Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 11(1): 970, 2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-32080200

RESUMO

Deregulation of mitochondrial network in terminally differentiated cells contributes to a broad spectrum of disorders. Methylmalonic acidemia (MMA) is one of the most common inherited metabolic disorders, due to deficiency of the mitochondrial methylmalonyl-coenzyme A mutase (MMUT). How MMUT deficiency triggers cell damage remains unknown, preventing the development of disease-modifying therapies. Here we combine genetic and pharmacological approaches to demonstrate that MMUT deficiency induces metabolic and mitochondrial alterations that are exacerbated by anomalies in PINK1/Parkin-mediated mitophagy, causing the accumulation of dysfunctional mitochondria that trigger epithelial stress and ultimately cell damage. Using drug-disease network perturbation modelling, we predict targetable pathways, whose modulation repairs mitochondrial dysfunctions in patient-derived cells and alleviate phenotype changes in mmut-deficient zebrafish. These results suggest a link between primary MMUT deficiency, diseased mitochondria, mitophagy dysfunction and epithelial stress, and provide potential therapeutic perspectives for MMA.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Erros Inatos do Metabolismo dos Aminoácidos/patologia , Erros Inatos do Metabolismo/metabolismo , Erros Inatos do Metabolismo/patologia , Metilmalonil-CoA Mutase/deficiência , Doenças Mitocondriais/metabolismo , Doenças Mitocondriais/patologia , Mitofagia/fisiologia , Alquil e Aril Transferases/deficiência , Alquil e Aril Transferases/genética , Erros Inatos do Metabolismo dos Aminoácidos/genética , Animais , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Técnicas de Inativação de Genes , Humanos , Masculino , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Erros Inatos do Metabolismo/genética , Metilmalonil-CoA Mutase/genética , Metilmalonil-CoA Mutase/metabolismo , Camundongos , Camundongos Knockout , Doenças Mitocondriais/genética , Mitofagia/genética , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Estresse Fisiológico , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Peixe-Zebra
2.
Gastroenterology ; 157(5): 1293-1309, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31302143

RESUMO

BACKGROUND & AIMS: It is not clear how regulation of T-cell function is altered during development of inflammatory bowel diseases (IBD). We studied the mechanisms by which geranylgeranyltransferase-mediated prenylation controls T-cell localization to the intestine and chronic inflammation. METHODS: We generated mice with T-cell-specific disruption of the geranylgeranyltransferase type I, beta subunit gene (Pggt1b), called Pggt1bΔCD4 mice, or the ras homolog family member A gene (Rhoa), called RhoaΔCD4 mice. We also studied mice with knockout of CDC42 or RAC1 and wild-type mice (controls). Intestinal tissues were analyzed by histology, multiphoton and confocal microscopy, and real-time polymerase chain reaction. Activation of CDC42, RAC1, and RHOA were measured with G-LISA, cell fractionation, and immunoblots. T cells and lamina propria mononuclear cells from mice were analyzed by flow cytometry or transferred to Rag1-/- mice. Mice were given injections of antibodies against integrin alpha4beta7 or gavaged with the RORC antagonist GSK805. We obtained peripheral blood and intestinal tissue samples from patients with and without IBD and analyzed them by flow cytometry. RESULTS: Pggt1bΔCD4 mice developed spontaneous colitis, characterized by thickening of the intestinal wall, edema, fibrosis, accumulation of T cells in the colon, and increased expression of inflammatory cytokines. Compared with control CD4+ T cells, PGGT1B-deficient CD4+ T cells expressed significantly higher levels of integrin alpha4beta7, which regulates their localization to the intestine. Inflammation induced by transfer of PGGT1B-deficient CD4+ T cells to Rag1-/- mice was blocked by injection of an antibody against integrin alpha4beta7. Lamina propria of Pggt1bΔCD4 mice had increased numbers of CD4+ T cells that expressed RORC and higher levels of cytokines produced by T-helper 17 cells (granulocyte-macrophage colony-stimulating factor, interleukin [IL]17A, IL17F, IL22, and tumor necrosis factor [TNF]). The RORC inverse agonist GSK805, but not antibodies against IL17A or IL17F, prevented colitis in Pggt1bΔCD4 mice. PGGT1B-deficient CD4+ T cells had decreased activation of RHOA. RhoAΔCD4 mice had a similar phenotype to Pggt1bΔCD4 mice, including development of colitis, increased numbers of CD4+ T cells in colon, increased expression of integrin alpha4beta7 by CD4+ T cells, and increased levels of IL17A and other inflammatory cytokines in lamina propria. T cells isolated from intestinal tissues from patients with IBD had significantly lower levels of PGGT1B than tissues from individuals without IBD. CONCLUSION: Loss of PGGT1B from T cells in mice impairs RHOA function, increasing CD4+ T-cell expression of integrin alpha4beta7 and localization to colon, resulting in increased expression of inflammatory cytokines and colitis. T cells isolated from gut tissues from patients with IBD have lower levels of PGGT1B than tissues from patients without IBD.


Assuntos
Alquil e Aril Transferases/deficiência , Quimiotaxia de Leucócito , Colite/enzimologia , Colo/enzimologia , Integrinas/metabolismo , Linfócitos T/enzimologia , Proteínas rho de Ligação ao GTP/metabolismo , Imunidade Adaptativa , Alquil e Aril Transferases/genética , Animais , Estudos de Casos e Controles , Células Cultivadas , Colite/genética , Colite/imunologia , Colite/patologia , Colo/imunologia , Colo/patologia , Citocinas/metabolismo , Modelos Animais de Doenças , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Imunidade Inata , Mediadores da Inflamação/metabolismo , Ativação Linfocitária , Camundongos Knockout , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Transdução de Sinais , Linfócitos T/imunologia , Linfócitos T/patologia , Proteína cdc42 de Ligação ao GTP/genética , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/deficiência , Proteínas rho de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP
3.
J Am Soc Nephrol ; 28(9): 2607-2617, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28428331

RESUMO

Clinical studies have identified patients with nephrotic syndrome caused by mutations in genes involved in the biosynthesis of coenzyme Q10 (CoQ10), a lipid component of the mitochondrial electron transport chain and an important antioxidant. However, the cellular mechanisms through which these mutations induce podocyte injury remain obscure. Here, we exploited the striking similarities between Drosophila nephrocytes and human podocytes to develop a Drosophila model of these renal diseases, and performed a systematic in vivo analysis assessing the role of CoQ10 pathway genes in renal function. Nephrocyte-specific silencing of Coq2, Coq6, and Coq8, which are genes involved in the CoQ10 pathway that have been associated with genetic nephrotic syndrome in humans, induced dramatic adverse changes in these cells. In particular, silencing of Coq2 led to an abnormal localization of slit diaphragms, collapse of lacunar channels, and more dysmorphic mitochondria. In addition, Coq2-deficient nephrocytes showed elevated levels of autophagy and mitophagy, increased levels of reactive oxygen species, and increased sensitivity to oxidative stress. Dietary supplementation with CoQ10 at least partially rescued these defects. Furthermore, expressing the wild-type human COQ2 gene specifically in nephrocytes rescued the defective protein uptake, but expressing the mutant allele derived from a patient with COQ2 nephropathy did not. We conclude that transgenic Drosophila lines carrying mutations in the CoQ10 pathway genes are clinically relevant models with which to explore the pathogenesis of podocyte injury and could serve as a new platform to test novel therapeutic approaches.


Assuntos
Alquil e Aril Transferases/genética , Síndrome Nefrótica/genética , Síndrome Nefrótica/metabolismo , Ubiquinona/análogos & derivados , Vitaminas/farmacologia , Alquil e Aril Transferases/deficiência , Alelos , Animais , Autofagia/efeitos dos fármacos , Linhagem Celular , Células Cultivadas , Modelos Animais de Doenças , Inativação Gênica , Humanos , Mitocôndrias/ultraestrutura , Mitofagia/efeitos dos fármacos , Organismos Geneticamente Modificados , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/genética , Ubiquinona/biossíntese , Ubiquinona/genética , Ubiquinona/farmacologia , Vitaminas/biossíntese
4.
Eur J Hum Genet ; 23(9): 1254-8, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25564041

RESUMO

Coenzyme Q10 deficiency is a clinically and genetically heterogeneous disorder, with manifestations that may range from fatal neonatal multisystem failure, to adult-onset encephalopathy. We report a patient who presented at birth with severe lactic acidosis, proteinuria, dicarboxylic aciduria, and hepatic insufficiency. She also had dilation of left ventricle on echocardiography. Her neurological condition rapidly worsened and despite aggressive care she died at 23 h of life. Muscle histology displayed lipid accumulation. Electron microscopy showed markedly swollen mitochondria with fragmented cristae. Respiratory-chain enzymatic assays showed a reduction of combined activities of complex I+III and II+III with normal activities of isolated complexes. The defect was confirmed in fibroblasts, where it could be rescued by supplementing the culture medium with 10 µM coenzyme Q10. Coenzyme Q10 levels were reduced (28% of controls) in these cells. We performed exome sequencing and focused the analysis on genes involved in coenzyme Q10 biosynthesis. The patient harbored a homozygous c.545T>G, p.(Met182Arg) alteration in COQ2, which was validated by functional complementation in yeast. In this case the biochemical and morphological features were essential to direct the genetic diagnosis. The parents had another pregnancy after the biochemical diagnosis was established, but before the identification of the genetic defect. Because of the potentially high recurrence risk, and given the importance of early CoQ10 supplementation, we decided to treat with CoQ10 the newborn child pending the results of the biochemical assays. Clinicians should consider a similar management in siblings of patients with CoQ10 deficiency without a genetic diagnosis.


Assuntos
Alquil e Aril Transferases/genética , Ataxia/diagnóstico , Ataxia/genética , Mitocôndrias Musculares/genética , Doenças Mitocondriais/diagnóstico , Doenças Mitocondriais/genética , Debilidade Muscular/diagnóstico , Debilidade Muscular/genética , Mutação Puntual , Ubiquinona/análogos & derivados , Ubiquinona/deficiência , Acidose Láctica/sangue , Acidose Láctica/genética , Acidose Láctica/patologia , Alquil e Aril Transferases/deficiência , Ataxia/sangue , Ataxia/patologia , Consanguinidade , Evolução Fatal , Feminino , Expressão Gênica , Insuficiência Hepática/sangue , Insuficiência Hepática/genética , Insuficiência Hepática/patologia , Humanos , Recém-Nascido , Deficiência Intelectual/sangue , Deficiência Intelectual/genética , Deficiência Intelectual/patologia , Mitocôndrias Musculares/enzimologia , Mitocôndrias Musculares/patologia , Doenças Mitocondriais/sangue , Doenças Mitocondriais/patologia , Debilidade Muscular/sangue , Debilidade Muscular/patologia , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Proteinúria/sangue , Proteinúria/genética , Proteinúria/patologia , Aminoacidúrias Renais/sangue , Aminoacidúrias Renais/genética , Aminoacidúrias Renais/patologia , Análise de Sequência de DNA , Ubiquinona/sangue , Ubiquinona/genética
5.
J Neurol Sci ; 326(1-2): 24-8, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23343605

RESUMO

Coenzyme Q10 (ubiquinone or CoQ10) serves as a redox carrier in the mitochondrial oxidative phosphorylation system. The reduced form of this lipid-soluble antioxidant (ubiquinol) is involved in other metabolic processes as well, such as preventing reactive oxygen species (ROS) induced damage from the mitochondrial membrane. Primary coenzyme Q10 deficiency is a rare, autosomal recessive disorder, often presenting with neurological and/or muscle involvement. Until now, five patients from four families have been described with primary coenzyme Q10 deficiency due to mutations in COQ2 encoding para-hydroxybenzoate polyprenyl transferase. Interestingly, four of these patients showed a distinctive renal involvement (focal segmental glomerular sclerosis, crescentic glomerulonephritis, nephrotic syndrome), which is only very rarely seen in correlation with mitochondrial disorders. The fifth patient deceases due to infantile multi organ failure, also with renal involvement. Here we report a novel homozygous mutation in COQ2 (c.905C>T, p.Ala302Val) in a dizygotic twin from consanguineous Turkish parents. The children were born prematurely and died at the age of five and six months, respectively, after an undulating disease course involving apneas, seizures, feeding problems and generalized edema, alternating with relative stable periods without the need of artificial ventilation. There was no evidence for renal involvement. We would like to raise awareness for this potentially treatable disorder which could be under diagnosed in patients with fatal neonatal or infantile multi-organ disease.


Assuntos
Alquil e Aril Transferases/deficiência , Alquil e Aril Transferases/genética , Doenças em Gêmeos/genética , Doenças Metabólicas/genética , Insuficiência de Múltiplos Órgãos/genética , Mutação/genética , Sequência de Aminoácidos , Doenças em Gêmeos/diagnóstico , Doenças em Gêmeos/enzimologia , Evolução Fatal , Feminino , Homozigoto , Humanos , Lactente , Masculino , Doenças Metabólicas/diagnóstico , Doenças Metabólicas/enzimologia , Dados de Sequência Molecular , Insuficiência de Múltiplos Órgãos/diagnóstico , Insuficiência de Múltiplos Órgãos/enzimologia
6.
FASEB J ; 27(2): 612-21, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23150520

RESUMO

Primary human CoQ(10) deficiencies are clinically heterogeneous diseases caused by mutations in PDSS2 and other genes required for CoQ(10) biosynthesis. Our in vitro studies of PDSS2 mutant fibroblasts, with <20% CoQ(10) of control cells, revealed reduced activity of CoQ(10)-dependent complex II+III and ATP synthesis, without amplification of reactive oxygen species (ROS), markers of oxidative damage, or antioxidant defenses. In contrast, COQ2 and ADCK3 mutant fibroblasts, with 30-50% CoQ(10) of controls, showed milder bioenergetic defects but significantly increased ROS and oxidation of lipids and proteins. We hypothesized that absence of oxidative stress markers and cell death in PDSS2 mutant fibroblasts were due to the extreme severity of CoQ(10) deficiency. Here, we have investigated in vivo effects of Pdss2 deficiency in affected and unaffected organs of CBA/Pdss2(kd/kd) mice at presymptomatic, phenotypic-onset, and end-stages of the disease. Although Pdss2 mutant mice manifest widespread CoQ(9) deficiency and mitochondrial respiratory chain abnormalities, only affected organs show increased ROS production, oxidative stress, mitochondrial DNA depletion, and reduced citrate synthase activity, an index of mitochondrial mass. Our data indicate that kidney-specific loss of mitochondria triggered by oxidative stress may be the cause of renal failure in Pdss2(kd/kd) mice.


Assuntos
Alquil e Aril Transferases/deficiência , Alquil e Aril Transferases/genética , Mitocôndrias/metabolismo , Ubiquinona/deficiência , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Transporte de Elétrons , Fibroblastos/metabolismo , Humanos , Rim/metabolismo , Rim/patologia , Camundongos , Camundongos Endogâmicos CBA , Camundongos Mutantes , Estresse Oxidativo , Distribuição Tecidual
7.
Mitochondrion ; 13(5): 417-26, 2013 09.
Artigo em Inglês | MEDLINE | ID: mdl-23261681

RESUMO

This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This retraction was suggested by the University of Cologne Investigation committee and seconded by the authors who the journal was able to contact (Wenz, T., Dillon, L., Diaz, F., Hida, A., and Moraes, C.T.). Following an investigation of the last author, Dr. Tina Wenz, by the University of Cologne, Germany, the university determined that data presented in this article have been inappropriately manipulated https://www.portal.uni-koeln.de/9015.html?&tx_news_pi1%5Bnews%5D=4335&tx_news_pi1%5Bcontroller%5D=News&tx_news_pi1%5Baction%5D=detail&cHash=1deb8399d7f796d65ca9f6ae4764a1ce. Specifically, western blot images in Figure 5F (tubulin in cortex), 2F (COXI in hippocampus) and 3B (Sod2 in hippocampus) were re-used from an earlier article published elsewhere [Increased muscle PGC-1alpha expression protects from sarcopenia and metabolic disease during aging" Wenz T, Rossi SG, Rotundo RL, Spiegelman BM, and Moraes CT. Proc Natl Acad Sci U S A. 2009;106:20405-10, doi: 10.1073/pnas.0911570106] representing different experimental findings. Therefore, whether or not the main conclusions are still valid, the authors request retraction of this publication because the scientific integrity of the study was compromised. The authors sincerely apologize to the scientific community.


Assuntos
Bezafibrato/administração & dosagem , Hipolipemiantes/administração & dosagem , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , Encefalomiopatias Mitocondriais/patologia , Encefalomiopatias Mitocondriais/fisiopatologia , Fármacos Neuroprotetores/administração & dosagem , Trifosfato de Adenosina/metabolismo , Alquil e Aril Transferases/deficiência , Animais , Astrócitos/fisiologia , Proliferação de Células , Modelos Animais de Doenças , Proteínas de Membrana/deficiência , Camundongos , Proteínas Mitocondriais/metabolismo , Resultado do Tratamento
8.
Pediatr Nephrol ; 26(7): 1157-61, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21365190

RESUMO

Renal manifestations of mitochondrial cytopathies have been described, but nephrotic syndrome with respiratory-chain disorders have been described extremely rarely. We report a 9-month-old boy with a mitochondrial cytopathy preceded by a 2-month history of steroid-resistant nephrotic syndrome. Percutaneous renal biopsy revealed diffuse mesangial sclerosis, and mutational analysis was compatible with PLCE1 mutation. However, electron microscopic findings of renal tissue, sensorineural hearing loss, and other ocular and neurologic findings led us to suspect mitochondrial cytopathy. Muscle tissue analysis showed a deficiency of the respiratory chain complex IV. The clinical presentation of our patient is not typical for primary cytochrome oxidase (COX) deficiency but showed similarities with patients carrying AR mutations in COX10. This was the first case in the literature with both PLCE1 mutation and COX deficiency. We could not identify pathogenic mutations in the COX10 gene, suggesting that PLCE1 deficiency could be the cause of the secondary deficiency of COX. Another, more likely, possibility is that the mitochondriopathy phenotype is caused by another mutation homozygous by descent in a yet unidentified recessive gene.


Assuntos
Alquil e Aril Transferases/genética , Deficiência de Citocromo-c Oxidase/diagnóstico , Proteínas de Membrana/genética , Síndrome Nefrótica/diagnóstico , Fosfoinositídeo Fosfolipase C/genética , Esclerose/diagnóstico , Alquil e Aril Transferases/deficiência , Biópsia , Deficiência de Citocromo-c Oxidase/complicações , Deficiência de Citocromo-c Oxidase/enzimologia , Deficiência de Citocromo-c Oxidase/genética , Deficiência de Citocromo-c Oxidase/terapia , Análise Mutacional de DNA , Complexo IV da Cadeia de Transporte de Elétrons , Predisposição Genética para Doença , Humanos , Lactente , Masculino , Proteínas de Membrana/deficiência , Mutação , Síndrome Nefrótica/enzimologia , Síndrome Nefrótica/genética , Síndrome Nefrótica/terapia , Fenótipo , Esclerose/enzimologia , Esclerose/genética , Esclerose/terapia
9.
J Clin Invest ; 121(2): 510-3, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21266773

RESUMO

Proteins that end with a CAAX sequence are targeted to cellular membranes by a series of posttranslational modifications that include prenylation, proteolysis, and carboxyl methylation. Two prenyltransferases modify CAAX proteins: farnesyltransferase and geranylgeranyltransferase type I (GGTase-I). Rho family GTPases that control the actin cytoskeleton and are therefore critical to inflammatory cell function are substrates for GGTase-I. In this issue of the JCI, Khan et al. examined mice in which GGTase-I was conditionally deleted in macrophages. Rather than obtunded cells, the authors found activated Rho proteins in fully functional macrophages that hypersecreted inflammatory cytokines and induced an erosive, inflammatory arthritis. This surprising result calls into question the role of protein geranylgeranylation in inflammatory cell signaling.


Assuntos
Alquil e Aril Transferases/deficiência , Alquil e Aril Transferases/genética , Sequência de Aminoácidos , Animais , Farnesiltranstransferase/metabolismo , Macrófagos/enzimologia , Macrófagos/imunologia , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Prenilação , Transdução de Sinais/imunologia , Proteínas rho de Ligação ao GTP/metabolismo
10.
J Clin Invest ; 121(2): 628-39, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21266780

RESUMO

RHO family proteins are important for the function of inflammatory cells. They are modified with a 20-carbon geranylgeranyl lipid in a process catalyzed by protein geranylgeranyltransferase type I (GGTase-I). Geranylgeranylation is viewed as essential for the membrane targeting and activity of RHO proteins. Consequently, inhibiting GGTase-I to interfere with RHO protein activity has been proposed as a strategy to treat inflammatory disorders. However, here we show that mice lacking GGTase-I in macrophages develop severe joint inflammation resembling erosive rheumatoid arthritis. The disease was initiated by the GGTase-I-deficient macrophages and was transplantable and reversible in bone marrow transplantation experiments. The cells accumulated high levels of active GTP-bound RAC1, CDC42, and RHOA, and RAC1 remained associated with the plasma membrane. Moreover, GGTase-I deficiency activated p38 and NF-κB and increased the production of proinflammatory cytokines. The results challenge the view that geranylgeranylation is essential for the activity and localization of RHO family proteins and suggest that reduced geranylgeranylation in macrophages can initiate erosive arthritis.


Assuntos
Alquil e Aril Transferases/deficiência , Artrite/imunologia , Artrite/patologia , Macrófagos/imunologia , Alquil e Aril Transferases/genética , Animais , Citocinas/imunologia , Macrófagos/citologia , Macrófagos/enzimologia , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína cdc42 de Ligação ao GTP/genética , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteínas rap1 de Ligação ao GTP/genética , Proteínas rap1 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo
11.
J Appl Physiol (1985) ; 106(5): 1712-9, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19286571

RESUMO

Defects in the mitochondrial ATP-generating system are one of the most commonly inherited neurological disorders, but they remain without treatment. We have recently shown that modulation of the peroxisome proliferator-activated receptor-gamma coactivator-1alpha (PGC-1alpha) level in skeletal muscle of a mitochondrial myopathy mouse model offers a therapeutic approach. Here we analyzed if endurance exercise, which is known to be associated with an increased PGC-1alpha level in muscle, offers the same beneficial effect. We subjected male and female mice that develop a severe mitochondrial myopathy due to a cytochrome-c oxidase deficiency at 3 mo of age to endurance exercise training and monitored phenotypical and metabolic changes. Sedentary myopathy and wild-type mice were used as controls. Exercise increased PGC-1alpha in muscle, resulting in increased mitochondrial biogenesis, and successfully stimulated residual respiratory capacity in muscle tissue. As a consequence, ATP levels were increased in exercised mice compared with sedentary myopathy animals, which resulted in a delayed onset of the myopathy and a prolonged lifespan of the exercised mice. As an added benefit, endurance exercise induced antioxidant enzymes. The overall protective effect of endurance exercise delayed the onset of the mitochondrial myopathy and increased life expectancy in the mouse model. Thus stimulating residual oxidative phosphorylation function in the affected muscle by inducing mitochondrial biogenesis through endurance exercise might offer a valuable therapeutic intervention for mitochondrial myopathy patients.


Assuntos
Mitocôndrias Musculares/fisiologia , Miopatias Mitocondriais/prevenção & controle , Condicionamento Físico Animal/fisiologia , Trifosfato de Adenosina/metabolismo , Alquil e Aril Transferases/deficiência , Alquil e Aril Transferases/genética , Animais , Deficiência de Citocromo-c Oxidase/metabolismo , Modelos Animais de Doenças , Feminino , Regulação Enzimológica da Expressão Gênica , Longevidade , Masculino , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miopatias Mitocondriais/genética , Miopatias Mitocondriais/patologia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Oxirredutases/biossíntese , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Descanso/fisiologia , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição
12.
J Clin Invest ; 117(5): 1223-5, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17476354

RESUMO

Posttranslational modification is critical for the function of the gene products of ras oncogenes, which are frequently mutated in cancer. Ras proteins are modified by farnesyltransferase (FTase), but many related small GTPases that also end in a CAAX motif (where C is cysteine, A is often an aliphatic amino acid, and X is any amino acid) are modified by a closely related enzyme known as geranylgeranyltransferase type I (GGTase-I). Accordingly, inhibitors for both of these enzymes have been developed, and those active against FTase are in clinical trials. In this issue of the JCI, Sjogren et al. report the development of a mouse strain homozygous for a conditional allele of the gene that encodes GGTase-I (see the related article beginning on page 1294). They found that ablation of the GGTase-I-encoding gene in cells destined to produce lung tumors driven by oncogenic K-Ras resulted in delayed onset and decreased severity of disease, validating in a genetic model the theory that GGTase-I is a good target for anti-cancer drug development.


Assuntos
Alquil e Aril Transferases/metabolismo , Antineoplásicos/administração & dosagem , Sistemas de Liberação de Medicamentos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/enzimologia , Alquil e Aril Transferases/deficiência , Alquil e Aril Transferases/genética , Animais , Modelos Animais de Doenças , Humanos , Neoplasias Pulmonares/genética , Camundongos
13.
J Clin Invest ; 117(5): 1294-304, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17476360

RESUMO

Protein geranylgeranyltransferase type I (GGTase-I) is responsible for the posttranslational lipidation of CAAX proteins such as RHOA, RAC1, and cell division cycle 42 (CDC42). Inhibition of GGTase-I has been suggested as a strategy to treat cancer and a host of other diseases. Although several GGTase-I inhibitors (GGTIs) have been synthesized, they have very different properties, and the effects of GGTIs and GGTase-I deficiency are unclear. One concern is that inhibiting GGTase-I might lead to severe toxicity. In this study, we determined the effects of GGTase-I deficiency on cell viability and K-RAS-induced cancer development in mice. Inactivating the gene for the critical beta subunit of GGTase-I eliminated GGTase-I activity, disrupted the actin cytoskeleton, reduced cell migration, and blocked the proliferation of fibroblasts expressing oncogenic K-RAS. Moreover, the absence of GGTase-I activity reduced lung tumor formation, eliminated myeloproliferative phenotypes, and increased survival of mice in which expression of oncogenic K-RAS was switched on in lung cells and myeloid cells. Interestingly, several cell types remained viable in the absence of GGTase-I, and myelopoiesis appeared to function normally. These findings suggest that inhibiting GGTase-I may be a useful strategy to treat K-RAS-induced malignancies.


Assuntos
Alquil e Aril Transferases/deficiência , Alquil e Aril Transferases/genética , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/prevenção & controle , Sobrevida/fisiologia , Proteínas ras/toxicidade , Alquil e Aril Transferases/antagonistas & inibidores , Animais , Inativação Gênica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidade , Camundongos
14.
Biochem J ; 382(Pt 2): 519-26, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15153069

RESUMO

The COQ2 gene in Saccharomyces cerevisiae encodes a Coq2 (p-hydroxybenzoate:polyprenyl transferase), which is required in the biosynthetic pathway of CoQ (ubiquinone). This enzyme catalyses the prenylation of p-hydroxybenzoate with an all-trans polyprenyl group. We have isolated cDNA which we believe encodes the human homologue of COQ2 from a human muscle and liver cDNA library. The clone contained an open reading frame of length 1263 bp, which encodes a polypeptide that has sequence homology with the Coq2 homologues in yeast, bacteria and mammals. The human COQ2 gene, when expressed in yeast Coq2 null mutant cells, rescued the growth of this yeast strain in the absence of a non-fermentable carbon source and restored CoQ biosynthesis. However, the rate of CoQ biosynthesis in the rescued cells was lower when compared with that in cells rescued with the yeast COQ2 gene. CoQ formed when cells were incubated with labelled decaprenyl pyrophosphate and nonaprenyl pyrophosphate, showing that the human enzyme is active and that it participates in the biosynthesis of CoQ.


Assuntos
Alquil e Aril Transferases/genética , Regulação Enzimológica da Expressão Gênica/genética , Ubiquinona/biossíntese , Alquil e Aril Transferases/deficiência , Alquil e Aril Transferases/metabolismo , Sequência de Aminoácidos/genética , Sequência de Bases/genética , Clonagem Molecular/métodos , Teste de Complementação Genética/métodos , Humanos , Fígado/química , Fígado/metabolismo , Proteínas Mitocondriais/genética , Dados de Sequência Molecular , Músculo Esquelético/química , Músculo Esquelético/metabolismo , Mutação/genética , Especificidade de Órgãos/genética , Valor Preditivo dos Testes , Sinais Direcionadores de Proteínas/genética , RNA Mensageiro/genética , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crescimento & desenvolvimento , Proteínas de Saccharomyces cerevisiae/genética , Análise de Sequência de DNA/métodos
15.
J Biol Chem ; 278(45): 44708-18, 2003 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-12944391

RESUMO

Ether phospholipids are major components of the membranes of humans and Leishmania. In protozoan parasites they occur separately or as part of the glycosylphosphatidylinositol (GPI) anchor of molecules implicated in virulence, such as lipophosphoglycan (LPG), smaller glycosylinositolphospholipids (GIPLs), and GPI-anchored proteins. We generated null mutants of the Leishmania major alkyldihydroxyacetonephosphate synthase (ADS), the first committed step of ether lipid synthesis. Enzymatic analysis and comprehensive mass spectrometric analysis showed that ads1- knock-outs lacked all ether phospholipids, including plasmalogens, LPG, and GIPLs. Leishmania ads1- thus represents the first ether lipid-synthesizing eukaryote for which a completely null mutant could be obtained. Remarkably ads1- grew well and maintained lipid rafts (detergent-resistant membranes). In virulence tests it closely resembled LPG-deficient L. major, including sensitivity to complement and an inability to survive the initial phase of macrophage infection. Likewise it retained the ability to inhibit host cell signaling and to form infectious amastigotes from the few parasites surviving the establishment defect. These findings counter current proposals that GIPLs are required for amastigote survival in the mammalian host or that parasite lyso-alkyl or alkylacyl-GPI anchors are solely responsible for inhibition of macrophage activation.


Assuntos
Glicosilfosfatidilinositóis/fisiologia , Leishmania major/patogenicidade , Ativação de Macrófagos/fisiologia , Éteres Fosfolipídicos , Alquil e Aril Transferases/deficiência , Alquil e Aril Transferases/genética , Alquil e Aril Transferases/fisiologia , Animais , Glicoesfingolipídeos/deficiência , Glicoesfingolipídeos/fisiologia , Glicosilfosfatidilinositóis/biossíntese , Glicosilfosfatidilinositóis/genética , Leishmania major/genética , Leishmania major/fisiologia , Macrófagos/parasitologia , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica , Microscopia de Fluorescência , Dados de Sequência Molecular , Mutagênese , Éteres Fosfolipídicos/análise , Espectrometria de Massas por Ionização por Electrospray
16.
J Leukoc Biol ; 69(4): 583-9, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11310844

RESUMO

Salmonella typhimurium (ST) can cause infection in man, and attenuated strains are under consideration as live vaccine vectors. However, little is known about the interaction of ST with human dendritic cells (DC). Here, we compared the consequences of exposure of human, monocyte-derived DC with different attenuated strains of ST. Infection was observed with all four strains tested (wild type, PhoP-, PhoPc, and AroA), but the PhoPc strain was by far the most efficient. Intracellular persistence of wild type and PhoP- was longer than that of PhoPc and AroA, both of which were largely eliminated within 24 h. Most DC survived infection by the attenuated strains, although apoptosis was observed in a fraction of the exposed cells. All strains induced DC maturation, independent from the extent of infection. Although all strains stimulated secretion of TNF-alpha and IL-12 strongly, PhoPc induced significantly less IL-10 than the other three strains and as much as 10 times less IL-10 than heat-killed PhoPc, suggesting that this mutant suppressed the secretion of IL-10 by the DC. These data indicate that infectivity, bacterial elimination, and cytokine secretion in human DC are controlled by the genetic background of ST.


Assuntos
Alquil e Aril Transferases/fisiologia , Proteínas de Bactérias/fisiologia , Citocinas/metabolismo , Células Dendríticas/microbiologia , Salmonella typhimurium/patogenicidade , 3-Fosfoshikimato 1-Carboxiviniltransferase , Alquil e Aril Transferases/deficiência , Alquil e Aril Transferases/genética , Apoptose , Proteínas de Bactérias/genética , Diferenciação Celular , Células Cultivadas , Células Dendríticas/metabolismo , Genes Bacterianos , Humanos , Interleucinas/metabolismo , Monócitos/citologia , Necrose , Salmonella typhimurium/genética , Fator de Necrose Tumoral alfa/metabolismo , Virulência/genética
17.
Hum Mol Genet ; 10(2): 127-36, 2001 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-11152660

RESUMO

The first steps of ether lipid biosynthesis are exclusively localized to peroxisomes and hence some peroxisomal disorders are characterized by a severe deficiency of plasmalogens, the main ether lipids in humans. Here we report on gene defects of plasmalogen biosynthesis, chromosomal localization of the corresponding genes and, as a consequence of plasmalogen deficiency, on structural alterations of caveolae, clathrin-coated pits, endoplasmic reticulum and Golgi cisternae, as well as on the reduced rate of transferrin receptor cycling. The data suggest that plasmalogens, analogous to cholesterol, are essential for correct membrane functioning and their deficiency results in impaired membrane trafficking.


Assuntos
Aciltransferases/genética , Alquil e Aril Transferases/genética , Transtornos Peroxissômicos/genética , Éteres Fosfolipídicos/metabolismo , Aciltransferases/metabolismo , Alquil e Aril Transferases/deficiência , Alquil e Aril Transferases/metabolismo , Animais , Cavéolas/ultraestrutura , Células Cultivadas , Mapeamento Cromossômico , Vesículas Revestidas por Clatrina/ultraestrutura , Endocitose , Retículo Endoplasmático/ultraestrutura , Feminino , Fibroblastos/metabolismo , Fibroblastos/ultraestrutura , Complexo de Golgi/ultraestrutura , Humanos , Masculino , Camundongos , Mutação , Transtornos Peroxissômicos/metabolismo , Fenótipo , Plasmalogênios/biossíntese , Transporte Proteico , Receptores da Transferrina/metabolismo , Pele/citologia
18.
J Lipid Res ; 40(11): 1998-2003, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10553003

RESUMO

Recent studies have indicated that two peroxisomal enzymes involved in ether lipid synthesis, i.e., dihydroxyacetonephosphate acyltransferase and alkyl-dihydroxyacetonephosphate synthase, are directed to peroxisomes by different targeting signals, i.e., peroxisomal targeting signal type 1 and type 2, respectively. In this study, we describe a new human fibroblast cell line in which alkyl-dihydroxyacetonephosphate synthase was found to be deficient both at the level of enzyme activity and enzyme protein. At the cDNA level, a 128 base pair deletion was found leading to a premature stop. Remarkably, dihydroxyacetonephosphate acyltransferase activity was strongly reduced to a level comparable to the activities measured in fibroblasts from patients affected by the classical form of rhizomelic chondrodysplasia punctata (caused by a defect in peroxisomal targeting signal type 2 import). Dihydroxyacetonephosphate acyltransferase activity was completely normal in another alkyl-dihydroxyacetonephosphate synthase activity-deficient patient. Fibroblasts from this patient showed normal levels of the synthase protein and inactivity results from a point mutation leading to an amino acid substitution. These results strongly suggest that the activity of dihydroxyacetonephosphate acyltransferase is dependent on the presence of alkyl-dihydroxyacetonephosphate synthase protein. This interpretation implies that the deficiency of dihydroxyacetonephosphate acyltransferase (targeted by a peroxisomal targeting signal type 1) in the classic form of rhizomelic chondrodysplasia punctata is a consequence of the absence of the alkyl-dihydroxyacetonephosphate synthase protein (targeted by a peroxisomal targeting signal type 2).


Assuntos
Éteres Fosfolipídicos/metabolismo , Plasmalogênios/biossíntese , Aciltransferases/metabolismo , Adulto , Alquil e Aril Transferases/deficiência , Pré-Escolar , Condrodisplasia Punctata Rizomélica/enzimologia , Condrodisplasia Punctata Rizomélica/patologia , Feminino , Humanos , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA