Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Leuk Res ; 125: 107001, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36566538

RESUMO

Vitamin C is an essential vitamin that acts as a co-factor for many enzymes involved in epigenetic regulation in humans. Low vitamin C levels in hematopoietic stem cells (HSC) promote self-renewal and vitamin C supplementation retards leukaemogenesis in vitamin C-deficient mouse models. Studies on vitamin C levels in patients with myeloid malignancies are limited. We thus conducted a retrospective analysis on a prospective cohort of patients with myeloid malignancies on whom plasma vitamin C levels were measured serially at diagnosis and during treatment. Baseline characteristics including hematological indices, cytogenetics, and molecular mutations are described in this cohort. Among 64 patients included in our study, 11 patients (17%) had low vitamin C levels. We noted a younger age at diagnosis for patients with myeloid malignancies who had low plasma vitamin C levels. Patients with low plasma vitamin C levels were more likely to have acute myeloid leukemia compared to other myeloid malignancies. Low vitamin C levels were associated with ASXL1 mutations. Our study calls for further multi-institutional studies to understand the relevance of low plasma vitamin C level in myeloid neoplasms, the role of vitamin C deficiency in leukemogenesis, and the potential benefit of vitamin C supplementation.


Assuntos
Deficiência de Ácido Ascórbico , Leucemia Mieloide Aguda , Transtornos Mieloproliferativos , Camundongos , Animais , Humanos , Epigênese Genética , Estudos Prospectivos , Estudos Retrospectivos , Transtornos Mieloproliferativos/genética , Mutação , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Ácido Ascórbico , Deficiência de Ácido Ascórbico/complicações , Deficiência de Ácido Ascórbico/genética
2.
Front Immunol ; 13: 898827, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36248829

RESUMO

Hematopoiesis, a process that results in the differentiation of all blood lineages, is essential throughout life. The production of 1x1012 blood cells per day, including 200x109 erythrocytes, is highly dependent on nutrient consumption. Notably though, the relative requirements for micronutrients during the perinatal period, a critical developmental window for immune cell and erythrocyte differentiation, have not been extensively studied. More specifically, the impact of the vitamin C/ascorbate micronutrient on perinatal as compared to adult hematopoiesis has been difficult to assess in animal models. Even though humans cannot synthesize ascorbate, due to a pseudogenization of the L-gulono-γ-lactone oxidase (GULO) gene, its generation from glucose is an ancestral mammalian trait. Taking advantage of a Gulo-/- mouse model, we show that ascorbic acid deficiency profoundly impacts perinatal hematopoiesis, resulting in a hypocellular bone marrow (BM) with a significant reduction in hematopoietic stem cells, multipotent progenitors, and hematopoietic progenitors. Furthermore, myeloid progenitors exhibited differential sensitivity to vitamin C levels; common myeloid progenitors and megakaryocyte-erythrocyte progenitors were markedly reduced in Gulo-/- pups following vitamin C depletion in the dams, whereas granulocyte-myeloid progenitors were spared, and their frequency was even augmented. Notably, hematopoietic cell subsets were rescued by vitamin C repletion. Consistent with these data, peripheral myeloid cells were maintained in ascorbate-deficient Gulo-/- pups while other lineage-committed hematopoietic cells were decreased. A reduction in B cell numbers was associated with a significantly reduced humoral immune response in ascorbate-depleted Gulo-/- pups but not adult mice. Erythropoiesis was particularly sensitive to vitamin C deprivation during both the perinatal and adult periods, with ascorbate-deficient Gulo-/- pups as well as adult mice exhibiting compensatory splenic differentiation. Furthermore, in the pathological context of hemolytic anemia, vitamin C-deficient adult Gulo-/- mice were not able to sufficiently increase their erythropoietic activity, resulting in a sustained anemia. Thus, vitamin C plays a pivotal role in the maintenance and differentiation of hematopoietic progenitors during the neonatal period and is required throughout life to sustain erythroid differentiation under stress conditions.


Assuntos
Deficiência de Ácido Ascórbico , Mustelidae , Escorbuto , Animais , Ácido Ascórbico/farmacologia , Deficiência de Ácido Ascórbico/genética , Eritropoese , Feminino , Glucose , Humanos , L-Gulonolactona Oxidase/genética , Camundongos , Gravidez , Vitaminas
3.
Nat Commun ; 13(1): 5883, 2022 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-36202795

RESUMO

Vitamin C deficiency disrupts the integrity of connective tissues including bone. For decades this function has been primarily attributed to Vitamin C as a cofactor for collagen maturation. Here, we demonstrate that Vitamin C epigenetically orchestrates osteogenic differentiation and function by modulating chromatin accessibility and priming transcriptional activity. Vitamin C regulates histone demethylation (H3K9me3 and H3K27me3) and promotes TET-mediated 5hmC DNA hydroxymethylation at promoters, enhancers and super-enhancers near bone-specific genes. This epigenetic circuit licenses osteoblastogenesis by permitting the expression of all major pro-osteogenic genes. Osteogenic cell differentiation is strictly and continuously dependent on Vitamin C, whereas Vitamin C is dispensable for adipogenesis. Importantly, deletion of 5hmC-writers, Tet1 and Tet2, in Vitamin C-sufficient murine bone causes severe skeletal defects which mimic bone phenotypes of Vitamin C-insufficient Gulo knockout mice, a model of Vitamin C deficiency and scurvy. Thus, Vitamin C's epigenetic functions are central to osteoblastogenesis and bone formation and may be leveraged to prevent common bone-degenerating conditions.


Assuntos
Deficiência de Ácido Ascórbico , Osteogênese , Animais , Ácido Ascórbico/farmacologia , Deficiência de Ácido Ascórbico/genética , Calcificação Fisiológica/genética , Diferenciação Celular/genética , Cromatina , DNA/metabolismo , Metilação de DNA , Histonas/metabolismo , Camundongos , Osteogênese/genética
4.
Int J Mol Sci ; 23(3)2022 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-35163133

RESUMO

The maternal diet during pregnancy is a key determinant of offspring health. Early studies have linked poor maternal nutrition during gestation with a propensity for the development of chronic conditions in offspring. These conditions include cardiovascular disease, type 2 diabetes and even compromised mental health. While multiple factors may contribute to these outcomes, disturbed epigenetic programming during early development is one potential biological mechanism. The epigenome is programmed primarily in utero, and during this time, the developing fetus is highly susceptible to environmental factors such as nutritional insults. During neurodevelopment, epigenetic programming coordinates the formation of primitive central nervous system structures, neurogenesis, and neuroplasticity. Dysregulated epigenetic programming has been implicated in the aetiology of several neurodevelopmental disorders such as Tatton-Brown-Rahman syndrome. Accordingly, there is great interest in determining how maternal nutrient availability in pregnancy might affect the epigenetic status of offspring, and how such influences may present phenotypically. In recent years, a number of epigenetic enzymes that are active during embryonic development have been found to require vitamin C as a cofactor. These enzymes include the ten-eleven translocation methylcytosine dioxygenases (TETs) and the Jumonji C domain-containing histone lysine demethylases that catalyse the oxidative removal of methyl groups on cytosines and histone lysine residues, respectively. These enzymes are integral to epigenetic regulation and have fundamental roles in cellular differentiation, the maintenance of pluripotency and development. The dependence of these enzymes on vitamin C for optimal catalytic activity illustrates a potentially critical contribution of the nutrient during mammalian development. These insights also highlight a potential risk associated with vitamin C insufficiency during pregnancy. The link between vitamin C insufficiency and development is particularly apparent in the context of neurodevelopment and high vitamin C concentrations in the brain are indicative of important functional requirements in this organ. Accordingly, this review considers the evidence for the potential impact of maternal vitamin C status on neurodevelopmental epigenetics.


Assuntos
Deficiência de Ácido Ascórbico/complicações , Ácido Ascórbico/farmacologia , Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento , Fenômenos Fisiológicos da Nutrição Materna , Transtornos do Neurodesenvolvimento/prevenção & controle , Neurogênese , Animais , Antioxidantes/farmacologia , Deficiência de Ácido Ascórbico/genética , Deficiência de Ácido Ascórbico/patologia , Feminino , Humanos , Transtornos do Neurodesenvolvimento/etiologia , Transtornos do Neurodesenvolvimento/patologia , Gravidez
5.
Br J Nutr ; 128(6): 993-1003, 2022 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-34725010

RESUMO

Vitamin E (α-tocopherol; VE) is known to be regenerated from VE radicals by vitamin C (L-ascorbic acid; VC) in vitro. However, their in vivo interaction in various tissues is still unclear. Therefore, we alternatively examined the in vivo interaction of VC and VE by measurement of their concentrations in various tissues of senescence marker protein-30 (SMP30) knockout (KO) mice as a VC synthesis deficiency model. Male SMP30-KO mice were divided into four groups (VC+/VE+, VC+/VE-, VC-/VE+ and VC-/VE-), fed diets with or without 500 mg/kg VE and given water with or without 1·5 g/l VC ad libitum. Then, VC and VE concentrations in the plasma and various tissues were determined. Further, gene expression levels of transporters associated with VC and VE, such as α-tocopherol transfer protein (α-TTP) and sodium-dependent vitamin C transporters (SVCTs), were examined. These results showed that the VE levels in the VC-depleted (VC-/VE+) group were significantly lower than those in the VC+/VE+ group in the liver and heart; the VC levels in the VE-depleted (VC+/VE-) group were significantly lower than those in the VC+/VE+ group in the kidneys. The α-TTP gene expression in the liver and kidneys was decreased by VC and/or VE depletion. Moreover, SVCT1 gene expression in the liver was decreased by both VC and VE depletion. In conclusion, these results indicate that VC spares VE mainly in the liver and heart and that VE spares VC in the kidneys of SMP30-KO mice. Thus, interaction between VC and VE is likely to be tissue specific.


Assuntos
Deficiência de Ácido Ascórbico , Ácido Ascórbico , Camundongos , Animais , Masculino , Vitamina E , Camundongos Knockout , Proteínas de Ligação ao Cálcio/genética , Deficiência de Ácido Ascórbico/complicações , Deficiência de Ácido Ascórbico/genética , Deficiência de Ácido Ascórbico/metabolismo , Vitaminas
6.
Nutr Metab Cardiovasc Dis ; 31(8): 2398-2406, 2021 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-34088583

RESUMO

BACKGROUND AND AIMS: The impact of vitamin C supplementation on the risk of cardiovascular diseases (CVDs) remains uncertain with inconsistent evidence obtained from observational studies and randomized clinical trials (RCTs). We aimed to assess possible causal associations of vitamin C with major CVD events as well as their risk factors using Mendelian randomization (MR) design. METHODS AND RESULTS: Nine genetic variants associated with vitamin C at genome-wide significance (p < 5 × 10-8) were used as instrumental variables to predict plasma vitamin C levels. The primary outcomes were coronary artery disease (Ncase = 122,733 and Ncontrol = 424,528), atrial fibrillation (Ncase = 60,620 and Ncontrol = 970,216), heart failure (Ncase = 47,309 and Ncontrol = 930,014), and ischemic stroke (Ncase = 40,585 and Ncontrol = 406,111). Several CVD risk factors were also evaluated in secondary analyses. Two-sample MR analyses were performed using the inverse variance weighted method, with several sensitivity analyses. Genetically determined higher levels of plasma vitamin C were not significantly associated with any of the four examined CVD events. Likewise, there is no convincing evidence for the associations between genetically determined vitamin C and CVD risk factors, including higher blood lipids, higher blood pressure, and abnormal body composition. Sensitivity analyses using different analytical approaches yielded consistent results. Additionally, MR assumptions did not seem to be violated. CONCLUSION: This MR study does not support a causal protective role to circulate vitamin C levels on various types of CVD events. In combination with previous RCT results, our findings suggest that vitamin C supplementation to increase circulating vitamin C levels may not help in CVD prevention.


Assuntos
Deficiência de Ácido Ascórbico/genética , Ácido Ascórbico/sangue , Doenças Cardiovasculares/etiologia , Polimorfismo de Nucleotídeo Único , Deficiência de Ácido Ascórbico/sangue , Deficiência de Ácido Ascórbico/complicações , Deficiência de Ácido Ascórbico/diagnóstico , Pressão Sanguínea , Composição Corporal , Doenças Cardiovasculares/sangue , Doenças Cardiovasculares/diagnóstico , Doenças Cardiovasculares/fisiopatologia , Predisposição Genética para Doença , Fatores de Risco de Doenças Cardíacas , Humanos , Lipídeos/sangue , Análise da Randomização Mendeliana , Fenótipo , Medição de Risco
7.
Nutrients ; 13(5)2021 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-34063417

RESUMO

Severe and long-term vitamin C deficiency can lead to fatal scurvy, which is fortunately considered rare today. However, a moderate state of vitamin C (vitC) deficiency (hypovitaminosis C)-defined as a plasma concentration below 23 µM-is estimated to affect up to 10% of the population in the Western world, albeit clinical hallmarks in addition to scurvy have not been linked to vitC deficiency. The brain maintains a high vitC content and uniquely high levels during deficiency, supporting vitC's importance in the brain. Actions include both antioxidant and co-factor functions, rendering vitamin C deficiency likely to affect several targets in the brain, and it could be particularly significant during development where a high cellular metabolism and an immature antioxidant system might increase sensitivity. However, investigations of a non-scorbutic state of vitC deficiency and effects on the developing young brain are scarce. This narrative review provides a comprehensive overview of the complex mechanisms that regulate vitC homeostasis in vivo and in the brain in particular. Functions of vitC in the brain and the potential consequences of deficiency during brain development are highlighted, based primarily on findings from experimental animal models. Perspectives for future investigations of vitC are outlined.


Assuntos
Deficiência de Ácido Ascórbico/sangue , Ácido Ascórbico/metabolismo , Encéfalo/metabolismo , Escorbuto/metabolismo , Animais , Antioxidantes/metabolismo , Ácido Ascórbico/sangue , Ácido Ascórbico/farmacocinética , Deficiência de Ácido Ascórbico/genética , Encéfalo/crescimento & desenvolvimento , Carnitina , Ácidos Graxos Insaturados/metabolismo , Homeostase , Humanos , Camundongos Knockout , Modelos Animais , Neuroglia/metabolismo , Neurônios/metabolismo , Transportadores de Sódio Acoplados à Vitamina C/genética
8.
J Cell Physiol ; 236(8): 5801-5817, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33432597

RESUMO

Glioblastoma multiforme (GBM) is the most common and aggressive brain tumor with a median survival of 14.6 months. GBM is highly resistant to radio- and chemotherapy, and remains without a cure; hence, new treatment strategies are constantly sought. Vitamin C, an essential micronutrient and antioxidant, was initially described as an antitumor molecule; however, several studies have shown that it can promote tumor progression and angiogenesis. Thus, considering the high concentrations of vitamin C present in the brain, our aim was to study the effect of vitamin C deficiency on the progression of GBM using a GBM model generated by the stereotactic injection of human GBM cells (U87-MG or HSVT-C3 cells) in the subventricular zone of guinea pig brain. Initial characterization of U87-MG and HSVT-C3 cells showed that HSVT-C3 are highly proliferative, overexpress p53, and are resistant to ferroptosis. To induce intraperiventricular tumors, animals received control or a vitamin C-deficient diet for 3 weeks, after which histopathological and confocal microscopy analyses were performed. We demonstrated that the vitamin C-deficient condition reduced the glomeruloid vasculature and microglia/macrophage infiltration in U87-MG tumors. Furthermore, tumor size, proliferation, glomeruloid vasculature, microglia/macrophage infiltration, and invasion were reduced in C3 tumors carried by vitamin C-deficient guinea pigs. In conclusion, the effect of the vitamin C deficiency was dependent on the tumor cell used for GBM induction. HSVT-C3 cells, a cell line with stem cell features isolated from a human subventricular GBM, showed higher sensitivity to the deficient condition; however, vitamin C deficiency displayed an antitumor effect in both GBM models analyzed.


Assuntos
Deficiência de Ácido Ascórbico/genética , Proliferação de Células/genética , Glioblastoma/genética , Células-Tronco Neoplásicas/metabolismo , Animais , Ácido Ascórbico/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/genética , Glioblastoma/patologia , Cobaias , Humanos , Células-Tronco Neoplásicas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
9.
J Neurochem ; 157(6): 1759-1773, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32219848

RESUMO

Ascorbic acid (vitamin C) is critical for Schwann cells to myelinate peripheral nerve axons during development and remyelination after injury. However, its exact mechanism remains elusive. Vitamin C is a dietary nutrient that was recently discovered to promote active DNA demethylation. Schwann cell myelination is characterized by global DNA demethylation in vivo and may therefore be regulated by vitamin C. We found that vitamin C induces a massive transcriptomic shift (n = 3,848 genes) in primary cultured Schwann cells while simultaneously producing a global increase in genomic 5-hydroxymethylcytosine (5hmC), a DNA demethylation intermediate which regulates transcription. Vitamin C up-regulates 10 pro-myelinating genes which exhibit elevated 5hmC content in both the promoter and gene body regions of these loci following treatment. Using a mouse model of human vitamin C metabolism, we found that maternal dietary vitamin C deficiency causes peripheral nerve hypomyelination throughout early development in resulting offspring. Additionally, dietary vitamin C intake regulates the expression of myelin-related proteins such as periaxin (PRX) and myelin basic protein (MBP) during development and remyelination after injury in mice. Taken together, these results suggest that vitamin C cooperatively promotes myelination through 1) increased DNA demethylation and transcription of pro-myelinating genes, and 2) its known role in stabilizing collagen helices to form the basal lamina that is necessary for myelination.


Assuntos
Ácido Ascórbico/administração & dosagem , Ácido Ascórbico/metabolismo , Desmetilação do DNA/efeitos dos fármacos , Proteínas da Mielina/metabolismo , Bainha de Mielina/metabolismo , Células de Schwann/fisiologia , Animais , Ácido Ascórbico/genética , Deficiência de Ácido Ascórbico/tratamento farmacológico , Deficiência de Ácido Ascórbico/genética , Deficiência de Ácido Ascórbico/metabolismo , Células Cultivadas , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas da Mielina/genética , Bainha de Mielina/efeitos dos fármacos , Bainha de Mielina/genética , Ratos Endogâmicos F344 , Células de Schwann/efeitos dos fármacos , Neuropatia Ciática/tratamento farmacológico , Neuropatia Ciática/genética , Neuropatia Ciática/metabolismo
10.
Nutrients ; 12(8)2020 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-32751086

RESUMO

Recent research studies have shown that vitamin C (ascorbic acid) may affect bone mineral density and that a deficiency of ascorbic acid leads to the development of osteoporosis. Patients suffering from an inflammatory bowel disease are at a risk of low bone mineral density. It is vital to notice that patients with Crohn's disease and ulcerative colitis also are at risk of vitamin C deficiency which is due to factors such as reduced consumption of fresh vegetables and fruits, i.e., the main sources of ascorbic acid. Additionally, some patients follow diets which may provide an insufficient amount of vitamin C. Moreover, serum vitamin C level also is dependent on genetic factors, such as SLC23A1 and SLC23A2 genes, encoding sodium-dependent vitamin C transporters and GSTM1, GSTP1 and GSTT1 genes which encode glutathione S-transferases. Furthermore, ascorbic acid may modify the composition of gut microbiota which plays a role in the pathogenesis of an inflammatory bowel disease.


Assuntos
Deficiência de Ácido Ascórbico/sangue , Ácido Ascórbico/sangue , Doenças Inflamatórias Intestinais/sangue , Osteoporose/etiologia , Deficiência de Ácido Ascórbico/etiologia , Deficiência de Ácido Ascórbico/genética , Densidade Óssea , Colite Ulcerativa/sangue , Colite Ulcerativa/complicações , Colite Ulcerativa/genética , Doença de Crohn/sangue , Doença de Crohn/complicações , Doença de Crohn/genética , Dieta/efeitos adversos , Feminino , Microbioma Gastrointestinal/fisiologia , Glutationa S-Transferase pi/sangue , Glutationa Transferase/sangue , Humanos , Doenças Inflamatórias Intestinais/complicações , Doenças Inflamatórias Intestinais/genética , Masculino , Osteoporose/genética , Fatores de Risco , Transportadores de Sódio Acoplados à Vitamina C/sangue
11.
Nutrients ; 12(7)2020 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-32630245

RESUMO

A recent review of global vitamin C status has indicated a high prevalence of deficiency, particularly in low- and middle-income countries, as well as in specific subgroups within high-income countries. Here, we provide a narrative review of potential factors influencing vitamin C status globally. The in vivo status of vitamin C is primarily affected by dietary intake and supplement use, with those who supplement having a higher mean status and a lower prevalence of deficiency. Dietary intake can be influenced by cultural aspects such as traditional cooking practices and staple foods, with many staple foods, such as grains, contributing negligible vitamin C to the diet. Environmental factors can also affect vitamin C intake and status; these include geographic region, season, and climate, as well as pollution, the latter partly due to enhanced oxidative stress. Demographic factors such as sex, age, and race are known to affect vitamin C status, as do socioeconomic factors such as deprivation, education and social class, and institutionalization. Various health aspects can affect vitamin C status; these include body weight, pregnancy and lactation, genetic variants, smoking, and disease states, including severe infections as well as various noncommunicable diseases such as cardiovascular disease and cancer. Some of these factors have changed over time; therefore, we also explore if vitamin C status has shown temporal changes. Overall, there are numerous factors that can affect vitamin C status to different extents in various regions of the world. Many of these factors are not taken into consideration during the setting of global dietary intake recommendations for vitamin C.


Assuntos
Deficiência de Ácido Ascórbico/epidemiologia , Ácido Ascórbico , Saúde Global/estatística & dados numéricos , Estado Nutricional , Ácido Ascórbico/administração & dosagem , Deficiência de Ácido Ascórbico/etnologia , Deficiência de Ácido Ascórbico/genética , Peso Corporal , Culinária/métodos , Cultura , Dieta , Suplementos Nutricionais , Meio Ambiente , Feminino , Nível de Saúde , Humanos , Masculino , Necessidades Nutricionais , Gravidez , Fatores Socioeconômicos
12.
Hum Mol Genet ; 29(9): 1476-1488, 2020 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-32307537

RESUMO

Arterial tortuosity syndrome (ATS) is a recessively inherited connective tissue disorder, mainly characterized by tortuosity and aneurysm formation of the major arteries. ATS is caused by loss-of-function mutations in SLC2A10, encoding the facilitative glucose transporter GLUT10. Former studies implicated GLUT10 in the transport of dehydroascorbic acid, the oxidized form of ascorbic acid (AA). Mouse models carrying homozygous Slc2a10 missense mutations did not recapitulate the human phenotype. Since mice, in contrast to humans, are able to intracellularly synthesize AA, we generated a novel ATS mouse model, deficient for Slc2a10 as well as Gulo, which encodes for L-gulonolactone oxidase, an enzyme catalyzing the final step in AA biosynthesis in mouse. Gulo;Slc2a10 double knock-out mice showed mild phenotypic anomalies, which were absent in single knock-out controls. While Gulo;Slc2a10 double knock-out mice did not fully phenocopy human ATS, histological and immunocytochemical analysis revealed compromised extracellular matrix formation. Transforming growth factor beta signaling remained unaltered, while mitochondrial function was compromised in smooth muscle cells derived from Gulo;Slc2a10 double knock-out mice. Altogether, our data add evidence that ATS is an ascorbate compartmentalization disorder, but additional factors underlying the observed phenotype in humans remain to be determined.


Assuntos
Artérias/anormalidades , Deficiência de Ácido Ascórbico/genética , Proteínas Facilitadoras de Transporte de Glucose/genética , Instabilidade Articular/genética , L-Gulonolactona Oxidase/genética , Dermatopatias Genéticas/genética , Malformações Vasculares/genética , Animais , Artérias/metabolismo , Artérias/patologia , Ácido Ascórbico/biossíntese , Ácido Ascórbico/genética , Deficiência de Ácido Ascórbico/metabolismo , Deficiência de Ácido Ascórbico/patologia , Modelos Animais de Doenças , Homozigoto , Humanos , Instabilidade Articular/metabolismo , Instabilidade Articular/patologia , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Respiração/genética , Transdução de Sinais/genética , Dermatopatias Genéticas/metabolismo , Dermatopatias Genéticas/patologia , Malformações Vasculares/metabolismo , Malformações Vasculares/patologia
13.
Biochem Biophys Res Commun ; 511(3): 705-710, 2019 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-30827506

RESUMO

Cav3.2 T-type Ca2+ channels are expressed in the primary afferents and play a pronociceptive role. The activity of Cav3.2 is enhanced by H2S, a gasotransmitter, and suppressed by ascorbic acid (vitamin C) through metal-catalyzed oxidation of the Zn2+-binding His191 in Cav3.2. Since rodents, but not humans, are capable of synthesizing ascorbic acid, the present study examined the role of ascorbic acid in nociceptive processing, using the mice lacking GNL/SMP30, an enzyme essential for ascorbic acid biosynthesis. Intraplantar and intracolonic administration of NaHS, an H2S donor, caused somatic allodynia and referred hyperalgesia, respectively, and repeated treatment with paclitaxel produced neuropathic allodynia in wild-type mice, all of which were suppressed by ascorbic acid or T-type Ca2+ channel blockers. Dietary ascorbic acid restriction caused dramatic decreases in plasma and tissue ascorbic acid levels in GNL/SMP30-knockout, but not wild-type, mice. The ascorbic acid restriction enhanced the somatic and visceral hypersensitivity following intraplantar and intracolonic NaHS, respectively, and paclitaxel-induced neuropathy in GNL/SMP30-knockout mice, while it had no such effect in wild-type mice. Together, our data unveil the critical role of ascorbic acid in regulating somatic and visceral pain sensitivity and support accumulating clinical evidence for the usefulness of ascorbic acid in pain management.


Assuntos
Deficiência de Ácido Ascórbico/metabolismo , Ácido Ascórbico/metabolismo , Proteínas de Ligação ao Cálcio/genética , Hiperalgesia/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Dor Visceral/metabolismo , Animais , Deficiência de Ácido Ascórbico/complicações , Deficiência de Ácido Ascórbico/genética , Canais de Cálcio Tipo T/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Hiperalgesia/etiologia , Hiperalgesia/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Limiar da Dor , Dor Visceral/etiologia , Dor Visceral/genética
14.
J Nutr Sci Vitaminol (Tokyo) ; 64(6): 404-411, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30606963

RESUMO

The aim of this study was to verify the protective effects of ascorbic acid (AsA) against lipopolysaccharide (LPS)-induced sepsis. The study was conducted using osteogenic disorder Shionogi (ODS) rats, which are unable to synthesize AsA. Male ODS rats (6 wk old) were fed either an AsA-free diet (AsA-deficient group), a diet supplemented with 300 mg/kg AsA (control group), or a diet supplemented with 3,000 mg/kg AsA (high-AsA group) for 8 d. On day 8, all the rats were intraperitoneally injected with LPS (15 mg/kg body weight). Forty-eight hours after the injection, the survival rates of the rats in the control (39%) and the high-AsA (61%) groups were significantly higher than that in the AsA-deficient group (5.5%). Next, we measured several inflammatory parameters during 10 h after administering LPS. At 6 h, elevated serum levels of markers for hepatic and systemic injuries were suppressed in rats fed AsA. Similarly, 10 h after LPS injection, the elevation in the serum levels of markers for renal injury were also suppressed proportionally to the amount of AsA in the diet. The elevated serum concentrations of TNFα and IL-1ß by LPS in the AsA-deficient group decreased in groups fed AsA. Hematic TNFα mRNA levels at 6 h after the LPS injection were also lowered by feeding AsA. These results demonstrated that the dietary intake of AsA improved the survival rates and suppressed the inflammatory damage, in a dose-dependent manner, caused during sepsis induced by LPS in ODS rats.


Assuntos
Ácido Ascórbico/uso terapêutico , Suplementos Nutricionais , Inflamação/prevenção & controle , Rim/efeitos dos fármacos , Fígado/efeitos dos fármacos , Sepse/tratamento farmacológico , Vitaminas/uso terapêutico , Animais , Anti-Inflamatórios/administração & dosagem , Anti-Inflamatórios/sangue , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Ácido Ascórbico/administração & dosagem , Ácido Ascórbico/sangue , Ácido Ascórbico/farmacologia , Deficiência de Ácido Ascórbico/sangue , Deficiência de Ácido Ascórbico/complicações , Deficiência de Ácido Ascórbico/genética , Deficiência de Ácido Ascórbico/prevenção & controle , Dieta , Inflamação/induzido quimicamente , Interleucina-1beta/sangue , Nefropatias/sangue , Nefropatias/etiologia , Nefropatias/prevenção & controle , Lipopolissacarídeos , Hepatopatias/sangue , Hepatopatias/etiologia , Hepatopatias/prevenção & controle , Masculino , Estado Nutricional , Osteogênese/fisiologia , Ratos , Ratos Endogâmicos , Sepse/induzido quimicamente , Sepse/complicações , Fator de Necrose Tumoral alfa/sangue , Vitaminas/administração & dosagem , Vitaminas/sangue , Vitaminas/farmacologia
15.
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
16.
In Vivo ; 31(4): 599-607, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28652426

RESUMO

BACKGROUND/AIM: Senescence marker protein-30/gluconolactonase knockout mice (SMP-30/GNL-KO) are a very useful model for clarifying the involvement of vitamin C (VC) in aging-related diseases. In this study, the effects of VC deficiency on skin and hair growth were investigated using SMP-30/GNL-KO mice by RNA sequencing. MATERIALS AND METHODS: SMP-30/GNL-KO mice were given water containing 1.5 g/l VC until up to 8 weeks after birth to maintain a VC concentration in their organs and plasma equivalent to that in wild-type mice. The mice were then divided into two groups: a VC(+) group, where VC was administered, and a VC(-) group, where VC was not administered. Skin samples were collected at 4 and 8 weeks after the treatment. RNA was extracted from each skin sample, followed by cDNA synthesis and RNA-seq. In addition, hair growth was compared between the VC(-) and VC(+) groups after shaving. Skin samples were collected from the shaved area for histological examination by hematoxylin & eosin (HE) staining. RESULTS: RNA-seq revealed that there were 1,736 (FDR<0.001) differentially expressed genes in the VC(-) and VC(+) groups. From the functional analysis of the differentially expressed genes in the VC(-) and VC(+) groups, predicted functionalities including cell death and cytotoxicity increased in the VC(+) group. Furthermore, it was predicted that the difference in hair growth between the VC(-) and VC(+) groups was caused by the expression of genes including keratin-related genes and the Sonic hedgehog gene. It was confirmed that hair growth was significantly promoted; hair growth from hair papilla cells was also confirmed by HE staining of the shaved backs of SMP-30/GNL-KO mice in the VC(+) group. CONCLUSION: RNA-seq of the skin from VC-deficient mice showed the effects of VC deficiency on the expression of genes involved in cell growth and the hair cycle. Visual inspection suggested that changes in the expression of the genes are involved in delaying hair growth in the VC(-) group. Further research on the relationship among VC deficiency, the hair cycle, and skin cell growth may contribute to research on hair restoration and skin aging.


Assuntos
Envelhecimento/genética , Proteínas de Ligação ao Cálcio/genética , Hidrolases de Éster Carboxílico/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Pele/metabolismo , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Ácido Ascórbico/genética , Ácido Ascórbico/metabolismo , Deficiência de Ácido Ascórbico/genética , Deficiência de Ácido Ascórbico/patologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/genética , Cabelo/crescimento & desenvolvimento , Cabelo/metabolismo , Cabelo/patologia , Humanos , Camundongos , Camundongos Knockout , Pele/crescimento & desenvolvimento , Pele/patologia
17.
Oncotarget ; 8(5): 7357-7369, 2017 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-28060768

RESUMO

The AKR1A1 protein is a member of the aldo-keto reductase superfamily that is responsible for the conversion of D-glucuronate to L-gulonate in the ascorbic acid (vitamin C) synthesis pathway. In a pCAG-eGFP transgenic mouse line that was produced by pronuclear microinjection, the integration of the transgene resulted in a 30-kb genomic DNA deletion, including the Akr1A1 gene, and thus caused the knockout (KO) of the Akr1A1 gene and targeting of the eGFP gene. The Akr1A1 KO mice (Akr1A1eGFP/eGFP) exhibited insufficient serum ascorbic acid levels, abnormal bone development and osteoporosis. Using micro-CT analysis, the results showed that the microarchitecture of the 12-week-old Akr1A1eGFP/eGFP mouse femur was shorter in length and exhibited less cortical bone thickness, enlargement of the bone marrow cavity and a complete loss of the trabecular bone in the distal femur. The femoral head and neck of the proximal femur also showed a severe loss of bone mass. Based on the decreased levels of serum osteocalcin and osteoblast activity in the Akr1A1eGFP/eGFP mice, the osteoporosis might be caused by impaired bone formation. In addition, administration of ascorbic acid to the Akr1A1eGFP/eGFP mice significantly prevented the condition of osteoporotic femurs and increased bone formation. Therefore, through ascorbic acid administration, the Akr1A1 KO mice exhibited controllable osteoporosis and may serve as a novel model for osteoporotic research.


Assuntos
Aldeído Redutase/genética , Deficiência de Ácido Ascórbico/genética , Fêmur/patologia , Técnicas de Inativação de Genes , Osteogênese , Osteoporose/genética , Aldeído Redutase/deficiência , Animais , Ácido Ascórbico/administração & dosagem , Ácido Ascórbico/sangue , Deficiência de Ácido Ascórbico/enzimologia , Deficiência de Ácido Ascórbico/patologia , Deficiência de Ácido Ascórbico/prevenção & controle , Modelos Animais de Doenças , Feminino , Fêmur/diagnóstico por imagem , Fêmur/enzimologia , Predisposição Genética para Doença , Camundongos Knockout , Osteoblastos/enzimologia , Osteoblastos/patologia , Osteocalcina/sangue , Osteoporose/enzimologia , Osteoporose/patologia , Osteoporose/prevenção & controle , Fenótipo , Fatores de Tempo , Microtomografia por Raio-X
18.
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
19.
Arch Soc Esp Oftalmol ; 90(10): 458-66, 2015 Oct.
Artigo em Inglês, Espanhol | MEDLINE | ID: mdl-26188624

RESUMO

PURPOSE: Climatic droplets keratopathy (CDK) is closely associated with superficial corneal erosions and lack of protective mechanisms against the harmful effects of ultraviolet radiation (UVR) during a prolonged period of time. One of the difficulties in studying the pathogenic mechanisms involved in this human disease is the lack of an experimental animal model. In this paper, a study is conducted on the effects of 4 types of lasers at various powers and time conditions on the normal guinea pig corneas in order to select only one laser condition that reversibly injures the epithelium and superficial stroma, without leaving scarring. METHODS: Damage was induced in the cornea of Guinea pigs using different powers and exposure times of 4 types of laser: argon, CO2, diode and Nd-Yag, and any injuries were evaluated by biomicroscopy (BM) and optical microscopy. Corneas from other normal animals were exposed to argon laser (350 mW, 0.3s, 50 µm of diameter), and the induced alterations were studied at different times using BM, optical coherence tomography (OCT) and transmission electron microscopy (TEM). RESULTS: Only argon laser at 350 mW, 0.3s, 50 µm of diameter produced epithelium and superficial stroma lesions. Some leukomas were observed by BM, and they disappeared by day 15. Corneal thickness measured by OCT decreased in the eyes treated with argon laser during the first week. Using TEM, different ultra structural alterations in corneal epithelium and stroma were observed during the early days, which disappeared by day 15. CONCLUSIONS: It was possible to develop reproducible corneal epithelium and anterior stroma injuries using Argon laser at 350 mW, 0.3s, 50 µm of diameter. In vivo and in vitro studies showed that injured corneas with these laser conditions did not leave irreversible microscopic or ultra structural alterations. This protocol of corneal erosion combined with exposure to UVR and partial deficiency of ascorbate in the diets of the animals for an extended period of time has been used in order to try to develop an experimental model of CDK.


Assuntos
Lesões da Córnea/etiologia , Opacidade da Córnea/etiologia , Modelos Animais de Doenças , Cobaias , Lasers/efeitos adversos , Animais , Deficiência de Ácido Ascórbico/complicações , Deficiência de Ácido Ascórbico/genética , Córnea/efeitos da radiação , Córnea/ultraestrutura , Opacidade da Córnea/complicações , Opacidade da Córnea/imunologia , Relação Dose-Resposta à Radiação , Exposição Ambiental , Feminino , Cobaias/genética , Humanos , Lasers de Gás/efeitos adversos , Material Particulado/efeitos adversos , Reprodutibilidade dos Testes , Lâmpada de Fenda , Raios Ultravioleta/efeitos adversos
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...