1
|
Liu ZM, Zhang YN, Zhang HF, Zhang XY. Combined PD-1 and IL-10 blockade reinvigorates mucosal CD8 +T exhaustion and relieves liver damage after intestinal ischemia reperfusion attack. Biochem Biophys Res Commun 2025; 742:151137. [PMID: 39647455 DOI: 10.1016/j.bbrc.2024.151137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/27/2024] [Accepted: 12/04/2024] [Indexed: 12/10/2024]
Abstract
Currently, impairments in gut mucosal immunity following intestinal ischemia reperfusion (IR) remain unclear. Mucosal CD8+T cells are critical for host defense against bacterial translocation from the gut lumen, and exhausted T cells lose robust effector functions. The present study was designed to verify the hypothesis that intestinal IR leads to mucosal CD8+T cell exhaustion, and that reinvigoration of exhausted CD8+T cell attenuates IR-induced bacterial translocation and liver damage. The intestinal IR model was performed through clamping the superior mesenteric artery in mice. The percent of exhausted CD8+T cells and the effector function of CD8+T cells were examined to determine the occurrence of intestinal mucosal CD8+T cell exhaustion. Subsequently, PD-1 blockade or combined PD-1 and IL-10 blockade was respectively used to reinvigorate exhausted CD8+T cells. Serum biomarkers, bacterial RNA and colonies, and inflammatory factors were examined to determine bacterial translocation and liver damage. The results indicated that intestinal IR induced CD8+T cell exhaustion in mucosal tissues, as evidenced by increased PD-1+ and PD-1+LAG-3+CD8+T cells and decreased IL-2 and TNF-α expression in CD8+T cells. Combined PD-1 and IL-10 blockade, but not PD-1 blockade alone, reinvigorated CD8+T cell exhaustion, as evidenced by increased generation of exhausted CD8+T cells with cytotoxicity and effector function, and elevated production of IFN-γ. Moreover, combined blockade significantly reduced the translocation of gut bacteria and injury to the liver after IR. In conclusion, intestinal IR leads to mucosal CD8+T cell exhaustion. Combined PD-1 and IL-10 blockade reinvigorates exhausted CD8+T cells, and ameliorates bacterial translocation and liver damage following IR.
Collapse
Affiliation(s)
- Zi-Meng Liu
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yi-Nan Zhang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hu-Fei Zhang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xu-Yu Zhang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
2
|
Takayama H, Fukatsu K, Noguchi M, Takahashi K, Watkins A, Matsumoto N, Narita T, Murakoshi S. Effects of parenteral nutrition supplemented with beta-hydroxy-beta-methylbutyrate on gut-associated lymphoid tissue and morphology in mice. JPEN J Parenter Enteral Nutr 2024; 48:927-939. [PMID: 39302343 DOI: 10.1002/jpen.2686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/08/2024] [Accepted: 08/29/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Parenteral nutrition (PN) without enteral nutrition (EN) leads to marked atrophy of gut-associated lymphoid tissue (GALT), causing mucosal defense failure in both the gut and the extraintestinal mucosal system. We evaluated the effects of beta-hydroxy-beta-methylbutyrate (HMB) on GALT and gut morphology in PN-fed mice. METHODS Experiment 1: male Institute of Cancer Research mice were assigned to the Chow (n = 12), Control (standard PN: n = 10), or H600 and H2000 (PN containing 600 mg/kg or H2000 mg/kg body weight of Ca-HMB: n = 12 and 10, respectively) groups. After 5 days of dietary manipulation, all mice were killed and the whole small intestine was harvested. GALT lymphocyte cell numbers and phenotypes of Peyer patch (PP), intraepithelial space, and lamina propria lymphocytes were evaluated. Experiment 2: 47 mice (Chow: n = 12; Control: n = 14; H600: n = 11; and H2000: n = 10) were fed for 5 days as in experiment 1. Proliferation and apoptosis of gut immune cells and mucosa, and protein expressions (mammalian target of rapamycin [mTOR], caspase-3, and Bcl2) were evaluated in the small intestine. RESULTS Compared with the Controls, the Chow and HMB groups showed significantly higher PP cell numbers, prevented gut mucosal atrophy, inhibited apoptosis of gut cells, and increased their proliferation in association with increased mTOR activity and Bcl2 expression. CONCLUSION HMB-supplemented PN is a potentially novel method of preserving GALT mass and gut morphology in the absence of EN.
Collapse
Affiliation(s)
- Haruka Takayama
- Operating Room Management and Surgical Metabolism, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazuhiko Fukatsu
- Operating Room Management and Surgical Metabolism, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Surgical Center, The University of Tokyo Hospital, Tokyo, Japan
| | - Midori Noguchi
- Surgical Center, The University of Tokyo Hospital, Tokyo, Japan
| | - Kazuya Takahashi
- Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ayako Watkins
- Operating Room Management and Surgical Metabolism, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Nana Matsumoto
- Operating Room Management and Surgical Metabolism, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomonori Narita
- Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Satoshi Murakoshi
- Operating Room Management and Surgical Metabolism, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Surgical Center, The University of Tokyo Hospital, Tokyo, Japan
| |
Collapse
|
3
|
Tian Y, Dhara S, Barrett CD, Richman AP, Brahmbhatt TS. Antibiotic use in acute mesenteric ischemia: a review of the evidence and call to action. Thromb J 2023; 21:39. [PMID: 37041639 PMCID: PMC10088293 DOI: 10.1186/s12959-023-00486-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 04/03/2023] [Indexed: 04/13/2023] Open
Abstract
Acute mesenteric ischemia (AMI) is a life-threatening condition with a high mortality rate. The standard practice after making the diagnosis includes aggressive resuscitation, anticoagulation, followed by revascularization and resection of necrotic bowel. The role of empiric antibiotics in the management of AMI is not well defined in the literature. This review article aims to examine our current understanding on this matter, based on bench research and clinical studies. It is demonstrated in animal study model that the ischemia/reperfusion (I/R) injury damages intestinal epithelium, and subsequently lead to barrier dysfunction, a condition that can support bacterial translocation through a complex interplay between the intestinal epithelium, the intestinal immune system and the intestine's endogenous bacterial population. Based on this mechanism, it is possible that the use of antibiotics may help mitigate the consequences of I/R injury, which is examined in few animal studies. In clinical practice, many guidelines support the use of prophylactic antibiotics, based on a meta-analysis of randomized control trials (RCTs) demonstrating the benefit of antibiotics in multi-organ dysfunction syndrome. However, there is no direct reference to AMI in this meta-analysis. Most clinical studies that focus on AMI and mentions the use of antibiotics are retrospective and single institution, and very few comments on the role of antibiotics in their discussions. We conclude that there is limited evidence in literature to support the use of prophylactic antibiotic in AMI to improve outcome. More clinical studies with high level of evidence and basic science research are needed to improve our understanding on this topic and ultimately help build a better clinical pathway for patients with AMI.
Collapse
Affiliation(s)
- Yuqian Tian
- Division of Acute Care Surgery and Surgical Critical Care, Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sanjeev Dhara
- Division of Vascular Surgery, Department of Surgery, Emory University, Atlanta, GA, USA
| | - Christopher D Barrett
- Division of Acute Care Surgery and Surgical Critical Care, Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA
| | - Aaron P Richman
- Divisions of Trauma and Acute Care Surgery and Surgical Critical Care, Department of Surgery, Boston University Medical Center, Boston University School of Medicine, Boston, MA, USA
| | - Tejal S Brahmbhatt
- Divisions of Trauma and Acute Care Surgery and Surgical Critical Care, Department of Surgery, Boston University Medical Center, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
4
|
Dmytriiev D, Nazarchuk O, Melnychenko M, Levchenko B. Optimization of the target strategy of perioperative infusion therapy based on monitoring data of central hemodynamics in order to prevent complications. Front Med (Lausanne) 2022; 9:935331. [PMID: 36262276 PMCID: PMC9573976 DOI: 10.3389/fmed.2022.935331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
Enhanced Recovery After Surgery (ERAS) protocols are increasingly used in the perioperative period around the world. The concept of goal-directed fluid therapy (GDT) is a key element of the ERAS protocols. Inadequate perioperative infusion therapy can lead to a number of complications, including the development of an infectious process, namely surgical site infections, pneumonia, urinary tract infections. Optimal infusion therapy is difficult to achieve with standard parameters (e.g., heart rate, blood pressure, central venous pressure), so there are various methods of monitoring central hemodynamics - from invasive, minimally invasive to non-invasive. The latter are increasingly used in clinical practice. The current evidence base shows that perioperative management, specifically the use of GDT guided by real-time, continuous hemodynamic monitoring, helps clinicians maintain a patient's optimal fluid balance. The manuscript presents the analytical data, which describe the benefits and basic principles of perioperative targeted infusion therapy based on central hemodynamic parameters to reduce the risk of complications.
Collapse
Affiliation(s)
- Dmytro Dmytriiev
- Department of Anesthesiology and Intensive Care, National Pirogov Memorial Medical University, Vinnytsya, Ukraine
| | - Oleksandr Nazarchuk
- Department of Microbiology, National Pirogov Memorial Medical University, Vinnytsya, Ukraine
| | - Mykola Melnychenko
- Department of Anesthesiology and Intensive Care, National Pirogov Memorial Medical University, Vinnytsya, Ukraine
| | - Bohdan Levchenko
- Department of Anesthesiology and Intensive Care, National Pirogov Memorial Medical University, Vinnytsya, Ukraine
| |
Collapse
|
5
|
Takayama H, Fukatsu K, Takahashi K, Noguchi M, Watkins A, Matsumoto N, Murakoshi S. Influences of a fermented milk with Lactobacillus bulgaricus and Streptococcus thermophiles on gut associated lymphoid tissue, mucosal IgA, and gut flora in mice. CLINICAL NUTRITION OPEN SCIENCE 2022. [DOI: 10.1016/j.nutos.2022.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
6
|
Hundscheid IHR, Schellekens DHSM, Grootjans J, Den Dulk M, Van Dam RM, Beets GL, Buurman WA, Lenaerts K, Derikx JPM, Dejong CHC. Evaluating the safety of two human experimental intestinal ischemia reperfusion models: A retrospective observational study. PLoS One 2021; 16:e0253506. [PMID: 34143845 PMCID: PMC8213171 DOI: 10.1371/journal.pone.0253506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 06/05/2021] [Indexed: 11/18/2022] Open
Abstract
Background We developed a jejunal and colonic experimental human ischemia-reperfusion (IR) model to study pathophysiological intestinal IR mechanisms and potential new intestinal ischemia biomarkers. Our objective was to evaluate the safety of these IR models by comparing patients undergoing surgery with and without in vivo intestinal IR. Methods A retrospective study was performed comparing complication rates and severity, based on the Clavien-Dindo classification system, in patients undergoing pancreatoduodenectomy with (n = 10) and without (n = 20 matched controls) jejunal IR or colorectal surgery with (n = 10) and without (n = 20 matched controls) colon IR. Secondary outcome parameters were operative time, blood loss, 90-day mortality and length of hospital stay. Results Following pancreatic surgery, 63% of the patients experienced one or more postoperative complications. There was no significant difference in incidence or severity of complications between patients undergoing pancreatic surgery with (70%) or without (60%, P = 0.7) jejunal IR. Following colorectal surgery, 60% of the patients experienced one or more postoperative complication. Complication rate and severity were similar in patients with (50%) and without (65%, P = 0.46) colonic IR. Operative time, amount of blood loss, postoperative C-reactive protein, length of hospital stay or mortality were equal in both intervention and control groups for jejunal and colon IR. Conclusion This study showed that human experimental intestinal IR models are safe in patients undergoing pancreatic or colorectal surgery.
Collapse
Affiliation(s)
- Inca H. R. Hundscheid
- Department of Pathology, Maastricht University Medical Centre+, Maastricht, The Netherlands
- NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
- * E-mail:
| | - Dirk H. S. M. Schellekens
- NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
- Department of Surgery, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Joep Grootjans
- NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, the Netherlands
| | - Marcel Den Dulk
- Department of Surgery, Maastricht University Medical Centre+, Maastricht, The Netherlands
- Department of Surgery, RWTH University Hospital Aachen, Aachen, Germany
| | - Ronald M. Van Dam
- Department of Surgery, Maastricht University Medical Centre+, Maastricht, The Netherlands
- Department of Surgery, RWTH University Hospital Aachen, Aachen, Germany
| | - Geerard L. Beets
- Department of Surgery, The Antoni van Leeuwenhoek Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Wim A. Buurman
- MHeNs School for Mental Healthy and Neuroscience, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Kaatje Lenaerts
- NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
- Department of Surgery, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Joep P. M. Derikx
- Department of Surgery, Maastricht University Medical Centre+, Maastricht, The Netherlands
- Department of Pediatric Surgery, Emma Children’s Hospital, Amsterdam University Medical Centre, University of Amsterdam, Free University of Amsterdam, Amsterdam, The Netherlands
| | - Cornelis H. C. Dejong
- NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
- Department of Surgery, Maastricht University Medical Centre+, Maastricht, The Netherlands
- Department of Surgery, RWTH University Hospital Aachen, Aachen, Germany
| |
Collapse
|
7
|
Wang C, Li Q, Ren J. Microbiota-Immune Interaction in the Pathogenesis of Gut-Derived Infection. Front Immunol 2019; 10:1873. [PMID: 31456801 PMCID: PMC6698791 DOI: 10.3389/fimmu.2019.01873] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 07/24/2019] [Indexed: 12/12/2022] Open
Abstract
Gut-derived infection is among the most common complications in patients who underwent severe trauma, serious burn, major surgery, hemorrhagic shock or severe acute pancreatitis (SAP). It could cause sepsis and multiple organ dysfunction syndrome (MODS), which are regarded as a leading cause of mortality in these cases. Gut-derived infection is commonly caused by pathological translocation of intestinal bacteria or endotoxins, resulting from the dysfunction of the gut barrier. In the last decades, the studies regarding to the pathogenesis of gut-derived infection mainly focused on the breakdown of intestinal epithelial tight junction and increased permeability. Limited information is available on the roles of intestinal microbial barrier in the development of gut-derived infection. Recently, advances of next-generation DNA sequencing techniques and its utilization has revolutionized the gut microecology, leading to novel views into the composition of the intestinal microbiota and its connections with multiple diseases. Here, we reviewed the recent progress in the research field of intestinal barrier disruption and gut-derived infection, mainly through the perspectives of the dysbiosis of intestinal microbiota and its interaction with intestinal mucosal immune cells. This review presents novel insights into how the gut microbiota collaborates with mucosal immune cells to involve the development of pathological bacterial translocation. The data might have important implication to better understand the mechanism underlying pathological bacterial translocation, contributing us to develop new strategies for prevention and treatment of gut-derived sepsis.
Collapse
Affiliation(s)
| | - Qiurong Li
- Research Institute of General Surgery, Jinling Hospital, Medical School, Nanjing University, Nanjing, China
| | - Jianan Ren
- Research Institute of General Surgery, Jinling Hospital, Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
8
|
Fukatsu K. Role of nutrition in gastroenterological surgery. Ann Gastroenterol Surg 2019; 3:160-168. [PMID: 30923785 PMCID: PMC6422822 DOI: 10.1002/ags3.12237] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 11/25/2018] [Accepted: 12/16/2018] [Indexed: 12/14/2022] Open
Abstract
Nutrition plays important roles in recovery after gastroenterological surgery. Severe surgical stress increases muscle breakdown and lipolysis, thereby accelerating wound healing and enhancing host defense against microbes. Malnourished patients have insufficient amounts of muscle and body fat. Therefore, they may not appropriately respond to surgical stress. Perioperative nutritional therapy maintaining nutritional status reduces postoperative complications and accelerates recovery after surgery, particularly for malnourished patients. In addition, perioperative oral or enteral nutrition is now recommended for preserving host defense mechanisms against microbes. Lack of enteral nutrition impairs gut and hepatic immunity, systemic mucosal defense and peritoneal host defense, even when nutrient amounts supplied by parenteral nutrition are adequate. Thus, surgeons should avoid no oral or enteral nutrition periods. Supplemental administration of specific nutrients such as glutamine, arginine and ω-3 fatty acids is termed "immunonutrition", and is expected to reduce the morbidity of infectious complications and length of hospital stay. Nutritional therapy is important even after discharge to maintain body weight and compensate for abnormalities in the digestion and absorption of nutrients. Understanding the significance of nutrition in gastroenterological patients leads to better outcomes.
Collapse
|
9
|
Activation of PD-1 Protects Intestinal Immune Defense Through IL-10/miR-155 Pathway After Intestinal Ischemia Reperfusion. Dig Dis Sci 2018; 63:3307-3316. [PMID: 30206757 DOI: 10.1007/s10620-018-5282-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 09/08/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND To date, mechanisms of intestinal immunoglobulin (Ig) dysfunction following intestinal ischemia/reperfusion (I/R) remain unclear. Programmed death 1 (PD-1) is associated with immune responses of lymphocytes. AIM We aimed to verify the hypothesis that activation of PD-1 may improve intestinal immune dysfunction by regulating IL-10/miR-155 production after intestinal IR injury. METHODS Intestinal I/R injury was induced in mice by clamping the superior mesenteric artery for 1 h followed by 2-h reperfusion. PD-L1 fusion Ig, anti-interleukin (IL)-10 monoclonal antibody (mAb), and microRNA (miR)-155 agomir were administered. PD-1 expression, IL-10 mRNA, and protein expression in Peyer's patches (PP) CD4+ cells were measured. MiR-155 levels, tumor necrosis factor (TNF)-α and IL-1β concentration, and activation-induced cytidine deaminase (AID), a key enzyme for intestinal immune antibodies, in PP tissues were measured, respectively. Importantly, the production and cecal bacteria-binding capacity of IgA and IgM were detected. RESULTS Intestinal I/R led to decreased PD-1 expression, imbalanced production, and impaired bacteria-binding capacity of IgA and IgM. Activating PD-1 by PD-L1 Ig facilitated IL-10 synthesis, then decreased miR-155 levels, and subsequently promoted AID expression and reduced TNF-α, IL-1β concentration. Upregulation of AID improved the disruptions of intestinal immune barrier caused by IgA and IgM dysfunction. Anti-IL-10 mAb and miR-155 agomir abolished the protective effects of PD-L1 Ig on the intestinal immune defense. CONCLUSION Activation of PD-1 with PD-L1 Ig relieves intestinal immune defensive injury through IL-10/miR-155 pathway following intestinal I/R attack. PD-1, IL-10, and miR-155 may be potential targets for the damages of intestinal barrier and immunity.
Collapse
|
10
|
Higashizono K, Fukatsu K, Watkins A, Watanabe T, Noguchi M, Ri M, Murakoshi S, Yasuhara H, Seto Y. Influences of Short-Term Fasting and Carbohydrate Supplementation on Gut Immunity and Mucosal Morphology in Mice. JPEN J Parenter Enteral Nutr 2018; 43:516-524. [PMID: 30260489 DOI: 10.1002/jpen.1446] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 08/16/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Preoperative carbohydrate (CHO) supplementation has been recommended in enhanced recovery after surgery protocols. However, the effects of CHO supplementation on gut and systemic immunity are not well understood. METHODS Mice (n = 60) were randomized to 1 of the following 5 groups: control (ad lib feeding), 12-hour fasting without CHO administration (fasting), and 12 hours of fasting with CHO administration at 2, 4, and 8 hours before sacrifice. Then, lymphocytes were isolated from gut-associated lymphoid tissue, that is, Peyer's patches, the intraepithelial space, and the lamina propria of the small intestine. These lymphocyte numbers and phenotypes were evaluated. IgA levels in respiratory and small-intestinal washings were determined by ELISA. Morphology, proliferation, and apoptosis of the intestinal epithelium were also evaluated histologically. RESULTS Although there were no significant differences in IgA levels among the 5 groups, fasting decreased intraepithelial and lamina propria, but not Peyer's patches lymphocyte numbers. CHO at 2 hours prevented lymphocyte loss in intraepithelial, whereas CHO at 4 hours reversed lamina propria lymphocytes numbers. Percentages of lymphocyte phenotypes were similar in each site among the 5 groups. Fasting caused villous atrophy; however, CHO at 2 hours restored villous structure along with maintenance of epithelial cell proliferation rate. CONCLUSIONS Only 12 hours of fasting causes marked gut-associated lymphoid tissue cell loss along with gut atrophy. However, CHO at 2 hours preserves gut immunity and morphology not completely but moderately.
Collapse
Affiliation(s)
- Kazuya Higashizono
- Department of Gastrointestinal Surgery, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan.,Surgical Center, the University of Tokyo Hospital, Tokyo, Japan
| | | | - Ayako Watkins
- Surgical Center, the University of Tokyo Hospital, Tokyo, Japan
| | - Tomoki Watanabe
- Surgical Center, the University of Tokyo Hospital, Tokyo, Japan
| | - Midori Noguchi
- Surgical Center, the University of Tokyo Hospital, Tokyo, Japan
| | - Motonari Ri
- Department of Gastrointestinal Surgery, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | | | | | - Yasuyuki Seto
- Department of Gastrointestinal Surgery, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| |
Collapse
|
11
|
|
12
|
Effects of short-term fasting on gut-associated lymphoid tissue and intestinal morphology in mice. CLINICAL NUTRITION EXPERIMENTAL 2018. [DOI: 10.1016/j.yclnex.2017.12.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
13
|
Schellekens DHSM, Hundscheid IHR, Leenarts CAJI, Grootjans J, Lenaerts K, Buurman WA, Dejong CHC, Derikx JPM. Human small intestine is capable of restoring barrier function after short ischemic periods. World J Gastroenterol 2017; 23:8452-8464. [PMID: 29358855 PMCID: PMC5752707 DOI: 10.3748/wjg.v23.i48.8452] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 08/08/2017] [Accepted: 08/15/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To assess intestinal barrier function during human intestinal ischemia and reperfusion (IR).
METHODS In a human experimental model, 6 cm of jejunum was selectively exposed to 30 min of ischemia (I) followed by 30 and 120 min of reperfusion (R). A sham procedure was also performed. Blood and tissue was sampled at all-time points. Functional barrier function was assessed using dual-sugar absorption tests with lactulose (L) and rhamnose (R). Plasma concentrations of citrulline, an amino acid described as marker for enterocyte function were measured as marker of metabolic enterocytes restoration. Damage to the epithelial lining was assessed by immunohistochemistry for tight junctions (TJs), by plasma marker for enterocytes damage (I-FABP) and analyzed by electron microscopy (EM) using lanthanum nitrate as an electrondense marker.
RESULTS Plasma L/R ratio’s were significantly increased after 30 min of ischemia (30I) followed by 30 min of reperfusion (30R) compared to control (0.75 ± 0.10 vs 0.20 ± 0.09, P < 0.05). At 120 min of reperfusion (120R), ratio’s normalized (0.17 ± 0.06) and were not significantly different from control. Plasma levels of I-FABP correlated with plasma L/R ratios measured at the same time points (correlation: 0.467, P < 0.01). TJs staining shows distortion of staining at 30I. An intact lining of TJs was again observed at 30I120R. Electron microscopy analysis revealed disrupted TJs after 30I with paracellular leakage of lanthanum nitrate, which restored after 30I120R. Furthermore, citrulline concentrations closely paralleled the histological perturbations during intestinal IR.
CONCLUSION This study directly correlates histological data with intestinal permeability tests, revealing that the human gut has the ability of to withstand short episodes of ischemia, with morphological and functional recovery of the intestinal barrier within 120 min of reperfusion.
Collapse
Affiliation(s)
- Dirk HSM Schellekens
- Department of Surgery, Maastricht University Medical Center, Maastricht 6200 MD, the Netherlands
- NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht 6200 MD, the Netherlands
| | - Inca HR Hundscheid
- Department of Surgery, Maastricht University Medical Center, Maastricht 6200 MD, the Netherlands
- NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht 6200 MD, the Netherlands
| | - Claire AJI Leenarts
- Department of Surgery, Maastricht University Medical Center, Maastricht 6200 MD, the Netherlands
- NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht 6200 MD, the Netherlands
| | - Joep Grootjans
- NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht 6200 MD, the Netherlands
- Department of Gastroenterology, Academic Medical Center, Amsterdam 1105 AZ, the Netherlands
| | - Kaatje Lenaerts
- Department of Surgery, Maastricht University Medical Center, Maastricht 6200 MD, the Netherlands
- NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht 6200 MD, the Netherlands
| | - Wim A Buurman
- NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht 6200 MD, the Netherlands
- MHeNs School of Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht 6200 MD, the Netherlands
| | - Cornelis HC Dejong
- Department of Surgery, Maastricht University Medical Center, Maastricht 6200 MD, the Netherlands
- NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht 6200 MD, the Netherlands
| | - Joep PM Derikx
- Department of Surgery, Maastricht University Medical Center, Maastricht 6200 MD, the Netherlands
- NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht 6200 MD, the Netherlands
- Pediatric Surgical Center of Amsterdam, Emma Children's Hospital Academic Medical Center and VU University Medical Center, Amsterdam 1100 DE, the Netherlands
| |
Collapse
|
14
|
Fukatsu K, Moriya T, Ikezawa F, Maeshima Y, Omata J, Yaguchi Y, Okamoto K, Mochizuki H, Hiraide H, Hardy G. Interleukin-7 Dose-Dependently Restores Parenteral Nutrition–Induced Gut-Associated Lymphoid Tissue Cell Loss but Does Not Improve Intestinal Immunoglobulin A Levels. JPEN J Parenter Enteral Nutr 2017; 30:388-93; discussion 393-4. [PMID: 16931606 DOI: 10.1177/0148607106030005388] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Without enteral nutrition, the mass and function of gut-associated lymphoid tissue (GALT), a center of systemic mucosal immunity, are reduced. Therefore, new therapeutic methods, designed to preserve mucosal immunity during parenteral nutrition (PN), are needed. Our recent study revealed that exogenous interleukin-7 (IL-7; 1 microg/kg twice a day) restores the GALT cell mass lost during intravenous (IV) PN but does not improve secretory immunoglobulin A (IgA) levels. Herein, we studied the IL-7 dose response to determine the optimal IL-7 dose for recovery of GALT mass and function during IV PN. We hypothesized that a high dose of IL-7 would increase intestinal IgA levels, as well as GALT cell numbers. METHODS Male mice (n = 42) were randomized to chow, IL-7-0, IL-7-0.1, IL-7-0.33, IL-7-1 and IL-7-3.3 groups and underwent jugular vein catheter insertion. The IL-7 groups were fed a standard PN solution and received IV injections of normal saline (IL-7-0), 0.1, 0.33, 1, or 3.3 microg/kg of IL-7 twice a day. The chow group was fed chow ad libitum. After 5 days of treatment, the entire small intestine was harvested and lymphocytes were isolated from Peyer's patches (PPs), intraepithelial (IE) spaces, and the lamina propria (LP). The lymphocytes were counted and phenotypes determined by flow cytometry (alphabetaTCR, gammadeltaTCR, CD4, CD8, B cell). IgA levels of small intestinal washings were also examined using ELISA (enzyme-linked immunoabsorbent assay). RESULTS IL-7 dose-dependently increased total lymphocyte numbers in PPs and the LP. The number of lymphocytes harvested from IE spaces reached a plateau at 1 microg/kg of IL-7. There were no significant differences in any phenotype percentages at any GALT sites among the groups. IgA levels of intestinal washings were significantly higher in the chow group than in any of the IL-7 groups, with similar levels in all IL-7 groups. CONCLUSIONS Exogenous IL-7 dose-dependently reverses PN-induced GALT cell loss, with no major changes in small intestinal IgA levels. IL-7 treatment during PN appears to have beneficial effects on gut immunity, but other therapeutic methods are needed to restore secretory IgA levels.
Collapse
Affiliation(s)
- Kazuhiko Fukatsu
- Division of Basic Traumatology, National Defense Medical College Research Institute, Tokorozawa, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Tumor Necrosis Factor α-Dependent Neutrophil Priming Prevents Intestinal Ischemia/Reperfusion-Induced Bacterial Translocation. Dig Dis Sci 2017; 62:1498-1510. [PMID: 28144894 DOI: 10.1007/s10620-017-4468-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 01/20/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND Intestinal ischemia/reperfusion (I/R) causes barrier impairment and bacterial influx. Protection against I/R injury in sterile organs by hypoxic preconditioning (HPC) had been attributed to erythropoietic and angiogenic responses. Our previous study showed attenuation of intestinal I/R injury by HPC for 21 days in a neutrophil-dependent manner. AIM To investigate the underlying mechanisms of neutrophil priming by HPC, and explore whether adoptive transfer of primed neutrophils is sufficient to ameliorate intestinal I/R injury. METHODS Rats raised in normoxia (NM) and HPC for 3 or 7 days were subjected to sham operation or superior mesenteric artery occlusion for I/R challenge. Neutrophils isolated from rats raised in NM or HPC for 21 days were intravenously injected into naïve controls prior to I/R. RESULTS Similar to the protective effect of HPC-21d, I/R-induced mucosal damage was attenuated by HPC-7d but not by HPC-3d. Naïve rats reconstituted with neutrophils of HPC-21d rats showed increase in intestinal phagocytic infiltration and myeloperoxidase activity, and barrier protection against I/R insult. Elevated free radical production, and higher bactericidal and phagocytic activity were observed in HPC neutrophils compared to NM controls. Moreover, increased serum levels of tumor necrosis factor α (TNFα) and cytokine-induced neutrophil chemoattractant-1 (CINC-1) were seen in HPC rats. Naïve neutrophils incubated with HPC serum or recombinant TNFα, but not CINC-1, exhibited heightened respiratory burst and bactericidal activity. Lastly, neutrophil priming effect was abolished by neutralization of TNFα in HPC serum. CONCLUSIONS TNFα-primed neutrophils by HPC act as effectors cells for enhancing barrier integrity under gut ischemia.
Collapse
|
16
|
Rao G, Yadav VR, Awasthi S, Roberts PR, Awasthi V. Effect of liposome-encapsulated hemoglobin resuscitation on proteostasis in small intestinal epithelium after hemorrhagic shock. Am J Physiol Gastrointest Liver Physiol 2016; 311:G180-91. [PMID: 27288424 PMCID: PMC4967179 DOI: 10.1152/ajpgi.00157.2016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 05/29/2016] [Indexed: 01/31/2023]
Abstract
Gut barrier dysfunction is the major trigger for multiorgan failure associated with hemorrhagic shock (HS). Although the molecular mediators responsible for this dysfunction are unclear, oxidative stress-induced disruption of proteostasis contributes to the gut pathology in HS. The objective of this study was to investigate whether resuscitation with nanoparticulate liposome-encapsulated hemoglobin (LEH) is able to restore the gut proteostatic mechanisms. Sprague-Dawley rats were recruited in four groups: control, HS, HS+LEH, and HS+saline. HS was induced by withdrawing 45% blood, and isovolemic LEH or saline was administered after 15 min of shock. The rats were euthanized at 6 h to collect plasma and ileum for measurement of the markers of oxidative stress, unfolded protein response (UPR), proteasome function, and autophagy. HS significantly increased the protein and lipid oxidation, trypsin-like proteasome activity, and plasma levels of IFNγ. These effects were prevented by LEH resuscitation. However, saline was not able to reduce protein oxidation and plasma IFNγ in hemorrhaged rats. Saline resuscitation also suppressed the markers of UPR and autophagy below the basal levels; the HS or LEH groups showed no effect on the UPR and autophagy. Histological analysis showed that LEH resuscitation significantly increased the villus height and thickness of the submucosal and muscularis layers compared with the HS and saline groups. Overall, the results showed that LEH resuscitation was effective in normalizing the indicators of proteostasis stress in ileal tissue. On the other hand, saline-resuscitated animals showed a decoupling of oxidative stress and cellular protective mechanisms.
Collapse
Affiliation(s)
- Geeta Rao
- 1Department of Pharmaceutical Sciences, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma; and
| | - Vivek R. Yadav
- 1Department of Pharmaceutical Sciences, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma; and
| | - Shanjana Awasthi
- 1Department of Pharmaceutical Sciences, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma; and
| | - Pamela R. Roberts
- 2Department of Anesthesiology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma
| | - Vibhudutta Awasthi
- Department of Pharmaceutical Sciences, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma; and
| |
Collapse
|
17
|
Zhang X, Liu Z, Zhang H, Li Y, Wen S, Shen J, Huang W, Liu K. TGF-β1 improves mucosal IgA dysfunction and dysbiosis following intestinal ischaemia-reperfusion in mice. J Cell Mol Med 2016; 20:1014-1023. [PMID: 26820382 PMCID: PMC4882980 DOI: 10.1111/jcmm.12789] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 12/13/2015] [Indexed: 12/14/2022] Open
Abstract
Intestinal ischaemia/reperfusion (I/R) severely disrupts gut barriers and leads to high mortality in the critical care setting. Transforming growth factor (TGF)-β1 plays a pivotal role in intestinal cellular and immune regulation. However, the effects of TGF-β1 on intestinal I/R injury remain unclear. Thus, we aimed to investigate the effects of TGF-β1 on gut barriers after intestinal I/R and the molecular mechanisms. Intestinal I/R model was produced in mice by clamping the superior mesenteric artery for 1 hr followed by reperfusion. Recombinant TGF-β1 was intravenously infused at 15 min. before ischaemia. The results showed that within 2 hrs after reperfusion, intestinal I/R disturbed intestinal immunoglobulin A class switch recombination (IgA CSR), the key process of mucosal IgA synthesis, and resulted in IgA dysfunction, as evidenced by decreased production and bacteria-binding capacity of IgA. Meanwhile, the disruptions of intestinal microflora and mucosal structure were exhibited. Transforming growth factor-β1 activated IgA CSR as evidenced by the increased activation molecules and IgA precursors. Strikingly, TGF-β1 improved intestinal mucosal IgA dysfunction, dysbiosis and epithelial damage at the early stage after reperfusion. In addition, SB-431542, a specific inhibitor of activating mothers against decapentaplegic homologue (SMAD) 2/3, totally blocked the inductive effect of TGF-β1 on IgA CSR and almost abrogated the above protective effects on intestinal barriers. Taken together, our study demonstrates that TGF-β1 protects intestinal mucosal IgA immunity, microbiota and epithelial integrity against I/R injury mainly through TGF-β receptor 1/SMAD 2/3 pathway. Induction of IgA CSR may be involved in the protection conferred by TGF-β1.
Collapse
Affiliation(s)
- Xu‐Yu Zhang
- Department of AnesthesiologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Zi‐Meng Liu
- Surgical Intensive Care UnitThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Hu‐Fei Zhang
- Department of AnesthesiologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Yun‐Sheng Li
- Department of AnesthesiologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Shi‐Hong Wen
- Department of AnesthesiologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Jian‐Tong Shen
- Department of AnesthesiologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Wen‐Qi Huang
- Department of AnesthesiologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Ke‐Xuan Liu
- Department of AnesthesiologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
18
|
Zhang XY, Liu ZM, Zhang HF, Li YS, Wen SH, Shen JT, Liu KX. Decreased PD-1/PD-L1 Expression Is Associated with the Reduction in Mucosal Immunoglobulin A in Mice with Intestinal Ischemia Reperfusion. Dig Dis Sci 2015; 60:2662-2669. [PMID: 25944714 DOI: 10.1007/s10620-015-3684-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 04/23/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND Intestinal ischemia/reperfusion (I/R) disrupts intestinal mucosal integrity and immunoglobulin A (IgA) generation. It has recently been shown that the programmed cell death-1 receptor (PD-1) plays a crucial role in regulating intestinal secreted IgA (sIgA). AIMS To evaluate changes in PD-1 and PD-ligand 1 (PD-L1) expression on Peyer's patches (PP) CD4(+) T cells and to investigate the correlation between PD-1/PD-L1 and intestinal IgA production/mucosal integrity in mice following intestinal I/R. METHODS I/R injury was induced by clamping the superior mesenteric artery for 1 h followed by 2-h reperfusion. PD-1/PD-L1 expression on PP CD4(+) T cells was measured in I/R and sham-operated mice. Additionally, transforming growth factor-β1 (TGF-β1) and interleukin-21 (IL-21) mRNA in CD4(+) T cells and IgA(+) and IgM(+) in PP B cells, as well as intestinal mucosal injury and sIgA levels, were assessed. RESULTS PD-1/PD-L1, TGF-β1, and IL-21 expression was down-regulated after intestinal I/R. Furthermore, IgA(+) B cells decreased and IgM(+) B cells increased in mice with intestinal I/R. Importantly, decreased PD-1/PD-L1 expression was correlated with increased mucosal injury and decreased IgA levels, as well as with decreased TGF-β1 and IL-21 expression. CONCLUSIONS Intestinal I/R inhibits PD-1/PD-L1 expression on PP CD4(+) T cells, which was associated with an impaired intestinal immune system and mechanical barriers. Our study indicates that PD-1/PD-L1 expression on CD4(+) T cells may be involved in the pathogenesis of intestinal I/R injury.
Collapse
Affiliation(s)
- Xu-Yu Zhang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan 2nd Road, Guangzhou, 510080, China
| | | | | | | | | | | | | |
Collapse
|
19
|
Fan J, Li G, Wu L, Tao S, Wang W, Sheng Z, Meng Q. Parenteral glutamine supplementation in combination with enteral nutrition improves intestinal immunity in septic rats. Nutrition 2014; 31:766-74. [PMID: 25837225 DOI: 10.1016/j.nut.2014.11.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 11/15/2014] [Accepted: 11/29/2014] [Indexed: 01/27/2023]
Abstract
OBJECTIVES The gut-associated lymphoid tissue is continuously exposed to antigens in the gut lumen and becomes the first line of defense against enteric bacteria and associated toxin. The aim of this study was to investigate the effects of parenteral glutamine (GLN) supplementation in combination with enteral nutrition (EN) on intestinal mucosal immunity in septic rats by cecal ligation and puncture (CLP). METHODS Male Sprague-Dawley rats were randomly assigned into four groups: A sham CLP + EN + saline group (n = 10), a sham CLP + EN + GLN group (n = 10), a CLP + EN + saline group (n = 10), and a CLP + EN + GLN group (n = 10). At 2 h after CLP or sham CLP, all rats in each of the four groups received an identical enteral nutrition solution as their base formula. Then, the rats in the sham CLP + EN + GLN group and CLP + EN + GLN group were given 0.35 g GLN/kg body weight daily for 7 d, all at the same time, via a tail vein injection; whereas those in the sham CLP + EN + saline group and CLP + EN + saline group were daily administered isovolumic sterile 0.9% saline for comparison. All rats in each of the four groups were given 290 kcal/kg body wt/d for 7 d. At the end of the seventh day after the nutritional program was finished, all rats were euthanized and the entire intestine was collected. Total Peyer's patches (PP) cell yield was counted by a hemocytometer. The percentage of PP lymphocyte subsets was analyzed by flow cytometry. The number of intestinal lamina propria IgA plasma cells was determined by the immunohistochemistry technique. The intestinal immunoglobulin A (IgA) levels were assessed by ELISA. PP apoptosis was evaluated by terminal deoxyuridine nick-end labeling. RESULTS The results revealed total PP cell yield, the numbers of PP lymphocyte subsets, intestinal lamina propria IgA plasma cells, and intestinal IgA levels in the CLP + EN + GLN group were significantly increased when compared with the CLP + EN + saline group (P < 0.05). On the other hand, the number of TUNEL-stained cells within PPs in the CLP + EN + GLN group was markedly decreased as compared with the CLP + EN + saline group (P < 0.05). CONCLUSION The results of this study show that parenteral glutamine supplementation in combination with enteral nutrition may attenuate PP apoptosis, increase PP cell yield and intestinal lamina propria IgA plasma cells, and subsequently improve intestinal mucosal immunity. Clinically, these results suggest therapeutic efforts at improving intestinal immunity may contribute to the prevention and treatment of sepsis.
Collapse
Affiliation(s)
- Jun Fan
- Department of Emergency and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Jiangxi, P.R. China.
| | - Guoping Li
- Department of Emergency and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Jiangxi, P.R. China
| | - Lidong Wu
- Department of Emergency and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Jiangxi, P.R. China
| | - Shaoyu Tao
- Department of Emergency and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Jiangxi, P.R. China
| | - Wei Wang
- Department of Emergency and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Jiangxi, P.R. China
| | - Zhiyong Sheng
- Department of Emergency and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Jiangxi, P.R. China
| | - Qingyan Meng
- Department of Burns, The Northern Hospital, Liaoning, P.R. China
| |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW Enteral nutrition is recommended as a standard nutritional therapy in clinical settings. The rationale behind enteral nutrition may be decreased infectious morbidities compared with parenteral nutrition. However, the mechanism may not be well understood. RECENT FINDINGS Animal studies have revealed that enteral nutrition, compared with parenteral nutrition, preserves the gut-associated lymphoid tissue mass and function with well controlled gut cytokine milieu and intracellular signaling pathway, leading to the maintenance of intestinal and extraintestinal acquired mucosal immunity. Moreover, enteral nutrition can enhance the gut innate immunity by increasing the antimicrobial peptides, such as secretory phospholipase A2. More importantly, a recent clinical study demonstrated preoperative parenteral nutrition without enteral nutrition to decrease the number of T cells, IgA-producing cells, and mature dendritic cells in human terminal ileum, which are consistent with the data obtained from animal studies. Investigation of the mechanism has given us some surrogates of enteral nutrition during parenteral nutrition, such as glutamine, butyric acid, cytokines, and other mediators. However, to date, no surrogates can restore parenteral-nutrition-induced impairment of host defense completely. SUMMARY Because enteral nutrition is a practical way to preserve gut immunity, clinicians should make any efforts to shorten the period of enteral nutrition absence and increase the dose according to the degree of tolerance.
Collapse
|
21
|
Lack of preoperative enteral nutrition reduces gut-associated lymphoid cell numbers in colon cancer patients: a possible mechanism underlying increased postoperative infectious complications during parenteral nutrition. Ann Surg 2014. [PMID: 23187750 DOI: 10.1097/sla.0b013e31827a0e05] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
OBJECTIVE To examine preoperative dietary influences on gut-associated lymphoid tissue (GALT) cell number in the context of postoperative infectious complications. BACKGROUND There is little clinical evidence regarding whether nutritional routes affect GALT size and/or phenotype. The influence of GALT atrophy on clinical outcomes is also unclear. METHOD Patients with complete obstruction of the colon due to a tumor were excluded from this study. Study 1. Resected terminal ileum specimens, from 62 patients [preoperative parenteral nutrition (PN): n = 15, preoperative oral feeding (OF): n = 47] who underwent right colectomy during the period from 1997 to 2004 at our department, were immunohistochemically stained for counting numbers of T, IgA-producing, and mature and immature dendritic cells (DCs) in the lamina propria (LP) and intraepithelial space.Study 2. We reviewed 341 patients (PN: n = 99, OF: n = 242) with colon cancer who underwent colectomy during this period for postoperative complications. RESULTS Study 1. T cell numbers in the LP and intraepithelial space and IgA-producing cell number in the LP were significantly lower in the PN than in the OF group. Mature DC number in the LP was significantly lower in the PN than in the OF group, whereas total DC numbers (both mature and immature DC) were similar in the 2 groups.Study 2. The PN group had significantly higher rates of total infectious complications, surgical site infection, pneumonia, infectious colitis, and central venous catheter infection. CONCLUSIONS Lack of enteral delivery of nutrients reduces numbers of T and IgA-producing cells, as well as mature DCs, in GALT of colon cancer patients, as it does in animal models. A close association between GALT changes and infectious complication morbidity was confirmed.
Collapse
|
22
|
Yang H, Jin Y, Wang CH, Tang CW. Effects of exogenous vasoactive intestinal peptide on mesenteric lymph pathway during early intestinal ischemia-reperfusion injury in rats. ACTA ACUST UNITED AC 2013; 186:36-42. [PMID: 23872373 DOI: 10.1016/j.regpep.2013.06.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 05/31/2013] [Accepted: 06/27/2013] [Indexed: 10/26/2022]
Abstract
Mesenteric lymph pathway serves as the primary route by which gut injury leads to systemic inflammation and distant organ injury. The inflammation of the intestinal tract is partially mediated by vasoactive intestinal peptide (VIP). Therefore, the aim of this study was to test whether exogenous VIP affects mesenteric lymph pathway during early intestinal ischemia-reperfusion (IIR) injury. Rats were randomized into control, control+VIP, IIR and IIR+VIP groups. The observation of mesenteric lymph flow was carried out by cannulation of mesenteric lymphatics. The distribution of in vivo lymphocyte trafficking was performed by (51)Cr labeled lymphocytes and was measured by γ-counter. Endotoxin concentration was assayed using the limulus test kit and TNF-α level was detected by ELISA. When IIR injury treated with VIP, the volumes of lymph flow increased by 80%, which caused the number of lymphocytes exiting in mesenteric lymphatic increased by 50% while the proportion of (51)Cr-lymphocytes in Peyer's patches, intestinal effector tissues, mesenteric nodes, large intestine, stomach decreased by 58%, 51%, 58%, 63%, 64% respectively at the 6th h after reperfusion following intestinal ischemia. Meanwhile, endotoxin and TNF-α levels in intestinal lymph decreased by 51% and 83%. These results suggest that exogenous VIP ameliorates IIR induced splanchnic organ damage via inhibition of toxic mediators reaching systemic circulation and reinforcement of the effective immune responses in gut-associated lymphoid tissues (GALT).
Collapse
Affiliation(s)
- Hui Yang
- Department of Gastroentrology, Nanjing Children's Hospital, Nanjing Medical University, 210008, China
| | | | | | | |
Collapse
|
23
|
Yang H, Jin Y, Wang C, Tang C. Role of mesenteric lymph pathway in the effects of exogenous somatostatin against early intestinal injury after ischemia-reperfusion in rats. Neuropeptides 2013; 47:237-43. [PMID: 23706875 DOI: 10.1016/j.npep.2013.04.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Revised: 04/19/2013] [Accepted: 04/22/2013] [Indexed: 12/22/2022]
Abstract
Intestinal ischemia-reperfusion (I/R)-induced gut injury remains a challenge for critically ill patients. This study aims to test whether mesenteric lymph pathway is involved in intestinal I/R injury and whether somatostatin (SST) affects mesenteric lymph pathway after mesenteric reperfusion. Intestinal I/R rats were treated with SST-14 by intravenous injection combined with intraperitoneal injection before occlusion of the SMA until the end of the experiment. When intestinal I/R injury treated with SST, the volumes of mesenteric lymph flow at the 6th h after reperfusion following intestinal ischemia were increased ([0.55±0.24] ml/h vs [0.25±0.09] ml/h, p<0.05) and the number of intestinal lymphocytes per milliliter ([2.30±0.72]×10⁷/ml vs [1.16±0.63]×10⁷/ml, p<0.05) was also increased, which caused the number of intestinal lymphocytes output at the same period of time was significantly increased compared with intestinal I/R group ([1.33±0.88]×10⁷/h vs [0.28±0.15]×10⁷/h, p<0.05). Meanwhile, the number of ⁵¹Cr-lymphocytes migration from systemic circulation to the effector sites in GALT was significantly increased ([1.93±0.23]×10⁵/h vs [0.90±0.25]×10⁵/h, p<0.05), although the percentage of ⁵¹Cr-lymphocytes in the effector sites ([1.45±0.26]% vs [3.23±1.69]%, p<0.05) was sharply decreased compared with intestinal I/R group. The accompanying decreases of the endotoxin concentration ([0.038±0.017] EU/mL vs [0.110±0.028] EU/mL, compared with intestinal I/R group p<0.05) and the TNF-α levels ([37.50±10.45] ρg/ml) vs ([74.93±14.77] ρg/ml), compared with intestinal I/R group p<0.05) in mesenteric lymph and the improvement of vital organ dysfunction happened during the early intestinal I/R injury. Suppression of gut-derived toxic mediators reaching systemic circulation and increases of the number of lymphocytes homing to the effector sites in GALT to strengthen the effective immune responses in intestinal mucosa account for the protective effects of exogenous SST against early intestinal I/R injury.
Collapse
Affiliation(s)
- Hui Yang
- Department of Gastroenterology, Nanjing Children's Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, PR China
| | | | | | | |
Collapse
|
24
|
Di Sabatino A, Brunetti L, Biancheri P, Ciccocioppo R, Guerci M, Casella C, Vidali F, MacDonald TT, Benazzo M, Corazza GR. Mucosal changes induced by ischemia-reperfusion injury in a jejunal loop transplanted in oropharynx. Intern Emerg Med 2013; 8:317-25. [PMID: 21553237 DOI: 10.1007/s11739-011-0615-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Accepted: 04/22/2011] [Indexed: 10/18/2022]
Abstract
Tissues exposed to ischemia and reperfusion develop an inflammatory response. We investigate the morphological and immunological changes occurring in the mucosa of a jejunal loop transplanted in the oropharynx of a man undergoing circular pharyngolaryngectomy. Jejunal biopsies were collected during the transplantation procedures (cold and warm ischemia, reperfusion), during the 7 post-operative days through an exteriorized jejunal segment for flap monitoring, and 45 days after transplantation through an upper endoscopy. Matrix metalloproteinase (MMP)-3 and MMP-12 increase was accompanied by a parallel rise in apoptotic enterocytes, and by a concomitant reduction of surface area to volume ratio and enterocyte height. Goblet cell hyperplasia is coupled with Paneth cell disappearance at the crypt base. CD8-positive intraepithelial lymphocytes initially decrease, then they increase in accordance with the peak of enterocyte apoptosis. We identified alterations in lymphocyte infiltration, mucosal architecture and epithelial cell turnover, which may give a window to mechanisms of small bowel ischemia-reperfusion in humans.
Collapse
Affiliation(s)
- Antonio Di Sabatino
- First Department of Medicine, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Piazzale Golgi 19, 27100, Pavia, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Jiang JW, Ren ZG, Chen LY, Jiang L, Xie HY, Zhou L, Zheng SS. Enteral supplementation with glycyl-glutamine improves intestinal barrier function after liver transplantation in rats. Hepatobiliary Pancreat Dis Int 2011; 10:380-385. [PMID: 21813386 DOI: 10.1016/s1499-3872(11)60064-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Most patients after liver transplantation (LT) suffer from intestinal barrier dysfunction. Glycyl-glutamine (Gly-Gln) by parenteral supplementation is hydrolyzed to release glutamine, which improves intestinal barrier function in intestinal injury. This study aimed to investigate the effect of Gly-Gln by enteral supplementation on intestinal barrier function in rats after allogenetic LT under immunosuppressive therapy. METHODS Twelve inbred Lewis rats were selected randomly as donors, and 24 inbred Brown Norway (BN) rats as recipients of allogenetic LT. The recipients were divided into a control group (Ala, n=12) and an experimental group (Gly-Gln, n=12). In each group, 6 normal BN rats were sampled for normal parameters on preoperative day 3. The 6 recipients in the control group received alanine (Ala) daily by gastric perfusion for 3 preoperative days and 7 postoperative days, and the 6 recipients in the experimental group were given Gly-Gln in the same manner. The 12 BN recipients underwent orthotopic LT under sterile conditions after a 3-day fast and were given immunosuppressive therapy for 7 days. They were harvested for sampling on postoperative day 8. The following parameters were assessed: intestinal mucosal protein content, mucosal ultrastructure, ileocecal sIgA content, portal plasma levels of endotoxin and TNF-alpha, and bacterial translocation. RESULTS All recipients were alive after LT. On preoperative day 3, all parameters were similar in the two groups. On postoperative day 8, all parameters in the two groups were remarkably changed from those on preoperative day 3. However, compared to the Ala group, supplementation with Gly-Gln increased the levels of intestinal mucosal protein and ileocecal sIgA, improved mucosal microvilli, and decreased portal plasma levels of endotoxin and TNF-alpha as well as bacterial translocation. CONCLUSION Enteral supplementation with Gly-Gln improved intestinal barrier function after allogenetic LT in rats.
Collapse
Affiliation(s)
- Jian-Wen Jiang
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health; Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | | | | | | | | | | | | |
Collapse
|
26
|
Murakami M, Sato N, Sato N, Nakamura T, Masunaga H. Changes in lymphocyte phenotypes and cytokine production by surgical stress in a rat small intestinal resection model. J Clin Biochem Nutr 2011; 40:216-20. [PMID: 18398499 PMCID: PMC2275767 DOI: 10.3164/jcbn.40.216] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2006] [Accepted: 11/07/2006] [Indexed: 11/22/2022] Open
Abstract
Small intestinal resection rats are used widely as a malabsorption model, but the immunological changes are unclear. We examined the changes in systemic and mucosal immune status after a small intestinal resection in rats with a controlled nutritional status. Rats had 60% of their small intestine removed. At 5 days after the surgery, spleen cells and intraepithelial lymphocytes (IEL) were isolated. The phenotypes of spleen cells and IEL, the production patterns of Th1 and Th2 cytokines, and the proinflammatory cytokine levels in the plasma were measured. CD4+ T cells in the blood and spleen were significantly decreased in the Resection group (p<0.05). In contrast, IEL subpopulations were not different between the two groups. Interferon-gamma production from the spleen cells was significantly decreased in the Resection group (p<0.05). Interleukin (IL)-4 production was not different between the two groups. Plasma IL-6 concentrations were significantly elevated in the Resection group 6 h after surgery (p<0.05). In conclusions, small intestinal resection in rats suppressed systemic immunity, and this model is useful as a surgical stress model.
Collapse
Affiliation(s)
- Mariko Murakami
- EN Otsuka Pharmaceutical Co., Ltd., Technical research laboratories, 4-3-5 Nimaibashi, Hanamaki, Iwate 025-0312, Japan
| | | | | | | | | |
Collapse
|
27
|
Dalfino L, Giglio MT, Puntillo F, Marucci M, Brienza N. Haemodynamic goal-directed therapy and postoperative infections: earlier is better. A systematic review and meta-analysis. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2011; 15:R154. [PMID: 21702945 PMCID: PMC3219028 DOI: 10.1186/cc10284] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Revised: 05/23/2011] [Accepted: 06/24/2011] [Indexed: 12/17/2022]
Abstract
Introduction Infectious complications are the main causes of postoperative morbidity. The early timing of their promoting factors is the rationale for perioperative strategies attempting to reduce them. Our aim was to determine the effects of perioperative haemodynamic goal-directed therapy on postoperative infection rates. Methods We performed a systematic review and meta-analysis. MEDLINE, EMBASE, The Cochrane Library and the DARE databases were searched up to March 2011. Randomised, controlled trials of major surgery in adult patients managed with perioperative goal-directed therapy or according to routine haemodynamic practice were included. Primary outcome measure was specific type of infection. Results Twenty-six randomised, controlled trials with a combined total of 4,188 participants met our inclusion criteria. Perioperative goal-directed therapy significantly reduced surgical site infections (pooled OR 0.58, 95% CI 0.46 to 0.74; P < 0.0001), pneumonia (pooled OR 0.71, 95% CI 0.55 to 0.92; P = 0.009), and urinary tract infections (pooled OR 0.44, 95% CI 0.22 to 0.84; P = 0.02). A significant benefit was found regarding total infectious episodes (OR 0.40, 95% CI 0.28 to 0.58; P < 0.00001). Conclusions Flow-directed haemodynamic therapy designed to optimise oxygen delivery protects surgical patients against postoperative hospital-acquired infections and must be strongly encouraged, particularly in the high-risk surgical population.
Collapse
Affiliation(s)
- Lidia Dalfino
- Anesthesia and Intensive Care Unit, Department of Emergency and Organ Transplantation, University of Bari, Piazza G, Cesare 11, I-70124 Bari, Italy
| | | | | | | | | |
Collapse
|
28
|
Gianotti L, Nespoli L, Rocchetti S, Vignali A, Nespoli A, Braga M. Gut oxygenation and oxidative damage during and after laparoscopic and open left-sided colon resection: a prospective, randomized, controlled clinical trial. Surg Endosc 2011; 25:1835-1843. [PMID: 21136109 DOI: 10.1007/s00464-010-1475-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Accepted: 10/13/2010] [Indexed: 01/15/2023]
Abstract
BACKGROUND Pneumoperitoneum (PP), established for laparoscopic (LPS) operation, has been associated with potential detrimental effects, such as mesenteric ischemia-reperfusion injury. The objective of the trial was to measure intestinal tissue oxygen pressure (PtiO2) and oxidative damage during laparoscopic (LPS) and open colon surgery and during the postoperative course. METHODS Forty patients candidate to left-sided colectomy were randomized to undergo open or LPS resection (20 patients/group). During the operation, PtiO2 was measured at established changes of PP pressure (from 0-15 mmHg) and for 6 days postoperatively. PtiO2 was determined by a polarographic microprobe implanted in the colon wall. Ischemia-reperfusion injury was assessed by plasma malondialdehyde (MDA). ClinicalTrial.gov registration number: NCT01040013. RESULTS LPS was associated with a higher PtiO2 at the beginning of surgery (73.9±9.4 vs. 64.3±6.4 in open; P=0.04) and at the end of the operation (57.7±7.9 vs. 53.1±4.7 in open; P=0.03). PtiO2 decreased significantly during mesentery traction vs. beginning in both groups (respectively 58.7±13.2 vs. 73.9±9.4 in LPS and 55.3±6.4 vs. 64.3±6.4 in open group; minimum P=0.02). During LPS, there was a significant decrease of PtiO2 only when PP was increased to 15 mmHg (63.2±7.5 vs. 76.6±10.7 at 10 mmHg; P=0.03). PtiO2 also was significantly better in the LPS group during the first 3 days after operation (minimum P=0.04 vs. open). MDA significantly increased in both groups after mesentery traction and at the end of operation vs. baseline levels with no difference between techniques. CONCLUSIONS LPS seems to be associated with a better intra- and postoperative PtiO2. High-pressure PP may impair PtiO2.
Collapse
Affiliation(s)
- Luca Gianotti
- Department of Surgery, San Gerardo Hospital (4° piano B), Milano-Bicocca University, Via Pergolesi 33, 20052, Monza, Italy.
| | | | | | | | | | | |
Collapse
|
29
|
The role of intestinal colonization with Gram-negative bacteria as a source for intensive care unit-acquired bacteremia*. Crit Care Med 2011; 39:961-6. [DOI: 10.1097/ccm.0b013e318208ee26] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
30
|
Fukatsu K, Tanabe K, Maeshima Y, Omata J, Yasuhara H, Saitoh D. Neutrophil elastase inhibitor restores gut ischemia reperfusion-induced impairment of gut immunity with reduced plasma interleukin-6 concentrations in mice. Surg Infect (Larchmt) 2011; 11:517-22. [PMID: 20969469 DOI: 10.1089/sur.2010.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Gut-associated lymphoid tissue (GALT) is regarded as central mucosa-associated lymphoid tissue that influences systemic mucosal immunity. Our previous study revealed that gut ischemia and reperfusion (I/R) reduces GALT lymphocyte numbers. Because gut hypoperfusion frequently occurs in trauma, shock, and surgery patients, establishment of a therapeutic method to preserve GALT mass after gut I/R may be important for the prevention of infections. We examined the effects of sivelestat sodium hydrate, a selective inhibitor of neutrophil elastase, on GALT mass and plasma cytokine concentrations in a murine gut I/R model. METHODS Seventy male ICR mice were randomized to the control (n = 34) or the sivelestat (n = 36) group. After intravenous cannulation of the animals, the superior mesenteric artery was occluded for 60 min. After reperfusion, physiologic saline or sivelestat 5 mg/kg hourly was infused for 24 h. Sixteen mice in the control and 22 in the sivelestat group were alive at 24 h. Twenty-six mice (n = 13 in each group) were chosen randomly for harvest of the small intestine. Lymphocytes from Peyer patches (PP), the intraepithelial space (IE), and the lamina propria (LP) were counted; and their phenotypes (αβT-cell receptor (TCR), γδTCR, CD4, CD8, B cell) were determined by flow cytometry. Cytokine concentrations (interleukin [IL]-6, IL-1β, IL-10) in the plasma and bronchoalveolar lavage fluid were measured by enzyme-linked immunosorbent assay. RESULTS Sivelestat treatment did not improve survival but increased PP and IE lymphocyte numbers significantly and reduced the LP CD8(+) cell percentage and plasma IL-6 concentration compared with controls. There were no significant differences between the two groups in other cell phenotypes or cytokine concentrations. CONCLUSION Sivelestat treatment after gut I/R may be useful for maintaining gut immunity and preventing systemic inflammatory responses.
Collapse
|
31
|
Varga J, Staško P, Tóth Š, Pristášová Z, Jonecová Z, Veselá J, Pomfy M. Morphological and apoptotic changes in the intestinal mucosa and lung parenchyma after ischaemic/reperfusion injury of the jejunum. Acta Vet Hung 2010; 58:243-56. [PMID: 20460223 DOI: 10.1556/avet.58.2010.2.10] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Ischaemic/reperfusion (IR) injury of the small intestine may lead to the development of multiple organ failure. Little is known about the morphological changes occurring in the organs during the subacute course of this syndrome. The objective of this study was to observe histopathological features and the role of apoptosis in the jejunal mucosa and lung parenchyma after intestinal IR injury in a long-term experiment. Wistar rats (n = 36) were divided into 4 experimental groups (IR(10), IR(20), IR(30), S). Groups IR(10), IR(20) and IR(30) (each n = 10) were subjected to 1-hour ischaemia of the cranial mesenteric artery followed by 10, 20 or 30 days of reperfusion, respectively. The control group S (n = 6) was not subjected to ischaemia. The jejunal mucosa remained intact after all periods of reperfusion. Apoptotic cells were found particularly in the lamina propria, with the most significant difference observed in the IR(30) group (P < 0.01). The lung parenchyma had lower regenerative capacity, which was confirmed by a high index of histological damage after 30 days of reperfusion (P < 0.01) and by the presence of an increased number of apoptotic cells, especially in the pulmonary interstitium. The number of apoptotic cells was ten times higher than in the control group (P < 0.001).
Collapse
Affiliation(s)
- Ján Varga
- 1 P. J. Šafárik University Department of Histology and Embryology, Faculty of Medicine Šrobárova 2 SK-04001 Košice Slovakia
| | - Pavel Staško
- 1 P. J. Šafárik University Department of Histology and Embryology, Faculty of Medicine Šrobárova 2 SK-04001 Košice Slovakia
| | - Štefan Tóth
- 1 P. J. Šafárik University Department of Histology and Embryology, Faculty of Medicine Šrobárova 2 SK-04001 Košice Slovakia
| | - Zuzana Pristášová
- 1 P. J. Šafárik University Department of Histology and Embryology, Faculty of Medicine Šrobárova 2 SK-04001 Košice Slovakia
| | - Zuzana Jonecová
- 1 P. J. Šafárik University Department of Histology and Embryology, Faculty of Medicine Šrobárova 2 SK-04001 Košice Slovakia
| | - Jarmila Veselá
- 1 P. J. Šafárik University Department of Histology and Embryology, Faculty of Medicine Šrobárova 2 SK-04001 Košice Slovakia
| | - Mikuláš Pomfy
- 1 P. J. Šafárik University Department of Histology and Embryology, Faculty of Medicine Šrobárova 2 SK-04001 Košice Slovakia
| |
Collapse
|
32
|
Arnalich F, Maldifassi MC, Ciria E, Quesada A, Codoceo R, Herruzo R, Garcia-Cerrada C, Montoya F, Vazquez JJ, López-Collazo E, Montiel C. Association of cell-free plasma DNA with perioperative mortality in patients with suspected acute mesenteric ischemia. Clin Chim Acta 2010; 411:1269-74. [PMID: 20478285 DOI: 10.1016/j.cca.2010.05.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2010] [Revised: 05/05/2010] [Accepted: 05/06/2010] [Indexed: 12/26/2022]
Abstract
BACKGROUND Diagnosing patients with acute mesenteric ischemia (AMI) in the emergency ward is challenging. This study assesses the usefulness of plasma DNA in patients with clinically suspected AMI. METHODS 130 consecutive patients who underwent laparotomy were studied. Cell-free plasma DNA was measured by real-time quantitative PCR assay for the beta-globin gene. The primary endpoint was the accuracy of plasma DNA for predicting 30-day mortality. RESULTS Surgery revealed AMI in 99 patients and alternative diagnoses in 31 patients. Forty-six patients with AMI died (46.6%) as compared to 6 (19.4%) in the non-AMI group (p<0.05). The DNA concentration at admission was significantly higher in patients with AMI (median 7340 GE/ml, versus, 2735 GE/ml, p<0.01) and in AMI patients who died (8830 GE/ml, versus 4970 GE/ml, p<0.05). The area under the ROC curves for plasma DNA as a marker for mesenteric ischemia and independent predictor for 30-day mortality were 0.708 (95% CI 0.701-0.890) and 0.815 (95% CI 0.735-0.894). Multiple logistic regression analysis showed that the risk of hospital mortality increased 1.52-fold for every 1000 GE/ml increase in plasma DNA. CONCLUSIONS Plasma DNA levels may be a useful biomarker in predicting the outcome of patients with AMI.
Collapse
Affiliation(s)
- Francisco Arnalich
- Emergency Medicine Service, Internal Medicine Department, Hospital Universitario La Paz, IdiPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Impact of selective decontamination of the digestive tract on multiple organ dysfunction syndrome: systematic review of randomized controlled trials. Crit Care Med 2010; 38:1370-6. [PMID: 20308882 DOI: 10.1097/ccm.0b013e3181d9db8c] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE We examined the impact of selective decontamination of the digestive tract on multiple organ dysfunction syndrome. DATA SOURCES We searched MEDLINE, EMBASE, the Cochrane Register of Controlled Trials, previous meta-analyses, and meetings proceedings. STUDY SELECTION We included all randomized trials comparing both oropharyngeal and intestinal administration of antibiotics in selective decontamination of the digestive tract with or without a parenteral component, with placebo or standard therapy used in the controls. DATA EXTRACTION Two reviewers independently applied selection criteria, performed quality assessment, and extracted the data. The primary end point was the number of patients with multiple organ dysfunction syndrome developing during intensive care unit stay. Secondary end points were overall mortality and multiple organ dysfunction syndrome-related mortality. Odds ratios were pooled with the random effect model. DATA SYNTHESIS We identified seven randomized trials including 1270 patients. Multiple organ dysfunction syndrome was found in 132 of 637 patients (20.7%) in the selective decontamination of the digestive tract group and in 219 of 633 patients (34.6%) in the control group (odds ratio, 0.50; 95% confidence interval, 0.34-0.74; p < .001). Overall mortality for selective decontamination of the digestive tract vs. control patients was 119 of 637 (18.7%) and 145 of 633 (22.9%), respectively, demonstrating a nonsignificant reduction in the odds of death (odds ratio, 0.82; 95% confidence interval, 0.51-1.32; p = .41). In five studies including 472 patients, multiple organ dysfunction syndrome-related mortality was demonstrated in 31 of 239 (13%) patients in selective decontamination of the digestive tract group and 37 of 233 (15.9%) in the controls (odds ratio, 0.84; 95% confidence interval, 0.48-1.41; p = .54). CONCLUSIONS Selective decontamination of the digestive tract reduces the number of patients with multiple organ dysfunction syndrome. Mortality was not significantly reduced, probably because of the small sample size.
Collapse
|
34
|
Kawasaki T, Kawasaki C, Sata T, Chaudry IH. Lidocaine suppresses mouse Peyer's Patch T cell functions and induces bacterial translocation. Surgery 2010; 149:106-13. [PMID: 20466400 DOI: 10.1016/j.surg.2010.03.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2009] [Accepted: 03/25/2010] [Indexed: 10/19/2022]
Abstract
BACKGROUND The gastrointestinal mucosa is an important route of entry for microbial pathogens. The immune cells of Peyer's patch (PP) compartments contribute to the active immune response against infection. Although local anesthetics are widely used in clinical practice, it remains unclear whether local anesthetics such as lidocaine affect PP T cell functions. METHODS The aim of this study was to examine if lidocaine has any effects on mouse PP T cell functions. To test this, freshly isolated mouse Peyer's patch T cells were incubated with lidocaine. The effects of lidocaine on concanavalin A-stimulated PP T cell proliferation and cytokine production were assessed. The effect of lidocaine on PP T cell mitogen-activated protein kinase (MAPK) activation was also assessed. RESULTS The results indicate that lidocaine suppresses cell proliferation, cytokine production, and MAPK activation in PP T cells. Furthermore, we found that the chronic in vivo exposure to lidocaine increases bacterial accumulation in PP. CONCLUSION The enhanced immunosuppressive effects of lidocaine on PP T cell functions could contribute to the host's enhanced susceptibility to infection.
Collapse
Affiliation(s)
- Takashi Kawasaki
- Center for Surgical Research and Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | |
Collapse
|
35
|
Kawasaki T, Suzuki T, Choudhry MA, Bland KI, Chaudry IH. Salutary effects of 17beta-estradiol on Peyer's patch T cell functions following trauma-hemorrhage. Cytokine 2010; 51:166-72. [PMID: 20400328 DOI: 10.1016/j.cyto.2010.03.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2010] [Accepted: 03/30/2010] [Indexed: 01/16/2023]
Abstract
Although 17beta-estradiol (E2) administration following trauma-hemorrhage (T-H) improves immune functions in male rodents, it remains unclear whether E2 has salutary effects on Peyer's patch (PP) T cell functions. We hypothesized that T-H induces PP T cell dysfunction and E2 administration following T-H will improve PP T cell function. T-H was induced in male C3H/HeN mice (6-8weeks) by midline laparotomy and approximately 90min of hemorrhagic shock (blood pressure 35mmHg), followed by fluid resuscitation (4x the shed blood volume in the form of Ringer's lactate). Estrogen receptor (ER)-alpha agonist propyl pyrazole triol (PPT; 5microg/kg), ER-beta agonist diarylpropionitrile (DPN; 5microg/kg), E2 (50microg/kg), or vehicle was injected subcutaneously at resuscitation onset. Two hours later, mice were sacrificed and PP T cells isolated. PP T cell capacity to produce cytokines in response to in vitro stimulation, PP T cell proliferation and MAPK (p38, ERK-1/2, JNK) activation were measured. Results indicate PP T cell proliferation, cytokine production and MAPK activation decreased significantly following T-H. E2, PPT or DPN administration normalized these parameters. Since PPT or DPN administration following T-H was effective in normalizing PP T cell functions, the salutary effects of E2 are mediated via ER-alpha and ER-beta.
Collapse
Affiliation(s)
- Takashi Kawasaki
- Center for Surgical Research and Department of Surgery, University of Alabama at Birmingham, G094 Volker Hall, 1670 University Boulevard, Birmingham, AL 35294, USA
| | | | | | | | | |
Collapse
|
36
|
Grootjans J, Lenaerts K, Derikx JPM, Matthijsen RA, de Bruïne AP, van Bijnen AA, van Dam RM, Dejong CHC, Buurman WA. Human intestinal ischemia-reperfusion-induced inflammation characterized: experiences from a new translational model. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:2283-91. [PMID: 20348235 DOI: 10.2353/ajpath.2010.091069] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human intestinal ischemia-reperfusion (IR) is a frequent phenomenon carrying high morbidity and mortality. Although intestinal IR-induced inflammation has been studied extensively in animal models, human intestinal IR induced inflammatory responses remain to be characterized. Using a newly developed human intestinal IR model, we show that human small intestinal ischemia results in massive leakage of intracellular components from ischemically damaged cells, as indicated by increased arteriovenous concentration differences of intestinal fatty acid binding protein and soluble cytokeratin 18. IR-induced intestinal barrier integrity loss resulted in free exposure of the gut basal membrane (collagen IV staining) to intraluminal contents, which was accompanied by increased arteriovenous concentration differences of endotoxin. Western blot for complement activation product C3c and immunohistochemistry for activated C3 revealed complement activation after IR. In addition, intestinal IR resulted in enhanced tissue mRNA expression of IL-6, IL-8, and TNF-alpha, which was accompanied by IL-6 and IL-8 release into the circulation. Expression of intercellular adhesion molecule-1 was markedly increased during reperfusion, facilitating influx of neutrophils into IR-damaged villus tips. In conclusion, this study for the first time shows the sequelae of human intestinal IR-induced inflammation, which is characterized by complement activation, production and release of cytokines into the circulation, endothelial activation, and neutrophil influx into IR-damaged tissue.
Collapse
Affiliation(s)
- Joep Grootjans
- Department of Surgery, NUTRIM School for Nutrition, Toxicology, and Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Derikx JPM, Matthijsen RA, de Bruïne AP, van Dam RM, Buurman WA, Dejong CHC. A new model to study intestinal ischemia-reperfusion damage in man. J Surg Res 2009; 166:222-6. [PMID: 20070978 DOI: 10.1016/j.jss.2009.09.046] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2009] [Revised: 08/31/2009] [Accepted: 09/30/2009] [Indexed: 12/13/2022]
Abstract
BACKGROUND This report describes a human in vivo ischemia reperfusion (IR) model of the small intestine. Animal models of intestinal IR are indispensable for our understanding of sequelae of IR induced organ damage. However, a functional experimental IR model of the human small intestine, allowing for translational research, can be considered critical for our pathophysiologic understanding of intestinal IR in man. MATERIALS AND METHODS Patients with a healthy gut undergoing abdominal surgery with a Roux-Y or similar reconstruction were included, creating the opportunity to study IR of an isolated jejunal segment in a harmless model. RESULTS Ischemia was induced by nontraumatic vascular clamping followed by reperfusion. This model can be adapted using variable ischemia and reperfusion times. Similarly, tissue and plasma can be collected at any given time point during ischemia until end of reperfusion, only determined by progress of the original, intended surgical procedure. CONCLUSION A unique and harmless human IR model of the jejunum was created, which enables the study of acute damage to the epithelial lining and its subsequent repair mechanisms.
Collapse
Affiliation(s)
- Joep P M Derikx
- Department of Surgery, University Hospital Maastricht and Nutrition and Toxicology Research Institute, Maastricht University, Maastricht, The Netherlands
| | | | | | | | | | | |
Collapse
|
38
|
Accumulation of DC in Lamina Propria Induced by FMS-Like Tyrosine Kinase 3 Ligand Aggravates the Intestinal Inflammatory Response During Endotoxemia. Inflammation 2009; 33:34-45. [DOI: 10.1007/s10753-009-9156-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
39
|
|
40
|
Kulik L, Fleming SD, Moratz C, Reuter JW, Novikov A, Chen K, Andrews KA, Markaryan A, Quigg RJ, Silverman GJ, Tsokos GC, Holers VM. Pathogenic natural antibodies recognizing annexin IV are required to develop intestinal ischemia-reperfusion injury. THE JOURNAL OF IMMUNOLOGY 2009; 182:5363-73. [PMID: 19380783 DOI: 10.4049/jimmunol.0803980] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Intestinal ischemia-reperfusion (IR) injury is initiated when natural IgM Abs recognize neo-epitopes that are revealed on ischemic cells. The target molecules and mechanisms whereby these neo-epitopes become accessible to recognition are not well understood. Proposing that isolated intestinal epithelial cells (IEC) may carry IR-related neo-epitopes, we used in vitro IEC binding assays to screen hybridomas created from B cells of unmanipulated wild-type C57BL/6 mice. We identified a novel IgM mAb (mAb B4) that reacted with the surface of IEC by flow cytometric analysis and was alone capable of causing complement activation, neutrophil recruitment and intestinal injury in otherwise IR-resistant Rag1(-/-) mice. mAb B4 was found to specifically recognize mouse annexin IV. Preinjection of recombinant annexin IV blocked IR injury in wild-type C57BL/6 mice, demonstrating the requirement for recognition of this protein to develop IR injury in the context of a complex natural Ab repertoire. Humans were also found to exhibit IgM natural Abs that recognize annexin IV. These data in toto identify annexin IV as a key ischemia-related target Ag that is recognized by natural Abs in a pathologic process required in vivo to develop intestinal IR injury.
Collapse
Affiliation(s)
- Liudmila Kulik
- Department of Medicine, University of Colorado Denver School of Medicine, Denver, CO 80045, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Fan J, Xie Y, Li X, Guo G, Meng Q, Xiu Y, Li T, Feng W, Ma L. The influence of Peyer's patch apoptosis on intestinal mucosal immunity in burned mice. Burns 2009; 35:687-94. [PMID: 19269747 DOI: 10.1016/j.burns.2008.10.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2008] [Accepted: 10/30/2008] [Indexed: 10/21/2022]
Abstract
The aim of this study was to investigate the influence of apoptosis on Peyer's patches and the intestinal immunoglobulin A (IgA) response in burned mice. Sixty male Balb/c mice were randomly assigned into the sham-burn (control) group (n=30) and the burn group (n=30). The mice in the burn group received a full-thickness scald burn over 20% of the total body surface area (TBSA), on the back. At 12, 24 and 72 h, respectively, after injury, the burned mice (n=10, at every time point) were anaesthetised and their entire intestines were collected. The mice in the sham-burn group were treated with the same procedure as above, except for the burn injury. The number of Peyer's patches on every entire intestine and the total Peyer's patches cell yield were counted. The changes of lymphocyte subpopulations in Peyer's patches were measured by flow cytometry (FCM). And the levels of intestinal IgA were examined by enzyme-linked immunosorbent assay (ELISA). Fluoresceinisothiocyanate (FITC)-conjugated Annexin-tau and propidium iodide (PI) double-staining cells were analysed by FCM for apoptotic ratio in Peyer's patches. The results showed that the total Peyer's patch cell yield and the numbers of CD3, CD4, CD8 and CD19 cells were significantly decreased at 12, 24 and 72 h after injury (P<0.05), and that the intestinal IgA levels were markedly reduced at 24 and 72 h (P<0.05). On the other hand, total apoptotic ratio and all cell subpopulation apoptosis in Peyer's patches were dramatically increased at 12, 24 and 72 h after injury (P<0.05). These results indicated that severe burns led to a significant decrease in the number of Peyer's patch cells and in intestinal IgA levels, which was closely associated with strongly increased apoptosis in Peyer's patches.
Collapse
Affiliation(s)
- Jun Fan
- Department of Burns, Northern Hospital, 83 Wenhua Road, Shenyang 110015, Liaoning, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Effects of enteral nutrition supplemented with glutamine on intestinal mucosal immunity in burned mice. Nutrition 2009; 25:233-9. [DOI: 10.1016/j.nut.2008.08.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2008] [Revised: 08/11/2008] [Accepted: 08/30/2008] [Indexed: 12/27/2022]
|
43
|
Abstract
OBJECTIVES To review the epidemiology of pediatric multiple organ dysfunction syndrome (MODS) and summarize current concepts regarding the pathophysiology of shock, organ dysfunction, and nosocomial infections in this population. DATA SOURCE A MEDLINE-based literature search using the keywords MODS and child, without any restriction to the idiom. MAIN RESULTS Critically ill children may frequently develop multisystemic manifestations during the course of severe infections, multiple trauma, surgery for congenital heart defects, or transplantations. Descriptive scores to estimate the severity of pediatric MODS have been validated. Young age and chronic health conditions have also been recognized as important contributors to the development of MODS. Unbalanced inflammatory processes and activation of coagulation may lead to the development of capillary leak and acute respiratory distress syndrome. Neuroendocrine and metabolic responses may result in insufficient adaptive immune response and the development of nosocomial infections, which may further threaten host homeostasis. CONCLUSIONS Over the last 20 yrs, there has been an increasing knowledge on the epidemiology of pediatric MODS and on the physiologic mechanisms involved in the genesis of organ dysfunction. Nevertheless, further studies are needed to more clearly evaluate what is the long-term outcome of pediatric MODS.
Collapse
|
44
|
Derikx JPM, Matthijsen RA, de Bruïne AP, van Bijnen AA, Heineman E, van Dam RM, Dejong CHC, Buurman WA. Rapid reversal of human intestinal ischemia-reperfusion induced damage by shedding of injured enterocytes and reepithelialisation. PLoS One 2008; 3:e3428. [PMID: 18927609 PMCID: PMC2561292 DOI: 10.1371/journal.pone.0003428] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2008] [Accepted: 09/22/2008] [Indexed: 02/07/2023] Open
Abstract
Background Intestinal ischemia-reperfusion (IR) is a phenomenon related to physiological conditions (e.g. exercise, stress) and to pathophysiological events (e.g. acute mesenteric ischemia, aortic surgery). Although intestinal IR has been studied extensively in animals, results remain inconclusive and data on human intestinal IR are scarce. Therefore, an experimental harmless model for human intestinal IR was developed, enabling us to clarify the sequelae of human intestinal IR for the first time. Methods and Findings In 30 patients undergoing pancreatico-duodenectomy we took advantage of the fact that in this procedure a variable length of jejunum is removed. Isolated jejunum (5 cm) was subjected to 30 minutes ischemia followed by reperfusion. Intestinal Fatty Acid Binding Protein (I-FABP) arteriovenous concentration differences across the bowel segment were measured before and after ischemia to assess epithelial cell damage. Tissue sections were collected after ischemia and at 25, 60 and 120 minutes reperfusion and stained with H&E, and for I-FABP and the apoptosis marker M30. Bonferroni's test was used to compare I-FABP differences. Mean (SEM) arteriovenous concentration gradients of I-FABP across the jejunum revealed rapidly developing epithelial cell damage. I-FABP release significantly increased from 290 (46) pg/ml before ischemia towards 3,997 (554) pg/ml immediately after ischemia (p<0.001) and declined gradually to 1,143 (237) pg/ml within 1 hour reperfusion (p<0.001). Directly after ischemia the intestinal epithelial lining was microscopically normal, while subepithelial spaces appeared at the villus tip. However, after 25 minutes reperfusion, enterocyte M30 immunostaining was observed at the villus tip accompanied by shedding of mature enterocytes into the lumen and loss of I-FABP staining. Interestingly, within 60 minutes reperfusion the epithelial barrier resealed, while debris of apoptotic, shedded epithelial cells was observed in the lumen. At the same time, M30 immunoreactivity was absent in intact epithelial lining. Conclusions This is the first human study to clarify intestinal IR induced cell damage and repair and its direct consequences. It reveals a unique, endogenous clearing mechanism for injured enterocytes: rapid detachment of damaged apoptotic enterocytes into the lumen. This process is followed by repair of the epithelial continuity within an hour, resulting in a normal epithelial lining.
Collapse
Affiliation(s)
- Joep P. M. Derikx
- Department of Surgery, School for Nutrition & Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, the Netherlands
- * E-mail:
| | - Robert A. Matthijsen
- Department of Surgery, School for Nutrition & Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Adriaan P. de Bruïne
- Department of Pathology, Cardiovascular Research Institute (CARIM), Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Annemarie A. van Bijnen
- Department of Surgery, School for Nutrition & Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Erik Heineman
- Department of Surgery, School for Nutrition & Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Ronald M. van Dam
- Department of Surgery, School for Nutrition & Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Cornelis H. C. Dejong
- Department of Surgery, School for Nutrition & Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Wim A. Buurman
- Department of Surgery, School for Nutrition & Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, the Netherlands
| |
Collapse
|
45
|
Detrimental Effects of Early Nutrition Administration After Severe Gut Ischemia-Reperfusion. J Surg Res 2008; 149:31-8. [DOI: 10.1016/j.jss.2007.12.749] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2007] [Revised: 10/25/2007] [Accepted: 12/06/2007] [Indexed: 11/22/2022]
|
46
|
|
47
|
Clark JA, Coopersmith CM. Intestinal crosstalk: a new paradigm for understanding the gut as the "motor" of critical illness. Shock 2008; 28:384-93. [PMID: 17577136 PMCID: PMC2084394 DOI: 10.1097/shk.0b013e31805569df] [Citation(s) in RCA: 345] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
For more than 20 years, the gut has been hypothesized to be the "motor" of multiple organ dysfunction syndrome. As critical care research has evolved, there have been multiple mechanisms by which the gastrointestinal tract has been proposed to drive systemic inflammation. Many of these disparate mechanisms have proved to be important in the origin and propagation of critical illness. However, this has led to an unusual situation where investigators describing the gut as a "motor" revving the systemic inflammatory response syndrome are frequently describing wholly different processes to support their claim (i.e., increased apoptosis, altered tight junctions, translocation, cytokine production, crosstalk with commensal bacteria, etc). The purpose of this review is to present a unifying theory as to how the gut drives critical illness. Although the gastrointestinal tract is frequently described simply as "the gut," it is actually made up of (1) an epithelium; (2) a diverse and robust immune arm, which contains most of the immune cells in the body; and (3) the commensal bacteria, which contain more cells than are present in the entire host organism. We propose that the intestinal epithelium, the intestinal immune system, and the intestine's endogenous bacteria all play vital roles driving multiple organ dysfunction syndrome, and the complex crosstalk between these three interrelated portions of the gastrointestinal tract is what cumulatively makes the gut a "motor" of critical illness.
Collapse
Affiliation(s)
- Jessica A Clark
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | | |
Collapse
|
48
|
Berger MM, Soguel Alexander L, Chioléro RL. Gut Absorption Capacity in the Critically III. Intensive Care Med 2007. [DOI: 10.1007/978-0-387-49518-7_56] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
49
|
Milano PM, Douillet CD, Riesenman PJ, Robinson WP, Beidler SK, Zarzaur BL, Rich PB. Intestinal ischemia-reperfusion injury alters purinergic receptor expression in clinically relevant extraintestinal organs. J Surg Res 2007; 145:272-8. [PMID: 17688885 PMCID: PMC2323452 DOI: 10.1016/j.jss.2007.03.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2007] [Revised: 03/01/2007] [Accepted: 03/13/2007] [Indexed: 01/21/2023]
Abstract
BACKGROUND Intestinal ischemia-reperfusion (IIR) injury is known to initiate the systemic inflammatory response syndrome, which often progresses to multiple organ failure. We investigated changes in purinoceptor expression in clinically relevant extra-intestinal organs following IIR injury. MATERIALS AND METHODS Anesthetized adult male BalbC mice were randomized to sham laparotomy (control, n = 5), or 15 min of superior mesenteric artery occlusion. Experimental ischemia was followed by a period of reperfusion [1 min (n = 6) or 1 h (n = 6)]. Mice were then sacrificed and lung, kidney, and intestinal tissues were harvested. Following RNA extraction, purinoceptor mRNA expression for P2Y2, A3, P2X7, A2b, P2Y4, and P2Y6 was analyzed using real-time RT-PCR. RESULTS Significant differences in purinoceptor expression were observed in the lungs and kidneys of mice exposed to IIR injury when compared to controls. Pulmonary P2Y2 receptor expression was increased in the 1 h IIR group when compared to control, while pulmonary A3 receptor expression was incrementally elevated following IIR injury. In the kidney, P2Y2 receptor expression was increased in the 1 h IIR group compared to both 1 min IIR and control, and A3 receptor expression was decreased in the 1 h IIR group compared to the 1 min IIR group. No significant changes were observed in the intestinal purinoceptor profiles. CONCLUSION Purinoceptor expression is altered in the murine lung and kidney, but not intestine following experimental IIR injury. These findings may implicate extracellular nucleotides and purinoceptors as possible mediators of the extra-intestinal organ dysfunction associated with IIR injury.
Collapse
Affiliation(s)
- Peter M. Milano
- Department of Surgery, Division of Trauma and Critical Care. The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Christelle D. Douillet
- Department of Surgery, Division of Trauma and Critical Care. The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Paul J. Riesenman
- Department of Surgery, Division of Trauma and Critical Care. The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - William P. Robinson
- Department of Surgery, Division of Trauma and Critical Care. The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Stephanie K. Beidler
- Department of Surgery, Division of Trauma and Critical Care. The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Ben L. Zarzaur
- Department of Surgery, Division of Trauma and Critical Care. The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Preston B. Rich
- Department of Surgery, Division of Trauma and Critical Care. The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
50
|
Ikezawa F, Fukatsu K, Moriya T, Maeshima Y, Okamoto K, Hara E, Hiraide H, Compher CW. Albumin infusion after reperfusion prevents gut ischemia-reperfusion-induced gut-associated lymphoid tissue atrophy. JPEN J Parenter Enteral Nutr 2006; 30:380-6; discussion 386-7. [PMID: 16931605 DOI: 10.1177/0148607106030005380] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Our recent study clarified that gut ischemia-reperfusion (I/R) causes gut-associated lymphoid tissue (GALT) mass atrophy, a possible mechanism for increased morbidity of infectious complications after severe surgical insults. Because albumin administration reportedly reduces hemorrhagic shock-induced lung injury, we hypothesized that albumin treatment prevents GALT atrophy due to gut I/R. METHODS Male mice (n = 37) were randomized to albumin, normal saline, and sham groups. All groups underwent jugular vein catheter insertion. The albumin and normal saline groups underwent 75-minute occlusion of the superior mesenteric artery. During gut ischemia, all mice received normal saline infusions at 1.0 mL/h. The albumin group was given 5% bovine serum albumin in normal saline at 1.0 mL/h for 60 minutes after reperfusion, whereas the normal saline group received 0.9% sodium chloride at 1.0 mL/h. The sham group underwent laparotomy only. Mice were killed on day 1 or 7, and the entire small intestine was harvested. GALT lymphocytes were isolated and counted. Their phenotypes (alphabetaTCR, gammadeltaTCR, CD4, CD8, B220) were determined by flow cytometry. RESULTS On day 1, the gut I/R groups showed significantly lower total lymphocyte and B cell numbers in Peyer's patches and the lamina propria than the sham group. However, the albumin infusion partially but significantly restored these cell numbers. On day 7, there were no significant differences in any of the parameters measured among the 3 groups. CONCLUSIONS Albumin infusion after a gut ischemic insult may maintain gut immunity by preventing GALT atrophy.
Collapse
Affiliation(s)
- Fumie Ikezawa
- Division of Basic Traumatology, National Defense Medical College Research Institute, Tokorozawa, Japan
| | | | | | | | | | | | | | | |
Collapse
|