1
|
Offersen SM, Henriksen NL, Brunse A. A weighted and cumulative point system for accurate scoring of intestinal pathology in a piglet model of necrotizing enterocolitis. Exp Mol Pathol 2024; 140:104936. [PMID: 39366159 DOI: 10.1016/j.yexmp.2024.104936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/06/2024]
Abstract
Necrotizing enterocolitis (NEC) is a serious condition in premature infants, in which a portion of the intestine undergoes inflammation and necrosis. The preterm pig develops NEC spontaneously, making it a suitable model for exploring novel NEC treatments. We aimed to revise the intestinal scoring system to more accurately describe the diversity of NEC lesions in the preterm piglet model. We included 333 preterm piglets from four experiments, each delivered via cesarean section. The piglets were fed either a gently processed (GP) or harshly processed (HP) milk formula for 96 h before euthanasia. At necropsy, the gastrointestinal tract was assessed with 1) an established 6-grade score and 2) a descriptive approach focusing on the distribution and severity of hyperemia, hemorrhage, pneumatosis intestinalis (intramural gas), and necrosis. Subsequently, the descriptive registrations were converted into a weighted and cumulative point (WCP) score. Compared to the 6-grade score, the WCP score enabled a greater segregation of severity levels, especially among organs with more prominent NEC lesions. IL-1β in small intestinal lesions and both IL-8 and IL-1β in colon lesions correlated positively with the WCP scale. A histopathological grade system (0-8) was established and revealed mucosal pathology in lesion biopsies, which were not recognized macroscopically. Finally, the WCP score showed a higher NEC-promoting effect of the HP formula compared to the GP formula. The descriptive registrations and extended score range of this revised intestinal scoring system enhance the accuracy of describing NEC lesions in preterm pigs. This approach may increase the efficiency of preclinical NEC experiments.
Collapse
Affiliation(s)
- Simone Margaard Offersen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Nicole Lind Henriksen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Anders Brunse
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark.
| |
Collapse
|
2
|
Wang Q, Wang Y, Wang Y, Zhang Q, Mi J, Ma Q, Li T, Huang S. Agaro-oligosaccharides mitigate deoxynivalenol-induced intestinal inflammation by regulating gut microbiota and enhancing intestinal barrier function in mice. Food Funct 2024; 15:3380-3394. [PMID: 38498054 DOI: 10.1039/d3fo04898e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Agarose-derived agaro-oligosaccharides (AgaroS) have been extensively studied in terms of structures and bioactivities; they reportedly possess antioxidant and anti-inflammatory activities that maintain intestinal homeostasis and host health. However, the protective effects of AgaroS on deoxynivalenol (DON)-induced intestinal dysfunction remain unclear. We investigated the effects of AgaroS on DON-induced intestinal dysfunction in mice and explored the underlying protective mechanisms. In total, 32 mice were randomly allocated to four treatments (n = 8 each) for 28 days. From day 1 to day 21, the control (CON) and DON groups received oral phosphate-buffered saline (200 μL per day); the AgaroS and AgaroS + DON groups received 200 mg AgaroS per kg body weight once daily by orogastric gavage. Experimental intestinal injury was induced by adding DON (4.8 mg per kg body weight) via gavage from day 21 to day 28. Phosphate-buffered saline was administered once daily by gavage in the CON and AgaroS groups. Herein, AgaroS supplementation led to a higher final body weight and smaller body weight loss and a lower concentration of plasma inflammatory cytokines, compared with the DON group. The DON group showed a significantly reduced ileal villus height and villus height/crypt depth, compared with the CON and AgaroS + DON groups. However, AgaroS supplementation improved DON-induced intestinal injury in mice. Compared with the DON group, ileal and colonic protein expression levels of claudin, occludin, Ki67, and mucin2 were significantly higher in the AgaroS supplementation group. Colonic levels of the anti-inflammatory cytokine IL-1β tended to be higher in the DON group than in the AgaroS + DON group. AgaroS altered the gut microbiota composition, accompanied by increased production of short-chain fatty acids in mice. In conclusion, our findings highlight a promising anti-mycotoxin approach whereby AgaroS alleviate DON-induced intestinal inflammation by modulating intestinal barrier functional integrity and gut microbiota in mice.
Collapse
Affiliation(s)
- Qingfeng Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
- Feed Safety and Healthy Livestock, Beijing Jingwa Agricultural Innovation Center, Beijing, China
| | - Yanwei Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
- Feed Safety and Healthy Livestock, Beijing Jingwa Agricultural Innovation Center, Beijing, China
- School of Life Sciences, Shanxi University, Taiyuan, 030006, Shanxi, China
| | - Yue Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
- Feed Safety and Healthy Livestock, Beijing Jingwa Agricultural Innovation Center, Beijing, China
| | - Qiyue Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
- Feed Safety and Healthy Livestock, Beijing Jingwa Agricultural Innovation Center, Beijing, China
- College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou, 21001, Liaoning, China
| | - Jinqiu Mi
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
- Feed Safety and Healthy Livestock, Beijing Jingwa Agricultural Innovation Center, Beijing, China
| | - Qiugang Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
- Feed Safety and Healthy Livestock, Beijing Jingwa Agricultural Innovation Center, Beijing, China
| | - Tiantian Li
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, China.
| | - Shimeng Huang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
- Feed Safety and Healthy Livestock, Beijing Jingwa Agricultural Innovation Center, Beijing, China
| |
Collapse
|
3
|
Kebbe M, Shankar K, Redman LM, Andres A. Human Milk Components and the Infant Gut Microbiome at 6 Months: Understanding the Interconnected Relationship. J Nutr 2024; 154:1200-1208. [PMID: 38442855 DOI: 10.1016/j.tjnut.2024.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/29/2024] [Accepted: 02/26/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Human milk oligosaccharides have been shown to relate to the infant gut microbiome. However, the impact of other human milk components on infant gut bacterial colonization remains unexplored. OBJECTIVES Our cross-sectional analysis aimed to investigate associations between human milk components (energy, macronutrients, free amino acids, inflammatory markers, and hormones) and infant gut microbiome diversity and composition (phylum, family, and genus) at 6 mo of age. METHODS Human milk and infant stool samples were collected at 6 mo postpartum. The infant gut microbiome was profiled using 16S rRNA sequencing. Linear regression models were performed to examine associations, adjusting for pregravid BMI (kg/m2), delivery mode, duration of human milk feeding, and infant sex, with q < 0.2 considered significant. RESULTS This analysis included a total of 54 mothers (100% exclusively feeding human milk) and infants (n = 28 male; 51.9%). Total energy in human milk showed a negative association with α-diversity measures (Chao1 and Shannon). Interleukin (IL)-8 in human milk was positively associated with Chao1 and observed operational taxonomic units. At the family level, human milk glutamine and serine levels showed a negative association with the abundance of Veillonellaceae, whereas isoleucine showed a positive association with Bacteroidaceae. Human milk IL-8 and IL-6 concentrations were positively associated with Bacteroidaceae abundance. IL-8 also had a positive relationship with Bifidobacteriaceae, whereas it had a negative relationship with Streptococcacea and Clostridiaceae. Human milk IL-8 was positively associated with the phylum Bacteroidetes, and negatively associated with Proteobacteria. At the genus level, human milk IL-8 exhibited a positive relationship with Bacteroides, whereas human milk isoleucine had a negative relationship with Bacteroides and Ruminococcus. Pregravid BMI and sex effects were observed. CONCLUSIONS IL-8 in human milk could potentially prepare the infant's immune system to respond effectively to various microorganisms, potentially promoting the growth of beneficial gut bacteria and protecting against pathogens.
Collapse
Affiliation(s)
- Maryam Kebbe
- Faculty of Kinesiology, University of New Brunswick, Fredericton, New Brunswick, Canada
| | - Kartik Shankar
- Department of Pediatrics, University of Colorado, Denver, CO, United States
| | - Leanne M Redman
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States
| | - Aline Andres
- Arkansas Children's Nutrition Center, Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States.
| |
Collapse
|
4
|
Solà Tapias N, Denadai-Souza A, Rolland-Fourcade C, Quaranta-Nicaise M, Blanpied C, Marcellin M, Edir A, Rolland C, Cirillo C, Dietrich G, Alric L, Portier G, Kirzin S, Bonnet D, Mas E, Burlet-Schiltz O, Deraison C, Bonnart C, Vergnolle N, Barreau F. Colitis Linked to Endoplasmic Reticulum Stress Induces Trypsin Activity Affecting Epithelial Functions. J Crohns Colitis 2021; 15:1528-1541. [PMID: 33609354 DOI: 10.1093/ecco-jcc/jjab035] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND AND AIMS Intestinal epithelial cells [IECs] from inflammatory bowel disease [IBD] patients exhibit an excessive induction of endoplasmic reticulum stress [ER stress] linked to altered intestinal barrier function and inflammation. Colonic tissues and the luminal content of IBD patients are also characterized by increased serine protease activity. The possible link between ER stress and serine protease activity in colitis-associated epithelial dysfunctions is unknown. We aimed to study the association between ER stress and serine protease activity in enterocytes and its impact on intestinal functions. METHODS The impact of ER stress induced by Thapsigargin on serine protease secretion was studied using either human intestinal cell lines or organoids. Moreover, treating human intestinal cells with protease-activated receptor antagonists allowed us to investigate ER stress-resulting molecular mechanisms that induce proteolytic activity and alter intestinal epithelial cell biology. RESULTS Colonic biopsies from IBD patients exhibited increased epithelial trypsin-like activity associated with elevated ER stress. Induction of ER stress in human intestinal epithelial cells displayed enhanced apical trypsin-like activity. ER stress-induced increased trypsin activity destabilized intestinal barrier function by increasing permeability and by controlling inflammatory mediators such as C-X-C chemokine ligand 8 [CXCL8]. The deleterious impact of ER stress-associated trypsin activity was specifically dependent on the activation of protease-activated receptors 2 and 4. CONCLUSIONS Excessive ER stress in IECs caused an increased release of trypsin activity that, in turn, altered intestinal barrier function, promoting the development of inflammatory process.
Collapse
Affiliation(s)
- Núria Solà Tapias
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | | | | | | | | | - Marlène Marcellin
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Anissa Edir
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | - Corinne Rolland
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | - Carla Cirillo
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France.,Laboratory for Enteric NeuroScience (LENS), TARGID, University of Leuven, Leuven, Belgium
| | - Gilles Dietrich
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | | | | | | | | | - Emmanuel Mas
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France.,Pole Digestif, CHU, Toulouse, France
| | - Odile Burlet-Schiltz
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Céline Deraison
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | | | - Nathalie Vergnolle
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France.,Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Frédérick Barreau
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| |
Collapse
|
5
|
De Fazio L, Beghetti I, Bertuccio SN, Marsico C, Martini S, Masetti R, Pession A, Corvaglia L, Aceti A. Necrotizing Enterocolitis: Overview on In Vitro Models. Int J Mol Sci 2021; 22:6761. [PMID: 34201786 PMCID: PMC8268427 DOI: 10.3390/ijms22136761] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 06/16/2021] [Accepted: 06/21/2021] [Indexed: 12/12/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a gut inflammatory disorder which constitutes one of the leading causes of morbidity and mortality for preterm infants. The pathophysiology of NEC is yet to be fully understood; several observational studies have led to the identification of multiple factors involved in the pathophysiology of the disease, including gut immaturity and dysbiosis of the intestinal microbiome. Given the complex interactions between microbiota, enterocytes, and immune cells, and the limited access to fetal human tissues for experimental studies, animal models have long been essential to describe NEC mechanisms. However, at present there is no animal model perfectly mimicking human NEC; furthermore, the disease mechanisms appear too complex to be studied in single-cell cultures. Thus, researchers have developed new approaches in which intestinal epithelial cells are exposed to a combination of environmental and microbial factors which can potentially trigger NEC. In addition, organoids have gained increasing attention as promising models for studying NEC development. Currently, several in vitro models have been proposed and have contributed to describe the disease in deeper detail. In this paper, we will provide an updated review of available in vitro models of NEC and an overview of current knowledge regarding its molecular underpinnings.
Collapse
Affiliation(s)
- Luigia De Fazio
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (L.D.F.); (S.N.B.); (C.M.); (S.M.); (R.M.); (A.P.); (L.C.); (A.A.)
- Pediatric Oncology and Hematology “Lalla Seragnoli”, Pediatric Unit-IRCCS Azienda Ospedaliero-Universitaria, 40138 Bologna, Italy
| | - Isadora Beghetti
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (L.D.F.); (S.N.B.); (C.M.); (S.M.); (R.M.); (A.P.); (L.C.); (A.A.)
- Neonatal Intensive Care Unit-IRCCS Azienda Ospedaliero-Universitaria, 40138 Bologna, Italy
| | - Salvatore Nicola Bertuccio
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (L.D.F.); (S.N.B.); (C.M.); (S.M.); (R.M.); (A.P.); (L.C.); (A.A.)
- Pediatric Oncology and Hematology “Lalla Seragnoli”, Pediatric Unit-IRCCS Azienda Ospedaliero-Universitaria, 40138 Bologna, Italy
| | - Concetta Marsico
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (L.D.F.); (S.N.B.); (C.M.); (S.M.); (R.M.); (A.P.); (L.C.); (A.A.)
- Neonatal Intensive Care Unit-IRCCS Azienda Ospedaliero-Universitaria, 40138 Bologna, Italy
| | - Silvia Martini
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (L.D.F.); (S.N.B.); (C.M.); (S.M.); (R.M.); (A.P.); (L.C.); (A.A.)
- Neonatal Intensive Care Unit-IRCCS Azienda Ospedaliero-Universitaria, 40138 Bologna, Italy
| | - Riccardo Masetti
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (L.D.F.); (S.N.B.); (C.M.); (S.M.); (R.M.); (A.P.); (L.C.); (A.A.)
- Pediatric Oncology and Hematology “Lalla Seragnoli”, Pediatric Unit-IRCCS Azienda Ospedaliero-Universitaria, 40138 Bologna, Italy
| | - Andrea Pession
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (L.D.F.); (S.N.B.); (C.M.); (S.M.); (R.M.); (A.P.); (L.C.); (A.A.)
- Pediatric Oncology and Hematology “Lalla Seragnoli”, Pediatric Unit-IRCCS Azienda Ospedaliero-Universitaria, 40138 Bologna, Italy
| | - Luigi Corvaglia
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (L.D.F.); (S.N.B.); (C.M.); (S.M.); (R.M.); (A.P.); (L.C.); (A.A.)
- Neonatal Intensive Care Unit-IRCCS Azienda Ospedaliero-Universitaria, 40138 Bologna, Italy
| | - Arianna Aceti
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (L.D.F.); (S.N.B.); (C.M.); (S.M.); (R.M.); (A.P.); (L.C.); (A.A.)
- Neonatal Intensive Care Unit-IRCCS Azienda Ospedaliero-Universitaria, 40138 Bologna, Italy
| |
Collapse
|
6
|
Splichalova A, Slavikova V, Splichalova Z, Splichal I. Preterm Life in Sterile Conditions: A Study on Preterm, Germ-Free Piglets. Front Immunol 2018; 9:220. [PMID: 29491864 PMCID: PMC5817058 DOI: 10.3389/fimmu.2018.00220] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 01/25/2018] [Indexed: 12/21/2022] Open
Abstract
Preterm infants born with immature organ systems, which can impede normal development, can also be highly sensitive to different biological and/or environmental factors. Animal models could aid in investigating and understanding the effects of different conditions on the health of these immunocompromised infants. The epitheliochorial placentation of the pig prevents the prenatal transfer of protective colostral immunoglobulins. Surgical colostrum-deprived piglets are free of maternal immunoglobulins, and the cells that are normally provided via colostrum. We bred preterm germ-free piglets in sterile conditions and compared them with their term counterparts. Enterocyte development and intestinal morphology, tight junction proteins claudin-1 and occludin, pattern-recognizing receptors, adaptor molecules and coreceptors (RAGE, TLR2, TLR4, TLR9, MyD88, TRIF, MD2, and CD14), and inflammasome NLRP3 transcription were all evaluated. The production of inflammatory mediators IFN-α, IL-4, IL-6, IL-8, IL-10, IL-12/23 p40, TNF-α, IFN-γ, and high mobility group box 1 (HMGB1) in the intestine of germ-free piglets was also assessed. In the preterm germ-free piglets, the ileum showed decreased lamina propria cellularity, reduced villous height, and thinner and less distinct stratification - especially muscle layer, in comparison with their term counterparts. Claudin-1 transcription increased in the intestine of the preterm piglets. The transcription levels of pattern-recognizing receptors and adaptor molecules showed ambiguous trends between the groups. The levels of IL-6, IL-8, IL-10, and TNF-α were increased in the preterm ileum numerically (though not significantly), with statistically significant increases in the colon. Additionally, IL-12/23 p40 and IFN-γ were statistically significantly higher in the preterm colon. Both blood plasma and intestinal HMGB1 levels were nonsignificantly higher in the preterm group. We propose that the intestine of the preterm germ-free piglets showed "mild inflammation in sterile conditions." This model, which establishes preterm, hysterectomy-derived germ-free piglets, without protective maternal immunoglobulins, can be used to study influences of microbiota, nutrition, and therapeutic interventions on the development and health of vulnerable immunocompromised preterm infants.
Collapse
Affiliation(s)
- Alla Splichalova
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Novy Hradek, Czechia
| | - Vera Slavikova
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Novy Hradek, Czechia
| | - Zdislava Splichalova
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Novy Hradek, Czechia
| | - Igor Splichal
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Novy Hradek, Czechia
| |
Collapse
|
7
|
Anders G, Hassiepen U, Theisgen S, Heymann S, Muller L, Panigada T, Huster D, Samsonov SA. The Intrinsic Pepsin Resistance of Interleukin-8 Can Be Explained from a Combined Bioinformatical and Experimental Approach. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2018; 15:300-308. [PMID: 28113517 DOI: 10.1109/tcbb.2016.2614821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Interleukin-8 (IL-8, CXCL8) is a neutrophil chemotactic factor belonging to the family of chemokines. IL-8 was shown to resist pepsin cleavage displaying its high resistance to this protease. However, the molecular mechanisms underlying this resistance are not fully understood. Using our in-house database containing the data on three-dimensional arrangements of secondary structure elements from the whole Protein Data Bank, we found a striking structural similarity between IL-8 and pepsin inhibitor-3. Such similarity could play a key role in understanding IL-8 resistance to the protease pepsin. To support this hypothesis, we applied pepsin assays confirming that intact IL-8 is not degraded by pepsin in comparison to IL-8 in a denaturated state. Applying 1H-15N Heteronuclear Single Quantum Coherence NMR measurements, we determined the putative regions at IL-8 that are potentially responsible for interactions with the pepsin. The results obtained in this work contribute to the understanding of the resistance of IL-8 to pepsin proteolysis in terms of its structural properties.
Collapse
|
8
|
Marin M, Holani R, Shah CB, Odeón A, Cobo ER. Cathelicidin modulates synthesis of Toll-like Receptors (TLRs) 4 and 9 in colonic epithelium. Mol Immunol 2017; 91:249-258. [PMID: 28988039 DOI: 10.1016/j.molimm.2017.09.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 09/12/2017] [Accepted: 09/21/2017] [Indexed: 12/13/2022]
Abstract
Cathelicidin are innate antimicrobial peptides with broad immunomodulatory functions; however, their role in regulating intestinal defenses is not well characterized. This study aimed to investigate the role of cathelicidin modulating expression of Toll-like receptors (TLRs) 4 and 9 in colonic epithelium in response to bacterial patterns. We demonstrated herein that intestinal epithelial cells, when primed by bacterial lipopolysaccharide (LPS), responded to cathelicidin by increased transcription and protein synthesis of TLR4. This cathelicidin-induced response required the interaction of LPS-TLR4 and activation of MAPK signalling pathways. However, cathelicidin blocked TLR9 responses induced by TLR9 ligand CpG oligodeoxynucleotide (CpG ODN) in these colonic epithelial cells. Modulations of TLRs triggered by cathelicidin in intestinal epithelium occurred mainly in the apical compartment of intestinal cells. Activation of TLR4 by ligands in combination with cathelicidin promoted CXCL8 chemokine secretion and epithelial antimicrobial defenses against Escherichia coli. We concluded that cathelicidin selectively modulated synthesis of TLR4 and 9 in intestinal epithelium, but only when cells were exposed to virulence factors, mostly from apical surfaces. Enhanced TLR4 expression promoted by cathelicidin in intestinal epithelium may be crucial for controlling enteric infectious diseases.
Collapse
Affiliation(s)
- Maia Marin
- National Scientific and Technical Research Council (CONICET), Argentina
| | - Ravi Holani
- Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Canada
| | | | - Anselmo Odeón
- Animal Production, Balcarce Experimental Station, National Institute of Agricultural Technology (INTA), Argentina
| | - Eduardo R Cobo
- Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Canada.
| |
Collapse
|
9
|
MohanKumar K, Namachivayam K, Ho TT, Torres BA, Ohls RK, Maheshwari A. Cytokines and growth factors in the developing intestine and during necrotizing enterocolitis. Semin Perinatol 2017; 41:52-60. [PMID: 27832931 PMCID: PMC5334139 DOI: 10.1053/j.semperi.2016.09.018] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cytokines and growth factors play diverse roles in the uninflamed fetal/neonatal intestinal mucosa and in the development of inflammatory bowel injury during necrotizing enterocolitis (NEC). During gestational development and the early neonatal period, the fetal/premature intestine is exposed to high levels of many "inflammatory" cytokines and growth factors, first via swallowed amniotic fluid in utero and then, after birth, in colostrum and mother's milk. This article reviews the dual, seemingly counter-intuitive roles of cytokines, where these agents play a "trophic" role and promote maturation of the uninflamed mucosa, but can also cause inflammation and promote intestinal injury during NEC.
Collapse
Affiliation(s)
| | | | - Thao T.B. Ho
- Department of Pediatrics, Morsani College of Medicine, Tampa, FL
| | | | - Robin K. Ohls
- Department of Pediatrics, University of New Mexico, Albuquerque, NM
| | - Akhil Maheshwari
- Department of Pediatrics, Morsani College of Medicine, Tampa, FL; Departments of Molecular Medicine, Morsani College of Medicine, Tampa, FL; Department of Community and Family Health, College of Public Health, University of South Florida, 1 Tampa General Circle, Suite F170, Tampa, FL.
| |
Collapse
|
10
|
Pierron A, Alassane-Kpembi I, Oswald IP. Impact of two mycotoxins deoxynivalenol and fumonisin on pig intestinal health. Porcine Health Manag 2016; 2:21. [PMID: 28405447 PMCID: PMC5382503 DOI: 10.1186/s40813-016-0041-2] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 07/25/2016] [Indexed: 11/16/2022] Open
Abstract
Mycotoxins are secondary metabolites of fungi that grow on a variety of substrates. Due to their high consumption of cereals and their sensitivity, pigs are highly impacted by the presence of mycotoxins. At the European level, regulations and recommendations exist for several mycotoxins in pig feed. Among these toxins, fumonisin B1 (FB1), and deoxynivalenol (DON) have a great impact on the intestine and the immune system. Indeed, the intestine is the first barrier to food contaminants and can be exposed to high concentrations of mycotoxins upon ingestion of contaminated feed. FB1 and DON alter the intestinal barrier, impair the immune response, reduce feed intake and weight gain. Their presence in feed increases the translocation of bacteria; mycotoxins can also impair the immune response and enhance the susceptibility to infectious diseases. In conclusion, because of their effect on the intestine, FB1 and DON are a major threat to pig health, welfare and performance.
Collapse
Affiliation(s)
- Alix Pierron
- ToxAlim Research Centre in Food Toxicology, INRA, UMR 1331, ENVT, INP Purpan, 180 chemin de Tournefeuille, BP93173, 31027 Toulouse, Cedex 03 France.,BIOMIN Research Center, Technopark 1, 3430 Tulln, Austria
| | - Imourana Alassane-Kpembi
- ToxAlim Research Centre in Food Toxicology, INRA, UMR 1331, ENVT, INP Purpan, 180 chemin de Tournefeuille, BP93173, 31027 Toulouse, Cedex 03 France
| | - Isabelle P Oswald
- ToxAlim Research Centre in Food Toxicology, INRA, UMR 1331, ENVT, INP Purpan, 180 chemin de Tournefeuille, BP93173, 31027 Toulouse, Cedex 03 France
| |
Collapse
|
11
|
|
12
|
Kan B, Razzaghian HR, Lavoie PM. An Immunological Perspective on Neonatal Sepsis. Trends Mol Med 2016; 22:290-302. [PMID: 26993220 PMCID: PMC5104533 DOI: 10.1016/j.molmed.2016.02.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 02/08/2016] [Indexed: 02/07/2023]
Abstract
Despite concerted international efforts, mortality from neonatal infections remains unacceptably high in some areas of the world, particularly for premature infants. Recent developments in flow cytometry and next-generation sequencing technologies have led to major discoveries over the past few years, providing a more integrated understanding of the developing human immune system in the context of its microbial environment. We review these recent findings, focusing on how in human newborns incomplete maturation of the immune system before a full term of gestation impacts on their vulnerability to infection. We also discuss some of the clinical implications of this research in guiding the design of more-accurate age-adapted diagnostic and preventive strategies for neonatal sepsis.
Collapse
Affiliation(s)
- Bernard Kan
- Child and Family Research Institute, Vancouver, British Columbia, Canada; Experimental Medicine Program, Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Hamid Reza Razzaghian
- Child and Family Research Institute, Vancouver, British Columbia, Canada; Experimental Medicine Program, Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Pascal M Lavoie
- Child and Family Research Institute, Vancouver, British Columbia, Canada; Experimental Medicine Program, Department of Medicine, University of British Columbia, Vancouver, Canada; Department of Pediatrics, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
13
|
Gökçe AM, Fındık H, Ata P, Gümrükçü G, Özel L, Gündoğdu K, Yazıcıoğlu B, Titiz MI. Relationship of Urothelial Gene Expressions in Urine-Deprived Bladders of Renal Recipients With Posttransplant Urinary Infections. Transplant Proc 2016; 47:1331-5. [PMID: 26093712 DOI: 10.1016/j.transproceed.2015.04.080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE In this study, we analyzed gene expression levels of apoptotic (Fas, FasL, Bcl-2, Bax) and survival (CXCR1, CXCR2, IL-8) signal pathways of the urine-deprived bladder tissues and the relation of urinary tract infections with these pathways. MATERIAL AND METHODS We included 37 patients admitted for renal transplantation between December 2009 and December 2012. Bladder mucosal samples were obtained at the time of transplantation and 6-8 weeks posttransplantation, at the time of ureteral catheter removal. RNA extraction and cDNA synthesis were done using guanidium-thiocyanate and colon filter methods. Expression analysis was studied with quantitative real-time polymerase chain reaction optimized with ROX dye and internal control β-actin. RESULTS According to our findings Fas, FasL, Bcl-2, and Bax expression was higher in urine-deprived bladder samples than those in the posttransplant samples (P < .05). Although Fas, FasL, Bcl-2, and Bax expression levels increased in pretransplant samples, there was an increase in posttransplant bladder samples; however, this increase was not as marked as those of pretransplant samples. IL-8, CXCR1, and CXCR2 expression was decreased at the pretransplant samples and increased in posttransplant bladder samples. CONCLUSIONS We have found an upregulated apoptotic process and decreased survival signals at the urine-deprived bladder tissue. After transplantation, bladder tissue survival parameters were increased, demonstrating the importance of urinary flow for bladder tissue.
Collapse
Affiliation(s)
- A M Gökçe
- Department of General Surgery and Transplantation, Haydarpasa Numune Training and Research Hospital, Uskudar, Istanbul, Turkey
| | - H Fındık
- Department of Molecular Genetics, Haliç University, Istanbul, Turkey
| | - P Ata
- Faculty of Medicine, Department of Medical Genetics, Marmara University, Istanbul, Turkey; Haydarpasa Numune Training and Research Hospital Tissue Typing Laboratory, Istanbul, Turkey.
| | - G Gümrükçü
- Department of General Surgery and Transplantation, Haydarpasa Numune Training and Research Hospital, Uskudar, Istanbul, Turkey
| | - L Özel
- Department of General Surgery and Transplantation, Haydarpasa Numune Training and Research Hospital, Uskudar, Istanbul, Turkey
| | - K Gündoğdu
- Department of Molecular Genetics, Haliç University, Istanbul, Turkey
| | - B Yazıcıoğlu
- Department of Molecular Genetics, Haliç University, Istanbul, Turkey
| | - M I Titiz
- Department of General Surgery and Transplantation, Haydarpasa Numune Training and Research Hospital, Uskudar, Istanbul, Turkey
| |
Collapse
|
14
|
Preterm Birth Reduces Nutrient Absorption With Limited Effect on Immune Gene Expression and Gut Colonization in Pigs. J Pediatr Gastroenterol Nutr 2015; 61:481-90. [PMID: 25883061 DOI: 10.1097/mpg.0000000000000827] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVES The primary risk factors for necrotizing enterocolitis (NEC) are preterm birth, enteral feeding, and gut colonization. It is unclear whether feeding and colonization induce excessive expression of immune genes that lead to NEC. Using a pig model, we hypothesized that reduced gestational age would upregulate immune-related genes and cause bacterial imbalance after birth. METHODS Preterm (85%-92% gestation, n = 53) and near-term (95%-99% gestation, n = 69) pigs were delivered by cesarean section and euthanized at birth or after 2 days of infant formula or bovine colostrum feeding. RESULTS At birth, preterm delivery reduced 5 of 30 intestinal genes related to nutrient absorption and innate immunity, relative to near-term pigs, whereas 2 genes were upregulated. Preterm birth also reduced ex vivo intestinal glucose and leucine uptake (40%-50%), but failed to increase cytokine secretions from intestinal explants relative to near-term birth. After 2 days of formula feeding, NEC incidence was increased in preterm versus near-term pigs (47% vs 0%-13%). A total of 6 of the 30 genes related to immunity (TLR2, IL1B, and IL8), permeability (CLDN3, and OCLN), and absorption (SGLT) decreased in preterm pigs without affecting Gram-negative bacteria-related responses (TLR4, IKBA, NFkB1, TNFAIP3, and PAFA). Bacterial abundance tended to be higher in preterm versus near-term pigs (P = 0.09), whereas the composition was unaffected. CONCLUSIONS Preterm birth predisposes to NEC and reduces nutrient absorption but does not induce upregulation of immune-related genes or cause bacterial dyscolonization in the neonatal period. Excessive inflammation and bacterial overgrowth may occur relatively late in NEC progression in preterm neonates.
Collapse
|
15
|
Abstract
Necrotizing enterocolitis (NEC) is a leading cause of mortality in preterm infants. This article reviews the immunologic and hematological abnormalities typically seen in infants with NEC, such as elevated plasma cytokine levels, thrombocytopenia, increased or decreased neutrophil counts, low monocyte counts, and anemia. Some of these findings may provide important diagnostic and prognostic information.
Collapse
Affiliation(s)
- Akhil Maheshwari
- Department of Pediatrics, Morsani College of Medicine, Tampa, FL 33606, USA; Department of Molecular Medicine, Morsani College of Medicine, Tampa, FL 33612, USA; Department of Community and Family Health, College of Public Health, University of South Florida, Tampa, FL 33612, USA.
| |
Collapse
|
16
|
Menckeberg CL, Hol J, Simons-Oosterhuis Y, Raatgeep HRC, de Ruiter LF, Lindenbergh-Kortleve DJ, Korteland-van Male AM, El Aidy S, van Lierop PPE, Kleerebezem M, Groeneweg M, Kraal G, Elink-Schuurman BE, de Jongste JC, Nieuwenhuis EES, Samsom JN. Human buccal epithelium acquires microbial hyporesponsiveness at birth, a role for secretory leukocyte protease inhibitor. Gut 2015; 64:884-93. [PMID: 25056659 DOI: 10.1136/gutjnl-2013-306149] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 06/29/2014] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Repetitive interaction with microbial stimuli renders epithelial cells (ECs) hyporesponsive to microbial stimulation. Previously, we have reported that buccal ECs from a subset of paediatric patients with Crohn's disease are not hyporesponsive and spontaneously released chemokines. We now aimed to identify kinetics and mechanisms of acquisition of hyporesponsiveness to microbial stimulation using primary human buccal epithelium. DESIGN Buccal ECs collected directly after birth and in later stages of life were investigated. Chemokine release and regulatory signalling pathways were studied using primary buccal ECs and the buccal EC line TR146. Findings were extended to the intestinal mucosa using murine model systems. RESULTS Directly after birth, primary human buccal ECs spontaneously produced the chemokine CXCL-8 and were responsive to microbial stimuli. Within the first weeks of life, these ECs attained hyporesponsiveness, associated with inactivation of the NF-κB pathway and upregulation of the novel NF-κB inhibitor SLPI but no other known NF-κB inhibitors. SLPI protein was abundant in the cytoplasm and the nucleus of hyporesponsive buccal ECs. Knock-down of SLPI in TR146-buccal ECs induced loss of hyporesponsiveness with increased NF-κB activation and subsequent chemokine release. This regulatory mechanism extended to the intestine, as colonisation of germfree mice elicited SLPI expression in small intestine and colon. Moreover, SLPI-deficient mice had increased chemokine expression in small intestinal and colonic ECs. CONCLUSIONS We identify SLPI as a new player in acquisition of microbial hyporesponsiveness by buccal and intestinal epithelium in the first weeks after microbial colonisation.
Collapse
Affiliation(s)
- Celia L Menckeberg
- Laboratory of Pediatrics, Division of Gastroenterology and Nutrition, Erasmus MC, Rotterdam, The Netherlands
| | - Jeroen Hol
- Laboratory of Pediatrics, Division of Gastroenterology and Nutrition, Erasmus MC, Rotterdam, The Netherlands Department of Pulmonary Diseases, Sophia Children's Hospital, Rotterdam, The Netherlands Department of Pediatrics, University Hospital Ghent, Ghent, Belgium
| | - Ytje Simons-Oosterhuis
- Laboratory of Pediatrics, Division of Gastroenterology and Nutrition, Erasmus MC, Rotterdam, The Netherlands
| | - H Rolien C Raatgeep
- Laboratory of Pediatrics, Division of Gastroenterology and Nutrition, Erasmus MC, Rotterdam, The Netherlands
| | - Lilian F de Ruiter
- Laboratory of Pediatrics, Division of Gastroenterology and Nutrition, Erasmus MC, Rotterdam, The Netherlands Department of Pulmonary Diseases, Sophia Children's Hospital, Rotterdam, The Netherlands
| | | | - Anita M Korteland-van Male
- Laboratory of Pediatrics, Division of Gastroenterology and Nutrition, Erasmus MC, Rotterdam, The Netherlands
| | - Sahar El Aidy
- Laboratory of Microbiology and Host Microbe Interactomics Group, Wageningen University, Wageningen, The Netherlands Department of Industrial Biotechnology, GEBRI, Sadat City University, Sadat City, Egypt
| | - Pieter P E van Lierop
- Laboratory of Pediatrics, Division of Gastroenterology and Nutrition, Erasmus MC, Rotterdam, The Netherlands
| | - Michiel Kleerebezem
- Laboratory of Microbiology and Host Microbe Interactomics Group, Wageningen University, Wageningen, The Netherlands
| | - Michael Groeneweg
- Department of Pediatrics, Maasstad Hospital, Rotterdam, The Netherlands
| | - Georg Kraal
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Johan C de Jongste
- Department of Pulmonary Diseases, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Edward E S Nieuwenhuis
- Laboratory of Pediatrics, Division of Gastroenterology and Nutrition, Erasmus MC, Rotterdam, The Netherlands Wilhelmina Children's Hospital, Utrecht, The Netherlands
| | - Janneke N Samsom
- Laboratory of Pediatrics, Division of Gastroenterology and Nutrition, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
17
|
Nguyen DN, Sangild PT, Ostergaard MV, Bering SB, Chatterton DEW. Transforming growth factor-β2 and endotoxin interact to regulate homeostasis via interleukin-8 levels in the immature intestine. Am J Physiol Gastrointest Liver Physiol 2014; 307:G689-99. [PMID: 25147235 DOI: 10.1152/ajpgi.00193.2014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
A balance between pro- and anti-inflammatory signals from milk and microbiota controls intestinal homeostasis just after birth, and an optimal balance is particularly important for preterm neonates that are sensitive to necrotizing enterocolitis (NEC). We suggest that the intestinal cytokine IL-8 plays an important role and hypothesize that transforming growth factor-β2 (TGF-β2) acts in synergy with bacterial lipopolysaccharide (LPS) to control IL-8 levels, thereby supporting intestinal homeostasis. Preterm pigs were fed colostrum (containing TGF-β2) or infant formula (IF) with or without antibiotics (COLOS, n = 27; ANTI, n = 11; IF, n = 40). Intestinal IL-8 levels and NEC incidence were much higher in IF than in COLOS and ANTI pigs (P < 0.001), but IL-8 levels did not correlate with NEC severity. Intestinal TGF-β2 levels were high in COLOS but low in IF and ANTI pigs. Based on these observations, the interplay among IL-8, TGF-β2, and LPS was investigated in a porcine intestinal epithelial cell line. TGF-β2 attenuated LPS-induced IL-6, IL-1β, and TNF-α release by reducing early ERK activation, whereas IL-8 secretion was synergistically induced by LPS and TGF-β2 via NF-κB. The TGF-β2/LPS-induced IL-8 levels stimulated cell proliferation and migration following epithelial injury, without continuous NF-κB activation and cyclooxygenase-2 expression. We suggest that a combined TGF-β2-LPS induction of IL-8 stimulates epithelial repair just after birth when the intestine is first exposed to colonizing bacteria and TGF-β2-containing milk. Moderate IL-8 levels may act to control intestinal inflammation, whereas excessive IL-8 production may enhance the damaging proinflammatory cascade leading to NEC.
Collapse
Affiliation(s)
- Duc Ninh Nguyen
- Faculty of Science, Department of Food Science, University of Copenhagen, Copenhagen, Denmark; and
| | - Per T Sangild
- Faculty of Science, Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Mette V Ostergaard
- Faculty of Science, Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Stine B Bering
- Faculty of Science, Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Dereck E W Chatterton
- Faculty of Science, Department of Food Science, University of Copenhagen, Copenhagen, Denmark; and Faculty of Science, Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
18
|
Liu M, Gao R, Meng Q, Zhang Y, Bi C, Shan A. Toxic effects of maternal zearalenone exposure on intestinal oxidative stress, barrier function, immunological and morphological changes in rats. PLoS One 2014; 9:e106412. [PMID: 25180673 PMCID: PMC4152245 DOI: 10.1371/journal.pone.0106412] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 08/06/2014] [Indexed: 01/25/2023] Open
Abstract
The present study was conducted to investigate the effects of maternal zearalenone (ZEN) exposure on the intestine of pregnant Sprague-Dawley (SD) rats and its offspring. Ninety-six pregnant SD rats were randomly divided into four groups and were fed with diets containing ZEN at concentrations of 0.3 mg/kg, 48.5 mg/kg, 97.6 mg/kg or 146.0 mg/kg from gestation days (GD) 1 to 7. All rats were fed with mycotoxin-free diet until their offspring were weaned at three weeks of age. The small intestinal fragments from pregnant rats at GD8, weaned dams and pups were collected and studied for toxic effects of ZEN on antioxidant status, immune response, expression of junction proteins, and morphology. The results showed that ZEN induced oxidative stress, affected the villous structure and reduced the expression of junction proteins claudin-4, occludin and connexin43 (Cx43) in a dose-dependent manner in pregnant rats. Different effects on the expression of cytokines were also observed both in mRNA and protein levels in these pregnant groups. Ingestion of high levels of ZEN caused irreversible damage in weaned dams, such as oxidative stress, decreased villi hight and low expression of junction proteins and cytokines. Decreased expression of jejunal interleukin-8 (IL-8) and increased expression of gastrointestinal glutathione peroxidase (GPx2) mRNA were detected in weaned offspring, indicating long-term damage caused by maternal ZEN. We also found that the Nrf2 expression both in mRNA and protein levels were up-regulated in the ZEN-treated groups of pregnant dams and the high-dose of ZEN group of weaned dams. The data indicate that modulation of Nrf2-mediated pathway is one of mechanism via which ZEN affects gut wall antioxidant and inflammatory responses.
Collapse
Affiliation(s)
- Min Liu
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, P. R. China
| | - Rui Gao
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, P. R. China
| | - Qingwei Meng
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, P. R. China
| | - Yuanyuan Zhang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, P. R. China
| | - Chongpeng Bi
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, P. R. China
| | - Anshan Shan
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, P. R. China
| |
Collapse
|
19
|
Cytokines associated with necrotizing enterocolitis in extremely-low-birth-weight infants. Pediatr Res 2014; 76:100-8. [PMID: 24732104 PMCID: PMC4062583 DOI: 10.1038/pr.2014.48] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Accepted: 01/01/2014] [Indexed: 11/09/2022]
Abstract
BACKGROUND The goal was to identify cytokines associated with necrotizing enterocolitis (NEC). Based on our earlier reports of decreased tissue expression of transforming growth factor (TGF)-β, we hypothesized that infants with NEC also have low blood TGF-β levels. We further hypothesized that because fetal inflammation increases the risk of NEC, infants who develop NEC have elevated blood cytokine levels in early neonatal period. METHODS Data on 104 extremely-low-birth-weight infants with NEC and 893 without NEC from 17 centers were analyzed. Clinical information was correlated with blood cytokine levels on postnatal day 1 (D1), D3, D7, D14, and D21. RESULTS Male gender, non-Caucasian/non-African American ethnicity, sepsis, lower blood TGF-β and interleukin (IL)-2 levels, and higher IL-8 levels were associated with NEC. The NEC group had lower TGF-β levels than controls since D1. The diagnosis of NEC was associated with elevated IL-1β, IL-6, IL-8, IL-10, monocyte chemoattractant protein-1/CC-motif ligand-2, macrophage inflammatory protein-1β/CC-motif ligand-3, and C-reactive protein. CONCLUSION Clinical characteristics, such as gender and ethnicity, and low blood TGF-β levels are associated with higher risk of NEC. Infants who developed NEC did not start with high blood levels of inflammatory cytokines, but these rose mainly after the onset of NEC.
Collapse
|
20
|
Pinton P, Oswald IP. Effect of deoxynivalenol and other Type B trichothecenes on the intestine: a review. Toxins (Basel) 2014; 6:1615-43. [PMID: 24859243 PMCID: PMC4052256 DOI: 10.3390/toxins6051615] [Citation(s) in RCA: 249] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 03/28/2014] [Accepted: 05/09/2014] [Indexed: 12/23/2022] Open
Abstract
The natural food contaminants, mycotoxins, are regarded as an important risk factor for human and animal health, as up to 25% of the world's crop production may be contaminated. The Fusarium genus produces large quantities of fusariotoxins, among which the trichothecenes are considered as a ubiquitous problem worldwide. The gastrointestinal tract is the first physiological barrier against food contaminants, as well as the first target for these toxicants. An increasing number of studies suggest that intestinal epithelial cells are targets for deoxynivalenol (DON) and other Type B trichothecenes (TCTB). In humans, various adverse digestive symptoms are observed on acute exposure, and in animals, these toxins induce pathological lesions, including necrosis of the intestinal epithelium. They affect the integrity of the intestinal epithelium through alterations in cell morphology and differentiation and in the barrier function. Moreover, DON and TCTB modulate the activity of intestinal epithelium in its role in immune responsiveness. TCTB affect cytokine production by intestinal or immune cells and are supposed to interfere with the cross-talk between epithelial cells and other intestinal immune cells. This review summarizes our current knowledge of the effects of DON and other TCTB on the intestine.
Collapse
Affiliation(s)
- Philippe Pinton
- INRA (Institut National de la Recherche Agronomique), UMR1331, Toxalim, Research Centre in Food Toxicology, Toulouse F-31027, France.
| | - Isabelle P Oswald
- INRA (Institut National de la Recherche Agronomique), UMR1331, Toxalim, Research Centre in Food Toxicology, Toulouse F-31027, France.
| |
Collapse
|
21
|
Lo MC, Yip TC, Ngan KC, Cheng WW, Law CK, Chan PS, Chan KC, Wong CKC, Wong RNS, Lo KW, Ng WT, Lee WM, Tsao SW, Kwong LW, Lung ML, Mak NK. Role of MIF/CXCL8/CXCR2 signaling in the growth of nasopharyngeal carcinoma tumor spheres. Cancer Lett 2013; 335:81-92. [DOI: 10.1016/j.canlet.2013.01.052] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 01/30/2013] [Accepted: 01/31/2013] [Indexed: 10/27/2022]
|
22
|
Abstract
This article provides an overview of the composition of human milk, its variation, and its clinical relevance. The composition of human milk is the biological norm for infant nutrition. Human milk also contains many hundreds to thousands of distinct bioactive molecules that protect against infection and inflammation and contribute to immune maturation, organ development, and healthy microbial colonization. Some of these molecules (eg, lactoferrin) are being investigated as novel therapeutic agents. Human milk changes in composition from colostrum to late lactation, within feeds, by gestational age, diurnally, and between mothers. Feeding infants with expressed human milk is increasing.
Collapse
Affiliation(s)
- Olivia Ballard
- Center for Interdisciplinary Research in Human Milk and Lactation & Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave., MLC 7009, Cincinnati, OH 45229.
| | - Ardythe L. Morrow
- Center for Interdisciplinary Research in Human Milk and Lactation, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave., MLC 7009, Cincinnati, OH 45229.
| |
Collapse
|
23
|
MohanKumar K, Kaza N, Jagadeeswaran R, Garzon SA, Bansal A, Kurundkar AR, Namachivayam K, Remon JI, Bandepalli CR, Feng X, Weitkamp JH, Maheshwari A. Gut mucosal injury in neonates is marked by macrophage infiltration in contrast to pleomorphic infiltrates in adult: evidence from an animal model. Am J Physiol Gastrointest Liver Physiol 2012; 303:G93-102. [PMID: 22538401 PMCID: PMC3404576 DOI: 10.1152/ajpgi.00016.2012] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Necrotizing enterocolitis (NEC) is an inflammatory bowel necrosis of premature infants. In tissue samples of NEC, we identified numerous macrophages and a few neutrophils but not many lymphocytes. We hypothesized that these pathoanatomic characteristics of NEC represent a common tissue injury response of the gastrointestinal tract to a variety of insults at a specific stage of gut development. To evaluate developmental changes in mucosal inflammatory response, we used trinitrobenzene sulfonic acid (TNBS)-induced inflammation as a nonspecific insult and compared mucosal injury in newborn vs. adult mice. Enterocolitis was induced in 10-day-old pups and adult mice (n = 25 animals per group) by administering TNBS by gavage and enema. Leukocyte populations were enumerated in human NEC and in murine TNBS-enterocolitis using quantitative immunofluorescence. Chemokine expression was measured using quantitative polymerase chain reaction, immunoblots, and immunohistochemistry. Macrophage recruitment was investigated ex vivo using intestinal tissue-conditioned media and bone marrow-derived macrophages in a microchemotaxis assay. Similar to human NEC, TNBS enterocolitis in pups was marked by a macrophage-rich leukocyte infiltrate in affected tissue. In contrast, TNBS-enterocolitis in adult mice was associated with pleomorphic leukocyte infiltrates. Macrophage precursors were recruited to murine neonatal gastrointestinal tract by the chemokine CXCL5, a known chemoattractant for myeloid cells. We also demonstrated increased expression of CXCL5 in surgically resected tissue samples of human NEC, indicating that a similar pathway was active in NEC. We concluded that gut mucosal injury in the murine neonate is marked by a macrophage-rich leukocyte infiltrate, which contrasts with the pleomorphic leukocyte infiltrates in adult mice. In murine neonatal enterocolitis, macrophages were recruited to the inflamed gut mucosa by the chemokine CXCL5, indicating that CXCL5 and its cognate receptor CXCR2 merit further investigation as potential therapeutic targets in NEC.
Collapse
Affiliation(s)
- Krishnan MohanKumar
- Departments of Pediatrics, 1Division of Neonatology and ,2Center for Neonatal and Pediatric Gastrointestinal Disease, ,Departments of 3Pediatrics and
| | - Niroop Kaza
- 4Pathology, University of Alabama at Birmingham, Birmingham, Alabama;
| | - Ramasamy Jagadeeswaran
- Departments of Pediatrics, 1Division of Neonatology and ,2Center for Neonatal and Pediatric Gastrointestinal Disease,
| | | | | | | | - Kopperuncholan Namachivayam
- Departments of Pediatrics, 1Division of Neonatology and ,2Center for Neonatal and Pediatric Gastrointestinal Disease,
| | - Juan I. Remon
- Departments of Pediatrics, 1Division of Neonatology and ,2Center for Neonatal and Pediatric Gastrointestinal Disease,
| | - C. Rekha Bandepalli
- Departments of Pediatrics, 1Division of Neonatology and ,2Center for Neonatal and Pediatric Gastrointestinal Disease,
| | - Xu Feng
- 4Pathology, University of Alabama at Birmingham, Birmingham, Alabama;
| | - Joern-Hendrik Weitkamp
- 6Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Akhil Maheshwari
- Departments of Pediatrics, 1Division of Neonatology and ,2Center for Neonatal and Pediatric Gastrointestinal Disease, ,7Pharmacology, University of Illinois at Chicago, Chicago, Illinois; ,Departments of 3Pediatrics and
| |
Collapse
|
24
|
Kelland EE, Gilmore W, Weiner LP, Lund BT. The dual role of CXCL8 in human CNS stem cell function: Multipotent neural stem cell death and oligodendrocyte progenitor cell chemotaxis. Glia 2011; 59:1864-78. [DOI: 10.1002/glia.21230] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Accepted: 07/20/2011] [Indexed: 12/16/2022]
|
25
|
Tseng-Rogenski S, Liebert M. Interleukin-8 is essential for normal urothelial cell survival. Am J Physiol Renal Physiol 2009; 297:F816-21. [PMID: 19535567 PMCID: PMC2739718 DOI: 10.1152/ajprenal.90733.2008] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2008] [Accepted: 06/11/2009] [Indexed: 11/22/2022] Open
Abstract
Interleukin-8 (IL-8; CXCL8) has been shown to play a role in multiple cellular processes. Here, we report an additional role of IL-8 as a growth and essential survival factor for normal human urothelial cells. Supplementing exogenous recombinant human IL-8 to normal urothelial cells promoted cell growth through the Akt pathway. Inhibition of IL-8 expression by small inhibitory RNA (siRNA) caused normal urothelial cells to die. Addition of recombinant human IL-8 rescued the normal urothelial cells treated with IL-8 siRNA. This rescue effect could be blocked by antibodies to the IL-8 receptor CXCR1 but not by CXCR2, suggesting that normal urothelial cells normally have IL-8 autocrine or paracrine activity for survival and growth mediated by CXCR1. IL-8 mRNA levels were lower in samples from patients with interstitial cystitis, a urinary bladder disorder associated with urothelial cell dysfunction and/or loss. Taken together, these results suggest that IL-8 is an important normal urothelial growth factor and is necessary for normal urothelial cell survival in vitro and in vivo. Lower IL-8 expression levels in the urinary bladder may contribute to pathophysiology of interstitial cystitis.
Collapse
|
26
|
Maheshwari A, Voitenok NN, Akalovich S, Shaik SS, Randolph DA, Sims B, Patel RP, Killingsworth CR, Fallon MB, Ohls RK. Developmental changes in circulating IL-8/CXCL8 isoforms in neonates. Cytokine 2009; 46:12-6. [PMID: 19232500 DOI: 10.1016/j.cyto.2008.12.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2008] [Revised: 11/20/2008] [Accepted: 12/29/2008] [Indexed: 01/21/2023]
Abstract
Interleukin-8 (IL-8/CXCL8) is widely expressed in fetal tissues although inflammatory changes are not seen. Circulating IL-8 is comprised of an endothelial-derived [ala-IL-8](77) isoform and another, more potent [ser-IL-8](72) secreted by most other cells; [ala-IL-8](77) can be converted into [ser-IL-8](72) by proteolytic removal of an N-terminal pentapeptide from [ala-IL-8](77). In this study, we show [ala-IL-8](77) is the predominant circulating isoform of IL-8 in premature neonates but not in term neonates/adults, who have [ser-IL-8](72) as the major isoform. This isoform switch from the less potent [ala-IL-8](77) to [ser-IL-8](72) correlates with a maturational increase in the neutrophil chemotactic potency of plasma IL-8. The emergence of [ser-IL-8](72) as the major isoform is likely due to increased plasma [ala-IL-8](77)-convertase activity and/or changes in the cellular sources of IL-8. Developmental changes in IL-8 isoforms may serve to minimize its inflammatory effects in the fetus and also provide a mechanism to restore its full activity after birth.
Collapse
Affiliation(s)
- Akhil Maheshwari
- Department of Pediatrics, University of Alabama, Birmingham, AL 35294, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
McPherson RJ, Juul SE. High-dose erythropoietin inhibits apoptosis and stimulates proliferation in neonatal rat intestine. Growth Horm IGF Res 2007; 17:424-430. [PMID: 17632025 DOI: 10.1016/j.ghir.2007.05.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2007] [Revised: 05/30/2007] [Accepted: 05/31/2007] [Indexed: 11/18/2022]
Abstract
BACKGROUND Erythropoietin (Epo) receptors are widely expressed in the small bowel of neonatal rats and evidence suggests Epo has important trophic effects in developing bowel. OBJECTIVE To compliment in vitro data, we directly examine in vivo the hypotheses that systemic Epo treatment can promote cell division and enterocyte migration, and arrest apoptosis in the ileum of neonatal rats. DESIGN Epo (5000 U/kg s.c.) or vehicle treatments were given to one week old Sprague-Dawley rats (n = 86) along with timed injections of the thymidine analog 5-bromo-2-deoxyuridine (BrdU, 50mg/kg s.c.) to label DNA synthesis and track newly proliferating cells. To characterize the time course of effects, animals were killed at scheduled times from 30 min to 24 h after treatment. BrdU-containing cells were immunostained and counted in intestinal crypts, villi, and muscle wall of ileum. Effects of Epo on apoptosis were analyzed by TUNEL staining. Calibrated measurements were made to determine the density or relative proportion of BrdU- and TUNEL-positive cells. RESULTS Systemic high-dose Epo promoted cell division in intestinal smooth muscle and enterocytes, stimulated migration of intestinal epithelial cells, and arrested apoptosis of enterocytes at the villous tips. CONCLUSION These data provide in vivo evidence that Epo functions trophically in developing intestine tissues.
Collapse
|
28
|
Bouhet S, Le Dorze E, Peres S, Fairbrother JM, Oswald IP. Mycotoxin fumonisin B1 selectively down-regulates the basal IL-8 expression in pig intestine: in vivo and in vitro studies. Food Chem Toxicol 2006; 44:1768-73. [PMID: 16843581 DOI: 10.1016/j.fct.2006.05.018] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2005] [Revised: 04/19/2006] [Accepted: 05/25/2006] [Indexed: 12/01/2022]
Abstract
Fumonisin B(1) (FB(1)) is a mycotoxin produced by Fusarium verticillioides and F. proliferatum, common contaminants of maize. FB(1) causes toxicological effects in laboratory and domestic animals including pigs. The gastrointestinal tract represents a barrier encountered by exogenous food compounds. The purpose of our study was to determine FB(1) effects on intestinal immune response. Nine recently weaned piglets orally received 0.5 mg of purified toxin/kg of body weight/day for 7 days, while eight other animals were kept as controls. After necropsy, ileal samples were analyzed for five pro-inflammatory cytokines mRNA expression by RT-PCR. No difference was observed for IL-1beta, IL-6, IL-12 and TNF-beta mRNA levels between control and FB(1)-treated animals. In contrast, FB(1) treatment induced a significant down-regulation of the expression of IL-8 mRNA in the pig ilea. The effect of FB(1) on the IL-8 expression was also examined in the porcine intestinal epithelial cell line IPEC-1. FB(1) decreases the expression of IL-8, both at the mRNA and protein levels, in a dose-dependant manner. Taken together, our data demonstrate that FB(1) alters the intestinal immune response by decreasing the level of IL-8. This may contribute to the increased intestinal colonization by pathogenic Escherichia coli that was observed in FB(1)-treated pigs and may have implications for humans/animals consuming FB(1)-contaminated food/feed.
Collapse
Affiliation(s)
- Sandrine Bouhet
- Laboratoire de Pharmacologie-Toxicologie, Institut National de Recherche Agronomique, 31931 Toulouse, France.
| | | | | | | | | |
Collapse
|
29
|
Maheshwari A, Lu W, Guida WC, Christensen RD, Calhoun DA. IL-8/CXC ligand 8 survives neonatal gastric digestion as a result of intrinsic aspartyl proteinase resistance. Pediatr Res 2005; 57:438-44. [PMID: 15585673 DOI: 10.1203/01.pdr.0000151317.08180.7e] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The human fetus and neonate swallow biologically significant quantities of IL-8/CXC ligand 8 (CXCL8) in amniotic fluid and breast milk, and this remains measurable through simulated neonatal gastric and proximal intestinal digestions. We sought to confirm the structural and functional integrity of IL-8/CXCL8 in digestates and determine the mechanisms underlying this protease resistance. We observed that in comparison with BSA, IL-8/CXCL8 is highly resistant to pepsin and can be detected intact in assays for structural, immunologic, and functional integrity. In a computational molecular docking simulation, IL-8/CXCL8 was observed to fit poorly in the pepsin active site. On the basis of simulated mutation analyses, we hypothesized that this protease resistance is due to disulfide bond-related tertiary folding in IL-8/CXCL8. This was confirmed on chemical reduction of these groups.
Collapse
Affiliation(s)
- Akhil Maheshwari
- Division of Neonatology, Deparment of Pediatrics, University of South Florida College of Medicine and All Children's Hospital, St. Petersburg, FL 33701, USA.
| | | | | | | | | |
Collapse
|
30
|
Abstract
Villous development of the intestine is beginning to be understood in terms of the underlying molecular mechanisms. There is increasing information on the role of cytokines as extrinsic regulators in this process. This article summarizes information available on various cytokines that have been studied in this context.
Collapse
Affiliation(s)
- Akhil Maheshwari
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Alabama at Birmingham, NHB 525, 619 19th Street South, Birmingham, AL 35233, USA.
| |
Collapse
|