Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Biol Chem ; 293(9): 3073-3087, 2018 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-29317503

RESUMEN

The gut microbiome contributes to inflammatory bowel disease (IBD), in which bacteria can be present within the epithelium. Epithelial barrier function is decreased in IBD, and dysfunctional epithelial mitochondria and endoplasmic reticulum (ER) stress have been individually associated with IBD. We therefore hypothesized that the combination of ER and mitochondrial stresses significantly disrupt epithelial barrier function. Here, we treated human colonic biopsies, epithelial colonoids, and epithelial cells with an uncoupler of oxidative phosphorylation, dinitrophenol (DNP), with or without the ER stressor tunicamycin and assessed epithelial barrier function by monitoring internalization and translocation of commensal bacteria. We also examined barrier function and colitis in mice exposed to dextran sodium sulfate (DSS) or DNP and co-treated with DAPK6, an inhibitor of death-associated protein kinase 1 (DAPK1). Contrary to our hypothesis, induction of ER stress (i.e. the unfolded protein response) protected against decreased barrier function caused by the disruption of mitochondrial function. ER stress did not prevent DNP-driven uptake of bacteria; rather, specific mobilization of the ATF6 arm of ER stress and recruitment of DAPK1 resulted in enhanced autophagic killing (xenophagy) of bacteria. Of note, epithelia with a Crohn's disease-susceptibility mutation in the autophagy gene ATG16L1 exhibited less xenophagy. Systemic delivery of the DAPK1 inhibitor DAPK6 increased bacterial translocation in DSS- or DNP-treated mice. We conclude that promoting ER stress-ATF6-DAPK1 signaling in transporting enterocytes counters the transcellular passage of bacteria evoked by dysfunctional mitochondria, thereby reducing the potential for metabolic stress to reactivate or perpetuate inflammation.


Asunto(s)
Proteínas Quinasas Asociadas a Muerte Celular/metabolismo , Estrés del Retículo Endoplásmico , Mitocondrias/metabolismo , Factor de Transcripción Activador 6/metabolismo , Anciano , Animales , Línea Celular Tumoral , Estrés del Retículo Endoplásmico/efectos de los fármacos , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Escherichia coli/efectos de los fármacos , Escherichia coli/fisiología , Femenino , Humanos , Masculino , Ratones , Mitocondrias/efectos de los fármacos , Fosforilación Oxidativa/efectos de los fármacos , Permeabilidad , Tunicamicina/farmacología
2.
Am J Physiol Gastrointest Liver Physiol ; 313(5): G467-G475, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28751424

RESUMEN

Cancer cell lines have been the mainstay of intestinal epithelial experimentation for decades, due primarily to their immortality and ease of culture. However, because of the inherent biological abnormalities of cancer cell lines, many cellular biologists are currently transitioning away from these models and toward more representative primary cells. This has been particularly challenging, but recent advances in the generation of intestinal organoids have brought the routine use of primary cells within reach of most epithelial biologists. Nevertheless, even with the proliferation of publications that use primary intestinal epithelial cells, there is still a considerable amount of trial and error required for laboratories to establish a consistent and reliable method to culture three-dimensional (3D) intestinal organoids and primary epithelial monolayers. We aim to minimize the time other laboratories spend troubleshooting the technique and present a standard method for culturing primary epithelial cells. Therefore, we have described our optimized, high-yield, cost-effective protocol to grow 3D murine colonoids for more than 20 passages and our detailed methods to culture these cells as confluent monolayers for at least 14 days, enabling a wide variety of potential future experiments. By supporting and expanding on the current literature of primary epithelial culture optimization and detailed use in experiments, we hope to help enable the widespread adoption of these innovative methods and allow consistency of results obtained across laboratories and institutions.NEW & NOTEWORTHY Primary intestinal epithelial monolayers are notoriously difficult to maintain culture, even with the recent advances in the field. We describe, in detail, the protocols required to maintain three-dimensional cultures of murine colonoids and passage these primary epithelial cells to confluent monolayers in a standardized, high-yield and cost-effective manner.


Asunto(s)
Colon , Células Epiteliales , Mucosa Intestinal , Organoides , Cultivo Primario de Células/métodos , Animales , Células Cultivadas , Colon/patología , Colon/fisiología , Células Epiteliales/patología , Células Epiteliales/fisiología , Mucosa Intestinal/patología , Mucosa Intestinal/fisiología , Ratones , Organoides/patología , Organoides/fisiología
3.
Gastroenterology ; 151(5): 933-944.e3, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27480173

RESUMEN

BACKGROUND & AIMS: The 5-hydroxytryptamine receptor 4 (5-HT4R or HTR4) is expressed in the colonic epithelium but little is known about its functions there. We examined whether activation of colonic epithelial 5-HT4R protects colons of mice from inflammation. METHODS: The 5-HT4R agonist tegaserod (1 mg/kg), the 5-HT4R antagonist GR113808 (1 mg/kg), or vehicle (control) were delivered by enema to wild-type or 5-HT4R knockout mice at the onset of, or during, active colitis, induced by administration of dextran sodium sulfate or trinitrobenzene sulfonic acid. Inflammation was measured using the colitis disease activity index and by histologic analysis of intestinal tissues. Epithelial proliferation, wound healing, and resistance to oxidative stress-induced apoptosis were assessed, as was colonic motility. RESULTS: Rectal administration of tegaserod reduced the severity of colitis compared with mice given vehicle, and accelerated recovery from active colitis. Rectal tegaserod did not improve colitis in 5-HT4R knockout mice, and intraperitoneally administered tegaserod did not protect wild-type mice from colitis. Tegaserod increased proliferation of crypt epithelial cells. Stimulation of 5-HT4R increased Caco-2 cell migration and reduced oxidative stress-induced apoptosis; these actions were blocked by co-administration of the 5-HT4R antagonist GR113808. In noninflamed colons of wild-type mice not receiving tegaserod, inhibition of 5-HT4Rs resulted in signs of colitis within 3 days. In these mice, epithelial proliferation decreased and bacterial translocation to the liver and spleen was detected. Daily administration of tegaserod increased motility in inflamed colons of guinea pigs and mice, whereas administration of GR113808 disrupted motility in animals without colitis. CONCLUSIONS: 5-HT4R activation maintains motility in healthy colons of mice and guinea pigs, and reduces inflammation in colons of mice with colitis. Agonists might be developed as treatments for patients with inflammatory bowel diseases.


Asunto(s)
Colitis/metabolismo , Colon/metabolismo , Mucosa Intestinal/metabolismo , Receptores de Serotonina 5-HT4/metabolismo , Agonistas del Receptor de Serotonina 5-HT4/farmacología , Antagonistas del Receptor de Serotonina 5-HT4/farmacología , Administración Rectal , Animales , Colitis/inducido químicamente , Colitis/patología , Colitis/prevención & control , Colon/efectos de los fármacos , Colon/patología , Sulfato de Dextran , Femenino , Cobayas , Indoles/farmacología , Indoles/uso terapéutico , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Noqueados , Agonistas del Receptor de Serotonina 5-HT4/uso terapéutico , Índice de Severidad de la Enfermedad , Sulfonamidas/farmacología , Ácido Trinitrobencenosulfónico
4.
J Neurosci ; 35(30): 10821-30, 2015 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-26224864

RESUMEN

Patients with systemic inflammatory diseases (e.g., rheumatoid arthritis, inflammatory bowel disease, chronic liver disease) commonly develop debilitating symptoms (i.e., sickness behaviors) that arise from changes in brain function. The microbiota-gut-brain axis alters brain function and probiotic ingestion can influence behavior. However, how probiotics do this remains unclear. We have previously described a novel periphery-to-brain communication pathway in the setting of peripheral organ inflammation whereby monocytes are recruited to the brain in response to systemic TNF-α signaling, leading to microglial activation and subsequently driving sickness behavior development. Therefore, we investigated whether probiotic ingestion (i.e., probiotic mixture VSL#3) alters this periphery-to-brain communication pathway, thereby reducing subsequent sickness behavior development. Using a well characterized mouse model of liver inflammation, we now show that probiotic (VSL#3) treatment attenuates sickness behavior development in mice with liver inflammation without affecting disease severity, gut microbiota composition, or gut permeability. Attenuation of sickness behavior development was associated with reductions in microglial activation and cerebral monocyte infiltration. These events were paralleled by changes in markers of systemic immune activation, including decreased circulating TNF-α levels. Our observations highlight a novel pathway through which probiotics mediate cerebral changes and alter behavior. These findings allow for the potential development of novel therapeutic interventions targeted at the gut microbiome to treat inflammation-associated sickness behaviors in patients with systemic inflammatory diseases. SIGNIFICANCE STATEMENT: This research shows that probiotics, when eaten, can improve the abnormal behaviors (including social withdrawal and immobility) that are commonly associated with inflammation. Probiotics are able to cause this effect within the body by changing how the immune system signals the brain to alter brain function. These findings broaden our understanding of how probiotics may beneficially affect brain function in the context of inflammation occurring within the body and may open potential new therapeutic alternatives for the treatment of these alterations in behavior that can greatly affect patient quality of life.


Asunto(s)
Encéfalo/efectos de los fármacos , Conducta de Enfermedad/efectos de los fármacos , Sistema Inmunológico/efectos de los fármacos , Neuroinmunomodulación/efectos de los fármacos , Probióticos/farmacología , Animales , Conducta Animal , Encéfalo/inmunología , Modelos Animales de Enfermedad , Inflamación/complicaciones , Masculino , Ratones , Ratones Endogámicos C57BL
5.
Gastroenterology ; 149(2): 445-55.e3, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25865048

RESUMEN

BACKGROUND & AIMS: Disturbances in the control of ion transport lead to epithelial barrier dysfunction in patients with colitis. Enteric glia regulate intestinal barrier function and colonic ion transport. However, it is not clear whether enteric glia are involved in epithelial hyporesponsiveness. We investigated enteric glial regulation of ion transport in mice with trinitrobenzene sulfonic acid- or dextran sodium sulfate-induced colitis and in Il10(-/-) mice. METHODS: Electrically evoked ion transport was measured in full-thickness segments of colon from CD1 and Il10(-/-) mice with or without colitis in Ussing chambers. Nitric oxide (NO) production was assessed using amperometry. Bacterial translocation was investigated in the liver, spleen, and blood of mice. RESULTS: Electrical stimulation of the colon evoked a tetrodotoxin-sensitive chloride secretion. In mice with colitis, ion transport almost completely disappeared. Inhibiting inducible NO synthase (NOS2), but not neuronal NOS (NOS1), partially restored the evoked secretory response. Blocking glial function with fluoroacetate, which is not a NOS2 inhibitor, also partially restored ion transport. Combined NOS2 inhibition and fluoroacetate administration fully restored secretion. Epithelial responsiveness to vasoactive intestinal peptide was increased after enteric glial function was blocked in mice with colitis. In colons of mice without colitis, NO was produced in the myenteric plexus almost completely via NOS1. NO production was increased in mice with colitis, compared with mice without colitis; a substantial proportion of NOS2 was blocked by fluoroacetate administration. Inhibition of enteric glial function in vivo reduced the severity of trinitrobenzene sulfonic acid-induced colitis and associated bacterial translocation. CONCLUSIONS: Increased production of NOS2 in enteric glia contributes to the dysregulation of intestinal ion transport in mice with colitis. Blocking enteric glial function in these mice restores epithelial barrier function and reduces bacterial translocation.


Asunto(s)
Colitis/metabolismo , Sistema Nervioso Entérico/citología , Transporte Iónico , Neuroglía/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Óxido Nítrico/metabolismo , Animales , Traslocación Bacteriana , Colitis/inducido químicamente , Colitis/genética , Modelos Animales de Enfermedad , Estimulación Eléctrica/métodos , Fluoroacetatos/administración & dosificación , Interleucina-10/deficiencia , Interleucina-10/genética , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Plexo Mientérico/citología , Neuroglía/citología
6.
Am J Pathol ; 184(9): 2516-27, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25034594

RESUMEN

Epithelial permeability is often increased in inflammatory bowel diseases. We hypothesized that perturbed mitochondrial function would cause barrier dysfunction and hence epithelial mitochondria could be targeted to treat intestinal inflammation. Mitochondrial dysfunction was induced in human colon-derived epithelial cell lines or colonic biopsy specimens using dinitrophenol, and barrier function was assessed by transepithelial flux of Escherichia coli with or without mitochondria-targeted antioxidant (MTA) cotreatment. The impact of mitochondria-targeted antioxidants on gut permeability and dextran sodium sulfate (DSS)-induced colitis in mice was tested. Mitochondrial superoxide evoked by dinitrophenol elicited significant internalization and translocation of E. coli across epithelia and control colonic biopsy specimens, which was more striking in Crohn's disease biopsy specimens; the mitochondria-targeted antioxidant, MitoTEMPO, inhibited these barrier defects. Increased gut permeability and reduced epithelial mitochondrial voltage-dependent anion channel expression were observed 3 days after DSS. These changes and the severity of DSS-colitis were reduced by MitoTEMPO treatment. In vitro DSS-stimulated IL-8 production by epithelia was reduced by MitoTEMPO. Metabolic stress evokes significant penetration of commensal bacteria across the epithelium, which is mediated by mitochondria-derived superoxide acting as a signaling, not a cytotoxic, molecule. MitoTEMPO inhibited this barrier dysfunction and suppressed colitis in DSS-colitis, likely via enhancing barrier function and inhibiting proinflammatory cytokine production. These novel findings support consideration of MTAs in the maintenance of epithelial barrier function and the management of inflammatory bowel diseases.


Asunto(s)
Colitis/patología , Mucosa Intestinal/metabolismo , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Antioxidantes/farmacología , Colitis/fisiopatología , Modelos Animales de Enfermedad , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/fisiopatología , Masculino , Ratones , Ratones Endogámicos BALB C , Mitocondrias/efectos de los fármacos , Permeabilidad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Am J Physiol Gastrointest Liver Physiol ; 303(1): G111-9, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22517768

RESUMEN

Proteinase-activated receptor (PAR)(2), a G protein-coupled receptor activated by serine proteinases, has been implicated in both intestinal inflammation and epithelial proliferation. Cyclooxygenase (COX)-2 is overexpressed in the gut during inflammation as well as in colon cancer. We hypothesized that PAR(2) drives COX-2 expression in intestinal epithelial cells. Treatment of Caco-2 colon cancer cells with the PAR(2)-activating peptide 2-furoyl-LIGRLO-NH(2) (2fLI), but not by its reverse-sequence PAR(2)-inactive peptide, for 3 h led to an increase in intracellular COX-2 protein expression accompanied by a COX-2-dependent increase in prostaglandin E(2) production. 2fLI treatment for 30 min significantly increased metalloproteinase activity in the culture supernatant. Increased epidermal growth factor receptor (EGFR) phosphorylation was observed in cell lysates following 40 min of treatment with 2fLI. The broad-spectrum metalloproteinase inhibitor marimastat inhibited both COX-2 expression and EGFR phosphorylation. The EGFR tyrosine kinase inhibitor PD153035 also abolished 2fLI-induced COX-2 expression. Although PAR(2) activation increased ERK MAPK phosphorylation, neither ERK pathway inhibitors nor a p38 MAPK inhibitor affected 2fLI-induced COX-2 expression. However, inhibition of either Src tyrosine kinase signaling by PP2, Rho kinase signaling by Y27632, or phosphatidylinositol 3 (PI3) kinase signaling by LY294002 prevented 2fLI-induced COX-2 expression. Trypsin increased COX-2 expression through PAR(2) in Caco-2 cells and in an EGFR-dependent manner in the noncancerous intestinal epithelial cell-6 cell line. In conclusion, PAR(2) activation drives COX-2 expression in Caco-2 cells via metalloproteinase-dependent EGFR transactivation and activation of Src, Rho, and PI3 kinase signaling. Our findings provide a mechanism whereby PAR(2) can participate in the progression from chronic inflammation to cancer in the intestine.


Asunto(s)
Ciclooxigenasa 2/biosíntesis , Células Epiteliales/metabolismo , Receptores ErbB/fisiología , Mucosa Intestinal/metabolismo , Receptor PAR-2/fisiología , Anfirregulina , Western Blotting , Células CACO-2 , Línea Celular , Cromatografía Líquida de Alta Presión , Ciclooxigenasa 2/genética , Dinoprostona/metabolismo , Familia de Proteínas EGF , Ensayo de Inmunoadsorción Enzimática , Receptores ErbB/genética , Glicoproteínas/análisis , Glicoproteínas/metabolismo , Humanos , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intercelular/análisis , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Mucosa Intestinal/citología , Metaloproteinasas de la Matriz/biosíntesis , Fosfatidilinositol 3-Quinasa/fisiología , Reacción en Cadena de la Polimerasa , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Transducción de Señal/fisiología , Activación Transcripcional/efectos de los fármacos , Tripsina/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Quinasas Asociadas a rho/fisiología , Familia-src Quinasas/fisiología
8.
Gastroenterology ; 137(2): 569-78, 578.e1, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19375422

RESUMEN

BACKGROUND & AIMS: Hydrogen sulfide (H(2)S) is an endogenous gaseous mediator of mucosal defense with antiinflammatory effects that promote ulcer healing. The effects of H(2)S during the pathogenesis of colitis have not been established. We analyzed the contribution of H(2)S to inflammation and ulceration of the colon in a rat model of colitis. METHODS: Colitis was induced by intracolonic administration of trinitrobenzene sulfonic acid. The ability of the colon to synthesize H(2)S was studied over the course of the resolution of the colitis. Expression of 2 enzymes involved in the synthesis of H(2)S and the effects of inhibitors of these enzymes were examined. We also examined the effects of H(2)S donors on the resolution of colitis. RESULTS: The capacity for the colon to produce H(2)S increased markedly over the first days after induction of colitis and then declined toward control levels as the colitis was resolved. Inhibition of colonic H(2)S synthesis markedly exacerbated the colitis, resulting in significant mortality. Inhibition of H(2)S synthesis in healthy rats resulted in inflammation and mucosal injury in the small intestine and colon along with down-regulation of cyclooxygenase-2 messenger RNA expression and prostaglandin synthesis. Intracolonic administration of H(2)S donors significantly reduced the severity of colitis and reduced colonic expression of messenger RNA for the proinflammatory cytokine tumor necrosis factor alpha. CONCLUSIONS: In rats, H(2)S modulates physiological inflammation and contributes to the resolution of colitis.


Asunto(s)
Colitis/tratamiento farmacológico , Colitis/patología , Sulfuro de Hidrógeno/metabolismo , Sulfuro de Hidrógeno/farmacología , Análisis de Varianza , Animales , Biomarcadores/metabolismo , Biopsia con Aguja , Colitis/enzimología , Colitis/mortalidad , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/metabolismo , Modelos Animales de Enfermedad , Gliburida/farmacología , Inmunohistoquímica , Masculino , Pinacidilo/farmacología , Probabilidad , Distribución Aleatoria , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sensibilidad y Especificidad , Estadísticas no Paramétricas , Tasa de Supervivencia , Resultado del Tratamiento , Ácido Trinitrobencenosulfónico/farmacología , Factor de Necrosis Tumoral alfa/análisis , Factor de Necrosis Tumoral alfa/metabolismo
9.
FASEB J ; 21(14): 4070-6, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17634391

RESUMEN

Hydrogen sulfide is an endogenous mediator that relaxes vascular smooth muscle, exhibits several antiinflammatory activities, and contributes to gastric mucosal defense. This study was performed to examine the role of hydrogen sulfide in the resolution of injury; specifically, the healing of gastric ulcers. Ulcers were induced in rats by serosal application of acetic acid. This elicited a marked increase in gastric expression of the two key enzymes in hydrogen sulfide synthesis (cystathionine-beta-synthase and cystathionine-gamma-lyase) and in hydrogen sulfide synthesis. Twice-daily treatment for a week with hydrogen sulfide donors significantly increased the extent of healing of gastric ulcers as compared to vehicle-treatment. Similar treatment with L-cysteine, a precursor for hydrogen sulfide, also accelerated healing of the ulcers, and the effect was abolished by cotreatment with an inhibitor of cystathionine-gamma-lyase. The beneficial effects of hydrogen sulfide on ulcer healing were not dependent on nitric oxide synthesis, nor did they appear to occur through activation of ATP-sensitive K+ channels. These results suggest that hydrogen sulfide is produced in the gastric mucosa in response to injury and acts to promote healing. The results further suggest that drugs releasing hydrogen sulfide could be employed to accelerate healing of gastric ulcers, and possibly of other wounds.


Asunto(s)
Sulfuro de Hidrógeno/farmacología , Úlcera Gástrica/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Ácido Acético/administración & dosificación , Ácido Acético/toxicidad , Animales , Sulfuro de Hidrógeno/metabolismo , Intubación Gastrointestinal , Masculino , Compuestos Organotiofosforados/farmacología , Ratas , Ratas Wistar , Úlcera Gástrica/inducido químicamente , Úlcera Gástrica/tratamiento farmacológico , Úlcera Gástrica/enzimología , Tioamidas/farmacología , Cicatrización de Heridas/fisiología
10.
Antioxid Redox Signal ; 28(16): 1533-1540, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28388861

RESUMEN

SIGNIFICANCE: Shortly after the discovery of the role of hydrogen sulfide (H2S) in many physiological and pathological processes, attempts were made to develop novel pharmaceuticals that may be of benefit for treatment or prevention of a wide range of disorders. The promise of H2S-based therapeutics is now being demonstrated in clinical trials. Recent Advances: H2S-releasing drugs, such as SG1002 for cardiovascular disorders, and ATB-346 for arthritis, have progressed into clinical trials and have shown considerable promise. Some older drugs, such as zofenopril, have now been recognized to produce at least some of the beneficial effects through release of H2S. CRITICAL ISSUES: There remains a need to better understand the underlying mechanisms for some of the observed effects of H2S-releasing drugs in a clinical setting, such as the marked increase in analgesic potency that has been observed with ATB-346. FUTURE DIRECTIONS: The proof-of-concept clinical studies reviewed herein pave the way for examination, in a clinical setting, of several other potential applications of H2S-based drugs in a wide range of disorders, including diabetes, hypertension, and cancer chemoprevention. Antioxid. Redox Signal. 28, 1533-1540.


Asunto(s)
Artritis/tratamiento farmacológico , Captopril/análogos & derivados , Fármacos Cardiovasculares/uso terapéutico , Enfermedades Cardiovasculares/tratamiento farmacológico , Sulfuro de Hidrógeno/uso terapéutico , Naproxeno/análogos & derivados , Animales , Captopril/uso terapéutico , Relación Dosis-Respuesta a Droga , Humanos , Sulfuro de Hidrógeno/metabolismo , Naproxeno/uso terapéutico
11.
J Mol Med (Berl) ; 96(10): 1081-1093, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30090948

RESUMEN

Aquaporin (AQP) 3 expression is altered in inflammatory bowel diseases, although the exact mechanisms regulating AQP abundance are unclear. Although interferon gamma (IFNγ) is centrally involved in intestinal inflammation, the effect of this cytokine on AQP3 expression remains unknown. HT-29 human colonic epithelial cells were treated with IFNγ to assess AQP3 mRNA expression by real-time RT-PCR and functional protein expression through the uptake of radiolabelled glycerol. Transient knockdown of signal transducer and activator of transcription 1 (STAT1), STAT3, Sp1, and Sp3 were performed to determine the involvement of these transcription factors in the IFNγ-induced signalling cascade. AQP3 promoter regions involved in the response to IFNγ were assessed using a luciferase reporter system. Likewise, enteroids derived from human colonic biopsies were also treated with IFNγ to assess for changes in AQP3 mRNA expression. IFNγ decreased AQP3 mRNA expression in HT-29 cells in a time- and concentration-dependent manner and reduced functional AQP3 protein expression (decreased 3H-labelled glycerol uptake). IFNγ also reduced AQP3 expression in enteroids derived from human colonic biopsies. Knockdown of STAT1 partially prevented the IFNγ-induced downregulation of AQP3 expression, whereas STAT3 and Sp3 knockdowns resulted in increased baseline expression of AQP3 but did not alter IFNγ-induced downregulation. Constitutive transcription of AQP3 is downregulated by IFNγ as demonstrated using the luciferase reporter system, with Sp3 bound to the AQP3 promoter as shown by chromatin immunoprecipitation. AQP3 constitutive transcription in intestinal epithelial cells is downregulated by IFNγ. This response requires STAT1 that is postulated to drive the downregulation of AQP3 expression through increased acetylation or decreased deacetylation the AQP3 promoter, ultimately resulting in decreased constitutive transcription of AQP3. KEY MESSAGES: • IFNγ suppresses the expression of AQP3 in intestinal epithelial cells. • Proximal AQP3 promoter elements are sufficient to drive constitutive expression and mediate the IFNγ-induced downregulation of AQP3 mRNA expression. • IFNγ-induced suppression of AQP3 is dependent upon STAT1 expression, but not STAT3, Sp1, or Sp3.


Asunto(s)
Acuaporina 3/genética , Interferón gamma/farmacología , Mucosa Intestinal/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Células HT29 , Humanos , Mucosa Intestinal/metabolismo , Transcripción Genética/efectos de los fármacos
12.
Physiol Rep ; 5(19)2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29038361

RESUMEN

Inflammatory diseases of the gut are associated with altered electrolyte and water transport, leading to the development of diarrhea. Epithelially expressed aquaporins (AQPs) are downregulated in inflammation, although the mechanisms involved are not known. We hypothesized that AQP3 expression in intestinal epithelial cells is altered in intestinal inflammation and that these changes are driven by tumor necrosis factor (TNF) α Human colonic adenocarcinoma (HT-29) cells were treated with TNFα to investigate signaling mechanisms in vitro. AQP3 expression was assessed by real-time PCR and radiolabeled glycerol uptake, with select inhibitors and a luciferase reporter construct used to further elucidate intracellular signaling. AQP3 expression was downregulated in HT-29 cells treated with TNFα Luciferase reporter construct experiments revealed that TNFα downregulated constitutive transcriptional activity of the AQP3 promoter, and inhibition of MEK/ERK and nuclear factor κB (NF-κB) signaling prevented the decrease in AQP3 mRNA expression. Constitutive AQP3 expression was suppressed by specificity protein (Sp) 3, and knockdown of this transcription factor bound to the AQP3 promoter was able to partially prevent the TNFα-induced downregulation of AQP3. TNFα signals through MEK/ERK and NF-κB to enhance the negative transcriptional control of AQP3 expression exerted by Sp3. Similar mechanisms regulate numerous ion channels, suggesting a common mechanism by which both ion and water transport are altered in inflammation.


Asunto(s)
Acuaporina 3/metabolismo , Enterocitos/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Acuaporina 3/genética , Enterocitos/efectos de los fármacos , Células HT29 , Humanos , Sistema de Señalización de MAP Quinasas , FN-kappa B/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Transcripción Sp3/metabolismo
13.
Br J Pharmacol ; 148(3): 274-8, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16565732

RESUMEN

1. Platelets contain an array of growth factors that can modulate healing processes, including both pro- (e.g., vascular endothelial growth factor (VEGF)) and antiangiogenic (e.g., endostatin) factors. Previous studies have shown that circulating platelets contribute significantly to gastric ulcer healing, acting as a delivery system for these growth factors to the site of injury. In this study, we examined the effects of orally administered human platelets on the healing of gastric ulcers in rats, and determined the contribution of VEGF and endostatin to healing in this model. 2. Twice-daily administration of human platelets significantly accelerated ulcer healing, but platelet-poor plasma (PPP), lysed platelets and serum failed to produce this effect. There was no correlation between ulcer healing and the levels of VEGF or endostatin in serum, PPP or platelet-rich plasma (PRP). 3. Accelerated ulcer healing could not be produced by oral administration of the angiogenic factors themselves, at concentrations matching those in PRP. 4. The accelerated healing induced by platelets could be reversed by immuno-neutralization of VEGF. In contrast, immuno-neutralization of endostatin did not affect PRP-induced ulcer healing. 5. These studies indicate that VEGF released from platelets accounts for the accelerated healing of gastric ulcers. However, as intact (rather than lysed) platelets were required for the accelerated healing, the presentation of VEGF by the platelet at the site of injury appears to be crucial for enhancement of the healing process.


Asunto(s)
Plaquetas/fisiología , Úlcera Gástrica/terapia , Factor A de Crecimiento Endotelial Vascular/administración & dosificación , Animales , Plaquetas/química , Sistemas de Liberación de Medicamentos , Endostatinas/administración & dosificación , Endostatinas/farmacología , Humanos , Masculino , Transfusión de Plaquetas , Ratas , Ratas Wistar , Factor A de Crecimiento Endotelial Vascular/farmacología
14.
PLoS One ; 10(3): e0118713, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25793528

RESUMEN

Inflammatory bowel diseases are associated with dysregulated electrolyte and water transport and resultant diarrhea. Aquaporins are transmembrane proteins that function as water channels in intestinal epithelial cells. We investigated the effect of the inflammatory cytokine, interferon-γ, which is a major player in inflammatory bowel diseases, on aquaporin-1 expression in a mouse colonic epithelial cell line, CMT93. CMT93 monolayers were exposed to 10 ng/mL interferon-γ and aquaporin-1 mRNA and protein expressions were measured by real-time PCR and western blot, respectively. In other experiments, CMT93 cells were pretreated with inhibitors or were transfected with siRNA to block the effects of Janus kinases, STATs 1 and 3, or interferon regulatory factor 2, prior to treatment with interferon-γ. Interferon-γ decreased aquaporin-1 expression in mouse intestinal epithelial cells in a manner that did not depend on the classical STAT1/JAK2/IRF-1 pathway, but rather, on an alternate Janus kinase (likely JAK1) as well as on STAT3. The pro-inflammatory cytokine, interferon-γ may contribute to diarrhea associated with intestinal inflammation in part through regulation of the epithelial aquaporin-1 water channel via a non-classical JAK/STAT receptor signalling pathway.


Asunto(s)
Acuaporina 1/genética , Células Epiteliales/metabolismo , Interferón gamma/farmacología , Intestinos/citología , Quinasas Janus/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Acuaporina 1/metabolismo , Línea Celular , Células Epiteliales/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Factor 1 Regulador del Interferón/metabolismo , Factor 2 Regulador del Interferón/metabolismo , Ratones , Modelos Biológicos , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Transcripción STAT1/metabolismo , Factores de Tiempo
15.
Inflamm Bowel Dis ; 18(6): 1137-45, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21953882

RESUMEN

BACKGROUND: Salvinorin A (SA) has a potent inhibitory action on mouse gastrointestinal (GI) motility and ion transport, mediated primarily by kappa-opioid receptors (KOR). The aim of the present study was to characterize possible antiinflammatory and antinociceptive effects of SA in the GI tract of mice. METHODS: Colonic damage scores and myeloperoxidase activity were determined after intraperitoneal (i.p.), intracolonic (i.c.), and oral (p.o.) administration of SA using the trinitrobenzene sulfonic acid (TNBS) and dextran sodium sulfate (DSS) models of colitis in mice. Additionally, KOR, cannabinoid (CB)1, and CB2 western blot analysis of colon samples was performed. The antinociceptive effect of SA was examined based on the number of behavioral responses to i.c. instillation of mustard oil (MO). RESULTS: The i.p. (3 mg/kg, twice daily) and p.o. (10 mg/kg, twice daily) administration of SA significantly attenuated TNBS and DSS colitis in mice. The effect of SA was blocked by KOR antagonist nor-binaltorphimine (10 mg/kg, i.p.). Western blot analysis showed no influence of SA on KOR, CB1, or CB2 levels. SA (3 mg/kg, i.p. and 10 mg/kg, i.c.) significantly decreased the number of pain responses after i.c. instillation of MO in the vehicle- and TNBS-treated mice. The antinociceptive action of SA was blocked by KOR and CB1 antagonists. The analgesic effect of i.c. SA was more potent in TNBS-treated mice compared to controls. CONCLUSIONS: Our results suggest that the drugs based on the structure of SA have the potential to become valuable antiinflammatory or analgesic therapeutics for the treatment of GI diseases.


Asunto(s)
Analgésicos/uso terapéutico , Antiinflamatorios/uso terapéutico , Colitis/tratamiento farmacológico , Diterpenos de Tipo Clerodano/uso terapéutico , Motilidad Gastrointestinal/efectos de los fármacos , Dolor/tratamiento farmacológico , Receptor Cannabinoide CB1/metabolismo , Receptor Cannabinoide CB2/metabolismo , Receptores Opioides kappa/metabolismo , Animales , Western Blotting , Colitis/inducido químicamente , Colitis/metabolismo , Colon/efectos de los fármacos , Colon/metabolismo , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Masculino , Ratones , Naltrexona/análogos & derivados , Dolor/metabolismo , Peroxidasa , Salvia/química , Ácido Trinitrobencenosulfónico/toxicidad
16.
Dig Liver Dis ; 42(2): 103-9, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19570733

RESUMEN

AIMS: Hydrogen sulphide (H2S) exerts several anti-inflammatory effects, accelerates the healing of experimental gastric ulcers, and can stimulate intestinal secretion. Little is known about H2S synthesis in the gastrointestinal tract. The aim of this study was to characterize H2S synthesis throughout the gastrointestinal tract. METHODS: H2S synthesis in various gastrointestinal tissues of rats and mice was determined. The effects and selectivity of inhibitors of two key enzymes for H2S synthesis, cystathionine-gamma-lyase and cystathionine-beta-synthase, were examined. Cystathionine-gamma-lyase and cystathionine-beta-synthase expression was evaluated by Western blotting and immunohistochemistry. Cystathionine-gamma-lyase and cystathionine-beta-synthase expression in biopsies of human colon was also examined. RESULTS: H2S synthesis was variable throughout the gastrointestinal tract in parallel with variations in cystathionine-gamma-lyase and cystathionine-beta-synthase expression. The efficacy of cystathionine-beta-synthase and cystathionine-gamma-lyase inhibitors to reduce H2S synthesis in these tissues was also variable. Cystathionine-beta-synthase is the predominant source of H2S synthesis in the colon of rodents. Cystathionine-gamma-lyase and cystathionine-beta-synthase were also expressed in healthy human colon biopsies. CONCLUSIONS: The capacity for H2S synthesis varies throughout the rodent gastrointestinal tract, as does the distribution and contribution of the two key enzymes. Investigation of additional enzymatic sources of H2S and the development of more selective inhibitors are suggested.


Asunto(s)
Cistationina betasintasa/metabolismo , Cistationina gamma-Liasa/metabolismo , Tracto Gastrointestinal/metabolismo , Sulfuro de Hidrógeno/metabolismo , Animales , Colon/metabolismo , Tracto Gastrointestinal/enzimología , Humanos , Inmunohistoquímica , Técnicas In Vitro , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos , Ratas , Ratas Wistar
17.
Proc Natl Acad Sci U S A ; 102(1): 216-20, 2005 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-15615851

RESUMEN

The roles of proteinase-activated receptors (PARs) in platelet functions other than aggregation are not well understood. Among these is the release of factors that regulate the process of angiogenesis, such as endostatin and VEGF, which, respectively, inhibit and promote angiogenesis. PAR1 and PAR4 are expressed on the surface of human platelets and can be activated by thrombin. In the present study, we have attempted to determine the roles of PAR1 and PAR4 in regulating release of endostatin and VEGF from human platelets. Aggregation and endostatin release could be elicited by a specific PAR4 agonist (AYPGKF-NH(2)). The PAR4 agonist concentration dependently suppressed VEGF release. A selective PAR1 agonist (TFLLR-NH(2)) induced platelet aggregation and VEGF release but suppressed endostatin release. Thrombin did not affect endostatin or VEGF release. However, in the presence of a selective PAR1 antagonist (SCH79797), thrombin stimulated endostatin release and suppressed VEGF release. Conversely, in the presence of a selective PAR4 antagonist (transcinnamoyl-YPGKF-NH(2)), thrombin stimulated VEGF release. In vivo, treatment of rats with established gastric ulcers with a PAR1 antagonist each day for 1 wk resulted in a significant retardation of healing. We conclude that PAR1 and PAR4 counter-regulate the release of endostatin and VEGF from platelets. These protease-activated receptors could therefore play a crucial role in regulating angiogenesis and in turn could regulate the processes of wound healing and tumor growth.


Asunto(s)
Plaquetas/metabolismo , Endostatinas/metabolismo , Receptor PAR-1/metabolismo , Receptores de Trombina/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Humanos , Receptores de Trombina/agonistas
18.
Am J Physiol Gastrointest Liver Physiol ; 286(1): G76-81, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14665439

RESUMEN

Aceylation of cyclooxygenase (COX)-2 by aspirin can trigger the formation of 15(R)-epilipoxin A4, or aspirin-triggered lipoxin (ATL). ATL exerts protective effects in the stomach. Selective COX-2 inhibitors block ATL synthesis and exacerbate aspirin-induced gastric damage. Nitric oxide-releasing aspirins, including NCX-4016, have antiplatelet effects similar to aspirin but do not cause gastric damage. In the present study, we examined whether or not NCX-4016 triggers ATL synthesis and/or upregulates gastric COX-2 expression and the effects of coadministration of NCX-4016 with a selective COX-2 inhibitor on gastric mucosal injury and inflammation. Rats were given aspirin or NCX-4016 orally and either vehicle or a selective COX-2 inhibitor (celecoxib) intraperitoneally. Gastric damage was blindly scored, and granulocyte infiltration into gastric tissue was monitored through measurement of myeloperoxidase activity. Gastric PG and ATL synthesis was measured as was COX-2 expression. Whereas celecoxib inhibited gastric ATL synthesis and increased the severity of aspirin-induced gastric damage and inflammation, coadministration of celecoxib and NCX-4016 did not result in damage or inflammation. NCX-4016 did not upregulate gastric COX-2 expression nor did it trigger ATL synthesis (in contrast to aspirin). Daily administration of aspirin for 5 days resulted in significantly less gastric damage than that seen with a single dose, as well as augmented ATL synthesis. Celecoxib reversed this effect. In contrast, repeated administration of NCX-4016 failed to cause gastric damage, whether given alone or with celecoxib. These studies support the notion that NCX-4016 may be an attractive alternative to aspirin for indications such as cardioprotection, including in individuals also taking selective COX-2 inhibitors.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Aspirina/farmacología , Inhibidores de la Ciclooxigenasa/farmacología , Mucosa Gástrica/patología , Gastritis/patología , Isoenzimas/metabolismo , Óxido Nítrico/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Animales , Aspirina/análogos & derivados , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Dinoprostona/metabolismo , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/enzimología , Isoenzimas/biosíntesis , Lipoxinas/biosíntesis , Masculino , Neutropenia/metabolismo , Infiltración Neutrófila/efectos de los fármacos , Peroxidasa/metabolismo , Prostaglandina-Endoperóxido Sintasas/biosíntesis , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Gastroenterology ; 123(5): 1598-606, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12404234

RESUMEN

BACKGROUND & AIMS: Cyclooxygenase-2 (COX-2) has been implicated as contributing to mucosal defense. Acetylation of COX-2 by aspirin can result in production of an antiinflammatory substance, 15(R)-epi-LXA4. We determined whether aspirin-triggered lipoxin (LX) production via COX-2 diminishes aspirin-induced damage in the rat stomach. METHODS: Rats were treated with aspirin plus or minus celecoxib or rofecoxib. Gastric generation of LXA4 was measured. Effect of exogenous LXA4 or an LXA4 receptor antagonist on gastric resistance to aspirin-induced damage was examined. Aspirin-induced leukocyte adherence in mesenteric venules, and the effects of LXA4, were examined by intravital microscopy. RESULTS: Celecoxib and rofecoxib significantly increased the severity of aspirin-induced gastric damage. Aspirin rapidly up-regulated COX-2 expression in the stomach and caused a significant increase in gastric 15(R)-epi-LXA4 production, which was abolished by celecoxib. LXA4 dose dependently (0.25-2.5 microg/kg, intraperitoneally) reduced the severity of aspirin-induced gastric damage and suppressed aspirin-induced leukocyte adherence, whereas an LXA4 antagonist had the opposite effects. CONCLUSIONS: Aspirin administration results in elevated production of 15(R)-epi-LXA4 via COX-2. LXA4 exerts very potent protective actions on the gastric mucosa. Co-administration of aspirin and a selective COX-2 inhibitor results in substantially more severe gastric injury than is produced with either agent alone.


Asunto(s)
Aspirina/efectos adversos , Inhibidores de la Ciclooxigenasa/efectos adversos , Ácidos Hidroxieicosatetraenoicos/fisiología , Isoenzimas/metabolismo , Lipoxinas , Prostaglandina-Endoperóxido Sintasas/metabolismo , Estómago/efectos de los fármacos , Animales , Aspirina/farmacología , Celecoxib , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Ciclooxigenasa 1 , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa/farmacología , Resistencia a Medicamentos/fisiología , Mucosa Gástrica/metabolismo , Ácidos Hidroxieicosatetraenoicos/biosíntesis , Ácidos Hidroxieicosatetraenoicos/farmacología , Isoenzimas/antagonistas & inhibidores , Lactonas/farmacología , Leucocitos/efectos de los fármacos , Leucocitos/fisiología , Masculino , Proteínas de la Membrana , Pirazoles , Ratas , Ratas Wistar , Estómago/patología , Sulfonamidas/farmacología , Sulfonas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA