1
|
Mao QL, Yu ZH, Nie L, Wang FX, Dong YH, Qi XF. Gastrointestinal injury in cardiopulmonary bypass: current insights and future directions. Front Pharmacol 2025; 16:1542995. [PMID: 40356958 PMCID: PMC12067416 DOI: 10.3389/fphar.2025.1542995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
Cardiopulmonary bypass (CPB) is an essential component of cardiac surgery. As CPB technology continues to advance and innovate, it has enabled the expansion of surgical boundaries and the resolution of many previously inoperable challenges. However, the occurrence of various complications during CPB warrants attention, with their prevention and management being paramount. The gastrointestinal tract, directly connected to the external environment, is vulnerable not only to external factors but also to internal changes that may induce damage. Both preclinical and clinical research have demonstrated the incidence of gastrointestinal injuries following CPB, often accompanied by dysbiosis and abnormal metabolic outputs. Currently, interventions addressing gastrointestinal injuries following CPB remain insufficient. Although recent years have not seen notable progress in this field, emerging academic research underscores the essential role of the gut microbiome and its metabolic products in sustaining overall health and internal equilibrium. Notably, their significance as the body's "second genome" is increasingly recognized. Consequently, reevaluating the gastrointestinal damage post-CPB, alongside the associated dysbiosis and metabolic disturbances, is imperative. This reassessment carries substantial theoretical and practical implications for enhancing treatment strategies and bettering patient outcomes after CPB. This review aims to deliver a comprehensive synthesis of the latest preclinical and clinical research on CPB, address current challenges and gaps, and explore potential future research directions.
Collapse
Affiliation(s)
- Qi-Long Mao
- Department of Anesthesiology, Bazhong Central Hospital, Bazhong, Sichuan, China
| | - Zi-Hang Yu
- Department of Anesthesiology, Fushun County People’s Hospital, Zigong, Sichuan, China
| | - Liang Nie
- Department of Anesthesiology, Fushun County People’s Hospital, Zigong, Sichuan, China
| | - Fei-Xiang Wang
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Yu-Hui Dong
- Department of Anesthesiology, Bazhong Central Hospital, Bazhong, Sichuan, China
| | - Xiao-Fei Qi
- Department of Anesthesiology, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong, China
- Department of Anesthesiology, Women and Children’s Medical Center, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong, China
| |
Collapse
|
2
|
Wang M, Wang Z, Li Z, Qu Y, Zhao J, Wang L, Zhou X, Xu Z, Zhang D, Jiang P, Fan B, Liu Y. Targeting programmed cell death in inflammatory bowel disease through natural products: New insights from molecular mechanisms to targeted therapies. Phytother Res 2025; 39:1776-1807. [PMID: 38706097 DOI: 10.1002/ptr.8216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 03/14/2024] [Accepted: 04/11/2024] [Indexed: 05/07/2024]
Abstract
Inflammatory bowel disease (IBD) is an autoimmune disorder primarily characterized by intestinal inflammation and recurrent ulceration, leading to a compromised intestinal barrier and inflammatory infiltration. This disorder's pathogenesis is mainly attributed to extensive damage or death of intestinal epithelial cells, along with abnormal activation or impaired death regulation of immune cells and the release of various inflammatory factors, which contribute to the inflammatory environment in the intestines. Thus, maintaining intestinal homeostasis hinges on balancing the survival and functionality of various cell types. Programmed cell death (PCD) pathways, including apoptosis, pyroptosis, autophagy, ferroptosis, necroptosis, and neutrophil extracellular traps, are integral in the pathogenesis of IBD by mediating the death of intestinal epithelial and immune cells. Natural products derived from plants, fruits, and vegetables have shown potential in regulating PCD, offering preventive and therapeutic avenues for IBD. This article reviews the role of natural products in IBD treatment by focusing on targeting PCD pathways, opening new avenues for clinical IBD management.
Collapse
Affiliation(s)
- Mengjie Wang
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhiyuan Wang
- People's Hospital of Zhengzhou, Zhengzhou, China
| | - Zhichao Li
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuan Qu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiting Zhao
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lei Wang
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xinpeng Zhou
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ziqi Xu
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Di Zhang
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ping Jiang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Bing Fan
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ying Liu
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
3
|
Zhang M, Li S, Ying J, Qu Y. Neutrophils: a key component in ECMO-related acute organ injury. Front Immunol 2024; 15:1432018. [PMID: 39346902 PMCID: PMC11427252 DOI: 10.3389/fimmu.2024.1432018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/27/2024] [Indexed: 10/01/2024] Open
Abstract
Extracorporeal membrane oxygenation (ECMO), as an extracorporeal life support technique, can save the lives of reversible critically ill patients when conventional treatments fail. However, ECMO-related acute organ injury is a common complication that increases the risk of death in critically ill patients, including acute kidney injury, acute brain injury, acute lung injury, and so on. In ECMO supported patients, an increasing number of studies have shown that activation of the inflammatory response plays an important role in the development of acute organ injury. Cross-cascade activation of the complement system, the contact system, and the coagulation system, as well as the mechanical forces of the circuitry are very important pathophysiological mechanisms, likely leading to neutrophil activation and the production of neutrophil extracellular traps (NETs). NETs may have the potential to cause organ damage, generating interest in their study as potential therapeutic targets for ECMO-related acute organ injury. Therefore, this article comprehensively summarized the mechanism of neutrophils activation and NETs formation following ECMO treatment and their actions on acute organ injury.
Collapse
Affiliation(s)
- Mingfu Zhang
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Chronobiology (National Health Commission), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Shiping Li
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Chronobiology (National Health Commission), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Junjie Ying
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Chronobiology (National Health Commission), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yi Qu
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Chronobiology (National Health Commission), West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Ludwig EK, Abraham N, Schaaf CR, McKinney CA, Freund J, Stewart AS, Veerasammy BA, Thomas M, Cardona DM, Garman K, Barbas AS, Sudan DL, Gonzalez LM. Comparison of the effects of normothermic machine perfusion and cold storage preservation on porcine intestinal allograft regenerative potential and viability. Am J Transplant 2024; 24:564-576. [PMID: 37918482 PMCID: PMC11082874 DOI: 10.1016/j.ajt.2023.10.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/27/2023] [Accepted: 10/28/2023] [Indexed: 11/04/2023]
Abstract
Intestinal transplantation (IT) is the final treatment option for intestinal failure. Static cold storage (CS) is the standard preservation method used for intestinal allografts. However, CS and subsequent transplantation induce ischemia-reperfusion injury (IRI). Severe IRI impairs epithelial barrier function, including loss of intestinal stem cells (ISC), critical to epithelial regeneration. Normothermic machine perfusion (NMP) preservation of kidney and liver allografts minimizes CS-associated IRI; however, it has not been used clinically for IT. We hypothesized that intestine NMP would induce less epithelial injury and better protect the intestine's regenerative ability when compared with CS. Full-length porcine jejunum and ileum were procured, stored at 4 °C, or perfused at 34 °C for 6 hours (T6), and transplanted. Histology was assessed following procurement (T0), T6, and 1 hour after reperfusion. Real-time quantitative reverse transcription polymerase chain reaction, immunofluorescence, and crypt culture measured ISC viability and proliferative potential. A greater number of NMP-preserved intestine recipients survived posttransplant, which correlated with significantly decreased tissue injury following 1-hour reperfusion in NMP compared with CS samples. Additionally, ISC gene expression, spheroid area, and cellular proliferation were significantly increased in NMP-T6 compared with CS-T6 intestine. NMP appears to reduce IRI and improve graft regeneration with improved ISC viability and proliferation.
Collapse
Affiliation(s)
- Elsa K Ludwig
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Nader Abraham
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Cecilia R Schaaf
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Caroline A McKinney
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - John Freund
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Amy S Stewart
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Brittany A Veerasammy
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Mallory Thomas
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Diana M Cardona
- Department of Pathology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Katherine Garman
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Andrew S Barbas
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Debra L Sudan
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA.
| | - Liara M Gonzalez
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA.
| |
Collapse
|
5
|
Govender K, Jani VP, Cabrales P. The Disconnect Between Extracorporeal Circulation and the Microcirculation: A Review. ASAIO J 2022; 68:881-889. [PMID: 35067580 DOI: 10.1097/mat.0000000000001618] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Extracorporeal circulation (ECC) procedures, such as cardiopulmonary bypass (CPB) and extracorporeal membrane oxygenation (ECMO), take over the function of one or more organs, providing clinicians time to treat underlying pathophysiological conditions. ECMO and CPB carry significant mortality rates for patients, despite prior decades of research focused on the resulting failure of critical organs. Since the focus of these procedures is to support blood flow and provide oxygen-rich blood to tissues, a shift in research toward the effects of ECMO and CPB on the microcirculation is warranted. Along with provoking systemic responses, both procedures disrupt the integrity of red blood cells, causing release of hemoglobin (Hb) from excessive foreign surface contact and mechanical stresses. The effects of hemolysis are especially pronounced in the microcirculation, where plasma Hb leads to nitric oxide scavenging, oxidization, formation of reactive oxygen species, and inflammatory responses. A limited number of studies have investigated the implications of ECMO in the microcirculation, but more work is needed to minimize ECMO-induced reduction of microcirculatory perfusion and consequently oxygenation. The following review presents existing information on the implications of ECMO and CPB on microvascular function and proposes future studies to understand and leverage key mechanisms to improve patient outcomes.
Collapse
Affiliation(s)
- Krianthan Govender
- From the Functional Cardiovascular Engineering Laboratory, University of California, San Diego, La Jolla, California
| | | | | |
Collapse
|
6
|
Visvalingam R, Ridley E, Barnett A, Rahman T, Fraser JF. Nutrition adequacy, gastrointestinal and hepatic function during extracorporeal membrane oxygenation in critically ill adults: a retrospective observational study. Artif Organs 2022; 46:1886-1892. [PMID: 35451130 DOI: 10.1111/aor.14269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 03/17/2022] [Accepted: 04/14/2022] [Indexed: 11/28/2022]
Abstract
AIMS To identify clinical and biochemical markers associated with nutrition adequacy and gastrointestinal and liver dysfunction in adults on extracorporeal membrane oxygenation (ECMO). METHODS A retrospective, observational, study was conducted at 2 centres in Australia. Adult patients who received ECMO from July 2011 to June 2015 were included. Mode of ECMO used, fluid balance, number of systemic inflammatory response syndrome (SIRS) criteria present, vasoactive-inotropic scores (VIS) and liver function tests (LFTs) were collected for the duration of ECMO until 7 days after ECMO cessation. Multiple regression models were used to determine if the collected variables were associated with nutrition adequacy. The mean LFTs during ECMO were also compared to mean LFTs post ECMO cessation. RESULTS During the first 5 days of ECMO commencement, mean nutrition adequacy was 10% higher in the veno-venous (VV) ECMO group than in the veno-arterial (VA) group (95% confidence interval [CI], 2% to 17%). For every 5,000 ml increase of fluid balance, an associated decrease in nutrition adequacy was observed (-8%, 95% CI, -15% to -2%). A doubling of bilirubin and VIS were associated with a mean reduction in nutrition adequacy of -5% (CI -8% to -2%) and -2% (CI, -3% to -1%), respectively. CONCLUSIONS In the first 5 days of ECMO commencement, higher nutrition adequacy was associated with the VV mode of ECMO and reduced nutrition adequacy with increased fluid balance, more vasopressor and inotropic support and raised bilirubin. Prospective investigation is required to confirm whether these associations have a causal relationship.
Collapse
Affiliation(s)
- R Visvalingam
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Australia
| | - E Ridley
- Australian and New Zealand Intensive Care Research Centre, School of Public Health and Preventative Medicine, Monash University, Melbourne, Australia.,Nutrition Department, The Alfred Hospital, Melbourne, Australia
| | - A Barnett
- Australian Centre for Health Services Innovation and Centre for Healthcare Transformation, School of Public Health & Social Work, Queensland University of Technology, Brisbane, Australia
| | - T Rahman
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Australia.,Gastroenterology and Hepatology, The Prince Charles Hospital, Brisbane, Australia
| | - J F Fraser
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Australia.,Adult Intensive Care Services, The Prince Charles Hospital, Brisbane, Australia.,Faculty of Medicine, University of Queensland, Brisbane, Australia
| |
Collapse
|
7
|
Khashu M, Dame C, Lavoie PM, De Plaen IG, Garg PM, Sampath V, Malhotra A, Caplan MD, Kumar P, Agrawal PB, Buonocore G, Christensen RD, Maheshwari A. Current Understanding of Transfusion-associated Necrotizing Enterocolitis: Review of Clinical and Experimental Studies and a Call for More Definitive Evidence. NEWBORN 2022; 1:201-208. [PMID: 35746957 PMCID: PMC9217573 DOI: 10.5005/jp-journals-11002-0005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
| | | | - Pascal M Lavoie
- University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Parvesh M Garg
- University of Mississippi, Jackson, Mississippi, United States of America
| | - Venkatesh Sampath
- University of Missouri–Kansas City, Kansas, United States of America
| | | | - Michael D Caplan
- University of Chicago, Chicago, Illinois, United States of America
| | - Praveen Kumar
- Postgraduate Institute of Medical Education and Research, Chandigarh, Punjab, India
| | - Pankaj B Agrawal
- Boston Children’s Hospital, Harvard University, Boston, Massachusetts, United States of America
| | | | | | - Akhil Maheshwari
- Global Newborn Society, Baltimore, Maryland, United States of America
| |
Collapse
|
8
|
Reed CR, Bonadonna D, Otto JC, McDaniel CG, Chabata CV, Kuchibhatla M, Frederiksen J, Layzer JM, Arepally GM, Sullenger BA, Tracy ET. Aptamer-based factor IXa inhibition preserves hemostasis and prevents thrombosis in a piglet model of ECMO. MOLECULAR THERAPY - NUCLEIC ACIDS 2022; 27:524-534. [PMID: 35036063 PMCID: PMC8728519 DOI: 10.1016/j.omtn.2021.12.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 12/09/2021] [Indexed: 11/30/2022]
Abstract
Extracorporeal membrane oxygenation (ECMO) requires anticoagulation to prevent clotting when the patient’s blood contacts the circuit. Unfractionated heparin (UFH) usually prevents clotting but can cause life-threatening bleeding. An anticoagulant that selectively inhibits the contact activation (intrinsic) pathway while sparing the tissue factor (extrinsic) pathway of coagulation might prevent clotting triggered by the circuit while permitting physiologic coagulation at surgical sites. DTRI-178 is an RNA anticoagulant aptamer conjugated to polyethylene glycol that increases its half-life in circulation. This aptamer is based on a previously described molecule (9.3t) that inhibits intrinsic tenase activity by binding to factor IXa on an exosite. Using a piglet model of pediatric venoarterial (VA) ECMO, we compared thromboprevention and blood loss using a single dose of DTRI-178 versus UFH. In each of five experiments, we subjected two litter-matched piglets, one anticoagulated with DTRI-178 and the other with UFH, to simultaneous 12-h periods of VA ECMO. Both anticoagulants achieved satisfactory and comparable thromboprotection. However, UFH piglets had increased surgical site bleeding and required significantly greater blood transfusion volumes than piglets anticoagulated with DTRI-178. Our results indicate that DTRI-178, an aptamer against factor IXa, may be feasible, safer, and result in fewer transfusions and clinical bleeding events in ECMO.
Collapse
Affiliation(s)
- Christopher R. Reed
- Department of Surgery, Duke University Medical Center and Health System, 2301 Erwin Road, Box 3443, Durham, NC 27710, USA
- Corresponding author Christopher R. Reed, MD, Department of Surgery, Duke University Medical Center and Health System, 2301 Erwin Road, Box 3443, Durham, NC 27710, USA
| | - Desiree Bonadonna
- Extracorporeal Life Support, Duke University Medical Center, Durham, NC 27710, USA
| | - James C. Otto
- Department of Surgery, Duke University Medical Center and Health System, 2301 Erwin Road, Box 3443, Durham, NC 27710, USA
| | | | - Charlene Vongai Chabata
- Departments of Surgery; and Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Maragatha Kuchibhatla
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC 27710, USA
| | - James Frederiksen
- Department of Surgery, Duke University Medical Center and Health System, 2301 Erwin Road, Box 3443, Durham, NC 27710, USA
| | - Juliana M. Layzer
- Duke University Clinical and Translational Science Institute, Durham, NC 27710, USA
| | - Gowthami M. Arepally
- Division of Hematology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Bruce A. Sullenger
- Department of Surgery, Duke University Medical Center and Health System, 2301 Erwin Road, Box 3443, Durham, NC 27710, USA
| | - Elisabeth T. Tracy
- Department of Surgery, Duke University Medical Center and Health System, 2301 Erwin Road, Box 3443, Durham, NC 27710, USA
- Division of Pediatric Surgery, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
9
|
Govender K, Cabrales P. Extracorporeal circulation impairs microcirculation perfusion and organ function. J Appl Physiol (1985) 2022; 132:794-810. [PMID: 35085033 PMCID: PMC8917920 DOI: 10.1152/japplphysiol.00726.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Extracorporeal membrane oxygenation (ECMO) is a procedure used to aid respiratory function in critical patients, involving extracorporeal circulation (ECC) of blood. There is a limited number of studies quantifying the hemodynamic effects of ECC procedures on the microcirculation. We sought to mimic veno-arterial-ECMO flow conditions by use of a scaled-down circuit primed with either lactate Ringer (LR) or 5% human serum albumin (HSA). The circuit was first tested using benchtop runs with blood, and subsequently used for in vivo experiments in Golden Syrian hamsters instrumented with a dorsal window chamber to allow for quantification of microvascular hemodynamics and functional capillary density (FCD). Results showed significant impairment in FCD, and a reduction of arteriolar and venular blood flow, with HSA providing significant higher blood flows and FCD compared with LR. Changes in hematocrit and RBC labeling after ECC reflected a shift in plasma volume, which may stem from a loss in intravascular oncotic pressure due to priming fluids. The distribution of hemoglobin oxygen saturation in the microvasculature showed a significant decrease in venules after ECC. In addition, major organs such as the kidney and heart showed increases in both inflammatory and damage markers. These results suggest that ECC impairs microvasculature function and promotes ischemia and hypoxia in the tissues, which can be vital to understanding comorbid clinical outcomes from ECC procedures such as acute kidney injury and multiorgan dysfunction.NEW & NOTEWORTHY ECC reduces microvascular perfusion, with no full recovery 24 h after ECC. HSA performed better as compared with LR in terms of FCD and venule flow, as well as venule oxygen saturation. Increases in inflammatory and damage markers in key organs were observed within all organs analyzed.
Collapse
Affiliation(s)
- Krianthan Govender
- Functional Cardiovascular Engineering Laboratory, Bioengineering Department, University of California San Diego, La Jolla, California
| | - Pedro Cabrales
- Functional Cardiovascular Engineering Laboratory, Bioengineering Department, University of California San Diego, La Jolla, California
| |
Collapse
|
10
|
A Pilot Study To Establish an In Vitro Model To Study Premature Intestinal Epithelium and Gut Microbiota Interactions. mSphere 2021; 6:e0080621. [PMID: 34643422 PMCID: PMC8513685 DOI: 10.1128/msphere.00806-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Intestinal microbiota has emerged as an important player in the health and disease of preterm infants. The interactions between intestinal flora and epithelium can lead to local injury and systemic diseases. A suitable in vitro cell model is needed to enhance our understanding of these interactions. In this study, we exposed fetal epithelial cell cultures (FHs-74 int cells, human, ATCC CCL 241) to sterile fecal filtrates derived from stool collected from preterm infants at <2 and at 3 to 4 weeks of age. We measured the cytokine levels from the culture media after 4, 24, and 48 h of exposure to the fecal filtrates. We analyzed the 16S rRNA V4 gene data of the fecal samples and transcriptome sequencing (RNA-seq) data from the fetal epithelial cells after 48 h of exposure to the same fecal filtrates. The results showed correlations between inflammatory responses (both cytokine levels and gene expression) and the Proteobacteria-to-Firmicutes ratio and between fecal bacterial genera and epithelial apoptosis-related genes. Our in vitro cell model can be further developed and applied to study how the epithelium responds to different microbial flora from preterm infants. Combining immature epithelial cells and preterm infant stool samples into one model allows us to investigate disease processes in preterm infants in a way that had not been previously reported. IMPORTANCE The gut bacterial flora influences the development of the immune system and long-term health outcomes in preterm infants. Studies of the mechanistic interactions between the gut bacteria and mucosal barrier are limited to clinical observations, animal models, and in vitro cell culture models for this vulnerable population. Most in vitro cell culture models of microbe-host interactions use single organisms or adult origin cell lines. Our study is innovative and significant in that we expose immature epithelial cells derived from fetal tissues to fecal filtrates from eight stool samples from four preterm infants to study the role of intestinal epithelial cells. In addition, we analyzed epithelial gene expression to examine multiple cellular processes simultaneously. This model can be developed into patient-derived two- or three-dimensional cell cultures exposed to their own fecal material to allow better prediction of patient physiological responses to support the growing field of precision medicine.
Collapse
|
11
|
Andrei S, Nguyen M, Berthoud V, Morgant MC, Bouhemad B, Guinot PG. Evaluation of the Oxiris Membrane in Cardiogenic Shock Requiring Extracorporeal Membrane Oxygenation Support: Study Protocol for a Single Center, Single-Blind, Randomized Controlled Trial. Front Cardiovasc Med 2021; 8:738496. [PMID: 34708091 PMCID: PMC8544809 DOI: 10.3389/fcvm.2021.738496] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/20/2021] [Indexed: 12/16/2022] Open
Abstract
Background: Veno-arterial extracorporeal membrane oxygenation (VA-ECMO) is the rescue treatment proposed to patients with refractory cardiogenic shock. The VA-ECMO implantation promotes inflammation and ischemia-reperfusion injuries through the VA-ECMO flow, causing digestive mucosa barrier disrupture and inducing translocation of bacterial wall components-Lipopolysaccharides (LPS) with further inflammation and circulatory impairment. LPS is a well-studied surrogate indicator of bacterial translocation. Oxiris membrane is a promising and well-tolerated device that can specifically remove LPS. The main study aim is to compare the LPS elimination capacity of Oxiris membrane vs. a non-absorbant classical renal replacement (RRT) membrane in patients with cardiogenic shock requiring VA-ECMO. Methods: ECMORIX is a randomized, prospective, single-center, single-blind, parallel-group, controlled study. It compares the treatment with Oxiris membrane vs. the standard continuous renal replacement therapy care in patients with cardiogenic shock support by peripheral VA-ECMO. Forty patients will be enrolled in both treatment groups. The primary endpoint is the value of LPS serum levels after 24 h of treatment. LPS serum levels will be monitored during the first 72 h of treatment, as clinical and cardiac ultrasound parameters, biological markers of inflammation and 30-day mortality. Discussion: Oxiris membrane appears to be beneficial in controlling the VA-ECMO-induced ischemia-reperfusion inflammation by LPS removal. ECMORIX results will be of major importance in the management of severe cases requiring VA-ECMO and will bring pathophysiological insights about the LPS role in this context. Clinical Trial Registration: www.ClinicalTrials.gov, identifier: NCT04886180.
Collapse
Affiliation(s)
- Stefan Andrei
- Anaesthesiology and Critical Care Department, Dijon Bourgogne University Hospital, Dijon, France
- Anaesthesiology and Critical Care Department, Carol Davila University of Medicine, Bucharest, Romania
| | - Maxime Nguyen
- Anaesthesiology and Critical Care Department, Dijon Bourgogne University Hospital, Dijon, France
- University of Burgundy Franche Comté, Dijon, France
| | - Vivien Berthoud
- Anaesthesiology and Critical Care Department, Dijon Bourgogne University Hospital, Dijon, France
| | | | - Belaid Bouhemad
- Anaesthesiology and Critical Care Department, Dijon Bourgogne University Hospital, Dijon, France
- University of Burgundy Franche Comté, Dijon, France
| | - Pierre-Grégoire Guinot
- Anaesthesiology and Critical Care Department, Dijon Bourgogne University Hospital, Dijon, France
- University of Burgundy Franche Comté, Dijon, France
| |
Collapse
|
12
|
Wang H, Li C, Peng M, Wang L, Zhao D, Wu T, Yi D, Hou Y, Wu G. N-Acetylcysteine improves intestinal function and attenuates intestinal autophagy in piglets challenged with β-conglycinin. Sci Rep 2021; 11:1261. [PMID: 33441976 PMCID: PMC7807065 DOI: 10.1038/s41598-021-80994-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 01/01/2021] [Indexed: 11/09/2022] Open
Abstract
β-Conglycinin (β-CG), an anti-nutritional factor, is a major allergen in soybeans to induce intestinal dysfunction and diarrhea in neonatal animals, including piglets and human infants. This study with a piglet model determined the effects of N-acetylcysteine (NAC) on intestinal function and autophagy in response to β-CG challenge. Twenty-four 12-day-old piglets (3.44 ± 0.28 kg), which had been weaned at 7 days of age and adapted for 5 days after weaning, were randomly allocated to the control, β-CG, and β-CG + NAC groups. Piglets in the control group were fed a liquid diet containing 10% casein, whereas those in the β-CG and β-CG + NAC groups were fed the basal liquid diets containing 9.5% casein and 0.5% β-CG for 2 days. Thereafter, pigs in the β-CG + NAC group were orally administrated with 50 mg (kg BW)-1 NAC for 3 days, while pigs in the other two groups were orally administrated with the same volume of sterile saline. NAC numerically reduced diarrhea incidence (- 46.2%) and the concentrations of hydrogen peroxide and malondialdehyde, but increased claudin-1 and intestinal fatty-acid binding protein (iFABP) protein abundances and activities of catalase and glutathione peroxidase in the jejunum of β-CG-challenged piglets. Although β-CG challenge decreased the villus height, villus height/crypt depth ratio, and mRNA levels of claudin-1 and occludin, no significant differences were observed in these indices between the control and β-CG + NAC groups, suggesting the positive effects of NAC supplementation on intestinal mucosal barrier function. Moreover, NAC increased the concentrations of citrulline and D-xylose in the plasma, as well as the expression of genes for aquaporin (AQP) 3, AQP4, peptide transporter 1 (PepT1), sodium/glucose co-transporter-1 (SGLT-1), potassium inwardly-rectifying channel, subfamily J, member 13 (KCNJ13), and solute carrier family 1 member 1 (SLC1A1) in the jejunum, demonstrating that NAC augmented intestinal metabolic activity and absorptive function. Remarkably, NAC decreased Atg5 protein abundance and the LC3II/LC3I ratio (an indicator of autophagy) in the jejunum of β-CG-challenged piglets. Taken together, NAC supplementation improved intestinal function and attenuated intestinal autophagy in β-CG-challenged piglets.
Collapse
Affiliation(s)
- Huiyun Wang
- Hubei International Scientific and Technological Cooperation Base of Animal Nutrition and Gut Health, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Chengcheng Li
- Hubei International Scientific and Technological Cooperation Base of Animal Nutrition and Gut Health, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Meng Peng
- Hubei International Scientific and Technological Cooperation Base of Animal Nutrition and Gut Health, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Lei Wang
- Hubei International Scientific and Technological Cooperation Base of Animal Nutrition and Gut Health, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Di Zhao
- Hubei International Scientific and Technological Cooperation Base of Animal Nutrition and Gut Health, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Tao Wu
- Hubei International Scientific and Technological Cooperation Base of Animal Nutrition and Gut Health, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Dan Yi
- Hubei International Scientific and Technological Cooperation Base of Animal Nutrition and Gut Health, Wuhan Polytechnic University, Wuhan, 430023, China.
| | - Yongqing Hou
- Hubei International Scientific and Technological Cooperation Base of Animal Nutrition and Gut Health, Wuhan Polytechnic University, Wuhan, 430023, China.
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, TX, 77843, USA
| |
Collapse
|
13
|
Coagulopathy Characterized by Rotational Thromboelastometry in a Porcine Pediatric ECMO Model. THE JOURNAL OF EXTRA-CORPOREAL TECHNOLOGY 2020; 52:203-211. [PMID: 32981958 DOI: 10.1182/ject-2000011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 07/16/2020] [Indexed: 11/20/2022]
Abstract
Venoarterial extracorporeal membrane oxygenation (VA-ECMO) is used to support patients with reversible cardiopulmonary insufficiency. Although it is a lifesaving technology, bleeding, inflammation, and thrombosis are well-described complications of ECMO. Adult porcine models of ECMO have been used to recapitulate the physiology and hemostatic consequences of ECMO cannulation in adults. However, these models lack the unique physiology and persistence of fetal forms of coagulation factors and fibrinogen as in human infants. We aimed to describe physiologic and coagulation parameters of piglets cannulated and supported with VA-ECMO. Four healthy piglets (5.7-6.4 kg) were cannulated via jugular vein and carotid artery by cutdown and supported for a maximum of 20 hours. Heparin was used with a goal activated clotting time of 180-220 seconds. Arterial blood gas (ABG) was performed hourly, and blood was transfused from an adult donor to maintain hematocrit (Hct) > 24%. Rotational thromboelastometry (ROTEM) was performed at seven time points. All animals achieved adequate flow with a patent circuit throughout the run (pre- and post-oxygenator pressure gradient <10 mmHg). There was slow but significant hemorrhage at cannulation, arterial line, and bladder catheter sites. All animals required the maximum blood transfusion volume available. All animals became anemic after exhaustion of blood for transfusion. ABG showed progressively declining Hct and adequate oxygenation. ROTEM demonstrated decreasing fibrin-only ROTEM (FIBTEM) clot firmness. Histology was overall unremarkable. Pediatric swine are an important model for the study of pediatric ECMO. We have demonstrated the feasibility of such a model while providing descriptions of physiologic, hematologic, and coagulation parameters throughout. Weak whole-blood clot firmness by ROTEM suggested defects in fibrinogen, and there was a clinical bleeding tendency in all animals studied. This model serves as an important means to study the complex derangements in hemostasis during ECMO.
Collapse
|
14
|
Guo M, Lu C, Li L, Yao D, Li Y. Normothermic extracorporeal membrane oxygenation support: Improving the function of intestinal grafts obtained from cardiac death donors. Artif Organs 2020; 44:1098-1106. [PMID: 32279328 DOI: 10.1111/aor.13697] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 02/26/2020] [Accepted: 03/28/2020] [Indexed: 12/01/2022]
Abstract
Extracorporeal membrane oxygenation (ECMO) could ameliorate the energy status and viability of bowel grafts from cardiac death donors. However, the function of these grafts after transplantation is not clear. The purpose of the study was to evaluate the early function of intestinal grafts after transplantation from expected cardiac death donors supported with normothermic extracorporeal support using a porcine allogeneic orthotopic segmental small bowel transplantation model. Eighteen domestic crossbred donor pigs were assigned to living donation (LD), donation after cardiac death (DCD), and ECMO groups. In the LD group, small bowels were harvested and preserved immediately in cold storage. In the other two groups, the donor pigs received conventional rapid recovery treatment or 1-hour normothermic extracorporeal support after 10-minutes expected cardiac arrest. Subsequently, the small bowels were removed and preserved in cold storage. After 5-6 hours of preservation, small bowel grafts were transplanted into the recipient pigs that underwent enterectomy. The pathology and electron microscopy results, cell apoptosis rate, tight junction protein expression level in the intestinal mucosa, and plasma endotoxin level were evaluated after transplantation. All grafts functioned on the basis of the maltose absorption test results at day 7 after transplantation. There were no significant differences in the morphological changes in the intestinal mucosa among the three groups at day 7 after transplantation. The cell apoptosis rate and plasma endotoxin level in the ECMO group did not differ significantly than those in the LD group, but were evidently lower than those in the DCD group (P < .001). The intestinal absorptive function improved significantly in the ECMO group in contrast with that in the DCD group (P < .001). Short-term ECMO intervention can alleviate ischemia-reperfusion injuries in intestinal grafts and improve intestinal absorptive function in the early stage after transplantation. Reducing caspase-3 protein expression and cell apoptosis in the intestinal mucosa may be one of the protective mechanisms of ECMO intervention.
Collapse
Affiliation(s)
- Mingxiao Guo
- Department of General Surgery, Linyi People's Hospital, Linyi, China
| | - Chunlei Lu
- Department of General Surgery, Linyi People's Hospital, Linyi, China
| | - Linlin Li
- Department of Surgery, Linyi Municipal Mental Health Center, Linyi, China
| | - Danhua Yao
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yousheng Li
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| |
Collapse
|
15
|
Targeted inhibition of thrombin attenuates murine neonatal necrotizing enterocolitis. Proc Natl Acad Sci U S A 2020; 117:10958-10969. [PMID: 32366656 DOI: 10.1073/pnas.1912357117] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is an inflammatory bowel necrosis of premature infants and an orphan disease with no specific treatment. Most patients with confirmed NEC develop moderate-severe thrombocytopenia requiring one or more platelet transfusions. Here we used our neonatal murine model of NEC-related thrombocytopenia to investigate mechanisms of platelet depletion associated with this disease [K. Namachivayam, K. MohanKumar, L. Garg, B. A. Torres, A. Maheshwari, Pediatr. Res. 81, 817-824 (2017)]. In this model, enteral administration of immunogen trinitrobenzene sulfonate (TNBS) in 10-d-old mouse pups produces an acute necrotizing ileocolitis resembling human NEC within 24 h, and these mice developed thrombocytopenia at 12 to 15 h. We hypothesized that platelet activation and depletion occur during intestinal injury following exposure to bacterial products translocated across the damaged mucosa. Surprisingly, platelet activation began in our model 3 h after TNBS administration, antedating mucosal injury or endotoxinemia. Platelet activation was triggered by thrombin, which, in turn, was activated by tissue factor released from intestinal macrophages. Compared to adults, neonatal platelets showed enhanced sensitivity to thrombin due to higher expression of several downstream signaling mediators and the deficiency of endogenous thrombin antagonists. The expression of tissue factor in intestinal macrophages was also unique to the neonate. Targeted inhibition of thrombin by a nanomedicine-based approach was protective without increasing interstitial hemorrhages in the inflamed bowel or other organs. In support of these data, we detected increased circulating tissue factor and thrombin-antithrombin complexes in patients with NEC. Our findings show that platelet activation is an important pathophysiological event and a potential therapeutic target in NEC.
Collapse
|
16
|
MohanKumar K, Namachivayam K, Song T, Jake Cha B, Slate A, Hendrickson JE, Pan H, Wickline SA, Oh JY, Patel RP, He L, Torres BA, Maheshwari A. A murine neonatal model of necrotizing enterocolitis caused by anemia and red blood cell transfusions. Nat Commun 2019; 10:3494. [PMID: 31375667 PMCID: PMC6677753 DOI: 10.1038/s41467-019-11199-5] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 06/27/2019] [Indexed: 12/23/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is an idiopathic, inflammatory bowel necrosis of premature infants. Clinical studies have linked NEC with antecedent red blood cell (RBC) transfusions, but the underlying mechanisms are unclear. Here we report a neonatal murine model to investigate this association. C57BL/6 mouse pups rendered anemic by timed phlebotomy and then given RBC transfusions develop NEC-like intestinal injury with prominent necrosis, inflammation, and submucosal edema/separation of the lamina propria in the ileocecal region and colon within 12-24 h. The anemic intestine is infiltrated by inflammatory macrophages, which are activated in situ by RBC transfusions via a Toll-like receptor (TLR)-4-mediated mechanism and cause bowel injury. Chelation of RBC degradation products with haptoglobin, absence of TLR4, macrophage depletion, and inhibition of macrophage activation is protective. Intestinal injury worsens with increasing severity and the duration of anemia prior to transfusion, indicating a need for the re-evaluation of current transfusion guidelines for premature infants.
Collapse
MESH Headings
- Anemia/complications
- Anemia/therapy
- Animals
- Animals, Newborn
- Cecum/pathology
- Colon/pathology
- Disease Models, Animal
- Enterocolitis, Necrotizing/etiology
- Enterocolitis, Necrotizing/pathology
- Erythrocyte Transfusion/adverse effects
- Humans
- Ileum/pathology
- Infant, Newborn
- Infant, Newborn, Diseases/etiology
- Infant, Newborn, Diseases/pathology
- Infant, Premature
- Intestinal Mucosa/pathology
- Macrophages/immunology
- Macrophages/metabolism
- Mice
- Toll-Like Receptor 4/immunology
- Toll-Like Receptor 4/metabolism
Collapse
Affiliation(s)
- Krishnan MohanKumar
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Kopperuncholan Namachivayam
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Tanjing Song
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Byeong Jake Cha
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Andrea Slate
- Department of Comparative Medicine, University of South Florida, Tampa, FL, 33612, USA
- Center for Comparative Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Jeanne E Hendrickson
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, 06520, USA
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Hua Pan
- Department of Cardiology, Morsani College of Medicine, University of South Florida, Tampa, FL, 33629, USA
| | - Samuel A Wickline
- Department of Cardiology, Morsani College of Medicine, University of South Florida, Tampa, FL, 33629, USA
| | - Joo-Yeun Oh
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Rakesh P Patel
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Ling He
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Benjamin A Torres
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Akhil Maheshwari
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA.
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD, 21287, USA.
| |
Collapse
|
17
|
Armacki M, Trugenberger AK, Ellwanger AK, Eiseler T, Schwerdt C, Bettac L, Langgartner D, Azoitei N, Halbgebauer R, Groß R, Barth T, Lechel A, Walter BM, Kraus JM, Wiegreffe C, Grimm J, Scheffold A, Schneider MR, Peuker K, Zeißig S, Britsch S, Rose-John S, Vettorazzi S, Wolf E, Tannapfel A, Steinestel K, Reber SO, Walther P, Kestler HA, Radermacher P, Barth TF, Huber-Lang M, Kleger A, Seufferlein T. Thirty-eight-negative kinase 1 mediates trauma-induced intestinal injury and multi-organ failure. J Clin Invest 2018; 128:5056-5072. [PMID: 30320600 DOI: 10.1172/jci97912] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 08/28/2018] [Indexed: 12/17/2022] Open
Abstract
Dysregulated intestinal epithelial apoptosis initiates gut injury, alters the intestinal barrier, and can facilitate bacterial translocation leading to a systemic inflammatory response syndrome (SIRS) and/or multi-organ dysfunction syndrome (MODS). A variety of gastrointestinal disorders, including inflammatory bowel disease, have been linked to intestinal apoptosis. Similarly, intestinal hyperpermeability and gut failure occur in critically ill patients, putting the gut at the center of SIRS pathology. Regulation of apoptosis and immune-modulatory functions have been ascribed to Thirty-eight-negative kinase 1 (TNK1), whose activity is regulated merely by expression. We investigated the effect of TNK1 on intestinal integrity and its role in MODS. TNK1 expression induced crypt-specific apoptosis, leading to bacterial translocation, subsequent septic shock, and early death. Mechanistically, TNK1 expression in vivo resulted in STAT3 phosphorylation, nuclear translocation of p65, and release of IL-6 and TNF-α. A TNF-α neutralizing antibody partially blocked development of intestinal damage. Conversely, gut-specific deletion of TNK1 protected the intestinal mucosa from experimental colitis and prevented cytokine release in the gut. Finally, TNK1 was found to be deregulated in the gut in murine and porcine trauma models and human inflammatory bowel disease. Thus, TNK1 might be a target during MODS to prevent damage in several organs, notably the gut.
Collapse
Affiliation(s)
- Milena Armacki
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | | | - Ann K Ellwanger
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Tim Eiseler
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Christiane Schwerdt
- Waldkrankenhaus "Rudolph Elle" Eisenberg, Lehrstuhl für Orthopädie Uniklinik Jena, Jena, Germany
| | - Lucas Bettac
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Dominik Langgartner
- Laboratory for Molecular Psychosomatics, Clinic for Psychosomatic Medicine and Psychotherapy, and
| | - Ninel Azoitei
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Rebecca Halbgebauer
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Rüdiger Groß
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Tabea Barth
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - André Lechel
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Benjamin M Walter
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | | | | | | | - Annika Scheffold
- Department of Internal Medicine III, University Hospital Ulm, Ulm, Germany
| | | | - Kenneth Peuker
- Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany
| | - Sebastian Zeißig
- Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany
| | - Stefan Britsch
- Institute of Molecular and Cellular Anatomy, Ulm University, Ulm, Germany
| | | | - Sabine Vettorazzi
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | | | | | - Konrad Steinestel
- Institute of Pathology and Molecular Pathology, Bundeswehrkrankenhaus Ulm, Ulm, Germany
| | - Stefan O Reber
- Laboratory for Molecular Psychosomatics, Clinic for Psychosomatic Medicine and Psychotherapy, and
| | - Paul Walther
- Central Facility for Electron Microscopy, University of Ulm, Ulm, Germany
| | | | - Peter Radermacher
- Institute of Anesthesiological Pathophysiology and Process Engineering, Ulm University, Ulm, Germany
| | | | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Alexander Kleger
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Thomas Seufferlein
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| |
Collapse
|
18
|
Endothelial Microvesicles and Soluble Markers of Endothelial Injury in Critically Ill Newborns. Mediators Inflamm 2018; 2018:1975056. [PMID: 30116143 PMCID: PMC6079510 DOI: 10.1155/2018/1975056] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 06/27/2018] [Indexed: 11/18/2022] Open
Abstract
Neonatal systemic inflammatory response and multiple organ dysfunction syndrome are the main postnatal insults influencing mortality and morbidity. Critically ill newborns with high predicted mortality are supported by extracorporeal membrane oxygenation (ECMO). Biomarkers of inflammatory response and endothelial injury can be used for early diagnosis and treatment of critical neonatal situations. The aim of our study was to explore plasma proteins and endothelial microvesicles as markers of inflammation and endothelial activation in newborns on ECMO and to compare them with healthy neonates. Thirteen newborns on ECMO and 13 healthy newborns were included in the study. Plasma soluble biomarkers were measured using multiplex immunoassay based on Luminex® xMAP multianalyte profiling platform. The total microvesicle count and plasma level of surface antigen-specific microvesicles were determined by flow cytometry. The plasma concentration of cell-derived microvesicles was measured using annexin-V labeling, and the endothelial origin of microvesicles was determined using lineage-specific antigen labeling of endothelial cell/microvesicle markers (endoglin/CD105, PECAM1/CD31, VEGFR2/CD309, and MadCAM1). Inflammatory markers (procalcitonin, IL-1β, IL-6, and IL-22) were increased in the ECMO group (P < 0.01). The assessment of endothelial markers showed higher concentrations of endocan and angiopoietin-2 (P < 0.01) in the ECMO group while VEGF in the ECMO group was significantly lower (P < 0.01). In the ECMO group, the concentration of annexin-V-positive microvesicles (total microvesicles) and endothelial microvesicles expressing mucosal vascular addressin cell adhesion molecule 1 (MadCAM1) was increased (P = 0.05). In summary, we found increased concentrations of soluble inflammatory and endothelial markers in the plasma of critically ill newborns with multiple organ dysfunction. Increased plasma concentrations of microvesicles may reflect the activation or damage of blood cells and vasculature including endothelial cells. The measurement of cell membrane-derived microvesicles may be added to the panel of established inflammatory markers in order to increase the sensitivity and specificity of the diagnostic process in critically ill newborns.
Collapse
|
19
|
Evaluation of combination therapy with hydrocortisone, vitamin C, and vitamin E in a rat model of intestine ischemia-reperfusion injury. ACTA ACUST UNITED AC 2017. [DOI: 10.1007/s00580-017-2610-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
20
|
Sunagawa G, Koprivanac M, Karimov JH, Moazami N, Fukamachi K. Is a pulse absolutely necessary during cardiopulmonary bypass? Expert Rev Med Devices 2016; 14:27-35. [DOI: 10.1080/17434440.2017.1265445] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Gengo Sunagawa
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Marijan Koprivanac
- Department of Thoracic and Cardiovascular Surgery, Kaufman Center for Heart Failure, Cardiac Transplantation and Mechanical Circulatory Support, Miller Family Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jamshid H. Karimov
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Nader Moazami
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Thoracic and Cardiovascular Surgery, Kaufman Center for Heart Failure, Cardiac Transplantation and Mechanical Circulatory Support, Miller Family Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Kiyotaka Fukamachi
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
21
|
Guo M, Yao D, Li L, Lu C, Li Y, Li J. Intestinal Conditioning After Cardiac Arrest: The Use of Normothermic Extracorporeal Membrane Oxygenation in the Non-Heart-Beating Animal Model. Artif Organs 2016; 40:738-45. [PMID: 27097758 DOI: 10.1111/aor.12691] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 12/03/2015] [Accepted: 12/11/2015] [Indexed: 12/12/2022]
Abstract
The effect of normothermic extracorporeal membrane oxygenation (NECMO) on small bowel preservation in a clinically relevant large animal model of expected donation after cardiac death (eDCD) was evaluated. Thirty domestic crossbred donor pigs were divided into five groups. The first group served as the live donation (LD) group, the second group served as the donation after cardiac death (DCD) group, and the remaining were further assigned into three subgroups: E1 group (1 h NECMO support), E3 group (3 h NECMO support), and E5 group (5 h NECMO support). Pathology, electron microscopy, energy metabolism, cell apoptosis, and tight junction (TJ) protein expression level of intestinal mucosa and the level of plasma d-lactic acid were evaluated in normal, cardiac death and at the end of extracorporeal support, respectively. The mean arterial pressure and PaO2 were maintained over 60 and 267 mm Hg during NECMO support, respectively. One hour of extracorporeal support could improve the energy status in intestines of the DCD group. Although the histologic damage and apoptosis of the E1 group had no significant difference with those of the LD and DCD groups (P > 0.05), the levels of intestinal mucosa TJ protein decreased (P < 0.05), and plasma d-lactic acid increased progressively (P < 0.05). With the extension of extracorporeal support, the degree of intestinal mucosa damage and intestinal permeability gradually increased, as well as the content of adenosine triphosphate in intestinal mucosa. The normothermic extracorporeal support for 1 h in DCD is beneficial for improving the energy status and viability of the bowel. However, the integrity of intestinal mucosa was destroyed gradually as extracorporeal support time went by. And the activation of intestinal epithelial apoptosis and hyperoxia might be the factors that lead to intestinal mucosa injury.
Collapse
Affiliation(s)
- Mingxiao Guo
- Department of Laparoscopic Surgery Center, Linyi People's Hospital, Shandong University, Linyi
| | - Danhua Yao
- Department of Surgery, Jinling Hospital, Nanjing University School of Medicine, Nanjing
| | - Linlin Li
- Department of Surgery, Linyi Mental Health Center, Linyi, China
| | - Chunlei Lu
- Department of Laparoscopic Surgery Center, Linyi People's Hospital, Shandong University, Linyi
| | - Yousheng Li
- Department of Surgery, Jinling Hospital, Nanjing University School of Medicine, Nanjing
| | - Jieshou Li
- Department of Surgery, Jinling Hospital, Nanjing University School of Medicine, Nanjing
| |
Collapse
|
22
|
Güney LH, Araz C, Beyazpınar DS, Arda İS, Arslan EE, Hiçsönmez A. Abdominal Problems in Children with Congenital Cardiovascular Abnormalities. Balkan Med J 2015; 32:285-90. [PMID: 26185717 DOI: 10.5152/balkanmedj.2015.151045] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 01/26/2015] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Congenital cardiovascular abnormality is an important cause of morbidity and mortality in childhood. Both the type of congenital cardiovascular abnormality and cardiopulmonary bypass are responsible for gastrointestinal system problems. AIMS Intra-abdominal problems, such as paralytic ileus, necrotizing enterocolitis, and intestinal perforation, are common in patients who have been operated or who are being followed for congenital cardiovascular abnormalities. Besides the primary congenital cardiovascular abnormalities, ischemia secondary to cardiac catheterization or surgery contributes to the incidence of these problems. STUDY DESIGN Cross-sectional study. METHODS In this study, we aimed to screen the intra-abdominal problems seen in patients with congenital cardiovascular abnormalities who had undergone surgical or angiographical intervention(s). Patients with congenital cardiovascular abnormalities who had been treated medically or surgically between 2000 and 2014 were analyzed retrospectively in terms of intra-abdominal problems. The patients' demographic data, type of congenital cardiovascular abnormalities, the intervention applied (surgical, angiographic), the incidence of intra-abdominal problem(s), the interventions applied for the intra-abdominal problems, and the results were evaluated. RESULTS Fourteen (Group I) of the 76 patients with congenital cardiovascular abnormalities diagnosis were operated due to intra-abdominal problems, and 62 (Group II) were followed-up clinically for intra-abdominal problems. In Group I (10 boys and 4 girls), 11 patients were aged between 0 and 12 months, and three patients were older than 12 months. Group II included 52 patients aged between 0 and 12 months and 10 patients older than 12 months. Cardiovascular surgical interventions had been applied to six patients in Group I and 40 patients in Group II. The most frequent intra-abdominal problems were necrotizing enterocolitis and intestinal perforation in Group I, and paralytic ileus in Group II. Seven of the Group I patients and 22 of the Group II patients died. The patients who died in both groups had more than three congenital cardiovascular abnormalities in the same patient, and 80% of these patients had been operated for congenital cardiovascular abnormalities. CONCLUSION The gastrointestinal system is involved in important complications experienced by patients with congenital cardiovascular abnormalities. The mortality rate was higher in operated patients due to gastrointestinal complications. Gastrointestinal complications are more frequent in patients with cyanotic anomalies. The presence of more than one congenital cardiovascular abnormality in a patient increased the mortality rate.
Collapse
Affiliation(s)
- Lütfi Hakan Güney
- Department of Pediatric Surgery, Başkent University Faculty of Medicine, Ankara, Turkey
| | - Coşkun Araz
- Department of Anesthesiology, Başkent University Faculty of Medicine, Ankara, Turkey
| | - Deniz Sarp Beyazpınar
- Department of Cardiovasculer Surgery, Başkent University Faculty of Medicine, Ankara, Turkey
| | - İrfan Serdar Arda
- Department of Pediatric Surgery, Başkent University Faculty of Medicine, Ankara, Turkey
| | - Esra Elif Arslan
- Department of Pediatric Surgery, Başkent University Faculty of Medicine, Ankara, Turkey
| | - Akgün Hiçsönmez
- Department of Pediatric Surgery, Başkent University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
23
|
Keschenau PR, Ribbe S, Tamm M, Hanssen SJ, Tolba R, Jacobs MJ, Kalder J. Extracorporeal circulation increases proliferation in the intestinal mucosa in a large animal model. J Vasc Surg 2015; 64:1121-33. [PMID: 26190050 DOI: 10.1016/j.jvs.2015.05.043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 05/29/2015] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Extracorporeal circulation induces ischemia/reperfusion injury in the small intestinal wall. One reason for this damage is a perfusion shift from the muscular toward the mucosal layer. This study investigated the effect of this perfusion shift on the small-intestinal apoptosis and proliferation. METHODS Twenty-eight pigs were randomly assigned to the following cohorts and underwent a thoracolaparotomy and a 1 hour main procedure: cohort I: control; cohort II: thoracic aortic cross-clamping (TAC) without perfusion; cohort III: TAC and distal aortic perfusion (DAP); cohort IV: TAC, DAP, and selective visceral perfusion. The main procedure was followed by 2 hours of reperfusion in all cohorts. Tissue samples were taken during the experiment, stained, and analyzed for apoptosis and proliferation (caspase-3, annexin-V, terminal deoxynucleotide transferase-mediated deoxy uridine triphosphate nick-end labeling, and proliferating cell nuclear antigen). Six animals died unexpectedly during the experiment and were excluded from the analysis. RESULTS Extensive tissue damage and necrosis was only found in cohort II after the main procedure. In the mucosa, the proliferation was increased in cohort III at the end of the experiment (P = .0157 cohort I vs II). In contrast, the annexin-V/proliferating cell nuclear antigen ratio was significantly higher in cohorts II and IV than in cohorts I and II at the end of the experiment (P = .0034). Furthermore, the caspase-3/annexin-V ratio was increased in all cohorts at the end of the experiment (P = .0015). CONCLUSIONS Mucosal proliferation is the early repair mechanism of the limited small intestinal ischemia/reperfusion injury after DAP. Furthermore, the extensive surgical trauma shifted the mucosal apoptosis into an advanced state.
Collapse
Affiliation(s)
- Paula Rosalie Keschenau
- European Vascular Center Aachen-Maastricht, Department of Vascular Surgery, Rheinisch-Westfälische Technische Hochschule University Hospital Aachen, Aachen, Germany
| | - Stefanie Ribbe
- European Vascular Center Aachen-Maastricht, Department of Vascular Surgery, Rheinisch-Westfälische Technische Hochschule University Hospital Aachen, Aachen, Germany
| | - Miriam Tamm
- Department of Medical Statistics, Rheinisch-Westfälische Technische Hochschule University Hospital Aachen, Aachen, Germany
| | - Sebastiaan J Hanssen
- European Vascular Center Aachen-Maastricht, Department of Surgery, Maastricht University Hospital, Maastricht, The Netherlands
| | - René Tolba
- Institute for Laboratory Animal Science and Experimental Surgery, Rheinisch-Westfälische Technische Hochschule University Hospital Aachen, Aachen, Germany
| | - Michael J Jacobs
- European Vascular Center Aachen-Maastricht, Department of Vascular Surgery, Rheinisch-Westfälische Technische Hochschule University Hospital Aachen, Aachen, Germany; European Vascular Center Aachen-Maastricht, Department of Surgery, Maastricht University Hospital, Maastricht, The Netherlands.
| | - Johannes Kalder
- European Vascular Center Aachen-Maastricht, Department of Vascular Surgery, Rheinisch-Westfälische Technische Hochschule University Hospital Aachen, Aachen, Germany
| |
Collapse
|