Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mediators Inflamm ; 2023: 2629262, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36704315

RESUMO

Salmonella Typhimurium infection of the gastrointestinal tract leads to damage that compromises the integrity of the intestinal epithelium and results in enterocolitis and inflammation. Salmonella infection promotes the expression of inflammasome NLRP3, leading to activation and release of proinflammatory cytokines such as IL-1ß, and the infected host often displays altered nutrient levels. To date, the effect of Salmonella infection and proinflammatory cytokine IL-1ß on the intestinal uptake of ascorbic acid (AA) is unknown. Our results revealed a marked decrease in the rate of AA uptake in mouse jejunum infected with Salmonella wild type (WT). However, the nonpathogenic mutant (Δ invA Δ spiB) strain did not affect AA uptake. The decrease in AA uptake due to Salmonella WT infection is accompanied by significantly lower expression of mouse (m)SVCT1 protein, mRNA, and hnRNA levels. NLRP3 and IL-1ß expression levels were markedly increased in Salmonella-infected mouse jejunum. IL-1ß-exposed Caco-2 cells displayed marked inhibition in AA uptake and significantly decreased hSVCT1 expression at both protein and mRNA levels. Furthermore, the activity of the SLC23A1 promoter was significantly inhibited by IL-1ß exposure. In addition, GRHPR (a known SVCT1 interactor) protein and mRNA expression levels were significantly reduced in Salmonella-infected mouse jejunum. These results indicate that Salmonella infection inhibits AA absorption in mouse jejunum and IL-1ß-exposed Caco-2 cells. The observed inhibitory effect may partially be mediated through transcriptional mechanisms.


Assuntos
Ácido Ascórbico , Infecções por Salmonella , Humanos , Animais , Camundongos , Ácido Ascórbico/metabolismo , Ácido Ascórbico/farmacologia , Salmonella typhimurium/metabolismo , Células CACO-2 , Proteína 3 que Contém Domínio de Pirina da Família NLR , Intestinos , Inflamassomos/metabolismo , Citocinas/farmacologia , RNA Mensageiro
2.
J Immunol ; 200(8): 2563-2570, 2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29531163

RESUMO

Biotin (vitamin B7) is essential for human health because of its involvement, as a cofactor, in a variety of critical cellular metabolic reactions. Previous studies have shown that biotin deficiency enhances inflammation, and certain chronic inflammatory diseases are associated with biotin deficiency; however, the mechanisms that mediate the association between biotin status and inflammation are not well understood. In this study, we examined the effect of biotin deficiency on human CD4+ T cell responses to determine their role in biotin deficiency-associated inflammation. Our investigations revealed that anti-CD3/CD28-stimulated CD4+ T cells cultured in biotin-deficient medium secreted significantly enhanced levels of the proinflammatory cytokines IFN-γ, TNF, and IL-17. Expression of the transcription factors T-bet and RORγt was increased, whereas Foxp3 expression was decreased, in biotin-deficient CD4+ T cells. The percentage of T regulatory cells was also decreased under biotin-deficient condition. A similar increase in T-bet, RORγt, and proinflammatory cytokine levels, as well as a decrease in Foxp3, was observed in inguinal lymph nodes of mice fed a biotin-deficient diet relative to pair-fed controls. Furthermore, differentiation of CD4+ T cells toward Th1 and Th17 cells was also enhanced. In vitro and in vivo investigations indicated that the increased inflammatory response was due to enhanced activation of the mammalian target of rapamycin signaling pathway in biotin-deficient CD4+ T cells. In summary, these results demonstrate that biotin deficiency enhances the inflammatory responses in CD4+ T cells, which may contribute to inflammation associated with biotin deficiency.

3.
Am J Physiol Gastrointest Liver Physiol ; 317(4): G518-G530, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31369292

RESUMO

The sodium-dependent multivitamin transporter (SMVT; SLC5A6) is involved in intestinal absorption of vitamin B7 (biotin). We have previously shown that mice with an embryonic intestinal-specific SMVT knockout (KO) develop biotin deficiency and severe spontaneous intestinal inflammation in addition to growth retardation, developmental delays, and death within the first 6-7 wk of life. The profound morbidity and mortality associated with the SMVT-KO has limited our ability to further characterize the intestinal inflammation and other sequelae of this deletion in adult mice with a mature gut microbiota. To overcome this limitation, we generated an intestine-specific, tamoxifen-inducible, conditional SMVT-KO (SMVT-icKO). Our results showed that adult SMVT-icKO mice have reduced body weight, biotin deficiency, shorter colonic length, and bloody diarrhea compared with age- and sex-matched control littermates. All SMVT-icKO mice also developed spontaneous intestinal inflammation associated with induction of calprotectin (S100a8/S100a9), proinflammatory cytokines (IL-1ß, TNF-α, IFN-γ, and IL-6), and an increase in intestinal permeability. Additionally, the intestines of SMVT-icKO showed activation of the NF-κB pathway and the nucleotide-binding domain and leucine-rich repeat pyrin 3 domain (NLRP3) inflammasome. Notably, administration of broad-spectrum antibiotics reduced lethality and led to normalization of intestinal inflammation, proinflammatory cytokines, altered mucosal integrity, and reduced expression of the NLRP3 inflammasome. Overall, these findings support our conclusion that the biotin transport pathway plays an important role in the maintenance of intestinal homeostasis, and that NF-κB and the NLRP3 inflammasome, as well as gut microbiota, drive the development of intestinal inflammation when SMVT is absent.NEW & NOTEWORTHY This study demonstrates that deletion of the intestinal biotin uptake system in adult mice leads to the development of spontaneous gut inflammation and that luminal microbiota plays a role in its development.


Assuntos
Enterite/genética , Antagonistas de Estrogênios/toxicidade , Microbioma Gastrointestinal/efeitos dos fármacos , Intestinos/efeitos dos fármacos , NF-kappa B/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Simportadores/metabolismo , Tamoxifeno/toxicidade , Envelhecimento , Animais , Biotina/metabolismo , Peso Corporal/efeitos dos fármacos , Colo/patologia , Citocinas/metabolismo , Diarreia/induzido quimicamente , Diarreia/microbiologia , Diarreia/patologia , Enterite/induzido quimicamente , Enterite/microbiologia , Intestinos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Simportadores/efeitos dos fármacos , Simportadores/genética
4.
Am J Physiol Cell Physiol ; 315(1): C73-C79, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29669219

RESUMO

Intestinal absorption of the water-soluble vitamins biotin and pantothenic acid is carrier mediated and involves the sodium-dependent multivitamin transporter (SMVT; product of the SLC5A6 gene). We recently observed that intestinal-specific (conditional) knockout of the mouse Slc5a6 gene (SMVT-cKO) is associated with growth retardation, the development of spontaneous and severe inflammation, abnormal histology in the large intestine, altered gut permeability, and early death. Our aim in this study was to examine the possibility that biotin and pantothenic acid oversupplementation (BPS) of the SMVT-cKO mice could reverse the above-described abnormalities. BPS was provided in the drinking water to mice before conception, to dams during pregnancy and lactation, and to the SMVT-cKO mice throughout their life. Our findings showed that such a regimen prevents early death, as well as normalizes the growth rate, intestinal integrity, pathology, and inflammation in SMVT-cKO mice. These findings provide clear evidence for a role for biotin and/or pantothenic acid in the maintenance of normal intestinal integrity and health.


Assuntos
Biotina/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Ácido Pantotênico/farmacologia , Simportadores/metabolismo , Animais , Feminino , Inflamação/metabolismo , Absorção Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Lactação/efeitos dos fármacos , Camundongos , Camundongos Knockout , Gravidez
6.
Am J Physiol Gastrointest Liver Physiol ; 310(4): G285-93, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26660539

RESUMO

Riboflavin (RF) is indispensable for normal cell metabolism, proliferation, and growth. The RFVT-3 protein (product of the Slc52a3 gene) is expressed in the gut with the expression being restricted to the apical membrane domain of the polarized intestinal epithelial cells. The relative contribution of RFVT-3 to total carrier-mediated RF uptake in the native intestine, however, is not clear. We addressed this issue in the current investigation using a conditional (intestinal-specific) RFVT-3 knockout (cKO) mouse model developed by the Cre/Lox approach. All RFVT-3 cKO mice were found to be RF deficient and showed a significant growth and development retardation; also, nearly two-thirds of them died prematurely between the age of 6 and 12 wk. In vivo (intestinal and colonic loops) and in vitro (native isolated intestinal epithelial cells) uptake studies showed a severe inhibition in carrier-mediated RF uptake in the cKO mice compared with control littermates. We also observed a significant increase in the level of expression of oxidative stress-responsive genes in the intestine of the cKO mice compared with control littermates. Supplementation of the RFVT-3 cKO mice with pharmacological doses of RF led to a complete correction of the growth retardation and to normalization in the level of expression of the oxidative stress-responsive genes in the gut. These results show, for the first time, that the RFVT-3 system is the main transporter involved in carrier-mediated RF uptake in the native mouse small and large intestine, and that its dysfunction impairs normal RF body homeostasis.


Assuntos
Absorção Intestinal/genética , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Riboflavina/metabolismo , Vitaminas/metabolismo , Animais , Colo/metabolismo , Deficiências do Desenvolvimento/genética , Células Epiteliais/metabolismo , Expressão Gênica/genética , Transtornos do Crescimento/genética , Homeostase/genética , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estresse Oxidativo/genética
7.
Am J Physiol Gastrointest Liver Physiol ; 311(3): G561-70, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27492331

RESUMO

Utilizing a conditional (intestinal-specific) knockout (cKO) mouse model, we have recently shown that the sodium-dependent multivitamin transporter (SMVT) (SLC5A6) is the only biotin uptake system that operates in the gut and that its deletion leads to biotin deficiency. Unexpectedly, we also observed that all SMVT-cKO mice develop chronic active inflammation, especially in the cecum. Our aim here was to examine the role of SMVT in the maintenance of intestinal mucosal integrity [permeability and expression of tight junction (TJ) proteins]. Our results showed that knocking out the mouse intestinal SMVT is associated with a significant increase in gut permeability and with changes in the level of expression of TJ proteins. To determine whether these changes are related to the state of biotin deficiency that develops in SMVT-cKO mice, we induced (by dietary means) biotin deficiency in wild-type mice and examined its effect on the above-mentioned parameters. The results showed that dietary-induced biotin deficiency leads to a similar development of chronic active inflammation in the cecum with an increase in the level of expression of proinflammatory cytokines, as well as an increase in intestinal permeability and changes in the level of expression of TJ proteins. We also examined the effect of chronic biotin deficiency on permeability and expression of TJ proteins in confluent intestinal epithelial Caco-2 monolayers but observed no changes in these parameters. These results show that the intestinal SMVT plays an important role in the maintenance of normal mucosal integrity, most likely via its role in providing biotin to different cells of the gut mucosa.


Assuntos
Mucosa Intestinal/fisiologia , Simportadores/metabolismo , Animais , Biotina/administração & dosagem , Deficiência de Biotinidase/metabolismo , Células CACO-2 , Regulação da Expressão Gênica/fisiologia , Homeostase/fisiologia , Humanos , Camundongos , Camundongos Knockout , Permeabilidade , Simportadores/genética , Proteínas de Junções Íntimas/genética , Proteínas de Junções Íntimas/metabolismo
8.
Am J Physiol Gastrointest Liver Physiol ; 306(4): G320-7, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24356885

RESUMO

Nonalcoholic fatty liver disease (NAFLD), a leading cause of liver damage, comprises a spectrum of liver abnormalities including the early fat deposition in the liver (hepatic steatosis) and advanced nonalcoholic steatohepatitis. Niacin decreases plasma triglycerides, but its effect on hepatic steatosis is elusive. To examine the effect of niacin on steatosis, rats were fed either a rodent normal chow, chow containing high fat (HF), or HF containing 0.5% or 1.0% niacin in the diet for 4 wk. For regression studies, rats were first fed the HF diet for 6 wk to induce hepatic steatosis and were then treated with niacin (0.5% in the diet) while on the HF diet for 6 wk. The findings indicated that inclusion of niacin at 0.5% and 1.0% doses in the HF diet significantly decreased liver fat content, liver weight, hepatic oxidative products, and prevented hepatic steatosis. Niacin treatment to rats with preexisting hepatic steatosis induced by the HF diet significantly regressed steatosis. Niacin had no effect on the mRNA expression of fatty acid synthesis or oxidation genes (including sterol-regulatory element-binding protein 1, acetyl-CoA carboxylase 1, fatty acid synthase, and carnitine palmitoyltransferase 1) but significantly inhibited mRNA levels, protein expression, and activity of diacylglycerol acyltrasferase 2, a key enzyme in triglyceride synthesis. These novel findings suggest that niacin effectively prevents and causes the regression of experimental hepatic steatosis. Approved niacin formulation(s) for other indications or niacin analogs may offer a very cost-effective opportunity for the clinical development of niacin for treating NAFLD and fatty liver disease.


Assuntos
Antioxidantes/farmacologia , Fígado Gorduroso/tratamento farmacológico , Fígado Gorduroso/prevenção & controle , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Niacina/farmacologia , Animais , Anti-Inflamatórios/uso terapêutico , Modelos Animais de Doenças , Progressão da Doença , Ácidos Graxos não Esterificados/metabolismo , Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Mediadores da Inflamação/metabolismo , Metabolismo dos Lipídeos/genética , Peroxidação de Lipídeos/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Masculino , Hepatopatia Gordurosa não Alcoólica , Oxirredução , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo , Fatores de Tempo , Triglicerídeos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
9.
Am J Physiol Gastrointest Liver Physiol ; 304(1): G64-71, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23104561

RESUMO

The Slc5a6 gene expresses a plasma membrane protein involved in the transport of the water-soluble vitamin biotin; the transporter is commonly referred to as the sodium-dependent multivitamin transporter (SMVT) because it also transports pantothenic acid and lipoic acid. The relative contribution of the SMVT system toward carrier-mediated biotin uptake in the native intestine in vivo has not been established. We used a Cre/lox technology to generate an intestine-specific (conditional) SMVT knockout (KO) mouse model to address this issue. The KO mice exhibited absence of expression of SMVT in the intestine compared with sex-matched littermates as well as the expected normal SMVT expression in other tissues. About two-thirds of the KO mice died prematurely between the age of 6 and 10 wk. Growth retardation, decreased bone density, decreased bone length, and decreased biotin status were observed in the KO mice. Microscopic analysis showed histological abnormalities in the small bowel (shortened villi, dysplasia) and cecum (chronic active inflammation, dysplasia) of the KO mice. In vivo (and in vitro) transport studies showed complete inhibition in carrier-mediated biotin uptake in the intestine of the KO mice compared with their control littermates. These studies provide the first in vivo confirmation in native intestine that SMVT is solely responsible for intestinal biotin uptake. These studies also provide evidence for a casual association between SMVT function and normal intestinal health.


Assuntos
Biotina/metabolismo , Absorção Intestinal/fisiologia , Mucosa Intestinal/metabolismo , Simportadores/genética , Animais , Western Blotting , Células-Tronco Embrionárias/transplante , Intestinos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ácido Pantotênico/metabolismo , Fenótipo , Reação em Cadeia da Polimerase em Tempo Real
10.
Histochem Cell Biol ; 139(6): 909-18, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23515787

RESUMO

The orexigenic peptide ghrelin and the anorexigenic peptide nesfatin-1 are expressed by the same endocrine cell of the rat stomach, the X/A-like cell. However, data in humans are lacking, especially under conditions of obesity. We collected gastric tissue of obese patients undergoing sleeve gastrectomy and investigated the expression of nesfatin-1 and ghrelin in the gastric oxyntic mucosa by immunofluorescence. Nesfatin-1 immunoreactivity was detected in the human oxyntic mucosa in cells with an endocrine phenotype. A major portion of nesfatin-1 immunoreactive cells (78 %) co-localized with ghrelin indicating the occurrence in human X/A-like cells. In patients with very high body mass index (BMI 55-65 kg/m(2)), the number of nesfatin-1 immunoreactive cells/low-power field was significantly higher than in obese patients with lower BMI (40-50 kg/m(2), 118 ± 10 vs. 82 ± 11, p < 0.05). On the other hand, the number of ghrelin immunoreactive cells was significantly reduced in obese patients with higher compared to lower BMI (96 ± 12 vs. 204 ± 21, p < 0.01). Also the ghrelin-acylating enzyme ghrelin-O-acyltransferase decreased with increasing BMI. In conclusion, nesfatin-1 immunoreactivity is also co-localized with ghrelin in human gastric X/A-like cells giving rise to a dual role of this cell type with differential effects on stimulation and inhibition of appetite dependent on the peptide released. The expression of these two peptides is differentially regulated under obese conditions with an increase of nesfatin-1 and a decrease of ghrelin immunoreactivity with rising BMI pointing towards an adaptive change of expression that may counteract further body weight increase.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ligação a DNA/metabolismo , Mucosa Gástrica/metabolismo , Grelina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Obesidade Mórbida/metabolismo , Aciltransferases/metabolismo , Adaptação Fisiológica , Adulto , Idoso , Western Blotting , Índice de Massa Corporal , Feminino , Mucosa Gástrica/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Nucleobindinas , Obesidade Mórbida/patologia , Obesidade Mórbida/cirurgia
11.
Cell Rep Med ; 4(7): 101101, 2023 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-37421947

RESUMO

Merkel cell carcinoma (MCC), a rare but aggressive skin cancer, remains a challenge in the era of precision medicine. Immune checkpoint inhibitors (ICIs), the only approved therapy for advanced MCC, are impeded by high primary and acquired resistance. Hence, we dissect transcriptomic heterogeneity at single-cell resolution in a panel of patient tumors, revealing phenotypic plasticity in a subset of treatment-naive MCC. The tumor cells in a "mesenchymal-like" state are endowed with an inflamed phenotype that portends a better ICI response. This observation is also validated in the largest whole transcriptomic dataset available from MCC patient tumors. In contrast, ICI-resistant tumors predominantly express neuroepithelial markers in a well-differentiated state with "immune-cold" landscape. Importantly, a subtle shift to "mesenchymal-like" state reverts copanlisib resistance in primary MCC cells, highlighting potential strategies in patient stratification for therapeutics to harness tumor cell plasticity, augment treatment efficacy, and avert resistance.


Assuntos
Carcinoma de Célula de Merkel , Neoplasias Cutâneas , Humanos , Carcinoma de Célula de Merkel/tratamento farmacológico , Carcinoma de Célula de Merkel/genética , Carcinoma de Célula de Merkel/patologia , Transcriptoma/genética , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Imunoterapia , Perfilação da Expressão Gênica
12.
Clin Dev Immunol ; 2012: 160724, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22899945

RESUMO

Lung cancers remain one of the most common and deadly cancers in the world today (12.5% of newly diagnosed cancers) despite current advances in chemo- and radiation therapies. Often, by the time these tumors are diagnosed, they have already metastasized. These tumors demonstrate the classic hallmarks of cancer in that they have advanced defensive strategies allowing them to escape various standard oncological treatments. Immunotherapy is making inroads towards effectively treating other fatal cancers, such as melanoma, glioblastoma multiforme, and castrate-resistant prostate cancers. This paper will cover the escape mechanisms of bronchogenic lung cancer that must be overcome before they can be successfully treated. We also review the history of immunotherapy directed towards lung cancers.


Assuntos
Vacinas Anticâncer , Imunoterapia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/terapia , Evasão Tumoral , Anticorpos/uso terapêutico , Progressão da Doença , Terapia Genética , Humanos , Neoplasias Pulmonares/patologia , Linfócitos/imunologia , Taxa de Sobrevida
13.
Am J Physiol Gastrointest Liver Physiol ; 301(2): G239-48, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21636529

RESUMO

Clinical studies are evaluating the efficacy of synthetic ghrelin agonists in postoperative ileus management. However, the control of ghrelin secretion under conditions of postoperative gastric ileus is largely unknown. Peripheral somatostatin inhibits ghrelin secretion in animals and humans. We investigated the time course of ghrelin changes postsurgery in fasted rats and whether somatostatin receptor subtype 2 (sst(2)) signaling is involved. Abdominal surgery (laparotomy and 1-min cecal palpation) induced a rapid and long-lasting decrease in plasma acyl ghrelin levels as shown by the 64, 67, and 59% reduction at 0.5, 2, and 5 h postsurgery, respectively, compared with sham (anesthesia alone for 10 min, P < 0.05). Levels were partly recovered at 7 h and fully restored at 24 h. The percentage of acyl ghrelin reduction was significantly higher than that of desacyl ghrelin at 2 h postsurgery and not at any other time point. This was associated with a 48 and 23% decrease in gastric and plasma ghrelin-O-acyltransferase protein concentrations, respectively (P < 0.001). Ghrelin-positive cells in the oxyntic mucosa expressed sst(2a) receptor and the sst(2) agonist S-346-011 inhibited fasting acyl ghrelin levels by 64 and 77% at 0.5 and 2 h, respectively. The sst(2) antagonist S-406-028 prevented the abdominal surgery-induced decreased circulating acyl ghrelin but not the delayed gastric emptying assessed 0.5 h postinjection. These data show that activation of sst(2) receptor located on gastric X/A-like cells plays a key role in the rapid inhibition of circulating acyl ghrelin induced by abdominal surgery while not being primarily involved in the early phase of postoperative gastric ileus.


Assuntos
Abdome/cirurgia , Aciltransferases/sangue , Esvaziamento Gástrico/fisiologia , Grelina/sangue , Íleus/fisiopatologia , RNA Mensageiro/metabolismo , Receptores de Somatostatina/metabolismo , Gastropatias/fisiopatologia , Aciltransferases/metabolismo , Análise de Variância , Animais , Mucosa Gástrica/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/antagonistas & inibidores , Transdução de Sinais/fisiologia , Fatores de Tempo
14.
Gastroenterology ; 138(5): 1802-9, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-19879271

RESUMO

BACKGROUND & AIMS: Intestinal thiamin uptake process is vital for maintaining normal body homeostasis of the vitamin; in vitro studies suggest that both thiamin transporter-1 (THTR-1) and -2 (THTR-2) are involved. Mutations in THTR-1 cause thiamin-responsive megaloblastic anemia, a tissue-specific disease associated with diabetes mellitus, megaloblastic anemia, and sensorineural deafness. However, in patients with thiamin-responsive megaloblastic anemia, plasma thiamin levels are within normal range, indicating that THTR-2 (or another carrier) could provide sufficient intestinal thiamin absorption. We tested this possibility and examined the role of THTR-2 in uptake of thiamin in the intestine of mice. METHODS: THTR-2-deficient mice were generated by SLC19A3 gene knockout and used to examine intestinal uptake of thiamin in vitro (isolated cells) and in vivo (intact intestinal loops). We also examined intestinal thiamin uptake in THTR-1-deficient mice. RESULTS: Intestine of THTR-2-deficient mice had reduced uptake of thiamin compared with those of wild-type littermate mice (P < .01); this reduction was associated with a decrease (P < .01) in blood thiamin levels in THTR-2-deficient mice. However, intestinal uptake of thiamin in THTR-1-deficient mice was not significantly different from that of wild-type littermate animals. Level of expression of THTR-1 was not altered in the intestine of THTR-2-deficient mice, but level of expression of THTR-2 was up-regulated in the intestine of THTR-1-deficient mice. CONCLUSIONS: THTR-2 is required for normal uptake of thiamin in the intestine and can fulfill normal levels of uptake in conditions associated with THTR-1 dysfunction.


Assuntos
Absorção Intestinal , Jejuno/metabolismo , Proteínas de Membrana Transportadoras/deficiência , Tiamina/metabolismo , Animais , Comportamento Animal , Transporte Biológico , Regulação da Expressão Gênica , Genótipo , Mucosa Intestinal/metabolismo , Rim/metabolismo , Rim/patologia , Fígado/metabolismo , Fígado/patologia , Proteínas de Membrana Transportadoras/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , RNA Mensageiro/metabolismo , Tiamina/sangue
15.
Curr Gastroenterol Rep ; 13(6): 513-7, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21931998

RESUMO

Ménétrier's disease is a rare hyperproliferative protein-losing gastropathy of the gastric foveolar epithelium. Most common symptoms include epigastric pain with fullness and vomiting, and generalized peripheral edema with hypoalbuminemia. Radiologically, the wall of the gastric body and fundus is diffusely thickened, often with antral sparing. Giant rugal edematous folds are seen on gastroscopy, and histology of biopsy material shows diffuse foveolar hyperplasia with cystic dilatation of the glandular portion of the gastric mucosa in the absence of significant inflammatory infiltrate. The recent discovery of transforming growth factor α overexpression opens the way of epidermal growth factor receptor blockade with cetuximab as first-line treatment modality in severe cases of Ménétrier's disease.


Assuntos
Gastrite Hipertrófica/diagnóstico , Gastrite Hipertrófica/etiologia , Gastrite Hipertrófica/metabolismo , Gastrite Hipertrófica/terapia , Humanos , Fator de Crescimento Transformador alfa/metabolismo
16.
Physiol Genomics ; 42(3): 348-60, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20551148

RESUMO

Prostanoids are an important class of intraocular pressure (IOP)-lowering antiglaucoma agents that act primarily via increased uveo-scleral aqueous humor outflow through the ciliary body. We have developed two novel PGE(2) analogs that are specific agonists for the PGE(2) receptor subtypes EP2 and EP4, respectively. To identify gene regulatory networks and key players that mediate the physiological effects observed in vivo, we performed genomewide expression studies using human ciliary smooth muscle cells. Quantitative real-time RT-PCR confirmed a largely overlapping gene expression profile subsequent to EP2 and EP4 agonist treatment, with 65 significantly regulated genes identified overall, 5 being specific for the EP2 agonist and 6 specific for the EP4 agonist. We found predicted functional cAMP-response elements in promoter regions of a large fraction of the predominantly upregulated genes, which suggests that the cAMP signaling pathway is the most important intracellular signaling pathway for these agonists in these cells. Several target genes were identified that, as part of complex regulatory networks, are implicated in tissue remodeling processes and osmoregulation (e.g., AREG, LOXL3, BMP2, AQP3) and thus may help elucidate the mechanism of action of these IOP-lowering drugs involving the uveo-scleral outflow path.


Assuntos
Corpo Ciliar/metabolismo , Regulação da Expressão Gênica , Miócitos de Músculo Liso/metabolismo , Receptores de Prostaglandina E Subtipo EP2/fisiologia , Receptores de Prostaglandina E Subtipo EP4/fisiologia , Células Cultivadas , Corpo Ciliar/efeitos dos fármacos , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Análise em Microsséries , Miócitos de Músculo Liso/efeitos dos fármacos , Prostaglandinas Sintéticas/farmacologia , Receptores de Prostaglandina E Subtipo EP2/agonistas , Receptores de Prostaglandina E Subtipo EP2/genética , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Receptores de Prostaglandina E Subtipo EP4/genética , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estudos de Validação como Assunto
17.
Biochem Biophys Res Commun ; 392(1): 67-71, 2010 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-20059966

RESUMO

The enzyme that acylates ghrelin was recently identified in mice as the fourth member of the membrane-bound O-acyltransferases superfamily (MBOAT4) and named ghrelin-O-acyltransferase (GOAT). Only one report showed GOAT mRNA expression in ghrelin-expressing cells of the mouse stomach. We investigated the distribution of GOAT protein in peripheral tissues and co-expression with endocrine markers in the gastric mucosa using a custom-made anti-GOAT antibody. Tissues were collected from male Sprague-Dawley rats and C57BL/6 mice. Western blot revealed two immunoreactive bands in rat and mouse gastric corpus mucosal proteins, a 50 kDa band corresponding to the GOAT protein and a 100 kDa band likely corresponding to a dimer. Western blot also detected GOAT in the plasma and levels were strongly increased after 24-h fasting in mice and slightly in rats. GOAT-immunoreactive cells were located in the gastric corpus mucosa and the anterior pituitary gland, whereas other peripheral tissues of rats and mice examined were negative. In mice, GOAT-immunoreactive cells were mainly distributed throughout the middle portion of the oxyntic glands, whereas in rats they were localized mainly in the lower portion of the glands. Double labeling showed that 95+/-1% of GOAT-immunoreactive cells in mice co-labeled with ghrelin, whereas in rats only 56+/-4% of GOAT-positive cells showed co-expression of ghrelin. The remainder of the GOAT-immunopositive cells in rats co-expressed histidine decarboxylase (44+/-3%). No co-localization was observed with somatostatin in rats or mice. These data suggest species differences between rats and mice in gastric GOAT expression perhaps resulting in a different role of the MBOAT4 enzyme in the rat stomach. Detection of GOAT in the plasma raises the possibility that ghrelin octanoylation may occur in the circulation and the fasting-induced increase in GOAT may contribute to the increase of acylated ghrelin after fasting.


Assuntos
Aciltransferases/metabolismo , Jejum/metabolismo , Células Parietais Gástricas/enzimologia , Aciltransferases/sangue , Animais , Anticorpos/imunologia , Masculino , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Sprague-Dawley
18.
Peptides ; 128: 170308, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32229144

RESUMO

Since its discovery in 2006 by Oh-I and colleagues, NUCB2/nesfatin-1 encoded by nucleobindin-2 (NUCB2) has drawn sustained attention as reflected in over 500 publications. Among those, more than half focused on the alterations of food intake, body weight and metabolism (glucose, fat) induced by nesfatin-1 and/or NUCB2/nesfatin-1. In the current review we discuss the existing literature focusing on NUCB2/nesfatin-1's influence on food intake, body weight and glucose as well as fat metabolism and highlight gaps in knowledge.


Assuntos
Peso Corporal/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Nucleobindinas/metabolismo , Nucleobindinas/farmacologia , Animais , Gorduras/metabolismo , Glucose/metabolismo , Humanos , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo
19.
Front Immunol ; 11: 561947, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33178189

RESUMO

Sickle cell disease (SCD) is a hemoglobinopathy affecting multiple organs and featuring acute and chronic pain. Purkinje cell damage and hyperalgesia have been demonstrated in transgenic sickle mice. Purkinje cells are associated with movement and neural function which may influence pain. We hypothesized that Purkinje cell damage and/or chronic pain burden provoke compensatory gait changes in sickle mice. We found that Purkinje cells undergoe increased apoptosis as shown by caspase-3 activation. Using an automated gait measurement system, MouseWalker, we characterized spatiotemporal gait characteristics of humanized transgenic BERK sickle mice in comparison to control mice. Sickle mice showed alteration in stance instability and dynamic gait parameters (walking speed, stance duration, swing duration and specific swing indices). Differences in stance instability may reflect motor dysfunction due to damaged Purkinje cells. Alterations in diagonal and all stance indices indicative of hesitation during walking may originate from motor dysfunction and/or arise from fear and/or anticipation of movement-evoked pain. We also demonstrate that stance duration, diagonal swing indices and all stance indices correlate with both mechanical and deep tissue hyperalgesia, while stance instability correlates with only deep tissue hyperalgesia. Therefore, objective analysis of gait in SCD may provide insights into neurological impairment and pain states.


Assuntos
Anemia Falciforme/fisiopatologia , Marcha/genética , Anemia Falciforme/complicações , Animais , Apoptose/genética , Encéfalo/patologia , Caspase 3/metabolismo , Dor Crônica/complicações , Modelos Animais de Doenças , Técnicas de Inativação de Genes , Humanos , Hiperalgesia/complicações , Camundongos , Camundongos Transgênicos , Fenótipo , Células de Purkinje/metabolismo , Células de Purkinje/patologia , Caminhada , alfa-Globinas/genética , alfa-Globinas/metabolismo , Globinas beta/genética , Globinas beta/metabolismo
20.
Endocrinology ; 150(1): 232-8, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18818289

RESUMO

Hypothalamic nesfatin-1, derived from the nucleobindin2 (NUCB2) precursor, inhibits nocturnal food intake and body weight gain in rats. Nesfatin-1 is able to cross the blood-brain barrier, suggesting a peripheral source of nesfatin-1. Many centrally acting food intake regulatory neuropeptides are also produced in the periphery, especially in the gastrointestinal tract. Therefore, we investigated the gene expression of NUCB2 and distribution of nesfatin-1-immunoreactive cells in the stomach. Microarray mRNA expression profiles in purified small endocrine cells of the gastric mucosa substantiated by quantitative RT-PCR showed significantly higher NUCB2 mRNA expression compared with brain and heart. Western blot confirmed the expression of NUCB2 protein and its transport into a secretory soluble fraction of gastric mucosal endocrine cell homogenates. Immunohistochemical colabeling for nesfatin-1 and ghrelin, histidine decarboxylase, or somatostatin revealed two subtypes of nesfatin-1-positive endocrine cells. Cells in the midportion of the glands coexpressed nesfatin-1 and ghrelin, whereas few cells in the glandular base coexpressed nesfatin-1 and somatostatin or histidine decarboxylase. High-resolution three-dimensional volume imaging revealed two separate populations of intracytoplasmic vesicles in these cells, one containing nesfatin-1 and the other ghrelin immunoreactivity. Microarray rat genome expression data of NUCB2 in small gastric endocrine cells confirmed by quantitative RT-PCR showed significant down-regulation of NUCB2 after 24 h fasting. In summary, NUCB2 mRNA expression as well as protein content is present in a specific subset of gastric endocrine cells, most of which coexpress ghrelin. NUCB2 gene expression is significantly regulated by nutritional status, suggesting a regulatory role of peripheral nesfatin-1 in energy homeostasis.


Assuntos
Mucosa Gástrica/citologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Células Parietais Gástricas/citologia , Animais , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação a DNA/genética , Ingestão de Energia , Grelina/metabolismo , Histidina Descarboxilase/metabolismo , Imuno-Histoquímica , Masculino , Nucleobindinas , Análise de Sequência com Séries de Oligonucleotídeos , Pâncreas/citologia , Hipófise/citologia , RNA/genética , RNA/isolamento & purificação , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Somatostatina/metabolismo , Aumento de Peso
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa