Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
Nutrients ; 11(7)2019 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-31340540

RESUMO

Selenium, an essential trace element known mainly for its antioxidant properties, is critical for proper brain function and regulation of energy metabolism. Whole-body knockout of the selenium recycling enzyme, selenocysteine lyase (Scly), increases susceptibility to metabolic syndrome and diet-induced obesity in mice. Scly knockout mice also have decreased selenoprotein expression levels in the hypothalamus, a key regulator of energy homeostasis. This study investigated the role of selenium in whole-body metabolism regulation using a mouse model with hypothalamic knockout of Scly. Agouti-related peptide (Agrp) promoter-driven Scly knockout resulted in reduced weight gain and adiposity while on a high-fat diet (HFD). Scly-Agrp knockout mice had reduced Agrp expression in the hypothalamus, as measured by Western blot and immunohistochemistry (IHC). IHC also revealed that while control mice developed HFD-induced leptin resistance in the arcuate nucleus, Scly-Agrp knockout mice maintained leptin sensitivity. Brown adipose tissue from Scly-Agrp knockout mice had reduced lipid deposition and increased expression of the thermogenic marker uncoupled protein-1. This study sheds light on the important role of selenium utilization in energy homeostasis, provides new information on the interplay between the central nervous system and whole-body metabolism, and may help identify key targets of interest for therapeutic treatment of metabolic disorders.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Dieta Hiperlipídica , Hipotálamo/enzimologia , Leptina/metabolismo , Liases/deficiência , Neurônios/metabolismo , Obesidade/prevenção & controle , Tecido Adiposo Marrom/enzimologia , Tecido Adiposo Marrom/fisiopatologia , Adiposidade , Animais , Modelos Animais de Doenças , Feminino , Técnicas de Inativação de Genes , Hipotálamo/fisiopatologia , Liases/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/enzimologia , Obesidade/genética , Obesidade/fisiopatologia , Transdução de Sinais , Proteína Desacopladora 1/metabolismo , Aumento de Peso
2.
Nutrients ; 10(2)2018 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-29385050

RESUMO

Selenium (Se) is an essential micronutrient known for its antioxidant properties and health benefits, attributed to its presence in selenoproteins as the amino acid, selenocysteine. Selenocysteine lyase (Scly) catalyzes hydrolysis of selenocysteine to selenide and alanine, facilitating re-utilization of Se for de novo selenoprotein synthesis. Previously, it was reported that male Scly-/- mice develop increased body weight and body fat composition, and altered lipid and carbohydrate metabolism, compared to wild type mice. Strikingly, females appeared to present with a less severe phenotype, suggesting the relationship between Scly and energy metabolism may be regulated in a sex-specific manner. Here, we report that while body weight and body fat gain occur in both male and female Scly-/- mice, strikingly, males are susceptible to developing glucose intolerance, whereas female Scly-/- mice are protected. Because Se is critical for male reproduction, we hypothesized that castration would attenuate the metabolic dysfunction observed in male Scly-/- mice by eliminating sequestration of Se in testes. We report that fasting serum insulin levels were significantly reduced in castrated males compared to controls, but islet area was unchanged between groups. Finally, both male and female Scly-/- mice exhibit reduced hypothalamic expression of selenoproteins S, M, and glutathione peroxidase 1.


Assuntos
Metabolismo Energético , Liases/deficiência , Caracteres Sexuais , Adiposidade , Fatores Etários , Animais , Glicemia/metabolismo , Metabolismo Energético/genética , Feminino , Genótipo , Intolerância à Glucose/sangue , Intolerância à Glucose/enzimologia , Intolerância à Glucose/genética , Glutationa Peroxidase/metabolismo , Hipotálamo/enzimologia , Insulina/sangue , Liases/genética , Masculino , Camundongos Knockout , Orquiectomia , Fenótipo , Selenoproteínas/metabolismo , Fatores de Tempo , Aumento de Peso , Glutationa Peroxidase GPX1
3.
J Am Heart Assoc ; 6(8)2017 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-28778941

RESUMO

BACKGROUND: Fetal growth impacts cardiovascular health throughout postnatal life in humans. Various animal models of intrauterine growth restriction exhibit reduced heart size at birth, which negatively influences cardiac function in adulthood. The mechanistic target of rapamycin complex 1 (mTORC1) integrates nutrient and growth factor availability with cell growth, thereby regulating organ size. This study aimed at elucidating a possible involvement of mTORC1 in intrauterine growth restriction and prenatal heart growth. METHODS AND RESULTS: We inhibited mTORC1 in fetal mice by rapamycin treatment of pregnant dams in late gestation. Prenatal rapamycin treatment reduces mTORC1 activity in various organs at birth, which is fully restored by postnatal day 3. Rapamycin-treated neonates exhibit a 16% reduction in body weight compared with vehicle-treated controls. Heart weight decreases by 35%, resulting in a significantly reduced heart weight/body weight ratio, smaller left ventricular dimensions, and reduced cardiac output in rapamycin- versus vehicle-treated mice at birth. Although proliferation rates in neonatal rapamycin-treated hearts are unaffected, cardiomyocyte size is reduced, and apoptosis increased compared with vehicle-treated neonates. Rapamycin-treated mice exhibit postnatal catch-up growth, but body weight and left ventricular mass remain reduced in adulthood. Prenatal mTORC1 inhibition causes a reduction in cardiomyocyte number in adult hearts compared with controls, which is partially compensated for by an increased cardiomyocyte volume, resulting in normal cardiac function without maladaptive left ventricular remodeling. CONCLUSIONS: Prenatal rapamycin treatment of pregnant dams represents a new mouse model of intrauterine growth restriction and identifies an important role of mTORC1 in perinatal cardiac growth.


Assuntos
Retardo do Crescimento Fetal/induzido quimicamente , Coração Fetal/efeitos dos fármacos , Alvo Mecanístico do Complexo 1 de Rapamicina/antagonistas & inibidores , Efeitos Tardios da Exposição Pré-Natal , Sirolimo/farmacologia , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Débito Cardíaco/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Feminino , Retardo do Crescimento Fetal/metabolismo , Retardo do Crescimento Fetal/patologia , Retardo do Crescimento Fetal/fisiopatologia , Coração Fetal/crescimento & desenvolvimento , Coração Fetal/patologia , Idade Gestacional , Proteína Homeobox Nkx-2.5/genética , Liases/deficiência , Liases/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Tamanho do Órgão , Organogênese/efeitos dos fármacos , Gravidez , Regiões Promotoras Genéticas , Função Ventricular Esquerda/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos
4.
J Mol Cell Cardiol ; 97: 1-14, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27106802

RESUMO

Whereas adult cardiomyocytes are highly susceptible to stress, cardiomyocytes in the prenatal heart appear to be rather resistant. To investigate how embryonic cardiomyocytes respond to metabolic stress in vivo, we utilized tissue mosaicism for mitochondrial dysfunction in 13.5dpc mouse hearts. The latter is based on inactivation of the X-linked gene encoding Holocytochrome c synthase (Hccs), which is essential for mitochondrial respiration. In heterozygous heart conditional Hccs knockout females (cHccs(+/-)) random X chromosomal inactivation results in a mosaic of healthy and HCCS deficient cells in the myocardium. Microarray RNA expression analyses identified genes involved in unfolded protein response (UPR) and programmed cell death as differentially expressed in cHccs(+/-) versus control embryonic hearts. Activation of the UPR is localized to HCCS deficient cardiomyocytes but does not involve ER stress pathways, suggesting that it is caused by defective mitochondria. Consistently, mitochondrial chaperones, such as HSP10 and HSP60, but not ER chaperones are induced in defective cells. Mitochondrial dysfunction can result in oxidative stress, but no evidence for excessive ROS (reactive oxygen species) production was observed in cHccs(+/-) hearts. Instead, the antioxidative proteins SOD2 and PRDX3 are induced, suggesting that ROS detoxification prevents oxidative damage in HCCS deficient cardiomyocytes. Mitochondrial dysfunction and unrestricted UPR can induce cell death, and we detected the initiation of upstream events of both intrinsic as well as extrinsic apoptosis in cHccs(+/-) hearts. Cell death is not executed, however, suggesting the activation of antiapoptotic mechanisms. Whereas most apoptosis inhibitors are either unchanged or downregulated in HCCS deficient cardiomyocytes, Bcl-2 and ARC (apoptosis repressor with caspase recruitment domain) are induced. Given that ARC can inhibit both apoptotic pathways as well as necrosis and attenuates UPR, we generated cHccs(+/-) embryos on an Arc knockout background (cHccs(+/-),Arc(-/-)). Surprisingly, the absence of ARC does not induce cell death in embryonic or postnatal HCCS deficient cardiomyocytes and adult cHccs(+/-),Arc(-/-) mice exhibit normal cardiac morphology and function. Taken together, our data demonstrate an impressive plasticity of embryonic cardiomyocytes to respond to metabolic stress, the loss of which might be involved in the high susceptibility of postnatal cardiomyocytes to cell death.


Assuntos
Mitocôndrias/metabolismo , Miócitos Cardíacos/metabolismo , Estresse Oxidativo , Animais , Antioxidantes/metabolismo , Apoptose/genética , Autofagia , Sobrevivência Celular/genética , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Feminino , Perfilação da Expressão Gênica , Genótipo , Coração/embriologia , Liases/deficiência , Liases/genética , Liases/metabolismo , Masculino , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Miocárdio/metabolismo , NF-kappa B/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Estresse Oxidativo/genética , Espécies Reativas de Oxigênio/metabolismo , Regeneração/genética , Transdução de Sinais , Resposta a Proteínas não Dobradas
5.
Tsitologiia ; 57(5): 353-9, 2015.
Artigo em Russo | MEDLINE | ID: mdl-26281212

RESUMO

The structural organization of cells of the Brc-1 mutant of the unicellular green algae Chlamydomonas reinhardtii grown in the light and in the dark has been studied. The Brc-1 mutant contains the brc-1 mutation in the nucleus gene LTS3. In the light, all membrane structures in mutant cells form normally and are well developed. In the dark under heterotrophic conditions, the mutant cells grew and divided well, however, all its cell membranes: plasmalemma, tonoplast, mitochondrial membranes, membranes of the nucleus shell and chloroplast, thylakoids, and the membranes of dictiosomes of the Golgi apparatus were not detected. In the dark under heterotrophic conditions, mutant cells well grow and divide. It were shown that a short-term (1-10 min) exposure of Brc-1 mutant cells to light leads to the restoration of all above-mentioned membrane structures. Possible reasons for the alterations of membrane structures are discussed.


Assuntos
Proteínas de Algas/genética , Membrana Celular/metabolismo , Chlamydomonas reinhardtii/metabolismo , Liases/genética , Fotossíntese/efeitos da radiação , Tilacoides/metabolismo , Proteínas de Algas/metabolismo , Divisão Celular , Membrana Celular/efeitos da radiação , Membrana Celular/ultraestrutura , Chlamydomonas reinhardtii/efeitos da radiação , Chlamydomonas reinhardtii/ultraestrutura , Clorofila/agonistas , Clorofila/metabolismo , Expressão Gênica , Complexo de Golgi/metabolismo , Complexo de Golgi/efeitos da radiação , Complexo de Golgi/ultraestrutura , Luz , Liases/deficiência , Microscopia Eletrônica , Mitocôndrias/metabolismo , Mitocôndrias/efeitos da radiação , Mitocôndrias/ultraestrutura , Mutação , Fotoperíodo , Fotossíntese/fisiologia , Tilacoides/efeitos da radiação , Tilacoides/ultraestrutura
6.
Cardiovasc Res ; 106(1): 43-54, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25661081

RESUMO

AIMS: Foetal growth has been proposed to influence cardiovascular health in adulthood, a process referred to as foetal programming. Indeed, intrauterine growth restriction in animal models alters heart size and cardiomyocyte number in the perinatal period, yet the consequences for the adult or challenged heart are largely unknown. The aim of this study was to elucidate postnatal myocardial growth pattern, left ventricular function, and stress response in the adult heart after neonatal cardiac hypoplasia in mice. METHODS AND RESULTS: Utilizing a new mouse model of impaired cardiac development leading to fully functional but hypoplastic hearts at birth, we show that myocardial mass is normalized until early adulthood by accelerated physiological cardiomyocyte hypertrophy. Compensatory hypertrophy, however, cannot be maintained upon ageing, resulting in reduced organ size without maladaptive myocardial remodelling. Angiotensin II stress revealed aberrant cardiomyocyte growth kinetics in adult hearts after neonatal hypoplasia compared with normally developed controls, characterized by reversible overshooting hypertrophy. This exaggerated growth mainly depends on STAT3, whose inhibition during angiotensin II treatment reduces left ventricular mass in both groups but causes contractile dysfunction in developmentally impaired hearts only. Whereas JAK/STAT3 inhibition reduces cardiomyocyte cross-sectional area in the latter, it prevents fibrosis in control hearts, indicating fundamentally different mechanisms of action. CONCLUSION: Impaired prenatal development leading to neonatal cardiac hypoplasia alters postnatal cardiac growth and stress response in vivo, thereby linking foetal programming to organ size control in the heart.


Assuntos
Animais Recém-Nascidos/crescimento & desenvolvimento , Desenvolvimento Embrionário/fisiologia , Desenvolvimento Fetal/fisiologia , Coração/embriologia , Coração/fisiopatologia , Estresse Fisiológico/fisiologia , Envelhecimento/fisiologia , Angiotensina II/farmacologia , Animais , Feminino , Coração/efeitos dos fármacos , Hipertrofia , Liases/deficiência , Liases/genética , Liases/fisiologia , Camundongos , Camundongos Knockout , Modelos Animais , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Tamanho do Órgão/fisiologia , Fator de Transcrição STAT3/fisiologia
7.
PLoS One ; 9(6): e99564, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24932524

RESUMO

Leaf-color is an effective marker to identify the hybridization of rice. Leaf-color related genes function in chloroplast development and the photosynthetic pigment biosynthesis of higher plants. The ygl7 (yellow-green leaf 7) is a mutant with spontaneous yellow-green leaf phenotype across the whole lifespan but with no change to its yield traits. We cloned gene Ygl7 (Os03g59640) which encodes a magnesium-chelatase ChlD protein. Expression of ygl7 turns green-leaves to yellow, whereas RNAi-mediated silence of Ygl7 causes a lethal phenotype of the transgenic plants. This indicates the importance of the gene for rice plant. On the other hand, it corroborates that ygl7 is a non-null mutants. The content of photosynthetic pigment is lower in Ygl7 than the wild type, but its light efficiency was comparatively high. All these results indicated that the mutational YGL7 protein does not cause a complete loss of original function but instead acts as a new protein performing a new function. This new function partially includes its preceding function and possesses an additional feature to promote photosynthesis. Chl1, Ygl98, and Ygl3 are three alleles of the OsChlD gene that have been documented previously. However, mutational sites of OsChlD mutant gene and their encoded protein products were different in the three mutants. The three mutants have suppressed grain output. In our experiment, plant materials of three mutants (ygl7, chl1, and ygl98) all exhibited mutational leaf-color during the whole growth period. This result was somewhat different from previous studies. We used ygl7 as female crossed with chl1 and ygl98, respectively. Both the F1 and F2 generation display yellow-green leaf phenotype with their chlorophyll and carotenoid content falling between the values of their parents. Moreover, we noted an important phenomenon: ygl7-NIL's leaf-color is yellow, not yellowy-green, and this is also true of all back-crossed offspring with ygl7.


Assuntos
Genes de Plantas , Liases/genética , Mutação de Sentido Incorreto , Oryza/genética , Fotossíntese/genética , Folhas de Planta/metabolismo , Proteínas de Plantas/genética , Mutação Puntual , Substituição de Aminoácidos , Carotenoides/análise , Clorofila/análise , Clorofila/biossíntese , Mapeamento Cromossômico , Cromossomos de Plantas/genética , Cor , Cruzamentos Genéticos , Éxons/genética , Técnicas de Silenciamento de Genes , Genes Letais , Genes Recessivos , Teste de Complementação Genética , Endogamia , Liases/química , Liases/deficiência , Liases/fisiologia , Oryza/metabolismo , Proteínas de Plantas/fisiologia , Plantas Geneticamente Modificadas , Estrutura Terciária de Proteína , Subunidades Proteicas , RNA Interferente Pequeno/farmacologia , Alinhamento de Sequência
8.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 30(4): 439-42, 2013 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-23926012

RESUMO

OBJECTIVE: To analyze CYP17A1 gene mutations in a child patient with 17 alpha-hydroxylase/17, 20-lyase deficiency (17OHD), and to review characteristics of CYP17A1 gene mutations in Chinese patients with 17OHD. METHODS: Clinical data were collected. PCR and DNA sequencing were performed to detect mutations in the patient. RESULTS: The patient has presented classical features of 17OHD including hypertension, hypokalemia, decreased sex hormones and plasma cortisol, and elevated blood adrenocorticotrophic hormone. A compound heterozygous mutation c.987C>A and c.985del was detected in the CYP17A1 gene, which resulted in two premature stop codons at positions 328 and 417. CONCLUSION: A compound mutation, c.987C>A and c.985del, has been identified in a patient with 17OHD. Among CYP17A1 gene mutations identified in Chinese patients, missence mutations have been most common, and exons 5 and 8 have been the mutation hotspots.


Assuntos
Hiperplasia Suprarrenal Congênita/enzimologia , Liases/genética , Mutação , Esteroide 17-alfa-Hidroxilase/genética , Adolescente , Hiperplasia Suprarrenal Congênita/genética , Sequência de Bases , Feminino , Humanos , Liases/deficiência , Dados de Sequência Molecular
9.
Circ Res ; 111(1): 37-49, 2012 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-22628578

RESUMO

RATIONALE: Hyperhomocysteinemia (HHcy) accelerates atherosclerosis and increases inflammatory monocytes (MC) in peripheral tissues. However, its causative role in atherosclerosis is not well established and its effect on vascular inflammation has not been studied. The underlying mechanism is unknown. OBJECTIVE: This study examined the causative role of HHcy in atherogenesis and its effect on inflammatory MC differentiation. METHODS AND RESULTS: We generated a novel HHcy and hyperlipidemia mouse model, in which cystathionine ß-synthase (CBS) and low-density lipoprotein receptor (LDLr) genes were deficient (Ldlr(-/-) Cbs(-/+)). Severe HHcy (plasma homocysteine (Hcy)=275 µmol/L) was induced by a high methionine diet containing sufficient basal levels of B vitamins. Plasma Hcy levels were lowered to 46 µmol/L from 244 µmol/L by vitamin supplementation, which elevated plasma folate levels. Bone marrow (BM)-derived cells were traced by the transplantation of BM cells from enhanced green fluorescent protein (EGFP) transgenic mice after sublethal irradiation of the recipient. HHcy accelerated atherosclerosis and promoted Ly6C(high) inflammatory MC differentiation of both BM and tissue origins in the aortas and peripheral tissues. It also elevated plasma levels of TNF-α, IL-6, and MCP-1; increased vessel wall MC accumulation; and increased macrophage maturation. Hcy-lowering therapy reversed HHcy-induced lesion formation, plasma cytokine increase, and blood and vessel inflammatory MC (Ly6C(high+middle)) accumulation. Plasma Hcy levels were positively correlated with plasma levels of proinflammatory cytokines. In primary mouse splenocytes, L-Hcy promoted rIFNγ-induced inflammatory MC differentiation, as well as increased TNF-α, IL-6, and superoxide anion production in inflammatory MC subsets. Antioxidants and folic acid reversed L-Hcy-induced inflammatory MC differentiation and oxidative stress in inflammatory MC subsets. CONCLUSIONS: HHcy causes vessel wall inflammatory MC differentiation and macrophage maturation of both BM and tissue origins, leading to atherosclerosis via an oxidative stress-related mechanism.


Assuntos
Aorta/enzimologia , Aterosclerose/etiologia , Células da Medula Óssea/enzimologia , Diferenciação Celular , Hiper-Homocisteinemia/complicações , Inflamação/etiologia , Liases/deficiência , Macrófagos/enzimologia , Receptores de LDL/deficiência , Animais , Antioxidantes/farmacologia , Aorta/efeitos dos fármacos , Aorta/imunologia , Aorta/patologia , Aterosclerose/sangue , Aterosclerose/enzimologia , Aterosclerose/genética , Aterosclerose/patologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/imunologia , Transplante de Medula Óssea , Células Cultivadas , Quimiocina CCL2/sangue , Modelos Animais de Doenças , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Hiper-Homocisteinemia/sangue , Hiper-Homocisteinemia/enzimologia , Hiper-Homocisteinemia/genética , Hiper-Homocisteinemia/imunologia , Hiperlipidemias/complicações , Hiperlipidemias/enzimologia , Hiperlipidemias/imunologia , Inflamação/sangue , Inflamação/enzimologia , Inflamação/imunologia , Mediadores da Inflamação/sangue , Interleucina-6/sangue , Lipídeos/sangue , Liases/genética , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Estresse Oxidativo , Receptores de LDL/genética , Índice de Gravidade de Doença , Superóxidos/metabolismo , Fator de Necrose Tumoral alfa/sangue , Complexo Vitamínico B/farmacologia
10.
Plant Cell ; 18(12): 3606-16, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17158606

RESUMO

Mg-chelatase catalyzes the insertion of Mg2+ into protoporphyrin IX at the first committed step of the chlorophyll biosynthetic pathway. It consists of three subunits: I, D, and H. The I subunit belongs to the AAA protein superfamily (ATPases associated with various cellular activities) that is known to form hexameric ring structures in an ATP-dependant fashion. Dominant mutations in the I subunit revealed that it functions in a cooperative manner. We demonstrated that the D subunit forms ATP-independent oligomeric structures and should also be classified as an AAA protein. Furthermore, we addressed the question of cooperativity of the D subunit with barley (Hordeum vulgare) mutant analyses. The recessive behavior in vivo was explained by the absence of mutant proteins in the barley cell. Analogous mutations in Rhodobacter capsulatus and the resulting D proteins were studied in vitro. Mixtures of wild-type and mutant R. capsulatus D subunits showed a lower activity compared with wild-type subunits alone. Thus, the mutant D subunits displayed dominant behavior in vitro, revealing cooperativity between the D subunits in the oligomeric state. We propose a model where the D oligomer forms a platform for the stepwise assembly of the I subunits. The cooperative behavior suggests that the D oligomer takes an active part in the conformational dynamics between the subunits of the enzyme.


Assuntos
Clorofila/biossíntese , Genes Dominantes , Genes Recessivos , Hordeum/enzimologia , Hordeum/genética , Liases/deficiência , Metaloendopeptidases/metabolismo , Sequência de Aminoácidos , Catálise , Regulação Enzimológica da Expressão Gênica , Regulação da Expressão Gênica de Plantas , Liases/química , Liases/genética , Liases/metabolismo , Modelos Biológicos , Dados de Sequência Molecular , Mutação/genética , Fenótipo , Folhas de Planta/enzimologia , Proteínas de Plantas/química , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Dobramento de Proteína , Estrutura Quaternária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/deficiência , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Rhodobacter capsulatus/ultraestrutura , Plântula/enzimologia
11.
Fertil Steril ; 83(5): 1548-51, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15866602

RESUMO

OBJECTIVE: To investigate the cause of hypergonadotropic hypogonadism. DESIGN: Case report and literature review. SETTING: University Departments of Pediatric Endocrinology and Obstetrics and Gynecology. PATIENT(S): A 13.5-year-old girl with absent puberty and growth retardation. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): Detailed biochemical, radiological, and molecular analysis, including pelvic ultrasound, basal steroid hormone analysis in serum and aspirated follicle fluid, serum steroid measurement after ACTH (Synachten) and human chorionic gonadotropin (hCG) stimulation, and molecular analysis of CYP17. RESULT(S): This girl with hypergonadotropic hypogonadism (LH 65 U/L, FSH 50 U/L) had a 46,XX karyotype, small uterus and enlarged cystic ovaries, and markedly delayed bone age (9 years). Basal (serum, follicular) and stimulated (serum) steroid hormone levels were consistent with isolated 17,20-lyase deficiency whereas relatively normal P and 17-hydroxyprogesterone concentrations were detected together with very low androstenedione, T, and E(2) levels. CONCLUSION(S): Isolated 17,20-lyase deficiency should be considered in the differential diagnosis of hypergonadotropic hypogonadism in 46,XX females, and follicular fluid steroid analysis is a useful adjuvant test. Failure to detect mutations in CYP17 raises the possibility of a novel association of these phenotypes.


Assuntos
Cromossomos Humanos X/genética , Liases/deficiência , Liases/genética , Puberdade Tardia/enzimologia , Puberdade Tardia/genética , Adolescente , Transtornos do Desenvolvimento Sexual/enzimologia , Transtornos do Desenvolvimento Sexual/genética , Feminino , Humanos
12.
Res Microbiol ; 156(4): 465-71, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15862443

RESUMO

The csdA, csdB and iscS genes encoding for cysteine desulfurase enzymatic activities in Escherichia coli were independently inactivated and potassium tellurite sensitivity, determined for each of the resulting mutant clones, was found to be iscS > csdB > csdA. Structural genes encoding for each of the wild-type cysteine desulfurases were cloned into a vector containing the regulated ara promoter and further introduced into the mutant strains. Desulfurase-deficient cells transformed with homolog or paralog desulfurase genes and grown in arabinose-amended media restored their basal tellurite resistance. While csdB gene complemented the auxotrophy of csdB and iscS mutants for nicotinic acid, the iscS gene only complemented the auxotrophy of iscS cells for thiamine. Introduction of the csdA gene into the desulfurase-deficient strains did not change tellurite resistance or nutritional requirement patterns of the recipient cells. Complementation analysis could not be performed under anaerobic conditions because the three mutants did not show tellurite hypersensitivity. These results indicate that oxidative stress is involved in tellurite toxicity in E. coli.


Assuntos
Liases de Carbono-Enxofre/genética , Escherichia coli/genética , Liases/genética , Telúrio/farmacologia , Liases de Carbono-Enxofre/deficiência , Farmacorresistência Bacteriana/genética , Escherichia coli/efeitos dos fármacos , Escherichia coli/crescimento & desenvolvimento , Teste de Complementação Genética , Liases/deficiência , Mutação , Regiões Promotoras Genéticas
13.
Plant Physiol Biochem ; 42(6): 557-64, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15246070

RESUMO

Magnesium chelatase (EC 6.6.1.1) catalyses the insertion of magnesium into protoporphyrin IX, the first unique step of the chlorophyll biosynthetic pathway. The enzyme is composed of three different subunits of approximately 40, 70 and 140 kDa. In barley (Hordeum vulgare L.) the subunits are encoded by the genes Xantha-h, Xantha-g and Xantha-f. In the 1950s, eight induced xantha-f mutants were isolated. In this work we characterized these mutations at the DNA level and provided explanations for their phenotypes. The xantha-f10 mutation is a 3 bp deletion, resulting in a polypeptide lacking the glutamate residue at position 424. The leaky mutation xantha-f26 has a missense mutation leading to a M632R exchange. The xantha-f27 and -f40 are deletions of 14 and 2 bp, respectively, resulting in truncated polypeptides of 1104 and 899 amino acid residues, respectively. Mutation xantha-f41 is an in-frame deletion that removes A439, L440, Q441 and V442 from the resulting protein. Mutation xantha-f58 is most likely a deletion of the whole Xantha-f gene, as no DNA fragments could be detected by PCR or southern blot experiments. The slightly leaky xantha-f60 and non-leaky -f68 mutations each have a missense mutation causing a P393L and G794E exchange in the polypeptide, respectively.


Assuntos
Hordeum/genética , Liases/deficiência , Liases/genética , Mutação/genética , Sequência de Aminoácidos , Mutação da Fase de Leitura/genética , Deleção de Genes , Hordeum/enzimologia , Magnésio/metabolismo , Dados de Sequência Molecular , Mutação de Sentido Incorreto/genética , Protoporfirinas/metabolismo
14.
Hum Mol Genet ; 11(25): 3237-48, 2002 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-12444108

RESUMO

Girls with MLS syndrome have microphthalmia with linear skin defects of face and neck, sclerocornea, corpus callosum agenesis and other brain anomalies. This X-linked dominant, male-lethal condition is associated with heterozygous deletions of a critical region in Xp22.31, from the 5' untranslated region of MID1 at the telomeric boundary to the ARHGAP6 gene at the centromeric boundary. HCCS, encoding human holocytochrome c-type synthetase, is the only gene located entirely inside the critical region. Because single gene analysis is not feasible in MLS patients (all have deletions), we generated a deletion of the equivalent region in the mouse to study the molecular basis of this syndrome. This deletion inactivates mouse Hccs, whose homologs in lower organisms (cytochrome c or c1 heme lyases) are essential for function of cytochrome c or c1 in the mitochondrial respiratory chain. Ubiquitous deletions generated in vivo lead to lethality of hemizygous, homozygous and heterozygous embryos early in development. This lethality is rescued by expression of the human HCCS gene from a transgenic BAC, resulting in viable homozygous, heterozygous and hemizygous deleted mice with no apparent phenotype. In the presence of the HCCS transgene, the deletion is easily transmitted to subsequent generations. We did obtain a single heterozygous deleted female that does not express human HCCS, which is analogous to the low prevalence of the heterozygous MLS deletion in humans. Through the study of these genetically engineered mice we demonstrate that loss of HCCS causes the male lethality of MLS syndrome.


Assuntos
Genes Dominantes/genética , Ligação Genética , Liases/deficiência , Microftalmia/genética , Anormalidades da Pele/genética , Cromossomo X/genética , Regiões 5' não Traduzidas/genética , Animais , Blastocisto/química , Blastocisto/metabolismo , Sobrevivência Celular/genética , Modelos Animais de Doenças , Éxons/genética , Feminino , Deleção de Genes , Genes Letais/genética , Variação Genética , Holoenzimas/deficiência , Holoenzimas/genética , Holoenzimas/fisiologia , Humanos , Liases/genética , Liases/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mosaicismo/genética , Fenótipo , Homologia de Sequência do Ácido Nucleico , Síndrome , Células-Tronco Totipotentes/química , Células-Tronco Totipotentes/metabolismo , Transgenes
15.
Neuroreport ; 9(7): 1431-4, 1998 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-9631442

RESUMO

Kynurenine aminotransferase I (KAT-I), which also shows glutamine transaminase K (GTK) activity, catalyses the conversion of kynurenine to kynurenic acid, an endogenous glutamate antagonist. Both the GTK and KAT enzyme activities were found to be significantly reduced in kidney, brain and medulla oblongata homogenates of spontaneously hypertensive (SHR) compared to Wistar-Kyoto (WKY) rats. Enzyme activity stains on native gel separations of partially purified kidney homogenates was associated with two major bands of GTK (KAT-I)-activity in WKY and Wistar rats, KAT-Ia and KAT-Ib. SHR rats however showed only KAT-Ia activity. These findings suggest that the absence of KAT-Ib activity may result in a reduced ability to synthesise kynurenic acid in SHR rats, this may help to explain the enhanced sensitivity to glutamate seen in this strain.


Assuntos
Encéfalo/enzimologia , Liases/metabolismo , Bulbo/enzimologia , Transaminases/metabolismo , Animais , Isoenzimas/deficiência , Isoenzimas/metabolismo , Rim/enzimologia , Liases/deficiência , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Ratos Wistar , Valores de Referência , Transaminases/deficiência
16.
Pharm World Sci ; 19(3): 126-32, 1997 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9259028

RESUMO

An elevated plasma homocysteine level may result from various environmental and genetic factors. Herediatary causes of severe hyperhomo-cysteinaemia are very rare and usually lead to disease in childhood or adolescence. Common pathology consists of early atherosclerotic vascular changes, arterioocclusive complications and venous thrombosis. Mildly elevated genetically determined plasma homocysteine levels are observed in 5% of the general population. In the last two decades research has shown mild hyperhomocysteinaemia to be linked to an increased risk of premature atherosclerosis, pregnancies complicated by neural tube defects and early pregnancy loss, and venous thrombosis. Homozygosity for thermolabile MTHFR deficiency has been identified as one important genetic factor, which expression is modified by dietary folate intake. Although mild hyperhomocysteinaemia can easily be treated by vitamin supplementation the beneficial effects of such treatment remains to be shown.


Assuntos
Homocisteína/sangue , Homocistinúria/fisiopatologia , Erros Inatos do Metabolismo/sangue , Oxirredutases/deficiência , 5,10-Metilenotetra-Hidrofolato Redutase (FADH2) , Arteriosclerose/epidemiologia , Arteriosclerose/etiologia , Feminino , Homocistinúria/sangue , Homocistinúria/tratamento farmacológico , Homocistinúria/genética , Homozigoto , Humanos , Liases/deficiência , Erros Inatos do Metabolismo/etiologia , Erros Inatos do Metabolismo/genética , Erros Inatos do Metabolismo/fisiopatologia , Metilenotetra-Hidrofolato Redutase (NADPH2) , Defeitos do Tubo Neural/epidemiologia , Defeitos do Tubo Neural/etiologia , Gravidez , Complicações na Gravidez/epidemiologia , Complicações na Gravidez/etiologia , Tromboflebite/epidemiologia , Tromboflebite/etiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA