@article{ludwig_abraham_mckinney_freund_stewart_garman_barbas_sudan_gonzalez_2023, title={45: Comparison of the Effects of Normothermic Machine Perfusion and Cold Storage Preservation on Porcine Intestinal Allograft Regenerative Potential and Viability}, volume={107}, ISSN={0041-1337}, url={http://dx.doi.org/10.1097/01.tp.0000945636.34372.db}, DOI={10.1097/01.tp.0000945636.34372.db}, abstractNote={Historically, intestinal transplantation (IT) has been reserved as the last treatment option for patients with irreversible intestinal failure who are unable to tolerate total parenteral nutrition. Successful IT is reliant upon graft health at the time of donation, minimizing graft injury that may occur during procurement, storage, and IT, and the ability of the graft to heal following insult. Unfortunately, the intestine is easily damaged by ischemia-reperfusion injury (IRI). IRI induces intestinal epithelial cell apoptosis and damages the mucosal barrier, which can result in bacterial translocation and activation of the local and systemic immune and inflammatory response, ultimately contributing to graft failure, rejection, and decreased recipient survival. The current, preferred method of intestinal preservation prior to IT is static cold storage (CS), however the prolonged hypothermic ischemia of CS causes cell injury and intensifies the IRI that occurs during transplantation. Furthermore, IRI to the epithelial crypt region diminishes the intestine’s ability to heal by inducing loss of the highly proliferative intestinal stem cells (ISCs) that are responsible for maintenance, regeneration, and repair of the epithelium, critical to graft health. Thus, the investigation of alternative organ preservation techniques that reduce IRI, cellular damage, and graft injury are warranted to overall improve IT success. Normothermic machine perfusion (NMP) is a preservation method that reduces inflammation and promotes graft regeneration in other organs by preventing CS-associated IRI. However, NMP has not been described for intestine. We hypothesized that, compared to CS, intestinal NMP will induce less epithelial injury and better protect ISC regenerative potential and viability. 15 porcine intestines were flushed with UW solution, stored at 4°C (CS), or perfused with 34°C perfusate (NMP) for 6hr, and transplanted (n=9). Recipient pigs were recovered from anesthesia. Jejunal and ileal segments were collected immediately after flushing, serving as control tissue (CO), after 6hr of CS or NMP, and after 1hr of reperfusion post-IT. Histologic injury was assessed. Crypts isolated after flushing (CO), 6hr CS or NMP, and 1hr of reperfusion post-IT were cultured. Spheroid number, size, and EdU staining quantified ISC viability and proliferation. Expression of ISC and cellular proliferation genes and proteins were measured. Histologically, NMP tissue had mild epithelial erosion and increased columnar cell attenuation and expression of ISC and proliferation genes/proteins was observed. NMP spheroid areas and proliferating cell numbers were significantly larger than control and CS. Apoptotic cells were increased following CS. Post-graft reperfusion, CS had increased injury compared to uninjured control and NMP tissue. Compared to CS, NMP may improve graft regenerative potential, resulting in transplantation of healthier bowel and superior recipient survival.}, number={7S}, journal={Transplantation}, publisher={Ovid Technologies (Wolters Kluwer Health)}, author={Ludwig, Elsa and Abraham, Nader and McKinney, Caroline and Freund, John and Stewart, Amy and Garman, Katherine and Barbas, Andrew and Sudan, Debra and Gonzalez, Liara}, year={2023}, month={Jun}, pages={25–25} } @article{schaaf_polkoff_carter_stewart_sheahan_freund_ginzel_snyder_roper_piedrahita_et al._2023, title={A LGR5 reporter pig model closely resembles human intestine for improved study of stem cells in disease}, volume={37}, ISSN={["1530-6860"]}, DOI={10.1096/fj.202300223R}, abstractNote={Abstract}, number={6}, journal={FASEB JOURNAL}, author={Schaaf, Cecilia R. and Polkoff, Kathryn M. and Carter, Amber and Stewart, Amy S. and Sheahan, Breanna and Freund, John and Ginzel, Joshua and Snyder, Joshua C. and Roper, Jatin and Piedrahita, Jorge A. and et al.}, year={2023}, month={Jun} } @article{ludwig_hobbs_mckinney-aguirre_gonzalez_2023, title={Biomarkers of Intestinal Injury in Colic}, volume={13}, ISSN={2076-2615}, url={http://dx.doi.org/10.3390/ani13020227}, DOI={10.3390/ani13020227}, abstractNote={Biomarkers are typically proteins, enzymes, or other molecular changes that are elevated or decreased in body fluids during the course of inflammation or disease. Biomarkers pose an extremely attractive tool for establishing diagnoses and prognoses of equine gastrointestinal colic, one of the most prevalent causes of morbidity and mortality in horses. This topic has received increasing attention because early diagnosis of some forms of severe colic, such as intestinal ischemia, would create opportunities for rapid interventions that would likely improve case outcomes. This review explores biomarkers currently used in equine medicine for colic, including acute phase proteins, proinflammatory cytokines, markers of endotoxemia, and tissue injury metabolites. To date, no single biomarker has been identified that is perfectly sensitive and specific for intestinal ischemia; however, L-lactate has been proven to be a very functional and highly utilized diagnostic tool. However, further exploration of other biomarkers discussed in this review may provide the key to accelerated identification, intervention, and better outcomes for horses suffering from severe colic.}, number={2}, journal={Animals}, publisher={MDPI AG}, author={Ludwig, Elsa K. and Hobbs, Kallie J. and McKinney-Aguirre, Caroline A. and Gonzalez, Liara M.}, year={2023}, month={Jan}, pages={227} } @article{veerasammy_gonzalez_báez‐ramos_schaaf_stewart_ludwig_mckinney‐aguirre_freund_robertson_gonzalez_2023, title={Changes in equine intestinal stem/progenitor cell number at resection margins in cases of small intestinal strangulation}, volume={55}, ISSN={0425-1644 2042-3306}, url={http://dx.doi.org/10.1111/evj.13927}, DOI={10.1111/evj.13927}, abstractNote={Abstract}, number={6}, journal={Equine Veterinary Journal}, publisher={Wiley}, author={Veerasammy, Brittany and Gonzalez, Gabriel and Báez‐Ramos, Patricia and Schaaf, Cecilia R. and Stewart, Amy Stieler and Ludwig, Elsa K. and McKinney‐Aguirre, Caroline and Freund, John and Robertson, James and Gonzalez, Liara M.}, year={2023}, month={Feb}, pages={995–1002} } @article{gonzalez_stampley_marcellin-little_kedrowicz_2023, title={Respondents to an American College of Veterinary Surgeons diplomate survey support the promotion of diversity, equity, and inclusion initiatives}, volume={261}, ISSN={["1943-569X"]}, DOI={10.2460/javma.23.06.0310}, abstractNote={Abstract}, number={12}, journal={JAVMA-JOURNAL OF THE AMERICAN VETERINARY MEDICAL ASSOCIATION}, author={Gonzalez, Liara M. and Stampley, Anita R. and Marcellin-Little, Denis J. and Kedrowicz, April A.}, year={2023}, month={Dec}, pages={1847–1852} } @article{stewart_schaaf_veerasammy_freund_gonzalez_2022, title={Culture of equine intestinal epithelial stem cells after delayed tissue storage for future applications}, volume={18}, ISSN={["1746-6148"]}, DOI={10.1186/s12917-022-03552-6}, abstractNote={Abstract}, number={1}, journal={BMC VETERINARY RESEARCH}, author={Stewart, Amy Stieler and Schaaf, Cecilia R. and Veerasammy, Brittany and Freund, John M. and Gonzalez, Liara M.}, year={2022}, month={Dec} } @article{tse_meganck_araba_yount_shaffer_hou_munt_adams_wykoff_morowitz_et al._2022, title={Genomewide CRISPR knockout screen identified PLAC8 as an essential factor for SADS-CoVs infection}, volume={119}, ISSN={["1091-6490"]}, DOI={10.1073/pnas.2118126119}, abstractNote={Significance}, number={18}, journal={PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA}, author={Tse, Longping V and Meganck, Rita M. and Araba, Kenza C. and Yount, Boyd L. and Shaffer, Kendall M. and Hou, Yixuan J. and Munt, Jennifer E. and Adams, Lily E. and Wykoff, Jason A. and Morowitz, Jeremy M. and et al.}, year={2022}, month={May} } @article{abraham_ludwig_schaaf_veerasammy_stewart_mckinney_freund_brassil_samy_gao_et al._2022, title={Orthotopic Transplantation of the Full-length Porcine Intestine After Normothermic Machine Perfusion}, volume={8}, ISSN={2373-8731}, url={http://dx.doi.org/10.1097/TXD.0000000000001390}, DOI={10.1097/TXD.0000000000001390}, abstractNote={ Background. Successful intestinal transplantation is currently hindered by graft injury that occurs during procurement and storage, which contributes to postoperative sepsis and allograft rejection. Improved graft preservation may expand transplantable graft numbers and enhance posttransplant outcomes. Superior transplant outcomes have recently been demonstrated in clinical trials using machine perfusion to preserve the liver. We hypothesized that machine perfusion preservation of intestinal allografts could be achieved and allow for transplantation in a porcine model. }, number={11}, journal={Transplantation Direct}, publisher={Ovid Technologies (Wolters Kluwer Health)}, author={Abraham, Nader and Ludwig, Elsa K. and Schaaf, Cecilia R. and Veerasammy, Brittany and Stewart, Amy S. and McKinney, Caroline and Freund, John and Brassil, John and Samy, Kannan P. and Gao, Qimeng and et al.}, year={2022}, month={Oct}, pages={e1390} } @misc{schaaf_gonzalez_2022, title={Use of Translational, Genetically Modified Porcine Models to Ultimately Improve Intestinal Disease Treatment}, volume={9}, ISSN={["2297-1769"]}, DOI={10.3389/fvets.2022.878952}, abstractNote={For both human and veterinary patients, non-infectious intestinal disease is a major cause of morbidity and mortality. To improve treatment of intestinal disease, large animal models are increasingly recognized as critical tools to translate the basic science discoveries made in rodent models into clinical application. Large animal intestinal models, particularly porcine, more closely resemble human anatomy, physiology, and disease pathogenesis; these features make them critical to the pre-clinical study of intestinal disease treatments. Previously, large animal model use has been somewhat precluded by the lack of genetically altered large animals to mechanistically investigate non-infectious intestinal diseases such as colorectal cancer, cystic fibrosis, and ischemia-reperfusion injury. However, recent advances and increased availability of gene editing technologies has led to both novel use of large animal models in clinically relevant intestinal disease research and improved testing of potential therapeutics for these diseases.}, journal={FRONTIERS IN VETERINARY SCIENCE}, author={Schaaf, Cecilia R. and Gonzalez, Liara M.}, year={2022}, month={May} } @article{stewart_schaaf_luff_freund_becker_tufts_robertson_gonzalez_2021, title={HOPX+ injury-resistant intestinal stem cells drive epithelial recovery after severe intestinal ischemia}, volume={321}, ISSN={["1522-1547"]}, url={https://doi.org/10.1152/ajpgi.00165.2021}, DOI={10.1152/ajpgi.00165.2021}, abstractNote={ This paper supports that rISCs are resistant to ischemic injury and likely an important source of cellular renewal following near-complete epithelial loss. Furthermore, we have evidence that HOPX controls ISC activity state and may be a critical signaling pathway during ISC-mediated repair. Finally, we use multiple novel methods to evaluate ISCs in a translationally relevant large animal model of severe intestinal injury and provide evidence for the potential role of rISCs as therapeutic targets. }, number={5}, journal={AMERICAN JOURNAL OF PHYSIOLOGY-GASTROINTESTINAL AND LIVER PHYSIOLOGY}, publisher={American Physiological Society}, author={Stewart, Amy Stieler and Schaaf, Cecilia Renee and Luff, Jennifer A. and Freund, John M. and Becker, Thomas C. and Tufts, Sara R. and Robertson, James B. and Gonzalez, Liara M.}, year={2021}, month={Oct}, pages={G588–G602} } @article{vermeire_gonzalez_jansens_cox_devriendt_2021, title={Porcine small intestinal organoids as a model to explore ETEC-host interactions in the gut}, volume={52}, ISSN={["1297-9716"]}, DOI={10.1186/s13567-021-00961-7}, abstractNote={Abstract}, number={1}, journal={VETERINARY RESEARCH}, author={Vermeire, Bjarne and Gonzalez, Liara M. and Jansens, Robert J. J. and Cox, Eric and Devriendt, Bert}, year={2021}, month={Jun} } @article{vermeire_gonzalez_jansens_cox_devriendt_2021, title={Porcine small intestinal organoids as a model to explore ETEC-host interactions in the gut (vol 52, 94, 2021)}, volume={52}, ISSN={["1297-9716"]}, DOI={10.1186/s13567-021-00977-z}, number={1}, journal={VETERINARY RESEARCH}, author={Vermeire, Bjarne and Gonzalez, Liara M. and Jansens, Robert J. J. and Cox, Eric and Devriendt, Bert}, year={2021}, month={Aug} } @article{singh_hung_shanahan_kanke_bonfini_dame_biraud_peck_oyesola_freund_et al._2020, title={Enteroendocrine Progenitor Cell-Enriched mir-7 Regulates Intestinal Epithelial Proliferation in an Xiap-Dependent Manner}, volume={9}, ISSN={["2352-345X"]}, DOI={10.1016/j.jcmgh.2019.11.001}, abstractNote={The enteroendocrine cell (EEC) lineage is important for intestinal homeostasis. It was recently shown that EEC progenitors contribute to intestinal epithelial growth and renewal, but the underlying mechanisms remain poorly understood. MicroRNAs are under-explored along the entire EEC lineage trajectory, and comparatively little is known about their contributions to intestinal homeostasis.We leverage unbiased sequencing and eight different mouse models and sorting methods to identify microRNAs enriched along the EEC lineage trajectory. We further characterize the functional role of EEC progenitor-enriched miRNA, miR-7, by in vivo dietary study as well as ex vivo enteroid in mice.First, we demonstrate that miR-7 is highly enriched across the entire EEC lineage trajectory and is the most enriched miRNA in EEC progenitors relative to Lgr5+ intestinal stem cells. Next, we show in vivo that in EEC progenitors miR-7 is dramatically suppressed under dietary conditions that favor crypt division and suppress EEC abundance. We then demonstrate by functional assays in mouse enteroids that miR-7 exerts robust control of growth, as determined by budding (proxy for crypt division), EdU and PH3 staining, and likely regulates EEC abundance also. Finally, we show by single-cell RNA sequencing analysis that miR-7 regulates Xiap in progenitor/stem cells and we demonstrate in enteroids that the effects of miR-7 on mouse enteroid growth depend in part on Xiap and Egfr signaling.This study demonstrates for the first time that EEC progenitor cell-enriched miR-7 is altered by dietary perturbations and that it regulates growth in enteroids via intact Xiap and Egfr signaling.}, number={3}, journal={CELLULAR AND MOLECULAR GASTROENTEROLOGY AND HEPATOLOGY}, author={Singh, Ajeet P. and Hung, Yu-Han and Shanahan, Michael T. and Kanke, Matt and Bonfini, Alessandro and Dame, Michael K. and Biraud, Mandy and Peck, Bailey C. E. and Oyesola, Oyebola O. and Freund, John M. and et al.}, year={2020}, pages={447–464} } @article{orr_baker_lynch_hughes_clark_slone_fogle_gonzalez_2020, title={Prognostic value of colonic and peripheral venous lactate measurements in horses with large colon volvulus}, url={https://doi.org/10.1111/vsu.13361}, DOI={10.1111/vsu.13361}, abstractNote={Abstract}, journal={Veterinary Surgery}, author={Orr, Kindra. E. and Baker, W. True and Lynch, Tim M. and Hughes, Faith E. and Clark, Carol K. and Slone, Donnie E., Jr and Fogle, Callie A. and Gonzalez, Liara M.}, year={2020}, month={Apr} } @article{gonzalez_stewart_freund_kucera_dekaney_magness_blikslager_2019, title={Preservation of reserve intestinal epithelial stem cells following severe ischemic injury}, volume={316}, ISSN={0193-1857 1522-1547}, url={http://dx.doi.org/10.1152/ajpgi.00262.2018}, DOI={10.1152/ajpgi.00262.2018}, abstractNote={ 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. }, number={4}, journal={American Journal of Physiology-Gastrointestinal and Liver Physiology}, publisher={American Physiological Society}, author={Gonzalez, Liara M. and Stewart, Amy Stieler and Freund, John and Kucera, Cecilia Renee and Dekaney, Christopher M. and Magness, Scott T. and Blikslager, Anthony T.}, year={2019}, month={Apr}, pages={G482–G494} } @article{ziegler_pridgen_mills_gonzalez_van landeghem_odle_blikslager_2018, title={Epithelial restitution defect in neonatal jejunum is rescued by juvenile mucosal homogenate in a pig model of intestinal ischemic injury and repair}, volume={13}, ISSN={1932-6203}, url={http://dx.doi.org/10.1371/journal.pone.0200674}, DOI={10.1371/journal.pone.0200674}, abstractNote={Intestinal ischemic injury results sloughing of the mucosal epithelium leading to host sepsis and death unless the mucosal barrier is rapidly restored. Neonatal necrotizing enterocolitis (NEC) and volvulus in infants is associated with intestinal ischemia, sepsis and high mortality rates. We have characterized intestinal ischemia/ repair using a highly translatable porcine model in which juvenile (6-8-week-old) pigs completely and efficiently restore barrier function by way of rapid epithelial restitution and tight junction re-assembly. In contrast, separate studies showed that younger neonatal (2-week-old) pigs exhibited less robust recovery of barrier function, which may model an important cause of high mortality rates in human infants with ischemic intestinal disease. Therefore, we aimed to further refine our repair model and characterize defects in neonatal barrier repair. Here we examine the defect in neonatal mucosal repair that we hypothesize is associated with hypomaturity of the epithelial and subepithelial compartments. Following jejunal ischemia in neonatal and juvenile pigs, injured mucosa was stripped from seromuscular layers and recovered ex vivo while monitoring transepithelial electrical resistance (TEER) and 3H-mannitol flux as measures of barrier function. While ischemia-injured juvenile mucosa restored TEER above control levels, reduced flux over the recovery period and showed 93±4.7% wound closure, neonates exhibited no change in TEER, increased flux, and a 11±23.3% increase in epithelial wound size. Scanning electron microscopy revealed enterocytes at the wound margins of neonates failed to assume the restituting phenotype seen in restituting enterocytes of juveniles. To attempt rescue of injured neonatal mucosa, neonatal experiments were repeated with the addition of exogenous prostaglandins during ex vivo recovery, ex vivo recovery with full thickness intestine, in vivo recovery and direct application of injured mucosal homogenate from neonates or juveniles. Neither exogenous prostaglandins, intact seromuscular intestinal layers, nor in vivo recovery enhanced TEER or restitution in ischemia-injured neonatal mucosa. However, ex vivo exogenous application of injured juvenile mucosal homogenate produced a significant increase in TEER and enhanced histological restitution to 80±4.4% epithelial coveragein injured neonatal mucosa. Thus, neonatal mucosal repair can be rescued through direct contact with the cellular and non-cellular milieu of ischemia-injured mucosa from juvenile pigs. These findings support the hypothesis that a defect in mucosal repair in neonates is due to immature repair mechanisms within the mucosal compartment. Future studies to identify and rescue specific defects in neonatal intestinal repair mechanisms will drive development of novel clinical interventions to reduce mortality in infants affected by intestinal ischemic injury.}, number={8}, journal={PLOS ONE}, publisher={Public Library of Science (PLoS)}, author={Ziegler, Amanda L. and Pridgen, Tiffany A. and Mills, Juliana K. and Gonzalez, Liara M. and Van Landeghem, Laurianne and Odle, Jack and Blikslager, Anthony T.}, editor={Karhausen, JörnEditor}, year={2018}, month={Aug}, pages={e0200674} } @article{blikslager_gonzalez_2018, title={Equine intestinal mucosal pathobiology}, volume={6}, journal={Annual review of animal biosciences, vol 6}, author={Blikslager, A. and Gonzalez, L.}, year={2018}, pages={157–175} } @article{fletcher_mansell_martin_borst_barnes_gonzalez_2018, title={Gross Morphometry, Histomorphometry, and Immunohistochemistry Confirm Early and Persistent Jejunal Crypt Hyperplasia in Poults with Enteritis and Depressed Growth}, volume={62}, ISSN={0005-2086 1938-4351}, url={http://dx.doi.org/10.1637/11759-101717-reg.1}, DOI={10.1637/11759-101717-reg.1}, abstractNote={SUMMARY Phosphorylated histone 3 (PH3) and cleaved caspase 3 (CCASP3) were used to detect proliferating and apoptotic cells, respectively, in the jejunums of female sibling poults, with and without enteritis and depressed growth, from hatch to day 35. Poults that developed enteritis and depressed growth (SIB flock) were raised on a commercial farm in eastern North Carolina, whereas poults with normal growth and no enteritis (TAU flock) were raised in the Teaching Animal Unit at North Carolina State University College of Veterinary Medicine. Beginning on day 5 through day 35 and at processing, TAU poults were significantly heavier than SIB poults. Jejunal weights, relative jejunal weights, and jejunal densities were greater in SIB poults from day 10 through 35. Jejunal efficiency (body weight /jejunal length) was higher in TAU poults at day 5 and days 10 through 35. Mucosal thickness was greater in SIB poults between days 7 and 21 but greater in TAU poults at days 28 and 35. From day 7 to 35, villus-to-crypt ratios were higher for TAU poults and lower for SIB poults because hyperplastic crypts formed a greater percentage of the mucosa in SIB poults. By day 7, PH3- and CCASP3-positive cells were increased in SIB poults, showing that mucosal changes resulted from combined crypt epithelial hyperplasia and increased apoptosis of villous enterocytes. Findings in this study confirm that enteritis, in the absence of clinical signs, and depressed growth in turkey poults begins by day 7, can be identified microscopically, persists for at least 35 days, is associated with lower processing weights, and has a profound negative effect on turkey growth.}, number={2}, journal={Avian Diseases}, publisher={American Association of Avian Pathologists (AAAP)}, author={Fletcher, O. J. and Mansell, R. and Martin, M. P. and Borst, L. B. and Barnes, H. John and Gonzalez, L. M.}, year={2018}, month={Jun}, pages={163–170} } @article{stieler stewart_freund_blikslager_gonzalez_2018, title={Intestinal Stem Cell Isolation and Culture in a Porcine Model of Segmental Small Intestinal Ischemia}, volume={5}, ISSN={1940-087X}, url={http://dx.doi.org/10.3791/57647}, DOI={10.3791/57647}, abstractNote={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.}, number={135}, journal={Journal of Visualized Experiments}, publisher={MyJove Corporation}, author={Stieler Stewart, Amy and Freund, John M and Blikslager, Anthony T and Gonzalez, Liara M}, year={2018}, month={May} } @article{gonzalez_2018, title={Large colon volvulus: The continued conundrum}, volume={30}, ISSN={["2042-3292"]}, url={https://doi.org/10.1111/eve.12748}, DOI={10.1111/eve.12748}, abstractNote={Equine Veterinary EducationVolume 30, Issue 6 p. 312-314 Clinical Commentary Large colon volvulus: The continued conundrum L. M. Gonzalez, Corresponding Author L. M. Gonzalez Lmgonza4@ncsu.edu orcid.org/0000-0003-3972-2812 Department of Clinical Sciences College of Veterinary Medicine, North Carolina State University, Raleigh, USACorresponding author email: Lmgonza4@ncsu.eduSearch for more papers by this author L. M. Gonzalez, Corresponding Author L. M. Gonzalez Lmgonza4@ncsu.edu orcid.org/0000-0003-3972-2812 Department of Clinical Sciences College of Veterinary Medicine, North Carolina State University, Raleigh, USACorresponding author email: Lmgonza4@ncsu.eduSearch for more papers by this author First published: 22 March 2017 https://doi.org/10.1111/eve.12748Read the full textAboutPDF ToolsExport citationAdd to favoritesTrack citation ShareShare Give accessShare full text accessShare full-text accessPlease review our Terms and Conditions of Use and check box below to share full-text version of article.I have read and accept the Wiley Online Library Terms and Conditions of UseShareable LinkUse the link below to share a full-text version of this article with your friends and colleagues. Learn more.Copy URL Share a linkShare onFacebookTwitterLinkedInRedditWechat No abstract is available for this article. Volume30, Issue6June 2018Pages 312-314 RelatedInformation}, number={6}, journal={EQUINE VETERINARY EDUCATION}, publisher={Wiley}, author={Gonzalez, L. M.}, year={2018}, month={Jun}, pages={312–314} } @article{royal_hunt_gonzalez_lewbart_bailey_2018, title={Veterinary Medical Students' Motivations for Exercise}, volume={45}, ISSN={0748-321X 1943-7218}, url={http://dx.doi.org/10.3138/jvme.0117-004r}, DOI={10.3138/jvme.0117-004r}, abstractNote={ The Centers for Disease Control (CDC) declares exercise to be one of the most important activities one can do to improve health. The benefits of exercise are well documented and include both physiologic and psychological health. Given the current landscape of wellness issues in veterinary medical education, it is necessary that students engage in exercise activities to manage stress and increase overall health. Therefore, to develop targeted interventions with the greatest likelihood for success, it is first necessary to understand what motivates veterinary medical students to exercise given their unique situational and environmental factors. This study is the first to explore this issue systematically in veterinary medical education, thus it is the authors' hope that the findings from this research will help identify exercise-related wellness interventions that could be implemented in veterinary medical schools. }, number={3}, journal={Journal of Veterinary Medical Education}, publisher={University of Toronto Press Inc. (UTPress)}, author={Royal, Kenneth D. and Hunt, Suzanne A. and Gonzalez, Liara M. and Lewbart, Gregory A. and Bailey, Kate M.}, year={2018}, month={Aug}, pages={367–373} } @article{royal_hunt_gonzalez_lewbart_bailey_2018, title={Veterinary Medical Students' Motivations for Exercise}, volume={1}, ISSN={0748-321X 1943-7218}, url={http://dx.doi.org/10.3138/jvme.0117-004}, DOI={10.3138/jvme.0117-004}, abstractNote={The Centers for Disease Control (CDC) declares exercise to be one of the most important activities one can do to improve health. The benefits of exercise are well documented and include both physiologic and psychological health. Given the current landscape of wellness issues in veterinary medical education, it is necessary that students engage in exercise activities to manage stress and increase overall health. Therefore, to develop targeted interventions with the greatest likelihood for success, it is first necessary to understand what motivates veterinary medical students to exercise given their unique situational and environmental factors. This study is the first to explore this issue systematically in veterinary medical education, thus it is the authors' hope that the findings from this research will help identify exercise-related wellness interventions that could be implemented in veterinary medical schools.}, journal={Journal of Veterinary Medical Education}, publisher={University of Toronto Press Inc. (UTPress)}, author={Royal, Kenneth D. and Hunt, Suzanne A. and Gonzalez, Liara M. and Lewbart, Gregory A. and Bailey, Kate M.}, year={2018}, month={Jan}, pages={1–7} } @article{dellon_joshi_blikslager_gonzalez_pridgen_whitlow_ivanovic_slaughter_garman_karp_et al._2017, title={A Novel Inflammation-Activated Drug Delivery System Using Self-Assembling Hydrogel Doubles Esophageal Dwell Time in an Esophageal Injury Porcine Model}, volume={152}, ISSN={0016-5085}, url={http://dx.doi.org/10.1016/S0016-5085(17)32956-6}, DOI={10.1016/S0016-5085(17)32956-6}, number={5}, journal={Gastroenterology}, publisher={Elsevier BV}, author={Dellon, Evan S. and Joshi, Nitin and Blikslager, Anthony and Gonzalez, Liara M. and Pridgen, Tiffany and Whitlow, Ann and Ivanovic, Marija and Slaughter, Kai and Garman, Katherine S. and Karp, Jeffrey M. and et al.}, year={2017}, month={Apr}, pages={S859–S860} } @article{stewart_freund_gonzalez_2017, title={Advanced three-dimensional culture of equine intestinal epithelial stem cells}, volume={50}, ISSN={0425-1644}, url={http://dx.doi.org/10.1111/evj.12734}, DOI={10.1111/evj.12734}, abstractNote={Summary}, number={2}, journal={Equine Veterinary Journal}, publisher={Wiley}, author={Stewart, A. Stieler and Freund, J. M. and Gonzalez, L. M.}, year={2017}, month={Sep}, pages={241–248} } @misc{stewart_pratt-phillips_gonzalez_2017, title={Alterations in Intestinal Permeability: The Role of the "Leaky Gut" in Health and Disease}, volume={52}, ISSN={["1542-7412"]}, url={https://doi.org/10.1016/j.jevs.2017.02.009}, DOI={10.1016/j.jevs.2017.02.009}, abstractNote={All species, including horses, suffer from alterations that increase intestinal permeability. These alterations, also known as “leaky gut,” may lead to severe disease as the normal intestinal barrier becomes compromised and can no longer protect against harmful luminal contents including microbial toxins and pathogens. Leaky gut results from a variety of conditions including physical stressors, decreased blood flow to the intestine, inflammatory disease, and pathogenic infections, among others. Several testing methods exist to diagnose these alterations in both a clinical and research setting. To date, most research has focused on regulation of the host immune response due to the wide variety of factors that can potentially influence the intestinal barrier. This article serves to review the normal intestinal barrier, measurement of barrier permeability, pathogenesis and main causes of altered permeability, and highlight potential alternative therapies of leaky gut in horses while relating what has been studied in other species. Conditions resulting in barrier dysfunction and leaky gut can be a major cause of decreased performance and also death in horses. A better understanding of the intestinal barrier in disease and ways to optimize the function of this barrier is vital to the long-term health and maintenance of these animals.}, journal={JOURNAL OF EQUINE VETERINARY SCIENCE}, publisher={Elsevier BV}, author={Stewart, Amy Stieler and Pratt-Phillips, Shannon and Gonzalez, Liara M.}, year={2017}, month={May}, pages={10–22} } @article{kruger_gonzalez_pridgen_mccall_furstenberg_harnden_carnighan_cox_blikslager_garman_et al._2017, title={Ductular and proliferative response of esophageal submucosal glands in a porcine model of esophageal injury and repair}, volume={313}, ISSN={0193-1857 1522-1547}, url={http://dx.doi.org/10.1152/ajpgi.00036.2017}, DOI={10.1152/ajpgi.00036.2017}, abstractNote={ Esophageal injury is a risk factor for diseases such as Barrett’s esophagus (BE) and esophageal adenocarcinoma. To improve understanding of signaling pathways associated with both normal and abnormal repair, animal models are needed. Traditional rodent models of esophageal repair are limited by the absence of esophageal submucosal glands (ESMGs), which are present in the human esophagus. Previously, we identified acinar ductal metaplasia in human ESMGs in association with both esophageal injury and cancer. In addition, the SOX9 transcription factor has been associated with generation of columnar epithelium and the pathogenesis of BE and is present in ESMGs. To test our hypothesis that ESMGs activate after esophageal injury with an increase in proliferation, generation of a ductal phenotype, and expression of SOX9, we developed a porcine model of esophageal injury and repair using radiofrequency ablation (RFA). The porcine esophagus contains ESMGs, and RFA produces a consistent and reproducible mucosal injury in the esophagus. Here we present a temporal assessment of this model of esophageal repair. Porcine esophagus was evaluated at 0, 6, 18, 24, 48, and 72 h and 5 and 7 days following RFA and compared with control uninjured esophagus. Following RFA, ESMGs demonstrated an increase in ductal phenotype, echoing our prior studies in humans. Proliferation increased in both squamous epithelium and ESMGs postinjury with a prominent population of SOX9-positive cells in ESMGs postinjury. This model promises to be useful in future experiments evaluating mechanisms of esophageal repair. }, number={3}, journal={American Journal of Physiology-Gastrointestinal and Liver Physiology}, publisher={American Physiological Society}, author={Kruger, L. and Gonzalez, Liara and Pridgen, T. A. and McCall, S. J. and Furstenberg, R. J. and Harnden, I. and Carnighan, G. E. and Cox, A. M. and Blikslager, Anthony and Garman, K. S. and et al.}, year={2017}, month={Sep}, pages={G180–G191} } @article{von furstenberg_li_stolarchuk_feder_campbell_kruger_gonzalez_blikslager_cardona_mccall_et al._2017, title={Porcine Esophageal Submucosal Gland Culture Model Shows Capacity for Proliferation and Differentiation}, volume={4}, ISSN={2352-345X}, url={http://dx.doi.org/10.1016/j.jcmgh.2017.07.005}, DOI={10.1016/j.jcmgh.2017.07.005}, abstractNote={Background & AimsAlthough cells comprising esophageal submucosal glands (ESMGs) represent a potential progenitor cell niche, new models are needed to understand their capacity to proliferate and differentiate. By histologic appearance, ESMGs have been associated with both overlying normal squamous epithelium and columnar epithelium. Our aim was to assess ESMG proliferation and differentiation in a 3-dimensional culture model.MethodsWe evaluated proliferation in human ESMGs from normal and diseased tissue by proliferating cell nuclear antigen immunohistochemistry. Next, we compared 5-ethynyl-2′-deoxyuridine labeling in porcine ESMGs in vivo before and after esophageal injury with a novel in vitro porcine organoid ESMG model. Microarray analysis of ESMGs in culture was compared with squamous epithelium and fresh ESMGs.ResultsMarked proliferation was observed in human ESMGs of diseased tissue. This activated ESMG state was recapitulated after esophageal injury in an in vivo porcine model, ESMGs assumed a ductal appearance with increased proliferation compared with control. Isolated and cultured porcine ESMGs produced buds with actively cycling cells and passaged to form epidermal growth factor–dependent spheroids. These spheroids were highly proliferative and were passaged multiple times. Two phenotypes of spheroids were identified: solid squamous (P63+) and hollow/ductal (cytokeratin 7+). Microarray analysis showed spheroids to be distinct from parent ESMGs and enriched for columnar transcripts.ConclusionsOur results suggest that the activated ESMG state, seen in both human disease and our porcine model, may provide a source of cells to repopulate damaged epithelium in a normal manner (squamous) or abnormally (columnar epithelium). This culture model will allow the evaluation of factors that drive ESMGs in the regeneration of injured epithelium. The raw microarray data have been uploaded to the National Center for Biotechnology Information Gene Expression Omnibus (accession number: GSE100543). Although cells comprising esophageal submucosal glands (ESMGs) represent a potential progenitor cell niche, new models are needed to understand their capacity to proliferate and differentiate. By histologic appearance, ESMGs have been associated with both overlying normal squamous epithelium and columnar epithelium. Our aim was to assess ESMG proliferation and differentiation in a 3-dimensional culture model. We evaluated proliferation in human ESMGs from normal and diseased tissue by proliferating cell nuclear antigen immunohistochemistry. Next, we compared 5-ethynyl-2′-deoxyuridine labeling in porcine ESMGs in vivo before and after esophageal injury with a novel in vitro porcine organoid ESMG model. Microarray analysis of ESMGs in culture was compared with squamous epithelium and fresh ESMGs. Marked proliferation was observed in human ESMGs of diseased tissue. This activated ESMG state was recapitulated after esophageal injury in an in vivo porcine model, ESMGs assumed a ductal appearance with increased proliferation compared with control. Isolated and cultured porcine ESMGs produced buds with actively cycling cells and passaged to form epidermal growth factor–dependent spheroids. These spheroids were highly proliferative and were passaged multiple times. Two phenotypes of spheroids were identified: solid squamous (P63+) and hollow/ductal (cytokeratin 7+). Microarray analysis showed spheroids to be distinct from parent ESMGs and enriched for columnar transcripts. Our results suggest that the activated ESMG state, seen in both human disease and our porcine model, may provide a source of cells to repopulate damaged epithelium in a normal manner (squamous) or abnormally (columnar epithelium). This culture model will allow the evaluation of factors that drive ESMGs in the regeneration of injured epithelium. The raw microarray data have been uploaded to the National Center for Biotechnology Information Gene Expression Omnibus (accession number: GSE100543).}, number={3}, journal={Cellular and Molecular Gastroenterology and Hepatology}, publisher={Elsevier BV}, author={von Furstenberg, Richard J. and Li, Joy and Stolarchuk, Christina and Feder, Rachel and Campbell, Alexa and Kruger, Leandi and Gonzalez, Liara M. and Blikslager, Anthony T. and Cardona, Diana M. and McCall, Shannon J. and et al.}, year={2017}, month={Nov}, pages={385–404} } @article{kucera_stranahan_hughes_blikslager_gonzalez_2017, title={Protein biomarker of cell proliferation determines survival to discharge in cases of equine large colon volvulus}, volume={50}, ISSN={0425-1644}, url={http://dx.doi.org/10.1111/evj.12767}, DOI={10.1111/evj.12767}, abstractNote={Summary}, number={4}, journal={Equine Veterinary Journal}, publisher={Wiley}, author={Kucera, C. R. and Stranahan, L. W. and Hughes, F. and Blikslager, A. T. and Gonzalez, L. M.}, year={2017}, month={Nov}, pages={452–456} } @article{lima_lin_jacobi_man_sommer_flowers_blikslager_gonzalez_odle_2017, title={Supplementation of Maternal Diets with Docosahexaenoic Acid and Methylating Vitamins Impacts Growth and Development of Fetuses from Malnourished Gilts}, volume={2}, ISSN={2475-2991}, url={http://dx.doi.org/10.3945/cdn.117.001958}, DOI={10.3945/cdn.117.001958}, abstractNote={Abstract Background Like many species, pregnant swine mobilize and repartition body nutrient stores during extreme malnutrition to support fetal development. Objective The objective of this study was to model chronic human maternal malnutrition and measure effects of methylating-vitamins (MVs, containing choline, folate, B-6, B-12, and riboflavin) and docosahexaenoic acid (DHA) supplementation on fetal growth and development. Methods Pregnant gilts (n = 24) were either fully nourished (2.0 kg/d) with a corn-plus-isolated-soy-protein basal diet (control) supplemented with MVs and DHA or nourishment was restricted throughout gestation. Basal diet fed to malnourished gilts was reduced progressively from 50% to 70% restriction (1.0 to 0.6 kg/d) and was supplemented following a 2 (±MVs) x 2 (±DHA) factorial design. Full-term c-sections were performed to assess impacts on low and normal birth weight (LBW/NBW) fetuses (n = 238). Results Body weight gain of malnourished gilts was 10% of full-fed control dams (P < 0.05), but offspring birth weight, length, girth, and percentage of LBW fetuses were not different between treatments. The number of pigs per litter was reduced by 30% in malnourished control dams. Fetal brain weights were reduced by 7% compared to positive controls (P < 0.05). Micronutrient supplementation to malnourished dams increased fetal brain weights back to full-fed control levels. Dams with DHA produced offspring with higher DHA concentrations in brain and liver (P < 0.05). Plasma choline concentration was 4-fold higher in fetuses from unsupplemented malnourished dams (P < 0.0001). Global DNA methylation status of fetuses from restricted dams was higher than in control fetuses, including brain, liver, heart, muscle, and placenta tissues (P < 0.05). Addition of DHA increased methylation in LBW fetal brains (P < 0.05). Conclusions Despite the mobilization of maternal stores, malnourished litters displayed reduced brain development that was fully mitigated by micronutrient supplementation. Severe maternal malnutrition increased global DNA methylation in several fetal tissues that was unaltered by choline and B-vitamin supplementation.}, number={3}, journal={Current Developments in Nutrition}, publisher={Oxford University Press (OUP)}, author={Lima, Hope K and Lin, Xi and Jacobi, Sheila K and Man, Caolai and Sommer, Jeffrey and Flowers, William and Blikslager, Anthony and Gonzalez, Liara and Odle, Jack}, year={2017}, month={Dec} } @article{simpson_gonzalez_chung_blikslager_magness_piedrahita_2016, title={27 AN IMPROVED LARGE ANIMAL MODEL FOR THE STUDY OF ADULT STEM CELLS}, volume={28}, ISSN={1031-3613}, url={http://dx.doi.org/10.1071/RDV28N2AB27}, DOI={10.1071/RDV28N2AB27}, abstractNote={ Murine models for the study of adult stem cell populations have broadened the understanding of previously uncharacterized stem cell niches. The development of murine reporter lines for the leucine-rich repeat-containing G-protein-coupled receptor-5 (Lgr5) has highlighted the importance of this gene as a stem cell marker in the stomach, intestine, hair follicle, liver, and kidney in mice. These models however have significant limitations in terms of translational applications because of anatomical and physiological differences between humans and mice. In order to overcome these limitations, we have sought to develop a porcine LGR5 reporter line. We report the generation of a porcine stem cell reporter line using the combination of transcription activator-like effector nucleases and somatic cell NT. Transcription activator-like effector nuclease-mediated homologous recombination was used to drive the integration of an internal ribosome entry site green fluorescent protein fusion into the 3′ untranslated region of the LGR5 locus in porcine fetal fibroblast cells. Multiple cell lines were developed and screened for the proper integration event. Upon confirmation of proper integration by genomic DNA sequencing, these lines were used as donors for somatic cell NT. Transfer of the somatic cell NT reconstructed embryos to a surrogate gilt resulted in 3 live births, and the establishment of a founder line of LGR5-green fluorescent protein reporter pigs. We have begun to characterise these lines, having observed fluorescent labelling of putative stem cell populations in the intestinal crypts and hair follicles from these animals. Many of these observations parallel the expression patterns observed in similar murine models. We have confirmed the fluorescent reporter signal by immunohistochemistry using an anti-green fluorescent protein antibody, and are working towards colocalization studies using anti-LGR5 antibodies and RNA in situ hybridization, as well as the characterisation of additional stem cell populations in the pig. The development of this line of transgenic pigs represents significant progress toward the study of adult stem cells, their progenitors, and the stem cell niche, using a large animal model with an anatomy, physiology, and ability to recapitulate human disease that overcomes the current limitations of rodent models. Funding was provided by NIH R21OD019738. }, number={2}, journal={Reproduction, Fertility and Development}, publisher={CSIRO Publishing}, author={Simpson, S. and Gonzalez, L. and Chung, J. and Blikslager, A. and Magness, S. and Piedrahita, J.}, year={2016}, pages={143} } @article{royal_gonzalez_2016, title={An Evaluation of the Psychometric Properties of an Advising Survey for Medical and Professional Program Students}, volume={6}, ISSN={1927-0534 1927-0526}, url={http://dx.doi.org/10.5539/jedp.v6n1p195}, DOI={10.5539/jedp.v6n1p195}, abstractNote={

The purpose of this study was to evaluate the psychometric properties of a newly developed instrument intended to measure faculty competence as it pertains to their role as advisors, particularly in medical and professional programs. A total of 166 students completed the Faculty Advisor’s Skills and Behaviors Inventory (FASBI). The psychometric properties of the FASBI were evaluated using the Rasch Rating Scale Model. Results indicate the FASBI is a psychometrically-sound instrument capable of producing valid and reproducible measures.

}, number={1}, journal={Journal of Educational and Developmental Psychology}, publisher={Canadian Center of Science and Education}, author={Royal, Kenneth D. and Gonzalez, Liara M.}, year={2016}, month={Apr}, pages={195} } @inbook{gonzalez_2016, place={Hoboken, NJ}, edition={3rd}, title={Intestinal Stem Cells}, booktitle={Equine Acute Abdomen}, publisher={Wiley & Sons, Inc}, author={Gonzalez, L.M.}, editor={Blikslager, A.T. and White, N.A. and Moore, J.N. and Mair, T.S.Editors}, year={2016} } @inbook{gonzalez_2016, place={Hoboken, NJ}, edition={3rd}, title={Intestinal Viability}, booktitle={Equine Acute Abdomen}, publisher={Wiley & Sons, Inc}, author={Gonzalez, L.M.}, editor={Blikslager, A.T. and White, N.A. and Moore, J.N. and Mair, T.S.Editors}, year={2016}, pages={570} } @article{ziegler_gonzalez_blikslager_2016, title={Large Animal Models: The Key to Translational Discovery in Digestive Disease Research}, volume={2}, ISSN={2352-345X}, url={http://dx.doi.org/10.1016/j.jcmgh.2016.09.003}, DOI={10.1016/j.jcmgh.2016.09.003}, abstractNote={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 (IPEC-J2) 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. 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 (IPEC-J2) 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. SummaryRodent models have been instrumental in furthering our understanding of gastrointestinal pathophysiology. However, there are important limitations to translating findings from rodent models to human digestive disease, and some of these can be overcome by large animal models. Rodent models have been instrumental in furthering our understanding of gastrointestinal pathophysiology. However, there are important limitations to translating findings from rodent models to human digestive disease, and some of these can be overcome by large animal models. Gastrointestinal diseases account for 10% of annual mortalities in the United States, and approximately 70 million Americans experience gastrointestinal illness each year.1Peery A.F. Dellon E.S. Lund J. et al.Burden of gastrointestinal disease in the United States: 2012 update.Gastroenterology. 2012; 143 (e1–3): 1179-1187Abstract Full Text Full Text PDF PubMed Scopus (1417) Google Scholar Because of the limitations of studying human gastrointestinal disease in a clinical environment, the use of animal models to examine the mechanisms of gastrointestinal disease has been of great importance. Rodent models remain the most commonly used animal model for the study of human disease because of their relatively low cost and maintenance requirements, rapid reproduction rates, and availability of research tools such as murine antibodies.2Low M.J. Mouse models in gastroenterology research.Gastroenterology. 2012; 143: 1410-1412Abstract Full Text Full Text PDF PubMed Scopus (4) Google Scholar, 3Chung S.K. Lee A.Y. Chung S.S. Mouse models for human diseases.Hong Kong Med J. 1997; 3: 201-209PubMed Google Scholar Transgenic and knockout mice, as well as chemically induced neoplastic and inflammatory disease models in mice, have provided a tremendous amount of information on the underlying mechanisms of clinical diseases.4Wagner S.J. Schmidt A. Effenberger M.J. et al.Semisynthetic diet ameliorates Crohn's disease-like ileitis in TNFDeltaARE/WT mice through antigen-independent mechanisms of gluten.Inflamm Bowel Dis. 2013; 19: 1285-1294Crossref PubMed Scopus (33) Google Scholar, 5Pizarro T.T. Pastorelli L. Bamias G. et al.SAMP1/YitFc mouse strain: a spontaneous model of Crohn's disease-like ileitis.Inflamm Bowel Dis. 2011; 17: 2566-2584Crossref PubMed Scopus (120) Google Scholar However, rodent models frequently fail to fully mimic clinical signs and significant pathologic hallmarks of human diseases.6Boivin G.P. Washington K. Yang K. et al.Pathology of mouse models of intestinal cancer: consensus report and recommendations.Gastroenterology. 2003; 124: 762-777Abstract Full Text Full Text PDF PubMed Scopus (415) Google Scholar, 7Flisikowska T. Merkl C. Landmann M. et al.A porcine model of familial adenomatous polyposis.Gastroenterology. 2012; 143 (e1–7): 1173-1175Abstract Full Text Full Text PDF PubMed Scopus (95) Google Scholar Because of this, there has been recent increased interest within the scientific community in developing large animal models that more closely approximate the clinical and pathologic features of human disease.8US Department of Health and Human Services. Opportunities and Challenges in Digestive Diseases Research: Recommendations of the National Commission on Digestive Diseases. March 2009; NIH Publication No. 08-6514.Google Scholar, 9Cibelli J. Emborg M.E. Prockop D.J. et al.Strategies for improving animal models for regenerative medicine.Cell Stem Cell. 2013; 12: 271-274Abstract Full Text Full Text PDF PubMed Scopus (61) Google Scholar Of the large animal species used for translational research, the pig offers a marked advantage because this species has important anatomic and physiological similarities with human beings, particularly in regard to the gastrointestinal tract.10Patterson J.K. Lei X.G. Miller D.D. The pig as an experimental model for elucidating the mechanisms governing dietary influence on mineral absorption.Exp Biol Med (Maywood). 2008; 233: 651-664Crossref PubMed Scopus (193) Google Scholar, 11Kararli T.T. Comparison of the gastrointestinal anatomy, physiology, and biochemistry of humans and commonly used laboratory animals.Biopharm Drug Dispos. 1995; 16: 351-380Crossref PubMed Scopus (1117) Google Scholar This review summarizes and evaluates the current uses of large animal models in gastrointestinal research with a particular emphasis on porcine models. In general, any nonrodent mammalian animal species used for translational research is considered a large animal model. Although rodent models have been invaluable in furthering the mechanistic understanding of human disease, there are circumstances in which choosing a large animal species in place of a rodent model for gastrointestinal work is scientifically justifiable. For instance, when investigating gastric disease, the dog, pig, and monkey all have a glandular-type stomach similar to that of human beings, whereas the murine stomach has both glandular and nonglandular regions and therefore may have differences in gastric pathophysiology (Figure 1).11Kararli T.T. Comparison of the gastrointestinal anatomy, physiology, and biochemistry of humans and commonly used laboratory animals.Biopharm Drug Dispos. 1995; 16: 351-380Crossref PubMed Scopus (1117) Google Scholar Of the large animal species used in translational gastrointestinal research, the dog has been used most extensively because canine gastrointestinal anatomy and physiology are considered to be highly similar to that of human beings, and dogs show spontaneous naturally occurring diseases in common with human beings, particularly neoplasia.12Kirk A.D. Crossing the bridge: large animal models in translational transplantation research.Immunol Rev. 2003; 196: 176-196Crossref PubMed Scopus (123) Google Scholar However, dogs are highly sensitive to some disease models, particularly intestinal ischemia, and experience high rates of mortality.13Block T. Isaksson H.S. Acosta S. et al.Altered mRNA expression due to acute mesenteric ischaemia in a porcine model.Eur J Vasc Endovasc Surg. 2011; 41: 281-287Abstract Full Text Full Text PDF PubMed Scopus (12) Google Scholar In addition, there is growing social aversion to the use of the dog as a laboratory animal.14Yandza T. Tauc M. Saint-Paul M.C. et al.The pig as a preclinical model for intestinal ischemia-reperfusion and transplantation studies.J Surg Res. 2012; 178: 807-819Abstract Full Text Full Text PDF PubMed Scopus (28) Google Scholar Calves have been established as a model for human enteric infectious disease, particularly Salmonellosis. Cattle may become naturally infected with Salmonella enterica serotype typhimurium and they show a disease state very analogous to human salmonellosis. Therefore, the calf commonly is used to study Salmonella species infection and the host–pathogen interaction, translating findings to human disease as well as to veterinary medicine and agriculture.15Elfenbein J.R. Endicott-Yazdani T. Porwollik S. et al.Novel determinants of intestinal colonization of Salmonella enterica serotype typhimurium identified in bovine enteric infection.Infect Immun. 2013; 81: 4311-4320Crossref Scopus (18) Google Scholar This recently was called Dual with Dual Benefit in a Program Announcement from the National Institutes of Health (http://grants.nih.gov/grants/guide/pa-files/PAR-16-366.html). However, the application of ruminant models for the study of other human gastrointestinal biology is limited owing to the fundamental difference in digestive anatomy and physiology. Alternatively, the pig is becoming progressively appreciated as a distinctly advantageous model for human beings in numerous fields of science, and an increasing number of textbooks, articles, and proceedings are being published that outline pig models in biomedical research, including digestive disease research (Table 1).16Swindle M.M. Makin A. Herron A.J. et al.Swine as models in biomedical research and toxicology testing.Vet Pathol. 2012; 49: 344-356Crossref PubMed Scopus (868) Google Scholar The pig has many fundamental anatomic, physiological, genomic, proteomic, immunologic, and nutritional similarities to human beings.12Kirk A.D. Crossing the bridge: large animal models in translational transplantation research.Immunol Rev. 2003; 196: 176-196Crossref PubMed Scopus (123) Google Scholar, 16Swindle M.M. Makin A. Herron A.J. et al.Swine as models in biomedical research and toxicology testing.Vet Pathol. 2012; 49: 344-356Crossref PubMed Scopus (868) Google Scholar, 17Roura E. Koopmans S.J. Lallès J.P. et al.Critical review evaluating the pig as a model for human nutritional physiology.Nutr Res Rev. 2016; 29: 60-90Crossref Scopus (168) Google Scholar, 18Rothkotter H.J. Sowa E. Pabst R. The pig as a model of developmental immunology.Hum Exp Toxicol. 2002; 21: 533-536Crossref PubMed Scopus (94) Google Scholar, 19Bendixen E. Danielsen M. Larsen K. et al.Advances in porcine genomics and proteomics–a toolbox for developing the pig as a model organism for molecular biomedical research.Brief Funct Genomics. 2010; 9: 208-219Crossref PubMed Scopus (128) Google Scholar, 20Lunney J.K. Advances in swine biomedical model genomics.Int J Biol Sci. 2007; 3: 179-184Crossref PubMed Scopus (416) Google Scholar, 21Hart E.A. Caccamo M. Harrow J.L. et al.Lessons learned from the initial sequencing of the pig genome: comparative analysis of an 8 Mb region of pig chromosome 17.Genome Biol. 2007; 8: R168Crossref PubMed Scopus (37) Google Scholar, 22Ibrahim Z. Busch J. Awwad M. et al.Selected physiologic compatibilities and incompatibilities between human and porcine organ systems.Xenotransplantation. 2006; 13: 488-499Crossref PubMed Scopus (147) Google Scholar The pig also shows potential for interspecies transplantation work, as well as the ability to fulfill United States Food and Drug Administration requirements for pharmaceutical testing.23Gonzalez L.M. Moeser A.J. Blikslager A.T. Porcine models of digestive disease: the future of large animal translational research.Transl Res. 2015; 166: 12-27Abstract Full Text Full Text PDF Scopus (118) Google Scholar These features of the pig combined with an increasing availability of biological tools and reagents for use to study porcine tissue make the pig arguably the best model available for translational biomedical research.Table 1Porcine Digestive Disease Models AvailableDisease or syndrome modeledReferencesEsophageal metaplasia and neoplasia24Garman K.S. Orlando R.C. Chen X. Review: experimental models for Barrett's esophagus and esophageal adenocarcinoma.Am J Physiol Gastrointest Liver Physiol. 2012; 302: G1231-G1243Crossref Scopus (25) Google Scholar, 38Kapoor H. Lohani K.R. Lee T.H. et al.Animal models of Barrett's esophagus and esophageal adenocarcinoma-past, present, and future.Clin Transl Sci. 2015; 8: 841-847Crossref Scopus (36) Google ScholarStress-induced intestinal dysfunction75Smith F. Clark J.E. Overman B.L. et al.Early weaning stress impairs development of mucosal barrier function in the porcine intestine.Am J Physiol Gastrointest Liver Physiol. 2010; 298: G352-G363Crossref PubMed Scopus (331) Google Scholar, 76Moeser A.J. Ryan K.A. Nighot P.K. et al.Gastrointestinal dysfunction induced by early weaning is attenuated by delayed weaning and mast cell blockade in pigs.Am J Physiol Gastrointest Liver Physiol. 2007; 293: G413-G421Crossref PubMed Scopus (175) Google Scholar, 77Moeser A.J. Klok C.V. Ryan K.A. et al.Stress signaling pathways activated by weaning mediate intestinal dysfunction in the pig.Am J Physiol Gastrointest Liver Physiol. 2007; 292: G173-G181Crossref PubMed Scopus (212) Google Scholar, 78Overman E.L. Rivier J.E. Moeser A.J. CRF induces intestinal epithelial barrier injury via the release of mast cell proteases and TNF-alpha.PLoS One. 2012; 7: e39935Crossref PubMed Scopus (150) Google Scholar, 79Schreiber K.L. Brown D.R. Adrenocorticotrophic hormone modulates Escherichia coli O157:H7 adherence to porcine colonic mucosa.Stress. 2005; 8: 185-190Crossref PubMed Scopus (21) Google Scholar, 80Chen C. Lyte M. Stevens M.P. et al.Mucosally-directed adrenergic nerves and sympathomimetic drugs enhance non-intimate adherence of Escherichia coli O157:H7 to porcine cecum and colon.Eur J Pharmacol. 2006; 539: 116-124Crossref PubMed Scopus (48) Google Scholar, 81Schmidt L.D. Xie Y. Lyte M. et al.Autonomic neurotransmitters modulate immunoglobulin A secretion in porcine colonic mucosa.J Neuroimmunol. 2007; 185: 20-28Abstract Full Text Full Text PDF PubMed Scopus (36) Google ScholarIntestinal ischemia/reperfusion injury and repair13Block T. Isaksson H.S. Acosta S. et al.Altered mRNA expression due to acute mesenteric ischaemia in a porcine model.Eur J Vasc Endovasc Surg. 2011; 41: 281-287Abstract Full Text Full Text PDF PubMed Scopus (12) Google Scholar, 14Yandza T. Tauc M. Saint-Paul M.C. et al.The pig as a preclinical model for intestinal ischemia-reperfusion and transplantation studies.J Surg Res. 2012; 178: 807-819Abstract Full Text Full Text PDF PubMed Scopus (28) Google Scholar, 50Blikslager A.T. Moeser A.J. Gookin J.L. et al.Restoration of barrier function in injured intestinal mucosa.Physiol Rev. 2007; 87: 545-564Crossref PubMed Scopus (417) Google Scholar, 54Blikslager A.T. Rhoads J.M. Bristol D.G. et al.Glutamine and transforming growth factor-alpha stimulate extracellular regulated kinases and enhance recovery of villous surface area in porcine ischemic-injured intestine.Surgery. 1999; 125: 186-194Abstract Full Text Full Text PDF PubMed Scopus (50) Google Scholar, 61Blikslager A.T. Roberts M.C. Argenzio R.A. Prostaglandin-induced recovery of barrier function in porcine ileum is triggered by chloride secretion.Am J Physiol. 1999; 276: G28-G36PubMed Google Scholar, 82Tang Z.H. Qiang J.W. Feng X.Y. et al.Acute mesenteric ischemia induced by ligation of porcine superior mesenteric vein: multidetector CT evaluations.Acad Radiol. 2010; 17: 1146-1152Abstract Full Text Full Text PDF PubMed Scopus (10) Google Scholar, 83Bruhn R.S. Distelmaier M.S. Hellmann-Sokolis M. et al.Early detection of acute mesenteric ischemia using diffusion-weighted 3.0-T magnetic resonance imaging in a porcine model.Invest Radiol. 2013; 48: 231-237PubMed Google Scholar, 84Klein H.M. Klosterhalfen B. Kinzel S. et al.CT and MRI of experimentally induced mesenteric ischemia in a porcine model.J Comput Assist Tomogr. 1996; 20: 254-261Crossref PubMed Scopus (26) Google Scholar, 85Jacobi S.K. Moeser A.J. Corl B.A. et al.Dietary long-chain PUFA enhance acute repair of ischemia-injured intestine of suckling pigs.J Nutr. 2012; 142: 1266-1271Crossref PubMed Scopus (34) Google Scholar, 86Acosta S. Nilsson T.K. Malina J. et al.L-lactate after embolization of the superior mesenteric artery.J Surg Res. 2007; 143: 320-328Abstract Full Text Full Text PDF PubMed Scopus (32) Google Scholar, 87Gookin J.L. Galanko J.A. Blikslager A.T. et al.PG-mediated closure of paracellular pathway and not restitution is the primary determinant of barrier recovery in acutely injured porcine ileum.Am J Physiol Gastrointest Liver Physiol. 2003; 285: G967-G979Crossref PubMed Scopus (52) Google ScholarNecrotizing enterocolitis40Azcarate-Peril M.A. Foster D.M. Cadenas M.B. et al.Acute necrotizing enterocolitis of preterm piglets is characterized by dysbiosis of ileal mucosa-associated bacteria.Gut Microbes. 2011; 2: 234-243Crossref PubMed Scopus (15) Google Scholar, 41Sangild P.T. Siggers R.H. Schmidt M. et al.Diet- and colonization-dependent intestinal dysfunction predisposes to necrotizing enterocolitis in preterm pigs.Gastroenterology. 2006; 130: 1776-1792Abstract Full Text Full Text PDF PubMed Scopus (215) Google Scholar, 88Thymann T. Moller H.K. Stoll B. et al.Carbohydrate maldigestion induces necrotizing enterocolitis in preterm pigs.Am J Physiol Gastrointest Liver Physiol. 2009; 297: G1115-G1125Crossref PubMed Scopus (98) Google Scholar, 89Van Haver E.R. Sangild P.T. Oste M. et al.Diet-dependent mucosal colonization and interleukin-1beta responses in preterm pigs susceptible to necrotizing enterocolitis.J Pediatr Gastroenterol Nutr. 2009; 49: 90-98Crossref PubMed Scopus (28) Google Scholar, 90Siggers R.H. Siggers J. Boye M. et al.Early administration of probiotics alters bacterial colonization and limits diet-induced gut dysfunction and severity of necrotizing enterocolitis in preterm pigs.J Nutr. 2008; 138: 1437-1444PubMed Scopus (124) Google Scholar, 91Sangild P.T. Petersen Y.M. Schmidt M. et al.Preterm birth affects the intestinal response to parenteral and enteral nutrition in newborn pigs.J Nutr. 2002; 132: 3786-3794PubMed Google ScholarShort-bowel syndrome92Pereira-Fantini P.M. Thomas S.L. Wilson G. et al.Short- and long-term effects of small bowel resection: a unique histological study in a piglet model of short bowel syndrome.Histochem Cell Biol. 2011; 135: 195-202Crossref PubMed Scopus (30) Google Scholar, 93Vegge A. Thymann T. Lund P. et al.Glucagon-like peptide-2 induces rapid digestive adaptation following intestinal resection in preterm neonates.Am J Physiol Gastrointest Liver Physiol. 2013; 305: G277-G285Crossref PubMed Scopus (44) Google ScholarIntestinal transplantation67Sala F.G. Kunisaki S.M. Ochoa E.R. et al.Tissue-engineered small intestine and stomach from autologous tissue in a preclinical large animal model.J Surg Res. 2009; 156: 205-212Abstract Full Text Full Text PDF PubMed Scopus (134) Google ScholarColorectal cancer94Aigner B. Renner S. Kessler B. et al.Transgenic pigs as models for translational biomedical research.J Mol Med (Berl). 2010; 88: 653-664Crossref PubMed Scopus (193) Google ScholarCystic fibrosis73Rogers C.S. Hao Y. Rokhlina T. et al.Production of CFTR-null and CFTR-DeltaF508 heterozygous pigs by adeno-associated virus-mediated gene targeting and somatic cell nuclear transfer.J Clin Invest. 2008; 118: 1571-1577Crossref PubMed Scopus (272) Google Scholar, 74Keiser N.W. Engelhardt J.F. New animal models of cystic fibrosis: what are they teaching us?.Curr Opin Pulm Med. 2011; 17: 478-483PubMed Google Scholar, 95Fan N. Lai L. Genetically modified pig models for human diseases.J Genet Genomics. 2013; 40: 67-73Crossref PubMed Scopus (75) Google Scholar, 96Whyte J.J. Prather R.S. Genetic modifications of pigs for medicine and agriculture.Mol Reprod Dev. 2011; 78: 879-891Crossref PubMed Scopus (145) Google Scholar, 97Fisher J.T. Zhang Y. Engelhardt J.F. Comparative biology of cystic fibrosis animal models.Methods Mol Biol. 2011; 742: 311-334Crossref PubMed Scopus (64) Google ScholarDiabetes mellitus98Grussner R. Nakhleh R. Grussner A. et al.Streptozotocin-induced diabetes mellitus in pigs.Horm Metab Res. 1993; 25: 199-203Crossref PubMed Scopus (62) Google Scholar, 99Barb C.R. Cox N.M. Carlton C.A. et al.Growth hormone secretion, serum, and cerebral spinal fluid insulin and insulin-like growth factor-I concentrations in pigs with streptozotocin-induced diabetes mellitus.Proc Soc Exp Biol Med. 1992; 201: 223-228Crossref PubMed Scopus (11) Google Scholar Open table in a new tab Despite the numerous advantages of large animal models, several key limitations have impeded their widespread use in biomedical research in favor of rodent models. The most significant limitation to large animal models is the increased cost of animal maintenance and husbandry. Large animal species require larger, more specialized housing and surgical facilities, with higher expenses related to feed, veterinary care, and surgery costs. In addition, their longer reproductive cycles and growth rates make large animal work slower and more expensive. This has hampered the development of transgenic animals. There are many notable similarities between the human and porcine gastrointestinal tracts, which make the porcine model a powerful tool for studying gastrointestinal disease. For example, the esophagus is very similar to that of human beings in that both species have esophageal submucosal glands as do human beings, whereas rodents do not.24Garman K.S. Orlando R.C. Chen X. Review: experimental models for Barrett's esophagus and esophageal adenocarcinoma.Am J Physiol Gastrointest Liver Physiol. 2012; 302: G1231-G1243Crossref Scopus (25) Google Scholar The stomach of the pig is entirely glandular, making it physiologically comparable with that of human beings.11Kararli T.T. Comparison of the gastrointestinal anatomy, physiology, and biochemistry of humans and commonly used laboratory animals.Biopharm Drug Dispos. 1995; 16: 351-380Crossref PubMed Scopus (1117) Google Scholar The structure of the small intestine is comparable in human beings and pigs, and the intestinal length (meters) per bodyweight (kilograms) ratio is approximately 0.1 in both species, compared with approximately 0.16 in mice.10Patterson J.K. Lei X.G. Miller D.D. The pig as an experimental model for elucidating the mechanisms governing dietary influence on mineral absorption.Exp Biol Med (Maywood). 2008; 233: 651-664Crossref PubMed Scopus (193) Google Scholar, 25Ranjay Chakraborty H.P. Tan C.C. Prunty M. et al.Contribution of body length on axial length during normal eye development in C57BL/6J and 129S1/SvJ wild-type mouse strains.Invest Ophthalmol Vis Sci. 2014; 55: 3614Google Scholar, 26Casteleyn C. Rekecki A. Van der Aa A. et al.Surface area assessment of the murine intestinal tract as a prerequisite for oral dose translation from mouse to man.Lab Anim. 2010; 44: 176-183Crossref Scopus (55) Google Scholar The epithelial cell population (cell lineages, phenotypes, and expression of distinct protein biomarkers) of the porcine small intestine is similar to that of human beings.27Gonzalez L.M. Williamson I. Piedrahita J.A. et al.Cell lineage identification and stem cell culture in a porcine model for the study of intestinal epithelial regeneration.PLoS One. 2013; 8: e66465Crossref PubMed Scopus (94) Google Scholar The villus structure is finger-like in pigs, mice, and human beings, whereas rats have a leaf-like villus structure.28Chen Y.M. Zhang J.S. Duan X.L. Changes of microvascular architecture, ultrastructure and permeability of rat jejunal villi at different ages.World J Gastroenterol. 2003; 9: 795-799Crossref PubMed Scopus (25) Google Scholar The subcellular structure of porcine enterocytes within the crypt base have been characterized and found to be similar to the description of these cells in human beings.23Gonzalez L.M. Moeser A.J. Blikslager A.T. Porcine models of digestive disease: the future of large animal translational research.Transl Res. 2015; 166: 12-27Abstract Full Text Full Text PDF Scopus (118) Google Scholar, 29Barker N. van Oudenaarden A. Clevers H. Identifying the stem cell of the intestinal crypt: strategies and pitfalls.Cell Stem Cell. 2012; 11: 452-460Abstract Full Text Full Text PDF PubMed Scopus (237) Google Scholar The colon of the pig and human beings both possess sacculations and longitudinal muscular bands (tenia) along their length, which results in similar transit times and thus comparable digestive physiology in the intestine, whereas the colon of the mouse and rat are nonsacculated.30Graham H. Aman P. The pig as a model in dietary fibre digestion studies.Scand J Gastroenterol Suppl. 1987; 129: 55-61Crossref PubMed Scopus (45) Google Scholar, 31Nguyen T.L.A. Vieira-Silva S. Liston A. et al.How informative is the mouse for human gut microbiota research?.Dis Model Mech. 2015; 8: 1-16Crossref PubMed Scopus (736) Google Scholar Pigs and human beings are capable of fermenting digesta within the colon, and have been shown to have similar microbial flora within the small intestine and large intestine.32Miller E.R. Ullrey D.E. The pig as a model for human nutrition.Annu Rev Nutr. 1987; 7: 361-382Crossref PubMed Scopus (539) Google Scholar, 33Pang X. Hua X. Yang Q. et al.Inter-species transplantation of gut microbiota from human to pigs.ISME J. 2007; 1: 156-162Crossref PubMed Scopus (130) Google Scholar Conversely, the microbiota of the mouse and human are significantly divergent, with approximately 85% of mouse gut microbiota being absent from human flora.34Ley R.E. Backhed F. Turnbaugh P. et al.Obesity alters gut microbial ecology.Proc Natl Acad Sci U S A. 2005; 102: 11070-11075Crossref PubMed Scopus (4363) Google Scholar Nonetheless, there are important differences in the pig as compared with human beings, including the absence of an appendix, a much larger and more developed cecum, and the spiral arrangement of the colon in the pig (Figure 1).11Kararli T.T. Comparison of the gastrointestinal anatomy, physiology, and biochemistry of humans and commonly used laboratory animals.Biopharm Drug Dispos. 1995; 16: 351-380Crossref PubMed Scopus (1117) Google Scholar, 16Swindle M.M. Makin A. Herron A.J. et al.Swine as models in biomedical research and toxicology testing.Vet Pathol. 2012; 49: 344-356Crossref PubMed Scopus (868) Google Scholar The dog and pig have been used extensively to study esophageal disease, with similarities in disease pathophysiology more similar to human beings than that of rodents.35Abdulnour-Nakhoul S. Nakhoul N.L. Wheeler S.A. et al.Characterization of esophageal submucosal glands in pig tissue and cultures.Dig Dis Sci. 2007; 52: 3054-3065Crossref PubMed Scopus (21) Google Scholar, 36Long J.D. Orlando R.C. Esophageal submucosal glands: structure and function.Am J Gastroenterol. 1999; 94: 2818-2824Crossref PubMed Scopus (99) Google Scholar For instance, in a chronic model of acid reflux disease, the dog develops a phenotype of esophageal adenocarcinoma that is similar to the glandular phenotype seen in human beings as opposed to the mucinous phenotype of mice and rats.37Kawaura Y. Tatsuzawa Y. Wakabayashi T. et al.Immunohistochemical study of p53, c-erbB-2, and PCNA in Barrett's esophagus with dysplasia and adenocarcinoma arising from experimental acid or alkaline reflux model.J Gastroenterol. 2001; 36: 595-600Crossref PubMed Scopus (27) Google Scholar In pigs, the esophageal and gastroesophageal junctional anatomy and physiology are comparable with that of human beings, both sharing the existence of large, dissectible submucosal glands, making the pig a preferred model for analogous clinical applications.11Kararli T.T. Comparison of the gastrointestinal anatomy, physiology, and biochemistry of humans and commonly used laboratory animals.Biopharm Drug Dispos. 1995; 16: 351-380Crossref PubMed Scopus (1117) Google Scholar, 24Garman K.S. Orlando R.C. Chen X. Review: experimental models for Barrett's esophagus and esophageal adenocarcinoma.Am J Physiol Gastrointest Liver Physiol. 2012; 302: G1231-G1243Crossref Scopus (25) Google Scholar, 38Kapoor H. Lohani K.R. Lee T.H. et al.Animal models of Barrett's esophagus and esophageal adenocarcinoma-past, present, and future.Clin Transl Sci. 2015; 8: 841-847Crossref Scopus (36) Google Scholar In particular, pigs are used commonly to test new surgical and endoscopic techniques in the esophagus. Recently, a pig model of esophageal injury and repair was developed in which endoscopic radiofrequency ablation was used to induce injury, followed by the study of cellular biology of the repair process, including the location and role of proliferative cells.24Garman K.S. Orlando R.C. Chen X. Review: experimental models for Barrett's esophagus and esophageal adenocarcinoma.Am J Physiol Gastrointest Liver Physiol. 2012; 302: G1231-G1243Crossref Scopus (25) Google Scholar This model promises}, number={6}, journal={Cellular and Molecular Gastroenterology and Hepatology}, publisher={Elsevier BV}, author={Ziegler, Amanda and Gonzalez, Liara and Blikslager, Anthony}, year={2016}, month={Nov}, pages={716–724} } @article{kruger_gonzalez_von furstenberg_henning_blikslager_garman_2016, title={Mo1253 Ductular and Proliferative Response of Esophageal Submucosal Glands in a Porcine Model of Esophageal Injury and Repair}, volume={150}, ISSN={0016-5085}, url={http://dx.doi.org/10.1016/S0016-5085(16)32312-5}, DOI={10.1016/S0016-5085(16)32312-5}, number={4}, journal={Gastroenterology}, publisher={Elsevier BV}, author={Kruger, Leandi and Gonzalez, Liara M. and von Furstenberg, Richard J. and Henning, Susan J. and Blikslager, Anthony and Garman, Katherine S.}, year={2016}, month={Apr}, pages={S679–S680} } @article{stewart_freund_magness_lund_blikslager_gonzalez_2016, title={Mo1298 Critical Contribution of Intestinal Stem Cells in the Repair of Ischemia Reperfusion Injury}, volume={150}, ISSN={0016-5085}, url={http://dx.doi.org/10.1016/S0016-5085(16)32355-1}, DOI={10.1016/S0016-5085(16)32355-1}, number={4}, journal={Gastroenterology}, publisher={Elsevier BV}, author={Stewart, Amy S. and Freund, John and Magness, Scott and Lund, Pauline K. and Blikslager, Anthony and Gonzalez, Liara M.}, year={2016}, month={Apr}, pages={S691} } @article{gonzalez_moeser_blikslager_2015, title={Animal models of ischemia-reperfusion-induced intestinal injury: progress and promise for translational research}, volume={308}, ISSN={0193-1857 1522-1547}, url={http://dx.doi.org/10.1152/ajpgi.00112.2013}, DOI={10.1152/ajpgi.00112.2013}, abstractNote={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.}, number={2}, journal={American Journal of Physiology-Gastrointestinal and Liver Physiology}, publisher={American Physiological Society}, author={Gonzalez, Liara M. and Moeser, Adam J. and Blikslager, Anthony T.}, year={2015}, month={Jan}, pages={G63–G75} } @article{gonzalez_kinnin_blikslager_2015, title={Characterization of discrete equine intestinal epithelial cell lineages}, volume={76}, ISSN={0002-9645}, url={http://dx.doi.org/10.2460/ajvr.76.4.358}, DOI={10.2460/ajvr.76.4.358}, abstractNote={Abstract}, number={4}, journal={American Journal of Veterinary Research}, publisher={American Veterinary Medical Association (AVMA)}, author={Gonzalez, Liara M. and Kinnin, Leslie A. and Blikslager, Anthony T.}, year={2015}, month={Apr}, pages={358–366} } @article{gonzalez_moeser_blikslager_2015, title={Porcine models of digestive disease: the future of large animal translational research}, volume={166}, ISSN={1931-5244}, url={http://dx.doi.org/10.1016/j.trsl.2015.01.004}, DOI={10.1016/j.trsl.2015.01.004}, abstractNote={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.}, number={1}, journal={Translational Research}, publisher={Elsevier BV}, author={Gonzalez, Liara M. and Moeser, Adam J. and Blikslager, Anthony T.}, year={2015}, month={Jul}, pages={12–27} } @article{gonzalez_2015, title={The mother of a gut cell: Intestinal epithelial stem cells}, volume={27}, ISSN={["2042-3292"]}, DOI={10.1111/eve.12456}, abstractNote={Equine Veterinary EducationVolume 27, Issue 11 p. 559-560 Editorial The mother of a gut cell: Intestinal epithelial stem cells L. M. Gonzalez, L. M. Gonzalez Clinical Sciences, North Carolina State University, Raleigh, USASearch for more papers by this author L. M. Gonzalez, L. M. Gonzalez Clinical Sciences, North Carolina State University, Raleigh, USASearch for more papers by this author First published: 13 October 2015 https://doi.org/10.1111/eve.12456Citations: 6Read the full textAboutPDF ToolsExport citationAdd to favoritesTrack citation ShareShare Give accessShare full text accessShare full-text accessPlease review our Terms and Conditions of Use and check box below to share full-text version of article.I have read and accept the Wiley Online Library Terms and Conditions of UseShareable LinkUse the link below to share a full-text version of this article with your friends and colleagues. Learn more.Copy URL Share a linkShare onEmailFacebookTwitterLinkedInRedditWechat No abstract is available for this article. References Barker, N., van Es, J.H., Kuipers, J., Kujala, P., van den Born, M., Cozijnsen, M., Haegebarth, A., Korving, J., Begthel, H., Peters, P.J. and Clevers, H. (2007) Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 449, 1003-1007. Cheng, H. and Leblond, C.P. (1974) Origin, differentiation and renewal of the four main epithelial cell types in the mouse small intestine. V. Tnitarian theory of the origin of the four epithelial cell types. Am. J. Anat. 141, 537-561. Dekaney, C.M., Fong, J.J., Rigby, R.J., Lund, P.K., Henning, S.J. and Helmrath, M.A. (2007) Expansion of intestinal stem cells associated with long-term adaptation following ileocecal resection in mice. Am. J. Physiol. Gastrointest. Liver Physiol. 293, G1013-G1022. Dekaney, C.M., Gulati, A.S., Garrison, A.P., Helmrath, M.A. and Henning, S.J. (2009) Regeneration of intestinal stem/progenitor cells following doxorubicin treatment of mice. Am. J. Physiol. Gastrointest. Liver Physiol. 297, G461-G470. van der Flier, L.G., Haegebarth, A., Stange, D.E., van de Wetering, M. and Clevers, H. (2009a) OLFM4 is a robust marker for stem cells in human intestine and marks a subset of colorectal cancer cells. Gastroenterology 137, 15-17. van der Flier, L.G., van Gijn, M.E., Hatzis, P., Kujala, P., Haegebarth, A., Stange, D.E., Begthel, H., van den Born, M., Guryev, V., Oving, I., van Es, J.H., Barker, N., Peters, P.J., van de Wetering, M. and Clevers, H. (2009b) Transcription factor achaete scute-like 2 controls intestinal stem cell fate. Cell 136, 903-912. Formeister, E.J., Sionas, A.L., Lorance, D.K., Barkley, C.L., Lee, G.H. and Magness, S.T. (2009) Distinct SOX9 levels differentially mark stem/progenitor populations and enteroendocrine cells of the small intestine epithelium. Am. J. Physiol. Gastrointest. Liver Physiol. 296, G1108-G1118. Gonzalez, L.M., Stranahan, L.W. and Blikslager, A.T. (2014) The proliferative pool of stem cells are decreased by large colon volvulus in horses. In: The 11th International Equine Colic Research Symposium, Dublin, Ireland. pp 128. Hua, G., Thin, T.H., Feldman, R., Haimovitz-Friedman, A., Clevers, H., Fuks, Z. and Kolesnick, R. (2012) Crypt base columnar stem cells in small intestines of mice are radioresistant. Gastroenterology 143, 1266-1276. Montgomery, R.K., Carlone, D.L., Richmond, C.A., Farilla, L., Kranendonk, M.E., Henderson, D.E., Baffour-Awuah, N.Y., Ambruzs, A.M., Fogli, L.K., Algra, S. and Breault, D.T. (2011) Mouse telomerase reverse transcriptase (mTert) expression marks slowly cycling intestinal stem cells. Proc. Natl Acad. Sci. USA 108, 179-184. Powell, A.E., Wang, Y., Li, Y., Poulin, E.J., Means, A.L., Washington, M.K., Higginbotham, J.N., Juchheim, A., Prasad, N., Levy, S.E., Guo, Y., Shyr, Y., Aronow, B.J., Haigis, K.M., Franklin, J.L. and Coffey, R.J. (2012) The pan-ErbB negative regulator Lrig1 is an intestinal stem cell marker that functions as a tumor suppressor. Cell 149, 146-158. Takeda, N., Jain, R., LeBoeuf, M.R., Wang, Q., Lu, M.M. and Epstein, J.A. (2011) Interconversion between intestinal stem cell populations in distinct niches. Science 334, 1420-1424. USDA:APHIS:VS Centers for Epidemiology and Animal Health (1998) Part I: Baseline Reference of 1998 Equine Health and Management. United States Department of Agriculture, Washington DC. Van Hoogmoed, L., Snyder, J.R., Pascoe, J.R. and Olander, H. (2000) Use of pelvic flexure biopsies to predict survival after large colon torsion in horses. Vet. Surg. 29, 572-577. Van Landeghem, L., Santoro, M.A., Krebs, A.E., Mah, A.T., Dehmer, J.J., Gracz, A.D., Scull, B.P., McNaughton, K., Magness, S.T. and Lund, P.K. (2012) Activation of two distinct Sox9-EGFP-expressing intestinal stem cell populations during crypt regeneration after irradiation. Am. J. Physiol. Gastrointest. Liver Physiol. 302, G1111-G1132. Yan, K.S., Chia, L.A., Li, X., Ootani, A., Su, J., Lee, J.Y., Su, N., Luo, Y., Hilshorn, S.C., Amieva, M.R., Sangiorgi, E., Capecchi, M.R. and Kuo, C.J. (2012) The intestinal stem cell markers Bmi1 and Lgr5 identify two functionally distinct populations. Proc. Natl Acad. Sci. USA 109, 466-471. Citing Literature Volume27, Issue11November 2015Pages 559-560 ReferencesRelatedInformation}, number={11}, journal={EQUINE VETERINARY EDUCATION}, author={Gonzalez, L. M.}, year={2015}, month={Nov}, pages={559–560} } @article{gonzalez_magness_lund_blikslager_2014, title={Mo1960 Intestinal Epithelial Cells Expressing Biomarkers of Crypt Base Columnar or Reserve Stem Cells Show Differential Resistance to Ischemia-Reperfusion Injury}, volume={146}, ISSN={0016-5085}, url={http://dx.doi.org/10.1016/S0016-5085(14)62550-6}, DOI={10.1016/S0016-5085(14)62550-6}, number={5}, journal={Gastroenterology}, publisher={Elsevier BV}, author={Gonzalez, Liara M. and Magness, Scott T. and Lund, Pauline K. and Blikslager, Anthony}, year={2014}, month={May}, pages={S-702} } @article{gonzalez_fogle_baker_hughes_law_motsinger-reif_blikslager_2014, title={Operative factors associated with short-term outcome in horses with large colon volvulus: 47 cases from 2006 to 2013}, volume={47}, ISSN={0425-1644}, url={http://dx.doi.org/10.1111/evj.12273}, DOI={10.1111/evj.12273}, abstractNote={Summary}, number={3}, journal={Equine Veterinary Journal}, publisher={Wiley}, author={Gonzalez, L. M. and Fogle, C. A. and Baker, W. T. and Hughes, F. E. and Law, J. M. and Motsinger-Reif, A. A. and Blikslager, A. T.}, year={2014}, month={May}, pages={279–284} } @article{gonzalez_williamson_piedrahita_blikslager_magness_2013, title={Cell lineage identification and stem cell culture in a porcine model for the study of intestinal epithelial regeneration}, volume={8}, number={6}, journal={PLoS One}, author={Gonzalez, L. M. and Williamson, I. and Piedrahita, J. A. and Blikslager, A. T. and Magness, S. T.}, year={2013} } @article{gonzalez_blikslager_piedrahita_magness_2013, title={Su1099 A Translational Porcine Model of Intestinal Stem Cells}, volume={144}, ISSN={0016-5085}, url={http://dx.doi.org/10.1016/S0016-5085(13)61466-3}, DOI={10.1016/S0016-5085(13)61466-3}, number={5}, journal={Gastroenterology}, publisher={Elsevier BV}, author={Gonzalez, Liara and Blikslager, Anthony and Piedrahita, Jorge A. and Magness, Scott T.}, year={2013}, month={May}, pages={S-398} } @article{gonzalez_schramme_robertson_thrall_redding_2010, title={MRI FEATURES OF METACARPO(TARSO)PHALANGEAL REGION LAMENESS IN 40 HORSES}, volume={51}, ISSN={1058-8183 1740-8261}, url={http://dx.doi.org/10.1111/j.1740-8261.2010.01676.x}, DOI={10.1111/j.1740-8261.2010.01676.x}, abstractNote={Lameness originating from the metacarpo(tarso)phalangeal (MP) joint has a significant effect on the use and athletic competitiveness of a horse. The identification of the cause of lameness originating from the MP joint can be challenging, given the limitations of radiography, ultrasonography, and nuclear scintigraphy. Our purpose was to describe the injury types and incidence in magnetic resonance imaging (MRI) studies from 40 horses with lameness attributable to the MP joint region where it was not possible to reach a clinically plausible diagnosis using other imaging modalities. Horses were examined in a 1.5 T magnet (Siemens Medical Solutions) under general anesthesia. The frequency of occurrence of MR lesions was subchondral bone injury (19), straight or oblique distal sesamoidean desmitis (13), articular cartilage injury and osteoarthritis (eight), suspensory branch desmitis (seven), osteochondral fragmentation (seven), proximal sesamoid bone injury (seven), inter-sesamoidean desmitis (four), deep digital flexor tendonitis (four), collateral desmitis (three), superficial digital flexor tendonitis (two), enostosis-like lesions of the proximal phalanx or MCIII (two), desmitis of the palmar annular ligament (one), desmitis of the proximal digital annular ligament (one), and dystrophic calcification of the lateral digital extensor tendon (one). Twenty-five horses had multiple MR abnormalities. MRI provided information that was complementary to radiography, ultrasonography, and nuclear scintigraphy and that allowed for a comprehensive evaluation of all structures in the MP joint region and a diagnosis in all 40 horses.}, number={4}, journal={Veterinary Radiology & Ultrasound}, publisher={Wiley}, author={Gonzalez, Liara M. and Schramme, Michael C. and Robertson, Ian D. and Thrall, Donald E. and Redding, Rich W.}, year={2010}, month={Apr}, pages={404–414} }