1
|
Archontakis-Barakakis P, Mavridis T, Chlorogiannis DD, Barakakis G, Laou E, Sessler DI, Gkiokas G, Chalkias A. Intestinal oxygen utilisation and cellular adaptation during intestinal ischaemia-reperfusion injury. Clin Transl Med 2025; 15:e70136. [PMID: 39724463 DOI: 10.1002/ctm2.70136] [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: 08/28/2024] [Revised: 11/06/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024] Open
Abstract
The gastrointestinal tract can be deranged by ailments including sepsis, trauma and haemorrhage. Ischaemic injury provokes a common constellation of microscopic and macroscopic changes that, together with the paradoxical exacerbation of cellular dysfunction and death following restoration of blood flow, are collectively known as ischaemia-reperfusion injury (IRI). Although much of the gastrointestinal tract is normally hypoxemic, intestinal IRI results when there is inadequate oxygen availability due to poor supply (pathological hypoxia) or abnormal tissue oxygen use and metabolism (dysoxia). Intestinal oxygen uptake usually remains constant over a wide range of blood flows and pressures, with cellular function being substantively compromised when ischaemia leads to a >50% decline in intestinal oxygen consumption. Restoration of perfusion and oxygenation provokes additional injury, resulting in mucosal damage and disruption of intestinal barrier function. The primary cellular mechanism for sensing hypoxia and for activating a cascade of cellular responses to mitigate the injury is a family of heterodimer proteins called hypoxia-inducible factors (HIFs). The HIF system is connected to numerous biochemical and immunologic pathways induced by IRI and the concentration of those proteins increases during hypoxia and dysoxia. Activation of the HIF system leads to augmented transcription of specific genes in various types of affected cells, but may also augment apoptotic and inflammatory processes, thus aggravating gut injury. KEY POINTS: During intestinal ischaemia, mitochondrial oxygen uptake is reduced when cellular oxygen partial pressure decreases to below the threshold required to maintain normal oxidative metabolism. Upon reperfusion, intestinal hypoxia may persist because microcirculatory flow remains impaired and/or because available oxygen is consumed by enzymes, intestinal cells and neutrophils.
Collapse
Affiliation(s)
| | - Theodoros Mavridis
- Department of Neurology, Tallaght University Hospital (TUH)/The Adelaide and Meath Hospital incorporating the National Children's Hospital (AMNCH), Dublin, Ireland
| | | | - Georgios Barakakis
- Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Eleni Laou
- Department of Anesthesiology, Agia Sophia Children's Hospital, Athens, Greece
| | - Daniel I Sessler
- Center for Outcomes Research and Department of Anesthesiology, UTHealth, Houston, Texas, USA
- Outcomes Research Consortium®, Houston, Texas, USA
| | - George Gkiokas
- Second Department of Surgery, Aretaieion University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasios Chalkias
- Outcomes Research Consortium®, Houston, Texas, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Department of Critical Care Medicine, Tzaneio General Hospital, Piraeus, Greece
| |
Collapse
|
2
|
McKinney-Aguirre CA, Schaaf CR, Goya-Jorge E, Freund JM, Gonzalez LM. Large animal models enhance the study of crypt-mediated epithelial recovery from prolonged intestinal ischemia reperfusion injury. Am J Physiol Gastrointest Liver Physiol 2024; 327:G783-G788. [PMID: 39404771 PMCID: PMC11684884 DOI: 10.1152/ajpgi.00236.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/12/2024] [Accepted: 10/03/2024] [Indexed: 11/14/2024]
Abstract
Intestinal ischemia and reperfusion injury (IRI) is a deadly and common condition. Death is associated with sepsis due to insufficient epithelial repair, requiring stem cell-driven regeneration, typically beginning 48 h after injury. Animal models are critical to advancing this field. To effectively study epithelial healing, models must survive clinically relevant intestinal ischemic injury extending to the crypt. Although mouse models are indispensable to intestinal research, their application for studying epithelial repair following severe IRI may be limited. Ischemic injury was induced in mouse and porcine jejunum for up to 3 h, with up to 72 h of reperfusion. Histologic damage was scored by Chiu-Park grade, and animal survival was assessed. Findings were compared between species. A mouse IRI literature review was performed to evaluate the purported degree of injury, duration of recovery, and reported survival rates. In mice and pigs, 3 h of ischemia induced severe, reliable injury extending into the crypt. However, at 48 h, mouse survival was only 23.5% compared with 100% survival in pigs. In literature, ischemia was induced for >1 h in only 4 of 102 mouse studies and none to 3 h. Recovery was attempted for 48 h in only six reports. Forty-seven studies reported intestinal crypt injury. Of those that featured histologic intestinal crypt damage, survival rates at 48 h ranged from 10 to 50% (median 30%). Mouse models are not ideal for studying intestinal stem cell-mediated recovery from severe IRI. Alternative large animal models, like pigs, are recommended.NEW & NOTEWORTHY Additional research is needed to improve recovery from severe intestinal ischemia. The selection of the ideal animal model is critical to facilitating this work. Based on our experimentation and literature review, porcine models, with increased translatability and an improved ability to survive both prolonged ischemia and the recovery period, appear to be the most appropriate choice for future studies.
Collapse
Affiliation(s)
- Caroline A McKinney-Aguirre
- Department of Clinical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, United States
| | - Cecilia R Schaaf
- Department of Clinical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, United States
| | - Elizabeth Goya-Jorge
- Department of Clinical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, United States
| | - John M Freund
- Department of Clinical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, United States
| | - Liara M Gonzalez
- Department of Clinical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, United States
| |
Collapse
|
3
|
Ziegler AL, Caldwell ML, Craig SE, Hellstrom EA, Sheridan AE, Touvron MS, Pridgen TA, Magness ST, Odle J, Van Landeghem L, Blikslager AT. Enteric glial cell network function is required for epithelial barrier restitution following intestinal ischemic injury in the early postnatal period. Am J Physiol Gastrointest Liver Physiol 2024; 326:G228-G246. [PMID: 38147796 PMCID: PMC11211042 DOI: 10.1152/ajpgi.00216.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 08/23/2023] [Accepted: 09/12/2023] [Indexed: 12/28/2023]
Abstract
Ischemic damage to the intestinal epithelial barrier, such as in necrotizing enterocolitis or small intestinal volvulus, is associated with higher mortality rates in younger patients. We have recently reported a powerful pig model to investigate these age-dependent outcomes in which mucosal barrier restitution is strikingly absent in neonates but can be rescued by direct application of homogenized mucosa from older, juvenile pigs by a yet-undefined mechanism. Within the mucosa, a postnatally developing network of enteric glial cells (EGCs) is gaining recognition as a key regulator of the mucosal barrier. Therefore, we hypothesized that the developing EGC network may play an important role in coordinating intestinal barrier repair in neonates. Neonatal and juvenile jejunal mucosa recovering from surgically induced intestinal ischemia was visualized by scanning electron microscopy and the transcriptomic phenotypes were assessed by bulk RNA sequencing. EGC network density and glial activity were examined by Gene Set Enrichment Analysis, three-dimensional (3-D) volume imaging, and Western blot and its function in regulating epithelial restitution was assessed ex vivo in Ussing chamber using the glia-specific inhibitor fluoroacetate (FA), and in vitro by coculture assay. Here we refine and elaborate our translational model, confirming a neonatal phenotype characterized by a complete lack of coordinated reparative signaling in the mucosal microenvironment. Furthermore, we report important evidence that the subepithelial EGC network changes significantly over the early postnatal period and demonstrate that the proximity of a specific functional population of EGC to wounded intestinal epithelium contributes to intestinal barrier restitution following ischemic injury.NEW & NOTEWORTHY This study refines a powerful translational pig model, defining an age-dependent relationship between enteric glia and the intestinal epithelium during intestinal ischemic injury and confirming an important role for enteric glial cell (EGC) activity in driving mucosal barrier restitution. This study suggests that targeting the enteric glial network could lead to novel interventions to improve recovery from intestinal injury in neonatal patients.
Collapse
Affiliation(s)
- Amanda L Ziegler
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States
| | - Madison L Caldwell
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States
| | - Sara E Craig
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States
| | - Emily A Hellstrom
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States
| | - Anastasia E Sheridan
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States
| | - Melissa S Touvron
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States
| | - Tiffany A Pridgen
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States
| | - Scott T Magness
- Joint Department of Biomedical Engineering, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States
| | - Jack Odle
- Department of Animal Science, College of Agriculture and Life Sciences, North Carolina State University, Raleigh, North Carolina, United States
| | - Laurianne Van Landeghem
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States
| | - Anthony T Blikslager
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States
| |
Collapse
|
4
|
Tanaka Y, Matsumura Y, Aoki M, Hayashi Y, Izawa Y, Endo K, Mato T. Establishment of a swine experimental model of non-occlusive mesenteric ischemia: Combining induced hemorrhagic shock and vasopressor administration. Acute Med Surg 2024; 11:e982. [PMID: 39045518 PMCID: PMC11263749 DOI: 10.1002/ams2.982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 07/05/2024] [Accepted: 07/11/2024] [Indexed: 07/25/2024] Open
Abstract
Aim Non-occlusive mesenteric ischemia (NOMI) is associated with high mortality rates, but definitive treatments have not yet been established. Although experimental animal models are worthwhile, reproducible models that reflect the pathophysiology of NOMI have not been developed. Methods We combined risk factors for NOMI, comprising hemorrhagic shock, systemic vasopressor infusion, and local vasopressor infusion from the superior mesenteric artery (SMA) in swine under maintained anesthesia. Experiment 1 involved full-intensity (40%) phlebotomy and systemic vasopressor (norepinephrine and epinephrine). Experiment 2 involved full-intensity (40%) phlebotomy, systemic norepinephrine, and local vasopressor infusion into the SMA. Experiment 3 involved moderate (27%) phlebotomy, systemic norepinephrine infusion, and local epinephrine infusion. We evaluated serum lactate levels, intestinal serosa color, computed tomography (CT) angiography, and pathological findings. Results After inducing hemorrhage, systemic vasopressor alone and in combination with local vasopressin or norepinephrine infusion did not induce ischemic color changes in the intestine. The combination of systemic norepinephrine and local epinephrine (0.5 μg/kg/min) after moderate (27% blood loss) hemorrhage induced gross color change, pathological destruction, and elevation of serum lactate. Patent flow in the SMA was confirmed on CT angiography. Conclusion We established a swine NOMI model with systemic norepinephrine infusion and local epinephrine with moderate hemorrhagic shock.
Collapse
Affiliation(s)
- Yasutaka Tanaka
- Department of Emergency and Critical Care MedicineJichi Medical UniversityShimotsukeJapan
| | - Yosuke Matsumura
- Department of Intensive CareChiba Emergency Medical CenterChibaJapan
- Department of Emergency and Critical Care MedicineChiba UniversityChibaJapan
| | - Makoto Aoki
- Division of TraumatologyNational Defense Medical College Research InstituteTokorozawaJapan
| | - Yosuke Hayashi
- Department of Emergency and Critical Care MedicineChiba UniversityChibaJapan
| | - Yoshimitsu Izawa
- Department of Emergency and Critical Care MedicineJichi Medical UniversityShimotsukeJapan
| | - Kazuhiro Endo
- Center for Development of Advanced Medical TechnologyJichi Medical UniversityShimotsukeJapan
| | - Takashi Mato
- Department of Emergency and Critical Care MedicineJichi Medical UniversityShimotsukeJapan
| |
Collapse
|
5
|
Kurup S, Tan C, Kume T. Cardiac and intestinal tissue conduct developmental and reparative processes in response to lymphangiocrine signaling. Front Cell Dev Biol 2023; 11:1329770. [PMID: 38178871 PMCID: PMC10764504 DOI: 10.3389/fcell.2023.1329770] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 12/08/2023] [Indexed: 01/06/2024] Open
Abstract
Lymphatic vessels conduct a diverse range of activities to sustain the integrity of surrounding tissue. Besides facilitating the movement of lymph and its associated factors, lymphatic vessels are capable of producing tissue-specific responses to changes within their microenvironment. Lymphatic endothelial cells (LECs) secrete paracrine signals that bind to neighboring cell-receptors, commencing an intracellular signaling cascade that preludes modifications to the organ tissue's structure and function. While the lymphangiocrine factors and the molecular and cellular mechanisms themselves are specific to the organ tissue, the crosstalk action between LECs and adjacent cells has been highlighted as a commonality in augmenting tissue regeneration within animal models of cardiac and intestinal disease. Lymphangiocrine secretions have been owed for subsequent improvements in organ function by optimizing the clearance of excess tissue fluid and immune cells and stimulating favorable tissue growth, whereas perturbations in lymphatic performance bring about the opposite. Newly published landmark studies have filled gaps in our understanding of cardiac and intestinal maintenance by revealing key players for lymphangiocrine processes. Here, we will expand upon those findings and review the nature of lymphangiocrine factors in the heart and intestine, emphasizing its involvement within an interconnected network that supports daily homeostasis and self-renewal following injury.
Collapse
Affiliation(s)
- Shreya Kurup
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Honors College, University of Illinois at Chicago, Chicago, IL, United States
| | - Can Tan
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Tsutomu Kume
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
6
|
Bardell D, Rocchigiani G, Ressel L, Milner P. Histological Evaluation of Resected Tissue as a Predictor of Survival in Horses with Strangulating Small Intestinal Disease. Animals (Basel) 2023; 13:2715. [PMID: 37684979 PMCID: PMC10486948 DOI: 10.3390/ani13172715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/19/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
Strangulating small intestinal disease (SSID) in horses carries a poor prognosis for survival, especially following resection of ischaemic tissue. The margins of a resection are principally based on visual appraisal of the intestine during surgery. We hypothesized that histological evaluation of resected tissue may identify occult changes indicative of prognosis. Small intestinal samples from 18 horses undergoing resection for SSID and 9 horses euthanised for reasons unrelated to gastrointestinal pathology were utilised. Histological appearance was used to generate a 'total damage score' (TDS) for the control tissue, grossly normal tissue at oral and aboral extremities (sections OR1 and AB1) of the resected intestine, and oral and aboral extremities of visually abnormal tissue (sections OR2 and AB2) from SSID horses. The relationship between TDS and long-term post-operative survival was investigated. TDS was not different between control tissues and OR1 and AB1 sections. Five surgical cases were alive at follow-up, the longest follow-up time being 2561 days. Based on the median scores for SSID cases versus controls, cut-off values were generated to evaluate post-operative survival versus TDS. Only OR2 TDS was significantly associated with survival, with a higher (worse) score indicating longer survival. More severe tissue insult may expedite rapid progression to surgery, improving post-operative outcomes.
Collapse
Affiliation(s)
- David Bardell
- Department of Equine Clinical Sciences, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Leahurst Campus, Chester High Road, Neston CH64 7TE, UK;
| | - Guido Rocchigiani
- Department of Veterinary Anatomy Physiology and Pathology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Leahurst Campus, Chester High Road, Neston CH64 7TE, UK; (G.R.); (L.R.)
| | - Lorenzo Ressel
- Department of Veterinary Anatomy Physiology and Pathology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Leahurst Campus, Chester High Road, Neston CH64 7TE, UK; (G.R.); (L.R.)
| | - Peter Milner
- Department of Equine Clinical Sciences, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Leahurst Campus, Chester High Road, Neston CH64 7TE, UK;
| |
Collapse
|
7
|
Rivera KR, Bliton RJ, Burclaff J, Czerwinski MJ, Liu J, Trueblood JM, Hinesley CM, Breau KA, Deal HE, Joshi S, Pozdin VA, Yao M, Ziegler AL, Blikslager AT, Daniele MA, Magness ST. Hypoxia Primes Human ISCs for Interleukin-Dependent Rescue of Stem Cell Activity. Cell Mol Gastroenterol Hepatol 2023; 16:823-846. [PMID: 37562653 PMCID: PMC10520368 DOI: 10.1016/j.jcmgh.2023.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND AND AIMS Hypoxia in the intestinal epithelium can be caused by acute ischemic events or chronic inflammation in which immune cell infiltration produces inflammatory hypoxia starving the mucosa of oxygen. The epithelium has the capacity to regenerate after some ischemic and inflammatory conditions suggesting that intestinal stem cells (ISCs) are highly tolerant to acute and chronic hypoxia; however, the impact of hypoxia on human ISC (hISC) function has not been reported. Here we present a new microphysiological system (MPS) to investigate how hypoxia affects hISCs from healthy donors and test the hypothesis that prolonged hypoxia modulates how hISCs respond to inflammation-associated interleukins (ILs). METHODS hISCs were exposed to <1.0% oxygen in the MPS for 6, 24, 48, and 72 hours. Viability, hypoxia-inducible factor 1a (HIF1a) response, transcriptomics, cell cycle dynamics, and response to cytokines were evaluated in hISCs under hypoxia. HIF stabilizers and inhibitors were screened to evaluate HIF-dependent responses. RESULTS The MPS enables precise, real-time control and monitoring of oxygen levels at the cell surface. Under hypoxia, hISCs maintain viability until 72 hours and exhibit peak HIF1a at 24 hours. hISC activity was reduced at 24 hours but recovered at 48 hours. Hypoxia induced increases in the proportion of hISCs in G1 and expression changes in 16 IL receptors. Prolyl hydroxylase inhibition failed to reproduce hypoxia-dependent IL-receptor expression patterns. hISC activity increased when treated IL1β, IL2, IL4, IL6, IL10, IL13, and IL25 and rescued hISC activity caused by 24 hours of hypoxia. CONCLUSIONS Hypoxia pushes hISCs into a dormant but reversible proliferative state and primes hISCs to respond to a subset of ILs that preserves hISC activity. These findings have important implications for understanding intestinal epithelial regeneration mechanisms caused by inflammatory hypoxia.
Collapse
Affiliation(s)
- Kristina R Rivera
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, North Carolina
| | - R Jarrett Bliton
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, North Carolina
| | - Joseph Burclaff
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, North Carolina
| | - Michael J Czerwinski
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jintong Liu
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jessica M Trueblood
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Caroline M Hinesley
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Keith A Breau
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Halston E Deal
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, North Carolina
| | - Shlok Joshi
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Vladimir A Pozdin
- Department of Electrical and Computer Engineering, North Carolina State University, Raleigh, North Carolina
| | - Ming Yao
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, North Carolina
| | - Amanda L Ziegler
- Comparative Medicine Institute, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Anthony T Blikslager
- Comparative Medicine Institute, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Michael A Daniele
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, North Carolina; Department of Electrical and Computer Engineering, North Carolina State University, Raleigh, North Carolina
| | - Scott T Magness
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, North Carolina; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| |
Collapse
|
8
|
Mork C, Gahl B, Eckstein F, Berdajs DA. Prolonged cardiopulmonary bypass time as predictive factor for bloodstream infection. Heliyon 2023; 9:e17310. [PMID: 37383209 PMCID: PMC10293714 DOI: 10.1016/j.heliyon.2023.e17310] [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: 04/16/2023] [Revised: 06/09/2023] [Accepted: 06/13/2023] [Indexed: 06/30/2023] Open
Abstract
Objectives To evaluate the correlation between patient characteristics, operative variables and the risk of blood stream infection as well as the association of primary blood stream infection and adverse outcomes. Methods Clinical records of 6500 adult patients who underwent open heart surgery between February 2008 and October 2020 were analyzed. The microbiological pattern of the primary BSI and its association with adverse events, such as mortality and major cardiovascular events, were evaluated. Results Primary bloodstream infection was diagnosed in 1.7% (n = 108) of patients following cardiac surgery with the application of cardiopulmonary bypass. Most isolated bacteria were gram-negative bacillus groups, such as the Enterobacteriaceae family with Serrata marcescens in 26.26%, followed by the Enterococcaceae family with the Enterococcus faecalis in 7.39% and Enterococcus faecium in 9.14% as the most frequently identified bacteria. The postprocedural mortality, stroke rate p < 0.001, the incidence of postoperative new renal failure p < 0.001, and the renal replacement therapy p < 0.001 were significantly higher in the primary BSI group. Aortic cross-clamp time >120 min, OR 2.31 95%CI 1.34 to 3.98, perfusion time >120 min, OR 2.45 95%CI 1.63 to 3.67, and duration of the intervention >300min, OR 2.78 95%CI 1.47 to 5.28, were significantly related to the primary BSI. Conclusion The gram-negative bacillus was the most common microorganism identified in BSI after cardiovascular operations using cardiopulmonary bypass. Patients on dialysis prior to cardiac surgery are at higher risk for having BSI. Enteric bacterial translocation after prolonged cardiopulmonary bypass is a possible mechanism of early primary bloodstream infection in these patients. In patients at high risk, prophylactic use of an antibiotic regimen with broader gram-negative bacteria coverage should be considered, especially in those with prolonged cardiopulmonary bypass and intervention time.
Collapse
Affiliation(s)
- Constantin Mork
- Department of Cardiac Surgery, University Hospital Basel, Switzerland
- Department of Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, Switzerland
| | - Brigita Gahl
- Department of Cardiac Surgery, University Hospital Basel, Switzerland
- Surgical Outcome Research Center Basel, University Hospital Basel, Switzerland
| | | | - Denis A. Berdajs
- Department of Cardiac Surgery, University Hospital Basel, Switzerland
| |
Collapse
|
9
|
Kural T, Grajciarová M, Rosendorf J, Pálek R, Červenková L, Malečková A, Šarčevic S, Liška V, Tonar Z. Histological mapping of healing of the small and large intestine – a quantitative study in a porcine model. Ann Anat 2023; 249:152095. [PMID: 37011825 DOI: 10.1016/j.aanat.2023.152095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/02/2023] [Accepted: 03/22/2023] [Indexed: 04/04/2023]
Abstract
BACKGROUND Gastrointestinal anastomoses are performed in many patients every year. The pathogenesis of aberrant anastomotic healing and the causes of intestinal leakage are not fully understood. The present study gathered and critically evaluated histological quantitative data to deepen current knowledge of anastomotic healing in the small and large intestine and its complications and outline the options for further experimental in vivo research in large porcine animal models. METHODS Three groups of porcine intestinal anastomoses were compared: small intestine without defect (SI; n=7), small intestine with an additional defect (SID; n=8), and large intestine (LI; n=7). Multilevel sampling (2,112 micrographs) and stereological methods were used for histological quantification of proliferation (Ki-67 immunohistochemistry), neutrophil infiltration (myeloperoxidase staining), vascularity (von Willebrand factor) and type I and type III collagen formation (picrosirius red in polarized light) within the region of anastomosis compared to the region outside of anastomosis. RESULTS Quantitative histological evaluation revealed the following results. i) Proliferation, vascularity, and collagen, but not neutrophils, were more highly expressed within the anastomosis than outside of the anastomosis region. ii) Porcine large and small intestine were not interchangeable based on histological evaluation of surgical experiments. The presence or absence of an additional experimental defect strongly affected healing, but the healing seemed complete after 21 days. iii) The microscopic structure of small intestine segments was more affected by their proximity to the anastomosis than the structure of large intestine segments. CONCLUSIONS Histological quantification was more laborious than the previously used semiquantitative scoring system evaluating the healing rate of intestinal anastomoses, but it provided detailed maps of biological processes within individual intestine layers. The primary data collected in the study are open and available for power sample analyses to calculate the minimum numbers of samples justified in future experiments on porcine intestines. The porcine intestine is a promising animal model with translational potential for human surgery.
Collapse
|
10
|
Rivera KR, Bliton RJ, Burclaff J, Czerwinski MJ, Liu J, Trueblood JM, Hinesley CM, Breau KA, Joshi S, Pozdin VA, Yao M, Ziegler AL, Blikslager AT, Daniele MA, Magness ST. A new microphysiological system shows hypoxia primes human ISCs for interleukin-dependent rescue of stem cell activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.31.524747. [PMID: 36778265 PMCID: PMC9915581 DOI: 10.1101/2023.01.31.524747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Background & Aims Hypoxia in the intestinal epithelium can be caused by acute ischemic events or conditions like Inflammatory Bowel Disease (IBD) where immune cell infiltration produces 'inflammatory hypoxia', a chronic condition that starves the mucosa of oxygen. Epithelial regeneration after ischemia and IBD suggests intestinal stem cells (ISCs) are highly tolerant to acute and chronic hypoxia; however, the impact of acute and chronic hypoxia on human ISC (hISC) properties have not been reported. Here we present a new microphysiological system (MPS) to investigate how hypoxia affects hISCs isolated from healthy human tissues. We then test the hypothesis that some inflammation-associated interleukins protect hISCs during prolonged hypoxia. Methods hISCs were exposed to <1.0% oxygen in the MPS for 6-, 24-, 48- & 72hrs. Viability, HIF1α response, transcriptomics, cell cycle dynamics, and hISC response to cytokines were evaluated. Results The novel MPS enables precise, real-time control and monitoring of oxygen levels at the cell surface. Under hypoxia, hISCs remain viable until 72hrs and exhibit peak HIF1α at 24hrs. hISCs lose stem cell activity at 24hrs that recovers at 48hrs of hypoxia. Hypoxia increases the proportion of hISCs in G1 and regulates hISC capacity to respond to multiple inflammatory signals. Hypoxia induces hISCs to upregulate many interleukin receptors and hISCs demonstrate hypoxia-dependent cell cycle regulation and increased organoid forming efficiency when treated with specific interleukins. Conclusions Hypoxia primes hISCs to respond differently to interleukins than hISCs in normoxia through a transcriptional response. hISCs slow cell cycle progression and increase hISC activity when treated with hypoxia and specific interleukins. These findings have important implications for epithelial regeneration in the gut during inflammatory events.
Collapse
Affiliation(s)
- Kristina R. Rivera
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill 911 Oval Dr., Raleigh, NC, 27695 (USA)
| | - R. Jarrett Bliton
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill 911 Oval Dr., Raleigh, NC, 27695 (USA)
| | - Joseph Burclaff
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill 911 Oval Dr., Raleigh, NC, 27695 (USA)
| | - Michael J. Czerwinski
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599 (USA)
| | - Jintong Liu
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599 (USA)
| | - Jessica M. Trueblood
- Center for Gastrointestinal Biology and Disease, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Caroline M. Hinesley
- Center for Gastrointestinal Biology and Disease, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Keith A Breau
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599 (USA)
| | - Shlok Joshi
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599 (USA)
| | - Vladimir A. Pozdin
- Department of Electrical & Computer Engineering, North Carolina State University, Raleigh, NC, 27695 (USA)
| | - Ming Yao
- Department of Mechanical & Aerospace Engineering, North Carolina State University, Raleigh, NC 27695 (USA)
| | - Amanda L. Ziegler
- Center for Gastrointestinal Biology and Disease, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Anthony T. Blikslager
- Center for Gastrointestinal Biology and Disease, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Michael A. Daniele
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill 911 Oval Dr., Raleigh, NC, 27695 (USA)
- Department of Electrical & Computer Engineering, North Carolina State University, Raleigh, NC, 27695 (USA)
| | - Scott T. Magness
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill 911 Oval Dr., Raleigh, NC, 27695 (USA)
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599 (USA)
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599 (USA)
| |
Collapse
|
11
|
Garzelli L, Nuzzo A, Hamon A, Ben Abdallah I, Gregory J, Raynaud L, Paulatto L, Dioguardi Burgio M, Castier Y, Panis Y, Vilgrain V, Corcos O, Ronot M. Reperfusion injury on computed tomography following endovascular revascularization of acute mesenteric ischemia: prevalence, risk factors, and patient outcome. Insights Imaging 2022; 13:194. [PMID: 36512135 PMCID: PMC9748024 DOI: 10.1186/s13244-022-01339-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 11/22/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Data about reperfusion injury (RI) following acute arterial mesenteric ischemia (AAMI) in humans are scarce. We aimed to assess the prevalence and risk factors of RI following endovascular revascularization of AMI and evaluate its impact on patient outcomes. METHODS Patients with AAMI who underwent endovascular revascularization (2016-2021) were included in this retrospective cohort. CT performed < 7 days after treatment was reviewed to identify features of RI (bowel wall hypoattenuation, mucosal hyperenhancement). Clinical, laboratory, imaging, and treatments were compared between RI and non-RI patients to identify factors associated with RI. Resection rate and survival were also compared. RESULTS Fifty patients (23 men, median 72-yrs [IQR 60-77]) were included, and 22 were diagnosed with RI (44%) after a median 28 h (22-48). Bowel wall hypoattenuation and mucosal hyperenhancement were found in 95% and 91% of patients with post-interventional RI, respectively. Patients with RI had a greater increase of CRP levels after endovascular treatment (p = 0.01). On multivariate analysis, a decreased bowel wall enhancement on baseline CT (HR = 8.2), an embolic cause (HR = 7.4), complete SMA occlusion (HR = 7.0), and higher serum lactate levels (HR = 1.4) were associated with RI. The three-month survival rate was 78%, with no difference between subgroups (p = 0.99). However, the resection rate was higher in patients with RI (32% versus 7%; p = 0.03). CONCLUSION RI is frequent after endovascular revascularization of AAMI, especially in patients who present with decreased bowel wall enhancement on pre-treatment CT, an embolic cause, and a complete occlusion of the SMA. However, its occurrence does not seem to negatively impact short-term survival.
Collapse
Affiliation(s)
- Lorenzo Garzelli
- grid.508487.60000 0004 7885 7602Université Paris Cité, Paris, France ,grid.411599.10000 0000 8595 4540Service de Radiologie, Hôpital Beaujon, APHP.Nord, 100 Blvd du Général Leclerc, 92118 Clichy, France
| | - Alexandre Nuzzo
- grid.411599.10000 0000 8595 4540Intestinal Stroke Center, Service de Gastroenterology, MICI Et Insuffisance Intestinale, Hôpital Beaujon, APHP.Nord, Clichy, France
| | - Annick Hamon
- grid.411599.10000 0000 8595 4540Intestinal Stroke Center, Service de Gastroenterology, MICI Et Insuffisance Intestinale, Hôpital Beaujon, APHP.Nord, Clichy, France
| | - Iannis Ben Abdallah
- grid.508487.60000 0004 7885 7602Université Paris Cité, Paris, France ,grid.411119.d0000 0000 8588 831XService de Chirurgie Vasculaire, Hôpital Bichat, APHP.Nord, Paris, France
| | - Jules Gregory
- grid.508487.60000 0004 7885 7602Université Paris Cité, Paris, France ,grid.411599.10000 0000 8595 4540Service de Radiologie, Hôpital Beaujon, APHP.Nord, 100 Blvd du Général Leclerc, 92118 Clichy, France
| | - Lucas Raynaud
- grid.508487.60000 0004 7885 7602Université Paris Cité, Paris, France ,grid.411599.10000 0000 8595 4540Service de Radiologie, Hôpital Beaujon, APHP.Nord, 100 Blvd du Général Leclerc, 92118 Clichy, France
| | - Luisa Paulatto
- grid.508487.60000 0004 7885 7602Université Paris Cité, Paris, France ,grid.411599.10000 0000 8595 4540Service de Radiologie, Hôpital Beaujon, APHP.Nord, 100 Blvd du Général Leclerc, 92118 Clichy, France
| | - Marco Dioguardi Burgio
- grid.508487.60000 0004 7885 7602Université Paris Cité, Paris, France ,grid.411599.10000 0000 8595 4540Service de Radiologie, Hôpital Beaujon, APHP.Nord, 100 Blvd du Général Leclerc, 92118 Clichy, France
| | - Yves Castier
- grid.508487.60000 0004 7885 7602Université Paris Cité, Paris, France ,grid.411119.d0000 0000 8588 831XService de Chirurgie Vasculaire, Hôpital Bichat, APHP.Nord, Paris, France
| | - Yves Panis
- grid.508487.60000 0004 7885 7602Université Paris Cité, Paris, France ,grid.411119.d0000 0000 8588 831XService de Chirurgie Digestive, Hôpital Bichat, APHP.Nord, Paris, France
| | - Valérie Vilgrain
- grid.508487.60000 0004 7885 7602Université Paris Cité, Paris, France ,grid.411599.10000 0000 8595 4540Service de Radiologie, Hôpital Beaujon, APHP.Nord, 100 Blvd du Général Leclerc, 92118 Clichy, France
| | - Olivier Corcos
- grid.411599.10000 0000 8595 4540Intestinal Stroke Center, Service de Gastroenterology, MICI Et Insuffisance Intestinale, Hôpital Beaujon, APHP.Nord, Clichy, France
| | - Maxime Ronot
- grid.508487.60000 0004 7885 7602Université Paris Cité, Paris, France ,grid.411599.10000 0000 8595 4540Service de Radiologie, Hôpital Beaujon, APHP.Nord, 100 Blvd du Général Leclerc, 92118 Clichy, France
| |
Collapse
|
12
|
Scott BB, Wang Y, Wu RC, Randolph MA, Redmond RW. Light-activated photosealing with human amniotic membrane strengthens bowel anastomosis in a hypotensive, trauma-relevant swine model. Lasers Surg Med 2022; 54:407-417. [PMID: 34664720 DOI: 10.1002/lsm.23485] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 09/27/2021] [Accepted: 10/04/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND Gastrointestinal anastomotic leakage is a dreaded complication despite advancements in surgical technique. Photochemical tissue bonding (PTB) is a method of sealing tissue surfaces utilizing photoactive dye. We evaluated if crosslinked human amniotic membrane (xHAM) photosealed over the enteroenterostomy would augment anastomotic strength in a trauma-relevant swine hemorrhagic shock model. METHODS Yorkshire swine (40-45 kg, n = 14) underwent midline laparotomy and sharp transection of the small intestine 120 cm proximal to the ileocecal fold. Immediately following intestinal transection, a controlled arterial bleed was performed to reach hemorrhagic shock. Intestinal repair was performed after 60 minutes and autotransfusion of the withdrawn blood was performed for resuscitation. Animals were randomized to small intestinal anastomosis by one of the following methods (seven per group): suture repair (SR), or SR with PTB augmentation. Animals were euthanized at postoperative Day 28 and burst pressure (BP) strength testing was performed on all excised specimens. RESULTS Mean BP for SR, PTB, and native tissue groups were 229 ± 40, 282 ± 21, and 282 ± 47 mmHg, respectively, with the SR group statistically significantly different on analysis of variance (p = 0.02). Post-hoc Tukey all-pairs comparison demonstrated a statistically significant difference in burst pressure strength between the SR only and the PTB group (p = 0.04). All specimens in SR group ruptured at the anastomosis upon burst pressure testing, while all specimens in the PTB group ruptured at least 2.5 cm from the anastomosis. CONCLUSION Photosealing with xHAM significantly augments the strength of small intestinal anastomosis performed in a trauma porcine model.
Collapse
Affiliation(s)
- Benjamin B Scott
- Wellman Center for Photomedicine, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
- Plastic Surgery Research Laboratory, Department of Surgery, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ying Wang
- Wellman Center for Photomedicine, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ruby C Wu
- Wellman Center for Photomedicine, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Mark A Randolph
- Plastic Surgery Research Laboratory, Department of Surgery, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Robert W Redmond
- Wellman Center for Photomedicine, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
13
|
Age-Dependent Intestinal Repair: Implications for Foals with Severe Colic. Animals (Basel) 2021; 11:ani11123337. [PMID: 34944114 PMCID: PMC8697879 DOI: 10.3390/ani11123337] [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: 11/03/2021] [Revised: 11/17/2021] [Accepted: 11/20/2021] [Indexed: 11/17/2022] Open
Abstract
Colic is a leading cause of death in horses, with the most fatal form being strangulating obstruction which directly damages the intestinal barrier. Following surgical intervention, it is imperative that the intestinal barrier rapidly repairs to prevent translocation of gut bacteria and their products and ensure survival of the patient. Age-related disparities in survival have been noted in many species, including horses, humans, and pigs, with younger patients suffering poorer clinical outcomes. Maintenance and repair of the intestinal barrier is regulated by a complex mucosal microenvironment, of which the ENS, and particularly a developing network of subepithelial enteric glial cells, may be of particular importance in neonates with colic. Postnatal development of an immature enteric glial cell network is thought to be driven by the microbial colonization of the gut and therefore modulated by diet-influenced changes in bacterial populations early in life. Here, we review the current understanding of the roles of the gut microbiome, nutrition, stress, and the ENS in maturation of intestinal repair mechanisms after foaling and how this may influence age-dependent outcomes in equine colic cases.
Collapse
|
14
|
Suzuki Y, Yamamoto M, Sugiyama K, Akai T, Suzuki K, Kawamura T, Sakata M, Morita Y, Kikuchi H, Hiramatsu Y, Kurachi K, Unno N, Takeuchi H. Usefulness of a finger-mounted tissue oximeter with near-infrared spectroscopy for evaluating the intestinal oxygenation and viability in rats. Surg Today 2021; 51:931-940. [PMID: 33108523 PMCID: PMC8141489 DOI: 10.1007/s00595-020-02171-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/26/2020] [Indexed: 12/04/2022]
Abstract
PURPOSE To investigate the utility of the device for evaluating intestinal oxygenation and viability using an animal model. METHODS Sprague-Dawley rats underwent laparotomy under general anesthesia, and the blood vessels in the terminal ileum were clamped to create ischemia. We measured the regional tissue oxygenation saturation (rSO2) using an oximeter after 1, 3, and 6 h of vessel clamping. Ischemic tissue damage was assessed using a histological score. The intestine was reperfused after each clamping period, and intestinal rSO2 and survival rate were evaluated. RESULTS When reperfusion was performed at 1 and 3 h after ischemia, rSO2 increased after 10 min, and it improved to the same level as for normal intestine after 1 h; all rats survived for 1 week. In contrast, after 6 h of ischemia, rSO2 did not increase after reperfusion, and all animals died within 2 days. The histological scores increased after 1 h of reperfusion, with longer clamping periods. CONCLUSION A finger-mounted tissue oximeter could evaluate intestinal ischemia and the viability, which is thus considered to be a promising result for future clinical application.
Collapse
Affiliation(s)
- Yuhi Suzuki
- Second Department of Surgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan.
| | - Masayoshi Yamamoto
- Second Department of Surgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Kosuke Sugiyama
- Second Department of Surgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Toshiya Akai
- Second Department of Surgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Katsunori Suzuki
- Second Department of Surgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Takafumi Kawamura
- Second Department of Surgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Mayu Sakata
- Second Department of Surgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Yoshifumi Morita
- Second Department of Surgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Hirotoshi Kikuchi
- Second Department of Surgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Yoshihiro Hiramatsu
- Second Department of Surgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Kiyotaka Kurachi
- Second Department of Surgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Naoki Unno
- Second Department of Surgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan
- Division of Vascular Surgery, Hamamatsu Medical Center, 328 Tomitsuka, Hamamatsu, Shizuoka, 432-8580, Japan
| | - Hiroya Takeuchi
- Second Department of Surgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan
| |
Collapse
|
15
|
Seeger B. Farm Animal-derived Models of the Intestinal Epithelium: Recent Advances and Future Applications of Intestinal Organoids. Altern Lab Anim 2020; 48:215-233. [PMID: 33337913 DOI: 10.1177/0261192920974026] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Farm animals play an important role in translational research as large animal models of the gastrointestinal (GI) tract. The mechanistic investigation of zoonotic diseases of the GI tract, in which animals can act as asymptomatic carriers, could provide important information for therapeutic approaches. In veterinary medicine, farm animals are no less relevant, as they can serve as models for the development of diagnostic and therapeutic approaches of GI diseases in the target species. However, farm animal-derived cell lines of the intestinal epithelium are rarely available from standardised cell banks and, in addition, are not usually specific for certain sections of the intestine. Immortalised porcine or bovine enterocytic cell lines are more widely available, compared to goat or sheep-derived cell lines; no continuous cell lines are available from the chicken. Other epithelial cell types with intestinal section-specific distribution and function, such as goblet cells, enteroendocrine cells, Paneth cells and intestinal stem cells, are not represented in those cell line-based models. Therefore, intestinal organoid models of farm animal species, which are already widely used for mice and humans, are gaining importance. Crypt-derived or pluripotent stem cell-derived intestinal organoid models offer the possibility to investigate the mechanisms of inter-cell or host-pathogen interactions and to answer species-specific questions. This review is intended to give an overview of cell culture models of the intestinal epithelium of farm animals, discussing species-specific differences, culture techniques and some possible applications for intestinal organoid models. It also highlights the need for species-specific pluripotent stem cell-derived or crypt-derived intestinal organoid models for promotion of the Three Rs principles (replacement, reduction and refinement).
Collapse
Affiliation(s)
- Bettina Seeger
- Department of Food Toxicology and Replacement/Complementary Methods to Animal Testing, Institute for Food Toxicology, 460510University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| |
Collapse
|
16
|
Søfteland JM, Casselbrant A, Biglarnia AR, Linders J, Hellström M, Pesce A, Padma AM, Jiga LP, Hoinoiu B, Ionac M, Oltean M. Intestinal Preservation Injury: A Comparison Between Rat, Porcine and Human Intestines. Int J Mol Sci 2019; 20:3135. [PMID: 31252560 PMCID: PMC6650817 DOI: 10.3390/ijms20133135] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 06/25/2019] [Indexed: 02/07/2023] Open
Abstract
Advanced preservation injury (PI) after intestinal transplantation has deleterious short- and long-term effects and constitutes a major research topic. Logistics and costs favor rodent studies, whereas clinical translation mandates studies in larger animals or using human material. Despite diverging reports, no direct comparison between the development of intestinal PI in rats, pigs, and humans is available. We compared the development of PI in rat, porcine, and human intestines. Intestinal procurement and cold storage (CS) using histidine-tryptophan-ketoglutarate solution was performed in rats, pigs, and humans. Tissue samples were obtained after 8, 14, and 24 h of CS), and PI was assessed morphologically and at the molecular level (cleaved caspase-3, zonula occludens, claudin-3 and 4, tricellulin, occludin, cytokeratin-8) using immunohistochemistry and Western blot. Intestinal PI developed slower in pigs compared to rats and humans. Tissue injury and apoptosis were significantly higher in rats. Tight junction proteins showed quantitative and qualitative changes differing between species. Significant interspecies differences exist between rats, pigs, and humans regarding intestinal PI progression at tissue and molecular levels. These differences should be taken into account both with regards to study design and the interpretation of findings when relating them to the clinical setting.
Collapse
Affiliation(s)
- John Mackay Søfteland
- The Transplant Institute, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
- Laboratory for Transplantation and Regenerative Medicine, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Sahlgrenska Science Park Medicinaregatan 8, 413 90 Gothenburg, Sweden
| | - Anna Casselbrant
- Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden
| | - Ali-Reza Biglarnia
- Department of Transplantation, Skåne University Hospital, 205 02 Malmö, Sweden
| | - Johan Linders
- Department of Transplantation, Skåne University Hospital, 205 02 Malmö, Sweden
| | - Mats Hellström
- Laboratory for Transplantation and Regenerative Medicine, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Sahlgrenska Science Park Medicinaregatan 8, 413 90 Gothenburg, Sweden
| | - Antonio Pesce
- Department of Medical and Surgical Sciences and Advanced Technologies, University of Catania, Via Santa Sofia 86, 95123 Catania, Italy
| | - Arvind Manikantan Padma
- Laboratory for Transplantation and Regenerative Medicine, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Sahlgrenska Science Park Medicinaregatan 8, 413 90 Gothenburg, Sweden
| | - Lucian Petru Jiga
- Department for Plastic, Aesthetic, Reconstructive and Hand Surgery, Evangelisches Krankenhaus Oldenburg, Medical Campus University of Oldenburg, Steinweg 13-17, 26122 Oldenburg, Germany
| | - Bogdan Hoinoiu
- Pius Branzeu Center for Laparoscopic Surgery and Microsurgery, University of Medicine and Pharmacy, P-ta. E. Murgu 2, 300041 Timisoara, Romania
| | - Mihai Ionac
- Pius Branzeu Center for Laparoscopic Surgery and Microsurgery, University of Medicine and Pharmacy, P-ta. E. Murgu 2, 300041 Timisoara, Romania
| | - Mihai Oltean
- The Transplant Institute, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden.
- Laboratory for Transplantation and Regenerative Medicine, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Sahlgrenska Science Park Medicinaregatan 8, 413 90 Gothenburg, Sweden.
| |
Collapse
|
17
|
Gonzalez LM, Stewart AS, Freund J, Kucera CR, Dekaney CM, Magness ST, Blikslager AT. Preservation of reserve intestinal epithelial stem cells following severe ischemic injury. Am J Physiol Gastrointest Liver Physiol 2019; 316:G482-G494. [PMID: 30714814 PMCID: PMC6483022 DOI: 10.1152/ajpgi.00262.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Intestinal ischemia is an abdominal emergency with a mortality rate >50%, leading to epithelial barrier loss and subsequent sepsis. Epithelial renewal and repair after injury depend on intestinal epithelial stem cells (ISC) that reside within the crypts of Lieberkühn. Two ISC populations critical to epithelial repair have been described: 1) active ISC (aISC; highly proliferative; leucine-rich-repeat-containing G protein-coupled receptor 5 positive, sex determining region Y-box 9 positive) and 2) reserve ISC [rISC; less proliferative; homeodomain only protein X (Hopx)+]. Yorkshire crossbred pigs (8-10 wk old) were subjected to 1-4 h of ischemia and 1 h of reperfusion or recovery by reversible mesenteric vascular occlusion. This study was designed to evaluate whether ISC-expressing biomarkers of aISCs or rISCs show differential resistance to ischemic injury and different contributions to the subsequent repair and regenerative responses. Our data demonstrate that, following 3-4 h ischemic injury, aISC undergo apoptosis, whereas rISC are preserved. Furthermore, these rISC are retained ex vivo in spheroids in which cell populations are enriched in the rISC biomarker Hopx. These cells appear to go on to provide a proliferative pool of cells during the recovery period. Taken together, these data indicate that Hopx+ cells are resistant to injury and are the likely source of epithelial renewal following prolonged ischemic injury. It is therefore possible that targeting reserve stem cells will lead to new therapies for patients with severe intestinal injury. NEW & NOTEWORTHY The population of reserve less-proliferative intestinal epithelial stem cells appears resistant to injury despite severe epithelial cell loss, including that of the active stem cell population, which results from prolonged mesenteric ischemia. These cells can change to an activated state and are likely indispensable to regenerative processes. Reserve stem cell targeted therapies may improve treatment and outcome of patients with ischemic disease.
Collapse
Affiliation(s)
- Liara M. Gonzalez
- 1Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina,4Center for Gastrointestinal Biologyand Disease, Joint Center at University of North Carolina Chapel Hill and North Carolina State University, Raleigh,North Carolina
| | - Amy Stieler Stewart
- 1Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - John Freund
- 1Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Cecilia Renee Kucera
- 1Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Christopher M. Dekaney
- 2Department of Molecular and Biological Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina,4Center for Gastrointestinal Biologyand Disease, Joint Center at University of North Carolina Chapel Hill and North Carolina State University, Raleigh,North Carolina
| | - Scott T. Magness
- 3University of North Carolina, Chapel Hill, North Carolina,4Center for Gastrointestinal Biologyand Disease, Joint Center at University of North Carolina Chapel Hill and North Carolina State University, Raleigh,North Carolina
| | - Anthony T. Blikslager
- 1Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina,4Center for Gastrointestinal Biologyand Disease, Joint Center at University of North Carolina Chapel Hill and North Carolina State University, Raleigh,North Carolina
| |
Collapse
|
18
|
Xiong X, Tan B, Song M, Ji P, Kim K, Yin Y, Liu Y. Nutritional Intervention for the Intestinal Development and Health of Weaned Pigs. Front Vet Sci 2019; 6:46. [PMID: 30847348 PMCID: PMC6393345 DOI: 10.3389/fvets.2019.00046] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 02/04/2019] [Indexed: 01/20/2023] Open
Abstract
Weaning imposes simultaneous stress, resulting in reduced feed intake, and growth rate, and increased morbidity and mortality of weaned pigs. Weaning impairs the intestinal integrity, disturbs digestive and absorptive capacity, and increases the intestinal oxidative stress, and susceptibility of diseases in piglets. The improvement of intestinal development and health is critically important for enhancing nutrient digestibility capacity and disease resistance of weaned pigs, therefore, increasing their survival rate at this most vulnerable stage, and overall productive performance during later stages. A healthy gut may include but not limited several important features: a healthy proliferation of intestinal epithelial cells, an integrated gut barrier function, a preferable or balanced gut microbiota, and a well-developed intestinal mucosa immunity. Burgeoning evidence suggested nutritional intervention are one of promising measures to enhance intestinal health of weaned pigs, although the exact protective mechanisms may vary and are still not completely understood. Previous research indicated that functional amino acids, such as arginine, cysteine, glutamine, or glutamate, may enhance intestinal mucosa immunity (i.e., increased sIgA secretion), reduce oxidative damage, stimulate proliferation of enterocytes, and enhance gut barrier function (i.e., enhanced expression of tight junction protein) of weaned pigs. A number of feed additives are marketed to assist in boosting intestinal immunity and regulating gut microbiota, therefore, reducing the negative impacts of weaning, and other environmental challenges on piglets. The promising results have been demonstrated in antimicrobial peptides, clays, direct-fed microbials, micro-minerals, milk components, oligosaccharides, organic acids, phytochemicals, and many other feed additives. This review summarizes our current understanding of nutritional intervention on intestinal health and development of weaned pigs and the importance of mechanistic studies focusing on this research area.
Collapse
Affiliation(s)
- Xia Xiong
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Bie Tan
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Minho Song
- Department of Animal Science and Biotechnology, Chungnam National University, Daejeon, South Korea
| | - Peng Ji
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Kwangwook Kim
- Department of Animal Science, University of California, Davis, Davis, CA, United States
| | - Yulong Yin
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Yanhong Liu
- Department of Animal Science, University of California, Davis, Davis, CA, United States
| |
Collapse
|
19
|
Stieler Stewart A, Freund JM, Blikslager AT, Gonzalez LM. Intestinal Stem Cell Isolation and Culture in a Porcine Model of Segmental Small Intestinal Ischemia. J Vis Exp 2018. [PMID: 29863654 PMCID: PMC6101266 DOI: 10.3791/57647] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Intestinal ischemia remains a major cause of morbidity and mortality in human and veterinary patients. Many disease processes result in intestinal ischemia, when the blood supply and therefore oxygen is decreased to the intestine. This leads to intestinal barrier loss and damage to the underlying tissue. Intestinal stem cells reside at the base of the crypts of Lieberkühn and are responsible for intestinal renewal during homeostasis and following injury. Ex vivo cell culture techniques have allowed for the successful study of epithelial stem cell interactions by establishing culture conditions that support the growth of three-dimensional epithelial organ-like systems (termed "enteroids" and "colonoids" from the small and large intestine, respectively). These enteroids are composed of crypt and villus-like domains and mature to contain all of the cell types found within the epithelium. Historically, murine models have been utilized to study intestinal injury. However, a porcine model offers several advantages including similarity of size as well as gastrointestinal anatomy and physiology to that of humans. By utilizing a porcine model, we establish a protocol in which segmental loops of intestinal ischemia can be created within a single animal, enabling the study of differing time points of ischemic injury and repair in vivo. Additionally, we describe a method to isolate and culture the intestinal stem cells from the ischemic loops of intestine, allowing for the continued study of epithelial repair, modulated by stem cells, ex vivo.
Collapse
Affiliation(s)
| | - John M Freund
- Department of Clinical Sciences, North Carolina State University
| | | | - Liara M Gonzalez
- Department of Clinical Sciences, North Carolina State University;
| |
Collapse
|
20
|
Strand-Amundsen RJ, Reims HM, Reinholt FP, Ruud TE, Yang R, Høgetveit JO, Tønnessen TI. Ischemia/reperfusion injury in porcine intestine - Viability assessment. World J Gastroenterol 2018; 24:2009-2023. [PMID: 29760544 PMCID: PMC5949714 DOI: 10.3748/wjg.v24.i18.2009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 04/20/2018] [Accepted: 04/23/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate viability assessment of segmental small bowel ischemia/reperfusion in a porcine model.
METHODS In 15 pigs, five or six 30-cm segments of jejunum were simultaneously made ischemic by clamping the mesenteric arteries and veins for 1 to 16 h. Reperfusion was initiated after different intervals of ischemia (1-8 h) and subsequently monitored for 5-15 h. The intestinal segments were regularly photographed and assessed visually and by palpation. Intraluminal lactate and glycerol concentrations were measured by microdialysis, and samples were collected for light microscopy and transmission electron microscopy. The histological changes were described and graded.
RESULTS Using light microscopy, the jejunum was considered as viable until 6 h of ischemia, while with transmission electron microscopy the ischemic muscularis propria was considered viable until 5 h of ischemia. However, following ≥ 1 h of reperfusion, only segments that had been ischemic for ≤ 3 h appeared viable, suggesting a possible upper limit for viability in the porcine mesenteric occlusion model. Although intraluminal microdialysis allowed us to closely monitor the onset and duration of ischemia and the onset of reperfusion, we were unable to find sufficient level of association between tissue viability and metabolic markers to conclude that microdialysis is clinically relevant for viability assessment. Evaluation of color and motility appears to be poor indicators of intestinal viability.
CONCLUSION Three hours of total ischemia of the small bowel followed by reperfusion appears to be the upper limit for viability in this porcine mesenteric ischemia model.
Collapse
Affiliation(s)
- Runar J Strand-Amundsen
- Department of Clinical and Biomedical Engineering, Oslo University Hospital, Oslo 0424, Norway
- Department of Physics, University of Oslo, Oslo 0316, Norway
| | - Henrik M Reims
- Department of Pathology, Oslo University Hospital, Oslo 0424, Norway
| | - Finn P Reinholt
- Department of Pathology, Oslo University Hospital, Oslo 0424, Norway
| | - Tom E Ruud
- Institute for Surgical Research, Oslo University Hospital, Oslo 0424, Norway
- Department of Surgery, Baerum Hospital, Vestre Viken Hospital Trust, Drammen 3004, Norway
| | - Runkuan Yang
- Department of Emergencies and Critical Care, Oslo University Hospital, Oslo 0424, Norway
| | - Jan O Høgetveit
- Department of Clinical and Biomedical Engineering, Oslo University Hospital, Oslo 0424, Norway
- Department of Physics, University of Oslo, Oslo 0316, Norway
| | - Tor I Tønnessen
- Department of Emergencies and Critical Care, Oslo University Hospital, Oslo 0424, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo 0424, Norway
| |
Collapse
|
21
|
|
22
|
Moeser AJ, Pohl CS, Rajput M. Weaning stress and gastrointestinal barrier development: Implications for lifelong gut health in pigs. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2017; 3:313-321. [PMID: 29767141 PMCID: PMC5941262 DOI: 10.1016/j.aninu.2017.06.003] [Citation(s) in RCA: 261] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 06/15/2017] [Accepted: 06/20/2017] [Indexed: 12/14/2022]
Abstract
The gastrointestinal (GI) barrier serves a critical role in survival and overall health of animals and humans. Several layers of barrier defense mechanisms are provided by the epithelial, immune and enteric nervous systems. Together they act in concert to control normal gut functions (e.g., digestion, absorption, secretion, immunity, etc.) whereas at the same time provide a barrier from the hostile conditions in the luminal environment. Breakdown of these critical GI functions is a central pathophysiological mechanism in the most serious GI disorders in pigs. This review will focus on the development and functional properties of the GI barrier in pigs and how common early life production stressors, such as weaning, can alter immediate and long-term barrier function and disease susceptibility. Specific stress-related pathophysiological mechanisms responsible for driving GI barrier dysfunction induced by weaning and the implications to animal health and performance will be discussed.
Collapse
Affiliation(s)
- Adam J. Moeser
- Gastrointestinal Stress Biology Laboratory, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
- Neuroscience Program, Michigan State University, East Lansing, MI 48824, USA
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
- Corresponding author.
| | - Calvin S. Pohl
- Gastrointestinal Stress Biology Laboratory, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Mrigendra Rajput
- Gastrointestinal Stress Biology Laboratory, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
23
|
Abstract
The equine intestinal mucosa is intimately involved in maintaining homeostasis both on a systemic level by controlling extracellular fluid movement and at the local level to maintain barrier function. Horses are particularly susceptible to the clinical syndrome of colic, with the most severe cases involving strangulating obstruction that induces ischemia. Because of the mucosal vascular architecture, the mucosal epithelium is particularly susceptible to ischemic injury. The potential for reperfusion injury has been investigated and found to play a minimal role. However, inflammation does affect mucosal repair. Mechanisms of repair, including villus contraction, epithelial restitution, and tight junction closure, are critical to reforming the mucosal barrier. Nonsteroidal anti-inflammatory drugs have an impact on this repair, particularly at the level of the tight junctions. Completion of mucosal regeneration requires proliferation, which is now being actively studied in equine enteroids. All of these aspects of equine mucosal pathobiology are reviewed in depth.
Collapse
Affiliation(s)
- Anthony Blikslager
- Center for Gastrointestinal Biology and Disease, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina 27607, USA;
| | - Liara Gonzalez
- Center for Gastrointestinal Biology and Disease, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina 27607, USA;
| |
Collapse
|
24
|
AlKukhun A, Caturegli G, Munoz-Abraham AS, Judeeba S, Patron-Lozano R, Morotti R, Rodriguez-Davalos MI, Geibel JP. Use of Fluorescein Isothiocyanate-Inulin as a Marker for Intestinal Ischemic Injury. J Am Coll Surg 2017; 224:1066-1073. [DOI: 10.1016/j.jamcollsurg.2016.12.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 10/09/2016] [Accepted: 12/05/2016] [Indexed: 12/14/2022]
|
25
|
Abstract
Gastrointestinal disease is a prevalent cause of morbidity and mortality and the use of animal models have been instrumental in studying mechanisms of digestive pathophysiology. As investigators attempt to translate the wealth of basic science information developed from rodent, models, large animal models provide a number of translational advantages. The pig, in particular, is arguably one of the most powerful models of human organ systems, including the gastrointestinal tract. The pig has provided important tools and insight into intestinal ischemia/reperfusion injury, intestinal mucosal repair, as well as new insights into esophageal injury and repair. Porcine model development has taken advantage of the size of the animal, allowing increased surgical and endoscopic access. In addition, cellular tools such as the intestinal porcine epithelial cell line and porcine enteroids are providing the methodology to translate basic science findings using in-depth mechanistic analyses. Further opportunities in porcine digestive disease modeling include developing additional transgenic pig strains. Collectively, porcine models hold great promise for the future of clinically relevant digestive disease research.
Collapse
|
26
|
Computed Tomography Perfusion Imaging Detection of Microcirculatory Dysfunction in Small Intestinal Ischemia-Reperfusion Injury in a Porcine Model. PLoS One 2016; 11:e0160102. [PMID: 27458696 PMCID: PMC4961382 DOI: 10.1371/journal.pone.0160102] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 07/13/2016] [Indexed: 01/19/2023] Open
Abstract
Objective To evaluate multi-slice computed tomography (CT) perfusion imaging (CTPI) for identifying microcirculatory dysfunction in small intestinal ischemia−reperfusion (IR) injury in a porcine model. Materials and Methods Fifty-two pigs were randomly divided into 4 groups: (1) the IR group (n = 24), where intestinal ischemia was induced by separating and clamping the superior mesenteric artery (SMA) for 2 h, followed by reperfusion for 1, 2, 3, and 4 h (IR-1h, IR-2h, IR-3h, and IR-4h; n = 6, respectively); (2) the sham-operated (SO) group (n = 20), where the SMA was separated without clamping and controlled at postoperative 3, 4, 5, and 6 h (SO-3h, SO-4h, SO-5h, and SO-6h; n = 5, respectively); (3) the ischemia group (n = 4), where the SMA was separated and clamped for 2 h, without reperfusion, and (4) baseline group (n = 4), an additional group that was not manipulated. Small intestinal CTPI was performed at corresponding time points and perfusion parameters were obtained. The distal ileum was resected to measure the concentrations of malondialdehyde (MDA) and superoxide dismutase (SOD) and for histopathological examination. Results The perfusion parameters of the IR groups showed significant differences compared with the corresponding SO groups and the baseline group (before ischemia). The blood flow (BF), blood volume (BV), and permeability surface (PS) among the 4 IR groups were significantly different. BF and BV were significantly negatively correlated with MDA, and significantly positively correlated with SOD in the IR groups. Histopathologically, the effects of the 2-h ischemic loops were not significantly exacerbated by reperfusion. Conclusion CTPI can be a valuable tool for detecting microcirculatory dysfunction and for dynamic monitoring of small intestinal IR injury.
Collapse
|
27
|
Gubernatorova EO, Perez-Chanona E, Koroleva EP, Jobin C, Tumanov AV. Murine Model of Intestinal Ischemia-reperfusion Injury. J Vis Exp 2016. [PMID: 27213580 DOI: 10.3791/53881] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Intestinal ischemia is a life-threatening condition associated with a broad range of clinical conditions including atherosclerosis, thrombosis, hypotension, necrotizing enterocolitis, bowel transplantation, trauma and chronic inflammation. Intestinal ischemia-reperfusion (IR) injury is a consequence of acute mesenteric ischemia, caused by inadequate blood flow through the mesenteric vessels, resulting in intestinal damage. Reperfusion following ischemia can further exacerbate damage of the intestine. The mechanisms of IR injury are complex and poorly understood. Therefore, experimental small animal models are critical for understanding the pathophysiology of IR injury and the development of novel therapies. Here we describe a mouse model of acute intestinal IR injury that provides reproducible injury of the small intestine without mortality. This is achieved by inducing ischemia in the region of the distal ileum by temporally occluding the peripheral and terminal collateral branches of the superior mesenteric artery for 60 min using microvascular clips. Reperfusion for 1 hr, or 2 hr after injury results in reproducible injury of the intestine examined by histological analysis. Proper position of the microvascular clips is critical for the procedure. Therefore the video clip provides a detailed visual step-by-step description of this technique. This model of intestinal IR injury can be utilized to study the cellular and molecular mechanisms of injury and regeneration.
Collapse
Affiliation(s)
| | - Ernesto Perez-Chanona
- Departments of Medicine and Infectious Diseases and Pathology, University of Florida
| | | | - Christian Jobin
- Departments of Medicine and Infectious Diseases and Pathology, University of Florida
| | | |
Collapse
|
28
|
Grootjans J, Lenaerts K, Buurman WA, Dejong CHC, Derikx JPM. Life and death at the mucosal-luminal interface: New perspectives on human intestinal ischemia-reperfusion. World J Gastroenterol 2016; 22:2760-2770. [PMID: 26973414 PMCID: PMC4777998 DOI: 10.3748/wjg.v22.i9.2760] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 09/24/2015] [Accepted: 12/14/2015] [Indexed: 02/06/2023] Open
Abstract
Intestinal ischemia is a frequently observed phenomenon. Morbidity and mortality rates are extraordinarily high and did not improve over the past decades. This is in part attributable to limited knowledge on the pathophysiology of intestinal ischemia-reperfusion (IR) in man, the paucity in preventive and/or therapeutic options and the lack of early diagnostic markers for intestinal ischemia. To improve our knowledge and solve clinically important questions regarding intestinal IR, we developed a human experimental intestinal IR model. With this model, we were able to gain insight into the mechanisms that allow the human gut to withstand short periods of IR without the development of severe inflammatory responses. The purpose of this review is to overview the most relevant recent advances in our understanding of the pathophysiology of human intestinal IR, as well as the (potential) future clinical implications.
Collapse
|
29
|
Porcine models of digestive disease: the future of large animal translational research. Transl Res 2015; 166:12-27. [PMID: 25655839 PMCID: PMC4458388 DOI: 10.1016/j.trsl.2015.01.004] [Citation(s) in RCA: 161] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 01/03/2015] [Accepted: 01/07/2015] [Indexed: 12/14/2022]
Abstract
There is increasing interest in nonrodent translational models for the study of human disease. The pig, in particular, serves as a useful animal model for the study of pathophysiological conditions relevant to the human intestine. This review assesses currently used porcine models of gastrointestinal physiology and disease and provides a rationale for the use of these models for future translational studies. The pig has proven its utility for the study of fundamental disease conditions such as ischemia-reperfusion injury, stress-induced intestinal dysfunction, and short bowel syndrome. Pigs have also shown great promise for the study of intestinal barrier function, surgical tissue manipulation and intervention, as well as biomaterial implantation and tissue transplantation. Advantages of pig models highlighted by these studies include the physiological similarity to human intestine and mechanisms of human disease. Emerging future directions for porcine models of human disease include the fields of transgenics and stem cell biology, with exciting implications for regenerative medicine.
Collapse
|
30
|
Gonzalez LM, Moeser AJ, Blikslager AT. Animal models of ischemia-reperfusion-induced intestinal injury: progress and promise for translational research. Am J Physiol Gastrointest Liver Physiol 2015; 308:G63-75. [PMID: 25414098 PMCID: PMC4297854 DOI: 10.1152/ajpgi.00112.2013] [Citation(s) in RCA: 177] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Research in the field of ischemia-reperfusion injury continues to be plagued by the inability to translate research findings to clinically useful therapies. This may in part relate to the complexity of disease processes that result in intestinal ischemia but may also result from inappropriate research model selection. Research animal models have been integral to the study of ischemia-reperfusion-induced intestinal injury. However, the clinical conditions that compromise intestinal blood flow in clinical patients ranges widely from primary intestinal disease to processes secondary to distant organ failure and generalized systemic disease. Thus models that closely resemble human pathology in clinical conditions as disparate as volvulus, shock, and necrotizing enterocolitis are likely to give the greatest opportunity to understand mechanisms of ischemia that may ultimately translate to patient care. Furthermore, conditions that result in varying levels of ischemia may be further complicated by the reperfusion of blood to tissues that, in some cases, further exacerbates injury. This review assesses animal models of ischemia-reperfusion injury as well as the knowledge that has been derived from each to aid selection of appropriate research models. In addition, a discussion of the future of intestinal ischemia-reperfusion research is provided to place some context on the areas likely to provide the greatest benefit from continued research of ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Liara M. Gonzalez
- 1Department of Clinical Sciences, Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, North Carolina; and
| | - Adam J. Moeser
- 2Department of Population Health and Pathobiology, Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, North Carolina
| | - Anthony T. Blikslager
- 1Department of Clinical Sciences, Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, North Carolina; and
| |
Collapse
|
31
|
Farias Rolim M, Riger CJ, Eleutherio EC, da Fonseca Colão C, Cotta Pereira G, Schanaider A. Colonic healing after portal ischemia and reperfusion: an experimental study with oxidative stress biomarkers. Redox Rep 2013; 12:267-74. [DOI: 10.1179/135100007x239261] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
32
|
Gonzalez LM, Williamson I, Piedrahita JA, Blikslager AT, Magness ST. Cell lineage identification and stem cell culture in a porcine model for the study of intestinal epithelial regeneration. PLoS One 2013; 8:e66465. [PMID: 23840480 PMCID: PMC3696067 DOI: 10.1371/journal.pone.0066465] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2013] [Accepted: 05/05/2013] [Indexed: 01/22/2023] Open
Abstract
Significant advances in intestinal stem cell biology have been made in murine models; however, anatomical and physiological differences between mice and humans limit mice as a translational model for stem cell based research. The pig has been an effective translational model, and represents a candidate species to study intestinal epithelial stem cell (IESC) driven regeneration. The lack of validated reagents and epithelial culture methods is an obstacle to investigating IESC driven regeneration in a pig model. In this study, antibodies against Epithelial Adhesion Molecule 1 (EpCAM) and Villin marked cells of epithelial origin. Antibodies against Proliferative Cell Nuclear Antigen (PCNA), Minichromosome Maintenance Complex 2 (MCM2), Bromodeoxyuridine (BrdU) and phosphorylated Histone H3 (pH3) distinguished proliferating cells at various stages of the cell cycle. SOX9, localized to the stem/progenitor cells zone, while HOPX was restricted to the +4/‘reserve’ stem cell zone. Immunostaining also identified major differentiated lineages. Goblet cells were identified by Mucin 2 (MUC2); enteroendocrine cells by Chromogranin A (CGA), Gastrin and Somatostatin; and absorptive enterocytes by carbonic anhydrase II (CAII) and sucrase isomaltase (SIM). Transmission electron microscopy demonstrated morphologic and sub-cellular characteristics of stem cell and differentiated intestinal epithelial cell types. Quantitative PCR gene expression analysis enabled identification of stem/progenitor cells, post mitotic cell lineages, and important growth and differentiation pathways. Additionally, a method for long-term culture of porcine crypts was developed. Biomarker characterization and development of IESC culture in the porcine model represents a foundation for translational studies of IESC-driven regeneration of the intestinal epithelium in physiology and disease.
Collapse
Affiliation(s)
- Liara M. Gonzalez
- Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, North Carolina, United States of America
- Department of Clinical Sciences, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Ian Williamson
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
- UNC/NCSU Biomedical Engineering, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Jorge A. Piedrahita
- Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, North Carolina, United States of America
- Molecular Biomedical Sciences, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Anthony T. Blikslager
- Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, North Carolina, United States of America
- Department of Clinical Sciences, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Scott T. Magness
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Cell Biology & Physiology, University of North Carolina, Chapel Hill, North Carolina, United States of America
- UNC/NCSU Biomedical Engineering, University of North Carolina, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
33
|
|
34
|
Jamel MJ, Pereira LDPM, Mello NB, Eleuthério ECA, Schanaider A. Blood carbonyl protein measurement as a specific oxidative stress biomarker after intestinal reperfusion in rats. Acta Cir Bras 2010; 25:59-62. [DOI: 10.1590/s0102-86502010000100014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2009] [Accepted: 11/10/2009] [Indexed: 11/22/2022] Open
Abstract
PURPOSE: An experimental study was performed to investigate the use of protein carbonyl group as a specific biological marker for oxidative stress in a rat model of intestinal ischaemia-reperfusion. METHODS: Twenty four male Wistar rats were randomly distributed into three groups with eight animals each: Group 1 - Control group; Group 2 - Sham; Group 3 - Intestinal ischaemia by clamping ileal branches of the superior mesenteric artery for one hour, followed by another hour of reperfusion. Blood samples were taken in order to analyze the protein carbonyl level by Slot blotting assay. RESULTS: In group 3 a significant increase of protein carbonyl level was observed if compared to the homogenous levels of groups 1 and 2. CONCLUSION: From the results it may be concluded that the protein carbonylation may be used as a specific marker for measuring oxidative stress in rat intestinal reperfusion model.
Collapse
|
35
|
Tschetter JR, Blikslager AT, Little D, Howard RD, Woody SL, Beex LM, Crisman MV. Detection of differentially regulated genes in ischaemic equine intestinal mucosa. Equine Vet J 2010; 37:319-24. [PMID: 16028620 DOI: 10.2746/0425164054529382] [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: 11/19/2022]
Abstract
REASONS FOR PERFORMING STUDY Colic is a serious disease syndrome in horses. Much of the mortality is associated with ischaemic-injured intestine during strangulating obstruction, yet there is limited understanding of the associated molecular events. Identification of differentially expressed genes during ischaemic injury should expand our understanding of colic and may lead to novel targeted therapeutic approaches in the future. OBJECTIVE To isolate and identify differentially expressed genes in equine jejunum following a 2 h ischaemic event compared to normally perfused jejunum. METHODS Suppressive subtractive hybridisation was used to clone genes that are differentially expressed in equine jejunum injured by 2 h of complete ischaemia as compared to time-matched control jejunal tissues. Expression of selected clones was further evaluated by northern blot analysis. RESULTS Of the 384 clones selected, 157 were confirmed to possess cDNAs corresponding differentially expressed genes by dot blot analysis. Two genes, fatty acid binding protein 2 and calcium-activated chloride channel 4 were further confirmed to be differentially expressed by northern blot analysis. CONCLUSIONS Suppressive subtractive hybridisation can be used to detect changes in expression of a broad array of genes, as confirmed by northern blot analysis of selected genes. POTENTIAL RELEVANCE These initial results have identified a pool of equine intestinal epithelial genes that are differentially expressed following a 2 h ischaemic event. In particular, genes indicative of deranged metabolic activity and those potentially involved in early repair events were identified and may ultimately provide clues as to the nature of epithelial ischaemic injury in horses.
Collapse
Affiliation(s)
- J R Tschetter
- Department of Large Animal Clinical Sciences, Virginia Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Ding W, Wu X, Gong G, Meng Q, Ni L, Li J. Establishment of an acute superior mesenteric artery injury model for damage control surgery. J Surg Res 2009; 152:249-257. [PMID: 18675994 DOI: 10.1016/j.jss.2008.02.058] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2007] [Revised: 02/21/2008] [Accepted: 02/26/2008] [Indexed: 10/22/2022]
Abstract
BACKGROUND Managements of superior mesenteric artery (SMA) injuries are difficult and often result in a disappointing outcome. Damage control surgery (DCS) has been approved to be an effective and reliable strategy for severe trauma victims. We aimed to build up a severe trauma-shock-hypothermia model of SMA injuries for DCS study and determine the optimal time to institute DCS. METHODS Pigs were anesthetized and instrumented with arterial and a thermodilution cardiac output catheter. SMA flow was interrupted while animals were hemorrhaged to 45% estimated blood volume. Pigs were maintained shock and intestine ischemia for three durations: intestine ischemia for 30 min (I-30; n = 6), 60 min (I-60; n = 6), and 90 min (I-90; n = 6). Cold lactated Ringer's (10 mL/kg) was infused to induce hypothermia. SMA was then declamped and kept in reperfusion for 6 h. Hemodynamic data and serum samples were collected during shock and resuscitation. Distal ileum was collected at the end of ischemia and reperfusion. RESULTS All animals presented with disastrous conditions at the end of ischemia: low temperature, severe acidosis, decreased blood pressure, depressed cardiac output, and oxygen delivery. I-90 animals suffered the lowest temperature, the most severe acidosis, lowest blood pressure, and depressed cardiac output and oxygen delivery. Coagulopathy developed in I-90, whereas normal prothrombin time and thrombin time were detected in I-30 and I-60. Aspartate aminotransferase, lactate dehydrogenase, creatine kinase, and alkaline phosphatase were equally within groups (P > 0.05). All (6/6) of I-30, 83.3% (5/6) of I-60, and 16.7% (1/6) of I-90 pigs survived (P < 0.01). Base excess in I-90 was much lower than that in I-30 and I-60 animals. CONCLUSIONS We first built up an acute SMA injury animal model for DCS investigations and determined that the optimal institution time of DCS was before 60 min after SMA injury in the trauma-shock-hypothermia swine model.
Collapse
Affiliation(s)
- Weiwei Ding
- Research Institute of General Surgery, School of Medicine, Nanjing University, Jinling Hospital, Jiangsu Province, Nanjing, China
| | | | | | | | | | | |
Collapse
|
37
|
Liu F, Yin J, Du M, Yan P, Xu J, Zhu X, Yu J. Heat-stress-induced damage to porcine small intestinal epithelium associated with downregulation of epithelial growth factor signaling. J Anim Sci 2009; 87:1941-9. [PMID: 19213704 DOI: 10.2527/jas.2008-1624] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Extreme heat during certain days of the summer renders pigs susceptible to severe heat stress, which negatively affects their growth performance. We hypothesized that such heat stress impaired the small intestinal mucosa, a site responsible for nutrient absorption. To simulate heat stress, Chinese experimental mini-pigs were treated with 5 h of continual 40 degrees C temperature each day for 10 d in succession. Pigs were killed at 1, 3, 6 and 10 d after treatment, and small intestinal epithelia were sampled for histochemical examination and biochemical analyses. The duodenum and jejunum were seriously damaged within 3 d of initiation of treatment. Subsequent study of the process of jejunum recovery showed that the initiation of recovery started within 6 d following heat stress. Such damage was associated with the downregulation of epithelial growth factor signaling. In conclusion, heat stress induced short-term damage to the epithelium of porcine intestine. Because the intestinal epithelium is crucial for nutrient uptake, such damage should partially account for the impairment of growth performance of pigs under heat stress.
Collapse
Affiliation(s)
- F Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China
| | | | | | | | | | | | | |
Collapse
|
38
|
Annecke T, Kubitz JC, Langer K, Hilberath JM, Kahr S, Krombach F, Bittmann I, Rehm M, Kemming GI, Conzen PF. Lung injury following thoracic aortic occlusion: comparison of sevoflurane and propofol anaesthesia. Acta Anaesthesiol Scand 2008; 52:977-86. [PMID: 18477071 DOI: 10.1111/j.1399-6576.2008.01665.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND Halogenated anaesthetics have been shown to reduce ischaemia-reperfusion injuries in various organs due to pre- and post-conditioning mechanisms. We compared volatile and total intravenous anaesthesia with regard to their effect on remote pulmonary injury after thoracic aortic occlusion and reperfusion. METHODS Eighteen pigs were randomized after sternotomy and laparotomy (fentanyl-midazolam anaesthesia) to receive either sevoflurane or propofol in an investigator-blinded fashion. Ninety minutes of thoracic aortic occlusion was induced by a balloon catheter. During reperfusion, a goal-directed resuscitation protocol was performed. After 120 min of reperfusion, the anaesthetic regimen was changed to fentanyl-midazolam again for another 180 min. The oxygenation index and intra-pulmonary shunt fractions were calculated. After 5 h of reperfusion, a bronchoalveolar lavage was performed. The total protein content and lactate dehydrogenase activity were measured in epithelial lining fluid (ELF). Alveolar macrophage oxidative burst was analysed. The wet to dry ratio was calculated and tissue injury was graded using a semi-quantitative score. Ten animals (n=5 for each anaesthetic) without aortic occlusion served as time controls. RESULTS The oxygenation index decreased and the intra-pulmonary shunt fraction increased significantly in both occlusion groups. There were no significant differences between sevoflurane and propofol with respect to the oxygenation index, ELF composition, morphologic lung damage, wet to dry ratio and alveolar macrophage burst activity. Differences were, however, seen in terms of systemic haemodynamic stability, where catecholamine requirements were less pronounced with sevoflurane. CONCLUSION We conclude that the severity of remote lung injury was not different between sevoflurane and propofol anaesthesia in this porcine model of severe lower-body ischaemia and reperfusion injury.
Collapse
Affiliation(s)
- T Annecke
- Department of Anaesthesiology, Ludwig-Maximilians-University of Munich, Munich, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Solligård E, Juel IS, Spigset O, Romundstad P, Grønbech JE, Aadahl P. Gut luminal lactate measured by microdialysis mirrors permeability of the intestinal mucosa after ischemia. Shock 2008; 29:245-51. [PMID: 17693938 DOI: 10.1097/shk.0b013e3180cab3ce] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The aim of the present study was to investigate the influence of a prolonged initial intestinal ischemic insult on transmucosal permeability after a subsequent ischemic event and whether microdialysis of biomarkers released to the gut lumen is able to reflect changes in intestinal permeability. The superior mesenteric artery was cross-clamped for 60 min followed by 4 h of reperfusion in 16 pigs. Nine pigs had a second cross-clamp of 60 min and 3 h of reperfusion, whereas seven pigs were observed for a further 4 h of reperfusion. Intestinal mucosal integrity was assessed by permeability of C-polyethylene glycol (PEG-4000) over the gut mucosa, luminal microdialysis of lactate, glucose and glycerol, and tonometry. During reperfusion, the PEG-4000 amount in venous blood was two times higher after the first than after the second ischemia (area under the curve, 44,780 [13,441-82,723] vs. 22,298 (12,213-49,698] counts min mL(-1), P=0.026 [mean {range}]). There was less lactate detected in the gut lumen after the second ischemia compared with the first (area under the curve, 797 [412-1,700] vs. 1,151 [880-1,969] mmol min L(-1), P=0.02) and a lower maximum concentration (4.8 [2.7-9.4] vs. 8.5 [5.0-14.9] mM, P=0.01). The same pattern was also seen for luminal glycerol and glucose. During the second ischemia, the intestinal mucosal/arterial CO2 gap was identical to the level during the first ischemic episode. A prolonged ischemic insult of the intestine confers protection, for reduced hyperpermeability against further ischemia. Microdialysis of biomarkers mirrors permeability changes associated with this type of protection. Lactate reflects permeability across the intestinal mucosa more precisely than glycerol.
Collapse
Affiliation(s)
- Erik Solligård
- Department of Anesthesiology and Intensive Care, St. Olav University Hospital, Trondheim, Norway.
| | | | | | | | | | | |
Collapse
|
40
|
EVALUATION OF INTESTINAL PRECONDITIONING IN A PORCINE MODEL USING CLASSIC ISCHEMIC PRECONDITIONING OR LUNG RECRUITMENT MANEUVERS. Shock 2008. [DOI: 10.1097/shk.0b013e318067d8b7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
41
|
Claesson J, Lehtipalo S, Johansson G, Abrahamsson P, Palmqvist R, Biber B, Winsö O. EVALUATION OF INTESTINAL PRECONDITIONING IN A PORCINE MODEL USING CLASSIC ISCHEMIC PRECONDITIONING OR LUNG RECRUITMENT MANEUVERS. Shock 2008; 29:98-103. [DOI: 10.1097/shk.0b013e3318057da56] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
42
|
Juel IS, Solligård E, Tvedt KE, Skogvoll E, Jynge P, Beisvag V, Erlandsen SE, Sandvik AK, Aadahl P, Grønbech JE. Post-ischaemic restituted intestinal mucosa is more resistant to further ischaemia than normal mucosa in the pig. Scandinavian Journal of Clinical and Laboratory Investigation 2007; 68:106-16. [PMID: 17852833 DOI: 10.1080/00365510701534833] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
OBJECTIVE Ischaemic preconditioning may protect the intestine from subsequent prolonged ischaemia. This study evaluates whether a much longer initial ischaemia, encountered clinically, may modify intestinal resistance to further ischaemia in a pig model. MATERIAL AND METHODS After cross-clamping of the superior mesenteric artery for 1 h, the intestine was either reperfused for 8 h or a second cross-clamping for 1 h was performed at 4 h of reperfusion. Based on microarray analysis of intestinal samples at 1, 4 and 8 h of reperfusion, mRNA of selected genes was measured with QRT-PCR. RESULTS The first ischaemic period caused exfoliation of surface epithelial cells from the basement membrane comprising about 90 % of the villi tips, a marked increase in permeability and depletion of ATP. The second ischaemic challenge caused about 30 % less denudation of the basement membrane (p = 0.008), no increase in permeability (p = 0.008) and less depletion of ATP (p = 0.039). mRNAs for superoxide dismutase 2, heat shock proteins and signal transducer and activator of transcription 3, which may protect against ischaemia/reperfusion injury, were up-regulated throughout the reperfusion period. mRNAs for matrix metalloproteinase 1, connexin 43 and peripheral myelin 22, which may be associated with cell migration or tight junctions, showed a particular up-regulation at 4 h of reperfusion. CONCLUSION One hour of initial ischaemia followed by 4 h of reperfusion is associated with increased intestinal resistance to further ischaemia. The differential regulation of genes identified in this study provides working hypotheses for mechanisms behind this observation.
Collapse
Affiliation(s)
- Ingebjørg S Juel
- Department of Surgery, St. Olav University Hospital, NO-7006 Trondheim, Norway.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Blikslager AT, Moeser AJ, Gookin JL, Jones SL, Odle J. Restoration of barrier function in injured intestinal mucosa. Physiol Rev 2007; 87:545-64. [PMID: 17429041 DOI: 10.1152/physrev.00012.2006] [Citation(s) in RCA: 409] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mucosal repair is a complex event that immediately follows acute injury induced by ischemia and noxious luminal contents such as bile. In the small intestine, villous contraction is the initial phase of repair and is initiated by myofibroblasts that reside immediately beneath the epithelial basement membrane. Subsequent events include crawling of healthy epithelium adjacent to the wound, referred to as restitution. This is a highly regulated event involving signaling via basement membrane integrins by molecules such as focal adhesion kinase and growth factors. Interestingly, however, ex vivo studies of mammalian small intestine have revealed the importance of closure of the interepithelial tight junctions and the paracellular space. The critical role of tight junction closure is underscored by the prominent contribution of the paracellular space to measures of barrier function such as transepithelial electrical resistance. Additional roles are played by subepithelial cell populations, including neutrophils, related to their role in innate immunity. The net result of reparative mechanisms is remarkably rapid closure of mucosal wounds in mammalian tissues to prevent the onset of sepsis.
Collapse
Affiliation(s)
- Anthony T Blikslager
- Department of Clinical Science, North Carolina State University, Raleigh 27606, USA.
| | | | | | | | | |
Collapse
|
44
|
Annecke T, Kubitz JC, Kahr S, Hilberath JM, Langer K, Kemming GI, Rehm M, Bittmann I, Conzen PF. Effects of sevoflurane and propofol on ischaemia–reperfusion injury after thoracic-aortic occlusion in pigs. Br J Anaesth 2007; 98:581-90. [PMID: 17371775 DOI: 10.1093/bja/aem049] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Thoraco-abdominal-aneurysm surgery predicts high mortality. Propofol and sevoflurane are commonly used anaesthetics for this procedure. Halogenated anaesthetics induce organ protection similar to ischaemic preconditioning. We investigated which anaesthetic regimen would lead to a better protection against ischaemia-reperfusion injury induced by temporary thoracic-aortic occlusion. METHODS Following initial fentanyl-midazolam anaesthesia for surgical preparation, 18 pigs were randomly assigned to two groups: group one received propofol (n=9) and group two sevoflurane (n=9) before, during, and after lower body ischaemia in an investigator blinded fashion. Ten animals without aortic occlusion served as time controls (propofol, n=5; sevoflurane, n=5). For induction of ischaemia, the thoracic aorta was occluded by a balloon-catheter for 90 min. After 120 min of reperfusion, the study anaesthetics were discontinued and fentanyl-midazolam re-established for an additional 180 min. Goal-directed therapy was performed during reperfusion. Fluid and catecholamine requirements were assessed. Serum samples and intestinal tissue specimens were obtained. RESULTS Severe declamping shock occurred in both study groups. While norepinephrine requirements in the sevoflurane group were significantly reduced during reperfusion (P<0.05), allowing cessation of catecholamine support in 4/9 animals, all 9/9 animals were still catecholamine dependent at the end of the experiment in the propofol group. Serum activities of lactate dehydrogenase, aspartate transaminase, and alanine aminotransferase were lower with sevoflurane (P<0.05). Small intestine tissue specimens did not differ histologically. CONCLUSIONS Use of sevoflurane compared with propofol attenuated the haemodynamic sequelae of reperfusion injury in our model. Release of serum markers of cellular injury was also attenuated.
Collapse
Affiliation(s)
- T Annecke
- Clinic of Anaesthesiology, Institute for Surgical Research, Ludwig-Maximilians-University, University Hospital, Munich, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Solligård E, Juel IS, Bakkelund K, Jynge P, Tvedt KE, Johnsen H, Aadahl P, Grønbech JE. Gut luminal microdialysis of glycerol as a marker of intestinal ischemic injury and recovery. Crit Care Med 2005; 33:2278-85. [PMID: 16215382 DOI: 10.1097/01.ccm.0000178187.84732.6c] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To evaluate microdialysis as a method to assess different degrees of intestinal damage and recovery during ischemia and reperfusion; to evaluate information obtained from microdialysis catheters in the peritoneum, the gut wall, and the gut lumen. DESIGN Randomized, controlled animal experiment. SETTING University laboratory animal center. SUBJECTS Twenty-seven domestic pigs. INTERVENTIONS The superior mesenteric artery was cross-clamped for 60 mins (n = 14) or 120 mins (n = 10) followed by 2 or 4 hrs of reperfusion. Three pigs served as controls. MEASUREMENTS AND MAIN RESULTS Intestinal mucosal integrity was assessed by morphometry, adenosine triphosphate in the gut wall, and permeability of C-polyethylene glycol. Lactate, glycerol, pyruvate, and glucose were measured by microdialysis. Changes in adenosine triphosphate, permeability, or lactate did not correlate to different extents of intestinal damage caused by 60 or 120 mins of ischemia. During the reperfusion period, pigs with 60 mins of intestinal ischemia showed a faster recovery of these variables than pigs with 120 mins of intestinal ischemia. Glycerol increased with increasing duration of the ischemic insult. After 60 mins of intestinal ischemia, glycerol in the gut lumen decreased toward baseline but remained high after 120 mins of intestinal ischemia. There was a good correlation between gut luminal glycerol and recovery of mucosal damage throughout the reperfusion period. In the peritoneal cavity, both glycerol and lactate decreased to baseline relatively shortly after onset of reperfusion independent of the duration of intestinal ischemia. CONCLUSIONS Microdialysis of glycerol provides information about the extent and severity of intestinal damage after ischemia and about the ensuing recovery. The gut lumen is to be preferred as a site for placement of microdialysis catheters.
Collapse
Affiliation(s)
- Erik Solligård
- Department of Anesthesiology and Intensive Care, St. Olav University Hospital, Norwegian University of Science and Technology, N-7006 Trondheim, Norway.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Douzinas EE, Kollias S, Tiniakos D, Evangelou E, Papalois A, Rapidis AD, Tsoukalas GD, Patsouris E, Roussos C. Hypoxemic reperfusion after 120 mins of intestinal ischemia attenuates the histopathologic and inflammatory response *. Crit Care Med 2004; 32:2279-83. [PMID: 15640642 DOI: 10.1097/01.ccm.0000145582.03345.c8] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE It has been suggested that reactive oxygen species play a pivotal role in the initial organ-tissue injury during reperfusion, eliciting inflammatory reaction and multiple organ failure. It was investigated if hypoxemic reperfusion attenuates tissue injury and inflammatory response. DESIGN Randomized animal study. SETTING Medical school laboratory. SUBJECTS Twenty-five male pigs weighing 25-28 kg. INTERVENTIONS Pigs were subjected to 120 mins of intestinal ischemia by clamping the superior mesenteric artery. Upon declamping, the animals were randomly assigned to receive either hypoxemic reperfusion (HR group, n = 9) reperfused with a Pao2 = 30-35 or normoxemic reperfusion (control group, n = 16) reperfused with a Pao2 = 100 mm Hg for 120 mins. Fluids without inotropes were given to combat circulatory shock during reperfusion. MEASUREMENTS AND MAIN RESULTS Portal blood and intestinal and lung biopsies were collected at baseline, end of ischemia, and end of reperfusion. Histopathologic changes were scored, and interleukin-1beta, qualitative Limulus amebocyte, lysate test, and Pao2/Fio2 were measured. Eight of 16 animals of the control group and seven of nine of the HR group survived (p = .22). At the end of reperfusion, the intestinal (p = .004) and lung (p = .028) pathologic scores were lower in the HR group compared with controls. The only significant difference in concentration of interleukin-1beta in the portal blood between the two animal groups occurred 120 mins after reperfusion (p = .006). The number of HR animals with a positive Limulus test was significantly smaller compared with controls at 60 (p = .041) and 120 (p = .07) mins of reperfusion. During the period of ischemia, the Pao2/Fio2 decreased similarly in the control and HR group, whereas after 120 mins of reperfusion the rate was significantly higher in the HR group. CONCLUSIONS Hypoxemic reperfusion represents an intervention that may attenuate the triggering of multifactorial cascade and organ tissue injury.
Collapse
Affiliation(s)
- Emmanuel E Douzinas
- Department of Critical Care, University of Athens Medical School, Athens, Greece
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Niewold TA, Meinen M, van der Meulen J. Plasma intestinal fatty acid binding protein (I-FABP) concentrations increase following intestinal ischemia in pigs. Res Vet Sci 2004; 77:89-91. [PMID: 15120958 DOI: 10.1016/j.rvsc.2004.02.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2004] [Indexed: 12/28/2022]
Abstract
Intestinal fatty acid binding protein (I-FABP) is an intracellular epithelial protein in the intestinal mucosa of many animals. I-FABP appears in the circulation following epithelial damage, and in humans, is proven to be a parameter for damage to the mucosa. In this paper, an ELISA test designed for human I-FABP analysis was used to assay pig blood samples. The test recognized I-FABP cloned from pig small intestine and expressed in Escherichia coli. Furthermore, in our experimental model of (low flow) intestinal ischemia and reperfusion a significant rise in plasma I-FABP concentrations 15-30 min after clamping of the mesenteric artery was demonstrated. This is the first report that in pigs circulating I-FABP is a useful marker for (mild) intestinal injury, and could possibly be used to monitor (intestinal) health in clinical practice.
Collapse
Affiliation(s)
- T A Niewold
- Animal Sciences Group, Division of Animal Resources Development, Research Group Animal Physiology and Health, P.O. Box 65, 8200 AB Lelystad, The Netherlands.
| | | | | |
Collapse
|
48
|
Juel IS, Solligård E, Lyng O, Strømholm T, Tvedt KE, Johnsen H, Jynge P, Saether OD, Aadahl P, Grønbech JE. Intestinal injury after thoracic aortic cross-clamping in the pig. J Surg Res 2004; 117:283-95. [PMID: 15047134 DOI: 10.1016/j.jss.2003.10.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2003] [Indexed: 10/26/2022]
Abstract
BACKGROUND The mucosal surface epithelium is an essential part of the functional intestinal barrier, but its structural response to ischemia/reperfusion is only partly characterized. The purpose of this study was to provide a detailed morphological evaluation of intestinal surface epithelium after aortic cross-clamping. MATERIAL AND METHODS Pigs were subjected to thoracic aortic cross-clamping for 60 min and subsequent reperfusion for 120 min. Tissue blood flow and high-energy phosphates were measured with microspheres and HPLC, respectively. Urinary excretion of (14)C polyethylene glycol (MW 4000 Da) (PEG-4000), loaded into an intestinal loop, provided an index of intestinal permeability. RESULTS Jejunal blood flow was restored at 10 min after aortic declamping. Denudation of the basement membrane of the intestinal villi tips, as a consequence of epithelial shedding, increased markedly during the initial 60 min of reperfusion (P = 0.002). During the following 45 min, the denuded basement membrane was partly covered with low cuboidal and squamous-shaped cells extending lamellipodia over a wavy basement membrane. Restoration of ATP at 60 min after aortic declamping correlated inversely to the extent of denuded basement membrane (r = 0.75, P = 0.032). Permeability of PEG-4000 increased markedly after aortic declamping and was linearly correlated to the area of denuded basement membrane (r = 0.87, P = 0.01). CONCLUSIONS Reperfusion for 2 h after aortic cross-clamping is associated with initial aggravation of ischemia-induced injury in the porcine jejunum, but thereafter with restitution of the surface epithelium. Restoration of ATP may be important to avoid intestinal injury after ischemia. Increased permeability of a macromolecule in response to reperfusion is closely correlated to injury of the surface epithelium.
Collapse
Affiliation(s)
- Ingebjørg S Juel
- Department of Surgery, St. Olav University Hospital, N-7006 Trondheim, Norway.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Ischemic injury is one of the most important causes of mortality in equine veterinary medicine. Although treatment of reperfusion injury has been attempted in a number of experimental trials to reduce the level of injury subsequent to an ischemic episode, this research has not resulted in the development of useful clinical treatments. Nevertheless, recent studies assessing intraluminal application of solutions containing antioxidants, nutrients, and vasodilators are promising. Furthermore, focusing on improving mucosal recovery after an ischemic event may provide an alternative method of reducing mortality. Potential treatments include administration of basement membrane components like hyaluronic acid, gut-specific nutrients like glutamine, and early return to feeding to stimulate endogenous repair mechanisms. Finally, recent studies evaluating NSAIDs have revealed the potential of flunixin meglumine to retard the mucosal repair process, indicating the need for judicious use of this drug.
Collapse
Affiliation(s)
- Anthony T Blikslager
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh 27606, USA.
| |
Collapse
|
50
|
|