Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int Ophthalmol ; 41(4): 1455-1465, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33481153

RESUMO

PURPOSE: Vitamin A deficiency (VAD) is associated with chalazion in young children. However, the underlying molecular mechanism remains unclear. In the present study, transcriptome data from rat meibomian glands (MGs) were analyzed to reveal specific molecular responses to VAD. METHODS: Total RNA was extracted and purified for library preparation and transcriptome sequencing. Differentially expressed genes (DEGs) between vitamin A normal (VAN) and VAD rats were analyzed using DESeq software. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses of DEGs were performed using the GO seq R package and KOBAS software. Real-time quantitative reverse transcription polymerase chain reaction was used to validate the RNA sequencing results. RESULTS: The number of DEGs in the VAD group compared to the VAN group was 3129 (1531 upregulated and 1598 downregulated) in the rat MGs. VAD upregulated a large number of lipid metabolism-related genes. GO analysis showed that the most enriched and meaningful terms were related to lipid metabolism (e.g., "oxidation-reduction process, GO: 0,055,114," "lipid metabolic process, GO: 000,662"). KEGG pathway analysis showed that most of the enriched signaling pathways were involved in lipid metabolism, including the PPAR signaling pathway associated with retinoic acid (RA)-mediated nuclear receptors. CONCLUSION: These findings demonstrate that VAD regulates the expression of numerous genes in the rat MG and that many of these genes are involved in lipid metabolic pathways.


Assuntos
Transcriptoma , Deficiência de Vitamina A , Animais , Perfilação da Expressão Gênica , Glândulas Tarsais , Ratos , Transdução de Sinais , Deficiência de Vitamina A/genética
2.
Am J Physiol Gastrointest Liver Physiol ; 318(5): G955-G965, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32200644

RESUMO

Functional fermentable fibers are considered essential for a healthy diet. Recently, we demonstrated that gut microbiota dysbiotic mice fed an inulin-containing diet (ICD) developed hepatocellular carcinoma (HCC) within 6 mo. In particular, a subset of Toll-like receptor 5-deficient (T5KO) mice prone to HCC exhibited rapid onset of hyperbilirubinemia (HB) and cholemia; these symptoms provide rationale that ICD induces cholestasis. Our objective in the present study was to determine whether inulin-fed T5KO-HB mice exhibit other known consequences of cholestasis, including essential fatty acid and fat-soluble vitamin deficiencies. Here, we measured hepatic fatty acids and serum vitamin A and D levels from wild-type (WT), T5KO low bilirubin (LB) and T5KO-HB mice fed ICD for 4 wk. Additionally, hepatic RNAseq and proteomics were performed to ascertain other metabolic alterations. Compared with WT and T5KO-LB, T5KO-HB mice exhibited steatorrhea, i.e., ~50% increase in fecal lipids. This could contribute to the significant reduction of linoleate in hepatic neutral lipids in T5KO-HB mice. Additionally, serum vitamins A and D were ~50% reduced in T5KO-HB mice, which was associated with metabolic compromises. Overall, our study highlights that fermentable fiber-induced cholestasis is further characterized by depletion of macro-and micronutrients.NEW & NOTEWORTHY Feeding a dietary, fermentable fiber diet to a subset of Toll-like receptor 5 deficient (T5KO) mice induces early onset hyperbilirubinemia and cholemia that later manifests to hepatocellular carcinoma (HCC). Our study highlights that fermentable fiber-induced cholestasis is characterized with modest macro- and micronutrient deficiencies that may further contribute to hepatic biliary disease. Compared with chemical induction, immunization, surgery, or genetic manipulation, these findings provide a novel approach to study the cholestatic subtype of HCC.


Assuntos
Fibras na Dieta , Fígado Gorduroso/metabolismo , Absorção Intestinal , Inulina , Metabolismo dos Lipídeos , Fígado/metabolismo , Síndromes de Malabsorção/metabolismo , Receptor 5 Toll-Like/deficiência , Deficiência de Vitamina A/metabolismo , Deficiência de Vitamina D/metabolismo , Animais , Ácidos e Sais Biliares/metabolismo , Colestase/genética , Colestase/metabolismo , Colestase/patologia , Modelos Animais de Doenças , Fígado Gorduroso/genética , Fígado Gorduroso/patologia , Fermentação , Fígado/patologia , Síndromes de Malabsorção/genética , Síndromes de Malabsorção/patologia , Masculino , Camundongos Knockout , Receptor 5 Toll-Like/genética , Deficiência de Vitamina A/genética , Deficiência de Vitamina D/genética
3.
Int J Mol Sci ; 21(15)2020 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-32759702

RESUMO

Vitamin A is an important regulator of immune protection, but it is often overlooked in studies of infectious disease. Vitamin A binds an array of nuclear receptors (e.g., retinoic acid receptor, peroxisome proliferator-activated receptor, retinoid X receptor) and influences the barrier and immune cells responsible for pathogen control. Children and adults in developed and developing countries are often vitamin A-deficient or insufficient, characteristics associated with poor health outcomes. To gain a better understanding of the protective mechanisms influenced by vitamin A, we examined immune factors and epithelial barriers in vitamin A deficient (VAD) mice, vitamin D deficient (VDD) mice, double deficient (VAD+VDD) mice, and mice on a vitamin-replete diet (controls). Some mice received insults, including intraperitoneal injections with complete and incomplete Freund's adjuvant (emulsified with PBS alone or with DNA + Fus-1 peptide) or intranasal inoculations with Sendai virus (SeV). Both before and after insults, the VAD and VAD+VDD mice exhibited abnormal serum immunoglobulin isotypes (e.g., elevated IgG2b levels, particularly in males) and cytokine/chemokine patterns (e.g., elevated eotaxin). Even without insult, when the VAD and VAD+VDD mice reached 3-6 months of age, they frequently exhibited opportunistic ascending bacterial urinary tract infections. There were high frequencies of nephropathy (squamous cell hyperplasia of the renal urothelium, renal scarring, and ascending pyelonephritis) and death in the VAD and VAD+VDD mice. When younger VAD mice were infected with SeV, the predominant lesion was squamous cell metaplasia of respiratory epithelium in lungs and bronchioles. Results highlight a critical role for vitamin A in the maintenance of healthy immune responses, epithelial cell integrity, and pathogen control.


Assuntos
Deficiência de Vitamina A/genética , Vitamina A/genética , Deficiência de Vitamina D/genética , Vitamina D/genética , Animais , Doenças Transmissíveis/genética , Doenças Transmissíveis/imunologia , Doenças Transmissíveis/metabolismo , Morte , Modelos Animais de Doenças , Humanos , Imunoglobulinas/genética , Imunoglobulinas/imunologia , Camundongos , Camundongos Knockout , Neoplasias de Células Escamosas/genética , Neoplasias de Células Escamosas/imunologia , Neoplasias de Células Escamosas/metabolismo , Proteínas Supressoras de Tumor/genética , Vitamina A/metabolismo , Deficiência de Vitamina A/imunologia , Deficiência de Vitamina A/metabolismo , Vitamina D/metabolismo , Deficiência de Vitamina D/imunologia , Deficiência de Vitamina D/metabolismo
4.
J Cell Mol Med ; 23(7): 4582-4591, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31044535

RESUMO

Congenital scoliosis (CS) is the result of anomalous vertebrae development, but the pathogenesis of CS remains unclear. Long non-coding RNAs (lncRNAs) have been implicated in embryo development, but their role in CS remains unknown. In this study, we investigated the role and mechanisms of a specific lncRNA, SULT1C2A, in somitogenesis in a rat model of vitamin A deficiency (VAD)-induced CS. Bioinformatics analysis and quantitative real-time PCR (qRT-PCR) indicated that SULT1C2A expression was down-regulated in VAD group, accompanied by increased expression of rno-miR-466c-5p but decreased expression of Foxo4 and somitogenesis-related genes such as Pax1, Nkx3-2 and Sox9 on gestational day (GD) 9. Luciferase reporter and small interfering RNA (siRNA) assays showed that SULT1C2A functioned as a competing endogenous RNA to inhibit rno-miR-466c-5p expression by direct binding, and rno-miR-466c-5p inhibited Foxo4 expression by binding to its 3' untranslated region (UTR). The spatiotemporal expression of SULT1C2A, rno-miR-466c-5p and Foxo4 axis was dynamically altered on GDs 3, 8, 11, 15 and 21 as detected by qRT-PCR and northern blot analyses, with parallel changes in Protein kinase B (AKT) phosphorylation and PI3K expression. Taken together, our findings indicate that SULT1C2A enhanced Foxo4 expression by negatively modulating rno-miR-466c-5p expression via the PI3K-ATK signalling pathway in the rat model of VAD-CS. Thus, SULT1C2A may be a potential target for treating CS.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Longo não Codificante/metabolismo , Escoliose/congênito , Escoliose/genética , Transdução de Sinais , Regiões 3' não Traduzidas/genética , Animais , Sequência de Bases , Regulação para Baixo/genética , Embrião de Mamíferos/metabolismo , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Células HEK293 , Humanos , Luciferases/metabolismo , MicroRNAs , Modelos Biológicos , Organogênese/genética , RNA Longo não Codificante/genética , Ratos Sprague-Dawley , Somitos/embriologia , Deficiência de Vitamina A/embriologia , Deficiência de Vitamina A/genética
5.
Indian J Med Res ; 150(6): 620-629, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-32048626

RESUMO

Background & objectives: Stearoyl-CoA desaturase 1 (SCD1) is a key lipogenic enzyme responsible for endogenous synthesis of monounsaturated fatty acids (MUFA) and plays a key role in various pathophysiology, including fatty liver diseases. In this experimental study the impact of vitamin A deficiency was assessed on SCD1 regulation in relation to kidney biology, under high fructose (HFr) diet-fed condition in rats. Methods: Forty male weanling (21 day old) Wistar rats were divided into four groups control, vitamin A-deficient (VAD), HFr, VAD with HFr consisting of eight rats each, except 16 for the VAD group. The groups received one of the following diets: control, VAD, HFr and VAD with HFr for 16 wk, except half of the VAD diet-fed rats were shifted to HFr diet, after eight week period. Results: Feeding of VAD diet (alone or with HFr) significantly reduced the kidney retinol (0.51, 0.44 µg/g vs. 2.1 µg/g; P < 0.05), while increased oleic (C18:1) and total MUFA levels (23.3, 22.2% and 27.3, 25.4% respectively vs. 14.7 and 16.6%; P < 0.05) without affecting the SCD1, both at protein and mRNA levels, when compared with HFr. Comparable, immunohistological staining for SCD1 was observed in the distal convoluted tubules. Despite an increase in MUFA, morphology, triglyceride content and markers of kidney function were not affected by VAD diet feeding. Interpretation & conclusions: Feeding of VAD diet either alone or under HFr condition increased the kidney oleic acid (C18:1) levels and thus total MUFA, which corroborated with elevated SCD1 activity index, without affecting its expression status. However, these changes did not alter the kidney morphology and function. Thus, nutrient-gene regulation in kidney biology seems to be divergent.


Assuntos
Rim/metabolismo , Ácido Oleico/metabolismo , Estearoil-CoA Dessaturase/genética , Deficiência de Vitamina A/metabolismo , Animais , Dieta/efeitos adversos , Ácidos Graxos Monoinsaturados/metabolismo , Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Frutose/efeitos adversos , Frutose/farmacologia , Regulação Enzimológica da Expressão Gênica/genética , Humanos , Rim/patologia , Ácido Oleico/genética , Ratos , Vitamina A/genética , Vitamina A/metabolismo , Deficiência de Vitamina A/genética
6.
J Cell Physiol ; 233(1): 607-616, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28322443

RESUMO

Activation of hepatic stellate cells (HSCs) is the effector factor of hepatic fibrosis and hepatocellular carcinoma (HCC) development. Accumulating evidence suggests that retinoic acids (RAs), derivatives of vitamin A, contribute to prevention of liver fibrosis and carcinogenesis, however, regulatory mechanisms of RAs still remain exclusive. To elucidate RA signaling pathway, we previously performed a genome-wide screening of RA-responsive genes by in silico analysis of RA-response elements, and identified 26 RA-responsive genes. We found that thioredoxin interacting protein (TXNIP), which inhibits antioxidant activity of thioredoxin (TRX), was downregulated by all-trans retinoic acid (ATRA). In the present study, we demonstrate that ATRA ameliorates activation of HSCs through TXNIP suppression. HSC activation was attenuated by TXNIP downregulation, whereas potentiated by TXNIP upregulation, indicating that TXNIP plays a crucial role in activation of HSCs. Notably, we showed that TXNIP-mediated HSC activation was suppressed by antioxidant N-acetylcysteine. In addition, ATRA treatment or downregulation of TXNIP clearly declined oxidative stress levels in activated HSCs. These data suggest that ATRA plays a key role in inhibition of HSC activation via suppressing TXNIP expression, which reduces oxidative stress levels.


Assuntos
Antioxidantes/farmacologia , Proteínas de Transporte/metabolismo , Células Estreladas do Fígado/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Tiorredoxinas/metabolismo , Tretinoína/farmacologia , Deficiência de Vitamina A/prevenção & controle , Animais , Proteínas de Transporte/genética , Linhagem Celular , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica , Células Estreladas do Fígado/metabolismo , Células Estreladas do Fígado/patologia , Humanos , Camundongos Endogâmicos C57BL , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Tiorredoxinas/genética , Transfecção , Deficiência de Vitamina A/genética , Deficiência de Vitamina A/metabolismo , Deficiência de Vitamina A/patologia
7.
Biochim Biophys Acta ; 1861(3): 156-65, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26597784

RESUMO

BACKGROUND/AIMS: Vitamin A and its metabolites are known to regulate lipid metabolism. However so far, no study has assessed, whether vitamin A deficiency per se aggravates or attenuates the development of non-alcoholic fatty liver disease (NAFLD). Therefore, here, we tested the impact of vitamin A deficiency on the development of NAFLD. METHODS: Male weanling Wistar rats were fed one of the following diets; control, vitamin A-deficient (VAD), high fructose (HFr) and VAD with HFr (VADHFr) of AIN93G composition, for 16weeks, except half of the VAD diet-fed rats were shifted to HFr diet (VAD(s)HFr), at the end of 8(th) week. RESULTS: Animals fed on VAD diet with HFr displayed hypotriglyceridemia (33.5mg/dL) with attenuated hepatic triglyceride accumulation (8.2mg/g), compared with HFr diet (89.5mg/dL and 20.6mg/g respectively). These changes could be partly explained by the decreased activity of glycerol 3-phosphate dehydrogenase (GPDH) and the down-regulation of stearoyl CoA desaturase 1 (SCD1), both at gene and protein levels, the key determinants of triglyceride biosynthesis. On the other hand, n-3 long chain polyunsaturated fatty acid, docosahexaenoic acid and its active metabolite; resolvin D1 (RvD1) levels were elevated in the liver and plasma of VAD diet-fed groups, which was negatively associated with triglyceride levels. All these factors confer vitamin A deficiency-mediated protection against the development of hepatic steatosis, which was also evident from the group shifted from VAD to HFr diet. CONCLUSIONS: Vitamin A deficiency attenuates high fructose-induced hepatic steatosis, by regulating triglyceride synthesis, possibly through GPDH, SCD1 and RvD1.


Assuntos
Ácidos Docosa-Hexaenoicos/metabolismo , Frutose , Fígado/metabolismo , Hepatopatia Gordurosa não Alcoólica/prevenção & controle , Triglicerídeos/biossíntese , Deficiência de Vitamina A/metabolismo , Adiposidade , Animais , Modelos Animais de Doenças , Regulação para Baixo , Glicerolfosfato Desidrogenase/genética , Glicerolfosfato Desidrogenase/metabolismo , Gordura Intra-Abdominal/metabolismo , Gordura Intra-Abdominal/fisiopatologia , Fígado/patologia , Masculino , Hepatopatia Gordurosa não Alcoólica/induzido quimicamente , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/fisiopatologia , Ratos Wistar , Estearoil-CoA Dessaturase/genética , Estearoil-CoA Dessaturase/metabolismo , Regulação para Cima , Deficiência de Vitamina A/genética , Deficiência de Vitamina A/fisiopatologia , Redução de Peso
8.
Cell Immunol ; 322: 49-55, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29042055

RESUMO

The present study extends an earlier report that retinoic acid (RA) down-regulates IgE Ab synthesis in vitro. Here, we show the suppressive activity of RA on IgE production in vivo and its underlying mechanisms. We found that RA down-regulated IgE class switching recombination (CSR) mainly through RA receptor α (RARα). Additionally, RA inhibited histone acetylation of germ-line ε (GL ε) promoter, leading to suppression of IgE CSR. Consistently, serum IgE levels were substantially elevated in vitamin A-deficient (VAD) mice and this was more dramatic in VAD-lecithin:retinol acyltransferase deficient (LRAT-/-) mice. Further, serum mouse mast cell protease-1 (mMCP-1) level was elevated while frequency of intestinal regulatory T cells (Tregs) were diminished in VAD LRAT-/- mice, reflecting that deprivation of RA leads to allergic immune response. Taken together, our results reveal that RA has an IgE-repressive activity in vivo, which may ameliorate IgE-mediated allergic disease.


Assuntos
Switching de Imunoglobulina/efeitos dos fármacos , Imunoglobulina E/biossíntese , Interleucina-4/metabolismo , Tretinoína/farmacologia , Deficiência de Vitamina A/sangue , Aciltransferases/deficiência , Aciltransferases/genética , Animais , Quimases/metabolismo , Hipersensibilidade Alimentar/tratamento farmacológico , Hipersensibilidade Alimentar/imunologia , Switching de Imunoglobulina/imunologia , Imunoglobulina E/sangue , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor alfa de Ácido Retinoico/imunologia , Linfócitos T Reguladores/imunologia , Vitamina A/genética , Deficiência de Vitamina A/genética
9.
Subcell Biochem ; 79: 311-43, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27485228

RESUMO

Carotenoids are essential components for human nutrition and health, mainly due to their antioxidant and pro-vitamin A activity. Foods with enhanced carotenoid content and composition are essential to ensure carotenoid feasibility in malnourished population of many countries around the world, which is critical to alleviate vitamin A deficiency and other health-related disorders. The pathway of carotenoid biosynthesis is currently well understood, key steps of the pathways in different plant species have been characterized and the corresponding genes identified, as well as other regulatory elements. This enables the manipulation and improvement of carotenoid content and composition in order to control the nutritional value of a number of agronomical important staple crops. Biotechnological and genetic engineering-based strategies to manipulate carotenoid metabolism have been successfully implemented in many crops, with Golden rice as the most relevant example of ß-carotene improvement in one of the more widely consumed foods. Conventional breeding strategies have been also adopted in the bio-fortification of carotenoid in staple foods that are highly consumed in developing countries, including maize, cassava and sweet potatoes, to alleviate nutrition-related problems. The objective of the chapter is to summarize major breakthroughs and advances in the enhancement of carotenoid content and composition in agronomical and nutritional important crops, with special emphasis to their potential impact and benefits in human nutrition and health.


Assuntos
Antioxidantes/metabolismo , Carotenoides/metabolismo , Deficiência de Vitamina A/dietoterapia , Vitamina A/biossíntese , Carotenoides/biossíntese , Carotenoides/genética , Humanos , Valor Nutritivo , Oryza/genética , Plantas Geneticamente Modificadas/genética , Plantas Geneticamente Modificadas/metabolismo , Vitamina A/metabolismo , Deficiência de Vitamina A/genética , Zea mays/genética , Zea mays/metabolismo
10.
Hum Mol Genet ; 23(20): 5402-17, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-24852372

RESUMO

Vitamin A must be adequately distributed within the body to maintain the functions of retinoids in the periphery and chromophore production in the eyes. Blood transport of the lipophilic vitamin is mediated by the retinol-binding protein, RBP4. Biochemical evidence suggests that cellular uptake of vitamin A from RBP4 is facilitated by a membrane receptor. This receptor, identified as the Stimulated by retinoic acid gene 6 (Stra6) gene product, is highly expressed in epithelia that constitute blood-tissue barriers. Here we established a Stra6 knockout mouse model to analyze the metabolic basis of vitamin A homeostasis in peripheral tissues. These mice were viable when bred on diets replete in vitamin A, but evidenced markedly reduced levels of ocular retinoids. Ophthalmic imaging and histology revealed malformations in the choroid and retinal pigmented epithelium, early cone photoreceptor cell death, and reduced lengths of rod outer segments. Similar to the blood-retina barrier in the RPE, vitamin A transport through the blood-cerebrospinal fluid barrier in the brain's choroid plexus was impaired. Notably, treatment with pharmacological doses of vitamin A restored vitamin A transport across these barriers and rescued the vision of Stra6(-/-) mice. Furthermore, under conditions mimicking vitamin A excess and deficiency, our analyses revealed that STRA6-mediated vitamin A uptake is a regulated process mandatory for ocular vitamin A uptake when RBP4 constitutes the only transport mode in vitamin A deficiency. These findings identifying STRA6 as a bona fide vitamin A transporter have important implications for disease states associated with impaired blood vitamin A homeostasis.


Assuntos
Olho/metabolismo , Olho/patologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Vitamina A/farmacocinética , Animais , Encéfalo/metabolismo , Plexo Corióideo/metabolismo , Olho/ultraestrutura , Homeostase , Camundongos , Camundongos Knockout , Mutação , Transporte Proteico , Retinoides/metabolismo , Proteínas Plasmáticas de Ligação ao Retinol/metabolismo , Vitamina A/uso terapêutico , Deficiência de Vitamina A/dietoterapia , Deficiência de Vitamina A/genética
11.
Am J Physiol Heart Circ Physiol ; 310(11): H1773-89, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27084391

RESUMO

To determine whether hepatic depletion of vitamin A (VA) stores has an effect on the postnatal heart, studies were carried out with mice lacking liver retinyl ester stores fed either a VA-sufficient (LRVAS) or VA-deficient (LRVAD) diet (to deplete circulating retinol and extrahepatic stores of retinyl esters). There were no observable differences in the weights or gross morphology of hearts from LRVAS or LRVAD mice relative to sex-matched, age-matched, and genetically matched wild-type (WT) controls fed the VAS diet (WTVAS), but changes in the transcription of functionally relevant genes were consistent with a state of VAD in LRVAS and LRVAD ventricles. In silico analysis revealed that 58/67 differentially expressed transcripts identified in a microarray screen are products of genes that have DNA retinoic acid response elements. Flow cytometric analysis revealed a significant and cell-specific increase in the number of proliferating Sca-1 cardiac progenitor cells in LRVAS animals relative to WTVAS controls. Before myocardial infarction, LRVAS and WTVAS mice had similar cardiac systolic function and structure, as measured by echocardiography, but, unexpectedly, repeat echocardiography demonstrated that LRVAS mice had less adverse remodeling by 1 wk after myocardial infarction. Overall, the results demonstrate that the adult heart is responsive to retinoids, and, most notably, reducing hepatic VA stores (while maintaining circulating levels of VA) impacts ventricular gene expression profiles, progenitor cell numbers, and response to injury.


Assuntos
Fígado/metabolismo , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Receptores do Ácido Retinoico/metabolismo , Retinoides/metabolismo , Deficiência de Vitamina A/metabolismo , Aciltransferases/genética , Aciltransferases/metabolismo , Animais , Ecocardiografia , Coração/fisiopatologia , Camundongos , Camundongos Knockout , Infarto do Miocárdio/fisiopatologia , Deficiência de Vitamina A/genética , Deficiência de Vitamina A/fisiopatologia , Receptor gama de Ácido Retinoico
12.
J Nutr ; 145(8): 1740-7, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26063065

RESUMO

BACKGROUND: The bioavailability of ß-carotene, the main dietary provitamin A carotenoid, varies among individuals. It is not known whether this variability can affect long-term ß-carotene, and hence vitamin A, status. OBJECTIVES: We hypothesized that variations in genes involved in ß-carotene absorption and postprandial metabolism could at least partially explain the high interindividual variability in ß-carotene bioavailability. Thus, the main objectives of this study were to identify associated single-nucleotide polymorphisms (SNPs), and to estimate whether populations with different allele frequencies at these SNPs could have different abilities to absorb provitamin A carotenoids. METHODS: In this single-group design, 33 healthy, nonobese adult men were genotyped with the use of whole-genome microarrays. After an overnight fast, they consumed a test meal containing 100 g tomato puree providing 0.4 mg ß-carotene. The postprandial plasma chylomicron ß-carotene concentration was then measured at regular time intervals over 8 h. Partial least squares (PLS) regression was used to identify the best combination of SNPs in or near candidate genes (54 genes representing 2172 SNPs) that was associated with the postprandial chylomicron ß-carotene response (incremental ß-carotene area-under-the-curve concentration over 8 h in chylomicrons). RESULTS: The postprandial chylomicron ß-carotene response was highly variable (CV = 105%) and was positively correlated with the fasting plasma ß-carotene concentration (r = 0.78; P < 0.0001). A significant (P = 6.54 × 10(-3)) multivalidated PLS regression model, which included 25 SNPs in 12 genes, explained 69% of the variance in the postprandial chylomicron ß-carotene response, i.e., ß-carotene bioavailability. CONCLUSIONS: Interindividual variability in ß-carotene bioavailability appears to be partially modulated by a combination of SNPs in 12 genes. This variability likely affects the long-term blood ß-carotene status. A theoretic calculation of ß-carotene bioavailability in 4 populations of the international HapMap project suggests that populations with different allele frequencies in these SNPs might exhibit a different ability to absorb dietary ß-carotene. This trial was registered at clinicaltrials.gov as NCT02100774.


Assuntos
Estudos de Associação Genética , Polimorfismo de Nucleotídeo Único , beta Caroteno/metabolismo , Adulto , Disponibilidade Biológica , Quilomícrons/sangue , Regulação da Expressão Gênica , Predisposição Genética para Doença , Humanos , Masculino , Deficiência de Vitamina A/genética
13.
Molecules ; 21(1): E46, 2015 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-26729079

RESUMO

This study was aimed to investigate whether vitamin A deficiency could alter P-GP expression and function in tissues of rats and whether such effects affected the drug distribution in vivo of vitamin A-deficient rats. We induced vitamin A-deficient rats by giving them a vitamin A-free diet for 12 weeks. Then, Abcb1/P-GP expression was evaluated by qRT-PCR and Western blot. qRT-PCR analysis revealed that Abcb1a mRNA levels were increased in hippocampus and liver. In kidney, it only showed an upward trend. Abcb1b mRNA levels were increased in hippocampus, but decreased in cerebral cortex, liver and kidney. Western blot results were in good accordance with the alterations of Abcb1b mRNA levels. P-GP function was investigated through tissue distribution and body fluid excretion of rhodamine 123 (Rho123), and the results proclaimed that P-GP activities were also in good accordance with P-GP expression in cerebral cortex, liver and kidney. The change of drug distribution was also investigated through the tissue distribution of vincristine, and the results showed a significantly upward trend in all indicated tissues of vitamin A-deficient rats. In conclusion, vitamin A deficiency may alter Abcb1/P-GP expression and function in rat tissues, and the alterations may increase drug activity/toxicity through the increase of tissue accumulation.


Assuntos
Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Vincristina/toxicidade , Deficiência de Vitamina A/induzido quimicamente , Animais , Córtex Cerebral/metabolismo , Modelos Animais de Doenças , Hipocampo/metabolismo , Rim/metabolismo , Fígado/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Vincristina/farmacocinética , Deficiência de Vitamina A/genética , Deficiência de Vitamina A/metabolismo
14.
J Biol Chem ; 288(47): 34081-34096, 2013 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-24106281

RESUMO

Mammalian genomes encode two provitamin A-converting enzymes as follows: the ß-carotene-15,15'-oxygenase (BCO1) and the ß-carotene-9',10'-oxygenase (BCO2). Symmetric cleavage by BCO1 yields retinoids (ß-15'-apocarotenoids, C20), whereas eccentric cleavage by BCO2 produces long-chain (>C20) apocarotenoids. Here, we used genetic and biochemical approaches to clarify the contribution of these enzymes to provitamin A metabolism. We subjected wild type, Bco1(-/-), Bco2(-/-), and Bco1(-/-)Bco2(-/-) double knock-out mice to a controlled diet providing ß-carotene as the sole source for apocarotenoid production. This study revealed that BCO1 is critical for retinoid homeostasis. Genetic disruption of BCO1 resulted in ß-carotene accumulation and vitamin A deficiency accompanied by a BCO2-dependent production of minor amounts of ß-apo-10'-carotenol (APO10ol). We found that APO10ol can be esterified and transported by the same proteins as vitamin A but with a lower affinity and slower reaction kinetics. In wild type mice, APO10ol was converted to retinoids by BCO1. We also show that a stepwise cleavage by BCO2 and BCO1 with APO10ol as an intermediate could provide a mechanism to tailor asymmetric carotenoids such as ß-cryptoxanthin for vitamin A production. In conclusion, our study provides evidence that mammals employ both carotenoid oxygenases to synthesize retinoids from provitamin A carotenoids.


Assuntos
Carotenoides/metabolismo , Dioxigenases/metabolismo , Vitamina A/metabolismo , beta-Caroteno 15,15'-Mono-Oxigenase/metabolismo , Animais , Carotenoides/genética , Criptoxantinas , Dioxigenases/genética , Células Hep G2 , Humanos , Camundongos , Camundongos Knockout , Vitamina A/genética , Deficiência de Vitamina A/enzimologia , Deficiência de Vitamina A/genética , Xantofilas/genética , Xantofilas/metabolismo , beta Caroteno/genética , beta Caroteno/metabolismo , beta-Caroteno 15,15'-Mono-Oxigenase/genética
15.
Development ; 137(4): 631-40, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20110328

RESUMO

Hirschsprung disease is a serious disorder of enteric nervous system (ENS) development caused by the failure of ENS precursor migration into the distal bowel. We now demonstrate that retinoic acid (RA) is crucial for GDNF-induced ENS precursor migration, cell polarization and lamellipodia formation, and that vitamin A depletion causes distal bowel aganglionosis in serum retinol-binding-protein-deficient (Rbp4(-/-)) mice. Ret heterozygosity increases the incidence and severity of distal bowel aganglionosis induced by vitamin A deficiency in Rbp4(-/-) animals. Furthermore, RA reduces phosphatase and tensin homolog (Pten) accumulation in migrating cells, whereas Pten overexpression slows ENS precursor migration. Collectively, these data support the hypothesis that vitamin A deficiency is a non-genetic risk factor that increases Hirschsprung disease penetrance and expressivity, suggesting that some cases of Hirschsprung disease might be preventable by optimizing maternal nutrition.


Assuntos
Sistema Nervoso Entérico/embriologia , Sistema Nervoso Entérico/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Vitamina A/metabolismo , Animais , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Modelos Animais de Doenças , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Sistema Nervoso Entérico/citologia , Feminino , Fator Neurotrófico Derivado de Linhagem de Célula Glial/farmacologia , Heterozigoto , Doença de Hirschsprung/etiologia , Doença de Hirschsprung/genética , Doença de Hirschsprung/metabolismo , Doença de Hirschsprung/prevenção & controle , Humanos , Técnicas In Vitro , Fenômenos Fisiológicos da Nutrição Materna , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , PTEN Fosfo-Hidrolase/genética , Gravidez , Receptores do Ácido Retinoico/antagonistas & inibidores , Receptores do Ácido Retinoico/metabolismo , Proteínas Plasmáticas de Ligação ao Retinol/deficiência , Proteínas Plasmáticas de Ligação ao Retinol/genética , Transdução de Sinais , Tretinoína/farmacologia , Deficiência de Vitamina A/complicações , Deficiência de Vitamina A/embriologia , Deficiência de Vitamina A/genética , Deficiência de Vitamina A/metabolismo
16.
Arch Biochem Biophys ; 539(2): 223-9, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23871845

RESUMO

We investigated the effect of ß-carotene (bC) supplementation during pregnancy in a mouse model of severe vitamin A deficiency, i.e. Lrat-/-Rbp-/- dams maintained on a vitamin A-deficient diet during gestation. bC, a provitamin A carotenoid, can be enzymatically cleaved to form vitamin A for use by the developing embryo. We found that an acute supplementation (13.5 days post coitum, dpc) of bC to Lrat-/-Rbp-/- dams on a vitamin A-deficient diet activated transcriptional mechanisms in the developing tissues to maximize the utilization of bC provided to the dams. Nevertheless, these regulatory mechanisms are inefficient under this regimen, as the embryonic phenotype was not improved. We further investigated the effect of a repeated supplementation of bC during a crucial developmental period (6.5-9.5 dpc) on the above-mentioned mouse model. This treatment improved the embryonic abnormalities, as 40% of the embryos showed a normal phenotype. In addition, analysis of retinoic acid-responsive genes, such as Cyp26a1 in these embryos suggests that bC cleavage results in the production of retinoic acid which then can be used by the embryo. Taken together, these in vivo studies show that bC can be used as a source of vitamin A for severely vitamin A-deficient mammalian embryos.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Troca Materno-Fetal , Fenótipo , Retinoides/metabolismo , Índice de Gravidade de Doença , Deficiência de Vitamina A/metabolismo , beta Caroteno/administração & dosagem , beta Caroteno/metabolismo , Animais , Sistema Enzimático do Citocromo P-450/biossíntese , Sistema Enzimático do Citocromo P-450/genética , Modelos Animais de Doenças , Feminino , Troca Materno-Fetal/genética , Camundongos , Gravidez , Ácido Retinoico 4 Hidroxilase , Retinoides/genética , Tretinoína/metabolismo , Deficiência de Vitamina A/genética , beta Caroteno/genética
17.
Br J Nutr ; 109(10): 1739-45, 2013 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-22950813

RESUMO

Vitamin A (VA) plays an important role in post-natal lung development and maturation. Previously, we have reported that a supplemental dose of VA combined with 10% of all-trans-retinoic acid (VARA) synergistically increases retinol uptake and retinyl ester (RE) storage in neonatal rat lung, while up-regulating several retinoid homeostatic genes including lecithin:retinol acyltransferase (LRAT) and the retinol-binding protein receptor, stimulated by retinoic acid 6 (STRA6). However, whether inflammation has an impact on the expression of these genes and thus compromises the ability of VARA to increase lung RE content is not clear. Neonatal rats, 7- to 8-d-old, were treated with VARA either concurrently with lipopolysaccharide (LPS; Expt 1) or 12 h after LPS administration (Expt 2); in both studies, lung tissue was collected 6 h after VARA treatment, when RE formation is maximal. Inflammation was confirmed by increased IL-6 and chemokine (C­C motif) ligand 2 (CCL2) gene expression in lung at 6 h and C-reactive protein in plasma at 18 h. In both studies, LPS-induced inflammation only slightly reduced, but did not prevent the VARA-induced increase in lung RE. Quantitative RT-PCR showed that co-administration of LPS with VARA slightly attenuated the VARA-induced increase of LRAT mRNA, but not of STRA6 or cytochrome P450 26B1, the predominant RA hydroxylase in lung. By 18 h post-LPS, expression had subsided and none of these genes differed from the level in the control group. Overall, the present results suggest that retinoid homeostatic gene expression is reduced modestly, if at all, by acute LPS-induced inflammation and that VARA is still effective in increasing lung RE under conditions of moderate inflammation.


Assuntos
Animais Recém-Nascidos/metabolismo , Inflamação/complicações , Pulmão/efeitos dos fármacos , Retinoides/metabolismo , Tretinoína/metabolismo , Vitamina A/metabolismo , Aciltransferases/genética , Aciltransferases/metabolismo , Animais , Proteína C-Reativa/metabolismo , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Ésteres/metabolismo , Expressão Gênica/efeitos dos fármacos , Inflamação/induzido quimicamente , Inflamação/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Lipopolissacarídeos , Pulmão/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , RNA Mensageiro/metabolismo , Ratos , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Retinoides/genética , Tretinoína/farmacologia , Vitamina A/farmacologia , Deficiência de Vitamina A/genética , Deficiência de Vitamina A/metabolismo , Deficiência de Vitamina A/prevenção & controle
18.
J Immunol ; 187(4): 1877-83, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21765014

RESUMO

Vitamin A deficiency is highly prevalent in much of the developing world, where vaccination programs are of paramount importance to public health. However, the impact of vitamin A deficiency on the immunogenicity and protective efficacy of vaccines has not been defined previously. In this article, we show that the vitamin A metabolite retinoic acid is critical for trafficking of vaccine-elicited T lymphocytes to the gastrointestinal mucosa and for vaccine protective efficacy in mice. Moderate vitamin A deficiency abrogated Ag-specific T lymphocyte trafficking to the gastrointestinal tract, gastrointestinal cellular immune responses, and protection against a mucosal challenge following immunization with a recombinant adenovirus vaccine vector. Oral vitamin A supplementation as well as retinoic acid administration fully restored the mucosal immune responses and vaccine protective efficacy. These data suggest that oral vitamin A supplementation may be important for optimizing the success of vaccines against HIV-1 and other mucosal pathogens in the developing world, highlighting a critical relationship between host nutritional status and vaccine efficacy.


Assuntos
Vacinas contra a AIDS/imunologia , Adenoviridae , Mucosa Gástrica/imunologia , Imunidade nas Mucosas/imunologia , Mucosa Intestinal/imunologia , Deficiência de Vitamina A/imunologia , Vacinas contra a AIDS/farmacologia , Animais , Movimento Celular/efeitos dos fármacos , Movimento Celular/imunologia , Imunidade nas Mucosas/efeitos dos fármacos , Imunidade nas Mucosas/genética , Camundongos , Camundongos Knockout , Linfócitos T/imunologia , Deficiência de Vitamina A/tratamento farmacológico , Deficiência de Vitamina A/genética
19.
Nat Genet ; 27(1): 74-8, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11138002

RESUMO

Mutations or rearrangements in the gene encoding the receptor tyrosine kinase RET result in Hirschsprung disease, cancer and renal malformations. The standard model of renal development involves reciprocal signaling between the ureteric bud epithelium, inducing metanephric mesenchyme to differentiate into nephrons, and metanephric mesenchyme, inducing the ureteric bud to grow and branch. RET and GDNF (a RET ligand) are essential mediators of these epithelial-mesenchymal interactions. Vitamin A deficiency has been associated with widespread embryonic abnormalities, including renal malformations. The vitamin A signal is transduced by nuclear retinoic acid receptors (RARs). We previously showed that two RAR genes, Rara and Rarb2, were colocalized in stromal mesenchyme, a third renal cell type, where their deletion led to altered stromal cell patterning, impaired ureteric bud growth and downregulation of Ret in the ureteric bud. Here we demonstrate that forced expression of Ret in mice deficient for both Rara and Rarb2 (Rara(-/-)Rarb2(-/-)) genetically rescues renal development, restoring ureteric bud growth and stromal cell patterning. Our studies indicate the presence of a new reciprocal signaling loop between the ureteric bud epithelium and the stromal mesenchyme, dependent on Ret and vitamin A. In the first part of the loop, vitamin-A-dependent signals secreted by stromal cells control Ret expression in the ureteric bud. In the second part of the loop, ureteric bud signals dependent on Ret control stromal cell patterning.


Assuntos
Proteínas de Drosophila , Epitélio/efeitos dos fármacos , Rim/embriologia , Mesoderma/efeitos dos fármacos , Vitamina A/farmacologia , Animais , Epitélio/metabolismo , Feminino , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial , Hibridização In Situ , Rim/anormalidades , Rim/efeitos dos fármacos , Rim/metabolismo , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Camundongos Transgênicos , Morfogênese/efeitos dos fármacos , Mutação , Técnicas de Cultura de Órgãos , Proteínas Proto-Oncogênicas , Proteínas Proto-Oncogênicas c-ret , RNA Mensageiro/análise , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Receptores Proteína Tirosina Quinases , Transdução de Sinais/efeitos dos fármacos , Vitamina A/administração & dosagem , Vitamina A/genética , Deficiência de Vitamina A/genética , Deficiência de Vitamina A/fisiopatologia
20.
Curr Top Dev Biol ; 152: 115-138, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36707209

RESUMO

Structural birth defects are a common cause of abnormalities in newborns. While there are cases of structural birth defects arising due to monogenic defects or environmental exposures, many birth defects are likely caused by a complex interaction between genes and the environment. A structural birth defect with complex etiology is congenital diaphragmatic hernias (CDH), a common and often lethal disruption in diaphragm development. Mutations in more than 150 genes have been implicated in CDH pathogenesis. Although there is generally less evidence for a role for environmental factors in the etiology of CDH, deficiencies in maternal vitamin A and its derivative embryonic retinoic acid are strongly associated with CDH. However, the incomplete penetrance of CDH-implicated genes and environmental factors such as vitamin A deficiency suggest that interactions between genes and environment may be necessary to cause CDH. In this review, we examine the genetic and environmental factors implicated in diaphragm and CDH development. In addition, we evaluate the potential for gene-environment interactions in CDH etiology, focusing on the potential interactions between the CDH-implicated gene, Gata4, and maternal vitamin A deficiency.


Assuntos
Hérnias Diafragmáticas Congênitas , Deficiência de Vitamina A , Recém-Nascido , Humanos , Hérnias Diafragmáticas Congênitas/genética , Hérnias Diafragmáticas Congênitas/patologia , Deficiência de Vitamina A/complicações , Deficiência de Vitamina A/genética , Deficiência de Vitamina A/patologia , Diafragma/anormalidades , Diafragma/patologia , Tretinoína , Mutação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA