Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Cancer Res ; 81(13): 3480-3494, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34127497

RESUMEN

Succinate dehydrogenase is a key enzyme in the tricarboxylic acid cycle and the electron transport chain. All four subunits of succinate dehydrogenase are tumor suppressor genes predisposing to paraganglioma, but only mutations in the SDHB subunit are associated with increased risk of metastasis. Here we generated an Sdhd knockout chromaffin cell line and compared it with Sdhb-deficient cells. Both cell types exhibited similar SDH loss of function, metabolic adaptation, and succinate accumulation. In contrast, Sdhb-/- cells showed hallmarks of mesenchymal transition associated with increased DNA hypermethylation and a stronger pseudo-hypoxic phenotype compared with Sdhd-/- cells. Loss of SDHB specifically led to increased oxidative stress associated with dysregulated iron and copper homeostasis in the absence of NRF2 activation. High-dose ascorbate exacerbated the increase in mitochondrial reactive oxygen species, leading to cell death in Sdhb-/- cells. These data establish a mechanism linking oxidative stress to iron homeostasis that specifically occurs in Sdhb-deficient cells and may promote metastasis. They also highlight high-dose ascorbate as a promising therapeutic strategy for SDHB-related cancers. SIGNIFICANCE: Loss of different succinate dehydrogenase subunits can lead to different cell and tumor phenotypes, linking stronger 2-OG-dependent dioxygenases inhibition, iron overload, and ROS accumulation following SDHB mutation.


Asunto(s)
Ácido Ascórbico/farmacología , Homeostasis , Hierro/metabolismo , Mutación , Estrés Oxidativo , Succinato Deshidrogenasa/fisiología , Animales , Antioxidantes/farmacología , Dioxigenasas/antagonistas & inhibidores , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Mitocondrias/patología , Fenotipo , Especies Reactivas de Oxígeno
2.
FASEB J ; 34(1): 303-315, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31914648

RESUMEN

Mutations in succinate dehydrogenase (SDH) lead to the development of tumors in a restricted subset of cell types, including chromaffin cells and paraganglia. The molecular basis for this specificity is currently unknown. We show that loss of SDH activity in a chromaffin cell model does not perturb complex I function, retaining the ability to oxidize NADH within the electron transport chain. This activity supports continued oxidation of substrates within the tricarboxylic acid (TCA) cycle. However, due to the block in the TCA cycle at SDH, the high glutamine oxidation activity is only maintained through an efflux of succinate. We also show that although the mitochondria of SDH-deficient cells are less active per se, their higher mass per cell results in an overall respiratory rate that is comparable with wild-type cells. Finally, we observed that when their mitochondria are uncoupled, SDH-deficient cells are unable to preserve their viability, suggesting that the mitochondrial metabolic network is unable to compensate when exposed to additional stress. We therefore show that in contrast to models of SDH deficiency based on epithelial cells, a chromaffin cell model retains aspects of metabolic "health," which could form the basis of cell specificity of this rare tumor type.


Asunto(s)
Células Cromafines/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Mitocondrias/metabolismo , Enfermedades Mitocondriales/metabolismo , Neoplasias/metabolismo , Succinato Deshidrogenasa/fisiología , Animales , Células Cromafines/patología , Humanos , Masculino , Ratones , Ratones Noqueados , Mitocondrias/patología , Enfermedades Mitocondriales/patología , Mutación , NAD/metabolismo , Neoplasias/patología , Succinato Deshidrogenasa/genética , Succinato Deshidrogenasa/metabolismo , Transcriptoma
3.
Invest Ophthalmol Vis Sci ; 60(13): 4187-4195, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31596927

RESUMEN

Purpose: Metastatic uveal melanoma (UM) has a very poor prognosis and no effective therapy. Despite remarkable advances in treatment of cutaneous melanoma, UM remains recalcitrant to chemotherapy, small-molecule kinase inhibitors, and immune-based therapy. Methods: We assessed two sets of oxidative phosphorylation (OxPhos) genes within 9858 tumors across 31 cancer types. An OxPhos inhibitor was used to characterize differential metabolic programming of highly metastatic monosomy 3 (M3) UM. Seahorse analysis and global metabolomics profiling were done to identify metabolic vulnerabilities. Analyses of UM TCGA data set were performed to determine expressions of key OxPhos effectors in M3 and non-M3 UM. We used targeted knockdown of succinate dehydrogenase A (SDHA) to determine the role of SDHA in M3 UM in conferring resistance to OxPhos inhibition. Results: We identified UM to have among the highest median OxPhos levels and showed that M3 UM exhibits a distinct metabolic profile. M3 UM shows markedly low succinate levels and has highly increased levels of SDHA, the enzyme that couples the tricarboxylic acid cycle with OxPhos by oxidizing (lowering) succinate. We showed that SDHA-high M3 UM have elevated expression of key OxPhos molecules, exhibit abundant mitochondrial reserve respiratory capacity, and are resistant to OxPhos antagonism, which can be reversed by SDHA knockdown. Conclusions: Our study has identified a critical metabolic program within poor prognostic M3 UM. In addition to the heightened mitochondrial functional capacity due to elevated SDHA, M3 UM SDHA-high mediate resistance to therapy that is reversible with targeted treatment.


Asunto(s)
Melanoma/metabolismo , Succinato Deshidrogenasa/fisiología , Neoplasias de la Úvea/metabolismo , Humanos , Fosforilación Oxidativa , Succinato Deshidrogenasa/metabolismo , Ácido Succínico/metabolismo , Células Tumorales Cultivadas
4.
Genet Med ; 21(3): 705-717, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30050099

RESUMEN

PURPOSE: Metabolic aberrations have been described in neoplasms with pathogenic variants (PV) in the Krebs cycle genes encoding succinate dehydrogenase (SDH), fumarate hydratase (FH) and isocitrate dehydrogenase (IDH). In turn, accumulation of oncometabolites succinate, fumarate, and 2-hydroxyglutarate can be employed to identify tumors with those PV . Additionally, such metabolic readouts may aid in genetic variant interpretation and improve diagnostics. METHODS: Using liquid chromatography-mass spectrometry, 395 pheochromocytomas and paragangliomas (PPGLs) from 391 patients were screened for metabolites to indicate Krebs cycle aberrations. Multigene panel sequencing was applied to detect driver PV in cases with indicative metabolite profiles but undetermined genetic drivers. RESULTS: Aberrant Krebs cycle metabolomes identified rare cases of PPGLs with germline PV in FH and somatic PV in IDHx and SDHx, including the first case of a somatic IDH2 PV in PPGL. Metabolomics also reliably identified PPGLs with SDHx loss-of-function (LOF) PV. Therefore we utilized tumor metabolite profiles to further classify variants of unknown significance in SDHx, thereby enabling missense variants associated with SDHx LOF to be distinguished from benign variants. CONCLUSION: We propose incorporation of metabolome data into the diagnostics algorithm in PPGLs to guide genetic testing and variant interpretation and to help identify rare cases with PV in FH and IDHx.


Asunto(s)
Genómica/métodos , Paraganglioma/genética , Feocromocitoma/genética , Neoplasias de las Glándulas Suprarrenales/genética , Cromatografía Liquida , Femenino , Fumarato Hidratasa/genética , Fumarato Hidratasa/fisiología , Humanos , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/fisiología , Masculino , Espectrometría de Masas , Metaboloma/genética , Succinato Deshidrogenasa/genética , Succinato Deshidrogenasa/fisiología
5.
J Biol Chem ; 292(31): 12744-12753, 2017 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-28615439

RESUMEN

Fe-S cofactors are composed of iron and inorganic sulfur in various stoichiometries. A complex assembly pathway conducts their initial synthesis and subsequent binding to recipient proteins. In this minireview, we discuss how discovery of the role of the mammalian cytosolic aconitase, known as iron regulatory protein 1 (IRP1), led to the characterization of the function of its Fe-S cluster in sensing and regulating cellular iron homeostasis. Moreover, we present an overview of recent studies that have provided insights into the mechanism of Fe-S cluster transfer to recipient Fe-S proteins.


Asunto(s)
Homeostasis , Proteína 1 Reguladora de Hierro/fisiología , Hierro/fisiología , Modelos Moleculares , Animales , Apoenzimas/química , Apoenzimas/metabolismo , Liasas de Carbono-Azufre/biosíntesis , Liasas de Carbono-Azufre/química , Liasas de Carbono-Azufre/fisiología , Transporte de Electrón , Regulación Enzimológica de la Expresión Génica , Proteínas HSP70 de Choque Térmico/biosíntesis , Proteínas HSP70 de Choque Térmico/química , Proteínas HSP70 de Choque Térmico/fisiología , Humanos , Proteína 1 Reguladora de Hierro/biosíntesis , Proteína 1 Reguladora de Hierro/química , Proteínas de Unión a Hierro/biosíntesis , Proteínas de Unión a Hierro/química , Proteínas de Unión a Hierro/fisiología , Proteínas Reguladoras del Hierro/biosíntesis , Proteínas Reguladoras del Hierro/química , Proteínas Reguladoras del Hierro/fisiología , Proteínas Hierro-Azufre/biosíntesis , Proteínas Hierro-Azufre/química , Proteínas Hierro-Azufre/fisiología , Proteínas Mitocondriales/biosíntesis , Proteínas Mitocondriales/química , Proteínas Mitocondriales/fisiología , Chaperonas Moleculares/biosíntesis , Chaperonas Moleculares/química , Chaperonas Moleculares/fisiología , Pliegue de Proteína , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Elementos de Respuesta , Succinato Deshidrogenasa/biosíntesis , Succinato Deshidrogenasa/química , Succinato Deshidrogenasa/fisiología , Frataxina
6.
J Ovarian Res ; 7: 115, 2014 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-25491408

RESUMEN

BACKGROUND: Ovarian carcinoma is one of the most common gynecological cancers with high mortality rates. Numerous evidences demonstrate that cancer cells undergo metabolic abnormality during tumorigenesis in tumor microenvironment and further facilitate tumor progression. Succinate dehydrogenase (SDH or Complex II) is one of the important enzymes in the tricarboxylic acid (TCA) cycle. Succinate dehydrogenase subunit B (SDHB) gene, which encodes one of the four subunits of SDH, has been recognized as a tumor suppressor. However the role of SDHB in ovarian cancer is still unclear. METHODS: Using the SDHB specific siRNA and overexpression plasmid, the expression of SDHB was silenced and conversely induced in ovarian cancer cell lines SKOV3 and A2780, respectively. The possible role of SDHB in ovarian cancer was investigated in vitro, using proliferation, migration and invasion assays. To explore the mechanism, proliferation and migration related proteins such as Bcl-2, cleaved caspase 3, p-ERK, MMP-2, and p-FAK were examined by western blot. P-P38, p-AMPKα, and HIF-1α were also examined by western blot. CoCl2 was used to induce HIF-1α expression in SKOV3 and A2780 cells. RESULTS: SDHB silencing promoted cell proliferation, invasion, and migration, but inhibited apoptosis of SKOV3 and A2780 cells. In contrast, overexpression of SDHB inhibited cell proliferation, invasion, migration, and promoted apoptosis in SKOV3 cells. It was observed that up-regulation of Bcl-2 and MMP-2, activation of p-P38, p-ERK, and p-FAK, inhibition of cleaved caspase 3 in SDHB-silenced cells. Meanwhile, decreased Bcl-2 and MMP-2, inhibition of p-P38, p-ERK, and p-FAK, activation of cleaved caspase 3 were shown in SDHB-overexpressed SKOV3 cells. HIF-1α, an essential factor in tumor progression, was up-regulated in SDHB-silenced cells with the activation of p-AMPKα and down-regulated in SDHB-overexpressed cancer cells with the decreased p-AMPKα. And SDHB was proved to be decreased due to upregulation of HIF-1α expression in CoCl2-treated cancer cells. CONCLUSIONS: Our results firstly revealed that SDHB played a key role in cell proliferation, invasion, migration, and apoptosis of human ovarian carcinoma via AMPK-HIF-1α pathway. SDHB-overexpression might be a new approach to inhibit tumor progression in human ovarian carcinoma.


Asunto(s)
Adenilato Quinasa/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Ováricas/enzimología , Succinato Deshidrogenasa/fisiología , Adenosina Trifosfato/metabolismo , Apoptosis , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Invasividad Neoplásica , Neoplasias Ováricas/patología , ARN Interferente Pequeño/genética , Transducción de Señal
7.
Hepatogastroenterology ; 60(126): 1383-6, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24298573

RESUMEN

BACKGROUND/AIMS: Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors in the gastrointestinal tract and are mostly driven by KIT and PDGFRA-activation mutations. However, other signaling pathways are involved in pathogenesis and proliferation of GISTs. This study investigates the prognostic significance of insulin-like growth factor 1 (IGF1) and IGF1 receptor (IGF1R) and the role of succinate dehydrogenase subunit B (SDHB) in GISTs. METHODOLOGY: Immunohistochemistry (IHC) for IGF1, IGF1R and SDHB was performed in total of 165 GISTs. RESULTS: The overexpression of IGF1 was evident in tumors with high mitotic count, large tumor size and was correlated with high risk of malignant behavior. IGF1R overexpression was correlated with IGF overexpression, high mitotic count and high risk of malignant behavior. Loss of expression for SDHB was found in only 2 gastric GISTs. CONCLUSIONS: The overexpression of IGF1 and IGF1R can be useful marker to predict relapse and aggressive behavior in GISTs and has prognostic implications.


Asunto(s)
Neoplasias Gastrointestinales/patología , Tumores del Estroma Gastrointestinal/patología , Factor I del Crecimiento Similar a la Insulina/análisis , Receptor IGF Tipo 1/análisis , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Neoplasias Gastrointestinales/química , Neoplasias Gastrointestinales/mortalidad , Tumores del Estroma Gastrointestinal/química , Tumores del Estroma Gastrointestinal/mortalidad , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Succinato Deshidrogenasa/fisiología
8.
Cancer Cell ; 23(6): 739-52, 2013 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-23707781

RESUMEN

Paragangliomas are neuroendocrine tumors frequently associated with mutations in RET, NF1, VHL, and succinate dehydrogenase (SDHx) genes. Methylome analysis of a large paraganglioma cohort identified three stable clusters, associated with distinct clinical features and mutational status. SDHx-related tumors displayed a hypermethylator phenotype, associated with downregulation of key genes involved in neuroendocrine differentiation. Succinate accumulation in SDH-deficient mouse chromaffin cells led to DNA hypermethylation by inhibition of 2-OG-dependent histone and DNA demethylases and established a migratory phenotype reversed by decitabine treatment. Epigenetic silencing was particularly severe in SDHB-mutated tumors, potentially explaining their malignancy. Finally, inactivating FH mutations were identified in the only hypermethylated tumor without SDHx mutations. These findings emphasize the interplay between the Krebs cycle, epigenomic changes, and cancer.


Asunto(s)
Metilación de ADN , Paraganglioma/patología , Succinato Deshidrogenasa/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Movimiento Celular/genética , Niño , Células Cromafines/citología , Células Cromafines/metabolismo , Neoplasias Colorrectales/genética , Epigénesis Genética , Femenino , Técnicas de Inactivación de Genes , Silenciador del Gen , Glioblastoma/genética , Histonas/metabolismo , Humanos , Masculino , Ratones , Persona de Mediana Edad , Paraganglioma/genética , Fenotipo , Feocromocitoma/genética , Feocromocitoma/patología , Succinato Deshidrogenasa/metabolismo , Succinato Deshidrogenasa/fisiología , Transcriptoma
9.
Cardiovasc Res ; 93(4): 702-10, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22215723

RESUMEN

AIMS: Alveolar hypoxia acutely elicits contraction of pulmonary arteries, leading to a rise in pulmonary arterial pressure (PAP) and shifting blood to better ventilated areas of the lung. The molecular mechanisms underlying this hypoxic pulmonary vasoconstriction (HPV) are still incompletely understood. Here, we investigated the role of succinate dehydrogenase (SDH; synonymous to mitochondrial complex II) in HPV, with particular emphasis on regional differences along the vascular bed and consequences for PAP and perfusion-to-ventilation matching, using mutant mice heterozygous for the SDHD subunit of complex II (SDHD(+/-)). METHODS AND RESULTS: Western blots revealed reduced protein content of complex II subunits SDHA, SDHB, and SDHC in lungs of SDHD(+/-) mice, despite unaffected mRNA content as determined by real-time PCR. Hypoxic pulmonary vasoconstriction of small (20-50 µm) intra-acinar and larger (51-100 µm) pre-acinar arteries was evaluated by videomorphometric analysis of precision-cut lung slices. The hypoxic response was detectable in pre-acinar arteries but absent from intra-acinar arteries of SDHD(+/-) mice. In isolated perfused lungs, basal PAP and its hypoxia-induced increase were indistinguishable between both mouse strains. Arterial oxygenation was measured after provocation of regional ventilatory failure by tracheal fluid instillation in anaesthetized mice, and it declined more in SDHD(+/-) than in wild-type mice. CONCLUSION: SDHD is required for the formation of a stable mitochondrial complex II and it is selectively important for HPV of intra-acinar vessels. This specialized vascular segment participates in perfusion-to-ventilation matching but does not significantly contribute to the acute hypoxic rise in PAP that results from more proximal vasoconstriction.


Asunto(s)
Hipoxia/fisiopatología , Pulmón/irrigación sanguínea , Arteria Pulmonar/fisiopatología , Succinato Deshidrogenasa/fisiología , Vasoconstricción/fisiología , Animales , Presión Sanguínea/fisiología , Complejo II de Transporte de Electrones/genética , Complejo II de Transporte de Electrones/fisiología , Heterocigoto , Pulmón/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Ratones , Ratones Mutantes , Modelos Animales , ARN Mensajero/metabolismo , Succinato Deshidrogenasa/genética
10.
Can J Physiol Pharmacol ; 89(11): 819-27, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22032694

RESUMEN

In this study, we have postulated that in healthy males, peak aerobic power ([Formula: see text]) would associate with muscle capillary density rather than oxidative potential, regardless of fibre type or subtype. To test this, active but untrained volunteers (n = 11) were separated into high (HI) and low (LO) groups based on [Formula: see text] obtained during a progressive cycle task to fatigue. The 26% higher (P < 0.05) [Formula: see text] observed in HI (40.8 ± 1.5 mL·kg(-1)·min(-1), mean ± SE) compared with LO ( 51.4 ± 0.90 mL·kg(-1)·min(-1), mean ± SE) was not accompanied by differences in age (21.3 ± 1.2 compared with 21.1 ± 0.63 years, respectively) or body mass (72.4 ± 4.6 compared with 71.6 ± 1.9 kg, respectively). Tissue samples obtained from the vastus lateralis indicated greater (P < 0.05) capillary counts per fibre (CC; +24%) in HI compared with LO, regardless of fibre type (I, IIA, IIX, IIAX). Capillary density (CD) as measured in a field of defined area was also elevated (+22%; P < 0.05), as was the number of capillaries per fibre (+22%; P < 0.05). No differences were observed between the 2 groups in the distribution, area, and the CC/fibre area ratio in the different fibre types and subtypes. Similarly, there was no difference between the HI and LO groups in oxidative potential, as measured by succinic dehydrogenase activity in the different fibre types. It is concluded that the higher capillary density may contribute to improved vascular conductance and the elevated [Formula: see text] observed in the untrained participants.


Asunto(s)
Capilares/fisiología , Ejercicio Físico , Fibras Musculares Esqueléticas/fisiología , Músculo Esquelético/fisiología , Músculo Cuádriceps/fisiología , Adulto , Prueba de Esfuerzo , Humanos , Masculino , Fibras Musculares Esqueléticas/citología , Músculo Esquelético/irrigación sanguínea , Músculos/fisiología , Oxidación-Reducción , Músculo Cuádriceps/citología , Proyectos de Investigación , Succinato Deshidrogenasa/análisis , Succinato Deshidrogenasa/fisiología , Adulto Joven
11.
Clin Auton Res ; 19(6): 335-42, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19768395

RESUMEN

The detection of hypoxia by the carotid bodies elicits a ventilatory response of utmost importance for tolerance to high altitude. Germline mutations in three genes encoding subunit B, C and D of succinate dehydrogenase (SDHB, SDHC and SDHD) have been associated with paragangliomas of the carotid body. We hypothesized that SDH dysfunction within the carotid body could result in low chemoresponsiveness and intolerance to high altitude. The frequency of polymorphisms of SDHs, hypoxia-inducible factor type 1 (HIF1alpha) and angiotensin converting enzyme (ACE) genes was compared between 40 subjects with intolerance to high altitude and a low hypoxic ventilatory response at exercise (HVRe < or = 0.5 ml min(-1) kg(-1); HVR- group) and 41 subjects without intolerance to high altitude and a high HVRe (> or = 0.80 ml min(-1) kg(-1); HVR+). We found no significant association between low or high HVRe and (1) the allele frequencies for nine single nucleotide polymorphisms (SNPs) in the SDHD and SDHB genes, (2) the ACE insertion/deletion polymorphism and (3) four SNPs in the HIF1alpha gene. However, a marginal significant association was found between the synonymous polymorphism c.18A>C of the SDHB gene and chemoresponsiveness: 8/40 (20%) in the HVR- group and 3/41 (7%) in the HVR+ group (p = 0.12). A principal component analysis showed that no subject carrying the 18C allele had both high ventilatory and cardiac response to hypoxia. In conclusion, no clear association was found between gene variants involved in oxygen sensing and chemoresponsiveness, although some mutations in the SDHB and SDHD genes deserve further investigations in a larger population.


Asunto(s)
Cuerpo Carotídeo/fisiología , Proteínas de la Membrana/fisiología , Oxígeno/fisiología , Ventilación Pulmonar/genética , Succinato Deshidrogenasa/fisiología , Adulto , Altitud , Anaerobiosis , Cuerpo Carotídeo/enzimología , Ejercicio Físico/fisiología , Femenino , Frecuencia de los Genes/genética , Mutación de Línea Germinal , Frecuencia Cardíaca/genética , Frecuencia Cardíaca/fisiología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Masculino , Proteínas de la Membrana/genética , Peptidil-Dipeptidasa A/genética , Polimorfismo de Nucleótido Simple/genética , Ventilación Pulmonar/fisiología , Succinato Deshidrogenasa/genética
12.
Am J Hum Genet ; 83(2): 261-8, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18678321

RESUMEN

Individuals with PTEN mutations have Cowden syndrome (CS), associated with breast, thyroid, and endometrial neoplasias. Many more patients with features of CS, not meeting diagnostic criteria (termed CS-like), are evaluated by clinicians for CS-related cancer risk. Germline mutations in succinate dehydrogenase subunits SDHB-D cause pheochromocytoma-paraganglioma syndrome. One to five percent of SDHB/SDHD mutation carriers have renal cell or papillary thyroid carcinomas, which are also CS-related features. SDHB-D may be candidate susceptibility genes for some PTEN mutation-negative individuals with CS-like cancers. To address this hypothesis, germline SDHB-D mutation analysis in 375 PTEN mutation-negative CS/CS-like individuals was performed, followed by functional analysis of identified SDH mutations/variants. Of 375 PTEN mutation-negative CS/CS-like individuals, 74 (20%) had increased manganese superoxide dismutase (MnSOD) expression, a manifestation of mitochondrial dysfunction. Among these, 10 (13.5%) had germline mutations/variants in SDHB (n = 3) or SDHD (7), not found in 700 controls (p < 0.001). Compared to PTEN mutation-positive CS/CS-like individuals, those with SDH mutations/variants were enriched for carcinomas of the female breast (6/9 SDH versus 30/107 PTEN, p < 0.001), thyroid (5/10 versus 15/106, p < 0.001), and kidney (2/10 versus 4/230, p = 0.026). In the absence of PTEN alteration, CS/CS-like-related SDH mutations/variants show increased phosphorylation of AKT and/or MAPK, downstream manifestations of PTEN dysfunction. Germline SDH mutations/variants occur in a subset of PTEN mutation-negative CS/CS-like individuals and are associated with increased frequencies of breast, thyroid, and renal cancers beyond those conferred by germline PTEN mutations. SDH testing should be considered for germline PTEN mutation-negative CS/CS-like individuals, especially in the setting of breast, thyroid, and/or renal cancers.


Asunto(s)
Regulación de la Expresión Génica , Variación Genética , Mutación de Línea Germinal , Síndrome de Hamartoma Múltiple/genética , Succinato Deshidrogenasa/genética , Adulto , Anciano , Femenino , Humanos , Masculino , Microscopía Confocal , Persona de Mediana Edad , Fosfohidrolasa PTEN/genética , Fenotipo , Succinato Deshidrogenasa/fisiología
13.
Proc Natl Acad Sci U S A ; 103(49): 18745-50, 2006 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-17124169

RESUMEN

Legionella pneumophila requires the Dot/Icm protein translocation system to replicate within host cells as a critical component of Legionnaire's pneumonia. None of the known individual substrates of the translocator have been shown to be essential for intracellular replication. We demonstrate here that mutants lacking the Dot/Icm substrate SdhA were severely impaired for intracellular growth within mouse bone marrow macrophages, with the defect absolute in triple mutants lacking sdhA and its two paralogs. The defect caused by the absence of the sdhA family was less severe during growth within Dictyostelium discoideum amoebae, indicating that the requirement for SdhA shows cell-type specificity. Macrophages harboring the L. pneumophila sdhA mutant showed increased nuclear degradation, mitochondrial disruption, membrane permeability, and caspase activation, indicating a role for SdhA in preventing host cell death. Defective intracellular growth of the sdhA(-) mutant could be partially suppressed by the action of caspase inhibitors, but caspase-independent cell death pathways eventually aborted replication of the mutant.


Asunto(s)
Apoptosis/fisiología , Proteínas Bacterianas/fisiología , Flavoproteínas/fisiología , Legionella pneumophila/crecimiento & desarrollo , Macrófagos/enzimología , Macrófagos/microbiología , Succinato Deshidrogenasa/metabolismo , Animales , Proteínas Bacterianas/genética , Transporte Biológico/genética , Células Cultivadas , Flavoproteínas/genética , Legionella pneumophila/enzimología , Legionella pneumophila/genética , Ratones , Ratones Endogámicos A , Mutación , Especificidad por Sustrato/genética , Succinato Deshidrogenasa/genética , Succinato Deshidrogenasa/fisiología
14.
Oncol Res ; 15(7-8): 393-8, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16491957

RESUMEN

Neuroblastoma and pheochromocytoma have the same embryonal origin. They originate from neural crest cells, and they usually affect suprarenal glands. The SDHB gene encodes the B subunit of succinate dehydrogenase, a protein implicated in the electron transport chain and Krebs cycle. Some mutations have been described in this gene in pheochromocytoma, and this gene could be an appropriate candidate for its study in neuroblastoma given its localization in 1p35-36. The aim of this study was to analyze neuroblastoma tumors in order to assess a possible implication of this gene in neuroblastoma development. We studied 28 neuroblastoma tumor samples from different stages. Mutation research in genomic DNA was carried out after individual amplification of each of the eight SDHB exons by SSCP analysis and sequencing of those samples with migration pattern variants. No variant was found except for three polymorphisms in four neuroblastoma samples. The first polymorphism was a synonymous A-->C change in the third position of codon 6 (exon 1). The other two polymorphisms were a TTC insert at the 5' flanking intron sequence of exon 5 in a stretch of seven TTC repeats. Upon the basis of posterior microsatellite instability and hypermethylation promoter studies, which were not significant, we can conclude that the SDHB gene, a positional candidate gene, is unlikely to be related to either initiation or tumoral progression in neuroblastoma.


Asunto(s)
Proteínas Hierro-Azufre/genética , Proteínas Hierro-Azufre/fisiología , Neuroblastoma/genética , Subunidades de Proteína/genética , Subunidades de Proteína/fisiología , Succinato Deshidrogenasa/genética , Succinato Deshidrogenasa/fisiología , Análisis Mutacional de ADN , ADN de Neoplasias , Progresión de la Enfermedad , Humanos , Estadificación de Neoplasias , Neuroblastoma/fisiopatología , Polimorfismo Genético , Polimorfismo Conformacional Retorcido-Simple
15.
Proc Natl Acad Sci U S A ; 101(32): 11880-5, 2004 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-15284438

RESUMEN

The mitochondrial ATP-sensitive K(+) (mitoK(ATP)) channel plays a central role in protection of cardiac and neuronal cells against ischemia and apoptosis, but its molecular structure is unknown. Succinate dehydrogenase (SDH) is inhibited by mitoK(ATP) activators, fueling the contrary view that SDH, rather than mitoK(ATP), is the target of cardioprotective drugs. Here, we report that SDH forms part of mitoK(ATP) functionally and structurally. Four mitochondrial proteins [mitochondrial ATP-binding cassette protein 1 (mABC1), phosphate carrier, adenine nucleotide translocator, and ATP synthase] associate with SDH. A purified IM fraction containing these proteins was reconstituted into proteoliposomes and lipid bilayers and shown to confer mitoK(ATP) channel activity. This channel activity is sensitive not only to mitoK(ATP) activators and blockers but also to SDH inhibitors. These results reconcile the controversy over the basis of ischemic preconditioning by demonstrating that SDH is a component of mitoK(ATP) as part of a macromolecular supercomplex. The findings also provide a tangible clue as to the structural basis of mitoK(ATP) channels.


Asunto(s)
Proteínas Mitocondriales/metabolismo , Canales de Potasio/metabolismo , Succinato Deshidrogenasa/fisiología , Adenosina Trifosfato/farmacología , Animales , Western Blotting , Electrofisiología , Precondicionamiento Isquémico , Liposomas , Sustancias Macromoleculares , Mitocondrias Hepáticas/química , Mitocondrias Hepáticas/enzimología , Mitocondrias Hepáticas/metabolismo , Proteínas Mitocondriales/aislamiento & purificación , Proteínas Mitocondriales/fisiología , Complejos Multiproteicos , Unión Proteica , Ratas , Succinato Deshidrogenasa/aislamiento & purificación , Succinato Deshidrogenasa/metabolismo
16.
Oncogene ; 22(9): 1358-64, 2003 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-12618761

RESUMEN

Phaeochromocytomas arising in adrenal or extra-adrenal sites and paragangliomas of the head and neck, in particular of the carotid bodies, occur sporadically and also in a familial setting. In addition to mutations in RET and VHL in familial disease, germline mutations in SDHD and SDHB genes that encode subunits of mitochondrial complex II have also been associated with the development of familial phaeochromocytomas. To further investigate the role of SDHD and SDHB in the development of these tumours we determined the occurrence of germline SDHD and SDHB mutations in four patients with a family history of phaeochromocytoma with associated head and neck paraganglioma, one patient with a family history of phaeochromocytoma only and two patients with apparently sporadic extra-adrenal phaeochromocytoma, one of whom had early onset disease. Secondly, we investigated whether somatic SDHB mutations correlated with loss of heterozygosity at 1p36 in a subgroup of 11 sporadic and three MEN 2-associated RET-mutation-positive phaeochromocytomas. Novel SDHB mutations were identified in the probands from four families and two apparently sporadic cases (six of seven probands studied), including two missense mutations, a single nonsense and frameshift mutation, as well as two splice site mutations, one of which was shown to have partial penetrance resulting in 'leaky' splicing. Further, five intronic polymorphisms in SDHB were found. No SDHD mutations were identified. In addition, no somatic SDHB mutations were found in the remaining allele of the 11 sporadic adrenal phaeochromocytomas with allelic loss at 1p36 or the three MEN 2-associated RET-mutation-positive phaeochromocytomas. Therefore, we conclude that SDHB has a major role in the pathogenesis of familial phaeochromocytomas, but the possible role of SDHB in sporadic tumours showing allelic loss at 1p36 has yet to be ascertained.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/genética , Proteínas de Neoplasias/genética , Paraganglioma/genética , Feocromocitoma/genética , Subunidades de Proteína/genética , Neoplasias Retroperitoneales/genética , Succinato Deshidrogenasa/genética , Adolescente , Neoplasias de las Glándulas Suprarrenales/enzimología , Adulto , Edad de Inicio , Australia/epidemiología , Niño , Cromosomas Humanos Par 1/genética , Análisis Mutacional de ADN , ADN de Neoplasias/genética , Complejo II de Transporte de Electrones , Mutación del Sistema de Lectura , Mutación de Línea Germinal , Neoplasias de Cabeza y Cuello/enzimología , Neoplasias de Cabeza y Cuello/epidemiología , Neoplasias de Cabeza y Cuello/genética , Humanos , Intrones/genética , Proteínas Hierro-Azufre , Pérdida de Heterocigocidad , Masculino , Persona de Mediana Edad , Complejos Multienzimáticos/genética , Neoplasia Endocrina Múltiple/enzimología , Neoplasia Endocrina Múltiple/epidemiología , Neoplasia Endocrina Múltiple/genética , Mutación Missense , Proteínas de Neoplasias/fisiología , Síndromes Neoplásicos Hereditarios/enzimología , Síndromes Neoplásicos Hereditarios/genética , Oxidorreductasas/genética , Paraganglioma/enzimología , Paraganglioma/epidemiología , Linaje , Feocromocitoma/enzimología , Feocromocitoma/epidemiología , Subunidades de Proteína/deficiencia , Subunidades de Proteína/fisiología , Sitios de Empalme de ARN/genética , Neoplasias Retroperitoneales/enzimología , Succinato Deshidrogenasa/deficiencia , Succinato Deshidrogenasa/fisiología
17.
Nat Rev Cancer ; 3(3): 193-202, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12612654

RESUMEN

Mitochondrial defects have been associated with neurological disorders, as well as cancers. Two ubiquitously expressed mitochondrial enzymes--succinate dehydrogenase (SDH) and fumarate hydratase (FH, fumarase)--catalyse sequential steps in the Krebs tricarboxylic-acid cycle. Inherited heterozygous mutations in the genes encoding these enzymes cause predispositions to two types of inherited neoplasia syndromes that do not share any component tumours. Homozygous mutations in the same genes result in severe neurological impairment. Understanding this link between inherited cancer syndromes and neurological disease could provide further insights into the mechanisms by which mitochondrial deficiencies lead to tumour development.


Asunto(s)
Ciclo del Ácido Cítrico/genética , Fumarato Hidratasa/fisiología , Mitocondrias/enzimología , Encefalomiopatías Mitocondriales/enzimología , Complejos Multienzimáticos/fisiología , Síndromes Neoplásicos Hereditarios/enzimología , Oxidorreductasas/fisiología , Succinato Deshidrogenasa/fisiología , Apoptosis/genética , Apoptosis/fisiología , Carcinoma de Células Renales/enzimología , Carcinoma de Células Renales/genética , Complejo II de Transporte de Electrones , Metabolismo Energético , Predicción , Radicales Libres , Fumarato Hidratasa/deficiencia , Fumarato Hidratasa/genética , Heterocigoto , Homocigoto , Humanos , Neoplasias Renales/enzimología , Neoplasias Renales/genética , Leiomiomatosis/enzimología , Leiomiomatosis/genética , Encefalomiopatías Mitocondriales/genética , Complejos Multienzimáticos/deficiencia , Complejos Multienzimáticos/genética , Mutación , Síndromes Neoplásicos Hereditarios/genética , Neovascularización Fisiológica/genética , Neovascularización Fisiológica/fisiología , Oxidorreductasas/deficiencia , Oxidorreductasas/genética , Paraganglioma/enzimología , Paraganglioma/genética , Feocromocitoma/enzimología , Feocromocitoma/genética , Subunidades de Proteína , Succinato Deshidrogenasa/deficiencia , Succinato Deshidrogenasa/genética
18.
Expert Opin Ther Targets ; 6(2): 135-46, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12223076

RESUMEN

Approximately 50% of the world's population carries Helicobacter pylori, a gastric bacterial pathogen linked to diseases including gastritis, ulcers and gastric cancer. Chemotherapies are being routinely used to treat systemic H. pylori infection. The common regimens consist of proton pump inhibitors (PPIs) or ranitidine bismuth citrate (RBC) and two antibiotics. Although these regimens efficiently eradicate H. pylori, the emergence of antibiotic-resistant H. pylori strains, their severe side effects and high costs are major drawbacks of these treatments. More efficient, economic and friendly drugs need to be developed. Fumarate reductase (FRD) catalyses the reduction of fumarate to succinate in the Krebs cycle and is also a key enzyme in anaerobic respiration with fumarate as the terminal electron acceptor for many facultative bacteria. H. pylori FRD contains three subunits, FrdA, FrdB and FrdC. Genome analysis and experimental evidence indicate that this enzyme appears to play an important role in the energy metabolism of H. pylori. In addition, FRD is essential for the colonisation of H. pylori in the acidic stomach as demonstrated in the mouse model of infection. Furthermore, three FRD inhibitors used to cure helminthic infection in animals and humans have both inhibitory and bactericidal effects on H. pylori. These lines of evidence indicate that FRD may be a promising chemotherapeutic target. Given that FrdA is strongly immunogenic in the sera from H. pylori-positive patients, this protein may also be used as a candidate for the development of an anti-H. pylori vaccine.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Gastritis/tratamiento farmacológico , Infecciones por Helicobacter/tratamiento farmacológico , Helicobacter pylori/enzimología , Succinato Deshidrogenasa/efectos de los fármacos , Animales , Antihelmínticos/farmacología , Antibacterianos/uso terapéutico , Anticuerpos Antibacterianos/inmunología , Proteínas Bacterianas/química , Proteínas Bacterianas/fisiología , Vacunas Bacterianas , Diseño de Fármacos , Inhibidores Enzimáticos/uso terapéutico , Helicobacter pylori/efectos de los fármacos , Helicobacter pylori/genética , Helicobacter pylori/inmunología , Humanos , Ratones , Subunidades de Proteína , Succinato Deshidrogenasa/química , Succinato Deshidrogenasa/fisiología
19.
Mol Pharmacol ; 48(5): 928-37, 1995 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7476924

RESUMEN

The role of complex II in the cellular protection against oxidative stress was investigated in freshly isolated rat renal proximal tubular cells (PTC) with the use of the nephrotoxin S-(1,2-dichlorovinyl)-L-cysteine (DCVC). DCVC caused oxidative stress in PTC as determined by flow cytometry with dihydrorhodamine-123; this fluorescent probe is readily oxidized by primary hydroperoxides such as those formed during lipid peroxidation. The oxidative stress could be prevented by inhibition of the beta-lyase-mediated formation and covalent binding to cellular macromolecules of reactive DCVC metabolites, with amino oxyacetic acid (AOA), or by the antioxidant N,N'-diphenyl-p-phenylenediamine. Both AOA and DPPD also prevented cell death. The DCVC-induced oxidative stress was associated with a decrease in the succinate:ubiquinone reductase (SQR) activity of complex II, whereas NADH:ubiquinone reductase activity of complex I remained unaffected. AOA prevented the effect on SQR activity, whereas N,N'-diphenyl-p-phenylenediamine did not. Inhibition of SQR activity with thenoyl trifluoracetone (TTFA) potentiated the DCVC-induced oxidative cell injury, suggesting the involvement of SQR activity in an antioxidant pathway. To investigate this in greater detail, PTC were treated with an inhibitor of cytochrome-c-oxidase, KCN, in a buffer containing glycine, which prevents cell death by KCN. Glycine did not affect cell death by DCVC. KCN prevented the DCVC-induced oxidative stress and cell death. KCN cytoprotection could be prevented by inhibition of SQR activity with oxaloacetate or TTFA, whereas inhibition of either complex I or III with rotenone and antimycin, respectively, did not prevent it. The effect of DCVC on complex II was associated with a decrease in the cellular amount of reduced ubiquinone (QH2); the KCN-mediated cytoprotection was related to a 60% increase of cellular QH2. Rotenone almost completely inhibited ubiquinone reduction even in the presence of KCN, whereas oxaloacetate in combination with KCN resulted in QH2 levels comparable to control. This suggests that the SQR activity by complex II rather than the cellular content of reduced ubiquinone (QH2) is important as a part of the cellular antioxidant machinery in the cyto-protection against oxidative stress.


Asunto(s)
Cisteína/análogos & derivados , Riñón/efectos de los fármacos , Complejos Multienzimáticos/fisiología , Estrés Oxidativo , Oxidorreductasas/fisiología , Succinato Deshidrogenasa/fisiología , Ubiquinona/análogos & derivados , Animales , Muerte Celular/efectos de los fármacos , Cisteína/metabolismo , Cisteína/toxicidad , Complejo I de Transporte de Electrón , Complejo II de Transporte de Electrones , Complejo IV de Transporte de Electrones/antagonistas & inhibidores , Peróxido de Hidrógeno/metabolismo , Riñón/metabolismo , Masculino , Complejos Multienzimáticos/antagonistas & inhibidores , NADH NADPH Oxidorreductasas/metabolismo , Oxidorreductasas/antagonistas & inhibidores , Cianuro de Potasio/farmacología , Ratas , Ratas Wistar , Succinato Deshidrogenasa/antagonistas & inhibidores , Ubiquinona/metabolismo
20.
J Neurochem ; 64(1): 430-6, 1995 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-7528265

RESUMEN

Although the mechanism of neuronal death in neurodegenerative diseases remains unknown, it has been hypothesized that relatively minor metabolic defects may predispose neurons to N-methyl-D-aspartate (NMDA) receptor-mediated excitotoxic damage in these disorders. To further investigate this possibility, we have characterized the excitotoxic potential of the reversible succinate dehydrogenase (SDH) inhibitor malonate. After its intrastriatal stereotaxic injection into male Sprague-Dawley rats, malonate produced a dose-dependent lesion when assessed 3 days after surgery using cytochrome oxidase histochemistry. This lesion was attenuated by coadministration of excess succinate, indicating that it was caused by specific inhibition of SDH. The lesion was also prevented by administration of the noncompetitive NMDA antagonist MK-801. MK-801 did not induce hypothermia, and hypothermia itself was not neuroprotective, suggesting that the neuroprotective effect of MK-801 was due to blockade of the NMDA receptor ion channel and not to any nonspecific effect. The competitive NMDA antagonist LY274614 and the glycine site antagonist 7-chlorokynurenate also profoundly attenuated malonate neurotoxicity, further indicating an NMDA receptor-mediated event. Finally, the alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA) antagonist NBQX (2,3-dihydroxy-6-nitro-7-sulfamoylbenzo(f)-quinoxaline) was ineffective at preventing malonate toxicity at a dose that effectively reduced S-AMPA toxicity, indicating that non-NMDA receptors are involved minimally, if at all, in the production of the malonate lesion. We conclude that inhibition of SDH by malonate results in NMDA receptor-mediated excitotoxic neuronal death. If this mechanism of "secondary" or "weak" excitotoxicity plays a role in neurodegenerative disease, NMDA antagonists and other "antiexcitotoxic" strategies may have therapeutic potential for these diseases.


Asunto(s)
Malonatos/farmacología , Succinato Deshidrogenasa/antagonistas & inhibidores , Animales , Temperatura Corporal/efectos de los fármacos , Encefalopatías/inducido químicamente , Encefalopatías/patología , Encefalopatías/prevención & control , Muerte Celular/efectos de los fármacos , Cuerpo Estriado/química , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/enzimología , Maleato de Dizocilpina/farmacología , Relación Dosis-Respuesta a Droga , Ácido Glutámico/metabolismo , Glicina/metabolismo , Isoquinolinas/farmacología , Masculino , Malonatos/efectos adversos , Neuronas/efectos de los fármacos , Neuronas/patología , Quinoxalinas/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Ácido Kaínico/fisiología , Receptores de N-Metil-D-Aspartato/análisis , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/fisiología , Succinato Deshidrogenasa/fisiología , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/antagonistas & inhibidores , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA