1
|
Zorzetti N, D'Andrea V, Lauro A. Proteomic biomarkers in short bowel syndrome : are we ready to use them in clinical activity? Expert Rev Proteomics 2021; 18:285-293. [PMID: 33910424 DOI: 10.1080/14789450.2021.1924063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: Short bowel syndrome (SBS) is a clinical condition that can affect childhood and adult patients. Biomarker research is expected to be a new frontier in the clinical application, helpful for patients and health-care systems.Areas covered: SBS is usually a consequence of a massive intestinal resection that leads to an intestinal failure because of the reduction of absorptive surface, bacterial overgrowth, and faster intestinal transit. This new condition requires a multidisciplinary expertise to achieve again digestive autonomy. Parental nutrition (PN) supports nutritional status in SBS patients while the new guidelines on intestinal transplantation confirm its strict indication only for patients at actual risk of death on PN. A PubMed literature review from the 1980s up to date was performed, highlighting proteomic biomarkers and growth factor therapies that have shown so far promising results in SBS patients.Expert opinion: Apart from a few specific biomarkers and growth factors, the discovery of specific molecular events is currently under investigation of the proteomic analysis and could potentially represent fundamental, future changes in prevention, diagnosis, therapeutic management, and experimental practices in SBS.
Collapse
Affiliation(s)
- Noemi Zorzetti
- Department of General Surgery, "Ospedale Civile Umberto I", Lugo, Ravenna, Italy
| | - Vito D'Andrea
- Department of Surgical Sciences, La Sapienza University, Rome, Italy
| | - Augusto Lauro
- Department of Surgical Sciences, La Sapienza University, Rome, Italy
| |
Collapse
|
2
|
Sukhotnik I, Ben-Shahar Y, Pollak Y, Cohen S, Moran-Lev H, Koppelmann T, Gorenberg M. Intestinal dysmotility after bowel resection in rats is associated with decreased ghrelin and vimentin expression and loss of intestinal cells of Cajal. Am J Physiol Gastrointest Liver Physiol 2021; 320:G283-G294. [PMID: 33325807 PMCID: PMC8609566 DOI: 10.1152/ajpgi.00223.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
This study provides novel insight into the mechanisms of intestinal dysmotility following massive small bowel resection. We show that 2 wk after bowel resection in rats, impaired intestinal motility was associated with loss of interstitial cells of Cajal (ICC; downregulation of transmembrane member 16A (TMEM16A) and c-kit expression) as well as with decreased vimentin, desmin, and ghrelin levels. Impaired intestinal motility led to a decrease in final body weight, suggesting less effective nutrient absorption. The purpose of this study was to evaluate the mechanisms of intestinal motility in a rat model of short bowel syndrome (SBS). Rats were divided into three groups: Sham rats underwent bowel transection; SBS-NSI rats underwent a 75% bowel resection and presented with normal intestinal size (NSI) at euthanasia and hypermotility patterns; SBS-DYS showed dysmotile (DYS) enlarged intestine and inhibited motility patterns. Animals were euthanized after 2 wk. Illumina's digital gene expression (DGE) analysis was used to determine the intestinal motility-related gene expression profiling in mucosal samples. Intestinal motility-related and ICC genes and protein expression in intestinal muscle layer were determined using real-time PCR, Western blotting, and immunohistochemistry. Gastrointestinal tract motility was studied by microcomputer tomography. From 10 Ca2+ signaling pathway-related genes, six genes in jejunum and seven genes in ileum were downregulated in SBS vs. Sham animals. Downregulation of TMEM16A mRNA and protein was confirmed by real-time PCR. Rapid intestinal transit time in SBS-NSI rats correlated with a mild decrease in TMEM16A, c-kit, and vimentin mRNA and protein expression (vs/. Sham animals). SBS-DYS rats demonstrated enlarged intestinal loops and delayed small intestinal emptying (on imaging studies) that were correlated with marked downregulation in TMEM16A, c-kit, vimentin, and ghrelin mRNA and protein levels compared with the other two groups. In conclusion, 2 wk following massive bowel resection in rats, impaired intestinal motility was associated with decreased vimentin and ghrelin gene and protein levels as well as loss of ICC (c-kit and TMEM16A).NEW & NOTEWORTHY This study provides novel insight into the mechanisms of intestinal dysmotility following massive small bowel resection. We show that 2 weeks after bowel resection in rats, impaired intestinal motility was associated with loss of interstitial cells of Cajal (downregulation of TMEM 16A, and c-kit expression) as well as with decreased vimentin, desmin, and ghrelin levels. Impaired intestinal motility led to decrease in final body weight, suggesting less effective nutrient absorption.
Collapse
Affiliation(s)
- Igor Sukhotnik
- 1Laboratory of Intestinal Adaptation and Recovery, Department of Pediatric Surgery, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel,3Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yoav Ben-Shahar
- 1Laboratory of Intestinal Adaptation and Recovery, Department of Pediatric Surgery, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel,4The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Yulia Pollak
- 1Laboratory of Intestinal Adaptation and Recovery, Department of Pediatric Surgery, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Shlomi Cohen
- 2Pediatric Gastroenterology Unit, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel,3Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Hadar Moran-Lev
- 2Pediatric Gastroenterology Unit, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel,3Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tal Koppelmann
- 1Laboratory of Intestinal Adaptation and Recovery, Department of Pediatric Surgery, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Migel Gorenberg
- 4The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
3
|
Abstract
PURPOSE OF REVIEW Recent studies have evaluated intestinal physiology following bowel resection; understanding changes in small bowel physiology after intestinal transplantation has received less attention. In this review, we will examine recent studies focused on changes in intestinal physiology following resection and intestinal transplantation. RECENT FINDINGS Absorption, immunity, and motility are fundamental components of small bowel physiology. Absorption after resection or transplantation is mediated by adaptation and enterocyte function. After resection, adaptation results in increased villus height and crypt depth. Hepatocyte growth factor and epidermal growth factors cause enterocyte hypertrophy and hyperplasia, allowing greater peptide uptake. Little is known about intestinal adaptation after transplant, but enteral autonomy is attainable. Immunity in small bowel after transplantation relies on a balance of innate and adaptive immune responses in the presence of the luminal microbiota. Intraepithelial lymphocytes are decreased following small bowel resection. After small bowel transplant, the number and the ratio of intraepithelial lymphocytes to enterocytes, as well as changes in the microbiota, can be used to identify rejection. After intestinal transplant, immune-mediated dysmotility is common. Perioperative infliximab in addition to tacrolimus may decrease the inflammation that contributes to dysmotility. SUMMARY As intestinal transplantation becomes more successful, understanding how absorption, immunity, and motility changes will allow for optimization of bowel function.
Collapse
|
4
|
Wang W, Xiao W, Sun L, Zhang C, Chen G, Yang H. Inhibition of ACE activity contributes to the intestinal structural compensation in a massive intestinal resection rat model. Pediatr Surg Int 2012; 28:533-41. [PMID: 22441330 DOI: 10.1007/s00383-012-3075-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/07/2012] [Indexed: 01/27/2023]
Abstract
BACKGROUND Intestinal adaptation in short bowel syndrome (SBS) consists of increased epithelial cells (ECs) proliferation as well as apoptosis. Angiotensin-converting enzyme (ACE) has been shown to regulate ECs apoptosis. In this study, we investigated the effect of ACE inhibition on intestinal adaptation after small bowel resection (SBR) in a rat model. METHODS Sprague-Dawley rats were used and were divided into four groups: (1) Sham group received an ileum transection (n = 6); (2) Sham + ACE-I group received an ileum transaction and lavage with ACE inhibitor (ACE-I, enalaprilat, 2 mg/kg/day) (n = 6); (3) SBS group received a 70 % mid-intestinal resection (n = 6); (4) SBS + ACE-I group received a 70 % mid-intestinal resection and lavage with enalaprilat (2 mg/kg/day) (n = 6). Sampling was done 10 days after surgery. ECs apoptosis was studied by TUNEL staining. ACE, angiotensin II (ANGII) receptor type 1 (AT1R) and receptor type 2 (AT2R) expressions were detected with RT-PCR and immunofluorescent confocal microscopy. RESULTS SBR leads to significant intestinal hypertrophy. The addition of ACE-I to SBS rat resulted in a significant decline in ECs apoptosis. ACE mRNA expression was significantly elevated after SBS creation (0.24 ± 0.07 vs. 0.42 ± 0.11), and ACE-I administration further increased mucosal ACE mRNA expression (0.54 ± 0.12). Interestingly, AT1R mRNA expression showed a significant decline in the SBS group compared to Sham levels, and ACE-I administration increased AT1R mRNA expression to Sham levels. No significant difference in AT2R mRNA expression was found between Sham and SBS group. CONCLUSION These results offer further insight into the role of ACE on intestinal mucosal remolding after massive bowel resection. ACE-I may be beneficial to SBS patients via a reduction of the apoptotic rate, thus facilitating the degree of adaptation.
Collapse
Affiliation(s)
- Wensheng Wang
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | | | | | | | | | | |
Collapse
|
5
|
Thompson JS, Rochling FA, Weseman RA, Mercer DF. Current management of short bowel syndrome. Curr Probl Surg 2012; 49:52-115. [PMID: 22244264 DOI: 10.1067/j.cpsurg.2011.10.002] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Jon S Thompson
- University of Nebraska Medical Center, Omaha, Nebraska, USA
| | | | | | | |
Collapse
|
6
|
Wang D, Zhou X, She R, Xiong J, Sun Q, Peng K, Liu L, Liu Y. Impaired intestinal mucosal immunity in specific-pathogen-free chickens after infection with very virulent infectious bursal disease virus. Poult Sci 2009; 88:1623-8. [PMID: 19590077 DOI: 10.3382/ps.2009-00124] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Diarrhea is one of the most important clinical side effects that occurs in chickens after infection with bursal disease virus. However, the kinetics of the intestinal mucosal immune parameters in infectious bursal disease virus (IBDV)-infected birds remains unclear. Forty-five specific-pathogen-free chickens were randomly divided into 2 groups [30 chickens in the very virulent IBDV (vvIBDV)-infected group and 15 chickens in the control group] to determine the effect of vvIBDV infection on intestinal mucosal immunity. All chickens were raised in negative-pressure isolators and fed the same diet. Results indicate that severe histologic lesions were observed in the intestinal tract in the vvIBDV-infected group, and the villus heights of the duodenum, jejunum, and ileum were decreased after vvIBDV infection (P < 0.01 or P < 0.05). The number of intestinal intraepithelial lymphocytes, mast cells, and goblet cells decreased significantly in the vvIBDV-infected group compared with the control (P < 0.01 or P < 0.05) on d 1, 2, and 3 postchallenge with vvIBDV. Very virulent IBDV infection reduced the expression of each fragment of intestinal secretory IgA at all 3 time points. Intestinal alkaline phosphatase decreased dramatically on d 2 and 3 postchallenge with vvIBDV. Taken together, these results indicate that vvIBDV infection contributes to considerable damage to the normal structure of the intestine and intestinal mucosal immune parameters of specific-pathogen-free chickens. Our research helps to increase an understanding of the pathogenesis of diarrhea induced by vvIBDV infection. These findings provide new insights into the pathogenesis and pathophysiology of vvIBDV infection.
Collapse
Affiliation(s)
- D Wang
- College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| | | | | | | | | | | | | | | |
Collapse
|
7
|
Wang D, Ma W, She R, Sun Q, Liu Y, Hu Y, Liu L, Yang Y, Peng K. Effects of swine gut antimicrobial peptides on the intestinal mucosal immunity in specific-pathogen-free chickens. Poult Sci 2009; 88:967-74. [DOI: 10.3382/ps.2008-00533] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
8
|
Intestinal specific overexpression of interleukin-7 attenuates the alternation of intestinal intraepithelial lymphocytes after total parenteral nutrition administration. Ann Surg 2008; 248:849-56. [PMID: 18948814 DOI: 10.1097/sla.0b013e31818a1522] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Total parenteral nutrition (TPN), with the complete removal of enteral nutrition, results in marked changes in intestinal intraepithelial lymphocyte (IEL) function and phenotype. Previous work shows that TPN results in a loss of intestinal epithelial cell-derived interleukin-7 (IL-7), and this loss may play an important role in development of such TPN-associated IEL changes. METHODS To further understand this relation, we generated a transgenic mouse (IL-7), which overexpresses IL-7 specifically in intestinal epithelial cells. We hypothesized that this localized overexpression would attenuate many of the observed TPN-associated IEL changes. RESULTS Our study showed that TPN administration led to significant changes in IEL phenotype, including a marked decline in the CD8alphabeta+, CD4+, and alphabeta-TCR+ populations. IEL basal proliferation decreased 1.7-fold compared with wild-type TPN mice. TPN administration in wild-type mice resulted in several changes in IEL-derived cytokine expression. IL-7 mice given TPN, however, maintained IEL proliferation, and sustained normal IEL numbers and phenotype. CONCLUSIONS We conclude that specific intestinal IL-7 overexpression significantly attenuated many IEL changes in phenotype and function after TPN administration. These findings suggest a mechanism by which TPN results in observed IEL changes.
Collapse
|
9
|
Lee CH, Chen JY, Li ML, Chou MC, Lo HC. Oral Antibiotics Attenuate Bowel Segment Reversal–Induced Alterations in Subpopulation and Function of Peripheral Blood Leukocytes, Thymocytes, and Splenocytes in Massive Bowel-Resected Rats. JPEN J Parenter Enteral Nutr 2008; 33:90-101. [DOI: 10.1177/0148607108322397] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Chien-Hsing Lee
- From the Division of Pediatric Surgery, Department of Surgery, Changhua Christian Hospital, Changhua, Taiwan; Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Medical Education and Research, Changhua Christian Hospital, Changhua, Taiwan; Department of Nutritional Science, Fu Jen Catholic University, Hsin Chuang City, Taipei, Taiwan
| | - Jing-Yi Chen
- From the Division of Pediatric Surgery, Department of Surgery, Changhua Christian Hospital, Changhua, Taiwan; Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Medical Education and Research, Changhua Christian Hospital, Changhua, Taiwan; Department of Nutritional Science, Fu Jen Catholic University, Hsin Chuang City, Taipei, Taiwan
| | - Mei-Ling Li
- From the Division of Pediatric Surgery, Department of Surgery, Changhua Christian Hospital, Changhua, Taiwan; Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Medical Education and Research, Changhua Christian Hospital, Changhua, Taiwan; Department of Nutritional Science, Fu Jen Catholic University, Hsin Chuang City, Taipei, Taiwan
| | - Ming-Chih Chou
- From the Division of Pediatric Surgery, Department of Surgery, Changhua Christian Hospital, Changhua, Taiwan; Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Medical Education and Research, Changhua Christian Hospital, Changhua, Taiwan; Department of Nutritional Science, Fu Jen Catholic University, Hsin Chuang City, Taipei, Taiwan
| | - Hui-Chen Lo
- From the Division of Pediatric Surgery, Department of Surgery, Changhua Christian Hospital, Changhua, Taiwan; Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Medical Education and Research, Changhua Christian Hospital, Changhua, Taiwan; Department of Nutritional Science, Fu Jen Catholic University, Hsin Chuang City, Taipei, Taiwan
| |
Collapse
|
10
|
Haxhija EQ, Yang H, Spencer AU, Koga H, Sun X, Teitelbaum DH. Modulation of mouse intestinal epithelial cell turnover in the absence of angiotensin converting enzyme. Am J Physiol Gastrointest Liver Physiol 2008; 295:G88-G98. [PMID: 18483182 PMCID: PMC2494725 DOI: 10.1152/ajpgi.00589.2007] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Angiotensin converting enzyme (ACE) has been shown to be involved in regulation of apoptosis in nonintestinal tissues. This study examined the role of ACE in the modulation of intestinal adaptation utilizing ACE knockout mice (ACE-/-). A 60% small bowel resection (SBR) was used, since this model results in a significant increase in intestinal epithelial cell (EC) apoptosis as well as proliferation. Baseline villus height, crypt depth, and intestinal EC proliferation were higher, and EC apoptosis rates were lower in ACE-/- compared with ACE+/+ mice. After SBR, EC apoptosis rates remained significantly lower in ACE-/- compared with ACE+/+ mice. Furthermore, villus height and crypt depth after SBR continued to be higher in ACE-/- mice. The finding of a lower bax-to-bcl-2 protein ratio in ACE-/- mice may account for reduced EC apoptotic rates after SBR in ACE-/- compared with ACE+/+ mice. The baseline higher rate of EC proliferation in ACE-/- compared with ACE+/+ mice may be due to an increase in the expression of several EC growth factor receptors. In conclusion, ACE appears to have an important role in the modulation of intestinal EC apoptosis and proliferation and suggests that the presence of ACE in the intestinal epithelium has a critical role in guiding epithelial cell adaptive response.
Collapse
Affiliation(s)
- Emir Q. Haxhija
- Section of Pediatric Surgery, Department of Surgery, University of Michigan Medical School, and C. S. Mott Children's Hospital, Ann Arbor, Michigan; and Department of Pediatric Surgery, Medical University Graz, Graz, Austria
| | - Hua Yang
- Section of Pediatric Surgery, Department of Surgery, University of Michigan Medical School, and C. S. Mott Children's Hospital, Ann Arbor, Michigan; and Department of Pediatric Surgery, Medical University Graz, Graz, Austria
| | - Ariel U. Spencer
- Section of Pediatric Surgery, Department of Surgery, University of Michigan Medical School, and C. S. Mott Children's Hospital, Ann Arbor, Michigan; and Department of Pediatric Surgery, Medical University Graz, Graz, Austria
| | - Hiroyuki Koga
- Section of Pediatric Surgery, Department of Surgery, University of Michigan Medical School, and C. S. Mott Children's Hospital, Ann Arbor, Michigan; and Department of Pediatric Surgery, Medical University Graz, Graz, Austria
| | - Xiaoyi Sun
- Section of Pediatric Surgery, Department of Surgery, University of Michigan Medical School, and C. S. Mott Children's Hospital, Ann Arbor, Michigan; and Department of Pediatric Surgery, Medical University Graz, Graz, Austria
| | - Daniel H. Teitelbaum
- Section of Pediatric Surgery, Department of Surgery, University of Michigan Medical School, and C. S. Mott Children's Hospital, Ann Arbor, Michigan; and Department of Pediatric Surgery, Medical University Graz, Graz, Austria
| |
Collapse
|
11
|
Wang J, Zhang H, Ma H, Lu B, Wang J, Li Y, Li J. Inhibitory effect of dietary n-3 polyunsaturated fatty acids to intestinal IL-15 expression is associated with reduction of TCRαβ+CD8α+CD8β− intestinal intraepithelial lymphocytes. J Nutr Biochem 2008; 19:475-81. [PMID: 17855067 DOI: 10.1016/j.jnutbio.2007.06.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2007] [Revised: 05/26/2007] [Accepted: 06/11/2007] [Indexed: 11/16/2022]
Abstract
Intestinal intraepithelial lymphocytes (IELs) and their cytokines play an important role in the regulation of gut immune response and take part in gut immune barrier function. n-3 polyunsaturated fatty acid (PUFA) is an immunoregulator that has been shown to influence the process of gut inflammation. Interleukin (IL)-15 is a T-cell growth factor that has been shown to influence the differentiation of IEL. The aim of this study was to analyze the effects of dietary n-3 PUFA on IEL. IEL phenotype and cytokine (TNF-alpha, IFN-gamma, IL-4, IL-10 and TGF-beta1) profile were measured by FACS and real-time RT-PCR in healthy adult rats fed with fish oil diet for 90 days. Rats fed with corn oil diet served as controls. Intestinal IL-15 expression was measured by immunohistochemistry and real-time RT-PCR. The results demonstrated a decrease of intestinal IL-15 expression in the fish oil group. Associated with this deduction, n-3 PUFA significantly decreased the proportion of TCRalphabeta+CD8alpha+CD8beta- cells and IEL-derived TNF-alpha, IFN-gamma, IL-4 and IL-10. In conclusion, n-3 PUFA could inhibit intestinal mucosal expression of IL-15 and may influence phenotype and function of IEL through this mechanism.
Collapse
Affiliation(s)
- Jian Wang
- Department of Surgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 21002, China
| | | | | | | | | | | | | |
Collapse
|
12
|
Liu T, She R, Wang K, Bao H, Zhang Y, Luo D, Hu Y, Ding Y, Wang D, Peng K. Effects of rabbit sacculus rotundus antimicrobial peptides on the intestinal mucosal immunity in chickens. Poult Sci 2008; 87:250-4. [PMID: 18212367 DOI: 10.3382/ps.2007-00353] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Ninety chickens were randomly divided into 2 groups (45 chickens in each group) to determine the effect of oral administration of rabbit sacculus rotundus antimicrobial peptides (RSRP) on the intestinal mucosal immune responses in chicken. On d 7, 14, 21, and 28, the animals received 0.1 mg of RSRP dissolved in 0.5 mL of physiological saline. The control groups received the same dose of physiological salt solution on the same day. The results showed that RSRP increased the villus height of the duodenum (P < 0.01) and jejunum (P < 0.05) at the ages of 28, 42, and 56 d. The numbers of intestinal intraepithelial lymphocytes in different parts of intestine of the RSRP group were increased significantly more than that of the control (P < 0.01 or P < 0.05) at the ages of 28, 42, and 56 d. The RSRP increased the area of IgA-secreting cells of each fragment of intestine at all 3 time points. These results indicated that the presence of RSRP affected and considerably modified the structure of the intestine and mucosal immune parameters in healthy chickens when compared with controls.
Collapse
Affiliation(s)
- T Liu
- College Of Veterinary Medicine, China Agricultural University, Beijing, 100094 China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Wildhaber BE, Yang H, Haxhija EQ, Spencer AU, Teitelbaum DH. Intestinal intraepithelial lymphocyte derived angiotensin converting enzyme modulates epithelial cell apoptosis. Apoptosis 2008; 10:1305-15. [PMID: 16215678 PMCID: PMC1351359 DOI: 10.1007/s10495-005-2138-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Intestinal adaptation in short bowel syndrome (SBS) consists of increased epithelial cell (EC) proliferation as well as apoptosis. Previous microarray analyses of intraepithelial lymphocytes (IEL) gene expression after SBS showed an increased expression of angiotensin converting enzyme (ACE). Because ACE has been shown to promote alveolar EC apoptosis, we examined if IEL-derived ACE plays a role in intestinal EC apoptosis. METHODS Mice underwent either a 70% mid-intestinal resection (SBS group) or a transection (Sham group) and were studied at 7 days. ACE expression was measured, and ACE inhibition (ACE-I, enalaprilat) was used to assess ACE function. RESULTS IEL-derived ACE was significantly elevated in SBS mice. The addition of an ACE-I to SBS mice resulted in a significant decline in EC apoptosis. To address a possible mechanism, tumor necrosis factor alpha (TNF-alpha) mRNA expression was measured. TNF-alpha was significantly increased in SBS mice, and decreased with ACE-I. Interestingly, ACE-I was not able to decrease EC apoptosis in TNF-alpha knockout mice. CONCLUSIONS This study shows a previously undescribed expression of ACE by IEL. SBS was associated with an increase in IEL-derived ACE. ACE appears to be associated with an up-regulation of intestinal EC apoptosis. ACE-I significantly decreased EC apoptosis.
Collapse
Affiliation(s)
| | | | | | | | - Daniel H. Teitelbaum
- Correspondence to: Daniel H. Teitelbaum, MD, Section of Pediatric Surgery, University of Michigan Hospitals, Mott F3970, Box 0245, Ann Arbor, MI, 48109, USA, Phone: 734 764 4151, fax :734 936 9784 e-mail:
| |
Collapse
|
14
|
Hise M, Compher C, Brown J. Inflammatory Mediators and Home Parenteral Nutrition. Nutr Clin Pract 2008; 23:42-8. [DOI: 10.1177/011542650802300142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Affiliation(s)
- Mary Hise
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, Kansas; University of Pennsylvania, School of Nursing, Philadelphia, Pennsylvania; and the Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas
| | - Charlene Compher
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, Kansas; University of Pennsylvania, School of Nursing, Philadelphia, Pennsylvania; and the Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas
| | - John Brown
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, Kansas; University of Pennsylvania, School of Nursing, Philadelphia, Pennsylvania; and the Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas
| |
Collapse
|
15
|
Haxhija EQ, Yang H, Spencer AU, Sun X, Teitelbaum DH. Intestinal epithelial cell proliferation is dependent on the site of massive small bowel resection. Pediatr Surg Int 2007; 23:379-90. [PMID: 17205293 DOI: 10.1007/s00383-006-1855-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Early intestinal adaptation after massive small bowel resection (SBR) is driven by increased epithelial cell (EC) proliferation. There is a clear clinical difference in the post-operative course of patients after the loss of proximal (P) compared to distal (D) small bowel. This study examined the effects of the site of SBR on post-resectional intestinal adaptation, and investigated the potential mechanisms involved. C57BL/6J mice (n = 7/group) underwent: (1) 60% P-SBR, (2) 60% D-SBR, (3) 60% mid (M)-SBR and (4) SHAM-operation (transection/reanastomosis). Mice were sacrificed at 7 days after surgery and ECs and adjacent mucosal lymphocytes (IELs) isolated. Adaptation was assessed in both jejunum and ileum by quantification of villus height, crypt depth, villus cell size, crypt cell size (microns), goblet cell number, and EC proliferation (%BrdU incorporation). Proliferation signalling pathways including keratinocyte growth factor (KGF)/KGFR(1), IL-7/IL-7R, and epidermal growth factor receptor (EGFR) were measured by RT-PCR. Expression of IL-7 was further analysed by immunofluorescence. Data were analyzed using ANOVA. All three SBR models led to significant increases in villus height, crypt depth, goblet cell numbers and EC proliferation rate when compared to respective SHAM groups. The strongest morphometric changes were found for jejunal segments after M-SBR and for ileal segments after P-SBR. Furthermore, morphometric analysis showed that at 1-week post-resection a tremendous increase in EC numbers occurred in jejunal villi (cell hyperplasia), whereas a significant increase in EC size predominated in ileal villi (cell hypertrophy). mRNA expression of KGF, KGFR(1), IL-7R, and EGFR showed a significant increase only after D-SBR, whereas IL-7 increased significantly after SBR in all investigated models, and this was confirmed by immunofluorescence studies. Early intestinal adaptation shows distinct differences depending on the site of SBR, and is predominately driven by cell hyperplasia in jejunal villi and cell hypertrophy in ileal villi. However, the exact mechanisms, which guide these signalling pathways are still unclear.
Collapse
Affiliation(s)
- Emir Q Haxhija
- Department of Pediatric Surgery, Medical University Graz, Graz, Austria
| | | | | | | | | |
Collapse
|
16
|
Olio DD, Gupte G, Sharif K, Murphy MS, Lloyd C, McKiernan PJ, Kelly DA, Beath SV. Immunosuppression in infants with short bowel syndrome undergoing isolated liver transplantation. Pediatr Transplant 2006; 10:677-81. [PMID: 16911490 DOI: 10.1111/j.1399-3046.2006.00504.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Little data exist on immunosuppressive drug absorption in children with short bowel syndrome and intestinal failure associated liver disease (SBS-IFALD). AIM To evaluate the absorption of immunosuppressive medications in children with SBS-IFALD undergoing isolated liver transplantation (iLTx). METHODS A retrospective review was performed in children with SBS-IFALD undergoing LTx and comparison made with weight, age-matched children undergoing iLTX (extra-hepatic biliary atresia (EHBA) and normal intestinal length and function). RESULTS Seven children with SBS-IFALD undergoing iLTx (median residual bowel length, 60 cm, range 40-80) were compared with 15 children undergoing LTx for EHBA. SBS-IFALD children had significantly lower trough tacrolimus levels at three months (5.8 vs. 7.9 ng/mL, p<0.05) and six months (5.0 vs. 8.0 ng/mL, p<0.05), but equivalent levels at 12 months after iLTx. The median calculated dose-normalized concentrations indicated that systemic availability of tacrolimus was comparable in two groups at 3, 6, 12 months (33.1 vs. 23.3; 42.4 vs. 36; 51 vs. 52.9) despite the differences in enteral function. The incidence of acute rejection was 1/7 (SBS-IFALD) and 10/15 (EHBA) group (p = 0.06). CONCLUSION Children with SBS-IFALD demonstrated adequate absorption of oral tacrolimus without significant acute rejection rate after iLTx suggesting that modification of immunosuppression is not necessary.
Collapse
Affiliation(s)
- Dominic Dell Olio
- The Liver Unit, Birmingham Children's Hospital (BCH), Birmingham, UK.
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Haxhija EQ, Yang H, Spencer AU, Sun X, Teitelbaum DH. Influence of the site of small bowel resection on intestinal epithelial cell apoptosis. Pediatr Surg Int 2006; 22:37-42. [PMID: 16307277 PMCID: PMC1509096 DOI: 10.1007/s00383-005-1576-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Massive small bowel resection (SBR) results in a significant increase in intestinal epithelial cell (EC) proliferation as well as apoptosis. Because the site of SBR (proximal (P) vs. distal (D)) affects the degree of intestinal adaptation, we hypothesized that different rates of EC apoptosis would also be found between P-SBR and D-SBR models. Wild-type C57BL/6J mice underwent: (1) 60% P-SBR, (2) 60% D-SBR, or (3) SHAM-operation (transaction-reanastomosis) at the mid-gut point. Mice were sacrificed after 7 days. EC apoptosis was measured by TUNEL staining. EC-related apoptotic gene expression including intrinsic and extrinsic pathways was measured with reverse transcriptase-polymerase chain reaction. Bcl-2 and bax protein expression were analyzed by Western immunoblotting. Both models of SBR led to significant increases in villus height and crypt depth; however, the morphologic adaptation was significantly higher after P-SBR compared to D-SBR (P<0.01). Both models of SBR led to significant increases in enterocyte apoptotic rates compared to respective sham levels; however, apoptotic rates were 2.5-fold higher in ileal compared to jejunal segments (P<0.01). P-SBR led to significant increases in bax (pro-apoptotic) and Fas expression, whereas D-SBR resulted in a significant increase in TNF-alpha expression (P<0.01). EC apoptosis seems to be an important component of intestinal adaptation. The significant difference in EC apoptotic rates between proximal and distal intestinal segments appeared to be due to utilization of different mechanisms of action.
Collapse
Affiliation(s)
- Emir Q. Haxhija
- Department of Pediatric Surgery, Medical University Graz, Graz, Austria
| | - Hua Yang
- Section of Pediatric Surgery, Department of Surgery, University of Michigan Medical School, and C. S. Mott Children’s Hospital, Ann Arbor, MI 48109
| | - Ariel U. Spencer
- Section of Pediatric Surgery, Department of Surgery, University of Michigan Medical School, and C. S. Mott Children’s Hospital, Ann Arbor, MI 48109
| | - Xiaoyi Sun
- Section of Pediatric Surgery, Department of Surgery, University of Michigan Medical School, and C. S. Mott Children’s Hospital, Ann Arbor, MI 48109
| | - Daniel H. Teitelbaum
- Section of Pediatric Surgery, Department of Surgery, University of Michigan Medical School, and C. S. Mott Children’s Hospital, Ann Arbor, MI 48109
| |
Collapse
|
18
|
Yang H, Spencer AU, Teitelbaum DH. Interleukin-7 administration alters intestinal intraepithelial lymphocyte phenotype and function in vivo. Cytokine 2005; 31:419-28. [PMID: 16102972 DOI: 10.1016/j.cyto.2005.06.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2004] [Revised: 06/01/2005] [Accepted: 06/21/2005] [Indexed: 01/05/2023]
Abstract
BACKGROUND Interleukin-7 (IL-7) plays a crucial role in controlling T-cell development and homeostasis. IL-7 knock out and IL-7 receptor knock out mice show distinct declines in absolute numbers of the intestinal intraepithelial lymphocytes (IEL). Therefore, we hypothesized that exogenous administration of IL-7 would alter IEL phenotype and function. METHODS Adult C57BL/6J mice were treated with IL-7 or saline. Mice were euthanized at day 7. Cytokine and keratinocyte growth factor (KGF) expressions were measured with RT-PCR. IEL phenotype was studied with flow cytometry. Finally, to address the association of endogenous epithelial cell (EC)-derived IL-7 and IEL, confocal microscopy was used to observe co-localization of IL-7 to IEL subpopulations. RESULTS IL-7 administration significantly increased IEL numbers. CD8alphabeta+ IEL increased 3.2-fold, CD8+CD44+ IEL increased 1.3-fold, and alphabeta-T-cell receptor (TCR)+ IEL increased 1.3-fold. IL-7 administration also significantly changed both alphabeta-TCR+ IEL- and gammadelta-TCR+ IEL-derived cytokine expressions. Interestingly, IL-7 administration also led to a significant increase in KGF expression. Confocal microscopy showed a high level of co-localization between the alphabeta-TCR+ IEL and EC-derived IL-7. gammadelta-TCR+ IEL showed a lower level, but still significant, co-localization. CONCLUSION IL-7 administration significantly affected IEL phenotype and function. The observed co-localization suggests that there is a close IEL-EC cross-communication mediated by EC-derived IL-7 expression.
Collapse
Affiliation(s)
- Hua Yang
- Section of Pediatric Surgery, Department of Surgery, The University of Michigan Medical School, Ann Arbor, 48109, USA
| | | | | |
Collapse
|