Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
J Pediatr Surg ; 53(6): 1203-1207, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29636182

RESUMO

BACKGROUND: Epidermal Growth Factor (EGF) reduces necrotizing enterocolitis (NEC). However, its high cost virtually prohibits clinical use. To reduce cost, soybean expressing human EGF was developed. Here we report effectiveness of soybean-derived EGF in experimental NEC. METHODS: Newborn rats were subjected to the NEC-inducing regimen of formula feeding and hypoxia. Formula was supplemented with extract from EGF-expressing or empty soybeans. NEC pathology was determined microscopically. Localization of tight junction proteins JAM-A and ZO-1 was examined by immunofluorescence and levels of mucosal COX-2 and iNOS mRNAs by real time PCR. RESULTS: Soybean extract amounts corresponding to 150µg/kg/day EGF caused considerable mortality, whereas those corresponding to 75µg/kg/day EGF were well tolerated. There was no significant difference in NEC scores between animals fed plain formula and formula supplemented with empty soybean extract. Soybean-EGF-supplemented formula at 75µg/kg/day EGF significantly decreased NEC, attenuated dissociation of JAM-A and ZO-1 proteins from tight junctions, and reduced intestinal expression of COX-2 and iNOS mRNAs. CONCLUSION: Supplementation with soybean-expressed EGF significantly decreased NEC in the rat model. Soybean-expressed EGF may provide an economical solution for EGF administration and prophylaxis of clinical NEC.


Assuntos
Enterocolite Necrosante/prevenção & controle , Fator de Crescimento Epidérmico/uso terapêutico , Glycine max , Extratos Vegetais/uso terapêutico , Substâncias Protetoras/uso terapêutico , Animais , Animais Recém-Nascidos , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , Enterocolite Necrosante/patologia , Humanos , Fórmulas Infantis , Recém-Nascido , Recém-Nascido Prematuro , Doenças do Prematuro/patologia , Doenças do Prematuro/prevenção & controle , Mucosa Intestinal/metabolismo , Intestinos/patologia , Moléculas de Adesão Juncional/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Proteínas Recombinantes/uso terapêutico , Proteínas da Zônula de Oclusão/metabolismo
2.
Sci Rep ; 7(1): 5580, 2017 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-28717211

RESUMO

Intestinal tuft cells are one of 4 secretory cell linages in the small intestine and the source of IL-25, a critical initiator of the type 2 immune response to parasite infection. When Raptor, a critical scaffold protein for mammalian target of rapamycin complex 1 (mTORC1), was acutely deleted in intestinal epithelium via Tamoxifen injection in Tritrichomonas muris (Tm) infected mice, tuft cells, IL-25 in epithelium and IL-13 in the mesenchyme were significantly reduced, but Tm burden was not affected. When Tm infected mice were treated with rapamycin, DCLK1 and IL-25 expression in enterocytes and IL-13 expression in mesenchyme were diminished. After massive small bowel resection, tuft cells and Tm were diminished due to the diet used postoperatively. The elimination of Tm and subsequent re-infection of mice with Tm led to type 2 immune response only in WT, but Tm colonization in both WT and Raptor deficient mice. When intestinal organoids were stimulated with IL-4, tuft cells and IL-25 were induced in both WT and Raptor deficient organoids. In summary, our study reveals that enterocyte specific Raptor is required for initiating a type 2 immune response which appears to function through the regulation of mTORC1 activity.


Assuntos
Enterócitos/citologia , Intestino Delgado/citologia , Infecções Protozoárias em Animais/imunologia , Proteína Regulatória Associada a mTOR/deficiência , Sirolimo/administração & dosagem , Tritrichomonas/imunologia , Animais , Quinases Semelhantes a Duplacortina , Regulação para Baixo , Enterócitos/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Imunidade nas Mucosas/efeitos dos fármacos , Interleucina-13/genética , Interleucina-13/metabolismo , Interleucinas/genética , Interleucinas/metabolismo , Intestino Delgado/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Infecções Protozoárias em Animais/tratamento farmacológico , Sirolimo/farmacologia , Tamoxifeno/administração & dosagem , Tamoxifeno/farmacologia
3.
Cell Mol Gastroenterol Hepatol ; 3(2): 231-244, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28275690

RESUMO

BACKGROUND & AIMS: Intestinal adaptation is a compensatory response to the massive loss of small intestine after surgical resection. We investigated the role of intestinal epithelial cell-specific mammalian target of rapamycin complex 1 (i-mTORC1) in intestinal adaptation after massive small bowel resection (SBR). METHODS: We performed 50% proximal SBR on mice to study adaptation. To manipulate i-mTORC1 activity, Villin-CreER transgenic mice were crossed with tuberous sclerosis complex (TSC)1flox/flox or Raptorflox/flox mice to inducibly activate or inactivate i-mTORC1 activity with tamoxifen. Western blot was used to confirm the activity of mTORC1. Crypt depth and villus height were measured to score adaptation. Immunohistochemistry was used to investigate differentiation and rates of crypt proliferation. RESULTS: After SBR, mice treated with systemic rapamycin showed diminished structural adaptation, blunted crypt cell proliferation, and significant body weight loss. Activating i-mTORC1 via TSC1 deletion induced larger hyperproliferative crypts and disorganized Paneth cells without a significant change in villus height. After SBR, ablating TSC1 in intestinal epithelium induced a robust villus growth with much stronger crypt cell proliferation, but similar body weight recovery. Acute inactivation of i-mTORC1 through deletion of Raptor did not change crypt cell proliferation or mucosa structure, but significantly reduced lysozyme/matrix metalloproteinase-7-positive Paneth cell and goblet cell numbers, with increased enteroendocrine cells. Surprisingly, ablation of intestinal epithelial cell-specific Raptor after SBR did not affect adaptation or crypt proliferation, but dramatically reduced body weight recovery after surgery. CONCLUSIONS: Systemic, but not intestinal-specific, mTORC1 is important for normal adaptation responses to SBR. Although not required, forced enterocyte mTORC1 signaling after resection causes an enhanced adaptive response.

4.
J Pediatr Surg ; 52(6): 1026-1030, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28343662

RESUMO

PURPOSE: Previously, we demonstrated enhanced adaptation after small bowel resection (SBR) in intestinal-specific retinoblastoma (Rb)-deficient mice along with elevated levels of insulin-like growth factor 2 (IGF2) expression within the villi. The purpose of this study was to verify that the insulin-like growth factor 1 receptor (IGF1R) plays a role in this phenomenon. METHODS: Inducible and intestinal specific Rb and IGF1R double knockout mice (iRb/IGF1R-IKO) (n=4) and Rb single knockout mice (iRb-IKO) (n=5) underwent 50% mid SBR. On post-operative day 28, mice were harvested, and structural adaptation was measured as changes in crypt depth and villus height. Rates of enterocyte proliferation were recorded. IGF2 expression within the remnant villi was measured via RT-PCR. RESULTS: Both iRb-IKO and iRb/IGF1R-IKO mice demonstrated enhanced adaptation with at least a 45% increase in both crypt depth and villus height in the proximal and distal remnant bowel. Both groups showed elevation of IGF2 expression in the remnant villi, but there were no differences between the two groups. CONCLUSION: Epithelial IGF1R is dispensable for IGF2-mediated enhanced intestinal adaptation in retinoblastoma-deficient mice. Our findings suggest that IGF2 signals for enhanced adaptation in cells outside of the epithelium. Further investigation is needed to study the IGF2/IGF1R signaling interaction within the mesenchyme. LEVEL OF EVIDENCE: Animal study - not clinical.


Assuntos
Adaptação Fisiológica , Fator de Crescimento Insulin-Like II/metabolismo , Mucosa Intestinal/metabolismo , Intestino Delgado/cirurgia , Receptor IGF Tipo 1/metabolismo , Proteína do Retinoblastoma/deficiência , Síndrome do Intestino Curto/metabolismo , Animais , Biomarcadores/metabolismo , Mucosa Intestinal/fisiologia , Intestino Delgado/metabolismo , Intestino Delgado/fisiologia , Camundongos , Camundongos Knockout , Período Pós-Operatório , Reação em Cadeia da Polimerase em Tempo Real , Síndrome do Intestino Curto/fisiopatologia
5.
J Pediatr Surg ; 52(6): 1014-1019, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28351520

RESUMO

BACKGROUND: A significant number of children with short bowel syndrome experience intestinal failure-associated liver disease. We recently demonstrated accelerated hepatic steatosis after 50% small bowel resection (SBR) in mice. Since SBR is associated with alterations in the gut microbiome, the purpose of this study was to determine whether TLR4 signaling is critical to the development of resection-associated hepatic steatosis. METHODS: Male C57BL6 (control) and TLR4-knockout (KO) mice underwent 50% proximal SBR. Liver sections were analyzed to obtain the percent lipid content, and Ileal sections were assessed for morphological adaptation. Intestinal TLR4 mRNA expression was measured at 7days and 10weeks. RESULTS: Compared to controls, TLR4 KO mice demonstrated similar weight gain and morphological adaptation after SBR. Hepatic steatosis was decreased 32-fold in the absence of TLR4. Intestinal TLR4 mRNA expression was significantly elevated 7days after SBR. We also found that TLR4 expression in the intestine was 20-fold higher in whole bowel sections compared with isolated enterocytes. CONCLUSIONS: TLR4 signaling is critical for the development of resection-associated steatosis, but not involved in intestinal adaptation after massive SBR. Further studies are needed to delineate the mechanism for TLR4 signaling in the genesis of resection-associated liver injury. LEVEL OF EVIDENCE: Animal study, not clinical.


Assuntos
Fígado Gorduroso/etiologia , Intestino Delgado/cirurgia , Síndrome do Intestino Curto/complicações , Receptor 4 Toll-Like/metabolismo , Animais , Biomarcadores/metabolismo , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Intestino Delgado/metabolismo , Intestino Delgado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Síndrome do Intestino Curto/metabolismo , Síndrome do Intestino Curto/patologia
6.
Surgery ; 160(6): 1485-1495, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27592213

RESUMO

BACKGROUND: Intestinal failure-associated liver disease causes significant mortality in patients with short bowel syndrome. Steatosis, a major component of intestinal failure-associated liver disease has been shown to persist even after weaning from parenteral nutrition. We sought to determine whether steatosis occurs in our murine model of short bowel syndrome and whether steatosis was affected by manipulation of the intestinal microbiome. METHODS: Male C57BL6 mice underwent 50% small bowel resection and orogastric gavage with vancomycin or vehicle for 10 weeks. DNA was extracted from stool samples then sequenced using 16s rRNA. Liver lipid content was analyzed. Bile acids were measured in liver and stool. RESULTS: Compared with unoperated mice, small bowel resection resulted in significant changes in the fecal microbiome and was associated with a >25-fold increase in steatosis. Oral vancomycin profoundly altered the gut microbiome and was associated with a 15-fold reduction in hepatic lipid content after resection. There was a 17-fold reduction in fecal secondary bile acids after vancomycin treatment. CONCLUSION: Massive small bowel resection in mice is associated with development of steatosis and prevented by oral vancomycin. These findings implicate a critical role for gut bacteria in intestinal failure-associated liver disease pathogenesis and illuminate a novel, operative model for future investigation into this important morbidity.


Assuntos
Antibacterianos/uso terapêutico , Fígado Gorduroso/etiologia , Fígado Gorduroso/prevenção & controle , Síndrome do Intestino Curto/complicações , Vancomicina/uso terapêutico , Administração Oral , Animais , Modelos Animais de Doenças , Fezes/microbiologia , Humanos , Intestino Delgado/microbiologia , Intestino Delgado/cirurgia , Camundongos , Camundongos Endogâmicos C57BL , Síndrome do Intestino Curto/microbiologia
7.
J Pediatr Surg ; 50(6): 943-7, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25818318

RESUMO

PURPOSE: Intestinal adaptation structurally represents increases in crypt depth and villus height in response to small bowel resection (SBR). Previously, we found that neither epidermal growth factor receptor (EGFR) nor insulin-like growth factor 1 receptor (IGF1R) function was individually required for normal adaptation. In this study, we sought to determine the effect of disrupting both EGFR and IGF1R expression on resection-induced adaptation. METHODS: Intestinal-specific EGFR and IGF1R double knockout mice (EGFR/IGF1R-IKO) (n=6) and wild-type (WT) control mice (n=7) underwent 50% proximal SBR. On postoperative day (POD) 7, structural adaptation was scored by measuring crypt depth and villus height. Rates of crypt cell proliferation, apoptosis, and submucosal capillary density were also compared. RESULTS: After 50% SBR, normal adaptation occurred in both WT and EGFR/IGF1R-IKO. Rates of proliferation and apoptosis were no different between the two groups. The angiogenic response was less in the EGFR/IGF1R-IKO compared to WT mice. CONCLUSION: Disrupted expression of EGFR and IGF1R in the intestinal epithelial cells does not affect resection-induced structural adaptation but attenuates angiogenesis after SBR. These findings suggest that villus growth is driven by receptors and pathways that occur outside the epithelial cell component, while angiogenic responses may be influenced by epithelial-endothelial crosstalk.


Assuntos
Adaptação Fisiológica , Receptores ErbB/metabolismo , Intestino Delgado/fisiologia , Intestino Delgado/cirurgia , Receptor IGF Tipo 1/metabolismo , Animais , Apoptose , Proliferação de Células , Células Epiteliais/fisiologia , Mucosa Intestinal/metabolismo , Intestino Delgado/irrigação sanguínea , Camundongos Knockout , Neovascularização Fisiológica
8.
J Pediatr Surg ; 50(6): 948-53, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25818317

RESUMO

PURPOSE: Intestinal adaptation involves villus lengthening, crypt deepening, and increased capillary density following small bowel resection (SBR). Mice lacking the proangiogenic chemokine CXCL5 have normal structural adaptation but impaired angiogenesis. This work evaluates the impact of incomplete adaptive angiogenesis on the functional capacity of the intestine after SBR. METHODS: CXCL5 knockout (KO) and C57BL/6 wild-type (WT) mice underwent 50% SBR. Magnetic resonance imaging measured weekly body composition. Intestinal absorptive capacity was evaluated through fecal fat analysis. Gene expression profiles for select macronutrient transporters were measured via RT-PCR. Postoperative crypt and villus measurements were assessed for structural adaptation. Submucosal capillary density was measured through CD31 immunohistochemistry. RESULTS: Comparable postoperative weight gain occurred initially. Diminished weight gain, impaired fat absorption, and elevated steatorrhea occurred in KO mice after instituting high-fat diet. Greater postoperative upregulation of ABCA1 fat transporter occurred in WT mice, while PEPT1 protein transporter was significantly downregulated in KO mice. KO mice had impaired angiogenesis but intact structural adaptation. CONCLUSION: After SBR, KO mice display an inefficient intestinal absorption profile with perturbed macronutrient transporter expression, impaired fat absorption, and slower postoperative weight gain. In addition to longer villi and deeper crypts, an intact angiogenic response may be required to achieve functional adaptation to SBR.


Assuntos
Adaptação Fisiológica , Intestino Delgado/irrigação sanguínea , Intestino Delgado/cirurgia , Neovascularização Fisiológica , Animais , Quimiocina CXCL5 , Absorção Intestinal , Mucosa Intestinal/patologia , Intestino Delgado/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout
9.
J Gastrointest Surg ; 19(3): 451-7, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25519080

RESUMO

INTRODUCTION: Short bowel syndrome (SBS) is a morbid clinical condition that results from massive small-bowel resection (SBR). After SBR, there is a dramatic weight loss in the acute postoperative period. Our aim was to determine the impact of a high-protein diet (HPD) on weight gain and body composition in mice after SBR. METHODS: C57BL/6 mice underwent 50 % proximal SBR. Postoperatively, mice were randomly selected to receive standard rodent liquid diet (LD) (n = 6) or an isocaloric HPD (n = 9) for 28 days. Mice weights were recorded daily. Body composition analyses were obtained weekly. Student's t test was used for statistical comparisons with p < 0.05 considered significant. RESULTS: Mice that were fed HPD after SBR returned to baseline weight on average at postoperative day (POD) 8 versus mice that were fed LD that returned to baseline weight on average at POD 22. Total fat mass and lean mass were significantly greater by POD 14 within the HPD group. Both groups of mice demonstrated normal structural adaptation. CONCLUSION: HPD results in greater weight gain and improved body composition in mice after SBR. This finding may be clinically important for patients with SBS since improved weight gain may reduce the time needed for parenteral nutrition.


Assuntos
Proteínas Alimentares/administração & dosagem , Procedimentos Cirúrgicos do Sistema Digestório/efeitos adversos , Intestino Delgado/cirurgia , Síndrome do Intestino Curto/dietoterapia , Aumento de Peso , Adaptação Fisiológica , Animais , Composição Corporal , Camundongos , Camundongos Endogâmicos C57BL , Período Pós-Operatório , Distribuição Aleatória , Síndrome do Intestino Curto/etiologia
10.
J Gastrointest Surg ; 18(11): 1887-93, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25002022

RESUMO

Previously, we have demonstrated that genetically disrupting retinoblastoma protein (Rb) expression in enterocytes results in taller villi, mimicking resection-induced adaption responses. Rb deficiency also results in elevated insulin-like growth factor-2 (IGF-2) expression in villus enterocytes. We propose that postoperative disruption of Rb results in enhanced adaptation which is driven by IGF-2. Inducible, intestine-specific Rb-null mice (iRbIKO) and wild-type (WT) littermates underwent a 50% proximal small-bowel resection (SBR) at 7-9 weeks of age. They were then given tamoxifen on postoperative days (PODs) 4-6 and harvested on POD 28. The experiment was then repeated on double knockouts of both IGF-2 and Rb (IGF-2 null/iRbIKO). iRbIKO mice demonstrated enhanced resection-induced adaptive villus growth after SBR and increased IGF-2 messenger RNA (mRNA) in ileal villus enterocytes compared to their WT littermates. In the IGF-2 null/iRbIKO double-knockout mice, there was no additional villus growth beyond what was expected of normal resection-induced adaptation. Adult mice in which Rb is inducibly deleted from the intestinal epithelium following SBR have augmented adaptive growth. IGF-2 expression is necessary for enhanced adaptation associated with acute intestinal Rb deficiency.


Assuntos
Enterócitos/metabolismo , Fator de Crescimento Insulin-Like II/metabolismo , Intestino Delgado/cirurgia , Proteína do Retinoblastoma/metabolismo , Adaptação Fisiológica/genética , Animais , Apoptose , Western Blotting , Proliferação de Células , Modelos Animais de Doenças , Imuno-Histoquímica , Mucosa Intestinal/metabolismo , Intestino Delgado/patologia , Modelos Lineares , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/análise , Distribuição Aleatória , Reação em Cadeia da Polimerase em Tempo Real , Proteína do Retinoblastoma/genética , Tamoxifeno/farmacologia , Coleta de Tecidos e Órgãos
11.
J Pediatr Surg ; 49(6): 966-70; discussion 970, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24888844

RESUMO

PURPOSE: Enhanced structural features of resection-induced intestinal adaptation have been demonstrated following the administration of multiple different growth factors and peptides. Among these, the insulin-like growth factor (IGF) system has been considered to be significant. In this study, we employ mutant mouse strains to directly test the contribution of IGF2 and its enterocyte receptor (IGF1R) toward the adaptation response to massive small bowel resection (SBR). METHODS: IGF2-knockout (IGF2-KO) (n=8) and intestine specific IGF1R-knockout mice (IGF1R-IKO) (n=9) and their wild type (WT) littermates (n=5, n=7, respectively) underwent 50% proximal SBR. At post-operative day 7, structural adaptation was measured as crypt depth and villus height. Rates of enterocyte proliferation and apoptosis were also recorded. RESULTS: The successful deletion of IGF2 and IGF1R expression in the enterocytes was confirmed by RT-PCR and Western blot, respectively. Normal adaptation occurred in both IGF2-KO and IGF1R-IKO mice after 50% SBR. Post-operative rates of proliferation and apoptosis in both IGF2-KO and IGF1R-IKO mice were no different than their respective controls. CONCLUSION: IGF2 and functional IGF1R signaling in enterocytes are both dispensable for resection-induced adaptation responses. The mechanism for IGF-stimulation of intestinal adaptation may involve other ligands or cellular compartments within the intestine.


Assuntos
Adaptação Fisiológica , Regulação da Expressão Gênica , Fator de Crescimento Insulin-Like II/genética , Intestino Delgado/metabolismo , RNA/genética , Receptor IGF Tipo 1/genética , Síndrome do Intestino Curto/genética , Animais , Apoptose , Western Blotting , Proliferação de Células , Modelos Animais de Doenças , Enterócitos/metabolismo , Enterócitos/patologia , Fator de Crescimento Insulin-Like II/biossíntese , Mucosa Intestinal , Intestino Delgado/patologia , Intestino Delgado/cirurgia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor IGF Tipo 1/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Síndrome do Intestino Curto/metabolismo , Síndrome do Intestino Curto/patologia
12.
J Pediatr Surg ; 49(6): 976-80; discussion 980, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24888846

RESUMO

PURPOSE: Intestinal adaptation is the compensatory response to massive small bowel resection (SBR) and characterized by lengthening of villi and deepening of crypts, resulting in increased mucosal surface area. Previous studies have demonstrated increased villus capillary blood vessel density after SBR, suggesting a role for angiogenesis in the development of resection-induced adaptation. Since we have previously shown enhanced expression of the proangiogenic chemokine CXCL5 after SBR, the purpose of this study was to determine the effect of disrupted CXCL5 expression on intestinal adaptation. METHODS: CXCL5 knockout (KO) and C57BL/6 wild type (WT) mice were subjected to either a 50% proximal SBR or sham operation. Ileal tissue was harvested on postoperative day 7. To assess for adaptation, villus height and crypt depth were measured. Submucosal capillary density was measured by CD31 immunohistochemistry. RESULTS: Both CXCL5-KO and WT mice demonstrated normal structural features of adaptation. Submucosal capillary density increased in the WT but not in the KO mice following SBR. CONCLUSION: CXCL5 is required for increased intestinal angiogenesis during resection-induced adaptation. Since adaptive villus growth occurs despite impaired CXCL5 expression and enhanced angiogenesis, this suggests that the growth of new blood vessels is not needed for resection-induced mucosal surface area expansion following massive SBR.


Assuntos
Adaptação Fisiológica , Quimiocina CXCL5/genética , Regulação da Expressão Gênica , Intestino Delgado/irrigação sanguínea , Neovascularização Fisiológica/genética , RNA/genética , Síndrome do Intestino Curto/genética , Animais , Quimiocina CXCL5/biossíntese , Modelos Animais de Doenças , Feminino , Imuno-Histoquímica , Intestino Delgado/cirurgia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microcirculação , Reação em Cadeia da Polimerase em Tempo Real , Síndrome do Intestino Curto/patologia
13.
Surgery ; 156(2): 412-8, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24713095

RESUMO

BACKGROUND: After massive small bowel resection (SBR), a postoperative diet high in fat is associated with enhanced villus growth. The purpose of this study was to further elucidate the quantity and composition of enteral fat in structural and metabolic changes after SBR. METHODS: C57/Bl6 mice underwent a 50% proximal SBR. Mice were then randomized to receive a low-fat diet (12% kcal fat), medium-fat diet (44% kcal fat), or high-fat diet (HFD; 71% kcal fat) ad libitum. In a separate experiment, mice underwent 50% proximal SBR and then were randomized to liquid diets of 42% kcal of fat in which the fat was composed of menhaden oil, milk fat, or olive oil. After 2 weeks, mice underwent body composition analysis and the small intestine was harvested. RESULTS: Mice that ingested the greatest amount of enteral fat (HFD) had the greatest percent lean mass. When the effects of the different kinds of enteral fat were analyzed, mice that consumed menhaden oil had the greatest percent lean mass with the greatest overall retention of preoperative weight. CONCLUSION: These findings suggest that enteral fat enriched in omega-3 fatty acids may offer clinically relevant metabolic advantages for patients with short gut syndrome.


Assuntos
Composição Corporal , Dieta Hiperlipídica , Intestino Delgado/cirurgia , Animais , Modelos Animais de Doenças , Ingestão de Energia , Nutrição Enteral , Ácidos Graxos Ômega-3/administração & dosagem , Óleos de Peixe/administração & dosagem , Transportador de Glucose Tipo 2/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Complicações Pós-Operatórias/dietoterapia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Síndrome do Intestino Curto/dietoterapia , Síndrome do Intestino Curto/genética , Síndrome do Intestino Curto/patologia , Transportador 1 de Glucose-Sódio/genética , Regulação para Cima , Redução de Peso
14.
Am J Physiol Gastrointest Liver Physiol ; 306(10): G909-15, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24742992

RESUMO

We previously demonstrated increased villus height following genetic deletion, or knockout, of retinoblastoma protein (Rb) in the intestinal epithelium (Rb-IKO). Here we determined the functional consequences of augmented mucosal growth on intestinal fat absorption and following a 50% small bowel resection (SBR). Mice with constitutively disrupted Rb expression in the intestinal epithelium (Rb-IKO) along with their floxed (wild-type, WT) littermates were placed on a high-fat diet (HFD, 42% kcal fat) for 54 wk. Mice were weighed weekly, and fat absorption, indirect calorimetry, and MRI body composition were measured. Rb-IKO mice were also subjected to a 50% SBR, followed by HFD feeding for 33 wk. In separate experiments, we examined intestinal fat absorption in mice with conditional (tamoxifen-inducible) intestinal Rb (inducible Rb-IKO) deletion. Microarray revealed that the transcriptional expression of lipid absorption/transport genes was significantly reduced in constitutive Rb-IKO mice. These mice demonstrated greater mucosal surface area yet manifested paradoxically impaired intestinal long-chain triglyceride absorption and decreased cholesterol absorption. Despite attenuated lipid absorption, there were no differences in metabolic rate, body composition, and weight gain in Rb-IKO and WT mice at baseline and following SBR. We also confirmed fat malabsorption in inducible Rb-IKO mice. We concluded that, despite an expanded mucosal surface area, Rb-IKO mice demonstrate impaired lipid absorption without compensatory alterations in energy homeostasis or body composition. These findings underscore the importance of delineating structural/functional relationships in the gut and suggest a previously unknown role for Rb in the regulation of intestinal lipid absorption.


Assuntos
Absorção Intestinal , Mucosa Intestinal/metabolismo , Metabolismo dos Lipídeos/genética , Proteína do Retinoblastoma/genética , Animais , Dieta Hiperlipídica , Intestino Delgado/cirurgia , Camundongos , Camundongos Knockout , Triglicerídeos/metabolismo
15.
J Gastrointest Surg ; 18(2): 286-94; discussion 294, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24002772

RESUMO

Previous studies have shown that high-fat diet (HFD) enhances adaptation if provided immediately following small bowel resection (SBR). The purpose of this study was to determine if HFD could further enhance villus growth after resection-induced adaptation had already taken place. C57/Bl6 mice underwent a 50 % proximal SBR or sham operation and were then provided a standard rodent liquid diet (LD) ad lib. After a typical period of adaptation (7 days), SBR and sham-operated mice were randomized to receive either LD or HFD (42 % kcal fat) for an additional 7 days. Mice were then harvested, and small intestine was collected for analysis. Adaptation occurred in both SBR groups; however, the SBR/HFD had significantly increased villus height compared to SBR/LD. Reverse transcription-polymerase chain reaction of villus enterocytes showed a marked increase in CD36 expression in the SBR/HFD group compared with SBR/LD mice. While exposure to increased enteral fat alone did not affect villus morphology in sham-operated mice, HFD significantly increased villus growth in the setting of resection-induced adaptation, supporting the clinical utility of enteral fat in augmenting adaptation. Increased expression of CD36 suggests a possible mechanistic role in dietary fat metabolism and villus growth in the setting of short gut syndrome.


Assuntos
Adaptação Fisiológica , Dieta Hiperlipídica , Íleo/cirurgia , Mucosa Intestinal/anatomia & histologia , Jejuno/cirurgia , Animais , Apolipoproteínas B/genética , Composição Corporal , Antígenos CD36/análise , Antígenos CD36/genética , Proteínas de Transporte/genética , Proliferação de Células , Enterócitos/química , Enterócitos/metabolismo , Mucosa Intestinal/metabolismo , Masculino , Camundongos Endogâmicos C57BL , RNA Mensageiro/metabolismo
16.
J Pediatr Surg ; 48(6): 1330-9, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23845627

RESUMO

PURPOSE: Massive small bowel resection (SBR) results in an adaptive response within the remnant bowel. We have previously shown an immediate reduction in intestinal blood flow and oxygen saturation (sO2) after SBR. We therefore sought to determine the duration of resection-induced intestinal hypoxia and expression of hypoxia-inducible factors (HIFs) following SBR. METHODS: C57B6 mice were subjected to 50% proximal SBR or a sham procedure. Photoacoustic microscopy (PAM) was used to measure blood flow and sO2 on postoperative days (PODs) 1, 3, and 7. Ileal tissue was harvested 6h postoperatively and on PODs 1 and 2, and HIF1α, HIF2α, and VEGF mRNA expression were assessed via RT-PCR. A p value of less than 0.05 was considered significant. RESULTS: Following SBR, reduction in intestinal blood flow persists for 24h and is followed with hyperemia by POD 3. The immediate reduction in venous sO2 and increased tissue oxygen utilization continued through POD 7. Enhanced expression of HIF1α was demonstrated 6h following SBR. CONCLUSION: Massive SBR results in an immediate relative hypoxic state within the remnant bowel with early enhanced expression of HIF1α. On POD 7, increased tissue oxygen extraction and elevated blood flow persist in the adapting intestine.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Velocidade do Fluxo Sanguíneo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Hipóxia/etiologia , Intestino Delgado/cirurgia , Complicações Pós-Operatórias , Anastomose Cirúrgica , Animais , Biomarcadores/metabolismo , Regulação para Baixo , Hipóxia/metabolismo , Hipóxia/fisiopatologia , Intestino Delgado/irrigação sanguínea , Intestino Delgado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia , Oxigênio/metabolismo , Técnicas Fotoacústicas , Complicações Pós-Operatórias/metabolismo , Complicações Pós-Operatórias/fisiopatologia , Período Pós-Operatório , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/metabolismo
17.
J Pediatr Surg ; 48(6): 1340-7, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23845628

RESUMO

PURPOSE: We have previously demonstrated a hyperplastic phenotype when Rb expression was disrupted within the intestinal epithelium. These findings mimic resection-induced adaptation suggesting a possible mechanistic role for Rb during adaptation. The purpose of the present study was to elucidate a mechanism for how Rb deficiency induces intestinal hyperplasia. METHODS: Enterocytes isolated from intestine-specific Rb knockout mice (Rb-IKO) underwent a microarray to elucidate their gene expression profile. IGF2 expression was significantly elevated, which was subsequently confirmed by RT-PCR and in situ mRNA hybridization. Mice with deficient expression of IGF2 or its receptor IGF1R were therefore crossed with Rb-IKO mice to determine the significance of IGF2 in mediating the Rb-IKO intestinal phenotype. RESULTS: Expression of IGF2 was significantly elevated in villus enterocytes of Rb-IKO mice. The mucosal hyperplasia in Rb-IKO mice was reversed when either IGF2 or IGF1R expression was genetically disrupted in Rb-IKO mice. CONCLUSION: IGF-2 expression is significantly elevated in villus enterocytes and is required for the hyperplastic intestinal mucosal phenotype of Rb-IKO mice. The trophic effects of IGF2 require intact IGF1R signaling within the intestinal epithelium. These findings reveal novel regulatory roles for Rb in expanding intestinal mucosal surface area.


Assuntos
Fator de Crescimento Insulin-Like II/metabolismo , Mucosa Intestinal/patologia , Proteína do Retinoblastoma/deficiência , Animais , Biomarcadores/metabolismo , Western Blotting , Proliferação de Células , Enterócitos/metabolismo , Enterócitos/patologia , Perfilação da Expressão Gênica , Hiperplasia , Hibridização In Situ , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase em Tempo Real , Receptor IGF Tipo 1/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
J Pediatr Surg ; 47(9): 1748-53, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22974617

RESUMO

PURPOSE: Intestinal adaptation after massive small bowel resection (SBR) permits improved absorption of enteral nutrition despite significant loss of bowel length. Epidermal growth factor (EGF) and its receptor (EGFR) have previously been established to play major roles in the pathogenesis of adaptation. This study tested the hypothesis that EGFR signaling within the epithelial cell compartment (enterocytes) is required for intestinal adaptation. METHODS: We developed a tamoxifen-inducible Villin-Cre/LoxP recombinant system for enterocyte-directed EGFR deletion using EGFR-floxed mice. Epidermal growth factor receptor-null mice and wild-type littermates underwent either 50% proximal SBR or sham operation. Ileal tissue was harvested on postoperative day 7. To assess for adaptation, villus height and crypt depth as well as rates of crypt cell proliferation and apoptosis were measured. RESULTS: Adaptation after SBR occurred normally, as demonstrated by significant increases in villus height, crypt depth, and crypt proliferative and apoptotic index in both the wild-type and EGFR-null mice. CONCLUSION: Enterocyte EGFR expression is not required for the adaptation response to massive SBR. This novel finding suggests that enterocyte proliferation during adaptation is regulated by EGFR signaling in cells other than enterocytes, perhaps within the mesenchymal cell compartment of the bowel wall via factor(s) that are presently unknown.


Assuntos
Adaptação Fisiológica , Enterócitos/metabolismo , Receptores ErbB/metabolismo , Intestino Delgado/cirurgia , Animais , Biomarcadores/metabolismo , Western Blotting , Proliferação de Células , Enterócitos/fisiologia , Feminino , Íleo/metabolismo , Íleo/patologia , Íleo/fisiologia , Intestino Delgado/metabolismo , Intestino Delgado/patologia , Intestino Delgado/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
Surgery ; 152(2): 286-93, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22828150

RESUMO

BACKGROUND: p38-Alpha mitogen-activated protein kinase (p38-MAPK) is a tumor suppressor often mutated in human cancers, but its specific role in colorectal cancer is not completely understood. Previous studies have found that p38-MAPK activity inhibits epithelial proliferation and promotes apoptosis in the intestine. Therefore, we sought to test the hypothesis that intestinal disruption of p38-MAPK would lead to increased tumorigenesis in the colon. METHODS: p38-MAPK was deleted in mice within the intestinal epithelium using a tamoxifen-inducible Cre system under control of the villin promoter [villin-Cre ERT2(+), MAPK14(f/f)]. An azoxymethane and dextran sodium sulfate protocol was used to drive intestinal tumor development. Tumor measurements were made using computer software from photographs of excised colon specimens. RESULTS: The number of mice that developed tumors was not statistically different when comparing wild-type mice (7/14) to inducible, intestine epithelial-deleted p38-MAPK (9/11) mice after azoxymethane/dextran sodium sulfate treatment (P = .21). However, the epithelial-deleted p38-MAPK mice developed significantly more tumors (3.7 vs 1.1; P = .008) and nearly 4 times the total tumor burden as wild-type mice (17.4 vs 4.8 mm(2); P = .03). Wild-type and epithelial-deleted p38-MAPK groups demonstrated a similar degree of colon inflammation. CONCLUSION: Deletion of p38-MAPK within the colonic mucosa leads to a hyperplastic state promoting greater tumor development. Because the severity of colitis was not augmented in mice with p38-MAPK deficiency, tumor development is likely mediated by impaired cell cycle regulation within the colonic epithelium. Manipulation of p38-MAPK activity may provide a novel treatment and/or prevention strategy in the management of colorectal cancer, particularly in the setting of inflammatory bowel disease.


Assuntos
Adenoma/etiologia , Transformação Celular Neoplásica , Neoplasias do Colo/etiologia , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Adenoma/enzimologia , Adenoma/patologia , Animais , Azoximetano , Carcinógenos , Colite/induzido quimicamente , Colite/enzimologia , Colo/enzimologia , Colo/patologia , Neoplasias do Colo/enzimologia , Neoplasias do Colo/patologia , Sulfato de Dextrana , Enterócitos/enzimologia , Fezes , Mucosa Intestinal/enzimologia , Camundongos , Camundongos Knockout , Redução de Peso
20.
J Pediatr Surg ; 47(6): 1143-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22703784

RESUMO

PURPOSE: Massive small bowel resection (SBR) results in villus angiogenesis and a critical adaptation response within the remnant bowel. Previous ex vivo studies of intestinal blood flow after SBR are conflicting. We sought to determine the effect of SBR on intestinal hemodynamics using photoacoustic microscopy, a noninvasive, label-free, high-resolution in vivo hybrid imaging modality. METHODS: Photoacoustic microscopy was used to image the intestine microvascular system and measure blood flow and oxygen saturation (So(2)) of the terminal mesenteric arteriole and accompanying vein in C57BL6 mice (n = 7) before and immediately after a 50% proximal SBR. A P value of less than .05 was considered significant. RESULTS: Before SBR, arterial and venous So(2) were similar. Immediately after SBR, the venous So(2) decreased with an increase in the oxygen extraction fraction. In addition, the arterial and venous blood flow significantly decreased. CONCLUSION: Massive SBR results in an immediate reduction in intestinal blood flow and increase in tissue oxygen utilization. These physiologic changes are observed throughout the remnant small intestine. The contribution of these early hemodynamic alterations may contribute to the induction of villus angiogenesis and the pathogenesis of normal intestinal adaptation responses.


Assuntos
Íleo/cirurgia , Microcirculação , Microscopia/métodos , Oximetria/métodos , Oxigênio/análise , Técnicas Fotoacústicas , Síndrome do Intestino Curto/fisiopatologia , Circulação Esplâncnica , Adaptação Fisiológica , Animais , Arteríolas , Íleo/irrigação sanguínea , Íleo/química , Íleo/diagnóstico por imagem , Masculino , Veias Mesentéricas , Camundongos , Camundongos Endogâmicos C57BL , Microvasos/diagnóstico por imagem , Microvasos/ultraestrutura , Oximetria/instrumentação , Oxigênio/farmacocinética , Oxiemoglobinas/análise , Síndrome do Intestino Curto/etiologia , Ultrassonografia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA