1
|
Halpern MD, Gupta A, Zaghloul N, Thulasingam S, Calton CM, Camp SM, Garcia JGN, Ahmed M. Extracellular Nicotinamide Phosphoribosyltransferase Is a Therapeutic Target in Experimental Necrotizing Enterocolitis. Biomedicines 2024; 12:970. [PMID: 38790933 PMCID: PMC11118767 DOI: 10.3390/biomedicines12050970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/21/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
Necrotizing enterocolitis (NEC) is the most common gastrointestinal emergency of prematurity. Postulated mechanisms leading to inflammatory necrosis of the ileum and colon include activation of the pathogen recognition receptor Toll-like receptor 4 (TLR4) and decreased levels of transforming growth factor beta (TGFβ). Extracellular nicotinamide phosphoribosyltransferase (eNAMPT), a novel damage-associated molecular pattern (DAMP), is a TLR4 ligand and plays a role in a number of inflammatory disease processes. To test the hypothesis that eNAMPT is involved in NEC, an eNAMPT-neutralizing monoclonal antibody, ALT-100, was used in a well-established animal model of NEC. Preterm Sprague-Dawley pups delivered prematurely from timed-pregnant dams were exposed to hypoxia/hypothermia and randomized to control-foster mother dam-fed rats, injected IP with saline (vehicle) 48 h after delivery; control + mAB-foster dam-fed rats, injected IP with 10 µg of ALT-100 at 48 h post-delivery; NEC-orally gavaged, formula-fed rats injected with saline; and NEC + mAb-formula-fed rats, injected IP with 10 µg of ALT-100 at 48 h. The distal ileum was processed 96 h after C-section delivery for histological, biochemical, molecular, and RNA sequencing studies. Saline-treated NEC pups exhibited markedly increased fecal blood and histologic ileal damage compared to controls (q < 0.0001), and findings significantly reduced in ALT-100 mAb-treated NEC pups (q < 0.01). Real-time PCR in ileal tissues revealed increased NAMPT in NEC pups compared to pups that received the ALT-100 mAb (p < 0.01). Elevated serum levels of tumor necrosis factor alpha (TNFα), interleukin 6 (IL-6), interleukin-8 (IL-8), and NAMPT were observed in NEC pups compared to NEC + mAb pups (p < 0.01). Finally, RNA-Seq confirmed dysregulated TGFβ and TLR4 signaling pathways in NEC pups that were attenuated by ALT-100 mAb treatment. These data strongly support the involvement of eNAMPT in NEC pathobiology and eNAMPT neutralization as a strategy to address the unmet need for NEC therapeutics.
Collapse
Affiliation(s)
- Melissa D. Halpern
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Akash Gupta
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Nahla Zaghloul
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Senthilkumar Thulasingam
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Christine M. Calton
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Sara M. Camp
- Center for Inflammation Science and Systems Medicine, University of Florida Scripps Research Institute, Jupiter, FL 33458, USA (J.G.N.G.)
| | - Joe G. N. Garcia
- Center for Inflammation Science and Systems Medicine, University of Florida Scripps Research Institute, Jupiter, FL 33458, USA (J.G.N.G.)
| | - Mohamed Ahmed
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
2
|
Calton CM, Carothers K, Ramamurthy S, Jagadish N, Phanindra B, Garcia A, Viswanathan VK, Halpern MD. Clostridium scindens exacerbates experimental necrotizing enterocolitis via upregulation of the apical sodium-dependent bile acid transporter. Am J Physiol Gastrointest Liver Physiol 2024; 326:G25-G37. [PMID: 37933481 PMCID: PMC11208032 DOI: 10.1152/ajpgi.00102.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 10/19/2023] [Accepted: 11/01/2023] [Indexed: 11/08/2023]
Abstract
Necrotizing enterocolitis (NEC) is the most common gastrointestinal emergency in premature infants. Evidence indicates that bile acid homeostasis is disrupted during NEC: ileal bile acid levels are elevated in animals with experimental NEC, as is expression of the apical sodium-dependent bile acid transporter (Asbt). In addition, bile acids, which are synthesized in the liver, are extensively modified by the gut microbiome, including via the conversion of primary bile acids to more cytotoxic secondary forms. We hypothesized that the addition of bile acid-modifying bacteria would increase susceptibility to NEC in a neonatal rat model of the disease. The secondary bile acid-producing species Clostridium scindens exacerbated both incidence and severity of NEC. C. scindens upregulated the bile acid transporter Asbt and increased levels of intraenterocyte bile acids. Treatment with C. scindens also altered bile acid profiles and increased hydrophobicity of the ileal intracellular bile acid pool. The ability of C. scindens to enhance NEC requires bile acids, as pharmacological sequestration of ileal bile acids protects animals from developing disease. These findings indicate that bile acid-modifying bacteria can contribute to NEC pathology and provide additional evidence for the role of bile acids in the pathophysiology of experimental NEC.NEW & NOTEWORTHY Necrotizing enterocolitis (NEC), a life-threatening gastrointestinal emergency in premature infants, is characterized by dysregulation of bile acid homeostasis. We demonstrate that administering the secondary bile acid-producing bacterium Clostridium scindens enhances NEC in a neonatal rat model of the disease. C. scindens-enhanced NEC is dependent on bile acids and driven by upregulation of the ileal bile acid transporter Asbt. This is the first report of bile acid-modifying bacteria exacerbating experimental NEC pathology.
Collapse
Affiliation(s)
- Christine M Calton
- Department of Pediatrics and Steele Children's Research Center, University of Arizona, Tucson, Arizona, United States
| | - Katelyn Carothers
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona, United States
| | - Shylaja Ramamurthy
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona, United States
| | - Neha Jagadish
- Department of Pediatrics and Steele Children's Research Center, University of Arizona, Tucson, Arizona, United States
| | - Bhumika Phanindra
- Department of Pediatrics and Steele Children's Research Center, University of Arizona, Tucson, Arizona, United States
| | - Anett Garcia
- Department of Pediatrics and Steele Children's Research Center, University of Arizona, Tucson, Arizona, United States
| | - V K Viswanathan
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona, United States
| | - Melissa D Halpern
- Department of Pediatrics and Steele Children's Research Center, University of Arizona, Tucson, Arizona, United States
| |
Collapse
|
3
|
Yang J, Chen X, Liu T, Shi Y. Potential role of bile acids in the pathogenesis of necrotizing enterocolitis. Life Sci 2024; 336:122279. [PMID: 37995935 DOI: 10.1016/j.lfs.2023.122279] [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: 09/01/2023] [Revised: 11/13/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023]
Abstract
Necrotizing enterocolitis (NEC) is one of the most common acute gastrointestinal diseases in preterm infants. Recent studies have found that NEC is not only caused by changes in the intestinal environment but also by the failure of multiple systems and organs, including the liver. The accumulation of bile acids (BAs) in the ileum and the disorder of ileal BA transporters are related to the ileum injury of NEC. Inflammatory factors such as tumor necrosis factor (TNF)-α and interleukin (IL)-18 secreted by NEC also play an important role in regulating intrahepatic BA transporters. As an important link connecting the liver and intestinal circulation, the bile acid metabolic pathway plays an important role in the regulation of intestinal microbiota, cell proliferation, and barrier protection. In this review, we focus on how bile acids explore the dynamic changes of bile acid metabolism in necrotizing enterocolitis and the potential therapeutic value of targeting the bile acid signaling pathways.
Collapse
Affiliation(s)
- Jiahui Yang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Xiaoyu Chen
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Tianjing Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Yongyan Shi
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
4
|
Wei J, Meng Z, Li Z, Dang D, Wu H. New insights into intestinal macrophages in necrotizing enterocolitis: the multi-functional role and promising therapeutic application. Front Immunol 2023; 14:1261010. [PMID: 37841247 PMCID: PMC10568316 DOI: 10.3389/fimmu.2023.1261010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/13/2023] [Indexed: 10/17/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is an inflammatory intestinal disease that profoundly affects preterm infants. Currently, the pathogenesis of NEC remains controversial, resulting in limited treatment strategies. The preterm infants are thought to be susceptible to gut inflammatory disorders because of their immature immune system. In early life, intestinal macrophages (IMφs), crucial components of innate immunity, demonstrate functional plasticity and diversity in intestinal development, resistance to pathogens, maintenance of the intestinal barrier, and regulation of gut microbiota. When the stimulations of environmental, dietary, and bacterial factors interrupt the homeostatic processes of IMφs, they will lead to intestinal disease, such as NEC. This review focuses on the IMφs related pathogenesis in NEC, discusses the multi-functional roles and relevant molecular mechanisms of IMφs in preterm infants, and explores promising therapeutic application for NEC.
Collapse
Affiliation(s)
- Jiaqi Wei
- Department of Neonatology, First Hospital of Jilin University, Changchun, China
| | - Zhaoli Meng
- Department of Translational Medicine Research Institute, First Hospital of Jilin University, Changchun, China
| | - Zhenyu Li
- Department of Neonatology, First Hospital of Jilin University, Changchun, China
| | - Dan Dang
- Department of Neonatology, First Hospital of Jilin University, Changchun, China
| | - Hui Wu
- Department of Neonatology, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
5
|
Wang Z, Chong Q, Zhou J, Gao T, Zhu K, Gong X, Sheng Q, Lv Z. Reduction of absolute monocyte counts is associated with the severity of preterm necrotizing enterocolitis. J Pediatr (Rio J) 2023; 99:449-455. [PMID: 37015323 PMCID: PMC10492144 DOI: 10.1016/j.jped.2023.02.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/18/2023] [Accepted: 02/23/2023] [Indexed: 04/06/2023] Open
Abstract
OBJECTIVE Necrotizing enterocolitis (NEC) is characterized by a rich infiltration of macrophages in the intestines, which is derived from monocytes in the blood. The authors aimed to explore the changing trend of absolute monocyte counts (AMC) over time in NEC infants and to verify whether the reduction of AMC correlates with the severity of NEC and whether it can be used to identify infants who need surgery. METHOD The authors collected the clinical data of 66 control and 222 NEC infants. The NEC infants were divided into medical NEC (M-NEC) and surgical NEC (S-NEC). The counting of monocyte and their percentage change were compared at the time of birth, before NEC (baseline), the onset of NEC and after NEC (recovery). In addition, the same comparison was made among stages 1, 2 and 3 of Bell's staging, respectively. RESULTS The authors found that the AMC in NEC infants decreased sharply at the onset. Further comparison was made between 172 cases of M-NEC and 50 cases of S-NEC. It was discovered that the AMC reduced more in S-NEC infants at onset, but it increased more at recovery. In addition, the authors found that among stage 1,2 and 3, stage 3 had the lowest AMC and the largest percentage decrease at the onset. CONCLUSION The AMC decreases sharply in NEC infants at onset, and the degree of decline is associated with the severity of NEC. AMC is expected to be a marker of NEC and provide a reference for clinicians in the diagnosis and treatment of NEC.
Collapse
Affiliation(s)
- Zhiru Wang
- Shanghai Jiao Tong University, School of Medicine, Shanghai Children's Hospital, Department of General Surgery, Shanghai, China
| | - Qingqi Chong
- Shanghai Jiao Tong University, School of Medicine, Shanghai Children's Hospital, Department of General Surgery, Shanghai, China
| | - Junmei Zhou
- Shanghai Jiao Tong University, School of Medicine, Shanghai Children's Hospital, Department of Central Laboratory, Shanghai, China
| | - Tingting Gao
- Shanghai Jiao Tong University, School of Medicine, Shanghai Children's Hospital, Department of General Surgery, Shanghai, China
| | - Kai Zhu
- Shanghai Jiao Tong University, School of Medicine, Shanghai Children's Hospital, Department of General Surgery, Shanghai, China
| | - Xiaohui Gong
- Shanghai Jiao Tong University, School of Medicine, Shanghai Children's Hospital, Department of Neonatology, Shanghai, China
| | - Qingfeng Sheng
- Shanghai Jiao Tong University, School of Medicine, Shanghai Children's Hospital, Department of General Surgery, Shanghai, China.
| | - Zhibao Lv
- Shanghai Jiao Tong University, School of Medicine, Shanghai Children's Hospital, Department of General Surgery, Shanghai, China.
| |
Collapse
|
6
|
Feng Z, Jia C, Lin X, Hao H, Li S, Li F, Cui Q, Chen Y, Wu F, Xiao X. The inhibition of enterocyte proliferation by lithocholic acid exacerbates necrotizing enterocolitis through downregulating the Wnt/β-catenin signalling pathway. Cell Prolif 2022; 55:e13228. [PMID: 35441471 PMCID: PMC9136529 DOI: 10.1111/cpr.13228] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/02/2022] [Accepted: 03/14/2022] [Indexed: 11/26/2022] Open
Abstract
Objectives Necrotizing enterocolitis (NEC) is a catastrophic gastrointestinal emergency in preterm infants, whose exact aetiology remains unknown. The role of lithocholic acid (LCA), a key component of secondary bile acids (BAs), in NEC is unclear. Methods Clinical data were collected to analyse the changes of BAs in NEC patients. In vitro studies, the cell proliferation and cell death were assessed. In vivo experiments, the newborn rats were administered with low or high dose of LCA and further induced NEC. Results Clinically, compared with control group, total BAs in the NEC patients were significantly higher when NEC occurred. In vitro, LCA treatment significantly inhibited the cell proliferation through arresting cell cycle at G1/S phase without inducing apoptosis or necroptosis. Mechanistically, the Wnt/β‐catenin pathway was involved. In vivo, LCA inhibited intestinal cell proliferation leading to disruption of intestinal barrier, and thereby increased the severity of NEC. Specifically, LCA supplementation caused higher levels of FITC‐labelled dextran in serum, reduced PCNA expression and inhibited the activity of Wnt/β‐catenin pathway in enterocytes. The LC–MS/MS test found that LCA was significantly higher in intestinal tissue of NEC group, and more obviously in the NEC‐L and NEC‐H group compared with the DM group. Conclusion LCA exacerbates NEC by inhibiting intestinal cell proliferation through downregulating the Wnt/β‐catenin pathway.
Collapse
Affiliation(s)
- Zhoushan Feng
- Department of Pediatrics, Sun Yat-sen University Sixth Affiliated Hospital, Guangzhou, China.,Department of Pediatrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chunhong Jia
- Department of Pediatrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Key Laboratory for Major Obstetric Diseases of Guangdong Province, Guangzhou, China
| | - Xiaojun Lin
- Department of Pediatrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hu Hao
- Department of Pediatrics, Sun Yat-sen University Sixth Affiliated Hospital, Guangzhou, China
| | - Sitao Li
- Department of Pediatrics, Sun Yat-sen University Sixth Affiliated Hospital, Guangzhou, China
| | - Fei Li
- Department of Pediatrics, Sun Yat-sen University Sixth Affiliated Hospital, Guangzhou, China
| | - Qiliang Cui
- Department of Pediatrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yaoyong Chen
- Department of Pediatrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fan Wu
- Department of Pediatrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Key Laboratory for Major Obstetric Diseases of Guangdong Province, Guangzhou, China
| | - Xin Xiao
- Department of Pediatrics, Sun Yat-sen University Sixth Affiliated Hospital, Guangzhou, China
| |
Collapse
|
7
|
Campion S, Inselman A, Hayes B, Casiraghi C, Joseph D, Facchinetti F, Salomone F, Schmitt G, Hui J, Davis-Bruno K, Van Malderen K, Morford L, De Schaepdrijver L, Wiesner L, Kourula S, Seo S, Laffan S, Urmaliya V, Chen C. The benefits, limitations and opportunities of preclinical models for neonatal drug development. Dis Model Mech 2022; 15:dmm049065. [PMID: 35466995 PMCID: PMC9066504 DOI: 10.1242/dmm.049065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Increased research to improve preclinical models to inform the development of therapeutics for neonatal diseases is an area of great need. This article reviews five common neonatal diseases - bronchopulmonary dysplasia, retinopathy of prematurity, necrotizing enterocolitis, perinatal hypoxic-ischemic encephalopathy and neonatal sepsis - and the available in vivo, in vitro and in silico preclinical models for studying these diseases. Better understanding of the strengths and weaknesses of specialized neonatal disease models will help to improve their utility, may add to the understanding of the mode of action and efficacy of a therapeutic, and/or may improve the understanding of the disease pathology to aid in identification of new therapeutic targets. Although the diseases covered in this article are diverse and require specific approaches, several high-level, overarching key lessons can be learned by evaluating the strengths, weaknesses and gaps in the available models. This Review is intended to help guide current and future researchers toward successful development of therapeutics in these areas of high unmet medical need.
Collapse
Affiliation(s)
- Sarah Campion
- Pfizer Worldwide Research, Development, and Medical, Groton, CT 06340, USA
| | - Amy Inselman
- U.S. Food and Drug Administration, National Center for Toxicological Research, Division of Systems Biology, Jefferson, AR 72079, USA
| | - Belinda Hayes
- U.S. Food and Drug Administration, Center for Drug Evaluation and Research, Office of New Drugs, Silver Spring, MD 20993, USA
| | - Costanza Casiraghi
- Department of Experimental Pharmacology and Translational Science, Chiesi Farmaceutici S.p.A., 43122 Parma, Italy
| | - David Joseph
- U.S. Food and Drug Administration, Center for Drug Evaluation and Research, Office of New Drugs, Silver Spring, MD 20993, USA
| | - Fabrizio Facchinetti
- Department of Experimental Pharmacology and Translational Science, Chiesi Farmaceutici S.p.A., 43122 Parma, Italy
| | - Fabrizio Salomone
- Department of Experimental Pharmacology and Translational Science, Chiesi Farmaceutici S.p.A., 43122 Parma, Italy
| | - Georg Schmitt
- Pharma Research and Early Development, Roche Innovation Center Basel, Pharmaceutical Sciences, F. Hoffmann-La Roche, 4070 Basel, Switzerland
| | - Julia Hui
- Bristol Myers Squibb, Nonclinical Research and Development, Summit, NJ 07901, USA
| | - Karen Davis-Bruno
- U.S. Food and Drug Administration, Center for Drug Evaluation and Research, Office of New Drugs, Silver Spring, MD 20993, USA
| | - Karen Van Malderen
- Federal Agency for Medicines and Health Products (FAMHP), Department DG PRE authorization, 1210 Brussels, Belgium
| | - LaRonda Morford
- Eli Lilly, Global Regulatory Affairs, Indianapolis, IN 46285, USA
| | | | - Lutz Wiesner
- Federal Institute for Drugs and Medical Devices, Clinical Trials, 53175 Bonn, Germany
| | - Stephanie Kourula
- Janssen R&D, Drug Metabolism & Pharmacokinetics, 2340 Beerse, Belgium
| | - Suna Seo
- U.S. Food and Drug Administration, Center for Drug Evaluation and Research, Office of New Drugs, Silver Spring, MD 20993, USA
| | - Susan Laffan
- GlaxoSmithKline, Non-Clinical Safety, Collegeville, PA 19406, USA
| | | | - Connie Chen
- Health and Environmental Sciences Institute, Washington, DC 20005, USA
| |
Collapse
|
8
|
Yang Z, Wei F, Zhang B, Luo Y, Xing X, Wang M, Chen R, Sun G, Sun X. Cellular Immune Signal Exchange From Ischemic Stroke to Intestinal Lesions Through Brain-Gut Axis. Front Immunol 2022; 13:688619. [PMID: 35432368 PMCID: PMC9010780 DOI: 10.3389/fimmu.2022.688619] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 03/16/2022] [Indexed: 12/24/2022] Open
Abstract
As a vital pivot for the human circulatory system, the brain-gut axis is now being considered as an important channel for many of the small immune molecules’ transductions, including interleukins, interferons, neurotransmitters, peptides, and the chemokines penetrating the mesentery and blood brain barrier (BBB) during the development of an ischemic stroke (IS). Hypoxia-ischemia contributes to pituitary and neurofunctional disorders by interfering with the molecular signal release and communication then providing feedback to the gut. Suffering from such a disease on a long-term basis may cause the peripheral system’s homeostasis to become imbalanced, and it can also lead to multiple intestinal complications such as gut microbiota dysbiosis (GMD), inflammatory bowel disease (IBD), necrotizing enterocolitis (NEC), and even the tumorigenesis of colorectal carcinoma (CRC). Correspondingly, these complications will deteriorate the cerebral infarctions and, in patients suffering with IS, it can even ruin the brain’s immune system. This review summarized recent studies on abnormal immunological signal exchange mediated polarization subtype changes, in both macrophages and microglial cells as well as T-lymphocytes. How gut complications modulate the immune signal transduction from the brain are also elucidated and analyzed. The conclusions drawn in this review could provide guidance and novel strategies to benefit remedies for both IS and relative gut lesions from immune-prophylaxis and immunotherapy aspects.
Collapse
Affiliation(s)
- Zizhao Yang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Fei Wei
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bin Zhang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yun Luo
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoyan Xing
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Min Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rongchang Chen
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guibo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Guibo Sun, ; Xiaobo Sun,
| | - Xiaobo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Guibo Sun, ; Xiaobo Sun,
| |
Collapse
|
9
|
Han N, Pan Z, Liu G, Yang R, Yujing B. Hypoxia: The "Invisible Pusher" of Gut Microbiota. Front Microbiol 2021; 12:690600. [PMID: 34367091 PMCID: PMC8339470 DOI: 10.3389/fmicb.2021.690600] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 06/25/2021] [Indexed: 12/20/2022] Open
Abstract
Oxygen is important to the human body. Cell survival and operations depend on oxygen. When the body becomes hypoxic, it affects the organs, tissues and cells and can cause irreversible damage. Hypoxia can occur under various conditions, including external environmental hypoxia and internal hypoxia. The gut microbiota plays different roles under hypoxic conditions, and its products and metabolites interact with susceptible tissues. This review was conducted to elucidate the complex relationship between hypoxia and the gut microbiota under different conditions. We describe the changes of intestinal microbiota under different hypoxic conditions: external environment and internal environment. For external environment, altitude was the mayor cause induced hypoxia. With the increase of altitude, hypoxia will become more serious, and meanwhile gut microbiota also changed obviously. Body internal environment also became hypoxia because of some diseases (such as cancer, neonatal necrotizing enterocolitis, even COVID-19). In addition to the disease itself, this hypoxia can also lead to changes of gut microbiota. The relationship between hypoxia and the gut microbiota are discussed under these conditions.
Collapse
Affiliation(s)
- Ni Han
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Zhiyuan Pan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Guangwei Liu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Ruifu Yang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Bi Yujing
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| |
Collapse
|
10
|
Quitadamo PA, Comegna L, Cristalli P. Anti-Infective, Anti-Inflammatory, and Immunomodulatory Properties of Breast Milk Factors for the Protection of Infants in the Pandemic From COVID-19. Front Public Health 2021; 8:589736. [PMID: 33738273 PMCID: PMC7960784 DOI: 10.3389/fpubh.2020.589736] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022] Open
Abstract
COVID-19 pandemic since the end of 2019 spreads worldwide, counting millions of victims. The viral invasion, systemic inflammation, and consequent organ failure are the gravest features of coronavirus disease 2019 (COVID-19), and they are associated with a high mortality rate. The aim of this study is to evaluate the role of breast milk in the COVID-19 pandemic, analyzing its antiviral, anti-inflammatory, and immunoregulatory effects due to its bioactive components, so numerous and important for the protection of infants. The study tried to demonstrate that all the components of human milk are capable of performing functions on all the pathogenic events recognized and described in COVID-19 disease. Those human milk factors are well-tolerated and practically free of side effects, so breast milk should become a research topic to discover therapies even in this epidemic. In the first part, the mechanisms of protection and defense of the breast milk elements will be delineated; in the second section, it will describe the human milk effects in viral infections and it will be hypothesized how the known mechanisms could act in COVID infection.
Collapse
Affiliation(s)
- Pasqua Anna Quitadamo
- NICU “Casa Sollievo della Sofferenza” Foundation, Scientific Research and Care Institute, San Giovanni Rotondo, Italy
| | | | | |
Collapse
|
11
|
Thai JD, Gregory KE. Bioactive Factors in Human Breast Milk Attenuate Intestinal Inflammation during Early Life. Nutrients 2020; 12:E581. [PMID: 32102231 PMCID: PMC7071406 DOI: 10.3390/nu12020581] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/15/2020] [Accepted: 02/16/2020] [Indexed: 12/14/2022] Open
Abstract
Human breast milk is well known as the ideal source of nutrition during early life, ensuring optimal growth during infancy and early childhood. Breast milk is also the source of many unique and dynamic bioactive components that play a key role in the development of the immune system. These bioactive components include essential microbes, human milk oligosaccharides (HMOs), immunoglobulins, lactoferrin and dietary polyunsaturated fatty acids. These factors all interact with intestinal commensal bacteria and/or immune cells, playing a critical role in establishment of the intestinal microbiome and ultimately influencing intestinal inflammation and gut health during early life. Exposure to breast milk has been associated with a decreased incidence and severity of necrotizing enterocolitis (NEC), a devastating disease characterized by overwhelming intestinal inflammation and high morbidity among preterm infants. For this reason, breast milk is considered a protective factor against NEC and aberrant intestinal inflammation common in preterm infants. In this review, we will describe the key microbial, immunological, and metabolic components of breast milk that have been shown to play a role in the mechanisms of intestinal inflammation and/or NEC prevention.
Collapse
Affiliation(s)
- Julie D. Thai
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Katherine E. Gregory
- Department of Pediatric Newborn Medicine, Department of Nursing, Brigham and Women’s Hospital, Boston, MA 02115, USA;
| |
Collapse
|
12
|
Elevated Coefficient of Variation in Total Fecal Bile Acids Precedes Diagnosis of Necrotizing Enterocolitis. Sci Rep 2020; 10:249. [PMID: 31937876 PMCID: PMC6959237 DOI: 10.1038/s41598-019-57178-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 12/24/2019] [Indexed: 12/15/2022] Open
Abstract
Accumulation of bile acids (BAs) may mediate development of necrotizing enterocolitis (NEC). Serial fecal samples were collected from premature infants with birth weight (BW) ≤ 1800 g, estimated gestational age (EGA) ≤ 32 weeks, and <30 days old prior to initiation of enteral feeding. Nine infants that developed Bell’s Stage ≥ II NEC were matched with control infants based on BW, EGA, day of life (DOL) enteral feeding was initiated and DOL of the first sample. From each subject, five samples matched by DOL collected were analyzed for BA levels and composition. Fifteen individual BA species were measured via LC-MS/MS and total BA levels were measured using the Diazyme Total Bile Acid Assay kit. No statistically significant differences in composition were observed between control and NEC at the level of individual species (p = 0.1133) or grouped BAs (p = 0.0742). However, there was a statistically significant difference (p = 0.000012) in the mean coefficient of variation (CV) between the two groups with infants developing NEC having more than four-fold higher mean CV than controls. Importantly, these variations occurred prior to NEC diagnosis. These data suggest fluctuations in total fecal BA levels could provide the basis for the first predictive clinical test for NEC.
Collapse
|
13
|
Meister AL, Doheny KK, Travagli RA. Necrotizing enterocolitis: It's not all in the gut. Exp Biol Med (Maywood) 2019; 245:85-95. [PMID: 31810384 DOI: 10.1177/1535370219891971] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Necrotizing enterocolitis is the leading cause of death due to gastrointestinal disease in preterm neonates, affecting 5–12% of neonates born at a very-low birth weight. Necrotizing enterocolitis can present with a slow and insidious onset, with some neonates displaying early symptoms such as feeding intolerance. Treatment during the early stages includes bowel rest and careful use of antibiotics, but surgery is required if pneumoperitoneum and intestinal perforation occur. Mortality rates among neonates requiring surgery are estimated to be 20–30%, mandating the development of non-invasive and reliable biomarkers to predict necrotizing enterocolitis before the onset of clinical signs. Such biomarkers would allow at-risk neonates to receive maximal preventative therapies such as careful nutritional consideration, probiotics, and increased skin-to-skin care.Impact statementNecrotizing enterocolitis (NEC) is a devastating gastrointestinal disease; its high mortality rate mandates the development of non-invasive biomarkers to predict NEC before its onset. This review summarizes the pathogenesis, prevention, unresolved issues, and long-term outcomes of NEC.
Collapse
Affiliation(s)
- Alissa L Meister
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, USA
| | - Kim K Doheny
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, USA.,Neonatal-Perinatal Medicine, Penn State College of Medicine, Hershey, PA, USA
| | - R Alberto Travagli
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, USA
| |
Collapse
|
14
|
Perez M, Robbins ME, Revhaug C, Saugstad OD. Oxygen radical disease in the newborn, revisited: Oxidative stress and disease in the newborn period. Free Radic Biol Med 2019; 142:61-72. [PMID: 30954546 PMCID: PMC6791125 DOI: 10.1016/j.freeradbiomed.2019.03.035] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 03/06/2019] [Accepted: 03/29/2019] [Indexed: 12/28/2022]
Abstract
Thirty years ago, there was an emerging appreciation for the significance of oxidative stress in newborn disease. This prompted a renewed interest in the impact of oxygen therapy for the newborn in the delivery room and beyond, especially in premature infants. Today, the complexity of oxidative stress both in normal regulation and pathology is better understood, especially as it relates to neonatal mitochondrial oxidative stress responses to hyperoxia. Mitochondria are recipients of oxidative damage and have a propensity for oxidative self-injury that has been implicated in the pathogenesis of neonatal lung diseases. Similarly, both intrauterine growth restriction (IUGR) and macrosomia are associated with mitochondrial dysfunction and oxidative stress. Additionally, reoxygenation with 100% O2 in a hypoxic-ischemic newborn lamb model increased the production of pro-inflammatory cytokines in the brain. Moreover, the interplay between inflammation and oxidative stress in the newborn is better understood because of animal studies. Transcriptomic analyses have found a number of genes to be differentially expressed in murine models of bronchopulmonary dysplasia (BPD). Epigenetic changes have also been detected both in animal models of BPD and premature infants exposed to oxygen. Antioxidant therapy to prevent newborn disease has not been very successful; however, new therapeutic principles, like melatonin, are under investigation.
Collapse
Affiliation(s)
- Marta Perez
- Division of Neonatology, Stanley Manne Children's Research Institute, Ann and Robert H Lurie Children's Hospital, Chicago, IL, United States; Department of Pediatrics, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States
| | - Mary E Robbins
- Division of Neonatology, Stanley Manne Children's Research Institute, Ann and Robert H Lurie Children's Hospital, Chicago, IL, United States; Department of Pediatrics, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States
| | - Cecilie Revhaug
- Department of Pediatric Research, University of Oslo, Oslo University Hospital, Norway
| | - Ola D Saugstad
- Department of Pediatrics, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States; Department of Pediatric Research, University of Oslo, Oslo University Hospital, Norway.
| |
Collapse
|
15
|
Burge K, Gunasekaran A, Eckert J, Chaaban H. Curcumin and Intestinal Inflammatory Diseases: Molecular Mechanisms of Protection. Int J Mol Sci 2019; 20:ijms20081912. [PMID: 31003422 PMCID: PMC6514688 DOI: 10.3390/ijms20081912] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/15/2019] [Accepted: 04/17/2019] [Indexed: 02/07/2023] Open
Abstract
Intestinal inflammatory diseases, such as Crohn’s disease, ulcerative colitis, and necrotizing enterocolitis, are becoming increasingly prevalent. While knowledge of the pathogenesis of these related diseases is currently incomplete, each of these conditions is thought to involve a dysfunctional, or overstated, host immunological response to both bacteria and dietary antigens, resulting in unchecked intestinal inflammation and, often, alterations in the intestinal microbiome. This inflammation can result in an impaired intestinal barrier allowing for bacterial translocation, potentially resulting in systemic inflammation and, in severe cases, sepsis. Chronic inflammation of this nature, in the case of inflammatory bowel disease, can even spur cancer growth in the longer-term. Recent research has indicated certain natural products with anti-inflammatory properties, such as curcumin, can help tame the inflammation involved in intestinal inflammatory diseases, thus improving intestinal barrier function, and potentially, clinical outcomes. In this review, we explore the potential therapeutic properties of curcumin on intestinal inflammatory diseases, including its antimicrobial and immunomodulatory properties, as well as its potential to alter the intestinal microbiome. Curcumin may play a significant role in intestinal inflammatory disease treatment in the future, particularly as an adjuvant therapy.
Collapse
Affiliation(s)
- Kathryn Burge
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, 1200 North Everett Drive, ETNP7504, Oklahoma City, OK 73104, USA.
| | - Aarthi Gunasekaran
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, 1200 North Everett Drive, ETNP7504, Oklahoma City, OK 73104, USA.
| | - Jeffrey Eckert
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, 1200 North Everett Drive, ETNP7504, Oklahoma City, OK 73104, USA.
| | - Hala Chaaban
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, 1200 North Everett Drive, ETNP7504, Oklahoma City, OK 73104, USA.
| |
Collapse
|
16
|
Arthur CM, Nalbant D, Feldman HA, Saeedi BJ, Matthews J, Robinson BS, Kamili NA, Bennett A, Cress GA, Sola-Visner M, Jones RM, Zimmerman MB, Neish AS, Patel RM, Nopoulos P, Georgieff MK, Roback JD, Widness JA, Josephson CD, Stowell SR. Anemia induces gut inflammation and injury in an animal model of preterm infants. Transfusion 2019; 59:1233-1245. [PMID: 30897226 DOI: 10.1111/trf.15254] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 11/12/2018] [Accepted: 11/28/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND While very low birth weight (VLBW) infants often require multiple red blood cell transfusions, efforts to minimize transfusion-associated risks have resulted in more restrictive neonatal transfusion practices. However, whether restrictive transfusion strategies limit transfusions without increasing morbidity and mortality in this population remains unclear. Recent epidemiologic studies suggest that severe anemia may be an important risk factor for the development of necrotizing enterocolitis (NEC). However, the mechanism whereby anemia may lead to NEC remains unknown. STUDY DESIGN AND METHODS The potential impact of anemia on neonatal inflammation and intestinal barrier disruption, two well-characterized predisposing features of NEC, was defined by correlation of hemoglobin values to cytokine levels in premature infants and by direct evaluation of intestinal hypoxia, inflammation and gut barrier disruption using a pre-clinical neonatal murine model of phlebotomy-induced anemia (PIA). RESULTS Increasing severity of anemia in the preterm infant correlated with the level of IFN-gamma, a key pro-inflammatory cytokine that may predispose an infant to NEC. Gradual induction of PIA in a pre-clinical model resulted in significant hypoxia throughout the intestinal mucosa, including areas where intestinal macrophages reside. PIA-induced hypoxia significantly increased macrophage pro-inflammatory cytokine levels, while reducing tight junction protein ZO-1 expression and increasing intestinal barrier permeability. Macrophage depletion reversed the impact of anemia on intestinal ZO-1 expression and barrier function. CONCLUSIONS Taken together, these results suggest that anemia can increase intestinal inflammation and barrier disruption likely through altered macrophage function, leading to the type of predisposing intestinal injury that may increase the risk for NEC.
Collapse
Affiliation(s)
- Connie M Arthur
- Departments of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Atlanta, Georgia
| | - Demet Nalbant
- Department of Pediatrics, University of Iowa, Iowa City, Iowa
| | - Henry A Feldman
- Division of Newborn Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Bejan J Saeedi
- Departments of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Atlanta, Georgia
| | - Jason Matthews
- Departments of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Atlanta, Georgia
| | - Brian S Robinson
- Departments of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Atlanta, Georgia
| | - Nourine A Kamili
- Departments of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Atlanta, Georgia
| | - Ashley Bennett
- Departments of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Atlanta, Georgia
| | | | - Martha Sola-Visner
- Division of Newborn Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Rheinallt M Jones
- Departments of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Atlanta, Georgia
| | | | - Andrew S Neish
- Departments of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Atlanta, Georgia
| | - Ravi M Patel
- Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Peggy Nopoulos
- Department of Psychiatry, College of Medicine, University of Iowa, Iowa City, Iowa
| | - Michael K Georgieff
- Department of Pediatrics, School of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - John D Roback
- Departments of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Atlanta, Georgia
| | - John A Widness
- Department of Pediatrics, University of Iowa, Iowa City, Iowa
| | - Cassandra D Josephson
- Departments of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Atlanta, Georgia.,Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Sean R Stowell
- Departments of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Atlanta, Georgia
| |
Collapse
|
17
|
Yin J, Sheng B, Yang K, Sun L, Xiao W, Yang H. The protective roles of NLRP6 in intestinal epithelial cells. Cell Prolif 2018; 52:e12555. [PMID: 30515917 PMCID: PMC6496424 DOI: 10.1111/cpr.12555] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 10/11/2018] [Accepted: 10/17/2018] [Indexed: 12/12/2022] Open
Abstract
The evolution of chronic inflammatory diseases is thought to be due to a combination of host genetic variations and environmental factors that include the alteration of intestinal flora, termed "dysbiosis." The intestinal mucosal barrier includes a chemical barrier and physical barrier that have important roles in protecting the intestine against inflammatory injury. The chemical barrier includes antimicrobial peptides (AMPs), and the physical barrier includes a mucous layer, a monolayer of intestinal epithelial cells and cell junctions. The intestinal mucosal barrier is not a static barrier, but rather, it strongly interacts with the gut microbiome and cells of the immune system. Correct expression of AMPs, together with mucus and balanced epithelial cell proliferation, prevents the occurrence of disease. NLRP6, a member of the nucleotide-binding domain, leucine-rich repeat-containing (NLR) innate immune receptor family, participates in the progression of intestinal inflammation and enteric pathogen infections. It has become apparent in recent years that NLRP6 is important in disease pathogenesis, as it responds to internal ligands that lead to the release of AMPs and mucus, thus regulating the regeneration of intestinal epithelial cells. This review summarizes the activation of NLRP6 and its protective role in the intestinal epithelial cell.
Collapse
Affiliation(s)
- Jiuheng Yin
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Baifa Sheng
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Kunqiu Yang
- Department of General Surgery, Navy General Hospital, Beijing, China
| | - Lihua Sun
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Weidong Xiao
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Hua Yang
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
18
|
Gross Margolis K, Vittorio J, Talavera M, Gluck K, Li Z, Iuga A, Stevanovic K, Saurman V, Israelyan N, Welch MG, Gershon MD. Enteric serotonin and oxytocin: endogenous regulation of severity in a murine model of necrotizing enterocolitis. Am J Physiol Gastrointest Liver Physiol 2017; 313:G386-G398. [PMID: 28774871 PMCID: PMC5792212 DOI: 10.1152/ajpgi.00215.2017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 07/28/2017] [Accepted: 07/28/2017] [Indexed: 01/31/2023]
Abstract
Necrotizing enterocolitis (NEC), a gastrointestinal inflammatory disease of unknown etiology that may also affect the liver, causes a great deal of morbidity and mortality in premature infants. We tested the hypothesis that signaling molecules, which are endogenous to the bowel, regulate the severity of intestinal and hepatic damage in an established murine NEC model. Specifically, we postulated that mucosal serotonin (5-HT), which is proinflammatory, would exacerbate experimental NEC and that oxytocin (OT), which is present in enteric neurons and is anti-inflammatory, would oppose it. Genetic deletion of the 5-HT transporter (SERT), which increases and prolongs effects of 5-HT, was found to increase the severity of systemic manifestations, intestinal inflammation, and associated hepatotoxicity of experimental NEC. In contrast, genetic deletion of tryptophan hydroxylase 1 (TPH1), which is responsible for 5-HT biosynthesis in enterochromaffin (EC) cells of the intestinal mucosa, and TPH inhibition with LP-920540 both decrease the severity of experimental NEC in the small intestine and liver. These observations suggest that 5-HT from EC cells helps to drive the inflammatory damage to the gut and liver that occurs in the murine NEC model. Administration of OT decreased, while the OT receptor antagonist atosiban exacerbated, the intestinal inflammation of experimental NEC. Data from the current investigation are consistent with the tested hypotheses-that the enteric signaling molecules, 5-HT (positively) and OT (negatively) regulate severity of inflammation in a mouse model of NEC. Moreover, we suggest that mucosally restricted inhibition of 5-HT biosynthesis and/or administration of OT may be useful in the treatment of NEC.NEW & NOTEWORTHY Serotonin (5-HT) and oxytocin reciprocally regulate the severity of intestinal inflammation and hepatotoxicity in a murine model of necrotizing enterocolitis (NEC). Selective depletion of mucosal 5-HT through genetic deletion or inhibition of tryptophan hydroxylase-1 ameliorates, while deletion of the 5-HT uptake transporter, which increases 5-HT availability, exacerbates the severity of NEC. In contrast, oxytocin reduces, while the oxytocin receptor antagonist atosiban enhances, NEC severity. Peripheral tryptophan hydroxylase inhibition may be useful in treatment of NEC.
Collapse
Affiliation(s)
- Kara Gross Margolis
- Department of Pediatrics, Morgan Stanley Children's Hospital, Columbia University Medical Center, New York, New York; .,Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York; and
| | - Jennifer Vittorio
- 1Department of Pediatrics, Morgan Stanley Children’s Hospital, Columbia University Medical Center, New York, New York; ,2Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York; and
| | - Maria Talavera
- 1Department of Pediatrics, Morgan Stanley Children’s Hospital, Columbia University Medical Center, New York, New York; ,2Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York; and
| | - Karen Gluck
- 1Department of Pediatrics, Morgan Stanley Children’s Hospital, Columbia University Medical Center, New York, New York; ,2Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York; and
| | - Zhishan Li
- 1Department of Pediatrics, Morgan Stanley Children’s Hospital, Columbia University Medical Center, New York, New York; ,2Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York; and
| | - Alina Iuga
- 2Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York; and
| | - Korey Stevanovic
- 1Department of Pediatrics, Morgan Stanley Children’s Hospital, Columbia University Medical Center, New York, New York; ,2Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York; and
| | - Virginia Saurman
- 1Department of Pediatrics, Morgan Stanley Children’s Hospital, Columbia University Medical Center, New York, New York; ,2Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York; and
| | - Narek Israelyan
- 1Department of Pediatrics, Morgan Stanley Children’s Hospital, Columbia University Medical Center, New York, New York; ,2Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York; and
| | - Martha G. Welch
- 1Department of Pediatrics, Morgan Stanley Children’s Hospital, Columbia University Medical Center, New York, New York; ,2Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York; and ,3Department of Psychiatry, Columbia University Medical Center, New York, New York
| | - Michael D. Gershon
- 2Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York; and
| |
Collapse
|
19
|
Shinyama S, Kaji T, Mukai M, Nakame K, Matsufuji H, Takamatsu H, Ieiri S. The novel preventive effect of Daikenchuto (TJ-100), a Japanese herbal drug, against neonatal necrotizing enterocolitis in rats. Pediatr Surg Int 2017; 33:1109-1114. [PMID: 28815293 DOI: 10.1007/s00383-017-4145-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/01/2017] [Indexed: 12/13/2022]
Abstract
PURPOSE Necrotizing enterocolitis (NEC) is a devastating gastrointestinal disease of premature infants. Daikenchuto, a Japanese herbal drug, has several effects on the digestive system, so we investigated its preventive effects in a rat model of NEC. METHODS NEC was induced in newborn rats via asphyxia (100% N2 for 90 s; every 4 h) + LPS (4 mg/kg/day [administered orally on days 0 and 1]). The effects of Daikenchuto were evaluated in four groups (control: 0 g/kg/day, I: 0.3 g/kg/day, II: 0.6 g/kg/day, and III: 1.0 g/kg/day). Daikenchuto was administered into the stomach through a microcatheter. The incidence and severity of NEC were pathologically assessed using the NEC grade in accordance with Dovorak's previous report. Cell positivity for inflammatory cytokine (IL-6) was also evaluated. RESULTS Daikenchuto reduced the incidence of NEC in control, Groups I, II, and III to 68.7, 30.0, 30.7, and 13.3%, respectively. High-dose Daikenchuto significantly improved the incidence of NEC, and the rate of IL-6 positive cells in group III was significantly lower than in the control group (p = 0.04). CONCLUSION We evaluated the effect of Daikenchuto against NEC and found that it reduced the incidence rate of NEC due to a decrease in the IL-6 production.
Collapse
Affiliation(s)
- Shin Shinyama
- Department of Pediatric Surgery, Research Field in Medicine and Health Sciences, Medical and Dental Sciences Area, Research and Education Assembly, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Tatsuru Kaji
- Department of Pediatric Surgery, Research Field in Medicine and Health Sciences, Medical and Dental Sciences Area, Research and Education Assembly, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Motoi Mukai
- Department of Pediatric Surgery, Research Field in Medicine and Health Sciences, Medical and Dental Sciences Area, Research and Education Assembly, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Kazuhiko Nakame
- Department of Pediatric Surgery, Research Field in Medicine and Health Sciences, Medical and Dental Sciences Area, Research and Education Assembly, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Hiroshi Matsufuji
- Ambulatory Care Center for Children, St. Luke's International Hospital, Tokyo, Japan
| | - Hideo Takamatsu
- Research and Education Assembly, Kagoshima University, Kagoshima, Japan
| | - Satoshi Ieiri
- Department of Pediatric Surgery, Research Field in Medicine and Health Sciences, Medical and Dental Sciences Area, Research and Education Assembly, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan.
| |
Collapse
|
20
|
Peroxisome proliferator-activated receptor-γ agonist pioglitazone reduces the development of necrotizing enterocolitis in a neonatal preterm rat model. Pediatr Res 2017; 81:364-368. [PMID: 27973471 DOI: 10.1038/pr.2016.214] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 08/25/2016] [Indexed: 11/09/2022]
Abstract
BACKGROUND Factors affecting innate immunity and acting as inflammatory regulators, such as the nuclear peroxisome proliferator-activated receptors (PPAR) could be crucial in the pathogenesis of necrotizing enterocolitis (NEC). We hypothesized that the PPARγ agonist pioglitazone (PIO) might be effective in preventing the development of NEC and/or reducing its severity. METHODS We studied preterm rats in which NEC was induced using the hypoxia-hypothermia model. The treatment group (TG; n = 30) received enteral PIO (10 mg/kg/d) for 72 h and the control group (CG; n = 30) did not. Animals were sacrificed 96 h after birth. NEC was diagnosed evaluating histological ileum changes, and mRNA levels of IL-4, IL-12, IL-6, IL-10, INF-γ, and TNF-α cytokines were measured. RESULTS NEC occurrence was higher in the CG (18/30; 60%) than in the TG (5/30; 16.7%) and was more severe. Proinflammatory IL-12 and INF-γ mRNA levels were significantly lower in the TG than in the CG; conversely, the anti-inflammatory IL-4 mRNA level was significantly higher in the TG than in the CG. CONCLUSION Our results demonstrate for the first time that PIO is effective in reducing the incidence and severity of NEC and in decreasing renal injuries in a preterm rat model.
Collapse
|
21
|
|
22
|
Martin VJ, Leonard MM, Fiechtner L, Fasano A. Transitioning From Descriptive to Mechanistic Understanding of the Microbiome: The Need for a Prospective Longitudinal Approach to Predicting Disease. J Pediatr 2016; 179:240-248. [PMID: 27634626 PMCID: PMC5479769 DOI: 10.1016/j.jpeds.2016.08.049] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 07/15/2016] [Accepted: 08/16/2016] [Indexed: 12/11/2022]
Affiliation(s)
| | | | | | - Alessio Fasano
- Department of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital for Children, Boston, MA.
| |
Collapse
|
23
|
Abstract
Necrotising enterocolitis (NEC) is an uncommon, but devastating intestinal inflammatory disease that predominantly affects preterm infants. NEC is sometimes dubbed the spectre of neonatal intensive care units, as its onset is insidiously non-specific, and once the disease manifests, the damage inflicted on the baby's intestine is already disastrous. Subsequent sepsis and multi-organ failure entail a mortality of up to 65%. Development of effective treatments for NEC has stagnated, largely because of our lack of understanding of NEC pathogenesis. It is clear, however, that NEC is driven by a profoundly dysregulated immune system. NEC is associated with local increases in pro-inflammatory mediators, e.g. Toll-like receptor (TLR) 4, nuclear factor-κB, tumour necrosis factor, platelet-activating factor (PAF), interleukin (IL)-18, interferon-gamma, IL-6, IL-8 and IL-1β. Deficiencies in counter-regulatory mechanisms, including IL-1 receptor antagonist (IL-1Ra), TLR9, PAF-acetylhydrolase, transforming growth factor beta (TGF-β)1&2, IL-10 and regulatory T cells likely facilitate a pro-inflammatory milieu in the NEC-afflicted intestine. There is insufficient evidence to conclude a predominance of an adaptive Th1-, Th2- or Th17-response in the disease. Our understanding of the accompanying regulation of systemic immunity remains poor; however, IL-1Ra, IL-6, IL-8 and TGF-β1 show promise as biomarkers. Here, we chart the emerging immunological landscape that underpins NEC by reviewing the involvement and potential clinical implications of innate and adaptive immune mediators and their regulation in NEC.
Collapse
|
24
|
Affiliation(s)
- Alessio Fasano
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital For Children, Harvard Medical School, Charlestown, MA 02129, USA; European Biomedical Research Institute of Salerno (EBRIS), Via De Renzi 50, 84125, Salerno (Italy).
| |
Collapse
|
25
|
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
|
26
|
Kitessa SM, Nattrass GS, Forder REA, McGrice HA, Wu SB, Hughes RJ. Mucin gene mRNA levels in broilers challenged with eimeria and/or Clostridium perfringens. Avian Dis 2015; 58:408-14. [PMID: 25518436 DOI: 10.1637/10757-122313-reg.1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The effects of Eimeria (EM) and Clostridium perfringens (CP) challenges on the mRNA levels of genes involved in mucin (Muc) synthesis (Muc2, Muc5ac, Muc13, and trefoil family factor-2 [TFF2]), inflammation (tumor necrosis factor alpha [TNF-alpha] and interleukin-18 [IL-18]), and metabolic processes (cluster of differentiation [CD]36) in the jejunum of broilers were investigated. Two parallel experiments involving 1) EM challenge and 2) EM and CP challenges were conducted. The first experiment was a 2 X 2 study with 12 birds per treatment (N = 48) involving fishmeal substitution (25%) in the diet (FM) and EM challenge. The treatments were: Control (FM-, EM-), Fishmeal (FM+, EM-), EM challenge (FM-, EM+), and fishmeal substitution and EM challenge (FM+, EM+). The second experiment was a 2 X 2 X 2 experiment with six birds per treatment (N = 48) involving fishmeal (FM-, FM+), Eimeria (EM-, EM+), and C perfringens (CP-, CP+). In both arms of the study, male broilers were given a starter diet for the whole period of 16 days, except those assigned to FM+, where 25% of the starter ration was replaced with fishmeal from days 8 to 14. EM inoculation was performed on day 9 and CP inoculation on days 14 and 15. The EM challenge birds were euthanatized for sampling on day 13; postmortem examination and sampling for the Eimeria plus C perfringens challenge arm of the study were on day 16. In the Eimeria challenge arm of the study, fishmeal supplementation significantly suppressed the mRNA levels of TNF-alpha, TFF2, and IL-18 pre-CP inoculation but simultaneously increased the levels of Muc13 and CD36 mRNAs. Birds challenged with Eimeria exhibited increased mRNA levels of Muc13, Muc5ac, TNF-alpha, and IL-18. In the Eimeria and C. perfringens challenge arm, birds exposed to EM challenge exhibited significantly lower mRNA levels of Muc2 and CD36. The mRNA levels of CD36 were also significantly suppressed by CP challenge. Our results showed that the transcription of mucin synthesis genes in the jejunum of broilers is modulated by fishmeal inclusion in the diet. Furthermore, we show for the first time suppression of CD36 mRNA levels in the intestine of broilers challenged with Eimeria or C. perfringens.
Collapse
|
27
|
Remon JI, Kampanatkosol R, Kaul R, Muraskas JK, Christensen RD, Maheshwari A. Acute drop in blood monocyte count differentiates NEC from other causes of feeding intolerance. J Perinatol 2014; 34:549-54. [PMID: 24674979 PMCID: PMC4074443 DOI: 10.1038/jp.2014.52] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 02/02/2014] [Accepted: 02/24/2014] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Necrotizing enterocolitis (NEC) is characterized by macrophage infiltration into affected tissues. Because intestinal macrophages are derived from recruitment and in situ differentiation of blood monocytes in the gut mucosa, we hypothesized that increased recruitment of monocytes to the intestine during NEC reduces the blood monocyte concentration and that this fall in blood monocytes can be a useful biomarker for NEC. STUDY DESIGN We reviewed medical records of very-low-birth-weight (VLBW) infants treated for NEC and compared them with a matched control group comprised of infants with feeding intolerance but no signs of NEC. Clinical characteristics and absolute monocyte counts (AMCs) were recorded. Diagnostic accuracy of AMC values was tested using receiver-operator characteristics (ROC). RESULT We compared 69 cases and 257 controls (median 27 weeks, range 26 to 29 in both the groups). In stage II NEC, AMCs decreased from median 1.7 × 10(9) l(-1) (interquartile range (IQR) 0.98 to 2.4) to 0.8 (IQR 0.62 to 2.1); P < 0.05. In stage III NEC, monocyte counts decreased from median 2.1 × 10(9) l(-1) (IQR 0.1.5 to 3.2) to 0.8 (IQR 0.6 to 1.9); P < 0.05. There was no change in AMCs in control infants. ROC of AMC values showed a diagnostic accuracy (area under the curve) of 0.76. In a given infant with feeding intolerance, a drop in AMCs of > 20% indicated NEC with sensitivity of 0.70 (95% confidence interval (CI) 0.57 to 0.81) and specificity of 0.71 (95% CI 0.64 to 0.77). CONCLUSION We have identified a fall in blood monocyte concentration as a novel biomarker for NEC in VLBW infants.
Collapse
Affiliation(s)
- Juan I. Remon
- Department of Pediatrics, Division of Neonatology, University of Illinois at Chicago, Chicago, Illinois
- Department of Pediatrics, Center for Neonatal and Pediatric Gastrointestinal Disease, University of Illinois at Chicago, Chicago, Illinois
| | - Richard Kampanatkosol
- Department of Pediatrics, Division of Neonatology, Loyola University Stritch School of Medicine, Maywood, Illinois
| | - Rajat Kaul
- Department of Pediatrics, Center for Neonatal and Pediatric Gastrointestinal Disease, University of Illinois at Chicago, Chicago, Illinois
| | - Jonathan K. Muraskas
- Department of Pediatrics, Division of Neonatology, Loyola University Stritch School of Medicine, Maywood, Illinois
| | - Robert D. Christensen
- Intermountain Healthcare Women and Newborns Clinical Program, Ogden, Utah
- McKay-Dee Hospital Center, Ogden, Utah
| | - Akhil Maheshwari
- Department of Pediatrics, Division of Neonatology, University of Illinois at Chicago, Chicago, Illinois
- Department of Pediatrics, Center for Neonatal and Pediatric Gastrointestinal Disease, University of Illinois at Chicago, Chicago, Illinois
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
28
|
Lu P, Sodhi CP, Jia H, Shaffiey S, Good M, Branca MF, Hackam DJ. Animal models of gastrointestinal and liver diseases. Animal models of necrotizing enterocolitis: pathophysiology, translational relevance, and challenges. Am J Physiol Gastrointest Liver Physiol 2014; 306:G917-28. [PMID: 24763555 PMCID: PMC4042110 DOI: 10.1152/ajpgi.00422.2013] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Necrotizing enterocolitis is the leading cause of morbidity and mortality from gastrointestinal disease in premature infants and is characterized by initial feeding intolerance and abdominal distention followed by the rapid progression to coagulation necrosis of the intestine and death in many cases. Although the risk factors for NEC development remain well accepted, namely premature birth and formula feeding, the underlying mechanisms remain incompletely understood. Current thinking indicates that NEC develops in response to an abnormal interaction between the mucosal immune system of the premature host and an abnormal indigenous microflora, leading to an exaggerated mucosal inflammatory response and impaired mesenteric perfusion. In seeking to understand the molecular and cellular events leading to NEC, various animal models have been developed. However, the large number and variability between the available animal models and the unique characteristics of each has raised important questions regarding the validity of particular models for NEC research. In an attempt to provide some guidance to the growing community of NEC researchers, we now seek to review the key features of the major NEC models that have been developed in mammalian and nonmammalian species and to assess the advantages, disadvantage, challenges and major scientific discoveries yielded by each. A strategy for model validation is proposed, the principal models are compared, and future directions and challenges within the field of NEC research are explored.
Collapse
Affiliation(s)
- Peng Lu
- 1Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania;
| | - Chhinder P. Sodhi
- 1Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania;
| | - Hongpeng Jia
- 1Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania;
| | - Shahab Shaffiey
- 1Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania;
| | - Misty Good
- 3Division of Newborn Medicine, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Maria F. Branca
- 1Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania;
| | - David J. Hackam
- 1Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; ,2Division of Pediatric Surgery, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania;
| |
Collapse
|
29
|
Sylvester KG, Ling XB, Liu GYG, Kastenberg ZJ, Ji J, Hu Z, Wu S, Peng S, Abdullah F, Brandt ML, Ehrenkranz RA, Harris MC, Lee TC, Simpson BJ, Bowers C, Moss RL. Urine protein biomarkers for the diagnosis and prognosis of necrotizing enterocolitis in infants. J Pediatr 2014; 164:607-12.e1-7. [PMID: 24433829 PMCID: PMC4161235 DOI: 10.1016/j.jpeds.2013.10.091] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Revised: 08/21/2013] [Accepted: 10/14/2013] [Indexed: 10/25/2022]
Abstract
OBJECTIVES To test the hypothesis that an exploratory proteomics analysis of urine proteins with subsequent development of validated urine biomarker panels would produce molecular classifiers for both the diagnosis and prognosis of infants with necrotizing enterocolitis (NEC). STUDY DESIGN Urine samples were collected from 119 premature infants (85 NEC, 17 sepsis, 17 control) at the time of initial clinical concern for disease. The urine from 59 infants was used for candidate biomarker discovery by liquid chromatography/mass spectrometry. The remaining 60 samples were subject to enzyme-linked immunosorbent assay for quantitative biomarker validation. RESULTS A panel of 7 biomarkers (alpha-2-macroglobulin-like protein 1, cluster of differentiation protein 14, cystatin 3, fibrinogen alpha chain, pigment epithelium-derived factor, retinol binding protein 4, and vasolin) was identified by liquid chromatography/mass spectrometry and subsequently validated by enzyme-linked immunosorbent assay. These proteins were consistently found to be either up- or down-regulated depending on the presence, absence, or severity of disease. Biomarker panel validation resulted in a receiver-operator characteristic area under the curve of 98.2% for NEC vs sepsis and an area under the curve of 98.4% for medical NEC vs surgical NEC. CONCLUSIONS We identified 7 urine proteins capable of providing highly accurate diagnostic and prognostic information for infants with suspected NEC. This work represents a novel approach to improving the efficiency with which we diagnose early NEC and identify those at risk for developing severe, or surgical, disease.
Collapse
Affiliation(s)
- Karl G. Sylvester
- Division of Pediatric Surgery, Lucile Packard Children’s Hospital, Palo Alto, CA,Department of Surgery, Stanford University School of Medicine, Baltimore, MD
| | - Xuefeng B. Ling
- Department of Surgery, Stanford University School of Medicine, Baltimore, MD
| | - Gigi Yuen-Gee Liu
- Department of Surgery, Stanford University School of Medicine, Baltimore, MD,Pediatric Surgery, Department of Surgery, Johns Hopkins University, Baltimore, MD
| | - Zachary J. Kastenberg
- Division of Pediatric Surgery, Lucile Packard Children’s Hospital, Palo Alto, CA,Department of Surgery, Stanford University School of Medicine, Baltimore, MD
| | - Jun Ji
- Department of Surgery, Stanford University School of Medicine, Baltimore, MD
| | - Zhongkai Hu
- Department of Surgery, Stanford University School of Medicine, Baltimore, MD
| | - Shuaibin Wu
- Department of Surgery, Stanford University School of Medicine, Baltimore, MD
| | - Sihua Peng
- Department of Surgery, Stanford University School of Medicine, Baltimore, MD
| | - Fizan Abdullah
- Pediatric Surgery, Department of Surgery, Johns Hopkins University, Baltimore, MD
| | - Mary L. Brandt
- Pediatric Surgery, Department of Surgery, Texas Children’s Hospital, Houston, TX
| | - Richard A. Ehrenkranz
- Department of Pediatrics, Division of Neonatology, Yale University School of Medicine, New Haven, CT
| | - Mary Catherine Harris
- Department of Pediatrics, Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Timothy C. Lee
- Pediatric Surgery, Department of Surgery, Texas Children’s Hospital, Houston, TX
| | - B. Joyce Simpson
- Department of Pediatrics, Division of Neonatology, Yale University School of Medicine, New Haven, CT
| | - Corinna Bowers
- Pediatric Surgery, Department of Surgery, Nationwide Children’s Hospital, Columbus, OH
| | - R. Lawrence Moss
- Pediatric Surgery, Department of Surgery, Nationwide Children’s Hospital, Columbus, OH
| |
Collapse
|
30
|
Yurttutan S, Ozdemir R, Canpolat FE, Oncel MY, Unverdi HG, Uysal B, Erdeve Ö, Dilmen U. Beneficial effects of Etanercept on experimental necrotizing enterocolitis. Pediatr Surg Int 2014; 30:71-7. [PMID: 24072202 DOI: 10.1007/s00383-013-3415-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/04/2013] [Indexed: 12/21/2022]
Abstract
PURPOSE Tissue damage in necrotizing enterocolitis (NEC) of infants occurs as a result of an uncontrolled inflammatory response. The aim of this study was to investigate any potential anti-inflammatory effects that Etanercept may have on the inflammatory response in an experimental NEC model in newborn rats. METHODS Newborn pups were randomized into three groups immediately after birth (Control, NEC + Placebo and NEC + Etanercept). Pups in the NEC + Placebo and NEC + Etanercept groups were subjected to an NEC-inducing protocol (hypercarbia, hypothermia and hyperoxia) twice a day for 3 days. Pups in the NEC + Etanercept group were given an intraperitoneal injection of Etanercept. Rats were harvested for biochemical and histopathological examinations. RESULTS The histopathological injury score of rats in the NEC + Placebo group was significantly higher compared to the NEC + Etanercept and Control groups (p < 0.05 for both comparisons). Tissue levels of tumor necrosis factor-α, interleukin-1β, and malondialdehyde were higher in the placebo group compared to the Etanercept group. CONCLUSION Our results suggest that Etanercept attenuates intestinal tissue damage in NEC by reducing inflammation and blocking the production of free-oxygen radicals, while also reducing tissue levels of tumor necrosis factor-α and interleukin-1β.
Collapse
Affiliation(s)
- Sadık Yurttutan
- Neonatal Intensive Care Unit, Zekai Tahir Burak Maternity Teaching Hospital, 06110, Hamamönü/Ankara, Turkey,
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Hunter CJ, De Plaen IG. Inflammatory signaling in NEC: Role of NF-κB, cytokines and other inflammatory mediators. ACTA ACUST UNITED AC 2013; 21:55-65. [PMID: 24388163 DOI: 10.1016/j.pathophys.2013.11.010] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Catherine J Hunter
- Department of Surgery, Division of Pediatric Surgery, Northwestern University Feinberg School of Medicine, Ann and Robert H. Lurie Children's Hospital of Chicago, United States
| | - Isabelle G De Plaen
- Department of Pediatrics, Division of Neonatology, Northwestern University Feinberg School of Medicine, Ann and Robert H. Lurie Children's Hospital of Chicago, 225 E Chicago Avenue, Box 45, Chicago, IL 60611, United States.
| |
Collapse
|
32
|
Mucosal loss with increased expression of IL-6, IL-8, and COX-2 in a formula-feeding only neonatal rat model of necrotizing enterocolitis. J Pediatr Surg 2013; 48:2301-7. [PMID: 24210203 DOI: 10.1016/j.jpedsurg.2013.04.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 04/08/2013] [Accepted: 04/14/2013] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The aim of our study is to establish a reliable neonatal rat model by formula feeding only for evaluation of early surgical intervention on the course of experimental necrotizing enterocolitis (NEC). MATERIAL AND METHODS Newborn Sprague-Dawley rats were divided into 50 breast-fed (group 1) and 38 formula fed (Similac/Esbilac, group 2) animals. The pups were sacrificed on the 4th, 5th, and 6th day of life and the terminal intestine examined for macroscopic and histologic changes as well as cytokine expression. RESULTS The histological mucosal damage was significantly higher of group 2 compared to group 1. The area of the vital mucosa of group 2 was significantly (58.57%, p<0.001) lower compared to group 1 (75.12%). The mRNA expression of the inflammatory cytokines IL-6, IL-8 and COX-2 was significantly 2-, 5- and 10-fold increased in group 2 compared to group 1. DISCUSSION Formula fed newborn rats displayed an inflammatory enterocolitis similar to human NEC. Our study demonstrates a significant loss of mucosa in animals with NEC having increased expression levels of IL-6, IL-8 and COX-2. Mucosal loss appears to be a distinct feature of experimental NEC and has to be correlated with the human disease.
Collapse
|
33
|
Nurses' perception of milk temperature at delivery compared to actual practice in the neonatal intensive care unit. Adv Neonatal Care 2013; 13:E1-E10. [PMID: 24042145 DOI: 10.1097/anc.0b013e3182a14cbd] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE The purposes of this study were to examine nurses' perception of feeding temperature practices and to compare the nurses' temperature estimation with the measured temperature of milk at the time of delivery to the infant. DESIGN A descriptive exploratory study was conducted in 3 level III neonatal intensive care units (NICUs). SUBJECTS A convenience sample of nurses from 3 level III NICUs in the Midwest. In addition, temperatures from bottle/syringe samples of formula/breast milk were measured and recorded. METHODS The Feeding Practices and Temperature Survey, a 10-item survey measuring nurses' perception of the effect of feeding temperature on infant condition, was distributed to subjects. Afterward for select feedings, researchers recorded the type of milk, delivery method, nurses' estimated temperature of the milk, and the measured infrared temperature of milk just before feeding delivery. To compare perception with actual practice patterns, the measured milk temperature was compared with the nurses' estimated temperature, standard room temperature, and body temperature using descriptive statistics of the survey responses and t test comparisons. MAIN OUTCOME MEASURES/PRINCIPAL RESULTS A total of 141 surveys were analyzed. More than 50% of respondents reported feeding temperature as clinically very significant. A range of 35.5°C to 37.2°C was reported as the ideal temperature of breast milk at delivery. Recordings of 419 temperatures were used for analysis. Measured milk temperature just before feeding ranged from 22°C to 46.4°C. The mean measured temperatures were 31.0°C (SD = 2.8°C) for warmed milk in a bottle and 30.5°C (SD = 2.5°C) for milk warmed in a syringe. The measured milk temperature and the nurse-estimated temperature were significantly lower than body temperature (P = 0.000) and significantly higher than room temperature (P = 0.000). CONCLUSION Current warming methods yield wide variation in milk temperature. Nurses' estimation of milk temperature was not consistent with measured temperature at the time of delivery. Future research is needed to establish guidelines for feeding temperature standardization assisting nurses to enhance evidence-based feeding practices.
Collapse
|
34
|
Rowland KJ, Choi PM, Warner BW. The role of growth factors in intestinal regeneration and repair in necrotizing enterocolitis. Semin Pediatr Surg 2013; 22:101-11. [PMID: 23611614 PMCID: PMC3635039 DOI: 10.1053/j.sempedsurg.2013.01.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Necrotizing enterocolitis (NEC) is a devastating intestinal disease resulting in major neonatal morbidity and mortality. The pathology is poorly understood, and the means of preventing and treating NEC are limited. Several endogenous growth factors have been identified as having important roles in intestinal growth as well as aiding intestinal repair from injury or inflammation. In this review, we will discuss several growth factors as mediators of intestinal regeneration and repair as well as potential therapeutic agents for NEC.
Collapse
Affiliation(s)
| | | | - Brad W. Warner
- Correspondence: Brad W. Warner, M.D. St. Louis Children's Hospital One Children's Place; Suite 5S40 St. Louis MO 63110 (314) 454-6022 - Phone (314) 454-2442 – Fax
| |
Collapse
|
35
|
Benkoe T, Baumann S, Weninger M, Pones M, Reck C, Rebhandl W, Oehler R. Comprehensive evaluation of 11 cytokines in premature infants with surgical necrotizing enterocolitis. PLoS One 2013; 8:e58720. [PMID: 23472217 PMCID: PMC3589358 DOI: 10.1371/journal.pone.0058720] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 02/05/2013] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE A prospective study to investigate the pattern of pro- and anti-inflammatory cytokine responses in neonates with surgical necrotizing enterocolitis (NEC) and identify those cytokines being the most promising for future research. METHODS A panel of 11 different cytokines were measured in 9 infants with proven NEC and compared with 18 age-matched healthy neonates. RESULTS The serum concentrations of the interleukins (IL)-6, IL-8, and IL-10 were significantly (32-fold to 56-fold) higher in NEC infants compared with controls. In contrast, IL-5, IFN gamma, IL-4 and IL-2 showed slightly (1.4-fold to 5.9-fold) lower levels in the NEC samples. However, these cytokines showed a very low absolute concentration in infants with NEC and in controls. The sum of the serum concentrations of IL-6, IL-8 and IL-10 was able to clearly separate infants with NEC from control samples. IL-1 beta and TNF-alpha showed no statistically different levels. The serum levels of TNF-beta and IL-12p70 were below the detection limit in more than 50% of all samples per group. CONCLUSION In spite of strong local inflammation only three out of eleven cytokines (IL-6, IL-8, and IL-10) showed strongly increased serum levels indicating an important role of them in the pathogenesis of NEC. At least two of these three cytokines were elevated in every single NEC patient. Thus, longitudinal monitoring of combined IL-8, IL-6, and IL-10 levels could reveal their potency in being clinical relevant markers in NEC.
Collapse
Affiliation(s)
- Thomas Benkoe
- Department of Pediatric Surgery, Medical University of Vienna, Vienna, Austria.
| | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
The pathogenesis of necrotizing enterocolitis (NEC) is complex and its speed of progression is variable. To gain understanding of the disease, researchers have examined tissues resected from patients with NEC; however, as these are obtained at late stages of the disease, they do not yield clues about the early pathogenic events leading to NEC. Therefore, animal models are used and have helped identify a role for several mediators of the inflammatory network in NEC. In this article, we discuss the evidence for the role of these inflammatory mediators and conclude with a current unifying hypothesis regarding NEC pathogenesis.
Collapse
Affiliation(s)
- Isabelle G. De Plaen
- Associate Professor of Pediatrics, Department of Pediatrics, Division of Neonatology, Northwestern University Feinberg School of Medicine, Children’s Hospital of Chicago Research Center, Ann and Robert H. Lurie Children’s Hospital of Chicago, 225 E. Chicago Ave, Box 45, Chicago, IL 60611-2605, U.S.A., Tel: (773)-755-6379; fax: (312)-227-9758
| |
Collapse
|
37
|
Cherrington NJ, Estrada TE, Frisk HA, Canet MJ, Hardwick RN, Dvorak B, Lux K, Halpern MD. The hepatic bile acid transporters Ntcp and Mrp2 are downregulated in experimental necrotizing enterocolitis. Am J Physiol Gastrointest Liver Physiol 2013; 304:G48-56. [PMID: 23125159 PMCID: PMC3543632 DOI: 10.1152/ajpgi.00317.2012] [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
Necrotizing enterocolitis (NEC) is the most common gastrointestinal emergency of premature infants and is characterized by an extensive hemorrhagic inflammatory necrosis of the distal ileum and proximal colon. We have previously shown that, during the development of experimental NEC, the liver plays an important role in regulating inflammation in the ileum, and accumulation of ileal bile acids (BA) along with dysregulation of ileal BA transporters contributes to ileal damage. Given these findings, we speculated that hepatic BA transporters would also be altered in experimental NEC. Using both rat and mouse models of NEC, levels of Cyp7a1, Cyp27a1, and the hepatic BA transporters Bsep, Ntcp, Oatp2, Oatp4, Mrp2, and Mrp3 were investigated. In addition, levels of hepatic BA transporters were also determined when the proinflammatory cytokines tumor necrosis factor (TNF)-α and interleukin (IL)-18, which are both elevated in NEC, are neutralized during disease development. Ntcp and Mrp2 were decreased in NEC, but elevated ileal BA levels were not responsible for these reductions. However, neutralization of TNF-α normalized Ntcp, whereas removal of IL-18 normalized Mrp2 levels. These data show that the hepatic transporters Ntcp and Mrp2 are downregulated, whereas Cyp27a1 is increased in rodent models of NEC. Furthermore, increased levels of TNF-α and IL-18 in experimental NEC may play a role in the regulation of Ntcp and Mrp2, respectively. These data suggest the gut-liver axis should be considered when therapeutic modalities for NEC are developed.
Collapse
Affiliation(s)
- Nathan J. Cherrington
- 1Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona; and
| | - Teresa E. Estrada
- 2Department of Pediatrics and the Steele Children's Research Center, University of Arizona, Tucson Arizona
| | - Harrison A. Frisk
- 2Department of Pediatrics and the Steele Children's Research Center, University of Arizona, Tucson Arizona
| | - Mark J. Canet
- 1Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona; and
| | - Rhiannon N. Hardwick
- 1Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona; and
| | - Bohuslav Dvorak
- 2Department of Pediatrics and the Steele Children's Research Center, University of Arizona, Tucson Arizona
| | - Katie Lux
- 2Department of Pediatrics and the Steele Children's Research Center, University of Arizona, Tucson Arizona
| | - Melissa D. Halpern
- 2Department of Pediatrics and the Steele Children's Research Center, University of Arizona, Tucson Arizona
| |
Collapse
|
38
|
Kim EK, Lee KY, Lee HJ, Lee JA, Choi CW, Kim HS, Kim BI, Choi JH. Heat shock pretreatment reduces intestinal injury in a neonatal rat model of early necrotizing enterocolitis. Neonatology 2013; 103:1-6. [PMID: 22948204 DOI: 10.1159/000339179] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 04/30/2012] [Indexed: 12/20/2022]
Abstract
BACKGROUND Increased pro-inflammatory cytokines are suggested in the pathogenesis of necrotizing enterocolitis (NEC). The transcription factor, nuclear factor-ĸB (NF-ĸB), is a central regulator of inflammatory and immune responses, and recent rodent NEC models have shown that NF-ĸB may have a critical role in the disease processes that underlie NEC. Heat shock proteins have important functions in response to stress-related events in a variety of systems, including digestive organs. OBJECTIVES We investigated whether heat shock pretreatment protects intestinal epithelial damage in the early NEC rat model. METHODS We generated human NEC-like lesions in neonatal rat ileum by administering oral endotoxin (10 mg/kg), intermittent 8% hypoxia, and hypertonic formula. Heat shock was administered by raising the chamber temperature to 42°C for 20 min, 3 h prior to endotoxin ingestion. RESULTS Heat shock pretreatment increased the expression of HSP70 in the ileal tissue and attenuated histological severity of early experimental NEC. NF-ĸB was activated in the ileal tissue of the NEC group and this activation was attenuated by heat shock pretreatment, which was determined by electrophoretic mobility shift assay and Western blot analysis of p50 in subcellular fractionated samples. CONCLUSIONS Heat shock pretreatment reduced the incidence and severity of early experimental NEC in rats. A possible mechanism underlying this protective effect includes inhibition of NF-ĸB activation through increased HSP70 expression.
Collapse
Affiliation(s)
- Ee-Kyung Kim
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Mirpuri J, Sotnikov I, Myers L, Denning TL, Yarovinsky F, Parkos CA, Denning PW, Louis NA. Lactobacillus rhamnosus (LGG) regulates IL-10 signaling in the developing murine colon through upregulation of the IL-10R2 receptor subunit. PLoS One 2012; 7:e51955. [PMID: 23272193 PMCID: PMC3525658 DOI: 10.1371/journal.pone.0051955] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Accepted: 11/07/2012] [Indexed: 12/23/2022] Open
Abstract
The intestinal microflora is critical for normal development, with aberrant colonization increasing the risk for necrotizing enterocolitis (NEC). In contrast, probiotic bacteria have been shown to decrease its incidence. Multiple pro- and anti-inflammatory cytokines have been identified as markers of intestinal inflammation, both in human patients with NEC and in models of immature intestine. Specifically, IL-10 signaling attenuates intestinal responses to gut dysbiosis, and disruption of this pathway exacerbates inflammation in murine models of NEC. However, the effects of probiotics on IL-10 and its signaling pathway, remain poorly defined. Real-time PCR profiling revealed developmental regulation of MIP-2, TNF-α, IL-12, IL-10 and the IL-10R2 subunit of the IL-10 receptor in immature murine colon, while the expression of IL-6 and IL-18 was independent of postnatal age. Enteral administration of the probiotic Lactobacillus rhamnosus GG (LGG) down-regulated the expression of TNF-α and MIP-2 and yet failed to alter IL-10 mRNA and protein expression. LGG did however induce mRNA expression of the IL-10R2 subunit of the IL-10 receptor. IL-10 receptor activation has been associated with signal transducer and activator of transcription (STAT) 3-dependent induction of members of the suppressors of cytokine signaling (SOCS) family. In 2 week-old mice, LGG also induced STAT3 phosphorylation, increased colonic expression of SOCS-3, and attenuated colonic production of MIP-2 and TNF-α. These LGG-dependent changes in phosphoSTAT3, SOCS3, MIP-2 and TNF-α were all inhibited by antibody-mediated blockade of the IL-10 receptor. Thus LGG decreased baseline proinflammatory cytokine expression in the developing colon through upregulation of IL-10 receptor-mediated signaling, most likely due to the combined induction of phospho-STAT3 and SOCS3. Furthermore, LGG-dependent increases in IL-10R2 were associated with reductions in TNF-α, MIP-2 and disease severity in a murine model of intestinal injury in the immature colon.
Collapse
Affiliation(s)
- Julie Mirpuri
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Emory University, Atlanta, Georgia, United States of America
- Department of Pediatrics, University of Texas Southwestern, Dallas, Texas, United States of America
- * E-mail: (JM); (NAL)
| | - Ilya Sotnikov
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Loren Myers
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Timothy L. Denning
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Emory University, Atlanta, Georgia, United States of America
- Epithelial Pathobiology Unit, Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Felix Yarovinsky
- Department of Immunology, University of Texas Southwestern, Dallas, Texas, United States of America
| | - Charles A. Parkos
- Epithelial Pathobiology Unit, Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Patricia W. Denning
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Nancy A. Louis
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Emory University, Atlanta, Georgia, United States of America
- * E-mail: (JM); (NAL)
| |
Collapse
|
40
|
Coursodon-Boyiddle CF, Snarrenberg CL, Adkins-Rieck CK, Bassaganya-Riera J, Hontecillas R, Lawrence P, Brenna JT, Jouni ZE, Dvorak B. Pomegranate seed oil reduces intestinal damage in a rat model of necrotizing enterocolitis. Am J Physiol Gastrointest Liver Physiol 2012; 303:G744-51. [PMID: 22821948 PMCID: PMC3468537 DOI: 10.1152/ajpgi.00248.2012] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Pomegranate seed oil (PSO), which is the major source of conjugated linolenic acids such as punicic acid (PuA), exhibits strong anti-inflammatory properties. Necrotizing enterocolitis (NEC) is a devastating disease associated with severe and excessive intestinal inflammation. The aim of this study was to evaluate the effects of orally administered PSO on the development of NEC, intestinal epithelial proliferation, and cytokine regulation in a rat model of NEC. Premature rats were divided into three groups: dam fed (DF), formula-fed rats (FF), or rats fed with formula supplemented with 1.5% of PSO (FF + PSO). All groups were exposed to asphyxia/cold stress to induce NEC. Intestinal injury, epithelial cell proliferation, cytokine production, and trefoil factor 3 (Tff3) production were evaluated in the terminal ileum. Oral administration of PSO (FF+PSO) decreased the incidence of NEC from 61 to 26%. Feeding formula with PSO improved enterocyte proliferation in the site of injury. Increased levels of proinflammatory IL-6, IL-8, IL-12, IL-23, and TNF-α in the ileum of FF rats were normalized in PSO-treated animals. Tff3 production in the FF rats was reduced compared with DF but not further affected by the PSO. In conclusion, administration of PSO protects against NEC in the neonatal rat model. This protective effect is associated with an improvement of intestinal epithelial homeostasis and a strong anti-inflammatory effect of PSO on the developing intestinal mucosa.
Collapse
Affiliation(s)
| | - Chelsea L. Snarrenberg
- 1Department of Pediatrics and Steele Children's Research Center, The University of Arizona, Tucson, Arizona;
| | - Camille K. Adkins-Rieck
- 1Department of Pediatrics and Steele Children's Research Center, The University of Arizona, Tucson, Arizona;
| | - Josep Bassaganya-Riera
- 2Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia;
| | - Raquel Hontecillas
- 2Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia;
| | - Peter Lawrence
- 3Division of Nutritional Sciences, Cornell University, Ithaca, New York;
| | - J. Thomas Brenna
- 3Division of Nutritional Sciences, Cornell University, Ithaca, New York;
| | | | - Bohuslav Dvorak
- 1Department of Pediatrics and Steele Children's Research Center, The University of Arizona, Tucson, Arizona;
| |
Collapse
|
41
|
Patel BK, Shah JS. Necrotizing enterocolitis in very low birth weight infants: a systemic review. ISRN GASTROENTEROLOGY 2012; 2012:562594. [PMID: 22997587 PMCID: PMC3444861 DOI: 10.5402/2012/562594] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 06/19/2012] [Indexed: 01/08/2023]
Abstract
Necrotizing enterocolitis (NEC) is the most common serious gastrointestinal disorder affecting very preterm or very low birth weight infants. The risk is inversely proportional to gestational age and weight at birth. Fetal growth restriction and compromise may be additional specific risk factors. Despite extensive research and animal studies etiopathogenesis, preventive strategies and management options remain controversial. The present paper reviews the literature for recent advances and newer insights for changing epidemiological trends, pathogenesis, role of inflammatory cytokines, and various preventive and management strategies.
Collapse
Affiliation(s)
- Bhoomika K. Patel
- Department of Clinical Pharmacy, Shri Sarvajanik Pharmacy College, Near Arvind Baug, Gujarat, Mehsana 384001, India
| | - Jigna S. Shah
- Department of Clinical Pharmacy, Shri Sarvajanik Pharmacy College, Near Arvind Baug, Gujarat, Mehsana 384001, India
| |
Collapse
|
42
|
Wu SF, Caplan M, Lin HC. Necrotizing enterocolitis: old problem with new hope. Pediatr Neonatol 2012; 53:158-63. [PMID: 22770103 DOI: 10.1016/j.pedneo.2012.04.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Revised: 09/23/2011] [Accepted: 04/26/2012] [Indexed: 12/11/2022] Open
Abstract
The incidence of necrotizing enterocolitis (NEC) and mortality rate associated with this disease are not decreasing despite more than three decades of intensive research investigation and advances in neonatal intensive care. Although the etiology of NEC is not clearly elucidated, the most accepted hypothesis at present is that enteral feeding in the presence of intestinal hypoxia-ischemia-reperfusion, and colonization with pathogens provokes an inappropriately accentuated inflammatory response by the immature intestinal epithelial cells of the preterm neonate. However, delayed colonization of commensal flora with dysbiotic flora with a predominance of pathologic microorganisms plays a fundamental role in the pathogenesis of NEC. Recent studies have further identified that NEC infants have less diverse flora compared to age-matched controls without NEC. Increased gastric residual volume may be an early sign of NEC. An absolute neutrophil count of <1.5 × 10(9)/L and platelets below 100 × 10(9)/L are associated with an increased risk for mortality and gastrointestinal morbidity. Nonspecific supportive medical management should be initiated promptly. Sudden changes in vital signs such as tachycardia or impending shock may indicate perforation. A recent meta-analysis investigating using probiotics for prevention of NEC with a total of 2176 preterm very low birth weight infants found a success rate of just 1/25. Careful monitoring of the residual volume, and of serious changes in hemograms and vital signs may help in early diagnosis and prediction of when to perform medical or early surgical intervention. In term of prevention, administration of oral probiotics containing Bifidobacterium and Lactobacillus is a simple and safe method that attempts to early establish of commensal flora balance to inhibit pathogenic flora and an inflammatory response.
Collapse
Affiliation(s)
- Shu-Fen Wu
- Department of Pediatrics, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | | | | |
Collapse
|
43
|
Abstract
PURPOSE OF REVIEW This review will summarize the clinical and experimental studies evaluating the role of epidermal growth factor (EGF) in prophylaxis and treatment of necrotizing enterocolitis (NEC). RECENT FINDINGS Clinical studies have suggested the importance of EGF in protection of the intestine against NEC, as well as its safety for infants suffering from NEC. The recent experimental studies identified the molecular mechanisms EGF uses for intestinal protection, which involves regulation of intestinal epithelial homeostasis and barrier function. Further studies are necessary to identify the optimal dose, timing, and route of administration of EGF to NEC patients. No clinical studies are currently underway. SUMMARY NEC is a devastating problem for preterm neonates, but the exact disease pathogenesis remains unclear. Growing clinical evidence supports the use of EGF as a predictive marker of NEC and its use for prevention and treatment of NEC. In addition, experimental data indicate potential mechanisms of EGF prevention against NEC. These include reduction of inflammation, improvement of barrier function, and regulation of epithelial apoptosis and autophagy.
Collapse
|
44
|
Liu Y, Fatheree NY, Mangalat N, Rhoads JM. Lactobacillus reuteri strains reduce incidence and severity of experimental necrotizing enterocolitis via modulation of TLR4 and NF-κB signaling in the intestine. Am J Physiol Gastrointest Liver Physiol 2012; 302:G608-17. [PMID: 22207578 PMCID: PMC3311308 DOI: 10.1152/ajpgi.00266.2011] [Citation(s) in RCA: 135] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Necrotizing enterocolitis (NEC) is the leading gastrointestinal cause of mortality and morbidity in the premature infant. Premature infants have a delay in intestinal colonization by commensal bacteria and colonization with potentially pathogenic organisms. Lactobacillus reuteri is a probiotic that inhibits enteric infections, modulates the immune system, and may be beneficial to prevent NEC. In previous studies, L. reuteri strains DSM 17938 and ATCC PTA 4659 differentially modulated inflammation in vitro; however, the strains had equivalent anti-inflammatory responses in LPS feeding-induced ileitis in neonatal rats in vivo. The impact of these two strains in the prevention of NEC has not been previously investigated. NEC was induced in newborn rats by orogastric formula feeding and exposure to hypoxia. L. reuteri was added to the formula to prevent NEC. NEC score, Toll-like receptor (TLR)-signaling genes, phospho-IκB activity, and cytokine levels in the intestine were examined. Both strains significantly increased survival rate and decreased the incidence and severity of NEC, with optimal effects from DSM 17938. In response to probiotic, mRNA expression of IL-6, TNF-α, TLR4, and NF-κB was significantly downregulated, while mRNA levels of anti-inflammatory cytokine IL-10 were significantly upregulated. In parallel, L. reuteri treatment led to decrease intestinal protein levels of TLR4 and cytokine levels of TNF-α and IL-1β in newborn rats with NEC. Both strains significantly inhibited not only intestinal LPS-induced phospho-IκB activity in an ex vivo study but also decreased the levels of phospho-IκB in the intestines of NEC rat model. Cow milk formula feeding produced a similar but milder proinflammatory profile in the intestine that was also ameliorated by 17938. Our studies demonstrate that each of the two L. reuteri strains has potential therapeutic value in our NEC model and in enteritis associated with cow milk feeding. These results support the concept that L. reuteri may represent a valuable treatment to prevent NEC.
Collapse
Affiliation(s)
- Yuying Liu
- Department of Pediatrics, Division of Gastroenterology, University of Texas Health Science Center at Houston Medical School, Houston, Texas
| | - Nicole Y. Fatheree
- Department of Pediatrics, Division of Gastroenterology, University of Texas Health Science Center at Houston Medical School, Houston, Texas
| | - Nisha Mangalat
- Department of Pediatrics, Division of Gastroenterology, University of Texas Health Science Center at Houston Medical School, Houston, Texas
| | - Jon Marc Rhoads
- Department of Pediatrics, Division of Gastroenterology, University of Texas Health Science Center at Houston Medical School, Houston, Texas
| |
Collapse
|
45
|
Zhang C, Sherman MP, Prince LS, Bader D, Weitkamp JH, Slaughter JC, McElroy SJ. Paneth cell ablation in the presence of Klebsiella pneumoniae induces necrotizing enterocolitis (NEC)-like injury in the small intestine of immature mice. Dis Model Mech 2012; 5:522-32. [PMID: 22328592 PMCID: PMC3380715 DOI: 10.1242/dmm.009001] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a leading cause of morbidity and mortality in premature infants. During NEC pathogenesis, bacteria are able to penetrate innate immune defenses and invade the intestinal epithelial layer, causing subsequent inflammation and tissue necrosis. Normally, Paneth cells appear in the intestinal crypts during the first trimester of human pregnancy. Paneth cells constitute a major component of the innate immune system by producing multiple antimicrobial peptides and proinflammatory mediators. To better understand the possible role of Paneth cell disruption in NEC, we quantified the number of Paneth cells present in infants with NEC and found that they were significantly decreased compared with age-matched controls. We were able to model this loss in the intestine of postnatal day (P)14-P16 (immature) mice by treating them with the zinc chelator dithizone. Intestines from dithizone-treated animals retained approximately half the number of Paneth cells compared with controls. Furthermore, by combining dithizone treatment with exposure to Klebsiella pneumoniae, we were able to induce intestinal injury and inflammatory induction that resembles human NEC. Additionally, this novel Paneth cell ablation model produces NEC-like pathology that is consistent with other currently used animal models, but this technique is simpler to use, can be used in older animals that have been dam fed, and represents a novel line of investigation to study NEC pathogenesis and treatment.
Collapse
Affiliation(s)
- Chunxian Zhang
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
|
47
|
Ran-Ressler RR, Khailova L, Arganbright KM, Adkins-Rieck CK, Jouni ZE, Koren O, Ley RE, Brenna JT, Dvorak B. Branched chain fatty acids reduce the incidence of necrotizing enterocolitis and alter gastrointestinal microbial ecology in a neonatal rat model. PLoS One 2011; 6:e29032. [PMID: 22194981 PMCID: PMC3237582 DOI: 10.1371/journal.pone.0029032] [Citation(s) in RCA: 149] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 11/19/2011] [Indexed: 01/05/2023] Open
Abstract
Introduction Branched chain fatty acids (BCFA) are found in the normal term human newborn's gut, deposited as major components of vernix caseosa ingested during late fetal life. We tested the hypothesis that premature infants' lack of exposure to gastrointestinal (GI) BCFA is associated with their microbiota and risk for necrotizing enterocolitis (NEC) using a neonatal rat model. Methods Pups were collected one day before scheduled birth. The pups were exposed to asphyxia and cold stress to induce NEC. Pups were assigned to one of three experimental treatments. DF (dam-fed) ; Control, hand-fed rat milk substitute ; BCFA, hand-fed rat milk substitute with 20%w/w BCFA. Total fat was equivalent (11%wt) for both the Control and BCFA groups. Cecal microbiota were characterized by 16S rRNA gene pyrosequencing, and intestinal injury, ileal cytokine and mucin gene expression, interleukin-10 (IL-10) peptide immunohistochemistry, and BCFA uptake in ileum phospholipids, serum and liver were assessed. Results NEC incidence was reduced by over 50% in the BCFA group compared to the Control group as assessed in ileal tissue; microbiota differed among all groups. BCFA-fed pups harbored greater levels of BCFA-associated Bacillus subtilis and Pseudomonas aeruginosa compared to Controls. Bacillus subtilis levels were five-fold greater in healthy pups compared to pups with NEC. BCFA were selectively incorporated into ileal phospholipids, serum and liver tissue. IL-10 expression increased three-fold in the BCFA group versus Controls and no other inflammatory or mucosal mRNA markers changed. Conclusion At constant dietary fat level, BCFA reduce NEC incidence and alter microbiota composition. BCFA are also incorporated into pup ileum where they are associated with enhanced IL-10 and may exert other specific effects.
Collapse
Affiliation(s)
- Rinat R. Ran-Ressler
- Division of Nutritional Sciences, Savage Hall, Cornell University, Ithaca, New York, United States of America
| | - Ludmila Khailova
- Department of Pediatrics, University of Arizona, Tucson, Arizona, United States of America
| | - Kelly M. Arganbright
- Department of Pediatrics, University of Arizona, Tucson, Arizona, United States of America
| | | | - Zeina E. Jouni
- Mead Johnson Nutrition, Evansville, Indiana, United States of America
| | - Omry Koren
- Department of Microbiology, Cornell University, Ithaca, New York, United States of America
| | - Ruth E. Ley
- Department of Microbiology, Cornell University, Ithaca, New York, United States of America
| | - J. Thomas Brenna
- Division of Nutritional Sciences, Savage Hall, Cornell University, Ithaca, New York, United States of America
- * E-mail: (JTB); (BD)
| | - Bohuslav Dvorak
- Department of Pediatrics, University of Arizona, Tucson, Arizona, United States of America
- * E-mail: (JTB); (BD)
| |
Collapse
|
48
|
Martin NA, Mount Patrick SK, Estrada TE, Frisk HA, Rogan DT, Dvorak B, Halpern MD. Active transport of bile acids decreases mucin 2 in neonatal ileum: implications for development of necrotizing enterocolitis. PLoS One 2011; 6:e27191. [PMID: 22162748 PMCID: PMC3230578 DOI: 10.1371/journal.pone.0027191] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Accepted: 10/11/2011] [Indexed: 12/18/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is the most common gastrointestinal emergency of premature infants, but its etiology remains unclear. We have previously shown that mucin 2 (Muc2) positive goblet cells are significantly decreased in NEC. We have also shown that ileal bile acids (BAs) are significantly increased during the development of this disease. Because BAs can affect mucins, we hypothesized that elevated ileal BAs contribute to decreased Muc2 in experimental NEC. The role of Muc2 in NEC was evaluated in Winnie +/+ mice, a strain that produces aberrant Muc2. Muc2 and trefoil factor 3 (Tff3) were assessed in neonatal rats subjected to the NEC protocol when bile acids were removed, and in ileal explants from newborn and older rats cultured with and without BAs. Further, the role of active transport of BAs was determined using neonatal rats given the apical sodium dependent bile acid transporter (Asbt) inhibitor SC-435 and in neonatal Asbt knockout mice subjected to the NEC protocol. Mice with aberrant Muc2 had significantly greater incidence and severity of NEC. Using both in vivo and ex vivo techniques, we determined that BAs decrease Muc2 positive cells in neonatal but not older ileum. However, Tff3 positive cells are not decreased by BAs. In addition, active transport of BAs is required for BAs to decrease Muc2 in immature ileum. These data show that functional Muc2 plays a critical role in the prevention of NEC and BAs can potentiate the decreased Muc2 in disease development. Further, BAs have a more profound effect on Muc2 in immature versus older ileum, which may explain at least in part why NEC occurs almost exclusively in premature infants.
Collapse
Affiliation(s)
- Nina A. Martin
- Department of Pediatrics and Steele Children's Research Center, University of Arizona, Tucson, Arizona
| | - Sarah K. Mount Patrick
- Department of Pediatrics and Steele Children's Research Center, University of Arizona, Tucson, Arizona
| | - Teresa E. Estrada
- Department of Pediatrics and Steele Children's Research Center, University of Arizona, Tucson, Arizona
| | - Harrison A. Frisk
- Department of Pediatrics and Steele Children's Research Center, University of Arizona, Tucson, Arizona
| | - Daniel T. Rogan
- Department of Pediatrics and Steele Children's Research Center, University of Arizona, Tucson, Arizona
| | - Bohuslav Dvorak
- Department of Pediatrics and Steele Children's Research Center, University of Arizona, Tucson, Arizona
| | - Melissa D. Halpern
- Department of Pediatrics and Steele Children's Research Center, University of Arizona, Tucson, Arizona
- * E-mail:
| |
Collapse
|
49
|
Okogbule-Wonodi AC, Gross GW, Sun CCJ, Agthe AG, Xiao L, Waites KB, Viscardi RM. Necrotizing enterocolitis is associated with ureaplasma colonization in preterm infants. Pediatr Res 2011; 69:442-7. [PMID: 21258263 PMCID: PMC3968774 DOI: 10.1203/pdr.0b013e3182111827] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The study objective was to determine whether Ureaplasma respiratory tract colonization of preterm infants <33 wk gestation is associated with an increased risk for necrotizing enterocolitis (NEC). One or more tracheal or nasopharyngeal aspirates for Ureaplasma culture and PCR were obtained during the first week of life from 368 infants <33 wk gestation enrolled from 1999 to 2003 or from 2007 to 2009. NEC Bell stage ≥ 2 was confirmed by radiological criteria, and pathology, if available. Cord serum samples were analyzed for IL-6 and IL-1β concentrations, and placentas were reviewed for histological chorioamnionitis in the first cohort. NEC was confirmed in 29 of 368 (7.9%) of the combined cohorts. The incidence of NEC was 2.2-fold higher in Ureaplasma-positive (12.3%) than Ureaplasma-negative (5.5%) infants <33 wk (OR, 2.43; 95% CI, 1.13-5.2; p = 0.023) and 3.3-fold higher in Ureaplasma-positive (14.6%) than Ureaplasma-negative (4.4%) infants ≤ 28 wk (OR, 3.67; 95% CI, 1.36-9.93; p = 0.01). Age of onset, hematologic parameters at onset, and NEC severity were similar between Ureaplasma-positive and negative infants. Cord serum IL-6 and IL-1β concentrations were significantly higher in Ureaplasma-positive than in Ureaplasma-negative NEC-affected infants. Ureaplasma may be a factor in NEC pathogenesis in preterm infants by contributing to intestinal mucosal injury and/or altering systemic or local immune responses.
Collapse
Affiliation(s)
- Adora C Okogbule-Wonodi
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Macrophage-produced IL-12p70 mediates hemorrhage-induced damage in a complement-dependent manner. Shock 2011; 35:134-40. [PMID: 20577145 DOI: 10.1097/shk.0b013e3181ed8ec9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Hemorrhage and hemorrhagic shock instigate intestinal damage and inflammation. Multiple components of the innate immune response, including complement and neutrophil infiltration, are implicated in this pathology. To investigate the interaction of complement activation and other components of the innate immune response during hemorrhage, we treated mice after hemorrhage with CR2-fH, a targeted inhibitor of the alternative complement pathway and assessed intestinal damage and inflammation 2 h after hemorrhage. In wild-type mice, CR2-fH attenuated hemorrhage-induced, midjejunal damage and inflammation as determined by decreased mucosal damage, macrophage infiltration, leukotriene B4, IL-12p40, and TNF-[alpha] production. The critical nature of intestinal macrophage infiltration and activation in the response to hemorrhage was further determined using mice pretreated with clodronate-containing liposomes. The absence of either macrophages or IL-12p70 attenuated intestinal damage. These data suggest that complement activation and macrophage infiltration with IL-12p70 production are critical to hemorrhage-induced midjejunal damage and inflammation.
Collapse
|