Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Pediatr Surg ; 47(9): 1748-53, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22974617

ABSTRACT

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.


Subject(s)
Adaptation, Physiological , Enterocytes/metabolism , ErbB Receptors/metabolism , Intestine, Small/surgery , Animals , Biomarkers/metabolism , Blotting, Western , Cell Proliferation , Enterocytes/physiology , Female , Ileum/metabolism , Ileum/pathology , Ileum/physiology , Intestine, Small/metabolism , Intestine, Small/pathology , Intestine, Small/physiology , Male , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction
2.
Am J Physiol Gastrointest Liver Physiol ; 302(9): G997-1005, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22383494

ABSTRACT

Increased apoptosis in crypt enterocytes is a key feature of intestinal adaptation following massive small bowel resection (SBR). Expression of the proapoptotic factor Bax has been shown to be required for resection-induced apoptosis. It has also been demonstrated that p38-α MAPK (p38) is necessary for Bax activation and apoptosis in vitro. The present studies were designed to test the hypothesis that p38 is a key regulator of Bax activation during adaptation after SBR in vivo. Enterocyte expression of p38 was deleted by tamoxifen administration to activate villin-Cre in adult mice with a floxed Mapk14 (p38-α) gene. Proximal 50% SBR or sham operations were performed on wild-type (WT) and p38 intestinal knockout (p38-IKO) mice under isoflurane anesthesia. Mice were killed 3 or 7 days after operation, and adaptation was analyzed by measuring intestinal morphology, proliferation, and apoptosis. Bax activity was quantified by immunoprecipitation, followed by Western blotting. After SBR, p38-IKO mice had deeper crypts, longer villi, and accelerated proliferation compared with WT controls. Rates of crypt apoptosis were significantly lower in p38-IKO mice, both at baseline and after SBR. Levels of activated Bax were twofold higher in WT mice after SBR relative to sham. In contrast, activated Bax levels were reduced by 67% in mice after p38 MAPK deletion. Deleted p38 expression within the intestinal epithelium leads to enhanced adaptation and reduced levels of enterocyte apoptosis after massive intestinal resection. p38-regulated Bax activation appears to be an important mechanism underlying resection-induced apoptosis.


Subject(s)
Apoptosis/physiology , Enterocytes/cytology , Enterocytes/physiology , Intestines/physiopathology , Short Bowel Syndrome/physiopathology , bcl-2-Associated X Protein/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cells, Cultured , Intestines/pathology , Intestines/surgery , Mice , Mice, Knockout , Short Bowel Syndrome/pathology
3.
J Pediatr Surg ; 45(6): 1274-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20620331

ABSTRACT

PURPOSE: The magnitude of intestinal adaptation is considered to correlate with the extent of small bowel resection (SBR). However, this association has never been tested in mice. We sought to test the hypothesis that a greater SBR will induce a greater adaptation response. METHODS: C57/B6 mice underwent 50% SBR, 75% SBR, or sham operation and were killed on postoperative day 7. The magnitude of adaptation was compared between 50% SBR and 75% SBR as changes in villus height, crypt depth, as well as rates of apoptosis and proliferation. RESULTS: Seventy-five percent SBR led to decreased survival and increased weight loss compared with 50% SBR. The remnant ileum of both 50% SBR and 75% SBR displayed similar crypt expansion, enhanced villi, and increased apoptotic indices. Proliferation rates increased after 50% and 75% SBR equally. CONCLUSION: Models of resection greater than 50% in mice result in greater morbidity and mortality and do not magnify the adaptation response to massive SBR. The use of more extreme resection models does not appear to provide added benefit for investigating mechanisms of intestinal adaptation.


Subject(s)
Adaptation, Physiological/physiology , Intestine, Small/surgery , Short Bowel Syndrome/pathology , Animals , Apoptosis , Cell Proliferation , Disease Models, Animal , Intestine, Small/pathology , Male , Mice , Mice, Inbred C57BL , Prognosis , Short Bowel Syndrome/metabolism
4.
Surgery ; 148(2): 364-70, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20471049

ABSTRACT

BACKGROUND: Adaptive villus growth after a massive small bowel resection (SBR) is an important response to the loss of intestinal surface area and is regulated via epidermal growth factor receptor (EGFR) signaling. Increased levels of the proangiogenic chemokine ligand 5 (CXCL5) have been found within the adapting bowel in which angiogenesis is increased. We sought to determine whether CXCL5 was expressed specifically in the villus mesenchymal zone (area of increased blood vessel growth) and whether this expression was affected by EGF. METHODS: C57BL/6J mice were subjected to sham operation (bowel transaction with reanastomosis) or 50% proximal SBR. The remnant intestine was harvested, and the villus lamina propria was isolated by laser capture microdissection. The expression of CXCL5 messenger RNA (mRNA) was analyzed using real-time polymerase chain reaction (RT-PCR). Furthermore, CXCL5 mRNA levels were determined in EGF-stimulated human umbilical vein endothelial cells (HUVECs). RESULTS: A 2.39-fold increase (P < .05) in CXCL5 mRNA occurred in the lamina propria after SBR. In addition, villus height was found to be related directly to the degree of CXCL5 mRNA (R(2) = 0.97) expression. HUVECs treated with EGF demonstrated a 9-fold increase in CXCL5 mRNA expression. CONCLUSION: The villus growth observed in resection-induced adaptation is associated with increased expression of the chemokine CXCL5 within the lamina propria. Because EGF enhances CXCL5 expression directly in endothelial cells, EGFR-directed proangiogenic gene expression may be a critical mechanism for adaptive ileal villus growth.


Subject(s)
Chemokine CXCL5/metabolism , ErbB Receptors/metabolism , Intestine, Small/blood supply , Intestine, Small/surgery , Neovascularization, Physiologic , Adaptation, Physiological , Animals , Cells, Cultured , Chemokine CXCL5/genetics , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Gene Expression , Humans , Intestinal Mucosa/blood supply , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Male , Mesoderm/blood supply , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
5.
Semin Pediatr Surg ; 19(1): 35-43, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20123272

ABSTRACT

The structural and functional changes during intestinal adaptation are necessary to compensate for the sudden loss of digestive and absorptive capacity after massive intestinal resection. When the adaptive response is inadequate, short bowel syndrome (SBS) ensues and patients are left with the requirement for parenteral nutrition and its associated morbidities. Several hormones have been studied as potential enhancers of the adaptation process. The effects of growth hormone, insulin-like growth factor-1, epidermal growth factor, and glucagon-like peptide 2 on adaptation have been studied extensively in animal models. In addition, growth hormone and glucagon-like peptide 2 have shown promise for the treatment of SBS in clinical trials in human beings. Several lesser studied hormones, including leptin, corticosteroids, thyroxine, testosterone, and estradiol, are also discussed.


Subject(s)
Adaptation, Physiological , Intercellular Signaling Peptides and Proteins/therapeutic use , Intestine, Small/drug effects , Peptide Hormones/therapeutic use , Short Bowel Syndrome/drug therapy , Animals , Cell Proliferation/drug effects , Enterocytes/drug effects , Epidermal Growth Factor/pharmacology , Epidermal Growth Factor/therapeutic use , Glucagon-Like Peptide 2/pharmacology , Glucagon-Like Peptide 2/therapeutic use , Growth Hormone/pharmacology , Growth Hormone/therapeutic use , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Intestine, Small/cytology , Intestine, Small/growth & development , Peptide Hormones/pharmacology , Rats , Short Bowel Syndrome/pathology , Somatomedins/pharmacology , Somatomedins/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...