Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 157
Filtrar
1.
Sci Rep ; 10(1): 9139, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32499563

RESUMEN

Duchenne Muscular Dystrophy (DMD) is a lethal muscle disorder, caused by mutations in the DMD gene and affects approximately 1:5000-6000 male births. In this report, we identified dysregulation of members of the Dlk1-Dio3 miRNA cluster in muscle biopsies of the GRMD dog model. Of these, we selected miR-379 for a detailed investigation because its expression is high in the muscle, and is known to be responsive to glucocorticoid, a class of anti-inflammatory drugs commonly used in DMD patients. Bioinformatics analysis predicts that miR-379 targets EIF4G2, a translational factor, which is involved in the control of mitochondrial metabolic maturation. We confirmed in myoblasts that EIF4G2 is a direct target of miR-379, and identified the DAPIT mitochondrial protein as a translational target of EIF4G2. Knocking down DAPIT in skeletal myotubes resulted in reduced ATP synthesis and myogenic differentiation. We also demonstrated that this pathway is GC-responsive since treating mice with dexamethasone resulted in reduced muscle expression of miR-379 and increased expression of EIF4G2 and DAPIT. Furthermore, miR-379 seric level, which is also elevated in the plasma of DMD patients in comparison with age-matched controls, is reduced by GC treatment. Thus, this newly identified pathway may link GC treatment to a mitochondrial response in DMD.


Asunto(s)
Glucocorticoides/uso terapéutico , MicroARNs/metabolismo , Mitocondrias/metabolismo , Distrofia Muscular de Duchenne/tratamiento farmacológico , Adenosina Trifosfato/metabolismo , Animales , Sitios de Unión , Dexametasona/farmacología , Modelos Animales de Enfermedad , Perros , Factor 4G Eucariótico de Iniciación/química , Factor 4G Eucariótico de Iniciación/genética , Factor 4G Eucariótico de Iniciación/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , MicroARNs/química , ATPasas de Translocación de Protón Mitocondriales/antagonistas & inhibidores , ATPasas de Translocación de Protón Mitocondriales/genética , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Mioblastos Esqueléticos/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo
2.
Cell Death Dis ; 5: e1487, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25341041

RESUMEN

During embryogenesis, the intrauterine milieu affects cell proliferation, differentiation, and function by modifying gene expression in susceptible cells, such as the pancreatic ß-cells. In this limited energy environment, mitochondrial dysfunction can lead to overproduction of reactive oxygen species (ROS) and to a decline in ß-cell function. In opposition to this toxicity, ROS are also required for insulin secretion. Here we investigated the role of ROS in ß-cell development. Surprisingly, decreasing ROS production in vivo reduced ß-cell differentiation. Moreover, in cultures of pancreatic explants, progenitors were highly sensitive to ROS stimulation and responded by generating ß-cells. ROS enhanced ß-cell differentiation through modulation of ERK1/2 signaling. Gene transfer and pharmacological manipulations, which diminish cellular ROS levels, also interfered with normal ß-cell differentiation. This study highlights the role of the redox balance on ß-cell development and provides information that will be useful for improving ß-cell production from embryonic stem cells, a step in cell therapy for diabetes.


Asunto(s)
Antioxidantes/farmacología , Diferenciación Celular/efectos de los fármacos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/enzimología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Oxidantes/toxicidad , Acetilcisteína/farmacología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carbonil Cianuro m-Clorofenil Hidrazona/farmacología , Catalasa/metabolismo , Embrión de Mamíferos/citología , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Femenino , Peróxido de Hidrógeno/toxicidad , Células Secretoras de Insulina/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Fosforilación/efectos de los fármacos , Ratas Wistar
3.
Cell Death Dis ; 3: e282, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22419111

RESUMEN

The Trans-activator protein (Tat) of human immunodeficiency virus (HIV) is a pleiotropic protein involved in different aspects of AIDS pathogenesis. As a number of viral proteins Tat is suspected to disturb mitochondrial function. We prepared pure synthetic full-length Tat by native chemical ligation (NCL), and Tat peptides, to evaluate their direct effects on isolated mitochondria. Submicromolar doses of synthetic Tat cause a rapid dissipation of the mitochondrial transmembrane potential (ΔΨ(m)) as well as cytochrome c release in mitochondria isolated from mouse liver, heart, and brain. Accordingly, Tat decreases substrate oxidation by mitochondria isolated from these tissues, with oxygen uptake being initially restored by adding cytochrome c. The anion-channel inhibitor 4,4'-diisothiocyanostilbene-2,2'-disulfonic acid (DIDS) protects isolated mitochondria against Tat-induced mitochondrial membrane permeabilization (MMP), whereas ruthenium red, a ryanodine receptor blocker, does not. Pharmacologic inhibitors of the permeability transition pore, Bax/Bak inhibitors, and recombinant Bcl-2 and Bcl-XL proteins do not reduce Tat-induced MMP. We finally observed that Tat inhibits cytochrome c oxidase (COX) activity in disrupted mitochondria isolated from liver, heart, and brain of both mouse and human samples, making it the first described viral protein to be a potential COX inhibitor.


Asunto(s)
Complejo IV de Transporte de Electrones/antagonistas & inhibidores , Mitocondrias/efectos de los fármacos , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/farmacología , Ácido 4,4'-Diisotiocianostilbeno-2,2'-Disulfónico/farmacología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Citocromos c/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Humanos , Transporte Iónico , Hígado/efectos de los fármacos , Hígado/enzimología , Potencial de la Membrana Mitocondrial , Ratones , Ratones Endogámicos BALB C , Mitocondrias/enzimología , Membranas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/metabolismo , Miocardio/enzimología , Fosforilación Oxidativa , Permeabilidad , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/fisiología
4.
Cell Death Dis ; 2: e203, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21881605

RESUMEN

Brain protection of the newborn remains a challenging priority and represents a totally unmet medical need. Pharmacological inhibition of caspases appears as a promising strategy for neuroprotection. In a translational perspective, we have developed a pentapeptide-based group II caspase inhibitor, TRP601/ORPHA133563, which reaches the brain, and inhibits caspases activation, mitochondrial release of cytochrome c, and apoptosis in vivo. Single administration of TRP601 protects newborn rodent brain against excitotoxicity, hypoxia-ischemia, and perinatal arterial stroke with a 6-h therapeutic time window, and has no adverse effects on physiological parameters. Safety pharmacology investigations, and toxicology studies in rodent and canine neonates, suggest that TRP601 is a lead compound for further drug development to treat ischemic brain damage in human newborns.


Asunto(s)
Inhibidores de Caspasas , Inhibidores de Cisteína Proteinasa/uso terapéutico , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Isquemia/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Oligopéptidos/uso terapéutico , Quinolinas/farmacología , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Sitios de Unión , Caspasas/metabolismo , Inhibidores de Cisteína Proteinasa/química , Citocromos c/metabolismo , Modelos Animales de Enfermedad , Hipoxia-Isquemia Encefálica/patología , Isquemia/patología , Ratones , Fármacos Neuroprotectores/química , Oligopéptidos/química , Oligopéptidos/farmacología , Quinolinas/química , Ratas
5.
Oncogene ; 30(37): 3918-29, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21460857

RESUMEN

The BH3 mimetic ABT737 induces autophagy by competitively disrupting the inhibitory interaction between the BH3 domain of Beclin 1 and the anti-apoptotic proteins Bcl-2 and Bcl-X(L), thereby stimulating the Beclin 1-dependent allosteric activation of the pro-autophagic lipid kinase VPS34. Here, we examined whether ABT737 stimulates other pro-autophagic signal-transduction pathways. ABT737 caused the activating phosphorylation of AMP-dependent kinase (AMPK) and of the AMPK substrate acetyl CoA carboxylase, the activating phosphorylation of several subunits of the inhibitor of NF-κB (IκB) kinase (IKK) and the hyperphosphorylation of the IKK substrate IκB, inhibition of the activity of mammalian target of rapamycin (mTOR) and consequent dephosphorylation of the mTOR substrate S6 kinase. In addition, ABT737 treatment dephosphorylates (and hence likewise inhibits) p53, glycogen synthase kinase-3 and Akt. All these effects were shared by ABT737 and another structurally unrelated BH3 mimetic, HA14-1. Functional experiments revealed that pharmacological or genetic inhibition of IKK, Sirtuin and the p53-depleting ubiquitin ligase MDM2 prevented ABT737-induced autophagy. These results point to unexpected and pleiotropic pro-autophagic effects of BH3 mimetics involving the modulation of multiple signalling pathways.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/agonistas , Autofagia/efectos de los fármacos , Compuestos de Bifenilo/farmacología , Proteínas de la Membrana/agonistas , Nitrofenoles/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Sulfonamidas/farmacología , Acetil-CoA Carboxilasa/metabolismo , Beclina-1 , Benzopiranos/farmacología , Línea Celular Tumoral , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Quinasa I-kappa B/metabolismo , Nitrilos/farmacología , Proteína Oncogénica v-akt/metabolismo , Fosforilación , Fosfotransferasas (Aceptor del Grupo Fosfato)/metabolismo , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteínas Quinasas S6 Ribosómicas/metabolismo , Transducción de Señal/efectos de los fármacos , Sirtuinas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
6.
Br J Dermatol ; 163(6): 1337-9, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20560959

RESUMEN

Multiple cutaneous and uterine leiomyomatosis (MCUL)/hereditary leiomyomatosis and renal cell cancer (HLRCC) (OMIM 150800/OMIM 605839) is a rare hereditary disorder leading to the development of benign cutaneous and uterine smooth muscle tumours in young adults.(1,2) This disease is characterized by an increased risk of developing renal cell carcinomas.(3) It results from dominantly inherited autosomal mutations in the fumarate hydratase (FH) gene.(4) This gene encodes a Krebs cycle enzyme, present in both cytosolic and mitochondrial compartments, and probably acts as a tumour suppressor gene. We report a 22-year-old man affected by cutaneous leiomyomatosis associated with cutis verticis gyrata, disseminated collagenoma and Charcot-Marie-Tooth disease, who was harbouring the novel FH gene mutation c.821C > T, p.Ala274Val.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/genética , Fumarato Hidratasa/genética , Leiomiomatosis/genética , Mutación , Enfermedades de la Piel/genética , Neoplasias Cutáneas/genética , Carcinoma de Células Renales/genética , Enfermedades del Colágeno/genética , Predisposición Genética a la Enfermedad , Humanos , Leiomiomatosis/enzimología , Leiomiomatosis/patología , Masculino , Dermatosis del Cuero Cabelludo/genética , Análisis de Secuencia de ADN , Neoplasias Cutáneas/enzimología , Neoplasias Cutáneas/patología , Adulto Joven
7.
Cell Death Differ ; 17(7): 1155-66, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20111043

RESUMEN

Apoptosis-inducing factor (AIF) has important supportive as well as potentially lethal roles in neurons. Under normal physiological conditions, AIF is a vital redox-active mitochondrial enzyme, whereas in pathological situations, it translocates from mitochondria to the nuclei of injured neurons and mediates apoptotic chromatin condensation and cell death. In this study, we reveal the existence of a brain-specific isoform of AIF, AIF2, whose expression increases as neuronal precursor cells differentiate. AIF2 arises from the utilization of the alternative exon 2b, yet uses the same remaining 15 exons as the ubiquitous AIF1 isoform. AIF1 and AIF2 are similarly imported to mitochondria in which they anchor to the inner membrane facing the intermembrane space. However, the mitochondrial inner membrane sorting signal encoded in the exon 2b of AIF2 is more hydrophobic than that of AIF1, indicating a stronger membrane anchorage of AIF2 than AIF1. AIF2 is more difficult to be desorbed from mitochondria than AIF1 on exposure to non-ionic detergents or basic pH. Furthermore, AIF2 dimerizes with AIF1, thereby preventing its release from mitochondria. Conversely, it is conceivable that a neuron-specific AIF isoform, AIF2, may have been 'designed' to be retained in mitochondria and to minimize its potential neurotoxic activity.


Asunto(s)
Factor Inductor de la Apoptosis/metabolismo , Encéfalo/metabolismo , Mitocondrias/metabolismo , Secuencia de Aminoácidos , Animales , Factor Inductor de la Apoptosis/química , Factor Inductor de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/química , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Humanos , Ratones , Proteínas Mitocondriales/química , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Datos de Secuencia Molecular , Neuronas/citología , Neuronas/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Alineación de Secuencia
8.
J Inherit Metab Dis ; 32(6): 684-698, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19821144

RESUMEN

Diabetes mellitus is occasionally observed in patients with skeletal muscle respiratory chain deficiency, suggesting that skeletal muscle mitochondrial dysfunction might play a pathogenic role in type 2 diabetes (T2D). In support of this hypothesis, decreased muscle mitochondrial activity has been reported in T2D patients and in mouse models of diabetes. However, recent work by several groups suggests that decreased muscle mitochondrial function may be a consequence rather than a cause of diabetes, since decreased mitochondrial function in mice affords protection from diabetes and obesity. We review the data on this controversial but important issue of potential links between mitochondrial dysfunction and diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/etiología , Mitocondrias Musculares/fisiología , Animales , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Humanos , Células Secretoras de Insulina/fisiología , Ratones , Mitocondrias Musculares/metabolismo , Enfermedades Mitocondriales/complicaciones , Modelos Biológicos , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , Transducción de Señal/fisiología
9.
Cell Death Differ ; 15(6): 1009-18, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18309327

RESUMEN

Apoptosis-inducing factor (AIF) is a phylogenetically conserved redox-active flavoprotein that contributes to cell death and oxidative phosphorylation in Saccharomyces cerevisiae, Caenorhabditis elegans, mouse and humans. AIF has been characterized as a caspase-independent death effector that is activated by its translocation from mitochondria to the cytosol and nucleus. Here, we report the molecular characterization of AIF in Drosophila melanogaster, a species in which most cell deaths occur in a caspase-dependent manner. Interestingly, knockout of zygotic D. melanogaster AIF (DmAIF) expression using gene targeting resulted in decreased embryonic cell death and the persistence of differentiated neuronal cells at late embryonic stages. Although knockout embryos hatch, they undergo growth arrest at early larval stages, accompanied by mitochondrial respiratory dysfunction. Transgenic expression of DmAIF misdirected to the extramitochondrial compartment (DeltaN-DmAIF), but not wild-type DmAIF, triggered ectopic caspase activation and cell death. DeltaN-DmAIF-induced death was not blocked by removal of caspase activator Dark or transgenic expression of baculoviral caspase inhibitor p35, but was partially inhibited by Diap1 overexpression. Knockdown studies revealed that DeltaN-DmAIF interacts genetically with the redox protein thioredoxin-2. In conclusion, we show that Drosophila AIF is a mitochondrial effector of cell death that plays roles in developmentally regulated cell death and normal mitochondrial function.


Asunto(s)
Factor Inductor de la Apoptosis/fisiología , Apoptosis , Proteínas de Drosophila/fisiología , Drosophila melanogaster/embriología , Secuencia de Aminoácidos , Animales , Factor Inductor de la Apoptosis/química , Factor Inductor de la Apoptosis/genética , Sistema Nervioso Central/embriología , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/anatomía & histología , Drosophila melanogaster/metabolismo , Metabolismo Energético , Ojo/anatomía & histología , Proteínas Mitocondriales/química , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/fisiología , Datos de Secuencia Molecular , Mutación , Homología de Secuencia de Aminoácido , Tiorredoxinas/metabolismo
10.
Artículo en Inglés | MEDLINE | ID: mdl-18814355

RESUMEN

Mitochondria contained in cancer cells exhibit two major alterations. First, they are often relatively resistant to the induction of mitochondrial membrane permeabilization (MMP), which is the rate-limiting step of the intrinsic pathway of apoptosis. The mechanisms of MMP resistance have come under close scrutiny because apoptosis resistance constitutes one of the essential hallmarks of cancer. Second, cancer cell mitochondria often exhibit a reduced oxidative phosphorylation, meaning that ATP is generated through the conversion of glucose to pyruvate and excess pyruvate is then eliminated as the waste product lactate. This glycolytic mode of energy production is even observed in conditions of high oxygen tension and is hence called anaerobic glycolysis. Here, we discuss the molecular mechanisms accounting for inhibition of the mitochondrial apoptosis pathway in neoplasia and discuss possible mechanistic links between MMP resistance and anaerobic glycolysis.


Asunto(s)
Mitocondrias/metabolismo , Neoplasias/metabolismo , Anaerobiosis , Animales , Apoptosis , Glucólisis , Humanos , Membranas Mitocondriales/metabolismo , Modelos Biológicos , Neoplasias/patología , Fosforilación Oxidativa , Permeabilidad
11.
Cell Death Differ ; 14(3): 422-35, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16888644

RESUMEN

The HIV-1 encoded apoptogenic protein Vpr induces mitochondrial membrane permeabilization (MMP) via interactions with the voltage-dependent anion channel (VDAC) and the adenine nucleotide translocator (ANT). We have designed a peptide, TEAM-VP, composed of two functional domains, one a tumor blood vessel RGD-like 'homing' motif and the other an MMP-inducing sequence derived from Vpr. When added to isolated mitochondria, TEAM-VP interacts with ANT and VDAC, reduces oxygen consumption and overcomes Bcl-2 protection to cause inner and outer MMP. TEAM-VP specifically recognizes cell-surface expressed alpha(V)beta(3) integrins, internalizes, temporarily localizes to lysosomes and progressively co-distributes with the mitochondrial compartment with no sign of lysosomal membrane permeabilization. Finally TEAM-VP reaches mitochondria of angiogenic endothelial cells to induce mitochondrial fission, dissipation of the mitochondrial transmembrane potential (DeltaPsi(m)), cytochrome c release and apoptosis hallmarks. Hence, this chimeric peptide constitutes the first example of a virus-derived mitochondriotoxic compound as a candidate to kill selectively tumor neo-endothelia.


Asunto(s)
Células Endoteliales/fisiología , Productos del Gen vpr/farmacocinética , Integrina alfaVbeta3/metabolismo , Mitocondrias/metabolismo , Péptidos/farmacocinética , Secuencia de Aminoácidos , Animales , Apoptosis , Supervivencia Celular , Relación Dosis-Respuesta a Droga , Células Endoteliales/metabolismo , Productos del Gen vpr/farmacología , Humanos , Lisosomas/metabolismo , Ratones , Ratones Endogámicos BALB C , Membranas Mitocondriales/metabolismo , Datos de Secuencia Molecular , Péptidos/farmacología , Permeabilidad
12.
Ir Med J ; 99(9): 262-4, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17144232

RESUMEN

Mitochondrial respiratory chain disorders account for significant and varied presentations in paediatric practice. The true prevalence of these disorders in the paediatric population is still not well documented with predicted geographic variation. We report a retrospective analysis over a seven year period of cases presenting to a tertiary care centre and associated clinical features. The overall prevalence of mitochondrial disorders in our population is higher than expected (1/9,000 births), explained in part by multiple presentations in a consanguineous subgroup of the population (Irish travellers).


Asunto(s)
Enfermedades Mitocondriales/epidemiología , Humanos , Incidencia , Irlanda/epidemiología , Enfermedades Mitocondriales/diagnóstico , Enfermedades Mitocondriales/genética , Fenotipo
13.
Arch Dis Child ; 91(2): 178-82, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16428367

RESUMEN

Cockayne syndrome is a multi-systemic, autosomal recessive disease characterised by postnatal growth failure and progressive multi-organ dysfunction. The main clinical features are severe dwarfism (<-2 SD), microcephaly (<-3 SD), psychomotor delay, sensorial loss (cataracts, pigmentary retinopathy, and deafness), and cutaneous photosensitivity. Here, 13 new cases of Cockayne syndrome are reported, which have been clinically diagnosed and confirmed using a biochemical transcription assay. The wide clinical variability, ranging from prenatal features to normal psychomotor development, is emphasised. When cardinal features are lacking, the diagnosis of Cockayne syndrome should be considered when presented with growth retardation, microcephaly, and one of the suggesting features such as enophthalmia, limb ataxia, abnormal auditory evoked responses, or increased ventricular size on cerebral imaging.


Asunto(s)
Síndrome de Cockayne/diagnóstico , Anomalías Múltiples/diagnóstico , Células Cultivadas , Niño , Preescolar , Síndrome de Cockayne/psicología , Discapacidades del Desarrollo/diagnóstico , Enoftalmia/diagnóstico , Femenino , Retardo del Crecimiento Fetal/diagnóstico , Trastornos del Crecimiento/diagnóstico , Humanos , Lactante , Masculino , Fenotipo , Desempeño Psicomotor
14.
Mol Genet Metab ; 86(4): 462-5, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16263314

RESUMEN

The activity of deoxyguanosine kinase (DGUOK), a mitochondrial enzyme involved in the anabolism of mitochondrial (mt) deoxyribonucleotides, governs the maintenance of the mtDNA. Deleterious mutations of the DGUOK gene are thus associated with mtDNA depletion and result in combined deficiencies of mtDNA-encoded respiratory chain enzymes. With the aim to estimate the prevalence of DGUOK mutations in a cohort of 30 patients with hepatocerebral disease and combined respiratory chain deficiencies, we studied the DGUOK gene and identified previously unreported mutations in five families. Two patients and their affected sibs, born to non-consanguineous parents, were homozygous for a missense mutation (M1T, and L250S, respectively). One patient presented a homozygous 4 pb insertion (796 insTGAT) and two other patients, and their affected sibs, were compound heterozygous (E165V/L266R and E211G/L266R, respectively). These findings allowed us to propose prenatal diagnosis in two families. In conclusion, we observed a high prevalence of DGUOK mutations (17%) in patients with hepatic involvement and combined respiratory chain deficiencies with hepatic involvement.


Asunto(s)
Enfermedades Mitocondriales/enzimología , Enfermedades Mitocondriales/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Secuencia de Aminoácidos , Secuencia de Bases , ADN/genética , Análisis Mutacional de ADN , ADN Mitocondrial/metabolismo , Femenino , Humanos , Lactante , Hígado/fisiopatología , Masculino , Enfermedades Mitocondriales/fisiopatología , Datos de Secuencia Molecular , Mutación , Homología de Secuencia de Aminoácido
15.
Pediatrics ; 116(5): 1170-7, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16264005

RESUMEN

OBJECTIVES: We sought to determine the long-term clinical and biochemical outcome of newborns with mitochondrial cytopathies (MCs) and to identify possible prognostic factors that may modify the course of these diseases. MATERIAL AND METHODS: Fifty-seven newborns with MCs were identified in a retrospective review (1983-2002). We defined 2 different outcome categories: clinical (neurologic, hepatic, myopathic, and multiorganic) and biochemical (lactate level normalization or initially normal remaining unchanged, decreased but not normalized, and persistently high). We used 2 different statistical approaches: (1) survival studies depending on the initial symptoms and lactate and enzymatic deficiencies using the Kaplan-Meier method; and (2) the same variables compared with different survival age groups and clinical and biochemical outcome categories using the chi2 test. RESULTS: Thirty-three patients died (57.8%), 12 remain alive (21%), and 12 were lost in the follow-up; 6 of them are currently older than 4 years. Most of the patients manifested multiorganic disease (64.8%) and high lactate level (77.1%) over time. Children surviving to 2.5 to 3 years of age were more likely to survive for a long period of time. Initial neurologic and hepatic presentation increased the risk to develop neurologic disease and severe persistent hyperlactacidemia, respectively. Initial severe hyperlactacidemia and combined enzyme deficiencies were significant risk factors for higher mortality and multiorganic disorders. Two patients with exclusively myopathic outcome are alive and cognitively normal at 12 years of life. CONCLUSIONS: Children with neonatal-onset MCs have very high mortality and poor prospects. However, some with life-threatening presentations may gradually improve, giving rise to less severe diseases. Those with exclusively myopathic symptoms have a better prognosis.


Asunto(s)
Enfermedades del Recién Nacido , Miopatías Mitocondriales/complicaciones , Preescolar , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Lactante , Recién Nacido , Enfermedades del Recién Nacido/sangre , Enfermedades del Recién Nacido/diagnóstico , Enfermedades del Recién Nacido/mortalidad , Ácido Láctico/sangre , Masculino , Miopatías Mitocondriales/sangre , Miopatías Mitocondriales/diagnóstico , Miopatías Mitocondriales/mortalidad , Pronóstico
16.
Cell Mol Life Sci ; 62(19-20): 2317-24, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16143825

RESUMEN

Mitochondrial succinate dehydrogenase (SDH) consists merely of four nuclearly encoded subunits. It participates in the electron transfer in the respiratory chain and in succinate catabolism in the Krebs cycle. Mutations in the four genes, SDHA, B, C and D, have been reported, resulting in strikingly diverse clinical presentations. So far, SDHA mutations have been reported to cause an encephalomyopathy in childhood, while mutations in the genes encoding the other three subunits have been associated only with tumour formation. Following a brief description of SDH genes and subunits, we examine the properties and roles of SDH in the mitochondria. This allows further discussion of the several hypotheses proposed to account for the different clinical presentations resulting from impaired activity of the enzyme. Finally we stress the importance of SDH as a target and/or marker in a number of diseases and the need to better delineate the consequences of SDH deficiency in humans.


Asunto(s)
Succinato Deshidrogenasa/deficiencia , Ácido Succínico/metabolismo , Humanos , Errores Innatos del Metabolismo/genética , Errores Innatos del Metabolismo/metabolismo , Mutación , Succinato Deshidrogenasa/genética
17.
Hum Mol Genet ; 14(15): 2231-9, 2005 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-15987702

RESUMEN

The nuclear-encoded Krebs cycle enzymes, fumarate hydratase (FH) and succinate dehydrogenase (SDHB, -C and -D), act as tumour suppressors. Germline mutations in FH predispose individuals to leiomyomas and renal cell cancer (HLRCC), whereas mutations in SDH cause paragangliomas and phaeochromocytomas (HPGL). In this study, we have shown that FH-deficient cells and tumours accumulate fumarate and, to a lesser extent, succinate. SDH-deficient tumours principally accumulate succinate. In situ analyses showed that these tumours also have over-expression of hypoxia-inducible factor 1alpha (HIF1alpha), activation of HIF1alphatargets (such as vascular endothelial growth factor) and high microvessel density. We found no evidence of increased reactive oxygen species in our cells. Our data provide in vivo evidence to support the hypothesis that increased succinate and/or fumarate causes stabilization of HIF1alpha a plausible mechanism, inhibition of HIF prolyl hydroxylases, has previously been suggested by in vitro studies. The basic mechanism of tumorigenesis in HPGL and HLRCC is likely to be pseudo-hypoxic drive, just as it is in von Hippel-Lindau syndrome.


Asunto(s)
Fumarato Hidratasa/genética , Mutación de Línea Germinal , Succinato Deshidrogenasa/genética , Succinato Deshidrogenasa/metabolismo , Carcinoma de Células Renales/metabolismo , Ciclo del Ácido Cítrico/fisiología , Femenino , Fumarato Hidratasa/metabolismo , Humanos , Leiomioma/genética , Leiomioma/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Paraganglioma/genética , Paraganglioma/metabolismo , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular/metabolismo
18.
Br J Cancer ; 91(10): 1835-41, 2004 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-15505628

RESUMEN

Germline mutations in the succinate dehydrogenase (SDH) (mitochondrial respiratory chain complex II) subunit B gene, SDHB, cause susceptibility to head and neck paraganglioma and phaeochromocytoma. Previously, we did not identify somatic SDHB mutations in sporadic phaeochromocytoma, but SDHB maps to 1p36, a region of frequent loss of heterozygosity (LOH) in neuroblastoma as well. Hence, to evaluate SDHB as a candidate neuroblastoma tumour suppressor gene (TSG) we performed mutation analysis in 46 primary neuroblastomas by direct sequencing, but did not identify germline or somatic SDHB mutations. As TSGs such as RASSF1A are frequently inactivated by promoter region hypermethylation, we designed a methylation-sensitive PCR-based assay to detect SDHB promoter region methylation. In 21% of primary neuroblastomas and 32% of phaeochromocytomas (32%) methylated (and unmethylated) alleles were detected. Although promoter region methylation was also detected in two neuroblastoma cell lines, this was not associated with silencing of SDHB expression, and treatment with a demethylating agent (5-azacytidine) did not increase SDH activity. These findings suggest that although germline SDHB mutations are an important cause of phaeochromocytoma susceptibility, somatic inactivation of SDHB does not have a major role in sporadic neural crest tumours and SDHB is not the target of 1p36 allele loss in neuroblastoma and phaeochromocytoma.


Asunto(s)
Metilación de ADN , Mutación , Neuroblastoma/genética , Feocromocitoma/genética , Subunidades de Proteína/genética , Succinato Deshidrogenasa/genética , Secuencia de Bases , Línea Celular Tumoral , Silenciador del Gen , Humanos , Proteínas Hierro-Azufre , Pérdida de Heterocigocidad , Datos de Secuencia Molecular , Cresta Neural , Regiones Promotoras Genéticas
19.
Eur J Paediatr Neurol ; 8(1): 55-9, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15023375

RESUMEN

An eight-week-old infant, the fourth child of consanguineous parents presented with intractable neonatal seizures. The mother had two previous miscarriages. The infant initially presented on day one with multifocal myoclonus, complex partial and generalised tonic-clonic seizures. On examination, there were dysmorphic hands and feet, with absent nails and terminal phalanges of the fingers and toes, hepatomegaly, marked axial and peripheral hypotonia and severe global developmental delay. Ophthalmological assessment showed 'salt and pepper' pigmentary retinopathy. The urinary organic acid profile revealed a marked increase in tricarboxylic acid metabolites. Urinary phosphate reabsorption was reduced at 84%. Type I fibre atrophy was seen on muscle histology, and a cytochrome c oxidase deficiency was found only on enzymology of liver tissue. Limb malformations associated with respiratory chain defects have rarely been reported. To our knowledge, this child has the most severe limb anomaly associated with a tissue-specific complex IV respiratory chain defect.


Asunto(s)
Anomalías Múltiples/genética , Anomalías Craneofaciales/genética , Deficiencia de Citocromo-c Oxidasa/genética , Complejo IV de Transporte de Electrones/genética , Epilepsia Benigna Neonatal/genética , Dedos/anomalías , Hígado/enzimología , Dedos del Pie/anomalías , Anomalías Múltiples/diagnóstico , Consanguinidad , Anomalías Craneofaciales/diagnóstico , Deficiencia de Citocromo-c Oxidasa/diagnóstico , Análisis Mutacional de ADN , Discapacidades del Desarrollo/diagnóstico , Discapacidades del Desarrollo/genética , Diagnóstico Diferencial , Epilepsias Mioclónicas/diagnóstico , Epilepsias Mioclónicas/genética , Epilepsia Benigna Neonatal/diagnóstico , Epilepsia Parcial Compleja/diagnóstico , Epilepsia Parcial Compleja/genética , Epilepsia Tónico-Clónica/diagnóstico , Epilepsia Tónico-Clónica/genética , Humanos , Lactante , Masculino , Proteínas de la Membrana/genética , Chaperonas Moleculares , Hipotonía Muscular/diagnóstico , Hipotonía Muscular/genética , Fenotipo , Estado Epiléptico/diagnóstico , Estado Epiléptico/genética , Ácidos Tricarboxílicos/orina
20.
J Med Genet ; 41(1): 14-7, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14729820

RESUMEN

Respiratory chain complex I deficiency represents a genetically heterogeneous group of diseases resulting from mutations in mitochondrial or nuclear genes. Mutations have been reported in 13 of the 14 subunits encoding the core of complex I (seven mitochondrial and six nuclear genes) and these result in Leigh or Leigh-like syndromes or cardiomyopathy. In this study, a combination of denaturing high performance liquid chromatography and sequence analysis was used to study the NDUFS3 gene in a series of complex I deficient patients. Mutations found in this gene (NADH dehydrogenase iron-sulphur protein 3), coding for the seventh and last subunit of complex I core, were shown to cause late onset Leigh syndrome, optic atrophy, and complex I deficiency. A biochemical diagnosis of complex I deficiency on cultured amniocytes from a later pregnancy was confirmed through the identification of disease causing NDUFS3 mutations in these cells. While mutations in the NDUFS3 gene thus result in Leigh syndrome, a dissimilar clinical phenotype is observed in mutations in the NDUFV2 and NDUFS2 genes, resulting in encephalomyopathy and cardiomyopathy. The reasons for these differences are uncertain.


Asunto(s)
Complejo I de Transporte de Electrón/genética , Enfermedad de Leigh/etiología , Enfermedad de Leigh/genética , Mutación/genética , NADH Deshidrogenasa/genética , Subunidades de Proteína/genética , Niño , Complejo I de Transporte de Electrón/deficiencia , Resultado Fatal , Humanos , Proteínas Hierro-Azufre/deficiencia , Proteínas Hierro-Azufre/genética , Enfermedad de Leigh/enzimología , Enfermedad de Leigh/patología , Masculino , NADH Deshidrogenasa/deficiencia , Subunidades de Proteína/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA