteínas de Neoplasias/fisiología , Neoplasias/patología , Neoplasias Experimentales/metabolismo , Proteínas Supresoras de Tumor/fisiología
13.
Hum Mol Genet ; 24(24): 6921-31, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26410888

RESUMEN

Leber's hereditary optic neuropathy (LHON) is a maternally inherited blinding disease characterized by degeneration of retinal ganglion cells (RGCs) and consequent optic nerve atrophy. Peculiar features of LHON are incomplete penetrance and gender bias, with a marked male prevalence. Based on the different hormonal metabolism between genders, we proposed that estrogens play a protective role in females and showed that these hormones ameliorate mitochondrial dysfunction in LHON through the estrogen receptors (ERs). We also showed that ERß localize to the mitochondria of RGCs. Thus, targeting ERß may become a therapeutic strategy for LHON specifically aimed at avoiding or delaying the onset of disease in mutation carriers. Here, we tested the effects of ERß targeting on LHON mitochondrial defective metabolism by treating LHON cybrid cells carrying the m.11778G>A mutation with a combination of natural estrogen-like compounds that bind ERß with high selectivity. We demonstrated that these molecules improve cell viability by reducing apoptosis, inducing mitochondrial biogenesis and strongly reducing the levels of reactive oxygen species in LHON cells. These effects were abolished in cells with ERß knockdown by silencing receptor expression or by using specific receptor antagonists. Our observations support the hypothesis that estrogen-like molecules may be useful in LHON prophylactic therapy. This is particularly important for lifelong disease prevention in unaffected LHON mutation carriers. Current strategies attempting to combat degeneration of RGCs during the acute phase of LHON have not been very effective. Implementing a different and preemptive approach with a low risk profile may be very helpful.


Asunto(s)
Receptor beta de Estrógeno/antagonistas & inhibidores , Atrofia Óptica Hereditaria de Leber/prevención & control , Fitoestrógenos/uso terapéutico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Respiración de la Célula , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Receptor beta de Estrógeno/genética , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mutación , Atrofia Óptica Hereditaria de Leber/metabolismo , Biogénesis de Organelos , Estrés Oxidativo/efectos de los fármacos , Consumo de Oxígeno , Células Ganglionares de la Retina/metabolismo
14.
Hum Mol Genet ; 23(6): 1453-66, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24163135

RESUMEN

Mitochondrial DNA mutations are currently investigated as modifying factors impinging on tumor growth and aggressiveness, having been found in virtually all cancer types and most commonly affecting genes encoding mitochondrial complex I (CI) subunits. However, it is still unclear whether they exert a pro- or anti-tumorigenic effect. We here analyzed the impact of three homoplasmic mtDNA mutations (m.3460G>A/MT-ND1, m.3571insC/MT-ND1 and m.3243A>G/MT-TL1) on osteosarcoma progression, chosen since they induce different degrees of oxidative phosphorylation impairment. In fact, the m.3460G>A/MT-ND1 mutation caused only a reduction in CI activity, whereas the m.3571insC/MT-ND1 and the m.3243A>G/MT-TL1 mutations induced a severe structural and functional CI alteration. As a consequence, this severe CI dysfunction determined an energetic defect associated with a compensatory increase in glycolytic metabolism and AMP-activated protein kinase activation. Osteosarcoma cells carrying such marked CI impairment displayed a reduced tumorigenic potential both in vitro and in vivo, when compared with cells with mild CI dysfunction, suggesting that mtDNA mutations may display diverse impact on tumorigenic potential depending on the type and severity of the resulting oxidative phosphorylation dysfunction. The modulation of tumor growth was independent from reactive oxygen species production but correlated with hypoxia-inducible factor 1α stabilization, indicating that structural and functional integrity of CI and oxidative phosphorylation are required for hypoxic adaptation and tumor progression.


Asunto(s)
ADN Mitocondrial/genética , Complejo I de Transporte de Electrón/genética , Metabolismo Energético , NADH Deshidrogenasa/metabolismo , Osteosarcoma/genética , ARN de Transferencia/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Línea Celular Tumoral , Progresión de la Enfermedad , Complejo I de Transporte de Electrón/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Mutagénesis Insercional , NADH Deshidrogenasa/genética , Osteosarcoma/patología , Fosforilación Oxidativa , Mutación Puntual , Especies Reactivas de Oxígeno/metabolismo
15.
Ann Neurol ; 78(1): 21-38, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25820230

RESUMEN

OBJECTIVE: Mounting evidence links neurodegenerative disorders such as Parkinson disease and Alzheimer disease with mitochondrial dysfunction, and recent emphasis has focused on mitochondrial dynamics and quality control. Mitochondrial dynamics and mtDNA maintenance is another link recently emerged, implicating mutations in the mitochondrial fusion genes OPA1 and MFN2 in the pathogenesis of multisystem syndromes characterized by neurodegeneration and accumulation of mtDNA multiple deletions in postmitotic tissues. Here, we report 2 Italian families affected by dominant chronic progressive external ophthalmoplegia (CPEO) complicated by parkinsonism and dementia. METHODS: Patients were extensively studied by optical coherence tomography (OCT) to assess retinal nerve fibers, and underwent muscle and brain magnetic resonance spectroscopy (MRS), and muscle biopsy and fibroblasts were analyzed. Candidate genes were sequenced, and mtDNA was analyzed for rearrangements. RESULTS: Affected individuals displayed a slowly progressive syndrome characterized by CPEO, mitochondrial myopathy, sensorineural deafness, peripheral neuropathy, parkinsonism, and/or cognitive impairment, in most cases without visual complains, but with subclinical loss of retinal nerve fibers at OCT. Muscle biopsies showed cytochrome c oxidase-negative fibers and mtDNA multiple deletions, and MRS displayed defective oxidative metabolism in muscle and brain. We found 2 heterozygous OPA1 missense mutations affecting highly conserved amino acid positions (p.G488R, p.A495V) in the guanosine triphosphatase domain, each segregating with affected individuals. Fibroblast studies showed a reduced amount of OPA1 protein with normal mRNA expression, fragmented mitochondria, impaired bioenergetics, increased autophagy and mitophagy. INTERPRETATION: The association of CPEO and parkinsonism/dementia with subclinical optic neuropathy widens the phenotypic spectrum of OPA1 mutations, highlighting the association of defective mitochondrial dynamics, mtDNA multiple deletions, and altered mitophagy with parkinsonism.


Asunto(s)
Demencia/genética , GTP Fosfohidrolasas/genética , Mutación Missense , Oftalmoplejía Externa Progresiva Crónica/genética , Trastornos Parkinsonianos/genética , Anciano , Demencia/complicaciones , Femenino , Predisposición Genética a la Enfermedad , Humanos , Italia , Masculino , Oftalmoplejía Externa Progresiva Crónica/complicaciones , Trastornos Parkinsonianos/complicaciones , Linaje
16.
Hum Mol Genet ; 22(11): 2141-51, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23418307

RESUMEN

Cytochrome b is the only mtDNA-encoded subunit of the mitochondrial complex III (CIII), the functional bottleneck of the respiratory chain. Previously, the human cytochrome b missense mutation m.15579A>G, which substitutes the Tyr 278 with Cys (p.278Y>C), was identified in a patient with severe exercise intolerance and multisystem manifestations. In this study, we characterized the biochemical properties of cybrids carrying this mutation and report that the homoplasmic p.278Y>C mutation caused a dramatic reduction in the CIII activity and in CIII-driven mitochondrial ATP synthesis. However, the CI, CI + CIII and CII + CIII activities and the rate of ATP synthesis driven by the CI or CII substrate were only partially reduced or unaffected. Consistent with these findings, mutated cybrids maintained the mitochondrial membrane potential in the presence of oligomycin, indicating that it originated from the respiratory electron transport chain. The p.278Y>C mutation enhanced superoxide production, as indicated by direct measurements in mitochondria and by the imbalance of glutathione homeostasis in intact cybrids. Remarkably, although the assembly of CI or CIII was not affected, the examination of respiratory supercomplexes revealed that the amounts of CIII dimer and III2IV1 were reduced, whereas those of I1III2IVn slightly increased. We therefore suggest that the deleterious effects of p.278Y>C mutation on cytochrome b are palliated when CIII is assembled into the supercomplexes I1III2IVn, in contrast to when it is found alone. These findings underline the importance of supramolecular interactions between complexes for maintaining a basal respiratory chain activity and shed light to the molecular basis of disease manifestations associated with this mutation.


Asunto(s)
Citocromos b/genética , Complejo IV de Transporte de Electrones/metabolismo , Mutación , Superóxidos/metabolismo , Adenosina Trifosfato/biosíntesis , Línea Celular , ADN Mitocondrial/genética , Transporte de Electrón/genética , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , Complejo IV de Transporte de Electrones/genética , Metabolismo Energético , Activación Enzimática , Glutatión/metabolismo , Homeostasis/fisiología , Humanos , Mitocondrias/genética , Mitocondrias/metabolismo
17.
Brain ; 137(Pt 2): 335-53, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24369379

RESUMEN

Leber's hereditary optic neuropathy is a maternally inherited blinding disease caused as a result of homoplasmic point mutations in complex I subunit genes of mitochondrial DNA. It is characterized by incomplete penetrance, as only some mutation carriers become affected. Thus, the mitochondrial DNA mutation is necessary but not sufficient to cause optic neuropathy. Environmental triggers and genetic modifying factors have been considered to explain its variable penetrance. We measured the mitochondrial DNA copy number and mitochondrial mass indicators in blood cells from affected and carrier individuals, screening three large pedigrees and 39 independently collected smaller families with Leber's hereditary optic neuropathy, as well as muscle biopsies and cells isolated by laser capturing from post-mortem specimens of retina and optic nerves, the latter being the disease targets. We show that unaffected mutation carriers have a significantly higher mitochondrial DNA copy number and mitochondrial mass compared with their affected relatives and control individuals. Comparative studies of fibroblasts from affected, carriers and controls, under different paradigms of metabolic demand, show that carriers display the highest capacity for activating mitochondrial biogenesis. Therefore we postulate that the increased mitochondrial biogenesis in carriers may overcome some of the pathogenic effect of mitochondrial DNA mutations. Screening of a few selected genetic variants in candidate genes involved in mitochondrial biogenesis failed to reveal any significant association. Our study provides a valuable mechanism to explain variability of penetrance in Leber's hereditary optic neuropathy and clues for high throughput genetic screening to identify the nuclear modifying gene(s), opening an avenue to develop predictive genetic tests on disease risk and therapeutic strategies.


Asunto(s)
ADN Mitocondrial/genética , Recambio Mitocondrial/genética , Atrofia Óptica Hereditaria de Leber/diagnóstico , Atrofia Óptica Hereditaria de Leber/genética , Penetrancia , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad , Linaje , Adulto Joven
18.
Hum Mutat ; 35(8): 954-8, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24863938

RESUMEN

A novel heteroplasmic mitochondrial DNA (mtDNA) microdeletion affecting the cytochrome b gene (MT-CYB) was identified in an Italian female patient with a multisystem disease characterized by sensorineural deafness, cataracts, retinal pigmentary dystrophy, dysphagia, postural and gait instability, and myopathy with prominent exercise intolerance. The deletion is 18-base pair long and encompasses nucleotide positions 15,649-15,666, causing the loss of six amino acids (Ile-Leu-Ala-Met-Ile-Pro) in the protein, but leaving the remaining of the MT-CYB sequence in frame. The defective complex III function was cotransferred with mutant mtDNA in cybrids, thus unequivocally establishing its pathogenic role. Maternal relatives failed to show detectable levels of the deletion in blood and urinary epithelium, suggesting a de novo mutational event. This is the second report of an in-frame intragenic deletion in MT-CYB, which most likely occurred in early stages of embryonic development, associated with a severe multisystem disorder with prominent exercise intolerance.


Asunto(s)
Secuencia de Bases , Citocromos b/genética , Fatiga/genética , Enfermedades Musculares/genética , Eliminación de Secuencia , Adulto , Catarata/genética , Catarata/patología , ADN Mitocondrial/genética , Trastornos de Deglución/genética , Trastornos de Deglución/patología , Fatiga/patología , Femenino , Expresión Génica , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/patología , Humanos , Datos de Secuencia Molecular , Enfermedades Musculares/patología , Epitelio Pigmentado Ocular/patología , Decoloración de Dientes/genética , Decoloración de Dientes/patología
19.
Biochim Biophys Acta ; 1832(3): 445-52, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23246842

RESUMEN

Complex I (CI) deficiency is a frequent cause of mitochondrial disorders and, in most cases, is due to mutations in CI subunit genes encoded by mitochondrial DNA (mtDNA). In this study, we establish the pathogenic role of the heteroplasmic mtDNA m.3890G>A/MT-ND1 (p.R195Q) mutation, which affects an extremely conserved amino acid position in ND1 subunit of CI. This mutation was found in a young-adult male with optic atrophy resembling Leber's hereditary optic neuropathy (LHON) and bilateral brainstem lesions. The only previously reported case with this mutation was a girl with fatal infantile Leigh syndrome with bilateral brainstem lesions. Transfer of the mutant mtDNA in the cybrid cell system resulted in a marked reduction of CI activity and CI-dependent ATP synthesis in the presence of a normally assembled enzyme. These findings establish the pathogenicity of the m.3890G>A/MT-ND1 mutation and remark the link between CI mutations affecting the mtDNA-encoded ND subunits and LHON-like optic atrophy, which may be complicated by bilateral and symmetric lesions affecting the central nervous system. Peculiar to this mutation is the distribution of the brainstem lesions, with sparing of the striatum in both patients.


Asunto(s)
Tronco Encefálico/metabolismo , ADN Mitocondrial/genética , Mutación Missense , NADH Deshidrogenasa/genética , Atrofias Ópticas Hereditarias/genética , Adulto , Secuencia de Aminoácidos , Tronco Encefálico/patología , Línea Celular Tumoral , Femenino , Humanos , Células Híbridas , Lactatos/sangre , Imagen por Resonancia Magnética , Espectroscopía de Resonancia Magnética , Masculino , Datos de Secuencia Molecular , Músculo Esquelético/metabolismo , NADH Deshidrogenasa/metabolismo , Atrofias Ópticas Hereditarias/sangre , Atrofias Ópticas Hereditarias/metabolismo , Linaje , Homología de Secuencia de Aminoácido
20.
Int J Biol Macromol ; 273(Pt 2): 133086, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38871105

RESUMEN

Variants found in the respiratory complex I (CI) subunit genes encoded by mitochondrial DNA can cause severe genetic diseases. However, it is difficult to establish a priori whether a single or a combination of CI variants may impact oxidative phosphorylation. Here we propose a computational approach based on coarse-grained molecular dynamics simulations aimed at investigating new CI variants. One of the primary CI variants associated with the Leber hereditary optic neuropathy (m.14484T>C/MT-ND6) was used as a test case and was investigated alone or in combination with two additional rare CI variants whose role remains uncertain. We found that the primary variant positioned in the E-channel region, which is fundamental for CI function, stiffens the enzyme dynamics. Moreover, a new mechanism for the transition between π- and α-conformation in the helix carrying the primary variant is proposed. This may have implications for the E-channel opening/closing mechanism. Finally, our findings show that one of the rare variants, located next to the primary one, further worsens the stiffening, while the other rare variant does not affect CI function. This approach may be extended to other variants candidate to exert a pathogenic impact on CI dynamics, or to investigate the interaction of multiple variants.


Asunto(s)
Complejo I de Transporte de Electrón , Simulación de Dinámica Molecular , Mutación Missense , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo , Humanos , Atrofia Óptica Hereditaria de Leber/genética , Biología Computacional/métodos , NADH Deshidrogenasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA