Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Am J Physiol Cell Physiol ; 325(1): C344-C361, 2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-37125773

RESUMO

Kidney stones (KSs) are very common, excruciating, and associated with tremendous healthcare cost, chronic kidney disease (CKD), and kidney failure (KF). Most KSs are composed of calcium oxalate and small increases in urinary oxalate concentration significantly enhance the stone risk. Oxalate also potentially contributes to CKD progression, kidney disease-associated cardiovascular diseases, and poor renal allograft survival. This emphasizes the urgent need for plasma and urinary oxalate lowering therapies, which can be achieved by enhancing enteric oxalate secretion. We previously identified Oxalobacter formigenes (O. formigenes)-derived factors secreted in its culture-conditioned medium (CM), which stimulate oxalate transport by human intestinal Caco2-BBE (C2) cells and reduce urinary oxalate excretion in hyperoxaluric mice by enhancing colonic oxalate secretion. Given their remarkable therapeutic potential, we now identified Sel1-like proteins as the major O. formigenes-derived secreted factors using mass spectrometry and functional assays. Crystal structures for six proteins were determined to confirm structures and better understand functions. OxBSel1-14-derived small peptides P8 and P9 were identified as the major factors, with P8 + 9 closely recapitulating the CM's effects, acting through the oxalate transporters SLC26A2 and SLC26A6 and PKA activation. Besides C2 cells, P8 + 9 also stimulate oxalate transport by human ileal and colonic organoids, confirming that they work in human tissues. In conclusion, P8 and P9 peptides are identified as the major O. formigenes-derived secreted factors and they have significant therapeutic potential for hyperoxalemia, hyperoxaluria, and related disorders, impacting the outcomes of patients suffering from KSs, enteric hyperoxaluria, primary hyperoxaluria, CKD, KF, and renal transplant recipients.NEW & NOTEWORTHY We previously identified Oxalobacter formigenes-derived secreted factors stimulating oxalate transport by human intestinal epithelial cells in vitro and reducing urinary oxalate excretion in hyperoxaluric mice by enhancing colonic oxalate secretion. We now identified Sel1-like proteins and small peptides as the major secreted factors and they have significant therapeutic potential for hyperoxalemia and hyperoxaluria, impacting the outcomes of patients suffering from kidney stones, primary and secondary hyperoxaluria, chronic kidney disease, kidney failure, and renal transplant recipients.


Assuntos
Hiperoxalúria , Cálculos Renais , Transplante de Rim , Insuficiência Renal Crônica , Insuficiência Renal , Humanos , Camundongos , Animais , Oxalobacter formigenes/metabolismo , Células CACO-2 , Oxalatos/metabolismo , Hiperoxalúria/metabolismo , Cálculos Renais/metabolismo , Células Epiteliais/metabolismo , Peptídeos/metabolismo , Insuficiência Renal/metabolismo , Insuficiência Renal Crônica/metabolismo
2.
Am J Physiol Cell Physiol ; 315(5): C687-C698, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30020825

RESUMO

Most kidney stones (KS) are composed of calcium oxalate, and small increases in urine oxalate affect the stone risk. Intestinal oxalate secretion mediated by anion exchanger SLC26A6 (PAT1) plays a crucial role in limiting net absorption of ingested oxalate, thereby preventing hyperoxaluria and related KS, reflecting the importance of understanding regulation of intestinal oxalate transport. We previously showed that ATP and UTP inhibit oxalate transport by human intestinal Caco2-BBE cells (C2). Since ATP is rapidly degraded to adenosine (ADO), we examined whether intestinal oxalate transport is regulated by ADO. We measured [14C]oxalate uptake in the presence of an outward Cl gradient as an assay of Cl-oxalate exchange activity, ≥49% of which is PAT1-mediated in C2 cells. We found that ADO significantly inhibited oxalate transport by C2 cells, an effect completely blocked by the nonselective ADO receptor antagonist 8- p-sulfophenyltheophylline. ADO also significantly inhibited oxalate efflux by C2 cells, which is important since PAT1 mediates oxalate efflux in vivo. Using pharmacological antagonists and A2B adenosine receptor (A2B AR) siRNA knockdown studies, we observed that ADO inhibits oxalate transport through the A2B AR, phospholipase C, and PKC. ADO inhibits oxalate transport by reducing PAT1 surface expression as shown by biotinylation studies. We conclude that ADO inhibits oxalate transport by lowering PAT1 surface expression in C2 cells through signaling pathways including the A2B AR, PKC, and phospholipase C. Given higher ADO levels and overexpression of the A2B AR in inflammatory bowel disease (IBD), our findings have potential relevance to pathophysiology of IBD-associated hyperoxaluria and related KS.


Assuntos
Adenosina/metabolismo , Sistemas de Transporte de Aminoácidos/genética , Doenças Inflamatórias Intestinais/genética , Receptor A2B de Adenosina/genética , Simportadores/genética , Adenosina/administração & dosagem , Antagonistas do Receptor A2 de Adenosina/administração & dosagem , Trifosfato de Adenosina/metabolismo , Transporte Biológico/genética , Células CACO-2 , Humanos , Hiperoxalúria/genética , Hiperoxalúria/metabolismo , Hiperoxalúria/patologia , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/patologia , Mucosa Intestinal/metabolismo , Intestinos/efeitos dos fármacos , Cálculos Renais/genética , Cálculos Renais/metabolismo , Cálculos Renais/patologia , Oxalatos/metabolismo , Receptor A2B de Adenosina/metabolismo , Fatores de Risco , Transdução de Sinais/efeitos dos fármacos , Teofilina/administração & dosagem , Fosfolipases Tipo C/genética
3.
Nature ; 557(7706): 580-584, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29769727

RESUMO

Somatic mutations in tet methylcytosine dioxygenase 2 (TET2), which encodes an epigenetic modifier enzyme, drive the development of haematopoietic malignancies1-7. In both humans and mice, TET2 deficiency leads to increased self-renewal of haematopoietic stem cells with a net developmental bias towards the myeloid lineage1,4,8,9. However, pre-leukaemic myeloproliferation (PMP) occurs in only a fraction of Tet2-/- mice8,9 and humans with TET2 mutations1,3,5-7, suggesting that extrinsic non-cell-autonomous factors are required for disease onset. Here we show that bacterial translocation and increased interleukin-6 production, resulting from dysfunction of the small-intestinal barrier, are critical for the development of PMP in mice that lack Tet2 expression in haematopoietic cells. Furthermore, in symptom-free Tet2-/- mice, PMP can be induced by disrupting intestinal barrier integrity, or in response to systemic bacterial stimuli such as the toll-like receptor 2 agonist. PMP was reversed by antibiotic treatment and failed to develop in germ-free Tet2-/- mice, which illustrates the importance of microbial signals in the development of this condition. Our findings demonstrate the requirement for microbial-dependent inflammation in the development of PMP and provide a mechanistic basis for the variation in PMP penetrance observed in Tet2-/- mice. This study will prompt new lines of investigation that may profoundly affect the prevention and management of haematopoietic malignancies.


Assuntos
Doenças Assintomáticas , Fenômenos Fisiológicos Bacterianos , Proliferação de Células , Proteínas de Ligação a DNA/deficiência , Leucemia/microbiologia , Leucemia/patologia , Proteínas Proto-Oncogênicas/deficiência , Animais , Infecções Bacterianas/imunologia , Infecções Bacterianas/microbiologia , Fenômenos Fisiológicos Bacterianos/imunologia , Proteínas de Ligação a DNA/genética , Dioxigenases , Feminino , Vida Livre de Germes , Inflamação/microbiologia , Interleucina-6/imunologia , Mucosa Intestinal/metabolismo , Lactobacillus/química , Lactobacillus/citologia , Lactobacillus/imunologia , Masculino , Camundongos , Penetrância , Permeabilidade , Proteínas Proto-Oncogênicas/genética , Receptor 2 Toll-Like/agonistas
4.
Pediatr Surg Int ; 34(3): 323-330, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29196880

RESUMO

BACKGROUND: Necrotizing enterocolitis (NEC) is a gastrointestinal disease of complex etiology resulting in devastating systemic inflammation and often death in premature newborns. We previously demonstrated that formula feeding inhibits ileal expression of heat shock protein-70 (Hsp70), a critical stress protein within the intestine. Barrier function for the premature intestine is critical. We sought to determine whether reduced Hsp70 protein expression increases neonatal intestinal permeability. METHODS: Young adult mouse colon cells (YAMC) were utilized to evaluate barrier function as well as intestine from Hsp70-/- pups (KO). Sections of intestine were analyzed by Western blot, immunohistochemistry, and real time PCR. YAMC cells were sub-lethally heated or treated with expressed milk (EM) to induce Hsp70. RESULTS: Immunostaining demonstrates co-localized Hsp70 and tight junction protein zona occludens-1 (ZO-1), suggesting physical interaction to protect tight junction function. The permeability of YAMC monolayers increases following oxidant injury and is partially blocked by Hsp70 induction either by prior heat stress or EM. RT-PCR analysis demonstrated that the Hsp70 isoforms, 70.1 and 70.3, predominate in WT pup; however, Hsp70.2 predominates in the KO pups. While Hsp70 is present in WT milk, it is not present in KO EM. Hsp70 associates with ZO-1 to maintain epithelial barrier function. CONCLUSION: Both induction of Hsp70 and exposure to EM prevent stress-induced increased permeability. Hsp70.2 is present in both WT and KO neonatal intestine, suggesting a crucial role in epithelial integrity. Induction of the Hsp70.2 isoform appears to be mediated by mother's milk. These results suggest that mother's milk feeding modulates Hsp70.2 expression and could attenuate injury leading to NEC. LEVEL OF EVIDENCE: Level III.


Assuntos
Proteínas de Choque Térmico HSP70/metabolismo , Mucosa Intestinal/metabolismo , Leite/metabolismo , Animais , Animais Recém-Nascidos , Citoproteção , Proteínas de Choque Térmico HSP70/genética , Camundongos , Permeabilidade , Isoformas de Proteínas , Junções Íntimas/metabolismo , Proteína da Zônula de Oclusão-1/metabolismo
5.
Sci Rep ; 7(1): 10411, 2017 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-28874832

RESUMO

Recent evidence suggests the commensal microbiome regulates host immunity and influences brain function; findings that have ramifications for neurodegenerative diseases. In the context of Alzheimer's disease (AD), we previously reported that perturbations in microbial diversity induced by life-long combinatorial antibiotic (ABX) selection pressure in the APPSWE/PS1ΔE9 mouse model of amyloidosis is commensurate with reductions in amyloid-ß (Aß) plaque pathology and plaque-localised gliosis. Considering microbiota-host interactions, specifically during early post-natal development, are critical for immune- and neuro-development we now examine the impact of microbial community perturbations induced by acute ABX exposure exclusively during this period in APPSWE/PS1ΔE9 mice. We show that early post-natal (P) ABX treatment (P14-P21) results in long-term alterations of gut microbial genera (predominantly Lachnospiraceae and S24-7) and reduction in brain Aß deposition in aged APPSWE/PS1ΔE9 mice. These mice exhibit elevated levels of blood- and brain-resident Foxp3+ T-regulatory cells and display an alteration in the inflammatory milieu of the serum and cerebrospinal fluid. Finally, we confirm that plaque-localised microglia and astrocytes are reduced in ABX-exposed mice. These findings suggest that ABX-induced microbial diversity perturbations during post-natal stages of development coincide with altered host immunity mechanisms and amyloidosis in a murine model of AD.


Assuntos
Doença de Alzheimer/etiologia , Precursor de Proteína beta-Amiloide/genética , Amiloidose/genética , Antibacterianos/farmacologia , Microbiota/efeitos dos fármacos , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Amiloidose/metabolismo , Amiloidose/patologia , Animais , Biodiversidade , Biomarcadores , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Microbioma Gastrointestinal , Mediadores da Inflamação/metabolismo , Masculino , Metagenoma , Metagenômica/métodos , Camundongos , Camundongos Transgênicos , Neuroimunomodulação/efeitos dos fármacos , Neuroimunomodulação/genética , Neuroimunomodulação/imunologia , Placa Amiloide/etiologia , Placa Amiloide/metabolismo , Placa Amiloide/patologia , RNA Ribossômico 16S/genética
6.
Phytother Res ; 31(1): 90-99, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27730672

RESUMO

Chemopreventative properties of traditional medicines and underlying mechanisms of action are incompletely investigated. This study demonstrates that dietary daikenchuto (TU-100), comprised of ginger, ginseng, and Japanese pepper effectively suppresses intestinal tumor development and progression in the azoxymethane (AOM) and APCmin/+ mouse models. For the AOM model, TU-100 was provided after the first of six biweekly AOM injections. Mice were sacrificed at 30 weeks. APCmin/+ mice were fed diet without or with TU-100 starting at 6 weeks, and sacrificed at 24 weeks. In both models, dietary TU-100 decreased tumor size. In APC min/+ mice, the number of small intestinal tumors was significantly decreased. In the AOM model, both TU-100 and Japanese ginseng decreased colon tumor numbers. Decreased Ki-67 and ß-catenin immunostaining and activation of numerous transduction pathways involved in tumor initiation and progression were observed. EGF receptor expression and stimulation/phosphorylation in vitro were investigated in C2BBe1 cells. TU-100, ginger, and 6-gingerol suppressed EGF receptor induced Akt activation. TU-100 and ginseng and to a lesser extent ginger or 6-gingerol inhibited EGF ERK1/2 activation. TU-100 and some of its components and metabolites of these components inhibit tumor progression in two mouse models of colon cancer by blocking downstream pathways of EGF receptor activation. Copyright © 2016 John Wiley & Sons, Ltd.


Assuntos
Azoximetano/química , Neoplasias do Colo/tratamento farmacológico , Extratos Vegetais/uso terapêutico , Animais , Azoximetano/farmacologia , Neoplasias do Colo/patologia , Modelos Animais de Doenças , Masculino , Medicina Tradicional , Camundongos , Panax , Extratos Vegetais/administração & dosagem , Zanthoxylum , Zingiberaceae
7.
J Am Soc Nephrol ; 28(3): 876-887, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27738124

RESUMO

Hyperoxaluria is a major risk factor for kidney stones and has no specific therapy, although Oxalobacter formigenes colonization is associated with reduced stone risk. O. formigenes interacts with colonic epithelium and induces colonic oxalate secretion, thereby reducing urinary oxalate excretion, via an unknown secretagogue. The difficulties in sustaining O. formigenes colonization underscore the need to identify the derived factors inducing colonic oxalate secretion. We therefore evaluated the effects of O. formigenes culture conditioned medium (CM) on apical 14C-oxalate uptake by human intestinal Caco-2-BBE cells. Compared with control medium, O. formigenes CM significantly stimulated oxalate uptake (>2.4-fold), whereas CM from Lactobacillus acidophilus did not. Treating the O. formigenes CM with heat or pepsin completely abolished this bioactivity, and selective ultrafiltration of the CM revealed that the O. formigenes-derived factors have molecular masses of 10-30 kDa. Treatment with the protein kinase A inhibitor H89 or the anion exchange inhibitor 4,4'-diisothiocyano-2,2'-stilbenedisulfonic acid completely blocked the CM-induced oxalate transport. Knockdown of the oxalate transporter SLC26A6 also significantly restricted the induction of oxalate transport by CM. In a mouse model of primary hyperoxaluria type 1, rectal administration of O. formigenes CM significantly reduced (>32.5%) urinary oxalate excretion and stimulated (>42%) distal colonic oxalate secretion. We conclude that O. formigenes-derived bioactive factors stimulate oxalate transport in intestinal cells through mechanisms including PKA activation. The reduction in urinary oxalate excretion in hyperoxaluric mice treated with O. formigenes CM reflects the in vivo retention of biologic activity and the therapeutic potential of these factors.


Assuntos
Fatores Biológicos/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Oxalatos/metabolismo , Oxalobacter formigenes , Animais , Humanos , Hiperoxalúria/metabolismo , Transporte de Íons , Masculino , Camundongos
8.
Biomed Chromatogr ; 31(4)2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27606833

RESUMO

After ingestion of ginseng, the bioavailability of its parent compounds is low and enteric microbiota plays an important role in parent compound biotransformation to their metabolites. Diet type can influence the enteric microbiota profile. When human subjects on different diets ingest ginseng, their different gut microbiota profiles may influence the metabolism of ginseng parent compounds. In this study, the effects of different diet type on gut microbiota metabolism of American ginseng saponins were investigated. We recruited six healthy adults who regularly consumed different diet types. These subjects received 7 days' oral American ginseng, and their biological samples were collected for LC-Q-TOF-MS analysis. We observed significant ginsenoside Rb1 (a major parent compound) and compound K (a major active metabolite) level differences in the samples from the subjects consuming different diets. Subjects on an Asian diet had much higher Rb1 levels but much lower compound K levels compared with those on a Western diet. Since compound K possesses much better cancer chemoprevention potential, our data suggested that consumers on a Western diet should obtain better cancer prevention effects with American ginseng intake compared with those on an Asian diet. Ginseng compound levels could be enhanced or reduced via gut microbiota manipulation for clinical utility.


Assuntos
Dieta , Microbioma Gastrointestinal , Panax/metabolismo , Saponinas/farmacocinética , Adulto , Cromatografia Líquida/métodos , Dieta Ocidental , Fezes/química , Microbioma Gastrointestinal/efeitos dos fármacos , Ginsenosídeos/análise , Ginsenosídeos/metabolismo , Humanos , Inativação Metabólica , Masculino , Pessoa de Meia-Idade , Saponinas/análise , Saponinas/metabolismo
9.
Sci Rep ; 6: 32094, 2016 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-27561676

RESUMO

Proliferation and spatial development of colonic epithelial cells are highly regulated along the crypt vertical axis, which, when perturbed, can result in aberrant growth and carcinogenesis. In this study, two key factors were identified that have important and counterbalancing roles regulating these processes: pericrypt myofibroblast-derived Wnt-5a and the microbial metabolite butyrate. Cultured YAMC cell proliferation and heat shock protein induction were analzyed after butryate, conditioned medium with Wnt5a activity, and FrzB containing conditioned medium. In vivo studies to modulate Hsp25 employed intra-colonic wall Hsp25 encoding lentivirus. To silence Wnt-5a in vivo, intra-colonic wall Wnt-5a silencing RNA was used. Wnt-5a, secreted by stromal myofibroblasts of the lower crypt, promotes proliferation through canonical ß-catenin activation. Essential to this are two key requirements: (1) proteolytic conversion of the highly insoluble ~40 kD Wnt-5a protein to a soluble 36 mer amino acid peptide that activates epithelial ß-catenin and cellular proliferation, and (2) the simultaneous inhibition of butyrate-induced Hsp25 by Wnt-5a which is necessary to arrest the proliferative process in the upper colonic crypt. The interplay and spatial gradients of these factors insures that crypt epithelial cell proliferation and development proceed in an orderly fashion, but with sufficient plasticity to adapt to physiological perturbations including inflammation.


Assuntos
Butiratos/farmacologia , Proliferação de Células/efeitos dos fármacos , Colo/metabolismo , Células Epiteliais/metabolismo , Mucosa Intestinal/metabolismo , Proteólise/efeitos dos fármacos , Proteína Wnt-5a/metabolismo , Animais , Linhagem Celular , Colo/citologia , Células Epiteliais/citologia , Proteínas de Choque Térmico/metabolismo , Mucosa Intestinal/citologia , Camundongos , Chaperonas Moleculares , Proteínas de Neoplasias/metabolismo
10.
Am J Physiol Gastrointest Liver Physiol ; 311(4): G634-G647, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27514476

RESUMO

Bifidobacterium breve and other Gram-positive gut commensal microbes protect the gastrointestinal epithelium against inflammation-induced stress. However, the mechanisms whereby these bacteria accomplish this protection are poorly understood. In this study, we examined soluble factors derived from Bifidobacterium breve and their impact on the two major protein degradation systems within intestinal epithelial cells, proteasomes and autophagy. Conditioned media from gastrointestinal Gram-positive, but not Gram-negative, bacteria activated autophagy and increased expression of the autophagy proteins Atg5 and Atg7 along with the stress response protein heat shock protein 27. Specific examination of media conditioned by the Gram-positive bacterium Bifidobacterium breve (Bb-CM) showed that this microbe produces small molecules (<3 kDa) that increase expression of the autophagy proteins Atg5 and Atg7, activate autophagy, and inhibit proteasomal enzyme activity. Upregulation of autophagy by Bb-CM was mediated through MAP kinase signaling. In vitro studies using C2BBe1 cells silenced for Atg7 and in vivo studies using mice conditionally deficient in intestinal epithelial cell Atg7 showed that Bb-CM-induced cytoprotection is dependent on autophagy. Therefore, this work demonstrates that Gram-positive bacteria modify protein degradation programs within intestinal epithelial cells to promote their survival during stress. It also reveals the therapeutic potential of soluble molecules produced by these microbes for prevention and treatment of gastrointestinal disease.


Assuntos
Autofagia/fisiologia , Mucosa Intestinal/microbiologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Estresse Fisiológico/fisiologia , Animais , Proteína 5 Relacionada à Autofagia/genética , Proteína 5 Relacionada à Autofagia/metabolismo , Proteína 7 Relacionada à Autofagia/genética , Proteína 7 Relacionada à Autofagia/metabolismo , Bifidobacterium breve , Linhagem Celular , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Inflamação/metabolismo , Inflamação/microbiologia , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Knockout , Transdução de Sinais/fisiologia
11.
Sci Rep ; 6: 30028, 2016 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-27443609

RESUMO

Severe amyloidosis and plaque-localized neuro-inflammation are key pathological features of Alzheimer's disease (AD). In addition to astrocyte and microglial reactivity, emerging evidence suggests a role of gut microbiota in regulating innate immunity and influencing brain function. Here, we examine the role of the host microbiome in regulating amyloidosis in the APPSWE/PS1ΔE9 mouse model of AD. We show that prolonged shifts in gut microbial composition and diversity induced by long-term broad-spectrum combinatorial antibiotic treatment regime decreases Aß plaque deposition. We also show that levels of soluble Aß are elevated and that levels of circulating cytokine and chemokine signatures are altered in this setting. Finally, we observe attenuated plaque-localised glial reactivity in these mice and significantly altered microglial morphology. These findings suggest the gut microbiota community diversity can regulate host innate immunity mechanisms that impact Aß amyloidosis.


Assuntos
Doença de Alzheimer/fisiopatologia , Amiloidose/fisiopatologia , Microbioma Gastrointestinal/imunologia , Imunidade Inata , Inflamação/fisiopatologia , Animais , Antibacterianos/administração & dosagem , Modelos Animais de Doenças , Disbiose/induzido quimicamente , Camundongos
12.
Am J Physiol Gastrointest Liver Physiol ; 310(11): G973-88, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27079612

RESUMO

Gut dysbiosis, host genetics, and environmental triggers are implicated as causative factors in inflammatory bowel disease (IBD), yet mechanistic insights are lacking. Longitudinal analysis of ulcerative colitis (UC) patients following total colectomy with ileal anal anastomosis (IPAA) where >50% develop pouchitis offers a unique setting to examine cause vs. effect. To recapitulate human IPAA, we employed a mouse model of surgically created blind self-filling (SFL) and self-emptying (SEL) ileal loops using wild-type (WT), IL-10 knockout (KO) (IL-10), TLR4 KO (T4), and IL-10/T4 double KO mice. After 5 wk, loop histology, host gene/protein expression, and bacterial 16s rRNA profiles were examined. SFL exhibit fecal stasis due to directional motility oriented toward the loop end, whereas SEL remain empty. In WT mice, SFL, but not SEL, develop pouchlike microbial communities without accompanying active inflammation. However, in genetically susceptible IL-10-deficient mice, SFL, but not SEL, exhibit severe inflammation and mucosal transcriptomes resembling human pouchitis. The inflammation associated with IL-10 required TLR4, as animals lacking both pathways displayed little disease. Furthermore, germ-free IL-10 mice conventionalized with SFL, but not SEL, microbiota populations develop severe colitis. These data support essential roles of stasis-induced, colon-like microbiota, TLR4-mediated colonic metaplasia, and genetic susceptibility in the development of pouchitis and possibly UC. However, these factors by themselves are not sufficient. Similarities between this model and human UC/pouchitis provide opportunities for gaining insights into the mechanistic basis of IBD and for identification of targets for novel preventative and therapeutic interventions.


Assuntos
Colite Ulcerativa/etiologia , Disbiose/complicações , Motilidade Gastrointestinal , Interleucina-10/genética , Receptor 4 Toll-Like/genética , Animais , Feminino , Humanos , Interleucina-10/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/microbiologia , Intestinos/patologia , Intestinos/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Microbiota , Receptor 4 Toll-Like/metabolismo
13.
PLoS One ; 9(5): e97456, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24857966

RESUMO

The Japanese traditional medicine daikenchuto (TU-100) has anti-inflammatory activities, but the mechanisms remain incompletely understood. TU-100 includes ginger, ginseng, and Japanese pepper, each component possessing bioactive properties. The effects of TU-100 and individual components were investigated in a model of intestinal T lymphocyte activation using anti-CD3 antibody. To determine contribution of intestinal bacteria, specific pathogen free (SPF) and germ free (GF) mice were used. TU-100 or its components were delivered by diet or by gavage. Anti-CD3 antibody increased jejunal accumulation of fluid, increased TNFα, and induced intestinal epithelial apoptosis in both SPF and GF mice, which was blocked by either TU-100 or ginger, but not by ginseng or Japanese pepper. TU-100 and ginger also blocked anti-CD3-stimulated Akt and NF-κB activation. A co-culture system of colonic Caco2BBE and Jurkat-1 cells was used to examine T-lymphocyte/epithelial cells interactions. Jurkat-1 cells were stimulated with anti-CD3 to produce TNFα that activates epithelial cell NF-κB. TU-100 and ginger blocked anti-CD3 antibody activation of Akt in Jurkat cells, decreasing their TNFα production. Additionally, TU-100 and ginger alone blocked direct TNFα stimulation of Caco2BBE cells and decreased activation of caspase-3 and polyADP ribose. The present studies demonstrate a new anti-inflammatory action of TU-100 that is microbe-independent and due to its ginger component.


Assuntos
Anticorpos Monoclonais/efeitos adversos , Enterite/tratamento farmacológico , NF-kappa B/metabolismo , Extratos Vegetais/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Linfócitos T/efeitos dos fármacos , Zingiber officinale/química , Animais , Anticorpos Monoclonais/imunologia , Apoptose/efeitos dos fármacos , Complexo CD3/imunologia , Linhagem Celular Tumoral , Enterite/induzido quimicamente , Enterite/imunologia , Enterite/metabolismo , Ativação Enzimática/efeitos dos fármacos , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia , Jejuno/efeitos dos fármacos , Jejuno/imunologia , Jejuno/patologia , Camundongos , Panax , Extratos Vegetais/uso terapêutico , Organismos Livres de Patógenos Específicos , Linfócitos T/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Zanthoxylum , Zingiberaceae
14.
J Vet Sci ; 15(2): 297-307, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24378587

RESUMO

This study was conducted to evaluate an adapter-modified Ussing chamber for assessment of transport physiology in endoscopically obtained duodenal biopsies from healthy cats and dogs, as well as dogs with chronic enteropathies. 17 duodenal biopsies from five cats and 51 duodenal biopsies from 13 dogs were obtained. Samples were transferred into an adapter-modified Ussing chamber and sequentially exposed to various absorbagogues and secretagogues. Overall, 78.6% of duodenal samples obtained from cats responded to at least one compound. In duodenal biopsies obtained from dogs, the rate of overall response ranged from 87.5% (healthy individuals; n = 8), to 63.6% (animals exhibiting clinical signs of gastrointestinal disease and histopathological unremarkable duodenum; n = 15), and 32.1% (animals exhibiting clinical signs of gastrointestinal diseases and moderate to severe histopathological lesions; n = 28). Detailed information regarding the magnitude and duration of the response are provided. The adapter-modified Ussing chamber enables investigation of the absorptive and secretory capacity of endoscopically obtained duodenal biopsies from cats and dogs and has the potential to become a valuable research tool. The response of samples was correlated with histopathological findings.


Assuntos
Biópsia/veterinária , Gatos/fisiologia , Cães/fisiologia , Duodenoscopia/veterinária , Duodeno/fisiologia , Animais , Doenças do Gato/fisiopatologia , Doenças do Cão/fisiopatologia , Duodenopatias/fisiopatologia , Duodeno/fisiopatologia
15.
J Clin Invest ; 123(9): 3983-96, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23945234

RESUMO

The inhibitory effects of vitamin D on colitis have been previously documented. Global vitamin D receptor (VDR) deletion exaggerates colitis, but the relative anticolitic contribution of epithelial and nonepithelial VDR signaling is unknown. Here, we showed that colonic epithelial VDR expression was substantially reduced in patients with Crohn's disease or ulcerative colitis. Moreover, targeted expression of human VDR (hVDR) in intestinal epithelial cells (IECs) protected mice from developing colitis. In experimental colitis models induced by 2,4,6-trinitrobenzenesulfonic acid, dextran sulfate sodium, or CD4(+)CD45RB(hi) T cell transfer, transgenic mice expressing hVDR in IECs were highly resistant to colitis, as manifested by marked reductions in clinical colitis scores, colonic histological damage, and colonic inflammation compared with WT mice. Reconstitution of Vdr-deficient IECs with the hVDR transgene completely rescued Vdr-null mice from severe colitis and death, even though the mice still maintained a hyperresponsive Vdr-deficient immune system. Mechanistically, VDR signaling attenuated PUMA induction in IECs by blocking NF-κB activation, leading to a reduction in IEC apoptosis. Together, these results demonstrate that gut epithelial VDR signaling inhibits colitis by protecting the mucosal epithelial barrier, and this anticolitic activity is independent of nonepithelial immune VDR actions.


Assuntos
Colite/metabolismo , Doenças Inflamatórias Intestinais/metabolismo , Mucosa Intestinal/patologia , Receptores de Calcitriol/metabolismo , Animais , Apoptose , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Colite/induzido quimicamente , Colo/metabolismo , Colo/patologia , Sulfato de Dextrana , Células Epiteliais/metabolismo , Células HCT116 , Humanos , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , NF-kappa B/metabolismo , Transdução de Sinais , Junções Íntimas/metabolismo , Ativação Transcricional , Ácido Trinitrobenzenossulfônico , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
16.
Int J Mol Sci ; 14(2): 2980-95, 2013 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-23434653

RESUMO

Compound K (20-O-beta-D-glucopyranosyl-20(S)-protopanaxadiol, CK), an intestinal bacterial metabolite of ginseng protopanaxadiol saponins, has been shown to inhibit cell growth in a variety of cancers. However, the mechanisms are not completely understood, especially in colorectal cancer (CRC). A xenograft tumor model was used first to examine the anti-CRC effect of CK in vivo. Then, multiple in vitro assays were applied to investigate the anticancer effects of CK including antiproliferation, apoptosis and cell cycle distribution. In addition, a qPCR array and western blot analysis were executed to screen and validate the molecules and pathways involved. We observed that CK significantly inhibited the growth of HCT-116 tumors in an athymic nude mouse xenograft model. CK significantly inhibited the proliferation of human CRC cell lines HCT-116, SW-480, and HT-29 in a dose- and time-dependent manner. We also observed that CK induced cell apoptosis and arrested the cell cycle in the G1 phase in HCT-116 cells. The processes were related to the upregulation of p53/p21, FoxO3a-p27/p15 and Smad3, and downregulation of cdc25A, CDK4/6 and cyclin D1/3. The major regulated targets of CK were cyclin dependent inhibitors, including p21, p27, and p15. These results indicate that CK inhibits transcriptional activation of multiple tumor-promoting pathways in CRC, suggesting that CK could be an active compound in the prevention or treatment of CRC.

17.
Dig Dis Sci ; 58(3): 668-77, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23329012

RESUMO

BACKGROUND: Lubiprostone has been used to treat constipation through its effects to stimulate Cl(-) secretion, resulting in water and electrolyte secretion. AIM: Potential associated changes in intestinal mucus and the colonizing bacteria (microbiome) have not been studied. As mucus obstructions may play a role in cystic fibrosis, the hypothesis that lubiprostone alters intestinal mucus and the microbiome was investigated. METHODS: Ion transport studies were performed ex vivo. For mucus and microbiome studies, mice were gavaged daily with lubiprostone or vehicle. Mucin from intestinal sections was analyzed in Carnoy's fixed tissues stained with Alcian blue. Microbiome composition was analyzed by 16S rRNA gene-based sequencing. RESULTS: Lubiprostone stimulated short circuit current in all mouse intestinal segments after both serosal and mucosal additions, albeit at lower concentrations in the latter. Current was Cl-dependent and blocked by mucosal diphenylcarboxylic acid, serosal bumetanide, and serosal Ba(++). The CFTR inhibitor CFTRinh172 had a marginal effect. Mucus near epithelial cells (inner layer mucus) was not present in the small intestine of any mice. Proximal colon inner mucus layer was thicker in ∆F/∆F compared with +/∆F and +/+ mice. Lubiprostone decreased inner mucus layer thickness in both proximal and distal colon of all mice. Furthermore, lubiprostone altered the intestinal microbiome by increasing abundance of Lactobacillus and Alistipes. CONCLUSIONS: Lubiprostone activates non-CFTR Cl(-) secretion and alters the colonic inner mucus layer, which is associated with changes in the composition of the enteric microbiome.


Assuntos
Alprostadil/análogos & derivados , Bactérias/efeitos dos fármacos , Colo/efeitos dos fármacos , Colo/microbiologia , Alprostadil/farmacologia , Animais , Ânions/metabolismo , Bactérias/classificação , Bactérias/genética , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Transporte de Íons/efeitos dos fármacos , Transporte de Íons/fisiologia , Lubiprostona , Camundongos , Camundongos Endogâmicos CFTR , Filogenia , RNA Bacteriano/genética , RNA Ribossômico 16S/genética , RNA Ribossômico 16S/metabolismo
18.
PLoS One ; 7(8): e44326, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22952955

RESUMO

The most feared complication following intestinal resection is anastomotic leakage. In high risk areas (esophagus/rectum) where neoadjuvant chemoradiation is used, the incidence of anastomotic leaks remains unacceptably high (≈ 10%) even when performed by specialist surgeons in high volume centers. The aims of this study were to test the hypothesis that anastomotic leakage develops when pathogens colonizing anastomotic sites become in vivo transformed to express a tissue destroying phenotype. We developed a novel model of anastomotic leak in which rats were exposed to pre-operative radiation as in cancer surgery, underwent distal colon resection and then were intestinally inoculated with Pseudomonas aeruginosa, a common colonizer of the radiated intestine. Results demonstrated that intestinal tissues exposed to preoperative radiation developed a significant incidence of anastomotic leak (>60%; p<0.01) when colonized by P. aeruginosa compared to radiated tissues alone (0%). Phenotype analysis comparing the original inoculating strain (MPAO1- termed P1) and the strain retrieved from leaking anastomotic tissues (termed P2) demonstrated that P2 was altered in pyocyanin production and displayed enhanced collagenase activity, high swarming motility, and a destructive phenotype against cultured intestinal epithelial cells (i.e. apoptosis, barrier function, cytolysis). Comparative genotype analysis between P1 and P2 revealed a single nucleotide polymorphism (SNP) mutation in the mexT gene that led to a stop codon resulting in a non-functional truncated protein. Replacement of the mutated mexT gene in P2 with mexT from the original parental strain P1 led to reversion of P2 to the P1 phenotype. No spontaneous transformation was detected during 20 passages in TSB media. Use of a novel virulence suppressing compound PEG/Pi prevented P. aeruginosa transformation to the tissue destructive phenotype and prevented anastomotic leak in rats. This work demonstrates that in vivo transformation of microbial pathogens to a tissue destroying phenotype may have important implications in the pathogenesis of anastomotic leak.


Assuntos
Fístula Anastomótica/microbiologia , Intestinos/microbiologia , Mutação/genética , Polimorfismo de Nucleotídeo Único/genética , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidade , Anastomose Cirúrgica/efeitos adversos , Fístula Anastomótica/patologia , Animais , Apoptose/efeitos dos fármacos , Sequência de Bases , Caenorhabditis elegans , Colo/efeitos dos fármacos , Colo/metabolismo , Colo/patologia , Intestinos/efeitos dos fármacos , Intestinos/patologia , Intestinos/ultraestrutura , Masculino , Dados de Sequência Molecular , Fenótipo , Fosfatos/farmacologia , Polietilenoglicóis/farmacologia , Substâncias Protetoras/farmacologia , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/isolamento & purificação , Radiação , Ratos , Ratos Wistar , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/metabolismo , Cicatrização/efeitos dos fármacos , Proteína da Zônula de Oclusão-1/metabolismo
19.
Res Vet Sci ; 93(3): 1454-61, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22721858

RESUMO

Adapter-modified Ussing chambers have been used for assessment of endoscopically obtained intestinal biopsies in humans. The aim of this study was to evaluate the feasibility of an adapter-modified Ussing chamber for assessment of intestinal transport physiology in endoscopically-obtained colonic biopsies from cats and dogs. Fifteen colonic biopsies from four cats and 13 colonic biopsies from four dogs were transferred into a modified Ussing chamber and sequentially exposed to several compounds. Baseline mean±SD conductance was measured. Changes of short circuit current (ΔIsc) were observed after exposure to glucose (number of feline biopsies that responded=0/number of canine biopsies that responded=4), phloridzin (n=0/n=7), histamine (n=5/n=12), serotonin (n=7/n=12), prostaglandin (n=5/n=7), forskolin (n=7/n=7), and ouabain (n=9/n=7). The adapter-modified Ussing chamber studied here enables investigation of transport physiology of endoscopically-obtained colonic biopsies from companion animals. However, we observed a large variability of results, suggesting that clinical use of this method is limited.


Assuntos
Doenças do Gato/patologia , Doenças do Colo/veterinária , Colonoscopia/veterinária , Doenças do Cão/patologia , Manejo de Espécimes/veterinária , Animais , Biópsia/métodos , Biópsia/veterinária , Gatos , Colforsina , Doenças do Colo/patologia , Cães , Condutividade Elétrica , Feminino , Glucose , Histamina , Masculino , Florizina , Serotonina , Manejo de Espécimes/instrumentação
20.
Int J Oncol ; 40(6): 1970-6, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22426808

RESUMO

Ginsenoside compound K (C-K) is an intestinal microbiota metabolite of ginsenoside Rb1, a major constituent in American ginseng. However, previous ginseng anti-cancer observations were largely focused on ginseng parent compounds but not metabolites, and anti-colorectal cancer studies on C-K were limited. This study investigated the anti-proliferative effects of C-K when compared to those of Rb1, and the related mechanisms of action, in HCT-116 and SW-480 colorectal cancer cells. The effects of Rb1 and C-K on the proliferation of HCT-116 and SW-480 human colorectal cancer cells were compared using an MTS assay. Cell cycle and cell apoptosis were assayed using flow cytometry. Enzymatic activities of caspases were determined by colorimetric assay, and interactions of C-K and caspases were explored by docking analysis. C-K showed significant anti-proliferative effects in HCT-116 and SW-480 cells at concentrations of 30-50 µM. At the same concentrations, Rb1 did not show any effects, while C-K arrested the cells in the G1 phase, and significantly induced cell apoptosis. Compared to HCT-116 (p53 wild-type), the p53 mutant cell line SW-480 was more sensitive to C-K as assessed by cell cycle regulation and apoptosis induction. C-K activated expression of caspases 8 and 9, consistent with docking analysis. The docking data suggested that C-K forms hydrogen bonds with Lys253, Thr904 and Gly362 in caspase 8, and with Thr62, Ser63 and Arg207 in caspase 9. C-K, but not its parent ginsenoside Rb1, showed significant anti-proliferative and pro-apoptotic effects in human colorectal cancer cells. These results suggest that C-K could be a potentially effective anti-colorectal cancer agent.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Colorretais/prevenção & controle , Ginsenosídeos/farmacologia , Animais , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Sítios de Ligação , Caspases/química , Caspases/metabolismo , Domínio Catalítico , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Simulação por Computador , Indução Enzimática , Ginsenosídeos/química , Humanos , Ligação de Hidrogênio , Camundongos , Modelos Moleculares , Ligação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA