Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.049
Filtrar
1.
Gastroenterology ; 162(3): 813-827.e8, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34767785

RESUMEN

BACKGROUND & AIMS: Because inflammatory bowel disease is increasing worldwide and can lead to colitis-associated carcinoma (CAC), new interventions are needed. We have shown that spermine oxidase (SMOX), which generates spermidine (Spd), regulates colitis. Here we determined whether Spd treatment reduces colitis and carcinogenesis. METHODS: SMOX was quantified in human colitis and associated dysplasia using quantitative reverse-transcription polymerase chain reaction and immunohistochemistry. We used wild-type (WT) and Smox-/- C57BL/6 mice treated with dextran sulfate sodium (DSS) or azoxymethane (AOM)-DSS as models of colitis and CAC, respectively. Mice with epithelial-specific deletion of Apc were used as a model of sporadic colon cancer. Animals were supplemented or not with Spd in the drinking water. Colonic polyamines, inflammation, tumorigenesis, transcriptomes, and microbiomes were assessed. RESULTS: SMOX messenger RNA levels were decreased in human ulcerative colitis tissues and inversely correlated with disease activity, and SMOX protein was reduced in colitis-associated dysplasia. DSS colitis and AOM-DSS-induced dysplasia and tumorigenesis were worsened in Smox-/- vs WT mice and improved in both genotypes with Spd. Tumor development caused by Apc deletion was also reduced by Spd. Smox deletion and AOM-DSS treatment were both strongly associated with increased expression of α-defensins, which was reduced by Spd. A shift in the microbiome, with reduced abundance of Prevotella and increased Proteobacteria and Deferribacteres, occurred in Smox-/- mice and was reversed with Spd. CONCLUSIONS: Loss of SMOX is associated with exacerbated colitis and CAC, increased α-defensin expression, and dysbiosis of the microbiome. Spd supplementation reverses these phenotypes, indicating that it has potential as an adjunctive treatment for colitis and chemopreventive for colon carcinogenesis.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Colitis/genética , Neoplasias del Colon/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Espermidina/uso terapéutico , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Azoximetano , Colitis/inducido químicamente , Colitis/enzimología , Colitis/prevención & control , Colitis Ulcerosa/enzimología , Colitis Ulcerosa/genética , Colon/enzimología , Colon/patología , Neoplasias del Colon/prevención & control , Sulfato de Dextran , Microbioma Gastrointestinal/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Mucosa Intestinal/enzimología , Mucosa Intestinal/patología , Masculino , Ratones , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Lesiones Precancerosas/enzimología , Factores Protectores , ARN Mensajero/metabolismo , Índice de Severidad de la Enfermedad , Espermidina/metabolismo , Espermidina/farmacología , Pérdida de Peso/efectos de los fármacos , alfa-Defensinas/genética , alfa-Defensinas/metabolismo , Poliamino Oxidasa
2.
Int J Mol Sci ; 22(22)2021 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-34830327

RESUMEN

Transglutaminases are protein-modifying enzymes involved in physiological and pathological processes with potent therapeutic possibilities. Human TG4, also called prostate transglutaminase, is involved in the development of autoimmune and tumour diseases. Although rodent TG4 is well characterised, biochemical characteristics of human TG4 that could help th e understanding of its way of action are not published. First, we analysed proteomics databases and found that TG4 protein is present in human tissues beyond the prostate. Then, we studied in vitro the transamidase activity of human TG4 and its regulation using the microtitre plate method. Human TG4 has low transamidase activity which prefers slightly acidic pH and a reducing environment. It is enhanced by submicellar concentrations of SDS suggesting that membrane proximity is an important regulatory event. Human TG4 does not bind GTP as tested by GTP-agarose and BODIPY-FL-GTPγS binding, and its proteolytic activation by dispase or when expressed in AD-293 cells was not observed either. We identified several potential human TG4 glutamine donor substrates in the AD-293 cell extract by biotin-pentylamine incorporation and mass spectrometry. Several of these potential substrates are involved in cell-cell interaction, adhesion and proliferation, suggesting that human TG4 could become an anticancer therapeutic target.


Asunto(s)
Colon/enzimología , Miocardio/enzimología , Próstata/enzimología , Transglutaminasas/metabolismo , Vejiga Urinaria/enzimología , Secuencia de Aminoácidos , Línea Celular Tumoral , Clonación Molecular , Estabilidad de Enzimas , Células Epiteliales/citología , Células Epiteliales/enzimología , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Cinética , Masculino , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Dodecil Sulfato de Sodio/química , Especificidad por Sustrato , Distribución Tisular , Transglutaminasas/genética
3.
Nat Immunol ; 22(11): 1440-1451, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34686860

RESUMEN

Intestinal epithelial cell (IEC) damage by T cells contributes to graft-versus-host disease, inflammatory bowel disease and immune checkpoint blockade-mediated colitis. But little is known about the target cell-intrinsic features that affect disease severity. Here we identified disruption of oxidative phosphorylation and an increase in succinate levels in the IECs from several distinct in vivo models of T cell-mediated colitis. Metabolic flux studies, complemented by imaging and protein analyses, identified disruption of IEC-intrinsic succinate dehydrogenase A (SDHA), a component of mitochondrial complex II, in causing these metabolic alterations. The relevance of IEC-intrinsic SDHA in mediating disease severity was confirmed by complementary chemical and genetic experimental approaches and validated in human clinical samples. These data identify a critical role for the alteration of the IEC-specific mitochondrial complex II component SDHA in the regulation of the severity of T cell-mediated intestinal diseases.


Asunto(s)
Colitis/enzimología , Colon/enzimología , Citotoxicidad Inmunológica , Complejo II de Transporte de Electrones/metabolismo , Células Epiteliales/enzimología , Enfermedad Injerto contra Huésped/enzimología , Mucosa Intestinal/enzimología , Mitocondrias/enzimología , Linfocitos T/inmunología , Animales , Estudios de Casos y Controles , Comunicación Celular , Células Cultivadas , Colitis/genética , Colitis/inmunología , Colitis/patología , Colon/inmunología , Colon/ultraestructura , Modelos Animales de Enfermedad , Complejo II de Transporte de Electrones/genética , Células Epiteliales/inmunología , Células Epiteliales/ultraestructura , Femenino , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Humanos , Inmunidad Mucosa , Mucosa Intestinal/inmunología , Mucosa Intestinal/ultraestructura , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias/inmunología , Mitocondrias/ultraestructura , Fosforilación Oxidativa , Ácido Succínico/metabolismo , Linfocitos T/metabolismo
4.
Am J Physiol Gastrointest Liver Physiol ; 321(5): G500-G512, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34494462

RESUMEN

Mouse and human data implicate the NOD1 and NOD2 sensors of the intestinal microbiome and the associated signal transduction via the receptor interacting protein kinase 2 (RIPK2) as a potential key signaling node for the development of inflammatory bowel disease (IBD) and an attractive target for pharmacological intervention. The TRUC mouse model of IBD was strongly indicated for evaluating RIPK2 antagonism for its effect on intestinal inflammation based on previous knockout studies with NOD1, NOD2, and RIPK2. We identified and profiled the BI 706039 molecule as a potent and specific functional inhibitor of both human and mouse RIPK2 and with favorable pharmacokinetic properties. We dosed BI 706039 in the spontaneous TRUC mouse model from age 28 to 56 days. Oral, daily administration of BI 706039 caused dose-responsive and significant improvement in colonic histopathological inflammation, colon weight, and terminal levels of protein-normalized fecal lipocalin (all P values <0.001). These observations correlated with dose responsively increasing systemic levels of the BI 706039 compound, splenic molecular target engagement of RIPK2, and modulation of inflammatory genes in the colon. This demonstrates that a relatively low oral dose of a potent and selective RIPK2 inhibitor can modulate signaling in the intestinal immune system and significantly improve disease associated intestinal inflammation.NEW & NOTEWORTHY The RIPK2 kinase at the apex of microbiome immunosensing is an attractive target for pharmacological intervention. A low oral dose of a RIPK2 inhibitor leads to significantly improved intestinal inflammation in the murine TRUC model of colitis. A selective and potent inhibitor of the RIPK2 kinase may represent a new class of therapeutics that target microbiome-driven signaling for the treatment of IBD.


Asunto(s)
Colitis Ulcerosa/tratamiento farmacológico , Colon/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/antagonistas & inhibidores , Animales , Disponibilidad Biológica , Células Cultivadas , Colitis Ulcerosa/enzimología , Colitis Ulcerosa/genética , Colitis Ulcerosa/patología , Colon/enzimología , Colon/patología , Enfermedad de Crohn/enzimología , Enfermedad de Crohn/patología , Citocinas/genética , Citocinas/metabolismo , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Heces/química , Humanos , Mediadores de Inflamación/metabolismo , Lipocalinas/metabolismo , Ratones Endogámicos BALB C , Ratones Noqueados , Modelos Biológicos , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Inhibidores de Proteínas Quinasas/farmacocinética , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/genética , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/metabolismo , Proteínas de Dominio T Box/genética
5.
Gastroenterology ; 161(4): 1270-1287.e19, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34224738

RESUMEN

BACKGROUND & AIMS: The molecular checkpoints driving T cell activation and cytokine responses in ulcerative colitis (UC) are incompletely understood. Here, we studied the Tec kinase ITK in UC. METHODS: We analyzed patients with inflammatory bowel disease (n = 223) and evaluated ITK activity as well as the functional effects of cyclosporine-A (CsA). In addition, 3 independent murine colitis models were used to investigate the functional role of ITK. Finally, the activity of ITK was blocked via pharmacological inhibitors and genetically engineered mice. Readout parameters were mini-endoscopy, histopathology, mucosal T cell apoptosis, and cytokine production. RESULTS: We found an expansion of pITK-expressing mucosal CD4+ T cells in UC rather than Crohn's disease that correlated with disease severity. CsA suppressed activation of ITK in cultured CD4+ T cells and calcineurin-containing microclusters adjacent to the T cell receptor signaling complex. Functionally, the capacity of CsA to suppress activity of experimental colitis was critically dependent on ITK. Genetic inactivation of Itk via gene targeting or induction of allele-sensitive Itk mutants prevented experimental colitis in 3 colitis models, and treatment with pharmacological ITK blockers suppressed established colitis. In addition, ITK controlled apoptosis and activation of mucosal Th2 and Th17 lymphocytes via NFATc2 signaling pathways. CONCLUSIONS: ITK activation was detected in UC and could be down-regulated in cultured T cells by CsA administration. Selective targeting of ITK emerges as an attractive approach for treatment of chronic intestinal inflammation and potentially UC by driving resolution of mucosal inflammation.


Asunto(s)
Antiinflamatorios/farmacología , Colitis Ulcerosa/prevención & control , Colon/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Linfocitos Intraepiteliales/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Colitis Ulcerosa/enzimología , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/patología , Colon/enzimología , Colon/inmunología , Colon/patología , Ciclosporina/farmacología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Mucosa Intestinal/enzimología , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Linfocitos Intraepiteliales/enzimología , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/patología , Ratones Noqueados , Terapia Molecular Dirigida , Fosforilación , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal
6.
Cells ; 10(5)2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-34067858

RESUMEN

Inflammatory bowel diseases (IBDs) are serious disorders of which the etiologies are not, as yet, fully understood. In this study, Peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (Pin1) protein was shown to be dramatically upregulated in the colons of dextran sodium sulfate (DSS)-induced ulcerative colitis model mice. Interestingly, Pin1 knockout (KO) mice exhibited significant attenuation of DSS-induced colitis compared to wild-type (WT) mice, based on various parameters, including body weight, colon length, microscopic observation of the intestinal mucosa, inflammatory cytokine expression, and cleaved caspase-3. In addition, a role of Pin1 in inflammation was suggested because the percentage of M1-type macrophages in the colon was decreased in the Pin1 KO mice while that of M2-type macrophages was increased. Moreover, Pin1 KO mice showed downregulation of both Il17 and Il23a expression in the colon, both of which have been implicated in the development of colitis. Finally, oral administration of Pin1 inhibitor partially but significantly prevented DSS-induced colitis in mice, raising the possibility of Pin1 inhibitors serving as therapeutic agents for IBD.


Asunto(s)
Colitis/enzimología , Colon/enzimología , Mucosa Intestinal/enzimología , Peptidilprolil Isomerasa de Interacción con NIMA/metabolismo , Animales , Antiinflamatorios/farmacología , Colitis/inducido químicamente , Colitis/patología , Colitis/prevención & control , Colon/efectos de los fármacos , Colon/patología , Citocinas/metabolismo , Sulfato de Dextran , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Mediadores de Inflamación/metabolismo , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Peptidilprolil Isomerasa de Interacción con NIMA/antagonistas & inhibidores , Peptidilprolil Isomerasa de Interacción con NIMA/genética , Naftoquinonas/farmacología
7.
Gut Microbes ; 13(1): 1938380, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34190032

RESUMEN

The gut microbiota is essential for human health. Microbial supply of short-chain fatty acids (SCFAs), particularly butyrate, is a well-established contributor to gut homeostasis and disease resistance. Reaching millimolar luminal concentrations, butyrate is sequestered and utilized in the colon as the favored energy source for intestinal epithelia. Given the steep oxygen gradient across the anoxic lumen and the highly oxygenated lamina propria, the colon provides a particularly interesting environment to study oxygen sensing. Previous studies have shown that the transcription factor hypoxia-inducible factor (HIF) is stabilized in healthy colonic epithelia. Here we show that butyrate directly inhibits HIF prolyl hydroxylases (PHDs) to stabilize HIF. We find that butyrate stabilizes HIF in vitro despite eliminating ß-oxidation and resultant oxygen consumption. Using recombinant PHD protein in combination with nuclear magnetic resonance and enzymatic biochemical assays, we identify butyrate to bind and function as a unique, noncompetitive inhibitor of PHDs relative to other SCFAs. Butyrate inhibited PHD with a noncompetitive Ki of 5.3 ± 0.5 mM, a physiologically relevant concentration. We also confirm that microbiota-derived butyrate is necessary to stabilize HIF in mice colonic tissue through antibiotic-induced butyrate depletion and reconstitution experiments. Our results suggest that the co-evolution of mammals and mutualistic microbiota has selected for butyrate to impact a critical gene regulation pathway that can be extended beyond the mammalian gut. As PHDs are a major target for drug development in the stabilization of HIF, butyrate holds great potential as a well-tolerated endogenous inhibitor with far-reaching therapeutic impact.


Asunto(s)
Bacterias/metabolismo , Butiratos/química , Colon/microbiología , Microbioma Gastrointestinal , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/química , Inhibidores de Prolil-Hidroxilasa/química , Animales , Bacterias/clasificación , Bacterias/genética , Butiratos/metabolismo , Colon/enzimología , Colon/metabolismo , Ácidos Grasos Volátiles/metabolismo , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Mucosa Intestinal/enzimología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción , Oxígeno/metabolismo , Inhibidores de Prolil-Hidroxilasa/metabolismo
8.
Pflugers Arch ; 473(8): 1199-1211, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34075446

RESUMEN

Obesogenic diets (ODs) can affect AMPK activation in several sites as the colon, liver, and hypothalamus. OD intake can impair the hypothalamic AMPK regulation of energy homeostasis. Despite consuming ODs, not all subjects have the propensity to develop or progress to obesity. The obesity propensity is more associated with energy intake than expenditure dysregulations and may have a link with AMPK activity. While the effects of ODs are studied widely, few evaluate the short-term effects of terminating OD intake. Withdrawing from OD (WTD) is thought to improve or reverse the damages caused by the intake. Therefore, here we applied an OD intake and WTD protocol aiming to evaluate AMPK protein content and phosphorylation in the colon, liver, and hypothalamus and their relationship with obesity propensity. To this end, male Wistar rats (60 days) received control or high-sugar/high-fat (HSHF) OD for 30 days. Half of the animals were OD-withdrawn and fed the control diet for 48 h. After intake, we found a reduction in AMPK phosphorylation in the hypothalamus and colon, and after WTD, we found an increase in its hepatic and hypothalamic phosphorylation. The decrease in colon pAMPK/AMPK could be linked with hypothalamic pAMPK/AMPK after HSHF intake, while the increase in hepatic pAMPK/AMPK could have prevented the increase in hypothalamic pAMPK/AMPK. In the obesity-prone rats, we found higher levels of hypothalamic and colon pAMPK/AMPK despite the higher body mass gain. Our results highlight the relevance in multi-organ investigations and animal phenotype evaluation when studying the energy metabolism regulations.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Eje Cerebro-Intestino , Colon/enzimología , Hipotálamo/enzimología , Hígado/enzimología , Obesidad/enzimología , Animales , Dieta Alta en Grasa/efectos adversos , Azúcares de la Dieta/efectos adversos , Masculino , Obesidad/etiología , Ratas Wistar
9.
Gastroenterology ; 161(1): 271-286.e11, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33819483

RESUMEN

BACKGROUND & AIMS: Sulfation is a conjugation reaction essential for numerous biochemical and cellular functions in mammals. The 3'-phosphoadenosine 5'-phosphosulfate (PAPS) synthase 2 (PAPSS2) is the key enzyme to generate PAPS, which is the universal sulfonate donor for all sulfation reactions. The goal of this study was to determine whether and how PAPSS2 plays a role in colitis and colonic carcinogenesis. METHODS: Tissue arrays of human colon cancer specimens, gene expression data, and clinical features of cancer patients were analyzed. Intestinal-specific Papss2 knockout mice (Papss2ΔIE) were created and subjected to dextran sodium sulfate-induced colitis and colonic carcinogenesis induced by a combined treatment of azoxymethane and dextran sodium sulfate or azoxymethane alone. RESULTS: The expression of PAPSS2 is decreased in the colon cancers of mice and humans. The lower expression of PAPSS2 in colon cancer patients is correlated with worse survival. Papss2ΔIE mice showed heightened sensitivity to colitis and colon cancer by damaging the intestinal mucosal barrier, increasing intestinal permeability and bacteria infiltration, and worsening the intestinal tumor microenvironment. Mechanistically, the Papss2ΔIE mice exhibited reduced intestinal sulfomucin content. Metabolomic analyses revealed the accumulation of bile acids, including the Farnesoid X receptor antagonist bile acid tauro-ß-muricholic acid, and deficiency in the formation of bile acid sulfates in the colon of Papss2ΔIE mice. CONCLUSIONS: We have uncovered an important role of PAPSS2-mediated sulfation in colitis and colonic carcinogenesis. Intestinal sulfation may represent a potential diagnostic marker and PAPSS2 may serve as a potential therapeutic target for inflammatory bowel disease and colon cancer.


Asunto(s)
Neoplasias Asociadas a Colitis/prevención & control , Colitis/prevención & control , Colon/enzimología , Mucosa Intestinal/enzimología , Mucinas/metabolismo , Complejos Multienzimáticos/metabolismo , Sulfato Adenililtransferasa/metabolismo , Animales , Ácidos y Sales Biliares/metabolismo , Colitis/enzimología , Colitis/genética , Colitis/patología , Neoplasias Asociadas a Colitis/enzimología , Neoplasias Asociadas a Colitis/genética , Neoplasias Asociadas a Colitis/patología , Colon/patología , Bases de Datos Genéticas , Modelos Animales de Enfermedad , Humanos , Mucosa Intestinal/patología , Metaboloma , Metabolómica , Ratones Endogámicos C57BL , Ratones Noqueados , Complejos Multienzimáticos/genética , Pronóstico , Receptores Citoplasmáticos y Nucleares/metabolismo , Sulfato Adenililtransferasa/genética
10.
Medicine (Baltimore) ; 100(17): e25658, 2021 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-33907129

RESUMEN

ABSTRACT: The aim of this study was to investigate the expression of phosphatase of regenerating live-3 (PRL-3) in human stage III colorectal cancer (CRC) and to evaluate its correlation with metachronous liver metastasis (MLM) and prognosis.The retrospective cohort study included 116 stage III CRC primary tumors and 60 normal colorectal tissues. PRL-3 expression was measured by immunohistochemistry. We investigated the correlation of PRL-3 with clinicopathologic features by the chi-square test. The association of PRL-3 expression with MLM was assessed by binary logistic regression. Overall survival (OS) and disease-free survival (DFS) between patients with positive PRL-3 expression and those with negative PRL-3 expression were compared by the Kaplan-Meier method and Cox proportional hazards regression model.We found that 32.8% of stage III CRC primary tumors were PRL-3 positive, and 15.0% of normal colorectal epithelia showed high PRL-3 expression (P = .012). Seventeen tumors (47.2%) among 36 cases that developed MLM were PRL-3 positive, and only 21 tumors (26.3%) in the 80 cases that did not develop MLM had positive PRL-3 expression (P = .026). PRL-3 expression was associated with MLM (P = .028). Patients with positive expression of PRL-3 showed a significantly shorter OS (40.32 ±â€Š3.97 vs 53.96 ±â€Š2.77 months, P = .009) and DFS (34.97 ±â€Š4.30 vs 44.48 ±â€Š2.89 months, P = .036). A multivariate analysis indicated that PRL-3 expression was an independent unfavorable prognostic factor for OS (P = .007).Our study suggested that high PRL-3 expression is an independent risk factor for MLM and poor prognosis. PRL-3 is expected to be a promising biomarker for predicting the incidence of MLM and prognosis in patients with stage III CRC.


Asunto(s)
Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/mortalidad , Proteínas de Neoplasias/metabolismo , Neoplasias Primarias Secundarias/enzimología , Neoplasias Primarias Secundarias/mortalidad , Proteínas Tirosina Fosfatasas/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/metabolismo , Colon/enzimología , Neoplasias Colorrectales/patología , Supervivencia sin Enfermedad , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Análisis Multivariante , Estadificación de Neoplasias , Neoplasias Primarias Secundarias/patología , Valor Predictivo de las Pruebas , Pronóstico , Modelos de Riesgos Proporcionales , Recto/enzimología , Estudios Retrospectivos , Factores de Riesgo
11.
Biomed Pharmacother ; 139: 111127, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33819810

RESUMEN

Inflammatory bowel disease (IBD), including Crohn's disease (CD) and ulcerative colitis (UC), is rising constantly all over the world. However, current medical treatments are not universally practical. Microcin J25 (MccJ25), a member of the lasso peptides class, has excellent antimicrobial activity both in vitro and in vivo. Here, we assessed the anti-inflammatory effects of MccJ25 through DSS-induced UC mouse model. MccJ25 significantly ameliorated the UC-associated parameters such as decreased body weight, increased disease activity index (DAI) and shortened colon length. MccJ25 also provides barrier protection by preserving structural integrity and reducing inflammatory infiltrates of colon epithelium. The underlying mechanism may be associated with gut microbiota. To test this uncertainty, co-housing experiment was performed, and results indicate homogenized microbiota could relief the inflammatory. Meanwhile, we also proved the prominent role of the possible targets of MccJ25, namely genus Lactobacillus, Bacteroides and Akkermansia (as well as the possible strains related to the important OTUs) in inflammation status through comprehensive analysis. In conclusion, MccJ25 effectively attenuates inflammation and improves disrupted barrier function, and the MccJ25-modified gut microbiota plays a central role in this process.


Asunto(s)
Antiinflamatorios/uso terapéutico , Bacteriocinas/uso terapéutico , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Proteínas Recombinantes/uso terapéutico , Animales , Colitis Ulcerosa/inducido químicamente , Colon/enzimología , Colon/patología , Sulfato de Dextran , Femenino , Mucosa Intestinal/enzimología , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Citotóxicas Formadoras de Poros
12.
Toxicol Appl Pharmacol ; 418: 115495, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33741346

RESUMEN

In the present study, the effects of NLRP3 on radiation-induced tissue damage, including colon and skin damage in mice, and the possible mechanisms were explored in vivo and in vitro. The mice were subjected to whole abdomen radiation by timed exposure to X-ray at a cumulative dose of 14 Gy. The survival rate showed that NLRP3 deficiency increased the mortality rate in mice. Furthermore, colon damage, evaluated by H&E staining and barrier function analysis, were significantly aggravated by NLRP3 deficiency. Enhanced phosphorylation of p-TBK1 and p-IRF3 in colonic tissue as well as elevated IFN-ß levels in the serum indicated hyperactivation of cGAS-STING signaling. Moreover, radiation-induced expression of p-TBK1, p-IRF3, and IFN-ß in BMDMs increased in vitro after NLRP3 knockout. Thus, our study outcomes suggest that NLRP3 may protect mice from radiation-induced tissue damage via attenuating cGAS-STING signaling.


Asunto(s)
Colon/efectos de la radiación , Macrófagos/efectos de la radiación , Proteínas de la Membrana/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Nucleotidiltransferasas/metabolismo , Traumatismos por Radiación/prevención & control , Úlcera Cutánea/prevención & control , Piel/efectos de la radiación , Animales , Células Cultivadas , Colon/enzimología , Colon/patología , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/metabolismo , Macrófagos/enzimología , Macrófagos/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Traumatismos por Radiación/enzimología , Traumatismos por Radiación/genética , Traumatismos por Radiación/patología , Transducción de Señal , Piel/enzimología , Piel/patología , Úlcera Cutánea/enzimología , Úlcera Cutánea/genética , Úlcera Cutánea/patología
13.
PLoS One ; 16(3): e0248730, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33725024

RESUMEN

COVID-19 (coronavirus disease 2019) patients exhibiting gastrointestinal symptoms are reported to have worse prognosis. Ace2 (angiotensin-converting enzyme 2), the gene encoding the host protein to which SARS-CoV-2 spike proteins bind, is expressed in the gut and therefore may be a target for preventing or reducing severity of COVID-19. Here we test the hypothesis that Ace2 expression in the gastrointestinal and respiratory tracts is modulated by the microbiome. We used quantitative PCR to profile Ace2 expression in germ-free mice, conventional raised specific pathogen-free mice, and gnotobiotic mice colonized with different microbiota. Intestinal Ace2 expression levels were significantly higher in germ-free mice compared to conventional mice. A similar trend was observed in the respiratory tract. Intriguingly, microbiota depletion via antibiotics partially recapitulated the germ-free phenotype, suggesting potential for microbiome-mediated regulation of Ace2 expression. Variability in intestinal Ace2 expression was observed in gnotobiotic mice colonized with different microbiota, partially attributable to differences in microbiome-encoded proteases and peptidases. Together, these data suggest that the microbiome may be one modifiable factor determining COVID-19 infection risk and disease severity.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Colon/enzimología , Microbioma Gastrointestinal , Intestino Delgado/enzimología , Pulmón/enzimología , Proteína de la Poliposis Adenomatosa del Colon/deficiencia , Proteína de la Poliposis Adenomatosa del Colon/genética , Enzima Convertidora de Angiotensina 2/genética , Animales , Femenino , Expresión Génica , Interleucina-10/deficiencia , Interleucina-10/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
14.
Glycoconj J ; 38(1): 129-134, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33608771

RESUMEN

Sialic acid and its associated metabolic enzymes have emerged as important components of the pathophysiology of type 2 diabetes (T2D). There is an elevation in the serum concentration of sialic acid in humans and animals with T2D. The present study investigated the modulation of mRNA expression level of UDP-N-acetylglucosamine-2-epimerase/N-acetylmannosamine kinase (GNE) and neuraminidase 1 (NEU1) genes in some organs of type 2 diabetic rats. T2D was induced using fructose-streptozotocin model and eight weeks after the induction of diabetes, sialic acid was assayed in the blood and organs (adipose tissue, brain, colon, kidney, liver, pancreas, skeletal muscle and spleen) followed by quantification of mRNA expression level of GNE and NEU1 genes by qPCR. The results showed a significant (P < 0.05) increase in sialic acid level in the serum and all the afore-mentioned organs investigated except in the adipose tissue and skeletal muscle of the diabetic rats compared the normal control. The expression GNE gene was only increased in the pancreas (1.8-fold) of the diabetic rats while there was a decrease in the expression of the gene in the colon. In contrast, the expression of NEU1 gene was increased in the spleen (3.5-fold), brain (2.2-fold), liver (1.9-fold), colon (1.5-fold) and kidney of the diabetic rats. It was concluded that the elevated level of sialic acid in the organs of diabetic rats, except the pancreas, might not be due to increased endogenous synthesis of sialic acid.


Asunto(s)
Diabetes Mellitus Tipo 2/enzimología , Complejos Multienzimáticos/genética , Animales , Encéfalo/enzimología , Colon/enzimología , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Tipo 2/genética , Regulación Enzimológica de la Expresión Génica , Hígado/enzimología , Ácido N-Acetilneuramínico/sangre , Ácido N-Acetilneuramínico/metabolismo , Neuraminidasa/genética , Páncreas/enzimología , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Bazo/enzimología
15.
Cancer Prev Res (Phila) ; 14(1): 65-76, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32917645

RESUMEN

Increased COX-2 and decreased 15-hydroxyprostaglandin dehydrogenase (15-HPGD) expression promote prostaglandin-mediated inflammation and colorectal carcinogenesis. Experimental studies suggest that vitamin D and calcium may inhibit these pathways, but their effects on colorectal tissue COX-2 and 15-HPGD expression in humans are unknown. We tested the effects of supplemental vitamin D (1,000 IU/day) and/or calcium (1,200 mg/day) on COX-2 and 15-HPGD expression in the morphologically normal rectal mucosa from 62 paients with colorectal adenoma in a placebo-controlled chemoprevention trial. We measured biomarker expression using automated IHC and quantitative image analysis at baseline and 1-year follow-up, and assessed treatment effects using mixed linear models. The primary outcome was the COX-2/15-HPGD expression ratio, because these enzymes function as physiologic antagonists. After 1 year of treatment, the mean COX-2/15-HPGD expression ratio in full-length crypts proportionately decreased 47% in the vitamin D group (P = 0.001), 46% in the calcium group (P = 0.002), and 34% in the calcium + vitamin D group (P = 0.03), relative to the placebo group. Among individuals with the functional vitamin D-binding protein isoform DBP2 (GC rs4588*A), the COX-2/15-HPDG ratio decreased 70% (P = 0.0006), 75% (P = 0.0002), and 60% (P = 0.006) in the vitamin D, calcium, and combined supplementation groups, respectively, relative to placebo. These results show that vitamin D and calcium favorably modulate the balance of expression of COX-2 and 15-HPGD-biomarkers of inflammation that are strongly linked to colorectal carcinogenesis-in the normal-appearing colorectal mucosa of patients with colorectal adenoma (perhaps especially those with the DBP2 isoform). PREVENTION RELEVANCE: Supplemental calcium and vitamin D reduce indicators of cancer-promoting inflammation in normal colorectal tissue in humans, thus furthering our understanding of how they may help prevent colorectal cancer.


Asunto(s)
Adenoma/prevención & control , Carbonato de Calcio/administración & dosificación , Neoplasias Colorrectales/prevención & control , Mucosa Intestinal/inmunología , Vitamina D/administración & dosificación , Adenoma/inmunología , Adenoma/patología , Anciano , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/metabolismo , Colon/efectos de los fármacos , Colon/enzimología , Colon/inmunología , Colon/patología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Ciclooxigenasa 2/análisis , Ciclooxigenasa 2/metabolismo , Suplementos Dietéticos , Femenino , Estudios de Seguimiento , Humanos , Hidroxiprostaglandina Deshidrogenasas/análisis , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Inflamación/diagnóstico , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/patología , Mucosa Intestinal/enzimología , Mucosa Intestinal/patología , Masculino , Persona de Mediana Edad , Recto/efectos de los fármacos , Recto/enzimología , Recto/inmunología , Recto/patología , Resultado del Tratamiento
16.
Eur J Pharmacol ; 892: 173763, 2021 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-33249075

RESUMEN

Colorectal cancer is one of the primary causes of cancer-related mortality worldwide. The tumor microenvironment contains growth factors; inflammatory chemokines, matrix metalloproteinases, and pro-oxidants leading to cancer development and progression. Phytochemicals have been used as the main source of anti-cancer agents. Accordingly, the effect of two natural flavonoids (Chrysin and Daidzein) was investigated on the level of amphiregulin (AREG), chemokine ligand (CXCL1), and matrix metalloproteinase-9 (MMP-9) in 1, 2-dimethylhydrazine dihydrochloride (DMH) induced colorectal cancer. Rats were injected by DMH (40 mg/kg/week S.C.) for 16 weeks concomitantly with 2% dextran sodium sulfate (DSS) in drinking water for three cycles. Rats were orally treated with chrysin (125 and 250 mg/kg) and daidzein (5 and10 mg/kg) three times/week for the last 8 weeks. DMH + DSS group showed a significant (P < 0.05) increase in the levels of AREG (2386 ± 18 vs 1377 ± 10 pg/ml), CXCL1 (18 ± 0.9 vs 6 ± 0.83 g/ml), MMP-9 (1355 ± 88 vs 452 ± 7 pg/ml) compared to normal rats. These findings were associated with a potent antioxidant activity against cytochrome P450 2E1; (CYP2E1). Histopathological findings of the DMH + DSS group showed focal hyperplasia of the mucosa lining overlying crypts with moderate inflammation, dysplastic epithelial cells, and loss of goblet cells. Chrysin and daidzein treatment significantly (P < 0.05) restored the biochemical alterations and reverted histopathological findings near to the normal status. Moreover, chrysin and daidzein exerted anticancer activity against SW620 cells that were associated with decreased the protein expression of p-ERK/ERK and p-AKT/AKT. In conclusion, this study highlighted the potential anticancer role of chrysin and daidzein in the treatment of colon cancer.


Asunto(s)
Anfirregulina/metabolismo , Antineoplásicos Fitogénicos/farmacología , Quimiocina CXCL1/metabolismo , Colon/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Flavonoides/farmacología , Isoflavonas/farmacología , Metaloproteinasa 9 de la Matriz/metabolismo , 1,2-Dimetilhidrazina , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica/inducido químicamente , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Colon/enzimología , Colon/patología , Neoplasias Colorrectales/inducido químicamente , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/patología , Citocromo P-450 CYP2E1/metabolismo , Sulfato de Dextran , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Masculino , Estrés Oxidativo/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Transducción de Señal
17.
Int Immunopharmacol ; 90: 107212, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33310666

RESUMEN

BACKGROUND: The pathogenesis of inflammatory bowel diseases (IBD), including Crohn's disease (CD) and ulcerative colitis (UC) has not been fully elucidated. However, a strong correlation between IBD and high T helper 17 (Th17) levels has been found. Sirtuin 2 (SIRT2) has recently been found to play an important role in metabolic reprogramming, but its potential anti-inflammatory properties remain unclear. METHODS: The expression levels of SIRT2 and glucose metabolism-related proteins in peripheral blood mononuclear cells (PBMCs) of IBD patients and healthy volunteers were detected. Human PBMCs were differentiated into Th17 cells in vitro and were treated with TM simultaneously. The ratio of Th17 cells and apoptotic cells and the production of Interleukin (IL)-17A and the expression levels of transcription factors of classical signaling pathway related to Th17 differentiation were determined. The acetylation of LDHA and glucose metabolism was assessed. Subsequently, C57BL/6J colitis mice induced by 2.5% dextran sulfatesodiumsalt (DSS) were treated with or without TM, Disease activity index, T cell subsets in the mice spleen, relevant inflammatory cytokines in serum, specific mRNA, and proteins in mice colon were evaluated respectively. RESULTS: SIRT2 and glucose metabolism-related proteins in PBMCs of patients were overexpressed. Compared with the positive control group, human PBMCs treated with TM had lower levels of IL-17A, percentage of Th17 cells, levels of phospho-signal transducer and activator of transcription (p-STAT) 3 and phospho-nuclear transcription factor-κB (p-NF-κB), but higher levels of acetylated LDHA. Compared with colitis mice, TM-treated colitis mice had longer colons, reduced weight-losses, and lower disease activity index and histopathologic scores. Interestingly, although the expression levels of interferon (IFN)-γ, IL-17A, and retinoic acid receptor-related orphan receptor (ROR)-γt were inhibited in the colons of TM-treated colitis mice, the expression of forkhead box protein P3 (FOXP3) didn't change. Consistently, relative to the high percentage of splenic Th17 cells in colitis mice, the percentage of splenic Th17 cells in TM-treated colitis mice was as normal as PBS-treated mice, while the percentage of Treg cells was not affected. Additionally, the TM group had reduced levels of IL-23 and hypoxiainduciblefactor-1α (HIF-1α), and an increased level of IL-10 in the colon, compared with the colitis group. CONCLUSION: Our results indicate that TM reduces UC progression by reducing metabolic reprogramming and T cell differentiation. Specifically, TM prevented Th17 differentiation by reducing the expression of related transcription factors and promoting acetylation of LDHA (weakening glycolysis). SIRT2 may be a potential target for IBD treatment.


Asunto(s)
Antiinflamatorios/farmacología , Diferenciación Celular/efectos de los fármacos , Reprogramación Celular/efectos de los fármacos , Colitis/prevención & control , Colon/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Sirtuina 2/metabolismo , Células Th17/efectos de los fármacos , Acetilación , Animales , Apoptosis/efectos de los fármacos , Estudios de Casos y Controles , Células Cultivadas , Colitis/enzimología , Colitis/inmunología , Colitis/patología , Colon/enzimología , Colon/inmunología , Colon/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Mediadores de Inflamación/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Sirtuina 2/genética , Células Th17/enzimología , Células Th17/inmunología
18.
Int Immunopharmacol ; 90: 107128, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33191180

RESUMEN

Sunitinib is a tyrosine kinase inhibitor for many tumors. Inflammation is one of the most important factors in the development of intestinal tumors. Many inflammation-related factors are regulated by tyrosine kinase receptors. It is reasonable to hypothesize that sunitinib can regulate the development of intestinal tumors by regulating the expression and/or activity of inflammation-related factors. Here, ApcMin/+ male mouse model was used to investigate the effect and mechanism of sunitinib malate against intestinal cancer. Results show that compared to vehicle, after sunitinib malate treatment, overall survival of ApcMin/+ mice was lengthened up to 25 days, with a gain of body weight, reduction of spleen/body weight index, and RBC, WBC and HGC regulated to normal levels of wild type mice, and a number of polyps no less than 1 mm significantly reduced. Meanwhile, in the intestines, the nuclear ß-Catenin protein and c-Myc mRNA were both down-regulated, and Bcl-6 was significantly reduced with Caspase-3 up regulated. Furthermore, inflammation-related factors including IL-6, TNF-α, IL-1α, IL-1ß and IFN-γ were down-regulated at mRNA levels in the intestines. These results suggest that sunitinib malate can significantly improve the survival status and inhibit intestinal tumor development in male ApcMin/+ mice, through inhibiting inflammation-related factors, while suppressing ß-cateinin/c-Myc pathway and re-balancing protein levels of Bcl-6 and Caspase-3.


Asunto(s)
Antiinflamatorios/farmacología , Anticarcinógenos/farmacología , Caspasa 3/metabolismo , Colon/efectos de los fármacos , Citocinas/metabolismo , Mediadores de Inflamación/metabolismo , Neoplasias Intestinales/prevención & control , Pólipos Intestinales/prevención & control , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Sunitinib/farmacología , beta Catenina/metabolismo , Animales , Colon/enzimología , Colon/patología , Citocinas/genética , Regulación de la Expresión Génica , Genes APC , Neoplasias Intestinales/enzimología , Neoplasias Intestinales/genética , Neoplasias Intestinales/patología , Pólipos Intestinales/enzimología , Pólipos Intestinales/genética , Pólipos Intestinales/patología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-myc/genética , Transducción de Señal
19.
Biol Pharm Bull ; 43(12): 1945-1949, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33268713

RESUMEN

Isoflavones have many biological activities and are major bioactive components of kakkonto, a traditional Japanese herbal medicine. We previously reported that the combined therapy of oral immune therapy (OIT) and kakkonto downregulates the mRNA expression of Cyp26b1, a major retinoic acid (RA)-degrading enzyme, in the colon of food allergy mice and thereby ameliorates allergic symptoms. In this study, we evaluated the effects of various isoflavones on Cyp26b1 expression in primary cultured lamina propria (LP) cells isolated from the mouse colon. The mRNA expression of Cyp26b1 was extremely downregulated by all isoflavones tested in the LP cells except for puerarin. In particular, genistein and genistin markedly suppressed Cyp26b1 mRNA expression without affecting RA-synthesizing enzyme expression. Moreover, to evaluate the effects of isoflavones on allergic reactions, genistein and genistin were administered to ovalbumin (OVA)-induced food allergy mice. Oral administration of genistin suppressed the development of allergic symptoms. These results raise the possibility that isoflavones elevated the level of RA in the colon by inhibiting RA degradation and then the high concentration of RA in the colon might exert immunosuppressive and antiallergic effects on food allergy mice.


Asunto(s)
Colon/efectos de los fármacos , Colon/enzimología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/enzimología , Isoflavonas/farmacología , Ácido Retinoico 4-Hidroxilasa/biosíntesis , Animales , Hipersensibilidad a los Alimentos/tratamiento farmacológico , Hipersensibilidad a los Alimentos/enzimología , Hipersensibilidad a los Alimentos/etiología , Regulación Enzimológica de la Expresión Génica , Isoflavonas/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Membrana Mucosa/efectos de los fármacos , Membrana Mucosa/enzimología , Ovalbúmina/toxicidad , Ácido Retinoico 4-Hidroxilasa/antagonistas & inhibidores
20.
Cell Physiol Biochem ; 54(6): 1132-1142, 2020 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-33175479

RESUMEN

BACKGROUND/AIMS: The colonic H+, K+ ATPase (HKA2) is a heterodimeric membrane protein that exchanges luminal K+ for intracellular H+ and is involved in maintaining potassium homeostasis. Under homeostatic conditions, the colonic HKA2 remains inactive, since most of the potassium is absorbed by the small intestine. In diarrheal states, potassium is secreted and compensatory potassium absorption becomes necessary. This study proposes a novel mechanism whereby the addition of penicillin G sodium salt (penG) to colonic crypts stimulates potassium uptake in the presence of intracellular nitric oxide (NO), under sodium-free (0-Na+) conditions. METHODS: Sprague Dawley rat colonic crypts were isolated and pHi changes were monitored through the ammonium prepulse technique. Increased proton extrusion in 0-Na+ conditions reflected heightened H+, K+ ATPase activity. Colonic crypts were exposed to penG, L-arginine (a NO precursor), and N-nitro l-arginine methyl ester (L-NAME, a NO synthase inhibitor). RESULTS: Isolated administration of penG significantly increased H+, K+ ATPase activity from baseline, p 0.0067. Co-administration of arginine and penG in 0-Na+ conditions further upregulated H+, K+ ATPase activity, p <0.0001. Crypt perfusion with L-NAME and penG demonstrated a significant reduction in H+, K+ ATPase activity, p 0.0058. CONCLUSION: Overall, acute exposure of colonic crypts to penG activates the H+, K+ ATPase in the presence of NO. This study provides new insights into colonic potassium homeostasis.


Asunto(s)
Colon/enzimología , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Óxido Nítrico/metabolismo , Penicilina G/farmacología , Animales , Arginina/farmacología , Masculino , NG-Nitroarginina Metil Éster/farmacología , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...