@article{ziegler_caldwell_craig_hellstrom_sheridan_touvron_pridgen_magness_odle_landeghem_et al._2024, title={Enteric glial cell network function is required for epithelial barrier restitution following intestinal ischemic injury in the early postnatal period}, url={https://doi.org/10.1152/ajpgi.00216.2022}, DOI={10.1152/ajpgi.00216.2022}, abstractNote={Ischemic damage to the intestinal epithelial barrier, such as in necrotizing enterocolitis or small intestinal volvulus, is associated with higher mortality rates in younger patients. We have recently reported a powerful pig model to investigate these age-dependent outcomes in which mucosal barrier restitution is strikingly absent in neonates but can be rescued by direct application of homogenized mucosa from older, juvenile pigs by a yet-undefined mechanism. Within the mucosa, a postnatally developing network of enteric glial cells (EGC) is gaining recognition as a key regulator of the mucosal barrier. Therefore, we hypothesized that the developing EGC network may play an important role in coordinating intestinal barrier repair in neonates. Neonatal and juvenile jejunal mucosa recovering from surgically induced intestinal ischemia was visualized by scanning electron microscopy and the transcriptomic phenotypes were assessed by bulk RNA sequencing. EGC network density and glial activity were examined by gene set enrichment analysis, three-dimensional volume imaging and western blot and its function in regulating epithelial restitution assessed ex vivo in Ussing chamber using the glia-specific inhibitor fluoroacetate, and in vitro by co-culture assay. Here we refine and elaborate our translational model, confirming a neonatal phenotype characterized by a complete lack of coordinated reparative signaling in the mucosal microenvironment. Further, we report important evidence that the subepithelial EGC network changes significantly over the early postnatal period and demonstrate that proximity of a specific functional population of EGC to wounded intestinal epithelium contributes to intestinal barrier restitution following ischemic injury.}, journal={American Journal of Physiology-Gastrointestinal and Liver Physiology}, author={Ziegler, Amanda L. and Caldwell, Madison L. and Craig, Sara E. and Hellstrom, Emily A. and Sheridan, Anastasia E. and Touvron, Melissa S. and Pridgen, Tiffany A. and Magness, Scott T. and Odle, Jack and Landeghem, Laurianne Van and et al.}, year={2024}, month={Mar} } @article{boston_wang_xi_kim_fellner_scott_ziegler_van landeghem_blikslager_odle_2023, title={Effects of Prebiotic Galactooligosaccharide (GOS) on Hindgut Microbial Composition of Pigs During the Peri-Weaning Period}, volume={101}, ISSN={["1525-3163"]}, DOI={10.1093/jas/skad281.228}, abstractNote={ Galactooligosaccharide (GOS) is a prebiotic containing beta-linked galactose oligomers of 2 to 8 units. Previously, GOS positively impacted post-weaning growth performance and altered jejunal morphology. In this study, GOS-enriched whey permeate (Milk Specialties Global, Eden Prairie MN) was supplemented to piglets in farrowing and nursery phases. To maximize pre-weaning GOS intake, novel gruel creep feeders were used. Piglets from 24 litters received either no creep feed (NC), creep without GOS (CG-) or creep with 5.0% GOS (CG+) followed by a phase 1 nursery diet without (NG-) or with 3.8% GOS (NG+). At d 22 (pre-weaning) and d 31 (post-weaning), 6 pigs per treatment were euthanized for sample collection. DNA was extracted from cecal and rectal swabs for downstream 16S rRNA sequencing via Illumina MiSeq (Zymo Research Corp, Irvine CA). An amplicon sequence variant (ASV) table was created using the DADA2 pipeline in R and taxonomy assignment was performed using the SILVA database. Average sequencing depth after filtering was 138,323 and low sequencing counts were discarded based on 20% prevalence. Data were analyzed using the lmer and adonis procedures in RStudio ver. 4.2.2. Analysis of 16S rRNA genes did not detect pre- or post-weaning GOS effects on Chao1, Simpson, or Shannon alpha diversity measures (P > 0.1). Similarly, no pre-weaning GOS effects were detected in Principal Coordinates Analysis (PCoA) beta diversity plots (P > 0.1); however, pigs fed GOS post-weaning segregated from controls (P < 0.01). The relative abundance of several genera including Fusicatenbacter and Collinsella increased by 2.0 and 1.3-fold, respectively, in the cecum of NG+ pigs, whereas, Bacteroides decreased by 3.8-fold (FDR, P < 0.05). In the rectum, relative abundance of Corprococcus decreased by 2.0-fold and Fusicatenbacter increased by 1.5-fold in NG+ pigs compared with NG- pigs (FDR, P < 0.05). Collectively, these genera in the cecum and rectum comprise of less than 3% of the total community. In contrast, lactobacillus in the cecum (33%) and rectum (21%) had a large relative abundance but was unaffected by treatment. We conclude that supplementation of GOS in the post-weaning phase 1 nursery diet alters the hindgut microbial community and may contribute to improvements in growth performance.}, journal={JOURNAL OF ANIMAL SCIENCE}, author={Boston, Timothy E. and Wang, Feng and Xi, Lin and Kim, Sung Woo and Fellner, Vivek and Scott, Mark F. and Ziegler, Amanda L. and Van Landeghem, Laurianne and Blikslager, Anthony T. and Odle, Jack}, year={2023}, month={Nov}, pages={187–188} } @article{boston_wang_xi_kim_fellner_scott_ziegler_van landeghem_blikslager_odle_2023, title={Effects of Prebiotic Galactooligosaccharide (Gos) on Piglet Growth and Jejunal Morphology During the Peri-Weaning Period}, volume={101}, ISSN={["1525-3163"]}, DOI={10.1093/jas/skad341.313}, abstractNote={ Galactooligosaccharide (GOS) is a specific prebiotic that is enzymatically synthesized from lactose to form beta-linked oligosaccharides containing 2-8 galactose units. In this study, GOS-enriched whey permeate (Milk Specialties Global, Eden Prairie MN) was supplemented to piglets in lactation and nursery phases. To maximize pre-weaning GOS intake, novel gruel creep feeders were utilized. Using a 3x2 factorial design, piglets from 24 litters received either no creep feed (NC), creep without GOS (CG-) or creep with 5% GOS (CG+) followed by a phase 1 nursery diet without (NG-) or with 3.8% GOS (NG+). After 1 week, pigs were fed common phase 2 and phase 3 diets. At d 22 (pre-weaning) and d 31 (post-weaning), 6 pigs per treatment were euthanized for intestinal measurements. Pre-weaning, creep fed pigs grew 19% faster than controls (P < 0.01) but GOS effects were not detected (P > 0.1). In contrast, pigs fed GOS in phase 1 grew 34 % faster than controls (P < 0.04), irrespective of creep treatment (interaction P > 0.1), and with corresponding greater intakes (P < 0.06). These GOS effects were sustained for overall nursery performance. Furthermore, overall ADG of CG+ piglets in the nursery tended to be greatest (P = 0.09), gaining 361g/d, followed by NC (324 g/d) and CG- (310 g/d) treatments (Table 1). No effects on jejunal morphology were detected at d 22, although there was an effect of age with decreased villi length, villus area, villi:crypt ratio and increased crypt depth at d 31 (P < 0.01). Supplementation of GOS in phase 1 increased villus length (36%) and area (51%) but only in pigs previously fed the control creep (CG-) diet (interaction, P < 0.01). Treatment effects on cecal pH and VFA concentrations were not detected, although there was an effect of age with a decrease in pH (P < 0.01) and increase in propionate and butyrate concentrations post-weaning (P < 0.01). We conclude that gruel creep feeding increases weight gain regardless of GOS treatment and that nursery growth and intestinal morphology are improved by post-weaning GOS supplementation. Funded in part by Milk Specialties Global, USDA-NIFA 2022-67015-37125 and Hatch 1016618. }, journal={JOURNAL OF ANIMAL SCIENCE}, author={Boston, Timothy E. and Wang, Feng and Xi, Lin and Kim, Sung Woo and Fellner, Vivek and Scott, Mark F. and Ziegler, Amanda L. and Van Landeghem, Laurianne and Blikslager, Anthony T. and Odle, Jack}, year={2023}, month={Oct}, pages={276–277} } @article{rivera_bilton_burclaff_czerwinski_liu_trueblood_hinesley_breau_deal_joshi_et al._2023, title={Hypoxia Primes Human ISCs for Interleukin-Dependent Rescue of Stem Cell Activity}, volume={16}, ISSN={["2352-345X"]}, DOI={10.1016/j.jcmgh.2023.07.012}, abstractNote={Hypoxia in the intestinal epithelium can be caused by acute ischemic events or chronic inflammation in which immune cell infiltration produces inflammatory hypoxia starving the mucosa of oxygen. The epithelium has the capacity to regenerate after some ischemic and inflammatory conditions suggesting that intestinal stem cells (ISCs) are highly tolerant to acute and chronic hypoxia; however, the impact of hypoxia on human ISC (hISC) function has not been reported. Here we present a new microphysiological system (MPS) to investigate how hypoxia affects hISCs from healthy donors and test the hypothesis that prolonged hypoxia modulates how hISCs respond to inflammation-associated interleukins (ILs).hISCs were exposed to <1.0% oxygen in the MPS for 6, 24, 48, and 72 hours. Viability, hypoxia-inducible factor 1a (HIF1a) response, transcriptomics, cell cycle dynamics, and response to cytokines were evaluated in hISCs under hypoxia. HIF stabilizers and inhibitors were screened to evaluate HIF-dependent responses.The MPS enables precise, real-time control and monitoring of oxygen levels at the cell surface. Under hypoxia, hISCs maintain viability until 72 hours and exhibit peak HIF1a at 24 hours. hISC activity was reduced at 24 hours but recovered at 48 hours. Hypoxia induced increases in the proportion of hISCs in G1 and expression changes in 16 IL receptors. Prolyl hydroxylase inhibition failed to reproduce hypoxia-dependent IL-receptor expression patterns. hISC activity increased when treated IL1β, IL2, IL4, IL6, IL10, IL13, and IL25 and rescued hISC activity caused by 24 hours of hypoxia.Hypoxia pushes hISCs into a dormant but reversible proliferative state and primes hISCs to respond to a subset of ILs that preserves hISC activity. These findings have important implications for understanding intestinal epithelial regeneration mechanisms caused by inflammatory hypoxia.}, number={5}, journal={CELLULAR AND MOLECULAR GASTROENTEROLOGY AND HEPATOLOGY}, author={Rivera, Kristina R. and Bilton, R. Jarrett and Burclaff, Joseph and Czerwinski, Michael J. and Liu, Jintong and Trueblood, Jessica M. and Hinesley, Caroline M. and Breau, Keith A. and Deal, Halston E. and Joshi, Shlok and et al.}, year={2023}, pages={823–846} } @article{ziegler_erwin_caldwell_touvron_pridgen_magness_odle_landeghem_blikslager_2022, title={Enteric Glial Cell Network Function is Required for Epithelial Barrier Restitution following Intestinal Ischemic Injury in the Early Postnatal Period}, url={https://doi.org/10.1101/2022.11.04.514575}, DOI={10.1101/2022.11.04.514575}, abstractNote={Ischemic damage to the intestinal epithelial barrier, such as in necrotizing enterocolitis or small intestinal volvulus, is associated with higher mortality rates in younger patients. We have recently reported a powerful pig model to investigate these age-dependent outcomes in which mucosal barrier restitution is strikingly absent in neonates but can be rescued by direct application of homogenized mucosa from older, juvenile pigs by a yet-undefined mechanism. Within the mucosa, a postnatally developing network of enteric glial cells (EGC) is gaining recognition as a key regulator of the mucosal barrier. Therefore, we hypothesized that the developing EGC network may play an important role in coordinating intestinal barrier repair in neonates. Neonatal and juvenile jejunal mucosa recovering from surgically induced intestinal ischemia was visualized by scanning electron microscopy and the transcriptomic phenotypes were assessed by bulk RNA sequencing. EGC network density and gliosis were examined by gene set enrichment analysis, three-dimensional volume imaging and western blot and its function in regulating epithelial restitution assessed ex vivo in Ussing chamber using the glia-specific inhibitor fluoroacetate, and in vivo by co-culture assay. Here we refine and elaborate our translational model, confirming a neonatal phenotype characterized by a complete lack of coordinated reparative signaling in the mucosal microenvironment. Further, we report important evidence that the subepithelial EGC network changes significantly over the early postnatal period and demonstrate that EGC function in close proximity to wounded intestinal epithelium is critical to intestinal barrier restitution following ischemic injury. NEW & NOTEWORTHY This study refines a powerful translational pig model, defining an age-dependent relationship between enteric glia and the intestinal epithelium during intestinal ischemic injury and confirming an important role of the enteric glial cell activity in driving mucosal barrier restitution. This study suggests that targeting the enteric glial network could lead to novel interventions to improve recovery from intestinal injury in neonatal patients.}, author={Ziegler, Amanda L. and Erwin, Sara and Caldwell, Madison L. and Touvron, Melissa S. and Pridgen, Tiffany A. and Magness, Scott T. and Odle, Jack and Landeghem, Laurianne Van and Blikslager, Anthony T.}, year={2022}, month={Nov} } @article{boger_sheridan_ziegler_blikslager_2022, title={Mechanisms and modeling of wound repair in the intestinal epithelium}, volume={6}, ISSN={["2168-8370"]}, url={https://doi.org/10.1080/21688370.2022.2087454}, DOI={10.1080/21688370.2022.2087454}, abstractNote={ABSTRACT The intestinal epithelial barrier is susceptible to injury from insults, such as ischemia or infectious disease. The epithelium’s ability to repair wounded regions is critical to maintaining barrier integrity. Mechanisms of intestinal epithelial repair can be studied with models that recapitulate the in vivo environment. This review focuses on in vitro injury models and intestinal cell lines utilized in such systems. The formation of artificial wounds in a controlled environment allows for the exploration of reparative physiology in cell lines modeling diverse aspects of intestinal physiology. Specifically, the use of intestinal cell lines, IPEC-J2, Caco-2, T-84, HT-29, and IEC-6, to model intestinal epithelium is discussed. Understanding the unique systems available for creating intestinal injury and the differences in monolayers used for in vitro work is essential for designing studies that properly capture relevant physiology for the study of intestinal wound repair.}, journal={TISSUE BARRIERS}, author={Boger, Kasey D. and Sheridan, Ana E. and Ziegler, Amanda L. and Blikslager, Anthony T.}, year={2022}, month={Jun} } @article{erwin_clark_dechant_aitken_hassel_blikslager_ziegler_2022, title={Multi-Institutional Retrospective Case-Control Study Evaluating Clinical Outcomes of Foals with Small Intestinal Strangulating Obstruction: 2000-2020}, volume={12}, ISSN={["2076-2615"]}, url={https://doi.org/10.3390/ani12111374}, DOI={10.3390/ani12111374}, abstractNote={Simple Summary Lower survival rates have been reported in foals than adults with severe colic lesions obstructing blood flow to the small intestine, but this has not been compared directly. These survival rates are important to horse owners making medical decisions surrounding colic, for both the foal’s wellbeing and the owner’s finances. In this retrospective case-control study, hospital records of surgical colic cases were collected from five US academic referral hospitals to directly compare foal and adult survival following surgery for specific colic lesions. It was hypothesized that foals would exhibit lower survival than case-matched adults. This study was limited by incomplete medical and surgical records, relatively small sample size, and lack of long-term follow-up. Short-term survival in foals was not significantly different than in adults with comparable colic lesions and may have been partly driven by decision-making on the farm prior to referral. More optimism toward surgical treatment of foals with suspected SISO may be warranted. Abstract Lower survival has been reported in foals than adults with small intestinal strangulating obstruction (SISO), but age-dependent outcomes have not been examined directly. Hospital records were collected from five US academic referral hospitals. It was hypothesized that foals would exhibit lower survival than case-matched adults. Foal cases 6-months-of-age or younger, and adult cases between 2- and 20-years-of-age were collected. Data revealed 24 of 25 (96.0%) foals and 66 of 75 (88.0%) adults that were recovered from surgery for SISO survived to hospital discharge. Sixteen of the total 41 (39.0%) foals studied were euthanized intraoperatively, whereas 30 of 105 (28.6%) adults were euthanized intraoperatively. Common lesions in foals that were recovered from surgery were volvulus (n = 13) and intussusception (n = 5), whereas common lesions in adults were volvulus (n = 25) and strangulating lipoma (n = 23). This study was limited by incomplete medical records, relatively small sample size, and lack of long-term follow-up. Unexpectedly, short-term survival tended to be higher in foals than adults and may have been partly driven by case selection prior to referral or surgery or decision-making intraoperatively. More optimism toward surgical treatment of foals with SISO may be warranted.}, number={11}, journal={ANIMALS}, publisher={MDPI AG}, author={Erwin, Sara J. and Clark, Marley E. and Dechant, Julie E. and Aitken, Maia R. and Hassel, Diana M. and Blikslager, Anthony T. and Ziegler, Amanda L.}, year={2022}, month={Jun} } @misc{rose_blikslager_ziegler_2022, title={Porcine Models of the Intestinal Microbiota: The Translational Key to Understanding How Gut Commensals Contribute to Gastrointestinal Disease}, volume={9}, ISSN={["2297-1769"]}, DOI={10.3389/fvets.2022.834598}, abstractNote={In the United States, gastrointestinal disorders account for in excess of $130 billion in healthcare expenditures and 22 million hospitalizations annually. Many of these disorders, including necrotizing enterocolitis of infants, obesity, diarrhea, and inflammatory bowel disease, are associated with disturbances in the gastrointestinal microbial composition and metabolic activity. To further elucidate the pathogenesis of these disease syndromes as well as uncover novel therapies and preventative measures, gastrointestinal researchers should consider the pig as a powerful, translational model of the gastrointestinal microbiota. This is because pigs and humans share striking similarities in their intestinal microbiota as well as gastrointestinal anatomy and physiology. The introduction of gnotobiotic pigs, particularly human-microbial associated pigs, has already amplified our understanding of many gastrointestinal diseases that have detrimental effects on human health worldwide. Continued utilization of these models will undoubtedly inform translational advancements in future gastrointestinal research and potential therapeutics.}, journal={FRONTIERS IN VETERINARY SCIENCE}, author={Rose, Elizabeth C. and Blikslager, Anthony T. and Ziegler, Amanda L.}, year={2022}, month={Mar} } @article{erwin_blikslager_ziegler_2021, title={Age-Dependent Intestinal Repair: Implications for Foals with Severe Colic}, volume={11}, ISSN={["2076-2615"]}, url={https://doi.org/10.3390/ani11123337}, DOI={10.3390/ani11123337}, abstractNote={Simple Summary Equine colic places a substantial financial burden on horse owners and the equine industry each year. Equine veterinary research is focused on preventing colic on the farm whenever possible and improving treatment options available to veterinarians in the field and referral hospitals. It is important for scientists to have a detailed understanding of the intestinal damage created by different types of colic in foals and adult horses so they can better target certain cell types or tissue systems when investigating new treatment options. This review article summarizes recent works in the field of intestinal injury research and describes the potential roles of various intestinal systems, such as the enteric nervous system (ENS), in repairing the intestine after colic injury and how these systems mature in early life. Abstract Colic is a leading cause of death in horses, with the most fatal form being strangulating obstruction which directly damages the intestinal barrier. Following surgical intervention, it is imperative that the intestinal barrier rapidly repairs to prevent translocation of gut bacteria and their products and ensure survival of the patient. Age-related disparities in survival have been noted in many species, including horses, humans, and pigs, with younger patients suffering poorer clinical outcomes. Maintenance and repair of the intestinal barrier is regulated by a complex mucosal microenvironment, of which the ENS, and particularly a developing network of subepithelial enteric glial cells, may be of particular importance in neonates with colic. Postnatal development of an immature enteric glial cell network is thought to be driven by the microbial colonization of the gut and therefore modulated by diet-influenced changes in bacterial populations early in life. Here, we review the current understanding of the roles of the gut microbiome, nutrition, stress, and the ENS in maturation of intestinal repair mechanisms after foaling and how this may influence age-dependent outcomes in equine colic cases.}, number={12}, journal={ANIMALS}, author={Erwin, Sara J. and Blikslager, Anthony T. and Ziegler, Amanda L.}, year={2021}, month={Dec} } @misc{hellstrom_ziegler_blikslager_2021, title={Postoperative Ileus: Comparative Pathophysiology and Future Therapies}, volume={8}, ISSN={["2297-1769"]}, DOI={10.3389/fvets.2021.714800}, abstractNote={Postoperative ileus (POI), a decrease in gastrointestinal motility after surgery, is an important problem facing human and veterinary patients. 37.5% of horses that develop POI following small intestinal (SI) resection will not survive to discharge. The two major components of POI pathophysiology are a neurogenic phase which is then propagated by an inflammatory phase. Perioperative care has been implicated, namely the use of opioid therapy, inappropriate fluid therapy and electrolyte imbalances. Current therapy for POI variably includes an early return to feeding to induce physiological motility, reducing the inflammatory response with agents such as non-steroidal anti-inflammatory drugs (NSAIDs), and use of prokinetic therapy such as lidocaine. However, optimal management of POI remains controversial. Further understanding of the roles of the gastrointestinal microbiota, intestinal barrier function, the post-surgical inflammatory response, as well as enteric glial cells, a component of the enteric nervous system, in modulating postoperative gastrointestinal motility and the pathogenesis of POI may provide future targets for prevention and/or therapy of POI.}, journal={FRONTIERS IN VETERINARY SCIENCE}, author={Hellstrom, Emily A. and Ziegler, Amanda L. and Blikslager, Anthony T.}, year={2021}, month={Sep} } @article{rose_odle_blikslager_ziegler_2021, title={Probiotics, Prebiotics and Epithelial Tight Junctions: A Promising Approach to Modulate Intestinal Barrier Function}, volume={22}, ISSN={1422-0067}, url={http://dx.doi.org/10.3390/ijms22136729}, DOI={10.3390/ijms22136729}, abstractNote={Disruptions in the intestinal epithelial barrier can result in devastating consequences and a multitude of disease syndromes, particularly among preterm neonates. The association between barrier dysfunction and intestinal dysbiosis suggests that the intestinal barrier function is interactive with specific gut commensals and pathogenic microbes. In vitro and in vivo studies demonstrate that probiotic supplementation promotes significant upregulation and relocalization of interepithelial tight junction proteins, which form the microscopic scaffolds of the intestinal barrier. Probiotics facilitate some of these effects through the ligand-mediated stimulation of several toll-like receptors that are expressed by the intestinal epithelium. In particular, bacterial-mediated stimulation of toll-like receptor-2 modulates the expression and localization of specific protein constituents of intestinal tight junctions. Given that ingested prebiotics are robust modulators of the intestinal microbiota, prebiotic supplementation has been similarly investigated as a potential, indirect mechanism of barrier preservation. Emerging evidence suggests that prebiotics may additionally exert a direct effect on intestinal barrier function through mechanisms independent of the gut microbiota. In this review, we summarize current views on the effects of pro- and prebiotics on the intestinal epithelial barrier as well as on non-epithelial cell barrier constituents, such as the enteric glial cell network. Through continued investigation of these bioactive compounds, we can maximize their therapeutic potential for preventing and treating gastrointestinal diseases associated with impaired intestinal barrier function and dysbiosis.}, number={13}, journal={International Journal of Molecular Sciences}, publisher={MDPI AG}, author={Rose, Elizabeth C. and Odle, Jack and Blikslager, Anthony T. and Ziegler, Amanda L.}, year={2021}, month={Jun}, pages={6729} } @article{sheridan_pridgen_odle_van landeghem_blikslager_ziegler_2020, title={A Glial Cell Inhibitor Blocks Epithelial Barrier Repair in a Pig Model of Intestinal Ischemia}, volume={34}, ISSN={["1530-6860"]}, DOI={10.1096/fasebj.2020.34.s1.02030}, abstractNote={The gut epithelium functions as a barrier against toxic luminal contents which can repair efficiently after injury to prevent systemic illness and death. However, we have shown that repair is severely hindered in neonates as compared to juveniles in our translational pig model of intestinal ischemia. The subepithelial enteric glial cell (EGC) network is known to promote epithelial repair by paracrine signaling mechanisms. This EGC network develops postnatally; therefore, we believe this repair defect in neonates is due to an underdeveloped EGC network. In support of this, we hypothesize that EGC inhibition in juveniles will block epithelial barrier repair after ischemic injury mimicking the neonatal phenotype. Ischemia‐injured jejunum of juvenile pigs was recovered ex vivo with and without fluoroacetate (FA), a glial inhibitor. Transepithelial electrical resistance (TEER) was monitored as a measure of barrier function, and tissues were collected for imaging analysis and primary EGC culture. FA inhibited TEER recovery at all tested concentrations (P<0.0001). Histology showed 500μM FA optimally inhibits epithelial repair (P=0.0196) without directly damaging the epithelium (P=0.5509). Cellular metabolism marker c‐fos has been optimized for immunofluorescence in control tissues and ongoing imaging work will validate selective inhibition of EGC metabolism by 500μM FA. In addition, pig EGC isolation and culture has been optimized, so that future work will confirm inhibitory effects of FA on EGC signaling functions in vitro. Understanding the development of glial‐epithelial crosstalk in barrier repair will ultimately guide novel clinical solutions to improve outcomes in neonatal patients affected by intestinal injury.}, number={S1}, journal={FASEB JOURNAL}, publisher={Wiley}, author={Sheridan, Ana and Pridgen, Tiffany and Odle, Jack and Van Landeghem, Laurianne and Blikslager, Anthony and Ziegler, Amanda}, year={2020}, month={Apr} } @inproceedings{sheridan_pridgen_odle_landeghem_blikslager_ziegler_2020, place={San Diego, CA, USA}, title={A Glial Cell Inhibitor Blocks Epithelial Barrier Repair in a Pig Model of Intestinal Ischemia}, booktitle={Experimental Biology}, author={Sheridan, A.E. and Pridgen, T.A. and Odle, J. and Landeghem, L.Van and Blikslager, A.T. and Ziegler, A.L.}, year={2020}, month={Apr} } @inproceedings{ziegler_sheridan_pridgen_odle_landeghem_blikslager_2020, place={Washington, DC, USA}, title={An age-dependent, rescuable defect in intestinal barrier repair is associated with an immature enteric glial network in a neonatal pig model of intestinal ischemia}, booktitle={Translational Science 2020}, author={Ziegler, A.L. and Sheridan, A.E. and Pridgen, T.A. and Odle, J. and Landeghem, LVan and Blikslager, A.T.}, year={2020}, month={Apr} } @article{ziegler_blikslager_2020, title={Effects of Environmental Acclimation versus Transport Stress on Barrier Recovery in a Pig Model of Intestinal Ischemia and Repair}, volume={34}, ISSN={["1530-6860"]}, DOI={10.1096/fasebj.2020.34.s1.09380}, abstractNote={The pig is a powerful model for intestinal barrier studies, and it is important to carefully plan animal care and handling for optimal study design as psychological and physiological stressors significantly impact mucosal barrier function. Here, we report the effects of a period of environmental acclimation versus acute transport stress on mucosal barrier repair after intestinal ischemic injury. Jejunal ischemia was induced for 30‐minutes in 8–10‐week‐old pigs which had been transported and allowed to acclimate to a biomedical research housing environment for 3‐days prior to injury or been transported immediately prior to injury. Jejunal mucosa was then allowed to recover ex vivo in Ussing chambers while transepithelial electrical resistance (TEER) and mannitol flux were measured, and epithelial integrity was assessed by histomorphometry. In uninjured mucosa, there was no difference in basal TEER (P=0.8801) or epithelial integrity on histology between groups (P=0.5686), however, acclimated pigs had increased flux as compared to transported pigs during the first hour of ex vivo incubation (P=0.0186). Ischemia‐injured mucosa of acclimated pigs had less robust TEER recovery ex vivo (P=0.0101) as compared to transported pigs, but an increased initial flux in this group was significantly reduced during recovery (P=0.0025). Ischemia induced greater epithelial loss in transported pigs as compared to acclimated pigs (68.5% versus 87.8% epithelial coverage, P<0.0001), yet both groups restored control levels of epithelial coverage after 120‐minutes ex vivo recovery. These results indicate that acute transport stress on the day of experimental intestinal injury modeling may increase mucosal susceptibility to epithelial loss, but also prime the tissue for a more robust barrier repair response. Brief environmental acclimation, on the other hand, appears to increase intestinal permeability to molecular flux in the absence of injury, while possibly having a protective effect on epithelial loss during injury. These are important considerations for appropriate study design when utilizing highly translational pig models for intestinal barrier research.}, journal={FASEB JOURNAL}, author={Ziegler, Amanda and Blikslager, Anthony}, year={2020}, month={Apr} } @inproceedings{ziegler_blikslager_2020, place={San Diego, CA, USA}, title={Effects of Environmental Acclimation versus Transport Stress on Barrier Recovery in a Pig Model of Intestinal Ischemia and Repair}, booktitle={Experimental Biology}, author={Ziegler, A.L. and Blikslager, A.T.}, year={2020}, month={Apr} } @article{shapiro_ziegler_odle_van landeghem_blikslager_2020, title={Effects of Oligosaccharide Supplementation on Intestinal Morphology and Enteric Glial Cell Marker Expression in a Neonatal Pig Model}, volume={34}, ISSN={["1530-6860"]}, DOI={10.1096/fasebj.2020.34.s1.09457}, abstractNote={We have shown that oligosaccharide supplementation can modulate the gut flora in neonates, and the microbiome is known to drive the maturation of the enteric glial cell (EGC) network postnatally. Here, we test the effects of dietary oligosaccharides on postnatal changes in the EGC network and intestinal morphology in a neonatal pig model.After suckling colostrum for 24‐hours, one‐day‐old pigs were grouped onto one of three formula‐based diets: control, high oligosaccharide (1:1 mixture of galactooligosaccharide and polydextrose, 8g/L), or low oligosaccharide (4g/L). Small intestine and colon samples were collected at 1‐ 7‐ 14‐ and 21‐ days‐of‐age for western blot and histological analysis. Preliminary histological results indicate a trend toward a decreased small intestinal villus length in the high oligosaccharide group at 7‐days‐of‐age, indicative of accelerated intestinal maturity. Expression of the EGC marker glial fibrillary acidic protein is increased in the small intestinal mucosa at 7‐ and 14‐days‐of‐age in the high oligosaccharide group based on a preliminary western blot (n=1). Following ongoing work to assess crypt morphology and quantify additional EGC markers S100B, PLP‐1, Sox10 in the small intestine and colon, we expect to find increased mucosal expression of EGC markers earlier in postnatal development in the high oligosaccharide group, as well as histological changes consistent with enhanced rates of gut maturation in pigs fed a high oligosaccharide diet. Understanding how dietary inputs drive intestinal development postnatally may improve practices for managing optimal gut heath early in life.}, number={S1}, journal={FASEB JOURNAL}, publisher={Wiley}, author={Shapiro, Lindsey and Ziegler, Amanda and Odle, Jack and Van Landeghem, Laurianne and Blikslager, Anthony}, year={2020}, month={Apr} } @inproceedings{shapiro_ziegler_odle_landeghem_blikslager_2020, place={San Diego, CA, USA}, title={Effects of Oligosaccharide Supplementation on Intestinal Morphology and Enteric Glial Cell Marker Expression in a Neonatal Pig Model. }, booktitle={Experimental Biology}, author={Shapiro, L. and Ziegler, A.L. and Odle, J. and Landeghem, L.Van and Blikslager, A.T.}, year={2020}, month={Apr} } @article{ziegler_pridgen_blikslager_2020, title={Environmental stressors affect intestinal permeability and repair responses in a pig intestinal ischemia model}, volume={8}, ISSN={["2168-8370"]}, url={https://doi.org/10.1080/21688370.2020.1832421}, DOI={10.1080/21688370.2020.1832421}, abstractNote={ABSTRACT The pig is a powerful model for intestinal barrier studies, and it is important to carefully plan animal care and handling for optimal study design as psychological and physiological stressors significantly impact intestinal mucosal barrier function. Here, we report the effects of a period of environmental acclimation versus acute transport stress on mucosal barrier repair after intestinal ischemic injury. Jejunal ischemia was induced in young pigs which had been allowed to acclimate to a biomedical research housing environment or had been transported immediately prior to experimental injury (non-acclimated). Mucosa was then incubated ex vivo on Ussing chambers. In uninjured mucosa, there was no difference in transepithelial electrical resistance (TEER) or epithelial integrity between groups. However, acclimated pigs had increased macromolecular flux as compared to non-acclimated pigs during the first hour of ex vivo incubation. Ischemia induced greater epithelial loss in non-acclimated pigs as compared to acclimated pigs, yet this group achieved greater wound healing during recovery. Non-acclimated pigs had more robust TEER recovery ex vivo following injury versus acclimated pigs. The expression pattern of the tight junction protein claudin-4 was disrupted in acclimated pigs following recovery but showed enhanced localization to the apical membrane in non-acclimated pigs following recovery. Acute transport stress increases mucosal susceptibility to epithelial loss but also primes the tissue for a more robust barrier repair response. Alternatively, environmental acclimation increases leak pathway and diminishes barrier repair responses after ischemic injury.}, number={4}, journal={TISSUE BARRIERS}, publisher={Informa UK Limited}, author={Ziegler, Amanda L. and Pridgen, Tiffany A. and Blikslager, Anthony T.}, year={2020}, month={Oct} } @inproceedings{erwin_dechant_aitken_hassel_ziegler_blikslager_2020, title={Multi-institutional retrospective case-control study evaluating clinical outcomes of foals with small intestinal strangulating obstruction: 2000-2019. }, booktitle={2020 American College of Veterinary Surgeons Surgery E-Summit}, publisher={American College of Veterinary Surgeons Surgery E-Summit}, author={Erwin, S.J. and Dechant, J.E. and Aitken, M.R. and Hassel, D.M. and Ziegler, A.L. and Blikslager, A.T.}, year={2020}, month={Oct} } @article{ziegler_blikslager_2020, title={Sparing the gut: COX‐2 inhibitors herald a new era for treatment of horses with surgical colic}, url={https://doi.org/10.1111/eve.13189}, DOI={10.1111/eve.13189}, abstractNote={Nonsteroidal anti-inflammatory drugs (NSAIDs) are commonly used to manage a wide variety of conditions in horses, including management of colic. Flunixin meglumine is by far the most commonly used drug in the control of colic pain and inflammation and has become a go-to for not only veterinarians but also horse-owners and nonmedical equine professionals. NSAID use, however, has always been controversial in critical cases due to a high risk of adverse effects associated with their potent cyclo-oxygenase (COX) inhibition. There are two important COX isoenzymes: COX-1 is generally beneficial for normal renal and gastrointestinal functions and COX-2 is associated with the pain and inflammation of disease. Newer selective NSAIDs can target COX-2-driven pathology while sparing important COX-1-driven physiology, which is of critical importance in horses with severe gastrointestinal disease. Emerging research suggests that firocoxib, a COX-2-selective NSAID labelled for use in horses, may be preferable for use in colic cases in spite of the decades-long dogma that flunixin saves lives.}, journal={Equine Veterinary Education}, author={Ziegler, A. L. and Blikslager, A. T.}, year={2020}, month={Nov} } @article{erwin_touvron_odle_van landeghem_blikslager_ziegler_2020, title={iDISCO Allows Complete Visualization and Analysis of Postnatal Enteric Nervous System Development in a Comparative Pig Model}, volume={34}, ISSN={["1530-6860"]}, DOI={10.1096/fasebj.2020.34.s1.03991}, abstractNote={The enteric nervous system consists of a dense, complex network of neurons and glia which are instrumental in the maturation of normal intestinal physiology after birth. Our lab uses a comparative pig model to study the postnatal development of the enteric glial network and its role in regulating intestinal barrier functions in neonates. Immunolabeling‐enabled three‐dimensional (3D) imaging of solvent‐cleared organs (iDISCO) is a method of preparing tissue samples for volume imaging with a light sheet microscope. iDISCO has been optimized for use in primarily mouse organs and embryos for the study of early development and offers a more complete picture of the tissue than traditional histological analysis. Our objective was to optimize the iDISCO protocol for use in porcine intestinal tissue to allow complete qualitative and quantitative analysis of postnatal development of the enteric glial network in our comparative pig model. Antibodies against glial cell markers S100b, Sox10, and glial fibrillary acidic protein (GFAP) were used to triple‐stain fixed full‐thickness 3mm by 5mm samples of porcine jejunum using the iDISCO protocol. Samples were imaged with a light‐sheet microscope (Ultra‐II, LaVision BioTec ® ) using three different fluorescent channels and datasets were visualized and analyzed in 3D with Imaris software (Oxford Instruments ® ). The percent volume of GFAP + glial cells was quantified by manually masking individual intestinal villi and optimizing a surface algorithm to identify glial network structures within those villi. Antibodies against all three markers tested produced the predicted staining pattern with minimal non‐specific staining. Percent of jejunal villus volume occupied by GFAP + glia is higher in 6‐week‐old versus 2‐week‐old pig (0.49% versus 0.23%, *P≤0.05). Ongoing work will optimize quantification techniques for S100b and Sox10, and assess co‐localization patterns of these glial markers in the jejunum at discrete timepoints postnatally. iDISCO is a powerful imaging modality which will allow our lab to directly assess the expansion, complexity, and localization of glial cell subtypes by marker co‐expression analysis at discrete postnatal timepoints, and will be utilized in future studies to explore effects of disease and external interventions on the enteric glial network. Support or Funding Information NIH P30 DK034987 UNC CGIBD Pilot Feasibility; NIH T32 5T32DK007737‐22 UNC Basic Science GI Research Training Fellowship; USDA National Institute of Food and Agriculture, Animal Health Projects 1007263 and 07985; 2017 NC State CMI TPP Seed Grant}, number={S1}, journal={FASEB JOURNAL}, publisher={Wiley}, author={Erwin, Sara and Touvron, Melissa and Odle, Jack and Van Landeghem, Laurianne and Blikslager, Anthony and Ziegler, Amanda}, year={2020}, month={Apr} } @inproceedings{erwin_touvron_odle_landeghem_blikslager_ziegler_2020, place={San Diego, CA, USA}, title={iDISCO allows complete visualization and analysis of postnatal enteric nervous system development in a comparative pig model}, booktitle={Experimental Biology}, author={Erwin, S.E. and Touvron, M. and Odle, J. and Landeghem, L.Van and Blikslager, A.T. and Ziegler, A.L.}, year={2020}, month={Apr} } @article{ziegler_fogle_burke_blikslager_2019, title={Letter to the Editor: Bias in statistics or bias in equine veterinary medicine?}, volume={51}, ISSN={0425-1644 2042-3306}, url={http://dx.doi.org/10.1111/evj.13081}, DOI={10.1111/evj.13081}, abstractNote={See Correspondence by Freeman}, number={3}, journal={Equine Veterinary Journal}, publisher={Wiley}, author={Ziegler, A. L. and Fogle, C. A. and Burke, M. and Blikslager, A. T.}, year={2019}, month={Feb}, pages={423–423} } @inproceedings{ziegler_pridgen_landeghem_odle_magness_blikslager_2019, place={San Diego, CA, USA}, title={Rescue of Restitution Defect in a Neonatal Pig Intestinal Ischemia Model is associated with an Immature Enteric Glial Network}, booktitle={Digestive Disease Week}, author={Ziegler, A.L. and Pridgen, T.A. and Landeghem, L.Van and Odle, J. and Magness, S.T. and Blikslager, A.T.}, year={2019}, month={May} } @inproceedings{ziegler_pridgen_landeghem_odle_magness_blikslager_2019, place={Chicago, IL, USA}, title={Suckling piglets have a rescuable defect in intestinal barrier repair associated with an immature enteric glial network}, booktitle={Conference of Research Workers in Animal Disease}, author={Ziegler, A.L. and Pridgen, T.A. and Landeghem, L.Van and Odle, J. and Magness, S.T. and Blikslager, A.T.}, year={2019}, month={Nov} } @article{ziegler_pridgen_mills_gonzalez_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={7}, url={https://doi.org/10.1101/361352}, DOI={10.1101/361352}, abstractNote={ABSTRACT 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 3 H-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.}, publisher={Cold Spring Harbor Laboratory}, author={Ziegler, Amanda L. and Pridgen, Tiffany A. and Mills, Juliana K. and Gonzalez, Liara M. and Landeghem, Laurianne Van and Odle, Jack and Blikslager, Anthony T.}, year={2018}, month={Jul} } @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{ziegler_freeman_fogle_burke_davis_cook_southwood_blikslager_2018, title={Multicentre, blinded, randomised clinical trial comparing the use of flunixin meglumine with firocoxib in horses with small intestinal strangulating obstruction}, volume={51}, ISSN={0425-1644 2042-3306}, url={http://dx.doi.org/10.1111/evj.13013}, DOI={10.1111/evj.13013}, abstractNote={Summary}, number={3}, journal={Equine Veterinary Journal}, publisher={Wiley}, author={Ziegler, A. L. and Freeman, C. K. and Fogle, C. A. and Burke, M. J. and Davis, J. L. and Cook, V. L. and Southwood, L. L. and Blikslager, A. T.}, year={2018}, month={Sep}, pages={329–335} } @inproceedings{ziegler_pridgen_landeghem_gonzalez_odle_blikslager_2018, place={College Station, TX, USA}, title={Neonatal Defect in Subacute Barrier Repair is Rescued by Juvenile Mucosal Homogenate in a Pig Model of Intestinal Ischemia and Repair}, volume={Aug 3}, booktitle={National Veterinary Scholars Symposium}, author={Ziegler, A.L. and Pridgen, T.A. and Landeghem, L.Van and Gonzalez, L.M. and Odle, J. and Blikslager, A.T.}, year={2018}, month={Aug} } @inproceedings{ziegler_freeman_fogle_burke_davis_southwood_cook_blikslager_2018, place={Oral Abstract}, title={Randomized control trial comparing flunixin meglumine and firocoxib in equine strangulating obstruction}, booktitle={American Association of Equine Practitioners Convention}, publisher={American Association of Equine Practitioners}, author={Ziegler, A.L. and Freeman, C.K. and Fogle, C.A. and Burke, M.J. and Davis, J. and Southwood, L.L. and Cook, V.L. and Blikslager, A.T.}, year={2018}, month={Dec} } @inproceedings{ziegler_pridgen_landeghem_gonzalez_odle_blikslager_2017, place={Lexington, KY, USA}, title={Neonates have a reduced ability to repair jejunal mucosal injury as compared to weanlings in a porcine model of small intestinal strangulating obstruction}, booktitle={12th International Colic Research Symposium}, author={Ziegler, A.L. and Pridgen, T.A. and Landeghem, L.Van and Gonzalez, L.M. and Odle, J. and Blikslager, A.T.}, year={2017}, month={Jul} } @inproceedings{ziegler_pridgen_landeghem_gonzalez_odle_blikslager_2017, place={CO, USA}, title={Neonates have reduced epithelial wound healing associated with fewer enteric glial cells as compared to juveniles in a pig model of intestinal ischemic injury and repair}, booktitle={FASEB Gastrointestinal Tract XVII}, author={Ziegler, A.L. and Pridgen, T.A. and Landeghem, L.Van and Gonzalez, L.M. and Odle, J. and Blikslager, A.T.}, year={2017}, month={Jul} } @inproceedings{ziegler_fogle_burke_davis_southwood_cook_blikslager_2017, place={Lexington, KY, USA}, title={Randomized control trial comparing flunixin and firocoxib in equine strangulating obstruction}, booktitle={12th International Colic Research Symposium}, author={Ziegler, A.L. and Fogle, C. and Burke, M. and Davis, J. and Southwood, L. and Cook, V. and Blikslager, A.T.}, year={2017}, month={Jul} } @article{ziegler_fogle_blikslager_2017, title={Update on the use of cyclooxygenase-2-selective nonsteroidal anti-inflammatory drugs in horses}, volume={250}, ISSN={0003-1488}, url={http://dx.doi.org/10.2460/javma.250.11.1271}, DOI={10.2460/javma.250.11.1271}, abstractNote={Abstract}, number={11}, journal={Journal of the American Veterinary Medical Association}, publisher={American Veterinary Medical Association (AVMA)}, author={Ziegler, Amanda and Fogle, Callie and Blikslager, Anthony}, year={2017}, month={Jun}, pages={1271–1274} } @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} } @inproceedings{ziegler_pridgen_carnighan_odle_gonzalez_blikslager_2016, place={San Diego, CA, USA}, title={Neonates Have a Reduced Ability to Repair Jejunal Mucosal Injury as Compared to Juveniles in a Pig Model of Ischemia/ Reperfusion Injury}, booktitle={Digestive Disease Week}, author={Ziegler, A.L. and Pridgen, T.A. and Carnighan, G. and Odle, J. and Gonzalez, L.M. and Blikslager, A.T.}, year={2016}, month={May} } @article{newman_sills_hanrahan_ziegler_tidd_cook_sannes_2015, title={Expression of WNT5A in Idiopathic Pulmonary Fibrosis and Its Control by TGF-β and WNT7B in Human Lung Fibroblasts}, volume={64}, ISSN={0022-1554 1551-5044}, url={http://dx.doi.org/10.1369/0022155415617988}, DOI={10.1369/0022155415617988}, abstractNote={ The wingless (Wnt) family of signaling ligands contributes significantly to lung development and is highly expressed in patients with usual interstitial pneumonia (UIP). We sought to define the cellular distribution of Wnt5A in the lung tissue of patients with idiopathic pulmonary fibrosis (IPF) and the signaling ligands that control its expression in human lung fibroblasts and IPF myofibroblasts. Tissue sections from 40 patients diagnosed with IPF or UIP were probed for the immunolocalization of Wnt5A. Further, isolated lung fibroblasts from normal or IPF human lungs, adenovirally transduced for the overexpression or silencing of Wnt7B or treated with TGF-β1 or its inhibitor, were analyzed for Wnt5A protein expression. Wnt5A was expressed in IPF lungs by airway and alveolar epithelium, smooth muscle cells, endothelium, and myofibroblasts of fibroblastic foci and throughout the interstitium. Forced overexpression of Wnt7B with or without TGF-β1 treatment significantly increased Wnt5A protein expression in normal human smooth muscle cells and fibroblasts but not in IPF myofibroblasts where Wnt5A was already highly expressed. The results demonstrate a wide distribution of Wnt5A expression in cells of the IPF lung and reveal that it is significantly increased by Wnt7B and TGF-β1, which, in combination, could represent key signaling pathways that modulate the pathogenesis of IPF. }, number={2}, journal={Journal of Histochemistry & Cytochemistry}, publisher={SAGE Publications}, author={Newman, Donna R. and Sills, W. Shane and Hanrahan, Katherine and Ziegler, Amanda and Tidd, Kathleen McGinnis and Cook, Elizabeth and Sannes, Philip L.}, year={2015}, month={Nov}, pages={99–111} } @inproceedings{welch_newman_sannes_2013, place={Lansing, MI, USA}, title={Porcine alveolar type II cells as a potential model for human alveolar epithelium in culture.}, booktitle={National Veterinary Scholars Symposium}, author={Welch, A.L. and Newman, D.R. and Sannes, P.L.}, year={2013}, month={Aug} }