Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 114
Filtrar
1.
Clin Infect Dis ; 77(8): 1157-1165, 2023 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-37264998

RESUMO

BACKGROUND: Reduced plasma vitamin C (vitC) concentrations in human immunodeficiency virus (HIV) may result from abnormal urinary excretion: a renal leak. VitC renal leak indicates underlying nutritional dysregulation independent of diet. We hypothesized that increased renal leak prevalence in HIV would be associated with deficient vitC concentrations. METHODS: We conducted an outpatient cross-sectional study of 96 women (40 HIV [PWH] and 56 without HIV [PWOH]) at the National Institutes of Health and Georgetown University. Renal leak was defined as abnormal urinary vitC excretion at fasting plasma concentrations <43.2µM, 2 SDs below vitC renal threshold in healthy women. To determine the primary outcome of renal leak prevalence, matched urine and plasma samples were collected the morning after overnight fast. Secondary outcomes assessed group differences in mean plasma vitC concentrations and prevalence of vitC deficiency. Exploratory outcomes assessed clinical parameters associated with renal leak. VitC was measured by high-performance liquid chromatography with coulometric electrochemical detection. RESULTS: PWH had significantly higher renal leak prevalence (73%vs14%; OR (odds ratio):16; P<.001), lower mean plasma vitC concentrations (14µMvs50µM; P<.001), and higher prevalence of vitC deficiency (43%vs7%; OR:10; P<.001) compared with PWOH, unchanged by adjustments for confounding factors. Significant predictors of renal leak included antiretroviral therapy (ART), Black race, older age, and metabolic comorbidities but not viral load or CD4 count. When compared with other chronic disease cohorts, PWH had the highest prevalence of renal leak and vitC deficiency (P<.001). CONCLUSIONS: High prevalence of vitC renal leak in HIV was associated with vitC deficiency, ART use, and race/ethnicity differences.


Assuntos
Deficiência de Ácido Ascórbico , Infecções por HIV , Feminino , Humanos , Ácido Ascórbico/metabolismo , Ácido Ascórbico/uso terapêutico , Estudos Transversais , Deficiência de Ácido Ascórbico/complicações , Deficiência de Ácido Ascórbico/metabolismo , HIV , Comorbidade , Infecções por HIV/complicações , Infecções por HIV/tratamento farmacológico , Infecções por HIV/epidemiologia
2.
Biomed Pharmacother ; 156: 113859, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36252352

RESUMO

The AKR1A1 protein is a member of the aldo-keto reductase superfamily that catalyzes the transformation of D-glucuronate to L-gulonate in the synthesis of L-ascorbic acid (vitamin C, Vit C). We previously demonstrated that AKR1A1 knockout mice (AKR1A1eGFP/eGFP) with Vit C deficiency exhibited aberrant bone formation and osteoporosis. In this study, we aimed to evaluate the osteoprotective effects of kefir peptides (KPs) in AKR1A1eGFP/eGFP mice and uncover the underlying mechanism of KPs in the modulation of bone remodeling. Six male CD-1 mice and 24 male AKR1A1eGFP/eGFP mice were used in this study, in which the AKR1A1eGFP/eGFP mice were randomly divided into four groups (n = 6). KPs treatment for 12 weeks exerted several effects in AKR1A1eGFP/eGFP mice including the reduction of serum proinflammatory cytokines (IL-1ß, IL-6, TNF-α), bone resorption markers (CTX-1, RANKL), and the increase of serum bone formation markers (P1NP, OPG, OC). µ-CT analysis indicated that KPs prevented the bone loss in the femurs of AKR1A1eGFP/eGFP mice by significantly increasing the trabecular parameters of bone mineral density, bone volume and bone number. Nanoindentation analysis demonstrated that KPs enhanced the elasticity and hardness of femoral cortical bones in AKR1A1eGFP/eGFP mice. KPs promoted bone marrow mesenchymal stem cells (BMMSCs)-derived osteoblast differentiation and mineralization by upregulating positive regulators of osteoblastogenesis (Runx2, ß-catenin, BMP-2, NFATc1). Conversely, KPs inhibited bone marrow macrophages (BMMs)-derived osteoclast differentiation and bone resorption, which was demonstrated by the facts that KPs suppressed RANKL-induced p38, NF-κB, Akt, PLCγ2 and CREB-1 phosphorylation, decreased the nuclear translocation of NFATc1 and c-Fos. Our findings demonstrate the efficacy of KPs in the prevention of osteoporosis in AKR1A1eGFP/eGFP mice and also unveil the dual effects of KPs in osteogenic promotion and osteoclastic inhibition. This study supports the use of KPs as nutritional supplements for the prevention of osteoporosis.


Assuntos
Deficiência de Ácido Ascórbico , Reabsorção Óssea , Kefir , Osteoporose , Masculino , Camundongos , Animais , Osteogênese , Camundongos Knockout , Ligante RANK/metabolismo , Osteoclastos , Deficiência de Ácido Ascórbico/metabolismo , Diferenciação Celular , Osteoporose/prevenção & controle , Osteoporose/metabolismo , Reabsorção Óssea/metabolismo , NF-kappa B/metabolismo , Fatores de Transcrição NFATC/metabolismo
3.
J Nutr Biochem ; 86: 108485, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32828899

RESUMO

We have previously demonstrated that ascorbic acid (AsA) deficiency causes inflammatory changes in the liver and intestine in Osteogenic Disorder Shionogi (ODS) rats, which are unable to synthesize AsA. We have suggested that AsA deficiency increased intestinal interleukine (IL)-6 production, stimulating hepatic acute phase proteins (APPs) expression via the portal vein. In this study, we determined whether these hepatic and intestinal inflammatory changes by AsA deficiency are induced in germ-free (GF) ODS rats. For 18 days, male specific pathogen-free (SPF) ODS rats were fed the basal diet containing 600 mg AsA/kg (control group) or the AsA-free diet (AsA-deficient group) in SPF conditions, while male GF ODS rats were fed the basal diet (control group) or the AsA-free diet (AsA-deficient group) in GF conditions. Firstly, AsA deficiency significantly elevated the hepatic expression of APPs in both SPF and GF rats. In hepatic mRNA levels of some APPs, significant interaction between GF and AsA-deficiency effects was observed. Secondly, AsA deficiency elevated intestinal IL-6 and IL-1ß mRNA levels in both SPF and GF rats, and significant interaction between GF and AsA-deficiency effects was observed in these mRNA levels of jejunum and cecum. In SPF and GF rats, AsA deficiency elevated portal IL-6 concentration. These results show that AsA deficiency caused hepatic and intestinal inflammatory changes in both the GF and SPF ODS rats and indicate that AsA deficiency could directly induce intestinal inflammatory changes without the involvement of gut microbiota.


Assuntos
Deficiência de Ácido Ascórbico/metabolismo , Perfilação da Expressão Gênica , Inflamação/metabolismo , Mucosa Intestinal/metabolismo , Fígado/metabolismo , Proteínas de Fase Aguda/metabolismo , Animais , Peso Corporal , Citocinas/metabolismo , Microbioma Gastrointestinal , Interleucina-6/metabolismo , Masculino , Tamanho do Órgão , Veia Porta/metabolismo , Ratos , Distribuição Tecidual
4.
J Nutr Sci Vitaminol (Tokyo) ; 66(1): 19-23, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32115449

RESUMO

Using rats, we previously found that vitamin C deficiency increases serum levels of interleukin-6 (IL-6) and glucocorticoid, and changes the gene expression of acute phase proteins (APP) in the liver. However, it remains unclear how vitamin C deficiency causes these inflammation-like responses. In this study, we investigated the possibility that changes in gut microbiota are involved in the induction of APP gene expression by vitamin C deficiency. ODS rats that cannot genetically synthesize vitamin C were divided into 4 groups based on the presence or absence of vitamin C or antibiotics and were raised for 15 d. Neomycin, vancomycin, and ampicillin were used as antibiotics, and 300 mg L-ascorbic acid/kg was added to the AIN93G diet. Vitamin C deficiency affected neither the wet tissue weights nor relative abundance of bacteria in the cecal contents. Antibiotic administration increased wet weights of the cecum, cecal contents, and colon, changed the relative abundance of some bacteria in the cecal contents, and decreased serum IL-6 level. However, antibiotic administration had no effect on serum concentrations of corticosterone and α1-acid glycoprotein (AGP), vitamin C concentration in the liver, and mRNA levels of haptoglobin and AGP in the liver. Therefore, disturbance of gut microbiota did not attenuate the increase in glucocorticoid level and induction of APP gene expression due to vitamin C deficiency. This suggests that gut microbiota is not involved in the inflammation-like responses caused by vitamin C deficiency.


Assuntos
Proteínas de Fase Aguda/metabolismo , Deficiência de Ácido Ascórbico/metabolismo , Microbioma Gastrointestinal/fisiologia , Proteínas de Fase Aguda/análise , Animais , Antibacterianos/farmacologia , Ácido Ascórbico/análise , Ácido Ascórbico/metabolismo , Corticosterona/sangue , Corticosterona/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Inflamação/metabolismo , Interleucina-6/sangue , Interleucina-6/metabolismo , Fígado/química , Fígado/metabolismo , Masculino , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Ratos
5.
Eur J Pharmacol ; 829: 1-11, 2018 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-29625084

RESUMO

Chronic low-grade inflammation plays a major role in the development of insulin resistance. The potential role and underlying mechanism of vitamin C, an antioxidant and anti-inflammatory agent, was investigated in tumor necrosis factor-α (TNF-α)-induced insulin resistance. Gulonolactone oxidase knockout (Gulo-/-) mice genetically unable to synthesize vitamin C were used to induce insulin resistance by continuously pumping small doses of TNF-α for seven days, and human liver hepatocellular carcinoma cells (HepG2 cells) were used to induce insulin resistance by treatment with TNF-α. Vitamin C deficiency aggravated TNF-α-induced insulin resistance in Gulo-/- mice, resulting in worse glucose tolerance test (GTT) results, higher fasting plasma insulin level, and the inactivation of the protein kinase B (AKT)/glycogen synthase kinase-3ß (GSK3ß) pathway in the liver. Vitamin C deficiency also worsened liver lipid accumulation and inflammation in TNF-α-treated Gulo-/- mice. In HepG2 cells, vitamin C reversed the TNF-α-induced reduction of glucose uptake and glycogen synthesis, which were mediated by increasing GLUT2 levels and the activation of the insulin receptor substrate (IRS-1)/AKT/GSK3ß pathway. Furthermore, vitamin C inhibited the TNF-α-induced activation of not only the mitogen-activated protein kinase (MAPKs), but also nuclear factor-kappa B (NF-κB) signaling. Taken together, vitamin C is essential for preventing and improving insulin resistance, and the supplementing with vitamin C may be an effective therapeutic intervention for metabolic disorders.


Assuntos
Deficiência de Ácido Ascórbico/metabolismo , Resistência à Insulina , Fator de Necrose Tumoral alfa/farmacologia , Animais , Ácido Ascórbico/farmacologia , Deficiência de Ácido Ascórbico/patologia , Ativação Enzimática/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Células Hep G2 , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Quinase I-kappa B/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Inibidor de NF-kappaB alfa/metabolismo , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
6.
Nature ; 549(7673): 476-481, 2017 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-28825709

RESUMO

Stem-cell fate can be influenced by metabolite levels in culture, but it is not known whether physiological variations in metabolite levels in normal tissues regulate stem-cell function in vivo. Here we describe a metabolomics method for the analysis of rare cell populations isolated directly from tissues and use it to compare mouse haematopoietic stem cells (HSCs) to restricted haematopoietic progenitors. Each haematopoietic cell type had a distinct metabolic signature. Human and mouse HSCs had unusually high levels of ascorbate, which decreased with differentiation. Systemic ascorbate depletion in mice increased HSC frequency and function, in part by reducing the function of Tet2, a dioxygenase tumour suppressor. Ascorbate depletion cooperated with Flt3 internal tandem duplication (Flt3ITD) leukaemic mutations to accelerate leukaemogenesis, through cell-autonomous and possibly non-cell-autonomous mechanisms, in a manner that was reversed by dietary ascorbate. Ascorbate acted cell-autonomously to negatively regulate HSC function and myelopoiesis through Tet2-dependent and Tet2-independent mechanisms. Ascorbate therefore accumulates within HSCs to promote Tet activity in vivo, limiting HSC frequency and suppressing leukaemogenesis.


Assuntos
Ácido Ascórbico/metabolismo , Carcinogênese/metabolismo , Células-Tronco Hematopoéticas/citologia , Leucemia/patologia , Animais , Ácido Ascórbico/análise , Deficiência de Ácido Ascórbico/genética , Deficiência de Ácido Ascórbico/metabolismo , Carcinogênese/genética , Proteínas de Ligação a DNA/metabolismo , Dioxigenases , Feminino , Células-Tronco Hematopoéticas/metabolismo , Humanos , Leucemia/genética , Masculino , Metabolômica , Camundongos , Mielopoese/genética , Proteínas Proto-Oncogênicas/metabolismo , Tirosina Quinase 3 Semelhante a fms/genética , Tirosina Quinase 3 Semelhante a fms/metabolismo
7.
Proc Natl Acad Sci U S A ; 113(37): 10238-44, 2016 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-27573823

RESUMO

Vitamin C deficiency is found in patients with cancer and might complicate various therapy paradigms. Here we show how this deficiency may influence the use of DNA methyltransferase inhibitors (DNMTis) for treatment of hematological neoplasias. In vitro, when vitamin C is added at physiological levels to low doses of the DNMTi 5-aza-2'-deoxycytidine (5-aza-CdR), there is a synergistic inhibition of cancer-cell proliferation and increased apoptosis. These effects are associated with enhanced immune signals including increased expression of bidirectionally transcribed endogenous retrovirus (ERV) transcripts, increased cytosolic dsRNA, and activation of an IFN-inducing cellular response. This synergistic effect is likely the result of both passive DNA demethylation by DNMTi and active conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) by ten-eleven translocation (TET) enzymes at LTR regions of ERVs, because vitamin C acts as a cofactor for TET proteins. In addition, TET2 knockout reduces the synergy between the two compounds. Furthermore, we show that many patients with hematological neoplasia are markedly vitamin C deficient. Thus, our data suggest that correction of vitamin C deficiency in patients with hematological and other cancers may improve responses to epigenetic therapy with DNMTis.


Assuntos
Ácido Ascórbico/administração & dosagem , Azacitidina/análogos & derivados , Inibidores Enzimáticos/administração & dosagem , Neoplasias Hematológicas/tratamento farmacológico , Apoptose/efeitos dos fármacos , Deficiência de Ácido Ascórbico/complicações , Deficiência de Ácido Ascórbico/tratamento farmacológico , Deficiência de Ácido Ascórbico/metabolismo , Deficiência de Ácido Ascórbico/patologia , Azacitidina/administração & dosagem , Proliferação de Células/efeitos dos fármacos , Metilação de DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/genética , Decitabina , Dioxigenases , Sinergismo Farmacológico , Retrovirus Endógenos/genética , Feminino , Neoplasias Hematológicas/complicações , Neoplasias Hematológicas/patologia , Humanos , Interferons/genética , Masculino , Metiltransferases/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , RNA de Cadeia Dupla/efeitos dos fármacos
8.
Bratisl Lek Listy ; 117(6): 305-7, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27546360

RESUMO

Epidemiology of bronchial asthma (BA) indicates a marked paradox: rapid rise in the prevalence.Simultaneous decline in mortality is mostly related to improvement in the diagnosis and therapy. In many economically developed countries the BA affects more than 10 per cent of the population, while mortality related to this respiratory disorder is below 1/100,000. Factors favorably influencing mortality of BA include new more effective medications, decline in smoking and also improved nutrition, based on awareness of protective role of vitamins. Vitamin D deficiency has a number of biological effects that are potentially instrumental in the pathogenesis and severity of BA. Increased number of randomized, controlled, interventional studies is showing positive effects of vitamin D supplementation in pediatric and in adult BA. Oxidative stress is potentially an important pathogenic factor in the progression of BA. Vitamin C (ascorbic acid) belongs to the most effective nutritional antioxidants. By counteracting oxidants, reducing generation of reactive oxygen species, vitamin C may inhibit external attacks in the respiratory tract, thus modulating the development of BA (Fig. 2, Ref. 15).


Assuntos
Deficiência de Ácido Ascórbico/epidemiologia , Asma/epidemiologia , Suplementos Nutricionais , Deficiência de Vitamina D/epidemiologia , Vitaminas/uso terapêutico , Adulto , Antioxidantes/uso terapêutico , Ácido Ascórbico/uso terapêutico , Deficiência de Ácido Ascórbico/tratamento farmacológico , Deficiência de Ácido Ascórbico/metabolismo , Asma/tratamento farmacológico , Asma/metabolismo , Criança , Humanos , Estresse Oxidativo , Fumar/epidemiologia , Vitamina D/uso terapêutico , Deficiência de Vitamina D/tratamento farmacológico , Deficiência de Vitamina D/metabolismo
9.
Cell Mol Life Sci ; 73(8): 1645-58, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26846695

RESUMO

Recent advances have uncovered a previously unknown function of vitamin C in epigenetic regulation. Vitamin C exists predominantly as an ascorbate anion under physiological pH conditions. Ascorbate was discovered as a cofactor for methylcytosine dioxygenases that are responsible for DNA demethylation, and also as a likely cofactor for some JmjC domain-containing histone demethylases that catalyze histone demethylation. Variation in ascorbate bioavailability thus can influence the demethylation of both DNA and histone, further leading to different phenotypic presentations. Ascorbate deficiency can be presented systematically, spatially and temporally in different tissues at the different stages of development and aging. Here, we review how ascorbate deficiency could potentially be involved in embryonic and postnatal development, and plays a role in various diseases such as neurodegeneration and cancer through epigenetic dysregulation.


Assuntos
Deficiência de Ácido Ascórbico/metabolismo , Ácido Ascórbico/metabolismo , Metilação de DNA/fisiologia , Desenvolvimento Embrionário/fisiologia , Proteínas F-Box/metabolismo , Histonas/metabolismo , Histona Desmetilases com o Domínio Jumonji/metabolismo , Envelhecimento , Deficiência de Ácido Ascórbico/genética , Dioxigenases/metabolismo , Epigênese Genética , Humanos , Neoplasias/patologia , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Escorbuto/patologia
10.
Br J Nutr ; 113(4): 603-9, 2015 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-25608928

RESUMO

Vitamin C (VC) is an essential nutrient for humans and certain other animals. It has antioxidant properties and has been reported to ameliorate oxidative damage to lipids, DNA and proteins. However, the effects of VC on immune function are poorly understood, especially the influence of long-term high-dose VC intake on the number and function of immune cells. In the present study, to evaluate the immune effects of VC, VC-deficient senescence marker protein-30 knockout (SMP30KO) mice were fed a diet containing the recommended level of VC (20 mg/kg per d; 0·02 % VC) or a high level of VC (200 mg/kg per d; 0·2 % VC) for 1 year. The plasma VC concentration of the 0·02 % group was the same as that of age-matched C57BL/6 mice after 1 year of feeding; however, plasma VC concentration and thymus weight were significantly higher in the 0·2 % VC group than in the 0·02 % VC group. The total counts of leucocytes, lymphocytes, granulocytes and monocytes in the peripheral blood, as well as the number of splenocytes and thymocytes, were all significantly higher in the 0·2 % VC group than in the 0·02 % VC group. In addition, the number of naive T cells in peripheral blood lymphocytes, the number of memory T-cell populations in splenocytes, and the number of cluster of differentiation (CD)4⁺CD8⁺ or CD4⁺CD8⁻ or CD4⁻CD8⁺ T cells in thymocytes were all markedly higher in the 0·2 % VC group than in the 0·02 % VC group after 1 year of dietary treatment. These results suggest that a long-term high-dose intake of VC is effective in the maintenance of immune cells, partly through the suppression of age-related thymic involution in VC-deficient SMP30KO mice.


Assuntos
Envelhecimento , Ácido Ascórbico/uso terapêutico , Proteínas de Ligação ao Cálcio/metabolismo , Suplementos Nutricionais , Fatores Imunológicos/uso terapêutico , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Doenças Linfáticas/prevenção & controle , Timo/patologia , Animais , Antioxidantes/administração & dosagem , Antioxidantes/efeitos adversos , Antioxidantes/análise , Antioxidantes/uso terapêutico , Ácido Ascórbico/administração & dosagem , Ácido Ascórbico/efeitos adversos , Ácido Ascórbico/sangue , Deficiência de Ácido Ascórbico/sangue , Deficiência de Ácido Ascórbico/dietoterapia , Deficiência de Ácido Ascórbico/metabolismo , Deficiência de Ácido Ascórbico/fisiopatologia , Atrofia , Proteínas de Ligação ao Cálcio/genética , Suplementos Nutricionais/efeitos adversos , Fatores Imunológicos/administração & dosagem , Fatores Imunológicos/efeitos adversos , Fatores Imunológicos/sangue , Peptídeos e Proteínas de Sinalização Intracelular/genética , Contagem de Leucócitos , Doenças Linfáticas/etiologia , Doenças Linfáticas/imunologia , Doenças Linfáticas/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tamanho do Órgão , Distribuição Aleatória , Organismos Livres de Patógenos Específicos , Baço/imunologia , Baço/metabolismo , Baço/patologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/patologia , Timo/imunologia , Timo/metabolismo
11.
Biochim Biophys Acta ; 1840(7): 2289-98, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24704458

RESUMO

BACKGROUND: We sought to elucidate the effect of an ascorbic acid (AA) deficiency on gene expression, because the water soluble antioxidant AA is an important bioactive substance in vivo. METHODS: We performed microarray analyses of the transcriptome in the liver from senescence marker protein-30 (SMP30)/gluconolactonase (GNL) knockout (KO) mice, which are unable to synthesize AA in vivo. RESULTS: Our microarray analysis revealed that the AA deficiency increased gene expression related to the oxidation-reduction process, i.e., the nuclear factor, erythroid derived 2, like 2 (Nrf2) gene, which is a reactive oxygen species-sensitive transcriptional factor. Moreover, this AA deficiency increased the expression of genes for lipid metabolism including the cytochrome P450, family 7, subfamily a, polypeptide 1 (Cyp7a1), which is a late-limiting enzyme of the primary bile acid biosynthesis pathway. Although an AA deficiency increased the Cyp7a1 protein level, bile acid levels in the liver and gallbladder decreased. Since Cyp7a1 has a heme iron at the active site, AA must function as a reductant of the iron required for the continuous activation of Cyp7a1. CONCLUSIONS: This experimental evidence strongly supports a role for AA in the physiologic oxidation-reduction process and lipid metabolism including bile acid biosynthesis. GENERAL SIGNIFICANCE: Although many effects of AA supplementation have been reported, no microarray analysis of AA deficiency in vivo is available. Results from using this unique model of AA deficiency, the SMP30/GNL-KO mouse, now provide new information about formerly unknown AA functions that will implement further study of AA in vivo.


Assuntos
Deficiência de Ácido Ascórbico/metabolismo , Ácido Ascórbico/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Metabolismo dos Lipídeos , Animais , Ácido Ascórbico/biossíntese , Deficiência de Ácido Ascórbico/genética , Proteínas de Ligação ao Cálcio/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Fígado/metabolismo , Camundongos , Camundongos Knockout , Análise em Microsséries , Oxirredução , Transcriptoma
12.
J Nutr ; 143(1): 1-11, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23173175

RESUMO

Dietary antioxidants are essential nutrients that inhibit the oxidation of biologically important molecules and suppress the toxicity of reactive oxygen or nitrogen species. When the total antioxidant capacity is insufficient to quench these reactive species, oxidative damage occurs and contributes to the onset and progression of chronic diseases, such as neurodegenerative diseases, cardiovascular diseases, and cancer. However, epidemiological studies that examine the relationship between antioxidants and disease outcome can only identify correlative associations. Additionally, many antioxidants also have prooxidant effects. Thus, clinically relevant animal models of antioxidant function are essential for improving our understanding of the role of antioxidants in the pathogenesis of complex diseases as well as evaluating the therapeutic potential and risks of their supplementation. Recent progress in gene knockout mice and virus-based gene expression has potentiated these areas of study. Here, we review the current genetically modified animal models of dietary antioxidant function and their clinical relevance in chronic diseases. This review focuses on the 3 major antioxidants in the human body: vitamin C, vitamin E, and uric acid. We examine genetic models of vitamin C synthesis (guinea pig, Osteogenic Disorder Shionogi rat, Gulo(-/-) and SMP30(-/-) mouse mutants) and transport (Slc23a1(-/-) and Slc23a2(-/-) mouse mutants), vitamin E transport (Ttpa(-/-) mouse mutant), and uric acid synthesis (Uox(-/-) mouse mutant). The application of these models to current research goals is also discussed.


Assuntos
Antioxidantes/administração & dosagem , Deficiências Nutricionais/fisiopatologia , Modelos Animais de Doenças , Estresse Oxidativo , Animais , Animais Geneticamente Modificados , Antioxidantes/efeitos adversos , Antioxidantes/metabolismo , Antioxidantes/uso terapêutico , Deficiência de Ácido Ascórbico/dietoterapia , Deficiência de Ácido Ascórbico/metabolismo , Deficiência de Ácido Ascórbico/fisiopatologia , Deficiências Nutricionais/dietoterapia , Deficiências Nutricionais/metabolismo , Humanos , Ácido Úrico/administração & dosagem , Ácido Úrico/efeitos adversos , Ácido Úrico/metabolismo , Ácido Úrico/uso terapêutico , Deficiência de Vitamina E/dietoterapia , Deficiência de Vitamina E/metabolismo , Deficiência de Vitamina E/fisiopatologia
13.
Int J Oncol ; 42(1): 55-64, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23175106

RESUMO

Degradation of the extracellular matrix (ECM) plays a critical role in the formation of tumors and metastasis and has been found to correlate with the aggressiveness of tumor growth and invasiveness of cancer. Ascorbic acid, which is known to be essential for the structural integrity of the intercellular matrix, is not produced by humans and must be obtained from the diet. Cancer patients have been shown to have very low reserves of ascorbic acid. Our main objective was to determine the effect of ascorbate supplementation on metastasis, tumor growth and tumor immunohistochemistry in mice unable to synthesize ascorbic acid [gulonolactone oxidase (gulo) knockout (KO)] when challenged with B16FO melanoma or 4T1 breast cancer cells. Gulo KO female mice 36-38 weeks of age were deprived of or maintained on ascorbate in food and water for 4 weeks prior to and 2 weeks post intraperitoneal (IP) injection of 5x105 B16FO murine melanoma cells or to injection of 5x105 4T1 breast cancer cells into the mammary pad of mice. Ascorbate-supplemented gulo KO mice injected with B16FO melanoma cells demonstrated significant reduction (by 71%, p=0.005) in tumor metastasis compared to gulo KO mice on the control diet. The mean tumor weight in ascorbate supplemented mice injected with 4T1 cells was reduced by 28% compared to tumor weight in scorbutic mice. Scorbutic tumors demonstrated large dark cores, associated with increased necrotic areas and breaches to the tumor surface, apoptosis and matrix metalloproteinase-9 (MMP-9), and weak, disorganized or missing collagen I tumor capsule. In contrast, the ascorbate-supplemented group tumors had smaller fainter colored cores and confined areas of necrosis/apoptosis with no breaches from the core to the outside of the tumor and a robust collagen I tumor capsule. In both studies, ascorbate supplementation of gulo KO mice resulted in profoundly decreased serum inflammatory cytokine interleukin (IL)-6 (99% decrease, p=0.01 in the B16F0 study and 85% decrease, p=0.08 in the 4T1 study) compared to the levels in gulo KO mice deprived of ascorbate. In the B16FO study, ascorbate supplementation of gulo KO mice resulted in profoundly decreased serum VEGF (98% decrease, p=0.019 than in the scorbutic gulo KO mice). As expected, mean serum ascorbate level in ascorbate-restricted mice was 2% (p<0.001) of the mean ascorbate levels in supplemented mice. In conclusion, ascorbate supplementation hinders metastasis, tumor growth and inflammatory cytokine secretion as well as enhanced encapsulation of tumors elicited by melanoma and breast cancer cell challenge in gulo KO mice.


Assuntos
Antioxidantes/administração & dosagem , Deficiência de Ácido Ascórbico/prevenção & controle , Ácido Ascórbico/administração & dosagem , Neoplasias da Mama/prevenção & controle , Suplementos Nutricionais , L-Gulonolactona Oxidase/fisiologia , Melanoma Experimental/prevenção & controle , Animais , Antioxidantes/metabolismo , Apoptose/efeitos dos fármacos , Ácido Ascórbico/metabolismo , Deficiência de Ácido Ascórbico/metabolismo , Deficiência de Ácido Ascórbico/patologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Técnicas Imunoenzimáticas , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Metástase Neoplásica , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular/metabolismo
14.
Basic Clin Pharmacol Toxicol ; 110(6): 524-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22212866

RESUMO

Increased oxidative stress in the brain has consistently been implied in ageing and in several degenerative brain disorders. Acting as a pivotal antioxidant in the brain, vitamin C is preferentially retained during deficiency and may play an essential role in neuroprotection during ageing. Thus, a lack of vitamin C could be associated with an increase in redox imbalance in the ageing brain. The present study compared oxidative stress of ageing to that of a long-term non-scorbutic vitamin C deficiency in guinea pigs. Adults (3-9 months old) were compared to old (36-42 months old) animals during a 6-month dietary intervention by assessing vitamin C transport and redox homoeostasis in the brain. In contrast to our hypothesis, chronic vitamin C deficiency did not affect the measured markers of oxidative stress in the brains of adult and aged animals. However, aged animals generally showed increased lipid oxidation (p < 0.001), decreased glutathione (p < 0.05), increased p53 mRNA expression (p < 0.01) and somewhat elevated DNA oxidation (p = 0.08) compared to adult counterparts irrespective of dietary vitamin C intake. Increased mRNA expression of sod1 (p < 0.05) and svct2 (p = 0.05) was observed in aged animals together with increased superoxide dismutase activity (p < 0.01) and cerebrospinal fluid vitamin C status (p < 0.001) suggesting a compensatory effort that did not counterbalance the effects of ageing. Essentially, no effects of age were observed in the liver demonstrating the brain's unique susceptibility to redox imbalance. Consistent with previous findings, we show that ageing per se constitutes a considerable oxidative insult in the brain. However, our data also suggest that a long-term poor vitamin C status does not accelerate this process.


Assuntos
Envelhecimento/metabolismo , Deficiência de Ácido Ascórbico/metabolismo , Encéfalo/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fatores Etários , Animais , Deficiência de Ácido Ascórbico/líquido cefalorraquidiano , Doença Crônica , Feminino , Regulação da Expressão Gênica , Glutationa/metabolismo , Cobaias , Homeostase , Peroxidação de Lipídeos , Fígado/metabolismo , Oxirredução , RNA Mensageiro/metabolismo , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1 , Fatores de Tempo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
15.
J Korean Med Sci ; 26(3): 317-24, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21394297

RESUMO

Hyperoxic ventilation induces detrimental effects on the respiratory system, and ambient oxygen may be harmful unless compensated by physiological anti-oxidants, such as vitamin C. Here we investigate the changes in electrolyte transport of airway epithelium in mice exposed to normobaric hyperoxia and in gulonolacton oxidase knock-out (gulo[-/-]) mice without vitamin C (Vit-C) supplementation. Short-circuit current (I(sc)) of tracheal epithelium was measured using Ussing chamber technique. After confirming amiloride-sensitive Na(+) absorption (ΔI(sc,amil)), cAMP-dependent Cl(-) secretion (ΔI(sc,forsk)) was induced by forskolin. To evaluate Ca(2+)-dependent Cl(-) secretion, ATP was applied to the luminal side (ΔI(sc,ATP)). In mice exposed to 98% PO(2) for 36 hr, ΔI(sc,forsk) decreased, ΔI(sc,amil) and ΔI(sc,ATP) was not affected. In gulo(-/-) mice, both ΔI(sc,forsk) and ΔI(sc,ATP) decreased from three weeks after Vit-C deprivation, while both were unchanged with Vit-C supplementation. At the fourth week, tissue resistance and all electrolyte transport activities were decreased. An immunofluorescence study showed that the expression of cystic fibrosis conductance regulator (CFTR) was decreased in gulo(-/-) mice, whereas the expression of KCNQ1 K(+) channel was preserved. Taken together, the CFTR-mediated Cl(-) secretion of airway epithelium is susceptible to oxidative stress, which suggests that supplementation of the antioxidant might be beneficial for the maintenance of airway surface liquid.


Assuntos
Deficiência de Ácido Ascórbico/metabolismo , Cloretos/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Hiperóxia/fisiopatologia , Mucosa Respiratória/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Colforsina/farmacologia , Regulador de Condutância Transmembrana em Fibrose Cística/antagonistas & inibidores , Regulador de Condutância Transmembrana em Fibrose Cística/efeitos dos fármacos , Oxigenoterapia Hiperbárica , Transporte de Íons/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout/metabolismo , Camundongos Transgênicos , Microscopia de Fluorescência , Estresse Oxidativo , Oxigênio/efeitos adversos , Oxigênio/farmacologia , Canais de Potássio/metabolismo , Mucosa Respiratória/efeitos dos fármacos , Sódio , Açúcares Ácidos/metabolismo
16.
Blood ; 117(20): 5485-93, 2011 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-21346252

RESUMO

Prolyl-4-hydroxylation is necessary for proper structural assembly of collagens and oxygen-dependent protein stability of hypoxia-inducible transcription factors (HIFs). In vitro function of HIF prolyl-4-hydroxylase domain (PHD) enzymes requires oxygen and 2-oxoglutarate as cosubstrates with iron(II) and vitamin C serving as cofactors. Although vitamin C deficiency is known to cause the collagen-disassembly disease scurvy, it is unclear whether cellular oxygen sensing is similarly affected. Here, we report that vitamin C-deprived Gulo(-/-) knockout mice show normal HIF-dependent gene expression. The systemic response of Gulo(-/-) animals to inspiratory hypoxia, as measured by plasma erythropoietin levels, was similar to that of animals supplemented with vitamin C. Hypoxic HIF induction was also essentially normal under serum- and vitamin C-free cell-culture conditions, suggesting that vitamin C is not required for oxygen sensing in vivo. Glutathione was found to fully substitute for vitamin C requirement of all 3 PHD isoforms in vitro. Consistently, glutathione also reduced HIF-1α protein levels, transactivation activity, and endogenous target gene expression in cells exposed to CoCl(2). A Cys201Ser mutation in PHD2 increased basal hydroxylation rates and conferred resistance to oxidative damage in vitro, suggesting that this surface-accessible PHD2 cysteine residue is a target of antioxidative protection by vitamin C and glutathione.


Assuntos
Ácido Ascórbico/metabolismo , Oxigênio/metabolismo , Substituição de Aminoácidos , Animais , Deficiência de Ácido Ascórbico/metabolismo , Hipóxia Celular , Linhagem Celular , Cobalto/farmacologia , Glutationa/metabolismo , Células HeLa , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Prolina Dioxigenases do Fator Induzível por Hipóxia , L-Gulonolactona Oxidase/deficiência , L-Gulonolactona Oxidase/genética , Camundongos , Camundongos Knockout , Mutagênese Sítio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Pró-Colágeno-Prolina Dioxigenase/genética , Pró-Colágeno-Prolina Dioxigenase/metabolismo
17.
Free Radic Biol Med ; 50(1): 27-36, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20974251

RESUMO

To test whether severe ascorbic acid deficiency in macrophages affects progression of early atherosclerosis, we used fetal liver cell transplantation to generate atherosclerosis-prone apolipoprotein E-deficient (apoE(-/-)) mice that selectively lacked the ascorbate transporter (SVCT2) in hematopoietic cells, including macrophages. After 13 weeks of chow diet, apoE(-/-) mice lacking the SVCT2 in macrophages had surprisingly less aortic atherosclerosis, decreased lesion macrophage numbers, and increased macrophage apoptosis compared to control-transplanted mice. Serum lipid levels were similar in both groups. Peritoneal macrophages lacking the SVCT2 had undetectable ascorbate; increased susceptibility to H(2)O(2)-induced mitochondrial dysfunction and apoptosis; decreased expression of genes for COX-2, IL1ß, and IL6; and decreased lipopolysaccharide-stimulated NF-κB and antiapoptotic gene expression. These changes were associated with decreased expression of both the receptor for advanced glycation end products and HIF-1α, either or both of which could have been the proximal cause of decreased macrophage activation and apoptosis in ascorbate-deficient macrophages.


Assuntos
Deficiência de Ácido Ascórbico/genética , Aterosclerose/genética , Aterosclerose/prevenção & controle , Macrófagos Peritoneais/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Simportadores/genética , Animais , Apolipoproteínas E/genética , Ácido Ascórbico/efeitos adversos , Ácido Ascórbico/metabolismo , Deficiência de Ácido Ascórbico/metabolismo , Deficiência de Ácido Ascórbico/patologia , Aterosclerose/etiologia , Aterosclerose/patologia , Células Cultivadas , Progressão da Doença , Feminino , Predisposição Genética para Doença/prevenção & controle , Macrófagos Peritoneais/patologia , Macrófagos Peritoneais/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Especificidade de Órgãos/genética , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/fisiologia , Transportadores de Sódio Acoplados à Vitamina C , Simportadores/metabolismo , Simportadores/fisiologia , Fatores de Tempo
18.
Arterioscler Thromb Vasc Biol ; 30(9): 1751-7, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20558818

RESUMO

OBJECTIVE: To assess the role of combined deficiencies of vitamins C and E on the earliest stages of atherosclerosis (an inflammatory condition associated with oxidative stress), 4 combinations of vitamin supplementation (low C/low E, low C/high E, high C/low E, and high C/high E) were studied in atherosclerosis-prone apolipoprotein E-deficient mice also unable to synthesize their own vitamin C (gulonolactone oxidase(-/-)); and to evaluate the effect of a more severe depletion of vitamin C alone in a second experiment using gulonolactone oxidase(-/-) mice carrying the hemizygous deletion of SVCT2 (the vitamin C transporter). METHODS AND RESULTS: After 8 weeks of a high-fat diet (16% lard and 0.2% cholesterol), atherosclerosis developed in the aortic sinus areas of mice in all diet groups. Each vitamin-deficient diet significantly decreased liver and brain contents of the corresponding vitamin. Combined deficiency of both vitamins increased lipid peroxidation, doubled plaque size, and increased plaque macrophage content by 2- to 3-fold in male mice, although only plaque macrophage content was increased in female mice. A more severe deficiency of vitamin C in gulonolactone oxidase(-/-) mice with defective cellular uptake of vitamin C increased both oxidative stress and atherosclerosis in apolipoprotein E(-/-) mice compared with littermates receiving a diet replete in vitamin C, again most clearly in males. CONCLUSIONS: Combined deficiencies of vitamins E and C are required to worsen early atherosclerosis in an apolipoprotein E-deficient mouse model. However, a more severe cellular deficiency of vitamin C alone promotes atherosclerosis when vitamin E is replete.


Assuntos
Doenças da Aorta/etiologia , Apolipoproteínas E/deficiência , Deficiência de Ácido Ascórbico/complicações , Aterosclerose/etiologia , Deficiência de Vitamina E/complicações , Animais , Doenças da Aorta/tratamento farmacológico , Doenças da Aorta/genética , Doenças da Aorta/metabolismo , Doenças da Aorta/patologia , Apolipoproteínas E/genética , Ácido Ascórbico/metabolismo , Ácido Ascórbico/farmacologia , Deficiência de Ácido Ascórbico/tratamento farmacológico , Deficiência de Ácido Ascórbico/metabolismo , Aterosclerose/tratamento farmacológico , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Encéfalo/metabolismo , Suplementos Nutricionais , Modelos Animais de Doenças , Progressão da Doença , Feminino , L-Gulonolactona Oxidase/deficiência , L-Gulonolactona Oxidase/genética , Peroxidação de Lipídeos , Fígado/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/deficiência , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Estresse Oxidativo , Índice de Gravidade de Doença , Fatores Sexuais , Transportadores de Sódio Acoplados à Vitamina C , Simportadores/deficiência , Simportadores/genética , Fatores de Tempo , Vitamina E/metabolismo , Vitamina E/farmacologia , Deficiência de Vitamina E/tratamento farmacológico , Deficiência de Vitamina E/metabolismo , Vitaminas/metabolismo , Vitaminas/farmacologia
19.
Arch Toxicol ; 84(8): 597-607, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20221587

RESUMO

Sulfonamide antimicrobials such as sulfamethoxazole (SMX) have been associated with drug hypersensitivity reactions, particularly in patients with AIDS. A reactive oxidative metabolite, sulfamethoxazole-nitroso (SMX-NO), forms drug-tissue adducts that elicit a T-cell response. Antioxidants such as ascorbic acid (AA) and glutathione (GSH) reduce SMX-NO to the less reactive hydroxylamine metabolite (SMX-HA), which is further reduced to the non-immunogenic parent compound by cytochrome b (5) (b5) and its reductase (b5R). We hypothesized that deficiencies in AA and GSH would enhance drug-tissue adduct formation and immunogenicity toward SMX-NO and that these antioxidant deficiencies might also impair the activity of the b5/b5R pathway. We tested these hypotheses in guinea pigs fed either a normal or AA-restricted diet, followed by buthionine sulfoximine treatment (250 mg/kg SC daily, or vehicle); and SMX-NO (1 mg/kg IP 4 days per week, or vehicle), for 2 weeks. Guinea pigs did not show any biochemical or histopathologic evidence of SMX-NO-related toxicity. Combined AA and GSH deficiency in this model did not significantly increase tissue-drug adduct formation, or splenocyte proliferation in response to SMX-NO. However, combined antioxidant deficiency was associated with decreased mRNA and protein expression of cytochrome b (5), as well as significant decreases in SMX-HA reduction in SMX-NO-treated pigs. These results suggest that SMX-HA detoxification may be down-regulated in combined AA and GSH deficiency. This mechanism could contribute to the higher risk of SMX hypersensitivity in patients with AIDS with antioxidant depletion.


Assuntos
Anti-Infecciosos/toxicidade , Deficiência de Ácido Ascórbico/metabolismo , Ácido Ascórbico/metabolismo , Citocromos b5/metabolismo , Glutationa/deficiência , Sulfametoxazol/análogos & derivados , Animais , Anti-Infecciosos/química , Anti-Infecciosos/metabolismo , Antioxidantes/metabolismo , Proliferação de Células/efeitos dos fármacos , Citocromo-B(5) Redutase/genética , Citocromo-B(5) Redutase/metabolismo , Citocromos b5/genética , Hipersensibilidade a Drogas/metabolismo , Glutationa/metabolismo , Cobaias , Inativação Metabólica , Fígado/metabolismo , Masculino , Sulfametoxazol/química , Sulfametoxazol/metabolismo , Sulfametoxazol/toxicidade , Linfócitos T/efeitos dos fármacos
20.
Biol Pharm Bull ; 31(9): 1673-9, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18758058

RESUMO

Carnitine is an essential cofactor in the transport of long-chain fatty acids into the mitochondrial matrix and plays an important role in energy production via beta-oxidation. Vitamin C (VC) has long been considered a requirement for the activities of two enzymes in the carnitine biosynthetic pathway, i.e., 6-N-trimethyllysine dioxygenase and gamma-butyrobetaine dioxygenase. Our present study using senescence marker protein-30 (SMP30)/gluconolactonase (GNL) knockout (KO) mice, which cannot synthesize VC in vivo, led to the conclusion that this notion is not true. After weaning at 40 d of age, SMP30/GNL KO mice were fed a diet lacking VC and carnitine, then given water containing 1.5 g/l VC (VC(+) mice) or no VC (VC(-) mice) for 75 d. Subsequently, total VC and carnitine levels were measured in the cerebrum, cerebellum, liver, kidney, soleus muscle, extensor digitorum longus muscle, heart, plasma and serum. The total VC levels in all tissues and plasma from VC(-) SMP30/GNL KO mice were negligible, i.e., <2% of the levels in SMP30/GNL KO VC(+) mice; however, the total carnitine levels of both groups were similar in all tissues and serum. In addition, carnitine was produced by incubated liver homogenates from the VC-depleted SMP30/GNL KO mice irrespective of the presence or absence of 1 mM VC. Collectively, these results indicate that VC is not essential for carnitine biosynthesis in vivo.


Assuntos
Deficiência de Ácido Ascórbico/metabolismo , Ácido Ascórbico/fisiologia , Proteínas de Ligação ao Cálcio/fisiologia , Hidrolases de Éster Carboxílico/fisiologia , Carnitina/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Animais , Peso Corporal/efeitos dos fármacos , Proteínas de Ligação ao Cálcio/genética , Hidrolases de Éster Carboxílico/genética , Carnitina/urina , Glutationa/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Camundongos , Camundongos Knockout , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA