1
|
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: 0.5] [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
|
2
|
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: 1.0] [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
|
3
|
Chen TT, Lv JJ, Chen L, Gao YW, Liu LP. Role of heparinase in the gastrointestinal dysfunction of sepsis (Review). Exp Ther Med 2022; 23:119. [PMID: 34970342 PMCID: PMC8713170 DOI: 10.3892/etm.2021.11042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 10/26/2021] [Indexed: 11/23/2022] Open
Abstract
Heparinase (HPA) is a β-D glucuronidase that belongs to the endoglycosidase enzyme family, and plays an important role in numerous pathological and physiological processes, including inflammation, angiogenesis and tumor metastasis. When the expression of HPA is abnormally high, the side chain of heparin sulfate proteoglycans degrades, destroying the cell barrier and leading to the occurrence and development of inflammation, with systemic inflammation occurring in severe cases. Sepsis is a major cause of mortality in critically ill patients. In sepsis, the gastrointestinal tract is the first and most frequently involved target organ, which often leads to gastrointestinal dysfunction. HPA overexpression has been determined to accelerate sepsis progression and gastrointestinal dysfunction; thus, it was hypothesized that HPA may play an important role and may serve as an index for the diagnosis of gastrointestinal dysfunction in sepsis. HPA inhibitors may therefore become applicable as targeted drugs for the treatment of gastrointestinal dysfunction in patients with sepsis. The present review mainly discussed the role of HPA in gastrointestinal dysfunction of sepsis.
Collapse
Affiliation(s)
- Ting-Ting Chen
- The First Clinical Medical School of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Jia-Jun Lv
- The First Clinical Medical School of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Ling Chen
- Department of Emergency Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
- Department of Emergency, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Yu-Wei Gao
- Department of Emergency Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
- Department of Emergency, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Li-Ping Liu
- Department of Emergency Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
- Department of Emergency, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
4
|
Maheshwari A, Traub TM, Garg PM, Ethawi Y, Buonocore G. Necrotizing Enterocolitis: Clinical Features, Histopathological Characteristics, and Genetic Associations. Curr Pediatr Rev 2022; 18:210-225. [PMID: 35125082 DOI: 10.2174/1573396318666220204113858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 11/25/2021] [Accepted: 11/30/2021] [Indexed: 11/22/2022]
Abstract
Necrotizing enterocolitis (NEC) is an inflammatory bowel necrosis seen in premature infants. Although the etiopathogenesis of NEC is unclear, genetic factors may alter a patient's susceptibility, clinical course, and outcomes. This review draws from existing studies focused on individual genes and others based on microarray-based high-throughput discovery techniques. We have included evidence from our own studies and from an extensive literature search in the databases PubMed, EMBASE, and Scopus. To avoid bias in the identification of studies, keywords were short-listed a priori from anecdotal experience and PubMed's Medical Subject Heading (MeSH) thesaurus.
Collapse
Affiliation(s)
| | - Terri M Traub
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Parvesh M Garg
- Global Newborn Society, Clarksville, Maryland, USA.,Department of Pediatrics, University of Mississippi, Jackson, Mississippi, USA
| | - Yahya Ethawi
- Global Newborn Society, Clarksville, Maryland, USA.,Department of Pediatrics, Saudi American Hospital, Ajman, United Arab Emirates
| | - Giuseppe Buonocore
- Global Newborn Society, Clarksville, Maryland, USA.,Department of Pediatrics/ Neonatology, University of Siena, Siena, Italy
| |
Collapse
|
5
|
Identification of the Molecular Basis of Nanocurcumin-Induced Telocyte Preservation within the Colon of Ulcerative Colitis Rat Model. Mediators Inflamm 2021; 2021:7534601. [PMID: 34373677 PMCID: PMC8349286 DOI: 10.1155/2021/7534601] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 06/10/2021] [Accepted: 06/25/2021] [Indexed: 12/25/2022] Open
Abstract
Background Telocytes (TCs) are a distinct type of interstitial cells that play a vital role in the pathogenesis of ulcerative colitis and colonic tissue hemostasis. The aim of this study was to examine the effect of nanocurcumin (NC) on the morphometric and immunohistochemical characterization of TCs in the ulcerative colitis (UC) rat model. Methods Forty rats were randomly divided into control, NC, UC, and UC+NC groups. At the end of the experiment, the colon was dissected and prepared for histopathological and immunohistochemical assessment. Tissue homogenates were prepared for real-time PCR assessment of interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), and transforming growth factor-beta (TGF-β) gene expression. Our results revealed extensive mucosal damage with inflammatory cell infiltration, significant reduction of CD34, and vimentin immunostained TCs in the colon of the UC group with significant elevation of expression of IL-6, TNF-α, and TGF-β. The UC+NC-treated group revealed significant elevation of TC count compared to the UC group besides, a significant reduction of the three gene expression. Conclusion NC successfully targeted the colonic tissue, improved the mucosal lesion, preserve TCs distribution, and count through its anti-inflammatory and fibrinolytic properties.
Collapse
|
6
|
Marônek M, Marafini I, Gardlík R, Link R, Troncone E, Monteleone G. Metalloproteinases in Inflammatory Bowel Diseases. J Inflamm Res 2021; 14:1029-1041. [PMID: 33790618 PMCID: PMC8001665 DOI: 10.2147/jir.s288280] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 03/09/2021] [Indexed: 12/13/2022] Open
Abstract
Inflammatory bowel diseases (IBD) are chronic inflammatory diseases of the gastrointestinal tract, encompassing two main disorders: Crohn's disease (CD) and ulcerative colitis (UC). In both these pathologies, excessive and local immune response against luminal antigens promotes a pathological process leading to various degrees of gut damage. Matrix metalloproteinases (MMPs) are a family of neutral proteases with the ability to degrade all components of extracellular matrix. In physiological conditions, MMPs are produced at very low level and generally in the latent form and are involved in the normal tissue turnover. Their function is inhibited by tissue inhibitors of metalloproteinases (TIMPs). However, in inflamed tissue of IBD patients, MMPs are produced in excess and/or the activity of TIMPs is not sufficient to block MMPs, thereby making a major contribution to the IBD-related mucosal degradation. In this review, we summarize the available evidence on the expression and role of MMPs in IBD.
Collapse
Affiliation(s)
- Martin Marônek
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, Bratislava, 81108, Slovakia
| | - Irene Marafini
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, 00133, Italy
| | - Roman Gardlík
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, Bratislava, 81108, Slovakia
| | - René Link
- Institute of Experimental Medicine, Faculty of Medicine, University of Pavol Jozef Šafárik, Košice, 040 11, Slovakia
| | - Edoardo Troncone
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, 00133, Italy
| | - Giovanni Monteleone
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, 00133, Italy
| |
Collapse
|
7
|
Jiang YN, Muk T, Stensballe A, Nguyen DN, Sangild PT, Jiang PP. Early Protein Markers of Necrotizing Enterocolitis in Plasma of Preterm Pigs Exposed to Antibiotics. Front Immunol 2020; 11:565862. [PMID: 33133078 PMCID: PMC7578346 DOI: 10.3389/fimmu.2020.565862] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/25/2020] [Indexed: 12/19/2022] Open
Abstract
Background: Most hospitalized preterm infants receive antibiotics in the first days of life to prevent or treat infections. Short-term, early antibiotic treatment may also prevent the microbiota-dependent gut inflammatory disorder, necrotizing enterocolitis (NEC). It remains a challenge to predict NEC, and a few early blood diagnostic markers exist. Using preterm pigs as model for infants, blood parameters and plasma proteins affected by early progression of NEC were profiled in preterm pigs subjected to oral, systemic, or no antibiotics after preterm birth. Methods: Preterm newborn pigs were treated with saline (CON) or antibiotics (ampicillin, gentamicin, and metronidazole) given enterally (ENT) or parenterally (PAR), and fed formula for 4 days to induce variable microbiome-dependent sensitivities to NEC. The gut was collected for macroscopic scoring of NEC lesions and blood for hematology, blood biochemistry, and LC/MS-based plasma proteomics. Statistical modeling was applied to detect plasma proteins affected by NEC and/or antibiotics. Results: Analyzed across different antibiotic regimens, NEC progression was associated with altered blood parameters and abundance of 89 plasma proteins that were functionally involved in extracellular membrane destruction, lipid metabolism, coagulopathy, and acute phase response. Large NEC-related changes were observed in abundance of RBP4, FGA, AHSG, C5, PTPRG, and A-1-antichymotrypsin 2, indicating potential serving as early markers of NEC. Conversely, antibiotic treatment, independent of NEC, affected only 4 proteins with main differences found between ENT and CON pigs. Conclusion: Early postnatal development of NEC lesions is associated with marked plasma protein changes that may be used for early NEC diagnosis.
Collapse
Affiliation(s)
- Yan-Nan Jiang
- School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Tik Muk
- Department of Veterinary and Animal Sciences, Section for Comparative Paediatrics and Nutrition, University of Copenhagen, Frederiksberg, Denmark
| | - Allan Stensballe
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Duc Ninh Nguyen
- Department of Veterinary and Animal Sciences, Section for Comparative Paediatrics and Nutrition, University of Copenhagen, Frederiksberg, Denmark
| | - Per Torp Sangild
- Department of Veterinary and Animal Sciences, Section for Comparative Paediatrics and Nutrition, University of Copenhagen, Frederiksberg, Denmark.,Department of Neonatology, Rigshospitalet, Copenhagen, Denmark
| | - Ping-Ping Jiang
- School of Public Health, Sun Yat-sen University, Guangzhou, China.,Department of Veterinary and Animal Sciences, Section for Comparative Paediatrics and Nutrition, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
8
|
Bertani L, Antonioli L, Fornai M, Tapete G, Baiano Svizzero G, Marchi S, Blandizzi C, Costa F. Evaluation of cytokine levels as putative biomarkers to predict the pharmacological response to biologic therapy in inflammatory bowel diseases. MINERVA GASTROENTERO 2019; 65:298-308. [PMID: 31646851 DOI: 10.23736/s1121-421x.19.02621-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cytokines play a central role in the pathogenesis of inflammatory bowel diseases. For this reason, the vast majority of biological therapies are aimed to block pro-inflammatory cytokines or their receptors. Although these drugs have modified the course of the disease due to their efficacy, a high rate of non-response or loss of response over time is still an important issue for clinicians. In this perspective, many studies have been conducted in recent years to individuate a reliable biomarker of therapeutic response. In this review, we discuss the role of cytokines involved in the pathogenesis and in the therapy of inflammatory bowel diseases, and their putative use as pharmacological biomarkers of therapy responsiveness.
Collapse
Affiliation(s)
- Lorenzo Bertani
- Unit of Gastroenterology, Department of Translational Research and New Technologies in Medicine and Surgery, Pisa University Hospital, Pisa, Italy -
| | - Luca Antonioli
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Matteo Fornai
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gherardo Tapete
- Unit of Gastroenterology, Department of Translational Research and New Technologies in Medicine and Surgery, Pisa University Hospital, Pisa, Italy
| | - Giovanni Baiano Svizzero
- Unit of Gastroenterology, Department of Translational Research and New Technologies in Medicine and Surgery, Pisa University Hospital, Pisa, Italy
| | - Santino Marchi
- Unit of Gastroenterology, Department of Translational Research and New Technologies in Medicine and Surgery, Pisa University Hospital, Pisa, Italy
| | - Corrado Blandizzi
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Francesco Costa
- Unit of Inflammatory Bowel Diseases, Department of General Surgery and Gastroenterology, Pisa University Hospital, Pisa, Italy
| |
Collapse
|
9
|
MohanKumar K, Namachivayam K, Song T, Jake Cha B, Slate A, Hendrickson JE, Pan H, Wickline SA, Oh JY, Patel RP, He L, Torres BA, Maheshwari A. A murine neonatal model of necrotizing enterocolitis caused by anemia and red blood cell transfusions. Nat Commun 2019; 10:3494. [PMID: 31375667 PMCID: PMC6677753 DOI: 10.1038/s41467-019-11199-5] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 06/27/2019] [Indexed: 12/23/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is an idiopathic, inflammatory bowel necrosis of premature infants. Clinical studies have linked NEC with antecedent red blood cell (RBC) transfusions, but the underlying mechanisms are unclear. Here we report a neonatal murine model to investigate this association. C57BL/6 mouse pups rendered anemic by timed phlebotomy and then given RBC transfusions develop NEC-like intestinal injury with prominent necrosis, inflammation, and submucosal edema/separation of the lamina propria in the ileocecal region and colon within 12-24 h. The anemic intestine is infiltrated by inflammatory macrophages, which are activated in situ by RBC transfusions via a Toll-like receptor (TLR)-4-mediated mechanism and cause bowel injury. Chelation of RBC degradation products with haptoglobin, absence of TLR4, macrophage depletion, and inhibition of macrophage activation is protective. Intestinal injury worsens with increasing severity and the duration of anemia prior to transfusion, indicating a need for the re-evaluation of current transfusion guidelines for premature infants.
Collapse
MESH Headings
- Anemia/complications
- Anemia/therapy
- Animals
- Animals, Newborn
- Cecum/pathology
- Colon/pathology
- Disease Models, Animal
- Enterocolitis, Necrotizing/etiology
- Enterocolitis, Necrotizing/pathology
- Erythrocyte Transfusion/adverse effects
- Humans
- Ileum/pathology
- Infant, Newborn
- Infant, Newborn, Diseases/etiology
- Infant, Newborn, Diseases/pathology
- Infant, Premature
- Intestinal Mucosa/pathology
- Macrophages/immunology
- Macrophages/metabolism
- Mice
- Toll-Like Receptor 4/immunology
- Toll-Like Receptor 4/metabolism
Collapse
Affiliation(s)
- Krishnan MohanKumar
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Kopperuncholan Namachivayam
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Tanjing Song
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Byeong Jake Cha
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Andrea Slate
- Department of Comparative Medicine, University of South Florida, Tampa, FL, 33612, USA
- Center for Comparative Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Jeanne E Hendrickson
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, 06520, USA
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Hua Pan
- Department of Cardiology, Morsani College of Medicine, University of South Florida, Tampa, FL, 33629, USA
| | - Samuel A Wickline
- Department of Cardiology, Morsani College of Medicine, University of South Florida, Tampa, FL, 33629, USA
| | - Joo-Yeun Oh
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Rakesh P Patel
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Ling He
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Benjamin A Torres
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Akhil Maheshwari
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA.
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD, 21287, USA.
| |
Collapse
|
10
|
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: 108] [Impact Index Per Article: 18.0] [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
|
11
|
Managlia E, Liu SXL, Yan X, Tan XD, Chou PM, Barrett TA, De Plaen IG. Blocking NF-κB Activation in Ly6c + Monocytes Attenuates Necrotizing Enterocolitis. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:604-618. [PMID: 30593820 PMCID: PMC6412404 DOI: 10.1016/j.ajpath.2018.11.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 11/06/2018] [Accepted: 11/28/2018] [Indexed: 12/31/2022]
Abstract
Necrotizing enterocolitis (NEC) is a devastating disease affecting premature infants with intestinal inflammation and necrosis. The neonatal intestinal inflammatory response is rich in macrophages, and blood monocyte count is low in human NEC. We previously found that NF-κB mediates the intestinal injury in experimental NEC. However, the role of NF-κB in myeloid cells during NEC remains unclear. Herein, inhibitor of kappaB kinase β (IKKβ), a critical kinase mediating NF-κB activation, was deleted in lysozyme M (Lysm)-expressing cells, which were found to be Cd11b+Ly6c+ monocytes but not Cd11b+Ly6c- macrophages in the dam-fed neonatal mouse intestine. NEC induced differentiation of monocytes into intestinal macrophages and up-regulation of monocyte recruitment genes (eg, L-selectin) in the macrophage compartment in wild-type mice, but not in pups with IKKβ deletion in Lysm+ cells. Thus, NF-κB is required for NEC-induced monocyte activation, recruitment, and differentiation in neonatal intestines. Furthermore, pups with Lysm-IKKβ deletion had improved survival and decreased incidence of severe NEC compared with littermate controls. Decreased NEC severity was not associated with an improved intestinal barrier. In contrast, NEC was unabated in mice with IKKβ deletion in intestinal epithelial cells. Together, these data suggest that recruitment of Ly6c+ monocytes into the intestine, NF-κB activation in these cells, and differentiation of Ly6c+ monocytes into macrophages are critical cellular and molecular events in NEC development to promote disease.
Collapse
Affiliation(s)
- Elizabeth Managlia
- Center for Intestinal and Liver Inflammation Research, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois; Division of Neonatology, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Shirley X L Liu
- Center for Intestinal and Liver Inflammation Research, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois; Division of Neonatology, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Xiaocai Yan
- Center for Intestinal and Liver Inflammation Research, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois; Division of Neonatology, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Xiao-Di Tan
- Center for Intestinal and Liver Inflammation Research, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois; Division of Gastroenterology, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois; Department of Pathology, Ann & Robert H. Lurie Children's Hospital of Chicago, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Pauline M Chou
- Department of Pathology, Ann & Robert H. Lurie Children's Hospital of Chicago, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Terrence A Barrett
- Division of Gastroenterology, Department of Internal Medicine, University of Kentucky, Lexington, Kentucky
| | - Isabelle G De Plaen
- Center for Intestinal and Liver Inflammation Research, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois; Division of Neonatology, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
12
|
Ginzel M, Feng X, Kuebler JF, Klemann C, Yu Y, von Wasielewski R, Park JK, Hornef MW, Vieten G, Ure BM, Kaussen T, Gosemann JH, Mayer S, Suttkus A, Lacher M. Dextran sodium sulfate (DSS) induces necrotizing enterocolitis-like lesions in neonatal mice. PLoS One 2017; 12:e0182732. [PMID: 28817583 PMCID: PMC5560643 DOI: 10.1371/journal.pone.0182732] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 07/24/2017] [Indexed: 12/12/2022] Open
Abstract
Background Necrotizing enterocolitis (NEC) is an inflammatory bowel disease of preterm human newborns with yet unresolved etiology. An established neonatal murine model for NEC employs oral administration of lipopolysaccharides (LPS) combined with hypoxia/hypothermia. In adult mice, feeding dextran sodium sulfate (DSS) represents a well-established model for experimental inflammatory bowel disease. Here we investigated the effect of DSS administration on the neonatal murine intestine in comparison with the established NEC model. Methods 3-day-old C57BL/6J mice were either fed formula containing DSS or LPS. LPS treated animals were additionally stressed by hypoxia/hypothermia twice daily. After 72 h, mice were euthanized, their intestinal tissue harvested and analyzed by histology, qRT-PCR and flow cytometry. For comparison, adult C57BL/6J mice were fed with DSS for 8 days and examined likewise. Untreated, age matched animals served as controls. Results Adult mice treated with DSS exhibited colonic inflammation with significantly increased Cxcl2 mRNA expression. In contrast, tissue inflammation in neonatal mice treated with DSS or LPS plus hypoxia/hypothermia was present in colon and small intestine as well. Comparative analysis of neonatal mice revealed a significantly increased lesion size and intestinal Cxcl2 mRNA expression after DSS exposure. Whereas LPS administration mainly induced local neutrophil recruitment, DSS treated animals displayed increased monocytes/macrophages infiltration. Conclusions Our study demonstrates the potential of DSS to induce NEC-like lesions accompanied by a significant humoral and cellular immune response in the small and large intestine of neonatal mice. The new model therefore represents a good alternative to LPS plus hypoxia/hypothermia administration requiring no additional physical stress.
Collapse
Affiliation(s)
- Marco Ginzel
- Center of Pediatric Surgery, Hannover Medical School, Hannover, Germany
- Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
| | - Xiaoyan Feng
- Center of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | | | - Christian Klemann
- Center of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | - Yi Yu
- Center of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | | | - Joon-Keun Park
- Center of Pediatric Surgery, Hannover Medical School, Hannover, Germany
- Department of Nephrology, Hannover Medical School, Hannover, Germany
| | | | - Gertrud Vieten
- Center of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | - Benno M. Ure
- Center of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | - Torsten Kaussen
- Department of Pediatric Cardiology and Pediatric Intensive Care, Hannover, Germany
| | | | - Steffi Mayer
- Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
| | - Anne Suttkus
- Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
| | - Martin Lacher
- Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
- * E-mail:
| |
Collapse
|
13
|
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
|
14
|
The viral dsRNA analogue poly (I:C) induces necrotizing enterocolitis in neonatal mice. Pediatr Res 2016; 79:596-602. [PMID: 26679153 DOI: 10.1038/pr.2015.261] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 09/29/2015] [Indexed: 11/08/2022]
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is a life-threatening gastrointestinal disease in premature infants with high mortality and morbidity with uncertain pathogenesis. Recent research focused on the role of intraluminal bacteria and lipopolysaccharide (LPS). However, an additional role of viral agents in the pathogenesis of NEC has recently been postulated. We assessed the role of polyinosinic:polycytidylic acid (pIC) mimicking viral dsRNA in contributing to the development of NEC in neonatal mice. METHODS Four-d-old C57BL/6J pups were stressed by asphyxia and hypothermia twice daily. Animals were either fed by formula only (FO), formula containing LPS or pIC. After 72 h, mice were euthanized, intestines harvested, and the severity of NEC was assessed. RESULTS Breastfed mice showed no evidence of NEC. Very mild NEC-like lesions were observed in mice fed by FO. Supplementation of LPS or pIC to the formula led to increased intestinal tissue damage and inflammation compared with FO in a similar manner. CONCLUSION Our study demonstrates the ability of viral factors to induce NEC in neonatal mice even in the absence of LPS. Furthermore, we present a new mouse model of pIC-induced NEC which may be used to obtain further mechanistic insights in the pathogenesis of this disease.
Collapse
|
15
|
Nadendla K, Sutton A, Kelly D, Dimmitt R, Wilkinson L, Harmon C, Martin C. Quintuplets: 5 Cases of NEC. Are There Other Risk Factors? Fetal Pediatr Pathol 2016; 35:425-433. [PMID: 27551982 DOI: 10.1080/15513815.2016.1214197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE To review a case of quintuplets with all babies developing necrotizing enterocolitis. METHODS A retrospective study of preterm quintuplets all developing necrotizing enterocolitis. Clinical outcomes were reviewed. RESULTS Quintuplets were born at 24 weeks gestation. Each baby developed NEC and was treated. One baby died. Currently the remaining 4 infants are on full enteral nutrition. CONCLUSION Further studies are needed to better understand this emerging population of multiple birth pregnancy and the frequency of NEC development.
Collapse
Affiliation(s)
- Kavita Nadendla
- a Surgery , University of Alabama at Birmingham , Birmingham , Alabama , USA
| | - Amelia Sutton
- b Department of Maternal Fetal Medicine , University of Alabama at Birmingham , Birmingham , Alabama , USA
| | - David Kelly
- c Pathology , Children's Hospital of Alabama , Birmingham , Alabama , USA
| | - Reed Dimmitt
- d Gastroenterology and Surgery , University of Alabama at Birmingham , Birmingham , Alabama , USA
| | - Linda Wilkinson
- a Surgery , University of Alabama at Birmingham , Birmingham , Alabama , USA
| | - Carroll Harmon
- e Surgery , University of Buffalo , Buffalo , New York , USA
| | - Colin Martin
- a Surgery , University of Alabama at Birmingham , Birmingham , Alabama , USA
| |
Collapse
|
16
|
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
|
17
|
TIMP-1 inhibition of occludin degradation in Caco-2 intestinal cells: a potential protective role in necrotizing enterocolitis. Pediatr Res 2015; 77:649-55. [PMID: 25665057 DOI: 10.1038/pr.2015.26] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 10/30/2014] [Indexed: 01/06/2023]
Abstract
BACKGROUND Necrotizing enterocolitis (NEC), a common intestinal disease affecting premature infants, is a major cause of morbidity and mortality. Previous reports indicate an upregulation of intestinal matrix metalloproteinases (MMPs) activity that may play key roles on the higher permeability of the intestinal barrier, typical to NEC. Recently, TIMP-1, a natural inhibitor of MMP's, was found to be over expressed in preterm human breast milk (HBM). Previous studies have shown that infants fed with HBM have a significant reduction in the incidence of NEC. The aim of the present study was to investigate the possible role that TIMP-1 may play on the maintenance of tight junctions and therefore the gut barrier integrity. METHODS Timp-1-treated Caco-2 intestinal cells were tested for MMP-2 enzymatic activity and cell junction integrity. RESULTS TIMP-1 inhibited MMP-2 activity, which induced a significant increase in the expression of occludin but not of claudin-4. TIMP-1 did not affect apoptosis. CONCLUSION One of the putative mechanisms associated with HBM protection against NEC is mediated by TIMP-1, which downregulates MMP-2 activity, inhibits the degradation of occluding, and preserves tight junctions and gut barrier integrity.
Collapse
|
18
|
Genome-wide expression profiles of necrotizing enterocolitis versus spontaneous intestinal perforation in human intestinal tissues: dysregulation of functional pathways. Ann Surg 2015; 260:1128-37. [PMID: 24368664 DOI: 10.1097/sla.0000000000000374] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To provide a comprehensive database of gene regulation and compare differentially regulated molecular networks in human tissues of necrotizing enterocolitis (NEC) and spontaneous intestinal perforation (SIP). BACKGROUND Both NEC and SIP are devastating surgical emergencies associated with high morbidity and mortality in preterm infants. Their pathophysiology and molecular mechanisms remain unclear. METHODS Differential whole genome microarray analysis was performed on intestinal tissues collected from NEC (n = 15) and SIP (n = 12) infants and compared with tissues collected from surgical-control patients with noninflammatory intestinal conditions (n = 14). Validation of 52 target gene expressions was performed by quantitative polymerase chain reaction. Regulatory networks of significantly affected genes were constructed according to functional pathways. RESULTS Extensive and significant changes of gene expression were observed in NEC tissues, which comprised multiple pathways of angiogenesis, arginine metabolism, cell adhesion and chemotaxis, extracellular matrix remodeling, hypoxia and oxidative stress, inflammation, and muscle contraction. These dysregulated genes could be networked downstream of key receptors, TLR2, TLR4, and TREM1, and mediated via NF-κB, AP-1, and HIF1A transcription factor pathways, indicating predominant microbial and inflammatory involvement. In contrast, SIP tissues exhibited much milder and less diversified expressional changes, with target genes significantly associated with G-protein-mediated muscle contraction and extracellular matrix remodeling. CONCLUSIONS The molecular evidence suggests that NEC and SIP are likely 2 different diseases caused by distinct etiology and pathophysiology. This first comprehensive database on differential gene expression profiles of human NEC and SIP tissues could lead to development of disease-specific diagnostic and prognostic biomarkers and new therapeutic strategies for improving outcomes.
Collapse
|
19
|
Is there new hope for therapeutic matrix metalloproteinase inhibition? Nat Rev Drug Discov 2014; 13:904-27. [DOI: 10.1038/nrd4390] [Citation(s) in RCA: 524] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
20
|
Premkumar MH, Sule G, Nagamani SC, Chakkalakal S, Nordin A, Jain M, Ruan MZ, Bertin T, Dawson B, Zhang J, Schady D, Bryan NS, Campeau PM, Erez A, Lee B. Argininosuccinate lyase in enterocytes protects from development of necrotizing enterocolitis. Am J Physiol Gastrointest Liver Physiol 2014; 307:G347-54. [PMID: 24904080 PMCID: PMC4121640 DOI: 10.1152/ajpgi.00403.2013] [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), the most common neonatal gastrointestinal emergency, results in significant mortality and morbidity, yet its pathogenesis remains unclear. Argininosuccinate lyase (ASL) is the only enzyme in mammals that is capable of synthesizing arginine. Arginine has several homeostatic roles in the gut and its deficiency has been associated with NEC. Because enterocytes are the primary sites of arginine synthesis in neonatal mammals, we evaluated the consequences of disruption of arginine synthesis in the enterocytes on the pathogenesis of NEC. We devised a novel approach to study the role of enterocyte-derived ASL in NEC by generating and characterizing a mouse model with enterocyte-specific deletion of Asl (Asl(flox/flox); VillinCre(tg/+), or CKO). We hypothesized that the presence of ASL in a cell-specific manner in the enterocytes is protective in the pathogenesis of NEC. Loss of ASL in enterocytes resulted in an increased incidence of NEC that was associated with a proinflammatory state and increased enterocyte apoptosis. Knockdown of ASL in intestinal epithelial cell lines resulted in decreased migration in response to lipopolysaccharide. Our results show that enterocyte-derived ASL has a protective role in NEC.
Collapse
Affiliation(s)
- M. H. Premkumar
- 1Division of Neonatology, Texas Children's Hospital, Baylor College of Medicine;
| | - G. Sule
- 2Department of Molecular and Human Genetics, Baylor College of Medicine;
| | - S. C. Nagamani
- 2Department of Molecular and Human Genetics, Baylor College of Medicine;
| | - S. Chakkalakal
- 2Department of Molecular and Human Genetics, Baylor College of Medicine;
| | - A. Nordin
- 2Department of Molecular and Human Genetics, Baylor College of Medicine;
| | - M. Jain
- 2Department of Molecular and Human Genetics, Baylor College of Medicine;
| | - M. Z. Ruan
- 2Department of Molecular and Human Genetics, Baylor College of Medicine;
| | - T. Bertin
- 2Department of Molecular and Human Genetics, Baylor College of Medicine;
| | - B. Dawson
- 2Department of Molecular and Human Genetics, Baylor College of Medicine;
| | - J. Zhang
- 2Department of Molecular and Human Genetics, Baylor College of Medicine;
| | - D. Schady
- 3Department of Pathology, Texas Children's Hospital, Houston;
| | - N. S. Bryan
- 4University of Texas Health Science Center at Houston, Texas;
| | - P. M. Campeau
- 2Department of Molecular and Human Genetics, Baylor College of Medicine;
| | - A. Erez
- 5Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel; and
| | - B. Lee
- 2Department of Molecular and Human Genetics, Baylor College of Medicine; ,6Howard Hughes Medical Institute, Houston, Texas
| |
Collapse
|
21
|
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: 2.8] [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
|
22
|
Cytokines associated with necrotizing enterocolitis in extremely-low-birth-weight infants. Pediatr Res 2014; 76:100-8. [PMID: 24732104 PMCID: PMC4062583 DOI: 10.1038/pr.2014.48] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Accepted: 01/01/2014] [Indexed: 11/09/2022]
Abstract
BACKGROUND The goal was to identify cytokines associated with necrotizing enterocolitis (NEC). Based on our earlier reports of decreased tissue expression of transforming growth factor (TGF)-β, we hypothesized that infants with NEC also have low blood TGF-β levels. We further hypothesized that because fetal inflammation increases the risk of NEC, infants who develop NEC have elevated blood cytokine levels in early neonatal period. METHODS Data on 104 extremely-low-birth-weight infants with NEC and 893 without NEC from 17 centers were analyzed. Clinical information was correlated with blood cytokine levels on postnatal day 1 (D1), D3, D7, D14, and D21. RESULTS Male gender, non-Caucasian/non-African American ethnicity, sepsis, lower blood TGF-β and interleukin (IL)-2 levels, and higher IL-8 levels were associated with NEC. The NEC group had lower TGF-β levels than controls since D1. The diagnosis of NEC was associated with elevated IL-1β, IL-6, IL-8, IL-10, monocyte chemoattractant protein-1/CC-motif ligand-2, macrophage inflammatory protein-1β/CC-motif ligand-3, and C-reactive protein. CONCLUSION Clinical characteristics, such as gender and ethnicity, and low blood TGF-β levels are associated with higher risk of NEC. Infants who developed NEC did not start with high blood levels of inflammatory cytokines, but these rose mainly after the onset of NEC.
Collapse
|
23
|
Xu J, Anderson V, Schwarz SM. Dietary GD3 ganglioside reduces the incidence and severity of necrotizing enterocolitis by sustaining regulatory immune responses. J Pediatr Gastroenterol Nutr 2013; 57:550-556. [PMID: 23783008 DOI: 10.1097/mpg.0b013e3182a027e1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVES Gangliosides are glycosphingolipids, rich in colostrum and in membrane microdomains, which promote enterocyte growth and differentiation, and modulate TH1/TH2 responses. In an in vitro intestinal explant model of necrotizing enterocolitis (NEC), gangliosides have been shown to ameliorate intestinal injury; however, possible immunomodulatory mechanisms associated with this observation, as well as potential in vivo protective effects of gangliosides, remain unknown. The present study evaluates the effects of dietary GD3, the predominant ganglioside in neonatal rat intestine, both on the clinicopathologic expression of disease and on ileal Foxp3+ T regulatory cell immune responses in an experimental NEC model. METHODS Newborn rat pups were fed gavage formula (NEC) or formula supplemented with 15 μg/mL GD3 (GD3-NEC). Dam-fed (DF) littermates served as controls. NEC was induced by asphyxia and cold stress. At 96 hours, ileal gross and histologic changes were evaluated, and ileal cytokine profiles, Foxp3 expression, and Foxp3+ cell numbers were determined. RESULTS GD3 decreased the incidence and gross and histopathologic severity of NEC. Ileal Foxp3 expression and Foxp3+ cell numbers were significantly decreased in the NEC group compared with DF. GD3 increased ileal Foxp3 expression and Foxp3+ cell numbers, in association with upregulation of anti-inflammatory cytokine interleukin (IL)-10 and chemokines, tissue inhibitor of metalloproteinases 1, IL-1 receptor antagonist (IL-1ra), and suppressed proinflammatory mediators. CONCLUSIONS These data suggest that dietary GD3 protects newborn rats from NEC, in part, by augmenting mucosal Foxp3+ T regulatory immune responses.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
- Biomarkers/metabolism
- Cell Count
- Colostrum/chemistry
- Cytokines/metabolism
- Dietary Supplements
- Disease Progression
- Enterocolitis, Necrotizing/immunology
- Enterocolitis, Necrotizing/metabolism
- Enterocolitis, Necrotizing/pathology
- Enterocolitis, Necrotizing/prevention & control
- Forkhead Transcription Factors/metabolism
- Gangliosides/therapeutic use
- Ileum/immunology
- Ileum/metabolism
- Ileum/pathology
- Immunologic Factors/therapeutic use
- Intestinal Mucosa/immunology
- Intestinal Mucosa/metabolism
- Intestinal Mucosa/pathology
- Lymphocyte Activation
- Random Allocation
- Rats
- Rats, Sprague-Dawley
- Severity of Illness Index
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/pathology
- Up-Regulation
Collapse
Affiliation(s)
- Jiliu Xu
- *Departments of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition †Department of Pathology, SUNY-Downstate Medical Center, Brooklyn, NY
| | | | | |
Collapse
|
24
|
Chatterton DE, Nguyen DN, Bering SB, Sangild PT. Anti-inflammatory mechanisms of bioactive milk proteins in the intestine of newborns. Int J Biochem Cell Biol 2013; 45:1730-47. [DOI: 10.1016/j.biocel.2013.04.028] [Citation(s) in RCA: 251] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 04/25/2013] [Accepted: 04/28/2013] [Indexed: 01/04/2023]
|
25
|
Zhang HY, Wang F, Feng JX. Intestinal microcirculatory dysfunction and neonatal necrotizing enterocolitis. Chin Med J (Engl) 2013; 126:1771-1778. [PMID: 23652066 DOI: 10.3760/cma.j.issn.0366-6999.20121741] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2023] Open
Abstract
OBJECTIVE Based on the observation that coagulation necrosis occurs in the majority of neonatal necrotizing enterocolitis (NEC) patients, it is clear that intestinal ischemia is a contributing factor to the pathogenesis of NEC. However, the published studies regarding the role of intestinal ischemia in NEC are controversial. The aim of this paper is to review the current studies regarding intestinal microcirculatory dysfunction and NEC, and try to elucidate the exact role of intestinal microcirculatory dysfunction in NEC. DATA SOURCES The studies cited in this review were mainly obtained from articles listed in Medline and PubMed. The search terms used were "intestinal microcirculatory dysfunction" and "neonatal necrotizing enterocolitis". STUDY SELECTION Mainly original milestone articles and critical reviews written by major pioneer investigators in the field were selected. RESULTS Immature regulatory control of mesentery circulation makes the neonatal intestinal microvasculature vulnerable. When neonates are subjected to stress, endothelial cell dysfunction occurs and results in vasoconstriction of arterioles, inflammatory cell infiltration and activation in venules, and endothelial barrier disruption in capillaries. The compromised vasculature increases circulation resistance and therefore decreases intestinal perfusion, and may eventually progress to intestinal necrosis. CONCLUSION Intestinal ischemia plays an important role through the whole course of NEC. New therapeutic agents targeting intestinal ischemia, like HB-EGF, are promising therapeutic agents for the treatment of NEC.
Collapse
Affiliation(s)
- Hong-yi Zhang
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College and Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | | | | |
Collapse
|
26
|
MohanKumar K, Kaza N, Jagadeeswaran R, Garzon SA, Bansal A, Kurundkar AR, Namachivayam K, Remon JI, Bandepalli CR, Feng X, Weitkamp JH, Maheshwari A. Gut mucosal injury in neonates is marked by macrophage infiltration in contrast to pleomorphic infiltrates in adult: evidence from an animal model. Am J Physiol Gastrointest Liver Physiol 2012; 303:G93-102. [PMID: 22538401 PMCID: PMC3404576 DOI: 10.1152/ajpgi.00016.2012] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Necrotizing enterocolitis (NEC) is an inflammatory bowel necrosis of premature infants. In tissue samples of NEC, we identified numerous macrophages and a few neutrophils but not many lymphocytes. We hypothesized that these pathoanatomic characteristics of NEC represent a common tissue injury response of the gastrointestinal tract to a variety of insults at a specific stage of gut development. To evaluate developmental changes in mucosal inflammatory response, we used trinitrobenzene sulfonic acid (TNBS)-induced inflammation as a nonspecific insult and compared mucosal injury in newborn vs. adult mice. Enterocolitis was induced in 10-day-old pups and adult mice (n = 25 animals per group) by administering TNBS by gavage and enema. Leukocyte populations were enumerated in human NEC and in murine TNBS-enterocolitis using quantitative immunofluorescence. Chemokine expression was measured using quantitative polymerase chain reaction, immunoblots, and immunohistochemistry. Macrophage recruitment was investigated ex vivo using intestinal tissue-conditioned media and bone marrow-derived macrophages in a microchemotaxis assay. Similar to human NEC, TNBS enterocolitis in pups was marked by a macrophage-rich leukocyte infiltrate in affected tissue. In contrast, TNBS-enterocolitis in adult mice was associated with pleomorphic leukocyte infiltrates. Macrophage precursors were recruited to murine neonatal gastrointestinal tract by the chemokine CXCL5, a known chemoattractant for myeloid cells. We also demonstrated increased expression of CXCL5 in surgically resected tissue samples of human NEC, indicating that a similar pathway was active in NEC. We concluded that gut mucosal injury in the murine neonate is marked by a macrophage-rich leukocyte infiltrate, which contrasts with the pleomorphic leukocyte infiltrates in adult mice. In murine neonatal enterocolitis, macrophages were recruited to the inflamed gut mucosa by the chemokine CXCL5, indicating that CXCL5 and its cognate receptor CXCR2 merit further investigation as potential therapeutic targets in NEC.
Collapse
Affiliation(s)
- Krishnan MohanKumar
- Departments of Pediatrics, 1Division of Neonatology and ,2Center for Neonatal and Pediatric Gastrointestinal Disease, ,Departments of 3Pediatrics and
| | - Niroop Kaza
- 4Pathology, University of Alabama at Birmingham, Birmingham, Alabama;
| | - Ramasamy Jagadeeswaran
- Departments of Pediatrics, 1Division of Neonatology and ,2Center for Neonatal and Pediatric Gastrointestinal Disease,
| | | | | | | | - Kopperuncholan Namachivayam
- Departments of Pediatrics, 1Division of Neonatology and ,2Center for Neonatal and Pediatric Gastrointestinal Disease,
| | - Juan I. Remon
- Departments of Pediatrics, 1Division of Neonatology and ,2Center for Neonatal and Pediatric Gastrointestinal Disease,
| | - C. Rekha Bandepalli
- Departments of Pediatrics, 1Division of Neonatology and ,2Center for Neonatal and Pediatric Gastrointestinal Disease,
| | - Xu Feng
- 4Pathology, University of Alabama at Birmingham, Birmingham, Alabama;
| | - Joern-Hendrik Weitkamp
- 6Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Akhil Maheshwari
- Departments of Pediatrics, 1Division of Neonatology and ,2Center for Neonatal and Pediatric Gastrointestinal Disease, ,7Pharmacology, University of Illinois at Chicago, Chicago, Illinois; ,Departments of 3Pediatrics and
| |
Collapse
|
27
|
Abstract
BACKGROUND AND AIMS Matrix metalloproteinases (MMPs) are a group of endopeptidases that play a key role in the degradation of the extracellular matrix. The natural inhibitors of MMPs are the tissue inhibitors of metalloproteinases (TIMPs). It has been shown that several MMPs may be major factors in tissue destruction and remodeling in necrotizing enterocolitis. We designed the present prospective observational study to determine whether specific MMPs activity and expression of their inhibitors are similar in the milk fed to preterm and term infants. METHODS We compared specific matrix metalloproteinases (MMP-2 and MMP-9) and 1 MMP inhibitor (TIMP-1) activities or expression in human milk (HM) fed to 18 preterm infants and 13 full-term infants, obtained at 72 hours and 1 and 2 weeks postpartum. RESULTS MMP-2 and MMP-9 activities were similar in both groups and did not vary over time. TIMP-1 was significantly higher in preterm HM. TIMP-1 expression increased significantly over time exclusively in the preterm group. CONCLUSIONS There are differences in the expression of TIMP-1 between colostrum and mature milk in preterm HM and differences in the expression of TIMP-1 between preterm and term milk.
Collapse
|
28
|
Chuang SL, Hayes PJ, Ogundipe E, Haddad M, MacDonald TT, Fell JM. Cow's milk protein-specific T-helper type I/II cytokine responses in infants with necrotizing enterocolitis. Pediatr Allergy Immunol 2009; 20:45-52. [PMID: 18298426 DOI: 10.1111/j.1399-3038.2008.00729.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Enteral feeding, in particular with formula feeds, is associated with necrotizing enterocolitis (NEC). In this study, we have examined, in the systemic and mucosal immune compartments, for evidence of bovine milk antigen sensitization in infants with NEC. Eleven newborns with Bell's staging 2-3 NEC [median post-conceptional age 31 wk (range 27-41 wk)], 21 neonatal controls [33 (28-40) wk] and 15 infants undergoing intestinal resection or mucosal biopsy for non-inflammatory conditions [39 (34-42) wk] were studied. Spontaneous and antigen or mitogen elicited interferon-gamma (IFN-gamma) [T-helper type I (Th1)], interleukin (IL)-4 and IL-5 [T-helper type II (Th2)] responses were enumerated using single-cell enzyme-linked immunospot (ELISPOT) assay in peripheral blood (PBMC) or lamina propria mononuclear cells. NEC infants, compared with controls, showed a significant elevation in baseline PBMC cytokine secreting cells, vigorous mitogen responses (20- to 120-fold increase) for IFN-gamma, IL-4 and IL-5 (p < 0.001), strong responses to beta-lactoglobulin (betalg) (IFN-gamma > IL-4/IL-5, p < or = 0.001), and somewhat smaller casein responses. Similarly, in the lamina propria, a small but significant increase in spontaneous cytokine-secreting cells was detected in NEC infants (p < 0.01), with an IFN-gamma/IL-4 predominant phytohemagglutinin (PHA)/concanavalin-A (ConA) response. Three of nine NEC infants (but no controls) also showed a positive ELISPOT response to betalg (IFN-gamma only) but none to casein. We have thus demonstrated significant cow's milk protein (CMP) sensitization in NEC, at least in the systemic compartment (mixed Th1/Th2), with minimal mucosal activation in some cases. These novel findings provide a potential mechanism for a direct contributory role of CMP in the pathogenesis of NEC.
Collapse
Affiliation(s)
- S-L Chuang
- Department of Neonatology, Chelsea and Westminster Hospital, London, UK.
| | | | | | | | | | | |
Collapse
|
29
|
Schnabl KL, Aerde JEV, Thomson ABR, Clandinin MT. Necrotizing enterocolitis: A multifactorial disease with no cure. World J Gastroenterol 2008; 14:2142-61. [PMID: 18407587 PMCID: PMC2703838 DOI: 10.3748/wjg.14.2142] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Necrotizing enterocolitis is an inflammatory bowel disease of neonates with significant morbidity and mortality in preterm infants. Due to the multifactorial nature of the disease and limitations in disease models, early diagnosis remains challenging and the pathogenesis elusive. Although preterm birth, hypoxic-ischemic events, formula feeding, and abnormal bacteria colonization are established risk factors, the role of genetics and vasoactive/inflammatory mediators is unclear. Consequently, treatments do not target the specific underlying disease processes and are symptomatic and surgically invasive. Breast-feeding is the most effective preventative measure. Recent advances in the prevention of necrotizing enterocolitis have focused on bioactive nutrients and trophic factors in human milk. Development of new disease models including the aspect of prematurity that consistently predisposes neonates to the disease with multiple risk factors will improve our understanding of the pathogenesis and lead to discovery of innovative therapeutics.
Collapse
|
30
|
Abstract
Necrotizing enterocolitis (NEC) is a devastating intra-abdominal emergency in the newborn period. The disease involves bowel wall inflammation, ischemic necrosis, eventual perforation, and the need for urgent surgical intervention. Unrecognized or left untreated, the neonate can decompensate quickly, often progressing to shock, multisystem organ failure, and eventual death. During the past several years, a number of basic science and clinical trials have been established in an attempt to understand the pathophysiology of NEC. As many researchers feel that NEC develops as an uncontrolled inflammatory response that leads to intestinal ischemia, a large number of studies have been focused on the inflammatory cascade and the role that cytokines play within that cascade. Although a large amount of data has been generated from these studies, the events leading to the ischemic injury of the intestine are still not fully understood. This article will therefore focus on the key cytokines involved with NEC, in an attempt to present the current literature and studies that support their involvement.
Collapse
Affiliation(s)
- Troy A Markel
- Department of Surgery, Indiana University School of Medicine, the Indiana Center for Vascular Biology and Medicine, Indianapolis, IN, USA
| | | | | | | | | | | |
Collapse
|
31
|
Leaphart CL, Qureshi F, Cetin S, Li J, Dubowski T, Baty C, Batey C, Beer-Stolz D, Guo F, Murray SA, Hackam DJ. Interferon-gamma inhibits intestinal restitution by preventing gap junction communication between enterocytes. Gastroenterology 2007; 132:2395-411. [PMID: 17570214 DOI: 10.1053/j.gastro.2007.03.029] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2006] [Accepted: 03/01/2007] [Indexed: 12/17/2022]
Abstract
BACKGROUND & AIMS Necrotizing enterocolitis (NEC) is characterized by interferon-gamma (IFN-gamma) release and inadequate intestinal restitution. Because enterocytes migrate together, mucosal healing may require interenterocyte communication via connexin 43-mediated gap junctions. We hypothesize that enterocyte migration requires interenterocyte communication, that IFN impairs migration by impairing connexin 43, and that impaired healing during NEC is associated with reduced gap junctions. METHODS NEC was induced in Swiss-Webster or IFN(-/-) mice, and restitution was determined in the presence of the gap junction inhibitor oleamide, or via time-lapse microscopy of IEC-6 cells. Connexin 43 expression, trafficking, and localization were detected in cultured or primary enterocytes or mouse or human intestine by confocal microscopy and (35)S-labeling, and gap junction communication was assessed using live microscopy with oleamide or connexin 43 siRNA. RESULTS Enterocytes expressed connexin 43 in vitro and in vivo, and exchanged fluorescent dye via gap junctions. Gap junction inhibition significantly reduced enterocyte migration in vitro and in vivo. NEC was associated with IFN release and loss of enterocyte connexin 43 expression. IFN inhibited enterocyte migration by reducing gap junction communication through the dephosphorylation and internalization of connexin 43. Gap junction inhibition significantly increased NEC severity, whereas reversal of the inhibitory effects of IFN on gap junction communication restored enterocyte migration after IFN exposure. Strikingly, IFN(-/-) mice were protected from the development of NEC, and showed restored connexin 43 expression and intestinal restitution. CONCLUSIONS IFN inhibits enterocyte migration by preventing interenterocyte gap junction communication. Connexin 43 loss may provide insights into the development of NEC, in which restitution is impaired.
Collapse
Affiliation(s)
- Cynthia L Leaphart
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh and University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Pathophysiological molecules in the extracellular environment offer excellent targets that can be exploited for designing drug targeting systems. Matrix metalloproteases (MMPs) are a family of extracellular proteolytic enzymes that are characterized by their overexpression or overactivity in several pathologies. Over the last two decades, the MMP literature reveals heightened interest in the research involving MMP biology, pathology and targeting. This review describes various strategies that have been designed to utilize MMPs for targeting therapeutic entities. Key factors that need to be considered in the successful design of such systems have been identified based on the analyses of these strategies. Development of targeted drug delivery using MMPs has been steadily pursued; however, drug delivery efforts using these targets need to be intensified and focused to realize the clinical application of the fast developing fundamental MMP research.
Collapse
Affiliation(s)
- Deepali G. Vartak
- The University of Illinois, Department of Biopharmaceutical Sciences, Chicago, IL 60612 USA
| | - Richard A. Gemeinhart
- The University of Illinois, Department of Biopharmaceutical Sciences, Chicago, IL 60612 USA
- The University of Illinois, Department of Bioengineering, Chicago, IL 60612 USA
- Corresponding Author:Richard A. Gemeinhart, Ph.D., Assistant Professor of Pharmaceutics and Bioengineering and Director of Graduate Studies, Department of Biopharmaceutical Sciences, College of Pharmacy, University of Illinois, 833 South Wood Street (MC 865), Chicago, IL 60612-7231, Voice: (312) 996-2253, Facsimile: (312) 996-2784,
| |
Collapse
|
33
|
Markel TA, Crisostomo PR, Wang M, Herring CM, Lahm T, Meldrum KK, Lillemoe KD, Rescorla FJ, Meldrum DR. Iron chelation acutely stimulates fetal human intestinal cell production of IL-6 and VEGF while decreasing HGF: the roles of p38, ERK, and JNK MAPK signaling. Am J Physiol Gastrointest Liver Physiol 2007; 292:G958-63. [PMID: 17204543 DOI: 10.1152/ajpgi.00502.2006] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Bacteria have developed mechanisms to sequester host iron via chelators such as deferoxamine (DFO). Interestingly, DFO has been shown to stimulate acute intestinal epithelial cell inflammatory cytokine production in the absence of bacteria; however, this mechanism has not been elucidated. Intestinal epithelial cell production of IL-6 and TNF-alpha is elevated in various gastrointestinal pathologies, including acute intestinal ischemia. Similarly, VEGF and HGF are essential to intestinal epithelial cell integrity. Therapeutic strategies that decrease IL-6 and TNF-alpha while increasing VEGF and HGF therefore have theoretical appeal. We hypothesized that 1) fetal human intestinal epithelial cells acutely produce increased IL-6, TNF-alpha, VEGF, and HGF during iron chelation and 2) the MAPK pathway mediates these effects. Fetal human intestinal epithelial cells were stimulated by iron chelation (1 mM DFO) with and without p38 MAPK, ERK, or JNK inhibition. Supernatants were harvested after 24 h of incubation, and IL-6, TNF-alpha, VEGF, and HGF levels were quantified by ELISA. Activation of MAPK pathways was confirmed by Western blot analysis. DFO stimulation resulted in a significant increase in epithelial cell IL-6 and VEGF production while yielding a decrease in HGF production (P<0.05). Unexpectedly, TNF-alpha was not detectable. p38 MAPK, ERK, and JNK inhibition significantly decreased IL-6, VEGF, and HGF production (P<0.05). In conclusion, DFO acutely increases fetal human intestinal epithelial cell IL-6 and VEGF expression while causing an unexpected decrease in HGF expression and no detectable TNF-alpha production. Furthermore, chelator-induced intestinal epithelial cell cytokine expression depends on p38, ERK, and JNK MAPK pathways.
Collapse
Affiliation(s)
- Troy A Markel
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Martin CR, Walker WA. Intestinal immune defences and the inflammatory response in necrotising enterocolitis. Semin Fetal Neonatal Med 2006; 11:369-77. [PMID: 16690363 DOI: 10.1016/j.siny.2006.03.002] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Necrotising enterocolitis is a devastating neonatal gastrointestinal emergency predominantly affecting low birth weight, premature infants and is accompanied by significant mortality and morbid sequelae. The pathophysiology remains obscure and the management of infants with necrotising enterocolitis has not changed since the recognition of this disease. Necrotising enterocolitis is most likely the clinical culmination of multiple different risk factors interacting with each other to produce bowel injury through a final, common inflammatory pathway. Here, we review intestinal immunity and the specific inflammatory mediators involved in this disease process.
Collapse
MESH Headings
- Enterocolitis, Necrotizing/immunology
- Enterocolitis, Necrotizing/mortality
- Enterocolitis, Necrotizing/pathology
- Humans
- Infant, Newborn
- Infant, Premature
- Infant, Premature, Diseases/immunology
- Infant, Premature, Diseases/mortality
- Infant, Premature, Diseases/pathology
- Intestines/immunology
- Prevalence
- Risk Factors
Collapse
Affiliation(s)
- Camilia R Martin
- Harvard Medical School, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Rose-318, Boston, MA 02215, USA.
| | | |
Collapse
|
35
|
Medina C, Radomski MW. Role of matrix metalloproteinases in intestinal inflammation. J Pharmacol Exp Ther 2006; 318:933-8. [PMID: 16644899 DOI: 10.1124/jpet.106.103465] [Citation(s) in RCA: 155] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Matrix metalloproteinases (MMPs) and their endogenous inhibitors, tissue inhibitors of MMPs (TIMPs), are produced in the gastrointestinal tract by several structural cells. The balance between MMPs and TIMPs is essential for many physiological processes in the gut. However, imbalance between MMPs and TIMPs plays an important role in the pathophysiology of diverse intestinal inflammatory conditions. We reviewed the role of the MMP/TIMP system in the pathogenesis of intestinal inflammatory diseases and pharmacologic perspectives for the use of compounds that restore the MMP/TIMP balance.
Collapse
Affiliation(s)
- Carlos Medina
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland
| | | |
Collapse
|
36
|
Seitz G, Warmann SW, Guglielmetti A, Heitmann H, Ruck P, Kreis ME, Fuchs J. Protective effect of tumor necrosis factor alpha antibody on experimental necrotizing enterocolitis in the rat. J Pediatr Surg 2005; 40:1440-5. [PMID: 16150346 DOI: 10.1016/j.jpedsurg.2005.05.043] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is a common and devastating disorder of premature infants. Elevated proinflammatory cytokines, especially tumor necrosis factor alpha (TNF-alpha), have been implicated in the pathogenesis of NEC. The aim of this study was to evaluate the effects of TNF-alpha on the inflammatory response in NEC by immunoneutralizing TNF-alpha with a selective antibody. METHODS Neonatal Sprague-Dawley rats were divided in 3 groups: group 1 (n = 20), a NEC-like enterocolitis was induced by formula feeding, asphyxia, and cold exposure; group 2 (n = 9), animals were treated like in group 1 and additionally received TNF-alpha antibody intraperitoneally; and group 3 (n = 17), animals were dam-fed (controls). Animals were killed in case of imminent death or after 96 hours. Specimens from small bowel were processed for blinded histologic (H&E) and immunhistologic (myeloperoxidase [MPO]) analysis. RESULTS In group 1, animals developed severe NEC (mean NEC score, 3.28 +/- 0.32; mean MPO, 65.85 +/- 9.46). In group 2, animals developed mild NEC (mean NEC score, 1.72 +/- 0.41; mean MPO, 34.33 +/- 9.69; P < .05). In group 3, no NEC was induced (mean NEC score, 0.0 +/- 0; mean MPO, 6 +/- 1.32; P < .05). CONCLUSION Tumor necrosis factor alpha antibody may have an attenuating effect on experimental NEC in rats.
Collapse
Affiliation(s)
- Guido Seitz
- Department of Pediatric Surgery, University Children's Hospital, 72076 Tübingen, Germany.
| | | | | | | | | | | | | |
Collapse
|