Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Am J Physiol Gastrointest Liver Physiol ; 322(1): G154-G168, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34816756

RESUMO

Loss of functional small bowel surface area causes short bowel syndrome (SBS), intestinal failure, and parenteral nutrition (PN) dependence. The gut adaptive response following resection may be difficult to predict, and it may take up to 2 yr to determine which patients will wean from PN. Here, we examined features of gut microbiota and bile acid (BA) metabolism in determining adaptation and ability to wean from PN. Stool and sera were collected from healthy controls and from patients with SBS (n = 52) with ileostomy, jejunostomy, ileocolonic, and jejunocolonic anastomoses fed with PN plus enteral nutrition or who were exclusively enterally fed. We undertook 16S rRNA gene sequencing, BA profiling, and 7α-hydroxy-4-cholesten-3-one (C4) quantitation with LC-MS/MS and serum amino acid analyses. Patients with SBS exhibited altered gut microbiota with reduced gut microbial diversity compared with healthy controls. We observed differences in the microbiomes of patients with SBS with ileostomy versus jejunostomy, jejunocolonic versus ileocolonic anastomoses, and PN dependence compared with those who weaned from PN. Stool and serum BA composition and C4 concentrations were also altered in patients with SBS, reflecting adaptive changes in enterohepatic BA cycling. Stools from patients who were weaned from PN were enriched in secondary BAs including deoxycholic acid and lithocholic aicd. Shifts in gut microbiota and BA metabolites may generate a favorable luminal environment in select patients with SBS, promoting the ability to wean from PN. Proadaptive microbial species and select BA may provide novel targets for patient-specific therapies for SBS.NEW & NOTEWORTHY Loss of intestinal surface area causes short bowel syndrome, intestinal failure, and parenteral nutrition dependence. We analyzed the gut microbiota and bile acid metabolome of a large cohort of short bowel syndrome adult patients with different postsurgical anatomies. We report a novel analysis of the microbiome of patients with ileostomy and jejunostomy. Enrichment of specific microbial and bile acid species may be associated with the ability to wean from parenteral nutrition.


Assuntos
Ácidos e Sais Biliares/metabolismo , Fezes/microbiologia , RNA Ribossômico 16S/metabolismo , Síndrome do Intestino Curto/metabolismo , Adaptação Fisiológica/fisiologia , Cromatografia Líquida , Microbioma Gastrointestinal/fisiologia , Humanos , Intestino Delgado/metabolismo , Metaboloma/fisiologia , Microbiota/fisiologia
2.
Dig Dis Sci ; 65(11): 3271-3279, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-31907775

RESUMO

BACKGROUND: Few studies have examined the metabolic consequences of short bowel syndrome (SBS) and its effects on body composition in adults. We hypothesized that body composition of SBS patients is altered compared to a normal age-, race-, and sex-matched population, regardless of parenteral nutrition (PN) dependence. AIM: To compare the body composition of adult patients with SBS to age-, sex-, and race-matched healthy controls. METHODS: Twenty patients with SBS underwent body composition analysis using the GE Lunar iDXA scanner. Patients were age-, sex-, and race-matched to controls from the National Health and Nutrition Examination Survey (1999-2004). Mean differences in body mass index, fat-free mass, fat mass, percent body fat, visceral adipose tissue mass and volume, and bone mineral density were measured. Statistical analysis was performed using SAS 9.4 software. RESULTS: Fifty-five percent of subjects had a history of PN use, and 30% were current PN users. Mean percent body fat for SBS patients was 35.1% compared to 30.9% for healthy controls (p = 0.043). Fat-free mass was reduced in SBS (p = 0.007). Patients with reduced bone mass had a trend toward significantly more years of PN exposure compared to those with normal bone mass (p = 0.094), and a trend toward older age (p = 0.075). CONCLUSIONS: SBS is associated with increased percent body fat and reduced fat-free mass, suggesting that improved dietary and therapeutic interventions are needed to restore normal metabolic indices and avoid risk of metabolic syndrome in SBS patients.


Assuntos
Adiposidade , Composição Corporal , Índice de Massa Corporal , Síndrome do Intestino Curto/metabolismo , Absorciometria de Fóton , Estudos de Casos e Controles , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Inquéritos Nutricionais
3.
Am J Physiol Gastrointest Liver Physiol ; 315(2): G185-G194, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29631377

RESUMO

Stem cell therapy is a potential therapeutic approach for disorders characterized by intestinal injury or loss of functional surface area. Stem cell function and proliferation are mediated by the stem cell niche. Stromal cells such as intestinal subepithelial myofibroblasts (ISEMFs) are important but poorly studied components of the stem cell niche. To examine the role of ISEMFs, we have previously generated mice with deletion of epimorphin ( Epim), an ISEMF protein and member of the syntaxin family of intracellular vesicle docking proteins that regulate cell secretion. Herein we explore the mechanisms for previous observations that Epim deletion increases gut crypt cell proliferation, crypt fission, and small bowel length in vivo. Stem cell-derived crypt culture techniques were used to explore the interaction between enteroids and myofibroblasts from Epim-/- and WT mice. Enteroids cocultured with ISEMFS had increased growth and crypt-like budding compared with enteroids cultured without stromal support. Epim deletion in ISEMFs resulted in increased enteroid budding and surface area compared with cocultures with wild-type (WT) ISEMFs. In primary crypt cultures, Epim-/- enteroids had significantly increased surface area and budding compared with WTs. However, stem cell assays comparing the number of Epim-/- vs. WT colony-forming units after first passage showed no differences in the absence of ISEMF support. Epim-/- vs. WT ISEMFs had increased Wnt4 expression, and addition of Wnt4 to WT cocultures enhanced budding. We conclude that ISEMFs play an important role in the stem cell niche. Epim regulates stem cell proliferation and differentiation via stromal contributions to the niche microenvironment. NEW & NOTEWORTHY The role of subepithelial intestinal myofibroblasts (ISEMFs) in the gut stem cell niche is controversial. We provide novel evidence supporting ISEMFs as important niche contributors. We show that the in vivo intestinal effects of deletion of myofibroblast Epim can be recapitulated in crypt stem cell cultures in vitro. ISEMFs support cocultured stem cell proliferation and enteroid growth, and these effects are augmented by deletion of Epim, a syntaxin that regulates myofibroblast cell secretion.


Assuntos
Mucosa Intestinal/metabolismo , Intestino Delgado/citologia , Glicoproteínas de Membrana/metabolismo , Miofibroblastos/fisiologia , Nicho de Células-Tronco/fisiologia , Animais , Diferenciação Celular , Proliferação de Células , Microambiente Celular/fisiologia , Camundongos
4.
Dig Dis Sci ; 62(12): 3460-3467, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29094309

RESUMO

BACKGROUND: Colon cancer (CRC) is the third most common cancer worldwide. CRC develops through combinations of genetic and epigenetic changes. However, there is marked heterogeneity in the "driver gene" mutational profiles within and among colon cancers from individual patients, and these are not sufficient to explain differences in colon cancer behavior and treatment response. Global modulation of the tumor landscape may play a role in cancer behavior. Interferon-related developmental regulator 1 (IFRD1) is a transcriptional co-regulator that modulates expression of large gene cassettes and plays a role in gut epithelial proliferation following massive intestinal resection. AIMS: We address the hypothesis that increased IFRD1 expression in colon cancers is associated with poorer patient survival. METHODS: Tumor and normal tissue from colon cancer patient cohorts from the USA, Spain, and China were used for this study. Cancers were scored for the intensity of IFRD1 immunostaining. The primary clinical outcome was overall survival defined as time from diagnosis to death due to cancer. Kaplan-Meier method and log-rank analysis were used to assess the association between IFRD1 expression and survival. RESULTS: Almost all (98.7%) colon cancers showed readily detectable IFRD1 expression, with immunoreactivity primarily in the tumor cytoplasm. High IFRD1 colon cancer expression was significantly associated with decreased 5-year patient survival. Patients in the American cohort with high IFRD1 expression had a poorer prognosis. CONCLUSIONS: We have demonstrated that high IFRD1 protein expression in colon cancer is associated with poorer patient prognosis, suggesting a potential role for IFRD1 in modulating tumor behavior.


Assuntos
Adenocarcinoma/etiologia , Neoplasias do Colo/etiologia , Proteínas Imediatamente Precoces/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/mortalidade , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias do Colo/metabolismo , Neoplasias do Colo/mortalidade , Feminino , Humanos , Mucosa Intestinal/metabolismo , Masculino , Pessoa de Meia-Idade , Missouri/epidemiologia
5.
Cell Physiol Biochem ; 38(4): 1532-43, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27050729

RESUMO

BACKGROUND/AIMS: Expression of the transcriptional co-regulator tis7 is markedly increased in the adaptive small intestine in a mouse model of short bowel syndrome. Transgenic mice with enterocytic overexpression of tis7 (tis7tg) have accelerated triglyceride absorption, with increased adiposity yet reduced skeletal muscle mass. To further explore this phenotype, we examined whether tis7 also regulates amino acid and carbohydrate absorption. METHODS: Small intestinal glucose and amino acid uptake were quantified in wild type (WT) and tis7tg mice. Amino acid transporter expression was assessed by qRT-PCR and immunoblot. Apical cell surface transporter expression was quantified by cell surface biotinylation. RESULTS: Active glucose uptake rates were unchanged. Uptake of proline but not leucine was significantly reduced in tis7tg vs. WT jejunum. Expression of serum and glucocorticoid-induced kinase 1 (SGK1), a solute carrier activator, was inhibited in tis7tg jejunum. Apical membrane expression of the proline transporter SLC6A20 was reduced in tis7tg jejunum. CONCLUSIONS: Tis7 overexpression in enterocytes inhibits proline uptake, associated with decreased expression of activated SGK1 and reduced cell surface expression of SLC6A20. Consistent with the observed tis7tg phenotype, tis7 overexpression increases triglyceride absorption but has adverse effects on the uptake of selected amino acids. Tis7 has pleiotropic effects on nutrient absorption.


Assuntos
Proteínas Imediatamente Precoces/metabolismo , Jejuno/metabolismo , Proteínas de Membrana/metabolismo , Prolina/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Sistemas de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos/metabolismo , Animais , Western Blotting , Peso Corporal , Dieta Hiperlipídica , Glucose/metabolismo , Proteínas Imediatamente Precoces/sangue , Proteínas Imediatamente Precoces/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Fosforilação , Proteínas Serina-Treonina Quinases/sangue , Proteínas Serina-Treonina Quinases/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
6.
Am J Physiol Gastrointest Liver Physiol ; 307(6): G642-54, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25059825

RESUMO

Effective therapies are limited for patients with parenteral nutrition-dependent short bowel syndrome. We previously showed that intestinal expression of the transcriptional coregulator tetradecanoyl phorbol acetate-induced sequence 7 (tis7) is markedly increased during the adaptive response following massive small bowel resection and tis7 plays a role in normal gut lipid metabolism. Here, we further explore the functional implications of tis7 deletion in intestinal lipid metabolism and the adaptive response following small bowel resection. Intestinal tis7 transgenic (tis7(tg)), tis7(-/-), and wild-type (WT) littermates were subjected to 50% small bowel resection. Mice were fed a control or a high-saturated-fat (42% energy) diet for 21 days. Survival, body weight recovery, lipid absorption, mucosal lipid analysis, and the morphometric adaptive response were analyzed. Quantitative real-time PCR was performed to identify tis7 downstream gene targets. Postresection survival was markedly reduced in high-fat, but not control, diet-fed tis7(-/-) mice. Decreased survival was associated with anastomotic inflammation and intestinal obstruction postresection. High-fat, but not control, diet-fed tis7(-/-) mice had increased intestinal IL-6 expression. Intestinal lipid trafficking was altered in tis7(-/-) compared with WT mice postresection. In contrast, high-fat diet-fed tis7(tg) mice had improved survival postresection compared with WT littermates. High-fat diet feeding in the setting of tis7 deletion resulted in postresection anastomotic inflammation and small bowel obstruction. Tolerance of a calorie-rich, high-fat diet postresection may require tis7 and its target genes. The presence of luminal fat in the setting of tis7 deletion promotes an intestinal inflammatory response postresection.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Enterite/etiologia , Proteínas Imediatamente Precoces/deficiência , Obstrução Intestinal/etiologia , Intestino Delgado/metabolismo , Proteínas de Membrana/deficiência , Síndrome do Intestino Curto/complicações , Anastomose Cirúrgica , Animais , Modelos Animais de Doenças , Enterite/genética , Enterite/metabolismo , Regulação da Expressão Gênica , Proteínas Imediatamente Precoces/genética , Interleucina-6/metabolismo , Absorção Intestinal , Obstrução Intestinal/genética , Obstrução Intestinal/metabolismo , Intestino Delgado/cirurgia , Metabolismo dos Lipídeos , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Síndrome do Intestino Curto/genética , Síndrome do Intestino Curto/metabolismo , Fatores de Tempo
7.
Cell Mol Gastroenterol Hepatol ; 18(5): 101390, 2024 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-39128652

RESUMO

BACKGROUND & AIMS: Human sporadic colorectal cancer (CRC) results from a multistep pathway with sequential acquisition of specific genetic mutations in the colorectal epithelium. Modeling CRC in vivo is critical for understanding the tumor microenvironment. To accurately recapitulate human CRC pathogenesis, mouse models must include these multi-step genetic abnormalities. The aim of this study was to generate a sporadic CRC model that more closely mimics this multi-step process and to use this model to study the role of a novel Let7 target PLAGL2 in CRC pathogenesis. METHODS: We generated a CRISPR/Cas9 somatic mutagenesis mouse model that is inducible and multiplexed for simultaneous inactivation of multiple genes involved in CRC pathogenesis. We used both a doxycycline-inducible transcriptional activator and a doxycycline-inactivated transcriptional repressor to achieve tight, non-leaky expression of the Cas9 nickase. This mouse has transgenic expression of multiple guide RNAs to induce sporadic inactivation in the gut epithelium of 4 tumor suppressor genes commonly mutated in CRC, Apc, Pten, Smad4, and Trp53. These were crossed to Vil-LCL-PLAGL2 mice, which have Cre-inducible overexpression of PLAGL2 in the gut epithelium. RESULTS: These mice exhibited random somatic mutations in all 4 targeted tumor suppressor genes, resulting in multiple adenomas and adenocarcinomas in the small bowel and colon. Crosses with Vil-LCL-PLAGL2 mice demonstrated that gut-specific PLAGL2 overexpression increased colon tumor growth. CONCLUSIONS: This conditional model represents a new CRISPR/Cas9-mediated mouse model of colorectal carcinogenesis. These mice can be used to investigate the role of novel, previously uncharacterized genes in CRC, in the context of multiple commonly mutated tumor suppressor genes and thus more closely mimic human CRC pathogenesis.

8.
Am J Physiol Gastrointest Liver Physiol ; 305(8): G564-72, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-23886856

RESUMO

Interactions between the epithelium and surrounding mesenchyme/stroma play an important role in normal gut morphogenesis, the epithelial response to injury, and epithelial carcinogenesis. The tumor microenvironment, composed of stromal cells including myofibroblasts and immune cells, regulates tumor growth and the cancer stem cell niche. Deletion of epimorphin (Epim), a syntaxin family member expressed in myofibroblasts and macrophages, results in partial protection from colitis and from inflammation-induced colon cancer in mice. We sought to determine whether epimorphin deletion protects from polyposis in the Apcmin/+ mouse model of intestinal carcinogenesis. Epim-/- mice were crossed to Apcmin/+ mice; Apcmin/+ and Apcmin/+/Epim-/- mice were killed at 3 mo of age. Polyp numbers and sizes were quantified in small intestine and colon, and gene expression analyses for pathways relevant to epithelial carcinogenesis were performed. Primary myofibroblast cultures were isolated, and expression and secretion of selected growth factors from Apcmin/+ and Apcmin/+/Epim-/- myofibroblasts were examined by ELISA. Small bowel polyposis was significantly inhibited in Apcmin/+/Epim-/- compared with Apcmin/+ mice. Apcmin/+/Epim-/- compared with Apcmin/+ polyps and adjacent uninvolved intestinal mucosa had increased transforming growth factor-ß (TGF-ß) expression and signaling with increased P-Smad2/3 expression. Myofibroblasts isolated from Apcmin/+/Epim-/- vs. Apcmin/+ mice had markedly decreased hepatocyte growth factor (HGF) expression and secretion. We concluded that Epim deletion inhibits polyposis in Apcmin/+ mice, associated with increased mucosal TGF-ß signaling and decreased myofibroblast HGF expression and secretion. Our data suggest that Epim deletion reduces tumorigenicity of the stromal microenvironment.


Assuntos
Proteína da Polipose Adenomatosa do Colo/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Glicoproteínas de Membrana/metabolismo , Miofibroblastos/metabolismo , Polipose Adenomatosa do Colo/genética , Polipose Adenomatosa do Colo/metabolismo , Polipose Adenomatosa do Colo/patologia , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Neoplasias do Colo/metabolismo , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Fator de Crescimento de Hepatócito/genética , Mucosa Intestinal/fisiologia , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
9.
Am J Pathol ; 180(3): 984-997, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22222225

RESUMO

Dendritic cells (DCs) use all-trans retinoic acid (ATRA) to promote characteristic intestinal responses, including Foxp3(+) Treg conversion, lymphocyte gut homing molecule expression, and IgA production. How this ability to generate ATRA is conferred to DCs in vivo remains largely unstudied. Here, we observed that among DCs, retinaldehyde dehydrogenase (ALDH1), which catalyzes the conversion of retinal to ATRA, was preferentially expressed by small intestine CD103(+) lamina propria (LP) DCs. Retinoids induced LP CD103(+) DCs to generate ATRA via ALDH1 activity. Either biliary or dietary retinoids were required to confer ALDH activity to LP DCs in vivo. Cellular retinol-binding protein II (CRBPII), a cytosolic retinoid chaperone that directs enterocyte retinol and retinal metabolism but is redundant to maintain serum retinol, was required to confer ALDH activity to CD103(+) LP DCs. CRBPII expression was restricted to small intestine epithelial cells, and ALDH activity in CRBPII(-/-) DCs was restored by transfer to a wild-type recipient. CD103(+) LP DCs from CRBPII(-/-) mice had a decreased capacity to promote IgA production. Moreover, CD103(+) DCs preferentially associated with the small intestine epithelium and LP CD103(+) DC ALDH activity, and the ability to promote IgA production was reduced in mice with impaired DC-epithelia associations. These findings demonstrate in vivo roles for the expression of epithelial CRBPII and lumenal retinoids to imprint local gut DCs with an intestinal phenotype.


Assuntos
Células Dendríticas/metabolismo , Imunoglobulina A/biossíntese , Intestino Delgado/metabolismo , Isoenzimas/metabolismo , Retinal Desidrogenase/metabolismo , Proteínas Celulares de Ligação ao Retinol/metabolismo , Tretinoína/metabolismo , Família Aldeído Desidrogenase 1 , Animais , Antígenos CD/metabolismo , Células Dendríticas/imunologia , Imunidade Celular/fisiologia , Cadeias alfa de Integrinas/metabolismo , Interleucina-6/metabolismo , Intestino Delgado/citologia , Intestino Delgado/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Fenótipo
10.
J Nutr ; 140(11): 1907-14, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20861213

RESUMO

After loss of intestinal surface area, the remaining bowel undergoes a morphometric and functional adaptive response. Enterocytic expression of the transcriptional coregulator tetradecanoyl phorbol acetate induced sequence 7 (Tis7) is markedly increased in a murine model of intestinal adaptation. Mice overexpressing Tis7 in intestine have greater triglyceride absorption and weight gain when fed a high-fat diet (42% energy) than their wild-type (WT) littermates fed the same diet. These and other data suggest that Tis7 has a unique role in nutrient absorptive and metabolic adaptation. Herein, male Tis7(-/-) and WT mice were fed a high-fat diet (42% energy) for 8 wk. Weight was monitored and metabolic analyses and hepatic and intestinal lipid concentrations were compared after 8 wk. Intestinal lipid absorption and metabolism studies and intestinal resection surgeries were performed in separate groups of Tis7(-/-) and WT mice. At 8 wk, weight gain was less and jejunal mucosal and hepatic triglyceride and cholesterol concentrations were lower in Tis7(-/-) mice than in the WT controls. Following corn oil gavage, serum cholesterol, triglyceride, and FFA concentrations were lower in the Tis7(-/-) mice than in the WT mice. Incorporation of oral (3)[H] triolein into intestinal mucosal cholesterol ester and FFA was less in Tis7(-/-) compared with WT mice. Following resection, crypt cell proliferation rates and villus heights were lower in Tis7(-/-) than in WT mice, indicating a blunted adaptive response. Our results suggest a novel physiologic function for Tis7 in the gut as a global regulator of lipid absorption and metabolism and epithelial cell proliferation.


Assuntos
Adaptação Fisiológica/genética , Gorduras na Dieta/efeitos adversos , Proteínas Imediatamente Precoces/genética , Intestinos/fisiopatologia , Metabolismo dos Lipídeos/genética , Proteínas de Membrana/genética , Síndrome do Intestino Curto/fisiopatologia , Aumento de Peso/genética , Animais , Proliferação de Células , Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Regulação da Expressão Gênica/genética , Absorção Intestinal/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Intestino Delgado/cirurgia , Intestinos/patologia , Lipídeos/análise , Lipídeos/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/metabolismo , Síndrome do Intestino Curto/metabolismo , Síndrome do Intestino Curto/patologia , Fatores de Tempo , Triglicerídeos/metabolismo
11.
JCI Insight ; 5(23)2020 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-33141758

RESUMO

Loss of functional small bowel surface area following surgical resection for disorders such as Crohn's disease, intestinal ischemic injury, radiation enteritis, and in children, necrotizing enterocolitis, atresia, and gastroschisis, may result in short bowel syndrome, with attendant high morbidity, mortality, and health care costs in the United States. Following resection, the remaining small bowel epithelium mounts an adaptive response, resulting in increased crypt cell proliferation, increased villus height, increased crypt depth, and enhanced nutrient and electrolyte absorption. Although these morphologic and functional changes are well described in animal models, the adaptive response in humans is less well understood. Clinically the response is unpredictable and often inadequate. Here we address the hypotheses that human intestinal stem cell populations are expanded and that the stem cell niche is regulated following massive gut resection in short bowel syndrome (SBS). We use intestinal enteroid cultures from patients with SBS to show that the magnitude and phenotype of the adaptive stem cell response are both regulated by stromal niche cells, including intestinal subepithelial myofibroblasts, which are activated by intestinal resection to enhance epithelial stem and proliferative cell responses. Our data suggest that myofibroblast regulation of bone morphogenetic protein signaling pathways plays a role in the gut adaptive response after resection.


Assuntos
Células-Tronco Adultas/metabolismo , Mucosa Intestinal/metabolismo , Síndrome do Intestino Curto/fisiopatologia , Células-Tronco Adultas/fisiologia , Idoso , Doença de Crohn/metabolismo , Enterite/metabolismo , Feminino , Humanos , Mucosa Intestinal/crescimento & desenvolvimento , Intestinos , Masculino , Pessoa de Meia-Idade , Miofibroblastos/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Síndrome do Intestino Curto/metabolismo , Transdução de Sinais
12.
J Clin Invest ; 116(6): 1535-46, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16710473

RESUMO

Dynamic and reciprocal epithelial-mesenchymal interactions are critical for the normal morphogenesis and maintenance of epithelia. Epimorphin has been identified as a unique molecule expressed by mesenchymal cells and myofibroblasts and has putative morphogenetic effects in multiple epithelial tissues, including intestine, skin, mammary gland, lung, gallbladder, and liver. To define the in vivo role of epimorphin, we created epimorphin-null mice by targeted inactivation of the epimorphin gene. Male epimorphin-/- mice are sterile due to abnormal testicular development and impaired spermatogenesis. Intestinal growth is increased in epimorphin-/- mice due to augmented crypt cell proliferation and crypt fission during the neonatal (suckling) period, mediated at least in part by changes in bone morphogenetic protein (Bmp) and Wnt/beta-catenin signaling pathways. Colonic mucosal injury and colitis induced by dextran sodium sulfate (DSS) are ameliorated in epimorphin-/- mice, probably due to the increased proliferative capacity of the epimorphin-/- colon. These in vivo findings support the notion that epimorphin is a key stromal regulator of epithelial cell proliferation and growth in the intestine. In addition, our studies demonstrate a novel and critical role for epimorphin in regulating testicular development and growth as well as spermatogenesis.


Assuntos
Colite/induzido quimicamente , Sulfato de Dextrana/toxicidade , Intestinos/crescimento & desenvolvimento , Glicoproteínas de Membrana/metabolismo , Espermatogênese/fisiologia , Animais , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Linhagem Celular , Colite/patologia , Feminino , Marcação de Genes , Indicadores e Reagentes/toxicidade , Intestinos/citologia , Intestinos/patologia , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Morfogênese , Testículo/citologia , Testículo/patologia , Testículo/fisiologia , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
13.
J Clin Invest ; 110(11): 1629-41, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12464668

RESUMO

The formation of the crypt-villus axis during gut ontogeny requires continued reciprocal interactions between the endoderm and mesenchyme. Epimorphin/syntaxin 2 (epimorphin) is a mesenchymal protein expressed in the fetal gastrointestinal tract during villus morphogenesis. To elucidate its role in gut ontogeny, the epimorphin cDNA was transfected, in sense and antisense orientations, into a rat intestinal myofibroblast cell line, MIC 216. To determine the effects of epimorphin on the epithelium, myofibroblasts were cocultured with the Caco2 cell line. Caco2 cells spread in a simple monolayer over antisense-transfected cells lacking epimorphin. In contrast, sense-transfected myofibroblasts induced Caco2 cells to form compact, round clusters with small lumens. These morphologic differences were preserved in Transwell cocultures in which cell-cell contact was prevented, suggesting that epimorphin's effects were mediated by secreted factor(s). To determine the effects of epimorphin on crypt-villus axis formation in an in vivo model, rat gut endoderm was combined with epimorphin-transfected myofibroblasts and implanted into the chick intracoelomic cavity. The grafts in which epimorphin was overexpressed revealed multiple well-formed villi with crypt-like units, whereas those in which epimorphin expression was inhibited developed into round cystic structures without crypts or villi. Of several potential secreted morphogens, only the expression of bone morphogenetic protein 4 (Bmp4) was increased in the epimorphin-transfected cells. Incubation with noggin partially blocked the transfected myofibroblasts' effects on Caco2 colony morphology. These results indicate that mesenchymal epimorphin has profound effects on crypt-villus morphogenesis, mediated in part by secreted factor(s) including the Bmp's.


Assuntos
Mucosa Intestinal/citologia , Intestinos/fisiologia , Glicoproteínas de Membrana/genética , Actinas/genética , Animais , Linhagem Celular , Técnicas de Cocultura , Regulação da Expressão Gênica , Humanos , Imuno-Histoquímica , Mucosa Intestinal/fisiologia , Glicoproteínas de Membrana/fisiologia , Morfogênese/fisiologia , Músculo Liso/fisiologia , Ratos , Sintaxina 1 , Células Tumorais Cultivadas
14.
Best Pract Res Clin Gastroenterol ; 30(2): 237-48, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27086888

RESUMO

Following loss of functional small bowel surface area due to surgical resection for therapy of Crohn's disease, ischemia, trauma or other disorders, the remnant gut undergoes a morphometric and functional compensatory adaptive response which has been best characterized in preclinical models. Increased crypt cell proliferation results in increased villus height, crypt depth and villus hyperplasia, accompanied by increased nutrient, fluid and electrolyte absorption. Clinical observations suggest that functional adaptation occurs in humans. In the immediate postoperative period, patients with substantial small bowel resection have massive fluid and electrolyte loss with reduced nutrient absorption. For many patients, the adaptive response permits partial or complete weaning from parenteral nutrition (PN), within two years following resection. However, others have life-long PN dependence. An understanding of the molecular mechanisms that regulate the gut adaptive response is critical for developing novel therapies for short bowel syndrome. Herein we present a summary of key studies that seek to elucidate the mechanisms that regulate post-resection adaptation, focusing on stem and crypt cell proliferation, epithelial differentiation, apoptosis, enterocyte function and the role of growth factors and the enteric nervous system.


Assuntos
Adaptação Fisiológica , Intestino Delgado/fisiopatologia , Síndrome do Intestino Curto/fisiopatologia , Sistema Nervoso Entérico/fisiopatologia , Humanos , Mucosa Intestinal , Nutrição Parenteral
15.
JPEN J Parenter Enteral Nutr ; 27(2): 123-31, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12665168

RESUMO

BACKGROUND: To define the molecular mechanisms underlying the intestinal adaptive response after partial small bowel resection, we previously identified a cohort of genes regulated in the remnant adaptive ileum. One is PC4/TIS7, an immediate early gene preferentially up-regulated during the first 48 hours after resection. To further the mechanisms that regulate gut adaptation, we sought to identify upstream regulators of PC4/TIS7 expression. METHODS: PC4/TIS7 expression in adaptive versus transection control mouse gut was examined at 48 hours after 50% intestinal resection, and its cellular localization was determined by immunohistochemistry. The effects of intestinotrophic peptides and growth factors on PC4/TIS7 expression were examined in vitro in the crypt epithelial cell line IEC 18 and in vivo in the mouse. RESULTS: PC4/TIS7 was expressed in the cytoplasm of IEC 18 cells and in adaptive mouse ileal crypt and villus enterocytes. Epidermal growth factor (EGF) and basic fibroblast growth factor (bFGF) increased PC4/TIS7 mRNA levels in postconfluent, quiescent IEC 18 cells, but insulin-like growth factor 1 (IGF-1) and nerve growth factor (NGF) had no effect. A stable derivative of glucagon-like peptide 2 (GLP-2), r(gly2)GLP-2, was most potent in increasing PC4/TIS7 expression; however, stimulation of proliferation and differentiation were not observed. To determine the effect of GLP-2 on PC4/TIS7 expression in vivo, r(gly2)GLP-2 was administered intraperitoneally to mice. PC4/TIS7 mRNA expression was increased in small bowel in response to GLP-2 compared with vehicle control. CONCLUSIONS: These results suggest that PC4/TIS7 plays a role in intracellular signaling in the intestinal epithelium during the adaptive response, possibly as a common downstream effector for several intestinotrophic growth factors.


Assuntos
Regulação da Expressão Gênica , Substâncias de Crescimento/farmacologia , Proteínas Imediatamente Precoces/genética , Intestino Delgado/fisiologia , Intestino Delgado/cirurgia , Proteínas de Membrana/genética , Adaptação Fisiológica/genética , Animais , Northern Blotting , Linhagem Celular , DNA Complementar , Fator de Crescimento Epidérmico/farmacologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Peptídeo 2 Semelhante ao Glucagon , Peptídeos Semelhantes ao Glucagon , Substâncias de Crescimento/fisiologia , Íleo/fisiologia , Íleo/cirurgia , Proteínas Imediatamente Precoces/biossíntese , Proteínas Imediatamente Precoces/fisiologia , Imuno-Histoquímica , Fator de Crescimento Insulin-Like I/farmacologia , Proteínas de Membrana/biossíntese , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Peptídeos/farmacologia , RNA Mensageiro/biossíntese , Distribuição Aleatória
16.
Transl Res ; 164(1): 70-83, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24731292

RESUMO

Epimorphin (Epim), a member of the syntaxin family of membrane-bound, intracellular vesicle-docking proteins, is expressed in intestinal myofibroblasts and macrophages. We demonstrated previously that Epimorphin(-/-)(Epim(-/-)) mice are protected, in part, from dextran sodium sulfate (DSS)-induced colitis. Although interleukin (IL)-6/p-Stat3 signaling has been implicated in the pathogenesis of colitis, the myofibroblast contribution to IL-6 signaling in colitis remains unexplored. Our aim was to investigate the IL-6 pathway in Epim(-/-) mice in the DSS colitis model. Whole colonic tissue, epithelium, and stroma of WT and congenic Epim(-/-) mice treated with 5% DSS for 7 days were analyzed for IL-6 and a downstream effector, p-Stat3, by immunostaining and immunoblot. Colonic myofibroblast and peritoneal macrophage IL-6 secretion were evaluated by enzyme-linked immunosorbent assay. IL-6 and p-Stat3 expression were decreased in Epim(-/-) vs WT colon. A relative increase in stromal vs epithelial p-Stat3 expression was observed in WT mice but not in Epim(-/-) mice. Epim deletion abrogates IL-6 secretion from colonic myofibroblasts treated with IL-1ß and decreases IL-6 secretion from peritoneal macrophages in a subset of DSS-treated mice. Epim deletion inhibits IL-6 secretion most profoundly from colonic myofibroblasts. Distribution of Stat3 activation is altered in DSS-treated Epim(-/-) mice. Our findings support the notion that myofibroblasts modulate IL-6/p-Stat3 signaling in DSS-treated Epim(-/-) mice.


Assuntos
Colite/induzido quimicamente , Interleucina-6/metabolismo , Glicoproteínas de Membrana/metabolismo , Transdução de Sinais , Animais , Sulfato de Dextrana/toxicidade , Regulação da Expressão Gênica/fisiologia , Interleucina-6/genética , Mucosa Intestinal/patologia , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
17.
Front Immunol ; 3: 107, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22586430

RESUMO

The inflammatory bowel diseases (IBD), including Crohn's disease (CD) and ulcerative colitis (UC), are chronic inflammatory disorders of the intestine. The prevalence in the United States is greater than 200 cases per 100,000, with the total number of IBD patients between 1 and 1.5 million. CD may affect all parts of the gastrointestinal tract, from mouth to anus, but most commonly involves the distal part of the small intestine or ileum, and colon. UC results in colonic inflammation that can affect the rectum only, or can progress proximally to involve part of or the entire colon. Clinical symptoms include diarrhea, abdominal pain, gastrointestinal bleeding, and weight loss. A serious long-term complication of chronic inflammation is the development of colorectal cancer. A genetic basis for IBD had long been recognized based on the increased familial risk. However, significant discordance for CD in twins, and a much less robust phenotypic concordance for UC, suggested additional factors play a role in disease pathogenesis, including environmental factors. In the past several years, progress in understanding the molecular basis of IBD has accelerated, beginning with the generation of animal models of colitis and progressing to the identification of specific genetic markers from candidate gene, gene linkage, and genome-wide association analyses. Genetic studies have also resulted in the recognition of the importance of environmental factors, particularly the crucial role of the gut microbiota in CD and UC. Altered immune responses to the normal intestinal flora are key factors in IBD pathogenesis. In this research topic, the genetic basis of IBD, the genetic and cellular alterations associated with colitis-associated colon cancer, and the emerging role of the intestinal microbiota and other environmental factors will be reviewed.

18.
J Clin Invest ; 120(6): 2081-93, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20458144

RESUMO

Epithelial-mesenchymal interactions regulate normal gut epithelial homeostasis and have a putative role in inflammatory bowel disease and colon cancer pathogenesis. Epimorphin is a mesenchymal and myofibroblast protein with antiproliferative, promorphogenic effects in intestinal epithelium. We previously showed that deletion of epimorphin partially protects mice from acute colitis, associated with an increase in crypt cell proliferation. Here we explored the potential therapeutic utility of modulating epimorphin expression by examining the effects of epimorphin deletion on chronic inflammation-associated colon carcinogenesis using the azoxymethane/dextran sodium sulfate (AOM/DSS) model. We found that mice in which epimorphin expression was absent had a marked reduction in incidence and extent of colonic dysplasia. Furthermore, epimorphin deletion in myofibroblasts altered the morphology and growth of cocultured epithelial cells. Loss of epimorphin affected secretion of soluble mesenchymal regulators of the stem cell niche such as Chordin. Importantly, IL-6 secretion from LPS-treated epimorphin-deficient myofibroblasts was completely inhibited, and stromal IL-6 expression was reduced in vivo. Taken together, these data show that epimorphin deletion inhibits chronic inflammation-associated colon carcinogenesis in mice, likely as a result of increased epithelial repair, decreased myofibroblast IL-6 secretion, and diminished IL-6-induced inflammation. Furthermore, we believe that modulation of epimorphin expression may have therapeutic benefits in appropriate clinical settings.


Assuntos
Colo/patologia , Neoplasias do Colo/patologia , Inflamação/metabolismo , Interleucina-6/metabolismo , Músculo Liso/metabolismo , Animais , Azoximetano/efeitos adversos , Azoximetano/metabolismo , Azoximetano/farmacologia , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Colite/induzido quimicamente , Colite/genética , Colite/patologia , Colo/efeitos dos fármacos , Colo/metabolismo , Neoplasias do Colo/complicações , Neoplasias do Colo/genética , Sulfato de Dextrana/efeitos adversos , Sulfato de Dextrana/metabolismo , Sulfato de Dextrana/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Inflamação/genética , Inflamação/patologia , Interleucina-6/genética , Interleucina-6/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso/efeitos dos fármacos , Músculo Liso/patologia , Deleção de Sequência
19.
JPEN J Parenter Enteral Nutr ; 33(6): 662-8, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19597188

RESUMO

BACKGROUND: Alcohol consumption is associated with oxidative stress in multiple tissues in vivo, yet the effect of chronic alcohol intake on intestinal redox state has received little attention. In this study, we investigated the redox status of 2 major intracellular redox regulating couples: glutathione (GSH)/glutathione disulfide (GSSG) and cysteine (Cys)/cystine (CySS) in a rat model of chronic alcohol ingestion. METHODS: Sprague-Dawley rats were fed the liquid Lieber-DeCarli diet consisting of 36% ethanol of total calories for 6 weeks. Control rats were pair-fed with an isocaloric, ethanol-free liquid diet. Defined mucosal samples from the jejunum, ileum, and colon were obtained and analyzed by high-performance liquid chromatography (HPLC) for GSH and Cys pool redox status. Mucosal free malondialdehyde (MDA) was measured as an indicator of lipid peroxidation. RESULTS: In the ethanol-fed rats, Cys and mixed disulfide (GSH-Cys) were significantly decreased in all 3 segments of intestinal mucosa. Free MDA was increased in jejunal but not in ileal or colonic mucosa. Chronic ethanol ingestion significantly increased mucosal GSH concentration in association with a more reducing GSH/GSSG redox potential in the jejunum, but these indices were unchanged in the ileum. In the colon, chronic ethanol ingestion increased oxidant stress as suggested by decreased GSH and oxidized GSH/GSSG redox potential. CONCLUSIONS: Chronic alcohol intake differentially alters the mucosal redox status in proximal to distal intestinal segments in rats. Such changes may reflect different adaptability of these intestinal segments to the oxidative stress challenge induced by chronic ethanol ingestion.


Assuntos
Alcoolismo/metabolismo , Antioxidantes/metabolismo , Etanol/efeitos adversos , Mucosa Intestinal/metabolismo , Peroxidação de Lipídeos , Oxirredução , Animais , Colo/metabolismo , Cisteína/metabolismo , Cistina/metabolismo , Modelos Animais de Doenças , Glutationa/metabolismo , Dissulfeto de Glutationa/metabolismo , Intestino Delgado/metabolismo , Masculino , Malondialdeído/metabolismo , Ratos , Ratos Sprague-Dawley , Compostos de Sulfidrila/metabolismo
20.
Am J Physiol Gastrointest Liver Physiol ; 292(6): G1559-69, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17307727

RESUMO

Following the loss of functional small bowel surface area, the intestine undergoes a compensatory adaptive response. The observation that adaptation is inhibited in vitamin A-deficient rats following submassive intestinal resection suggested that vitamin A is required for this response and raised the possibility that exogenous vitamin A could augment adaptation. Therefore, to directly assess whether chronically administered retinoic acid could stimulate gut adaptation in a model of short bowel syndrome and to address the mechanisms of any such effects, Sprague-Dawley rats were implanted with controlled release retinoic acid or control pellets and then subjected to mid-small bowel or sham resections. At 2 wk postoperation, changes in gut morphology, crypt cell proliferation and apoptosis, enterocyte migration, the extracellular matrix, and gene expression were assessed. Retinoic acid had significant trophic effects in resected and sham-resected rats. Retinoic acid markedly inhibited apoptosis and stimulated crypt cell proliferation and enterocyte migration postresection. Data presented indicate that these proadaptive effects of retinoic acid may be mediated via changes in the extracellular matrix (e.g., by increasing collagen IV synthesis, decreasing E-cadherin expression, and reducing integrin beta(3) levels), via affects on Hedgehog signaling (e.g., by reducing expression of the Hedgehog receptors Ptch and Ptch2 and the Gli1 transcription factor), by increasing expression of Reg1 and Pap1, and by modulation of retinoid and peroxisome proliferator-activated receptor signaling pathways. These studies are the first to demonstrate that retinoic acid can significantly enhance intestinal adaptation and suggest it may be beneficial in patients with short bowel syndrome.


Assuntos
Adaptação Fisiológica/efeitos dos fármacos , Intestino Delgado/efeitos dos fármacos , Receptores do Ácido Retinoico/agonistas , Síndrome do Intestino Curto/tratamento farmacológico , Tretinoína/farmacologia , Adaptação Fisiológica/genética , Animais , Apoptose/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Implantes de Medicamento , Enterócitos/efeitos dos fármacos , Enterócitos/patologia , Proteínas da Matriz Extracelular/metabolismo , Expressão Gênica/efeitos dos fármacos , Proteínas Hedgehog/metabolismo , Intestino Delgado/metabolismo , Intestino Delgado/patologia , Intestino Delgado/fisiopatologia , Intestino Delgado/cirurgia , Masculino , Proteínas Associadas a Pancreatite , Receptores Ativados por Proliferador de Peroxissomo/efeitos dos fármacos , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores do Ácido Retinoico/genética , Receptores do Ácido Retinoico/metabolismo , Síndrome do Intestino Curto/genética , Síndrome do Intestino Curto/metabolismo , Síndrome do Intestino Curto/patologia , Síndrome do Intestino Curto/fisiopatologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Tretinoína/administração & dosagem , Tretinoína/uso terapêutico , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA