1
|
Zhang D, Xie D, Qu Y, Mu D, Wang S. Digging deeper into necrotizing enterocolitis: bridging clinical, microbial, and molecular perspectives. Gut Microbes 2025; 17:2451071. [PMID: 39826099 DOI: 10.1080/19490976.2025.2451071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/26/2024] [Accepted: 01/02/2025] [Indexed: 01/20/2025] Open
Abstract
Necrotizing Enterocolitis (NEC) is a severe, life-threatening inflammatory condition of the gastrointestinal tract, especially affecting preterm infants. This review consolidates evidence from various biomedical disciplines to elucidate the complex pathogenesis of NEC, integrating insights from clinical, microbial, and molecular perspectives. It emphasizes the modulation of NEC-associated inflammatory pathways by probiotics and novel biologics, highlighting their therapeutic potential. We further critically examine dysbiotic alterations within the gut microbiota, with a particular focus on imbalances in bacterial and viral communities, which may contribute to the onset of NEC. The intricate interactions among toll-like receptor 4 (TLR4), microvascular integrity, immune activation, and the inflammatory milieu are meticulously summarized, offering a sophisticated understanding of NEC pathophysiology. This academic review aims to enhance the etiological comprehension of NEC, promote the development of targeted therapeutic interventions, and impart the significant impact of perinatal factors on the formulation of preventive and curative strategies for the disease.
Collapse
Affiliation(s)
- Deshuang Zhang
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
- Division of Neonatology/Pediatric Surgery, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Dongke Xie
- Division of Neonatology/Pediatric Surgery, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yi Qu
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Dezhi Mu
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Shaopu Wang
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
2
|
Sokou R, Mantzios P, Palioura AE, Tsantes AG, Lianou A, Piovani D, Tsante KA, Lampropoulou K, Iacovidou N, Bonovas S. Diagnostic and Prognostic Value of Hematological Parameters in Necrotizing Enterocolitis: A Systematic Review. J Clin Med 2025; 14:2530. [PMID: 40217979 PMCID: PMC11989880 DOI: 10.3390/jcm14072530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 03/24/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025] Open
Abstract
Background/Objectives: Necrotizing enterocolitis (NEC) is a severe, potentially fatal gastrointestinal disease that primarily affects preterm neonates, especially those with very low birth weight (<1500 g). Despite extensive research, its pathophysiology remains unclear, with NEC considered a spectrum of disorders driven by systemic inflammation, microbiota dysregulation, and intestinal hypoxic injury. Diagnosis is challenging due to its subtle presentation and reliance on clinical and radiographic findings, underscoring the urgent need for reliable early biomarkers. Complete blood count (CBC) is one of the most frequently performed laboratory tests in neonatal care, providing valuable insights associated with hematologic alterations associated with NEC. Given its cost-effectiveness, accessibility, and rapid turnaround time, CBC parameters have been increasingly investigated for their diagnostic and prognostic potential in NEC. This systematic review consolidates existing evidence on the diagnostic and prognostic utility of CBC parameters in NEC, examining their association with disease onset, progression, and outcomes. Methods: A systematic review of the literature in PubMed and Scopus databases was conducted, between February 25 and December 2024. Results: Following a PRISMA-compliant search strategy, 77 eligible studies were included, analyzing data from 295,195 neonates, of whom 14,570 had NEC. Among the 77 studies, 17 examined NEC-associated mortality as a primary outcome, while 13 studies focused on the development of predictive models incorporating CBC parameters alongside other clinical and laboratory data to assess NEC severity and prognosis in neonates. The findings highlight the potential of CBC-derived markers to facilitate early NEC detection and risk stratification. However, variations in study design and diagnostic criteria highlight the need for prospective studies to validate their clinical use. Conclusions: Despite advancements in understanding NEC, its diagnosis remains challenging due to the absence of fully reliable biomarkers. CBC parameters show promise in offering early diagnostic and prognostic insights. However, further validation is needed for their routine integration into NICU practice. Given the persistent challenges in NEC diagnosis and management, our findings highlight the necessity for integrated scoring systems that combine hematologic, clinical, and radiologic data to enhance early detection and optimize neonatal care. Further research is essential to refine these predictive models, enabling timely interventions and improving survival rates in NEC-affected neonates.
Collapse
Affiliation(s)
- Rozeta Sokou
- Neonatal Department, National and Kapodistrian University of Athens, Aretaieio Hospital, 11528 Athens, Greece;
| | - Petros Mantzios
- Immunology and Histocompatibility Department, Evangelismos General Hospital, 10676 Athens, Greece;
| | - Alexia Eleftheria Palioura
- Neonatal Intensive Care Unit, “Agios Panteleimon” General Hospital of Nikea, 18454 Piraeus, Greece; (A.E.P.); (A.L.)
| | - Andreas G. Tsantes
- Microbiology Department, “Saint Savvas” Oncology Hospital, 11522 Athens, Greece;
| | - Alexandra Lianou
- Neonatal Intensive Care Unit, “Agios Panteleimon” General Hospital of Nikea, 18454 Piraeus, Greece; (A.E.P.); (A.L.)
| | - Daniele Piovani
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy;
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | | | - Katerina Lampropoulou
- Neonatal Intensive Care Unit, School of Medicine, University of Ioannina, 45110 Ioannina, Greece;
| | - Nicoletta Iacovidou
- Neonatal Department, National and Kapodistrian University of Athens, Aretaieio Hospital, 11528 Athens, Greece;
| | - Stefanos Bonovas
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy;
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| |
Collapse
|
3
|
Li Z, Jiang Q, Wei J, Dang D, Meng Z, Wu H. Piezo1 promotes the progression of necrotizing enterocolitis by activating the Ca2(+)/CaMKII-dependent pathway. Commun Biol 2025; 8:417. [PMID: 40074811 PMCID: PMC11904196 DOI: 10.1038/s42003-025-07821-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Necrotizing enterocolitis (NEC) is a devastating inflammatory bowel necrosis of preterm infants with limited therapeutic approaches. Mounting evidence supports the role of Piezo1, namely, a widely distributed mechanosensor in intestinal epithelial cells (IECs), in intestinal inflammation but its underlying mechanism in the development of NEC remains unexplored. In this study, we demonstrated that Piezo1 expression was higher in preterm infants with lower gestational age. C57BL/6J mice wherein Piezo1 was deleted in IECs (villin-specific Piezo1 knockout mice; Piezo1flox/floxVillinCre+) and Piezo1flox/flox littermates were subjected to induce NEC, and Piezo1 knockout regulated the intestinal barrier function, restricted cytokines secretion, and diminished the inflammatory response in NEC mouse models. Piezo1 elevated cytosolic Ca2+ levels and activated Ca2+/calmodulin-dependent protein kinase II (CaMKII) to promote the CaMKII/NF-κB interaction and NF-κB activation in vitro. Finally, the effects of a CaMKII inhibitor, KN93, were evaluated both in vitro and in vivo in NEC models, and the functions of Piezo1 in IECs were suppressed partially by KN93. In this study, we characterise the undefined role of Piezo1 in the development of NEC, which may partially be attributed to the differential role of calcium under pathophysiological conditions.
Collapse
Affiliation(s)
- Zhenyu Li
- Department of Neonatology, Children's Medical Center, The First Hospital of Jilin University, Changchun, Jilin, China
- The Child Health Clinical Research Center of Jilin Province, Changchun, Jilin, China
| | - Qinlei Jiang
- Department of Neonatology, Children's Medical Center, The First Hospital of Jilin University, Changchun, Jilin, China
- The Child Health Clinical Research Center of Jilin Province, Changchun, Jilin, China
| | - Jiaqi Wei
- Department of Neonatology, Children's Medical Center, The First Hospital of Jilin University, Changchun, Jilin, China
- The Child Health Clinical Research Center of Jilin Province, Changchun, Jilin, China
| | - Dan Dang
- Department of Neonatology, Children's Medical Center, The First Hospital of Jilin University, Changchun, Jilin, China
- The Child Health Clinical Research Center of Jilin Province, Changchun, Jilin, China
| | - Zhaoli Meng
- Laboratory of Tumor Immunology, The First Hospital of Jilin University, Changchun, China
| | - Hui Wu
- Department of Neonatology, Children's Medical Center, The First Hospital of Jilin University, Changchun, Jilin, China.
- The Child Health Clinical Research Center of Jilin Province, Changchun, Jilin, China.
| |
Collapse
|
4
|
Younge N, Patel RM. Probiotics and the Risk of Infection. Clin Perinatol 2025; 52:87-100. [PMID: 39892956 PMCID: PMC11789005 DOI: 10.1016/j.clp.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Probiotic use has increased in preterm infants and may reduce the risk of necrotizing enterocolitis. Probiotic-associated infection is a concern for infants receiving probiotic supplementation in the neonatal intensive care unit, as highlighted by a recent case and subsequent action by the United States Food and Drug Administration. Based on reports to date, invasive infection is an infrequent but known risk of probiotic supplementation. In this article, we discuss the epidemiology and pathophysiology of invasive infection in preterm infants, review the benefits and risks of probiotic as regulations and available products continue to evolve.
Collapse
Affiliation(s)
- Noelle Younge
- Department of Pediatrics, Duke University, 2400 Pratt Street, DUMC Box 2739, Durham, NC 27705, USA
| | - Ravi M Patel
- Department of Pediatrics, Emory University and Children's Healthcare of Atlanta, Arthur M. Blank Hospital, 2220 North Druid Hills Road NE, CL.06323, Atlanta, GA 30329, USA.
| |
Collapse
|
5
|
Brown JA, Bashir H, Zeng MY. Lifelong partners: Gut microbiota-immune cell interactions from infancy to old age. Mucosal Immunol 2025:S1933-0219(25)00006-6. [PMID: 39862964 DOI: 10.1016/j.mucimm.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025]
Abstract
Our immune system and gut microbiota are intricately coupled from birth, both going through maturation during early life and senescence during aging almost in a synchronized fashion. The symbiotic relationship between the human host and microbiota is critically dependent on a healthy immune system to keep our microbiota in check, while the microbiota provides essential functions to promote the development and fitness of our immune system. The partnership between our immune system and microbiota is particularly important during early life, when microbial ligands and metabolites shape the development of the immune cells and immune tolerance; during aging, having sufficient beneficial gut bacteria is critical for the maintenance of intact mucosal barriers, immune metabolic fitness, and strong immunity against pathogens. The immune system during childhood is programmed, with the support of the microbiota, to develop robust immune tolerance, and limit autoimmunity and metabolic dysregulation, which are prevalent during aging. This review comprehensively explores the mechanistic underpinnings of gut microbiota-immune cell interactions during infancy and old age, with the goal to gain a better understanding of potential strategies to leverage the gut microbiota to combat age-related immune decline.
Collapse
Affiliation(s)
- Julia A Brown
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, United States; Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, United States
| | - Hilal Bashir
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, United States; Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, United States
| | - Melody Y Zeng
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, United States; Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, United States; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School, New York, NY 10065, United States.
| |
Collapse
|
6
|
Karadeniz Cerit K, Koyuncuoğlu T, Akcan B, Çağatay NS, Üçem S, Erdoğan Ö, Çevik Ö, Gökçeoğlu Kayalı D, Akakın D, Yeğen BÇ. Estrogen Alleviates Oxidative Bowel Injury and Neuroinflammation in Necrotizing Enterocolitis. J Surg Res 2025; 305:367-384. [PMID: 39733474 DOI: 10.1016/j.jss.2024.12.003] [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: 03/12/2024] [Revised: 11/28/2024] [Accepted: 12/05/2024] [Indexed: 12/31/2024]
Abstract
INTRODUCTION High mortality and morbidity of neonates with necrotizing enterocolitis (NEC) necessitates the investigation of novel therapies to improve outcomes. It was aimed to elucidate the potential therapeutic effect of estrogen receptor agonists on NEC-induced intestinal and brain injury in rats. METHODS Sprague-Dawley pups of both sexes were separated from their mothers at postnatal 5th d. Feeding with formula along with a single session of hypoxia was applied to induce NEC, while control pups were kept with their mothers. The NEC rats received either vehicle, estrogen receptor α (ERα) agonist propyl pyrazole triol (1 mg/kg/day), ERβ agonist diarylpropionitrile (1 mg/kg/day), or 17β-estradiol (1 mg/kg/day) during maternal separation. All pups were decapitated on postnatal 9th d to collect intestinal and brain tissue samples. RESULTS Elevation in proinflammatory cytokines, apoptosis, and microscopically and biochemically evident oxidative injury in both the intestinal and brain tissues were observed in NEC-induced pups. In both the intestinal and brain tissues, nerve growth factor and brain-derived neurotrophic factor protein levels were depleted, expressions of both the ESR1 and ESR2 genes were downregulated, while treatment with 17β-estradiol or ER agonists alleviated extent of oxidative injury of the intestines and brain tissue, upregulated nerve growth factor, brain-derived neurotrophic factor, and ER gene expressions, abolished NEC-induced decrease in claudin-3 expression, increased the survival rates, improved the clinical states of the survived pups at varying degrees. CONCLUSIONS Activation of estrogen signaling by receptor agonists alleviated NEC-induced intestinal and cerebral injury, implicating that estrogen agonists could be regarded as promising preventive/therapeutic agents for NEC.
Collapse
Affiliation(s)
| | - Türkan Koyuncuoğlu
- Department of Physiology, Marmara University, School of Medicine, İstanbul, Türkiye
| | - Beyza Akcan
- Marmara University, School of Medicine, İstanbul, Türkiye
| | | | - Selen Üçem
- Marmara University, School of Medicine, İstanbul, Türkiye
| | - Ömer Erdoğan
- Department of Biochemistry, Adnan Menderes University, School of Medicine, Aydın, Türkiye
| | - Özge Çevik
- Department of Biochemistry, Adnan Menderes University, School of Medicine, Aydın, Türkiye
| | - Damla Gökçeoğlu Kayalı
- Department of Histology and Embryology, Marmara University, School of Medicine, İstanbul, Türkiye
| | - Dilek Akakın
- Department of Histology and Embryology, Marmara University, School of Medicine, İstanbul, Türkiye
| | - Berrak Ç Yeğen
- Department of Physiology, Marmara University, School of Medicine, İstanbul, Türkiye
| |
Collapse
|
7
|
Li H, Lai J, Xiao D, Huang D, Zhang Y, Gu X, Li F, Hao H. Deoxycholic acid aggravates necrotizing enterocolitis through downregulation of mesenchymal-epithelial transition factor expression. Braz J Med Biol Res 2024; 57:e14046. [PMID: 39630809 DOI: 10.1590/1414-431x2024e14046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 09/17/2024] [Indexed: 12/07/2024] Open
Abstract
Bile acids are closely associated with necrotizing enterocolitis (NEC), and their accumulation has cytotoxic effects on cells. However, the specific bile acid subtype involved in NEC and its underlying mechanisms remains poorly understood, limiting the therapeutic potential of bile acids as treatment targets. In the present study, deoxycholic acid (DCA) accumulation in the intestinal lumen exacerbated NEC-induced intestinal damage. DCA suppressed the expression of mesenchymal-epithelial transition factor (MET), a proto-oncogene located on chromosome 7q31.2 that encodes c-Met, in the mouse intestine through transcription factors and increased nuclear translocation of p-STAT3. MET is a receptor tyrosine kinase that participates in cell proliferation and migration processes. Increasing concentrations of DCA downregulated MET expression and reduced the proliferation and migration of intestinal epithelial cells in vitro. MET knockdown reduced the proliferation and migration of intestinal epithelial cells but increased STAT3 phosphorylation. These findings indicated that MET mediated STAT3 involvement in intestinal epithelial cell proliferation and migration, demonstrating that the inhibitory effect of DCA on MET disrupted this process. These results elucidated the damaging effects and mechanisms of DCA accumulation in NEC, providing new insights into the use of DCA as a therapeutic target for NEC.
Collapse
Affiliation(s)
- Hongfu Li
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiahao Lai
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Dongfan Xiao
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dabin Huang
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yinchun Zhang
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xia Gu
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fei Li
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Inborn Errors of Metabolism Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hu Hao
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Inborn Errors of Metabolism Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
8
|
Klerk DH, Moore H, Scheese DJ, Tragesser C, Raouf Z, Duess JW, Tsuboi K, Sampah ME, Lopez CM, Williams-McLeod S, El Baassiri MG, Jang HS, Prindle T, Wang S, Wang M, Fulton WB, Sodhi CP, Hackam DJ. Multi-strain probiotic administration decreases necrotizing enterocolitis severity and alters the epigenetic profile in mice. Pediatr Res 2024:10.1038/s41390-024-03716-0. [PMID: 39562735 DOI: 10.1038/s41390-024-03716-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 09/29/2024] [Accepted: 10/14/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND Probiotic administration may decrease the incidence of necrotizing enterocolitis (NEC) through mechanisms that are largely unknown. We investigated the effects of probiotics on intestinal epigenetics and assessed their effects on intestinal inflammation and motility using both ileum-predominant and combined ileo-colitis mouse NEC models. METHODS C57BL/6 J mice were gavage-fed a multi-strain probiotic from postnatal days 3-11, consisting of B. infantis, B. lactis, and S. thermophilus. From p8, mice were exposed to ileo-colitis NEC involving formula containing NEC bacteria and 0.5% DSS. DNA methylation was measured using the Infinium Methylation Assay. Gastrointestinal motility was assessed by 70 Kd FITC-dextran transit time. Probiotic colonization was measured in probiotic-fed mice by qPCR. RESULTS Probiotic administration caused significant changes in the small intestine's epigenetic signature, a reduction in NEC severity, and improved intestinal motility. The effects of probiotics were more pronounced in the ileo-colitis NEC model. CONCLUSIONS These findings shed light on the role of probiotics in two clinically relevant models of NEC, add additional insights into their underlying mechanism of action, and reveal unanticipated epigenetic modifications to the intestinal mucosa after their use. IMPACT These findings shed light on the role of multi-strain probiotics in two clinically relevant animal models of NEC, and add additional insights into their underlying mechanism of action This study provides a new, clinically relevant model for the study of NEC including administration of 0.5% DSS, to include ileal dominant and ileo-colonic dominant phenotypes of the disease. These results reveal that clinically relevant strains of probiotic bacteria can exert epigenetic effects on the small intestine in mice, and can attenuate the epigenetic changes induced by NEC.
Collapse
Affiliation(s)
- Daphne H Klerk
- Division of Pediatric Surgery, Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Division of Neonatology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Hannah Moore
- Division of Pediatric Surgery, Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniel J Scheese
- Division of Pediatric Surgery, Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cody Tragesser
- Division of Pediatric Surgery, Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zachariah Raouf
- Division of Pediatric Surgery, Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Johannes W Duess
- Division of Pediatric Surgery, Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Koichi Tsuboi
- Division of Pediatric Surgery, Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Maame E Sampah
- Division of Pediatric Surgery, Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Carla M Lopez
- Division of Pediatric Surgery, Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sierra Williams-McLeod
- Division of Pediatric Surgery, Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mahmoud G El Baassiri
- Division of Pediatric Surgery, Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hee-Seong Jang
- Division of Pediatric Surgery, Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomas Prindle
- Division of Pediatric Surgery, Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sanxia Wang
- Division of Pediatric Surgery, Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Menghan Wang
- Division of Pediatric Surgery, Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - William B Fulton
- Division of Pediatric Surgery, Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chhinder P Sodhi
- Division of Pediatric Surgery, Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - David J Hackam
- Division of Pediatric Surgery, Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
9
|
Le H, Wang Y, Zhou J, Li D, Gong Z, Zhu F, Wang J, Tian C, Cai W, Wu J. Git2 deficiency promotes MDSCs recruitment in intestine via NF-κB-CXCL1/CXCL12 pathway and ameliorates necrotizing enterocolitis. Mucosal Immunol 2024; 17:1060-1071. [PMID: 39074614 DOI: 10.1016/j.mucimm.2024.07.006] [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: 11/10/2023] [Revised: 07/22/2024] [Accepted: 07/22/2024] [Indexed: 07/31/2024]
Abstract
Necrotizing enterocolitis (NEC) is a severe gastrointestinal disease in preterm infants and the most common cause of neonatal death, whereas the molecular mechanism of intestinal injury remains unclear accompanied by deficiency of effective therapeutic approaches. GIT2 (G-protein-coupled receptor kinase interacting proteins 2) can affect innate and adaptive immunity and has been involved in multiple inflammatory disorders. In this study, we investigated whether GIT2 participates in the pathogenesis of NEC. Here we found that intestinal Git2 gene expression was significantly increased in NEC patients and NEC mice, which positively correlated with the tissue damage severity, and Git2 deficiency could potently protect against NEC development in mice. Mechanistically, Git2 gene knockout dramatically increased the recruitment of MDSCs in the intestine, and in vivo depletion of MDSCs almost completely abrogated the protective effect of Git2 deficiency on NEC. Moreover, Git2 deficiency induced MDSCs intestinal accumulation mainly relied on CXCL1/CXCL12 signaling, as evidenced by the significant increment of CXCL1 and CXCL12 levels in intestinal epithelium of Git2-/- mice and dramatically decrease of MDSCs accumulation in intestine as well as increase of NEC severity upon treatment of CXCL1/CXCL12 pathway inhibitors. In addition, Git2 deficiency induced up-regulation of CXCL1 and CXCL12 is at least partially mediated through activating NF-κB signaling. Thus, our findings suggest that GIT2 is involved in the pathogenesis of NEC, and targeting GIT2 may be a potential preventive and therapeutic approach for NEC.
Collapse
Affiliation(s)
- Huijuan Le
- Department of pediatric Surgery, Xinhua hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China; Shanghai Institute for Pediatric Research, School of Medicine, Shanghai Jiaotong University, Shanghai, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Yanyan Wang
- Department of pediatric Surgery, Xinhua hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China; Shanghai Institute for Pediatric Research, School of Medicine, Shanghai Jiaotong University, Shanghai, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Jiefei Zhou
- Department of pediatric Surgery, Xinhua hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China; Shanghai Institute for Pediatric Research, School of Medicine, Shanghai Jiaotong University, Shanghai, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Dan Li
- Department of pediatric Surgery, Xinhua hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China; Shanghai Institute for Pediatric Research, School of Medicine, Shanghai Jiaotong University, Shanghai, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Zizhen Gong
- Department of pediatric Surgery, Xinhua hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China; Shanghai Institute for Pediatric Research, School of Medicine, Shanghai Jiaotong University, Shanghai, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Fangxinxing Zhu
- Department of pediatric Surgery, Xinhua hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China; Shanghai Institute for Pediatric Research, School of Medicine, Shanghai Jiaotong University, Shanghai, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Jian Wang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Chunyan Tian
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China.
| | - Wei Cai
- Department of pediatric Surgery, Xinhua hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China; Shanghai Institute for Pediatric Research, School of Medicine, Shanghai Jiaotong University, Shanghai, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China.
| | - Jin Wu
- Department of pediatric Surgery, Xinhua hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China; Shanghai Institute for Pediatric Research, School of Medicine, Shanghai Jiaotong University, Shanghai, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China.
| |
Collapse
|
10
|
Sarafidis K, Agakidou E, Kontou A, Agakidis C, Neu J. Struggling to Understand the NEC Spectrum-Could the Integration of Metabolomics, Clinical-Laboratory Data, and Other Emerging Technologies Help Diagnosis? Metabolites 2024; 14:521. [PMID: 39452903 PMCID: PMC11509608 DOI: 10.3390/metabo14100521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/14/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
Necrotizing enterocolitis (NEC) is the most prevalent and potentially fatal intestinal injury mainly affecting premature infants, with significant long-term consequences for those who survive. This review explores the scale of the problem, highlighting advancements in epidemiology, the understanding of pathophysiology, and improvements in the prediction and diagnosis of this complex, multifactorial, and multifaced disease. Additionally, we focus on the potential role of metabolomics in distinguishing NEC from other conditions, which could allow for an earlier and more accurate classification of intestinal injuries in infants. By integrating metabolomic data with other diagnostic approaches, it is hoped to enhance our ability to predict outcomes and tailor treatments, ultimately improving care for affected infants.
Collapse
Affiliation(s)
- Kosmas Sarafidis
- 1st Department of Neonatology, School of Medicine, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (E.A.); (A.K.)
| | - Eleni Agakidou
- 1st Department of Neonatology, School of Medicine, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (E.A.); (A.K.)
| | - Angeliki Kontou
- 1st Department of Neonatology, School of Medicine, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (E.A.); (A.K.)
| | - Charalampos Agakidis
- 1st Department of Pediatrics, School of Medicine, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece;
| | - Josef Neu
- Department of Pediatrics, Division of Neonatology, University of Florida, Gainesville, FL 32611, USA;
| |
Collapse
|
11
|
Wu S, Ren X, Zhuang Y, Shen C, Zhu H, Cao Y, Zhang X, Chen G. Peripheral Lymphocyte Changes Associate With the Progression of Necrotizing Enterocolitis in Infants. J Surg Res 2024; 301:215-223. [PMID: 38959630 DOI: 10.1016/j.jss.2024.04.046] [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: 10/18/2023] [Revised: 04/17/2024] [Accepted: 04/24/2024] [Indexed: 07/05/2024]
Abstract
INTRODUCTION Immune factors are important antecedents in the pathophysiology of necrotizing enterocolitis (NEC). However, studies on the peripheral blood lymphocyte subsets changes in NEC patients among different Bell stages and in patients requiring surgery are scarce. METHODS 34 infants with NEC and 33 age-matched controls were included. Peripheral blood was collected within 48 h after NEC diagnosis. Peripheral blood B and T lymphocytes subsets were detected by 12-color flow cytometry. Cell ratios were calculated, and their relationship to disease severity and their roles as indicators for surgery were assessed. RESULTS NEC patients showed elevated percentages of unSwB cells (CD27+IgD+ unswitched memory/activated B cells)/B cells, SwB cells (CD27+IgD-switched memory B cells)/B cells, CD8+ T (CD3+CD8+ T cells)/T cells, Tem (CD45RA-CCR7-effector memory T cells)/CD4+ T cells, Tem/CD8+ T cells and decreased Bn (CD27-IgD+ naïve B cells)/B cells, CD4+T (CD3+CD4+ T cells)/T cells, CD45RA+ CCR7+ naïve T cells (CD45RA+CCR7+ naïve T cells)/CD8+T cells. Compared to NEC patients at BELL stage I + II, patients at BELL stage III showed increased percentages of SwB cells/B cells, antibody secreting cell (ASC, CD3-CD20-CD27high CD38high ASCs)/B cells and Tem/CD4+ T cells, and decreased percentages of CD45RA+CCR7+ naïve T cells/CD4+ T cells. The Receiver Operating Characteristic Curve analysis showed that the sensitivity of ASC/B cells ratio (0.52%) is 86.67% and the specificity of Tem/CD4+T ratio (5.22%) is 100%, indicating that NEC patients required surgery. CONCLUSIONS The severity of NEC exhibits codirectional changes with the maturation of B and T lymphocytes, especially CD4+ T cells. The increased ASC/B and Tem/CD4+ T cells could serve as potential indicators for NEC patients requiring surgery.
Collapse
Affiliation(s)
- Shaojing Wu
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Xue Ren
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Yuxiu Zhuang
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Chun Shen
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Haitao Zhu
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Yun Cao
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Xiaoming Zhang
- Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.
| | - Gong Chen
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China.
| |
Collapse
|
12
|
Blum L, Vincent D, Boettcher M, Knopf J. Immunological aspects of necrotizing enterocolitis models: a review. Front Immunol 2024; 15:1434281. [PMID: 39104529 PMCID: PMC11298363 DOI: 10.3389/fimmu.2024.1434281] [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] [Received: 05/17/2024] [Accepted: 07/08/2024] [Indexed: 08/07/2024] Open
Abstract
Necrotizing enterocolitis (NEC) is one of the most devasting diseases affecting preterm neonates. However, despite a lot of research, NEC's pathogenesis remains unclear. It is known that the pathogenesis is a multifactorial process, including (1) a pathological microbiome with abnormal bacterial colonization, (2) an immature immune system, (3) enteral feeding, (3) an impairment of microcirculation, and (4) possibly ischemia-reperfusion damage to the intestine. Overall, the immaturity of the mucosal barrier and the increased expression of Toll-like receptor 4 (TLR4) within the intestinal epithelium result in an intestinal hyperinflammation reaction. Concurrently, a deficiency in counter-regulatory mediators can be seen. The sum of these processes can ultimately result in intestinal necrosis leading to very high mortality rates of the affected neonates. In the last decade no substantial advances in the treatment of NEC have been made. Thus, NEC animal models as well as in vitro models have been employed to better understand NEC's pathogenesis on a cellular and molecular level. This review will highlight the different models currently in use to study immunological aspects of NEC.
Collapse
Affiliation(s)
| | | | | | - Jasmin Knopf
- Department of Pediatric Surgery, University Medical Center Mannheim, University Heidelberg, Mannheim, Germany
| |
Collapse
|
13
|
Alwetaid MY, Almanaa TN, Bakheet SA, Ansari MA, Nadeem A, Attia SM, Hussein MH, Attia MSM, Ahmad SF. Aflatoxin B 1 exposure exacerbates chemokine receptor expression in the BTBR T + Itpr3 tf/J Mouse Model, unveiling insights into autism spectrum disorder: A focus on brain and spleen. Reprod Toxicol 2024; 126:108599. [PMID: 38679149 DOI: 10.1016/j.reprotox.2024.108599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 04/16/2024] [Accepted: 04/19/2024] [Indexed: 05/01/2024]
Abstract
OBJECTIVE Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by significant difficulties in social interaction, communication, and repeated stereotypic behaviour. Aflatoxin B1 (AFB1) is the most potent and well-known mycotoxin in various food sources. Despite its propensity to generate significant biochemical and structural changes in human and animal tissues, the influence of AFB1 on ASD has yet to be thoroughly studied. Mounting evidence indicates that chemokine receptors play a crucial function in the central nervous system and are implicated in developing several neuroinflammatory disorders. Chemokine receptors in individuals with ASD were elevated in the anterior cingulate gyrus astrocytes, cerebellum, and brain. METHODS The BTBR T+Itpr3tf/J (BTBR) mice are inbred strains that exhibit strong and consistently observed deficits in social interactions, characterized by excessive self-grooming and limited vocalization in social contexts. We examined the impact of AFB1 on CCR3-, CCR7-, CCR9-, CXCR3-, CXCR4-, and CXCR6-expressing I-A/I-E+ cells in the spleen of the BTBR mouse model of autism. We evaluated the mRNA levels of CCR3, CCR7, CCR9, CXCR3, CXCR4, and CXCR6 chemokine receptors in the brain. RESULTS The exposure to AFB1 in BTBR mice resulted in a significant rise in the number of I-A/I-E+CCR3+, I-A/I-E+CCR7+, I-A/I-E+CCR9+, I-A/I-E+CXCR3+, I-A/I-E+CXCR4+, and I-A/I-E+CXCR6+ cells. Furthermore, exposure to AFB1 increased mRNA expression levels of CCR3, CCR7, CCR9, CXCR3, CXCR4, and CXCR6 in the brain. CONCLUSIONS These findings highlight that AFB1 exposure increases the expression of chemokine receptors in BTBR mice, indicating the necessity for further research into AFB1's role in the development of ASD.
Collapse
Affiliation(s)
- Mohammad Y Alwetaid
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Taghreed N Almanaa
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mushtaq A Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Marwa H Hussein
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohamed S M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
14
|
Zheng X, Lei W, Zhang Y, Jin H, Han C, Wu F, Jia C, Zeng R, Chen Z, Zhang Y, Wang H, Liu Q, Yao Z, Yu Y, Zhou J. Neuropilin-1 high monocytes protect against neonatal inflammation. Cell Mol Immunol 2024; 21:575-588. [PMID: 38632385 PMCID: PMC11143335 DOI: 10.1038/s41423-024-01157-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 03/19/2024] [Indexed: 04/19/2024] Open
Abstract
Neonates are susceptible to inflammatory disorders such as necrotizing enterocolitis (NEC) due to their immature immune system. The timely appearance of regulatory immune cells in early life contributes to the control of inflammation in neonates, yet the underlying mechanisms of which remain poorly understood. In this study, we identified a subset of neonatal monocytes characterized by high levels of neuropilin-1 (Nrp1), termed Nrp1high monocytes. Compared with their Nrp1low counterparts, Nrp1high monocytes displayed potent immunosuppressive activity. Nrp1 deficiency in myeloid cells aggravated the severity of NEC, whereas adoptive transfer of Nrp1high monocytes led to remission of NEC. Mechanistic studies showed that Nrp1, by binding to its ligand Sema4a, induced intracellular p38-MAPK/mTOR signaling and activated the transcription factor KLF4. KLF4 transactivated Nos2 and enhanced the production of nitric oxide (NO), a key mediator of immunosuppression in monocytes. These findings reveal an important immunosuppressive axis in neonatal monocytes and provide a potential therapeutic strategy for treating inflammatory disorders in neonates.
Collapse
Affiliation(s)
- Xiaoqing Zheng
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, International Joint Laboratory of Ocular Diseases (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Immunology, Tianjin Medical University, Tianjin, 300070, China
- Institute of Pediatric Health and Disease, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Department of Immunology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Wen Lei
- Pediatric Immunity and Healthcare Biomedical Co., Ltd, Guangzhou, 510320, China
| | - Yongmei Zhang
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, International Joint Laboratory of Ocular Diseases (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Immunology, Tianjin Medical University, Tianjin, 300070, China
| | - Han Jin
- Department of Neurology, Institute of Neuroimmunology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Cha Han
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Fan Wu
- Institute of Pediatric Health and Disease, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Department of Neonatology, Guangzhou Key Laboratory of Neonatal Intestinal Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Chonghong Jia
- Institute of Pediatric Health and Disease, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Department of Neonatology, Guangzhou Key Laboratory of Neonatal Intestinal Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Ruihong Zeng
- Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Department of Immunology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Zhanghua Chen
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yuxia Zhang
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Haitao Wang
- Department of oncology, The Second Hospital of Tianjin Medical University, Tianjin Key Laboratory of Precision Medicine for Sex Hormones and Diseases, Tianjin, 300211, China
| | - Qiang Liu
- Department of Neurology, Institute of Neuroimmunology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Zhi Yao
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, International Joint Laboratory of Ocular Diseases (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Immunology, Tianjin Medical University, Tianjin, 300070, China
| | - Ying Yu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Jie Zhou
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, International Joint Laboratory of Ocular Diseases (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Immunology, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
15
|
Chong Q, Wang Z, Guo T, Zhang L, Lu L, Cai C, Gong X, Lv Z, Sheng Q. Gestational age-specific hematological features in preterm infants with necrotizing enterocolitis. Pediatr Res 2024; 95:1826-1836. [PMID: 38177247 DOI: 10.1038/s41390-023-02999-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND This study aimed to investigate gestational age-specific hematological features in preterm infants with necrotizing enterocolitis (NEC) and identify predictive hematological biomarkers for surgical NEC. METHODS We conducted a retrospective study comparing gestational age (GA)-specific clinical data between medical NEC (m-NEC) and surgical NEC (s-NEC) subgroups, stratified by GA as <28 weeks, 28 ≤ GA < 32 weeks, and 32 ≤ GA < 37 weeks. Multivariate logistic analysis and receiver operating characteristic curve were used to identify the independent predictors of s-NEC. RESULTS In comparison to m-NEC at NEC onset, s-NEC infants exhibited the following findings: In GA < 28 weeks, s-NEC infants had lower platelet counts. In 28 ≤ GA < 32 weeks, lower absolute lymphocyte counts, and significant percent drop in platelets, lymphocytes, and monocytes were observed. In 32 ≤ GA < 37 weeks, lower absolute lymphocyte counts and significant percent drop in lymphocytes were found. Independent predictors were able to distinguish s-NEC from m-NEC. The area under the curve (AUC) for platelet counts in GA < 28 weeks was 0.880, while C-reactive protein in 28 ≤ GA < 32 weeks had an AUC of 0.889. The AUC for lymphocyte counts in 32 ≤ GA < 37 weeks was 0.892. CONCLUSION This study identified hematological abnormalities in the development of NEC based on gestational age. Independent predictors may help clinicians distinguish surgical NEC from medical NEC. IMPACT Necrotizing enterocolitis (NEC) patients with different gestational ages (GA) exhibit different hematological features and independent predictors of surgical NEC differ among different GAs. Our research made the current studies about peripheral hematological features with NEC more complete by analyzing peripheral data collected within 24 h of birth, at day 5-7, day 3-4, day 1-2 before NEC onset, at the time of NEC onset, day 1, day 2, day 3, day 4-5, day 6-7 after NEC onset. Our study is helpful to clinicians in developing a more detailed diagnostic strategy based on GA for the early identification of surgical NEC.
Collapse
Affiliation(s)
- Qingqi Chong
- Department of General Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 355, Luding Rd, 200062, Shanghai, PR China
| | - Zhiru Wang
- Department of General Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 355, Luding Rd, 200062, Shanghai, PR China
| | - Ting Guo
- Department of General Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 355, Luding Rd, 200062, Shanghai, PR China
| | - Liaoran Zhang
- Department of General Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 355, Luding Rd, 200062, Shanghai, PR China
| | - Li Lu
- Department of General Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 355, Luding Rd, 200062, Shanghai, PR China
| | - Cheng Cai
- Department of Neonatology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 355, Luding Rd, 200062, Shanghai, PR China
| | - Xiaohui Gong
- Department of Neonatology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 355, Luding Rd, 200062, Shanghai, PR China
| | - Zhibao Lv
- Department of General Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 355, Luding Rd, 200062, Shanghai, PR China.
| | - Qingfeng Sheng
- Department of General Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 355, Luding Rd, 200062, Shanghai, PR China.
| |
Collapse
|
16
|
Nofi CP, Prince JM, Wang P, Aziz M. Chromatin as alarmins in necrotizing enterocolitis. Front Immunol 2024; 15:1403018. [PMID: 38881893 PMCID: PMC11176418 DOI: 10.3389/fimmu.2024.1403018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/20/2024] [Indexed: 06/18/2024] Open
Abstract
Necrotizing enterocolitis (NEC) is a severe gastrointestinal disease primarily affecting premature neonates, marked by poorly understood pro-inflammatory signaling cascades. Recent advancements have shed light on a subset of endogenous molecular patterns, termed chromatin-associated molecular patterns (CAMPs), which belong to the broader category of damage-associated molecular patterns (DAMPs). CAMPs play a crucial role in recognizing pattern recognition receptors and orchestrating inflammatory responses. This review focuses into the realm of CAMPs, highlighting key players such as extracellular cold-inducible RNA-binding protein (eCIRP), high mobility group box 1 (HMGB1), cell-free DNA, neutrophil extracellular traps (NETs), histones, and extracellular RNA. These intrinsic molecules, often perceived as foreign, have the potential to trigger immune signaling pathways, thus contributing to NEC pathogenesis. In this review, we unravel the current understanding of the involvement of CAMPs in both preclinical and clinical NEC scenarios. We also focus on elucidating the downstream signaling pathways activated by these molecular patterns, providing insights into the mechanisms that drive inflammation in NEC. Moreover, we scrutinize the landscape of targeted therapeutic approaches, aiming to mitigate the impact of tissue damage in NEC. This in-depth exploration offers a comprehensive overview of the role of CAMPs in NEC, bridging the gap between preclinical and clinical insights.
Collapse
Affiliation(s)
- Colleen P. Nofi
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Jose M. Prince
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| |
Collapse
|
17
|
Subramanian S, Bu HF, Chou PM, Wang X, Geng H, Akhtar S, Du C, Tan SC, Ideozu JE, Tulluri A, Sun Y, Ding WX, De Plaen IG, Tan XD. Scattered Crypt Intestinal Epithelial Cell Apoptosis Induces Necrotizing Enterocolitis Via Intricate Mechanisms. Cell Mol Gastroenterol Hepatol 2024; 18:101364. [PMID: 38788898 PMCID: PMC11278878 DOI: 10.1016/j.jcmgh.2024.05.012] [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: 12/07/2023] [Revised: 05/16/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND & AIMS Necrotizing enterocolitis (NEC) is a life-threatening disease affecting mostly the ileum of preemies. Intestinal epithelial cell (IEC) apoptosis contributes to NEC pathogenesis. However, how scattered crypt IEC apoptosis leads to NEC with excessive villus epithelial necrosis remains unclear. METHODS A novel triple-transgenic mouse model, namely, 3xTg-iAPcIEC (inducible apoptosis phenotype in crypt-IEC), was developed to induce IEC-specific overexpression of Fasl transgene using doxycycline (Dox)-inducible tetO-rtTA system and villin-cre technology. The 3-days-old neonatal 3xTg-iAPcIEC mice and their littermate controls were subcutaneously (s.c.) challenged with a single dose of Dox. Intestinal tissues were processed at different time points to examine scattered crypt IEC apoptosis-mediated NEC development. Gene knockout technology, antibody-mediated cell depletion, and antibiotic-facilitated Gram-positive bacteria depletion were used to study mechanisms. RESULTS Treatment of 3xTg-iAPcIEC mouse pups with Dox induces scattered crypt IEC apoptosis followed by crypt inflammation and excessive villous necrosis resembling NEC. This progression correlated with elevated Ifng, Rip3, CD8+ T cells, and Gram-positive bacteria in the ileum. Mechanistically, IFN-γ and RIP3-activated signals mediate the effect of scattered crypt IEC apoptosis on the induction of intestinal crypt inflammation and villous necrosis. Meanwhile, pathophysiological events of CD8+ T cell infiltration and dysbiosis with Gram-positive bacteria primarily contribute to excessive villous inflammation and necrosis. Notably, blocking any of these events protects against NEC development in 3xTg-iAPcIEC mouse pups, underlining their central roles in NEC pathogenesis. CONCLUSIONS Scattered crypt IEC apoptosis induces NEC in mouse pups via IFN-γ, RIP3, CD8+ T cells, and Gram-positive bacteria-mediated comprehensive pathophysiological events. Our findings may advance knowledge in the prevention and treatment of NEC.
Collapse
Affiliation(s)
- Saravanan Subramanian
- Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, Department of Pediatrics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Heng-Fu Bu
- Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, Department of Pediatrics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Pauline M Chou
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Xiao Wang
- Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, Department of Pediatrics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Hua Geng
- Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, Department of Pediatrics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Suhail Akhtar
- Department of Biochemistry, A.T. Still University of Health Sciences, Kirksville, Missouri
| | - Chao Du
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Stephanie C Tan
- Department of Medical Education, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
| | - Justin Eze Ideozu
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Aasrita Tulluri
- Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, Department of Pediatrics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, College Station, Texas
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Isabelle G De Plaen
- Division of Neonatology, Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Xiao-Di Tan
- Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, Department of Pediatrics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois; Department of Research & Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois.
| |
Collapse
|
18
|
Jones IH, Collins JE, Hall NJ, Heinson AI. Transcriptomic analysis of the effect of remote ischaemic conditioning in an animal model of necrotising enterocolitis. Sci Rep 2024; 14:10783. [PMID: 38734725 PMCID: PMC11088709 DOI: 10.1038/s41598-024-61482-9] [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: 01/11/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024] Open
Abstract
Necrotising enterocolitis (NEC) has a complex pathophysiology but the common end-point is ischaemia reperfusion injury (IRI) and intestinal necrosis. We have previously reported that RIC significantly reduces the intestinal injury in a rat model of NEC. Here we describe the changes in intestinal mRNA occurring in the intestine of animals exposed to IRI, both with and without RIC. Related rat-pups were randomly assigned to four groups: SHAM, IRI only, RIC only and RIC + IRI. IRI animals, underwent 40 min of intestinal ischaemia, and 90 min of reperfusion. Animals that underwent RIC had three cycles of 5 min of alternating ischaemia/reperfusion by means of a ligature applied to the hind limb. Samples from the terminal ileum were immediately stored in RNA-preserving media for later next generation sequencing and transciptome analysis using R v 3.6.1. Differential expression testing showed that 868 genes differentially expressed in animals exposed to RIC alone compared to SHAM and 135 in the IRI and RIC group compared to IRI alone. Comparison between these two sets showed that 25 genes were differentially expressed in both groups. Pro-inflammatory molecules: NF-ĸβ2, Cxcl1, SOD2 and Map3k8 all show reduced expression in response to RIC. Targeted gene analysis revealed increased expression in PI3K which is part of the so-called RISK-pathway which is a key part of the protective mechanisms of RIC in the heart. Overall, this transcriptomic analysis shows that RIC provides a protective effect to the intestine via anti-inflammatory pathways. This could be particularly relevant to treating and preventing NEC.
Collapse
Affiliation(s)
- Ian Howard Jones
- University Surgery Unit, Faculty of Medicine, University of Southampton, Tremona Road, Southampton, UK.
- Birmingham Children's Hospital, Steelhouse Lane, Birmingham, UK.
| | - Jane Elizabeth Collins
- University Surgery Unit, Faculty of Medicine, University of Southampton, Tremona Road, Southampton, UK
- Clinical and Experimental Sciences, University of Southampton School of Medicine, Southampton, UK
| | - Nigel John Hall
- University Surgery Unit, Faculty of Medicine, University of Southampton, Tremona Road, Southampton, UK
- Southampton Children's Hospital, Tremona Road, Southampton, UK
| | - Ashley Ivan Heinson
- University Surgery Unit, Faculty of Medicine, University of Southampton, Tremona Road, Southampton, UK
- Clinical Informatics Research Unit, Cancer Sciences, University of Southampton School of Medicine, Southampton, UK
| |
Collapse
|
19
|
Roberts AG, Younge N, Greenberg RG. Neonatal Necrotizing Enterocolitis: An Update on Pathophysiology, Treatment, and Prevention. Paediatr Drugs 2024; 26:259-275. [PMID: 38564081 DOI: 10.1007/s40272-024-00626-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/11/2024] [Indexed: 04/04/2024]
Abstract
Necrotizing enterocolitis (NEC) is a life-threatening disease predominantly affecting premature and very low birth weight infants resulting in inflammation and necrosis of the small bowel and colon and potentially leading to sepsis, peritonitis, perforation, and death. Numerous research efforts have been made to better understand, treat, and prevent NEC. This review explores a variety of factors involved in the pathogenesis of NEC (prematurity, low birth weight, lack of human breast milk exposure, alterations to the microbiota, maternal and environmental factors, and intestinal ischemia) and reports treatment modalities surrounding NEC, including pain medications and common antibiotic combinations, the rationale for these combinations, and recent antibiotic stewardship approaches surrounding NEC treatment. This review also highlights the effect of early antibiotic exposure, infections, proton pump inhibitors (PPIs), and H2 receptor antagonists on the microbiota and how these risk factors can increase the chances of NEC. Finally, modern prevention strategies including the use of human breast milk and standardized feeding regimens are discussed, as well as promising new preventative and treatment options for NEC including probiotics and stem cell therapy.
Collapse
|
20
|
Cui Z, Xu H, Wu F, Chen J, Zhu L, Shen Z, Yi X, Yang J, Jia C, Zhang L, Zhou P, Li MJ, Zhu L, Duan S, Yao Z, Yu Y, Liu Q, Zhou J. Maternal circadian rhythm disruption affects neonatal inflammation via metabolic reprograming of myeloid cells. Nat Metab 2024; 6:899-913. [PMID: 38561509 DOI: 10.1038/s42255-024-01021-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 03/05/2024] [Indexed: 04/04/2024]
Abstract
Disruption of circadian rhythm during pregnancy produces adverse health outcomes in offspring; however, the role of maternal circadian rhythms in the immune system of infants and their susceptibility to inflammation remains poorly understood. Here we show that disruption of circadian rhythms in pregnant mice profoundly aggravates the severity of neonatal inflammatory disorders in both male and female offspring, such as necrotizing enterocolitis and sepsis. The diminished maternal production of docosahexaenoic acid (DHA) and the impaired immunosuppressive function of neonatal myeloid-derived suppressor cells (MDSCs) contribute to this phenomenon. Mechanistically, DHA enhances the immunosuppressive function of MDSCs via PPARγ-mediated mitochondrial oxidative phosphorylation. Transfer of MDSCs or perinatal supplementation of DHA relieves neonatal inflammation induced by maternal rhythm disruption. These observations collectively demonstrate a previously unrecognized role of maternal circadian rhythms in the control of neonatal inflammation via metabolic reprograming of myeloid cells.
Collapse
Affiliation(s)
- Zhaohai Cui
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, International Joint Laboratory of Ocular Diseases, Ministry of Education, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- Institute of Pediatric Health and Disease, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangzhou, China
| | - Haixu Xu
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, International Joint Laboratory of Ocular Diseases, Ministry of Education, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Fan Wu
- Institute of Pediatric Health and Disease, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangzhou, China
- Department of Neonatology, Guangzhou Key Laboratory of Neonatal Intestinal Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiale Chen
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, International Joint Laboratory of Ocular Diseases, Ministry of Education, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Lin Zhu
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, International Joint Laboratory of Ocular Diseases, Ministry of Education, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Zhuxia Shen
- Department of Cardiology, Jing'an District Central Hospital of Shanghai, Fudan University, Shanghai, China
| | - Xianfu Yi
- Department of Bioinformatics, Tianjin, China
| | - Jinhao Yang
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, International Joint Laboratory of Ocular Diseases, Ministry of Education, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Chunhong Jia
- Institute of Pediatric Health and Disease, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangzhou, China
- Department of Neonatology, Guangzhou Key Laboratory of Neonatal Intestinal Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lijuan Zhang
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, International Joint Laboratory of Ocular Diseases, Ministry of Education, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Pan Zhou
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, International Joint Laboratory of Ocular Diseases, Ministry of Education, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | | | - Lu Zhu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Shengzhong Duan
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang, China
| | - Zhi Yao
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, International Joint Laboratory of Ocular Diseases, Ministry of Education, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ying Yu
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, International Joint Laboratory of Ocular Diseases, Ministry of Education, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| | - Qiang Liu
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, International Joint Laboratory of Ocular Diseases, Ministry of Education, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Jie Zhou
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, International Joint Laboratory of Ocular Diseases, Ministry of Education, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
21
|
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
|
22
|
Tuşat M, Eroz R, Bölükbaş F, Özkan E, Erdal H. Evaluation of the protective and therapeutic effects of extra virgin olive oil rich in phenol in experimental model of neonatal necrotizing enterocolitis by clinical disease score, ınflammation, apoptosis, and oxidative stress markers. Pediatr Surg Int 2024; 40:80. [PMID: 38493431 DOI: 10.1007/s00383-024-05669-1] [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] [Accepted: 02/29/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND AND AIM Necrotizing Enterocolitis (NEC) is an inflammation-associated ischemic necrosis of the intestine. To investigate the effects of extra virgin olive oil (EVOO) on inflammation, oxidative stress, apoptosis, and histological changes in NEC-induced newborn rats. MATERIALS AND METHODS 24 rats were randomly divided into three groups: control, NEC and NEC + EVOO. NEC induction was performed using hypoxia-hyperoxia, formula feeding, and cold stress. The NEC + EVOO group received 2 ml/kg EVOO with high phenolic content by gavage twice a day for 3 days. 3 cm of bowel including terminal ileum, cecum, and proximal colon was excised. RESULTS Weight gain and clinical disease scores were significantly higher in the NEC + EVOO group than in the NEC group (p < 0.001). EVOO treatment caused significant decreases in IL1β, IL6 levels (p = 0.016, p = 0.029 respectively) and EGF, MDA levels (p = 0.032, p = 0.013 respectively) compared to NEC group. Significant decreases were observed in IL6 gene expression in the NEC + EVOO group compared to the NEC group (p = 0.002). In the group NEC + EVOO, the number of Caspase-3 positive cells was found to be significantly reduced (p < 0.001) and histopathological examination revealed minimal changes and significantly lower histopathological scores (p < 0.001). CONCLUSION Phenol-rich EVOO prevents intestinal damage caused by NEC by inhibiting inflammation, oxidative stress, apoptosis.
Collapse
Affiliation(s)
- Mustafa Tuşat
- Department of Pediatric Surgery, Aksaray University Medical Faculty, Aksaray, Turkey.
| | - Recep Eroz
- Department of Medical Genetics, Aksaray University Medical Faculty, Aksaray, Turkey
| | - Ferhan Bölükbaş
- Department of Histology and Embryology, Aksaray University Medical Faculty, Aksaray, Turkey
| | - Erkan Özkan
- Faculty of Veterinary Medicine, Department of Parasitology, Aksaray University, Aksaray, Turkey
| | - Hüseyin Erdal
- Department of Medical Genetics, Aksaray University Medical Faculty, Aksaray, Turkey
| |
Collapse
|
23
|
Wang X, Li L, Liu T, Shi Y. More than nutrition: Therapeutic potential and mechanism of human milk oligosaccharides against necrotizing enterocolitis. Life Sci 2024; 339:122420. [PMID: 38218534 DOI: 10.1016/j.lfs.2024.122420] [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: 10/22/2023] [Revised: 12/31/2023] [Accepted: 01/08/2024] [Indexed: 01/15/2024]
Abstract
Human milk is the most valuable source of nutrition for infants. The structure and function of human milk oligosaccharides (HMOs), which are key components of human milk, have long been attracting particular research interest. Several recent studies have found HMOs to be efficacious in the prevention and treatment of necrotizing enterocolitis (NEC). Additionally, they could be developed in the future as non-invasive predictive markers for NEC. Based on previous findings and the well-defined functions of HMOs, we summarize potential protective mechanisms of HMOs against neonatal NEC, which include: modulating signal receptor function, promoting intestinal epithelial cell proliferation, reducing apoptosis, restoring intestinal blood perfusion, regulating microbial prosperity, and alleviating intestinal inflammation. HMOs supplementation has been demonstrated to be protective against NEC in both animal studies and clinical observations. This calls for mass production and use of HMOs in infant formula, necessitating more research into the safety of industrially produced HMOs and the appropriate dosage in infant formula.
Collapse
Affiliation(s)
- Xinru Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, San Hao Street, Heping District, Shenyang, Liaoning 110004, China
| | - Ling Li
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, San Hao Street, Heping District, Shenyang, Liaoning 110004, China
| | - Tianjing Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, San Hao Street, Heping District, Shenyang, Liaoning 110004, China.
| | - Yongyan Shi
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, San Hao Street, Heping District, Shenyang, Liaoning 110004, China.
| |
Collapse
|
24
|
Liu J, Joseph S, Manohar K, Lee J, Brokaw JP, Shelley WC, Markel TA. Role of innate T cells in necrotizing enterocolitis. Front Immunol 2024; 15:1357483. [PMID: 38390341 PMCID: PMC10881895 DOI: 10.3389/fimmu.2024.1357483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 01/16/2024] [Indexed: 02/24/2024] Open
Abstract
Necrotizing enterocolitis (NEC) is a destructive gastrointestinal disease primarily affecting preterm babies. Despite advancements in neonatal care, NEC remains a significant cause of morbidity and mortality in neonatal intensive care units worldwide and the etiology of NEC is still unclear. Risk factors for NEC include prematurity, very low birth weight, feeding with formula, intestinal dysbiosis and bacterial infection. A review of the literature would suggest that supplementation of prebiotics and probiotics prevents NEC by altering the immune responses. Innate T cells, a highly conserved subpopulation of T cells that responds quickly to stimulation, develops differently from conventional T cells in neonates. This review aims to provide a succinct overview of innate T cells in neonates, encompassing their phenotypic characteristics, functional roles, likely involvement in the pathogenesis of NEC, and potential therapeutic implications.
Collapse
Affiliation(s)
- Jianyun Liu
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Sharon Joseph
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Krishna Manohar
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jasmine Lee
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - John P. Brokaw
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - W. Christopher Shelley
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- Riley Hospital for Children at Indiana University Health, Indianapolis, IN, United States
| | - Troy A. Markel
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- Riley Hospital for Children at Indiana University Health, Indianapolis, IN, United States
| |
Collapse
|
25
|
Chen Z, Chen H, Huang W, Guo X, Yu L, Shan J, Deng X, Liu J, Li W, Shen W, Fan H. Bacteroides fragilis alleviates necrotizing enterocolitis through restoring bile acid metabolism balance using bile salt hydrolase and inhibiting FXR-NLRP3 signaling pathway. Gut Microbes 2024; 16:2379566. [PMID: 39013030 PMCID: PMC11253882 DOI: 10.1080/19490976.2024.2379566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 07/09/2024] [Indexed: 07/18/2024] Open
Abstract
Necrotizing enterocolitis (NEC) is a leading cause of morbidity and mortality in premature infants with no specific treatments available. We aimed to identify the molecular mechanisms underlying NEC and investigate the therapeutic effects of Bacteroides fragilis on NEC. Clinical samples of infant feces, bile acid-targeted metabolomics, pathological staining, bioinformatics analysis, NEC rat model, and co-immunoprecipitation were used to explore the pathogenesis of NEC. Taxonomic characterization of the bile salt hydrolase (bsh) gene, enzyme activity assays, 16S rRNA sequencing, and organoids were used to explore the therapeutic effects of B. fragilis on NEC-related intestinal damage. Clinical samples, NEC rat models, and in vitro experiments revealed that total bile acid increased in the blood but decreased in feces. Moreover, the levels of FXR and other bile acid metabolism-related genes were abnormal, resulting in disordered bile acid metabolism in NEC. Taurochenodeoxycholic acid accelerated NEC pathogenesis and taurodeoxycholate alleviated NEC. B. fragilis displayed bsh genes and enzyme activity and alleviated intestinal damage by restoring gut microbiota dysbiosis and bile acid metabolism abnormalities by inhibiting the FXR-NLRP3 signaling pathway. Our results provide valuable insights into the therapeutic role of B. fragilis in NEC. Administering B. fragilis may substantially alleviate intestinal damage in NEC.
Collapse
MESH Headings
- Enterocolitis, Necrotizing/metabolism
- Enterocolitis, Necrotizing/microbiology
- Enterocolitis, Necrotizing/drug therapy
- Animals
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- Bacteroides fragilis/metabolism
- Bacteroides fragilis/genetics
- Signal Transduction/drug effects
- Bile Acids and Salts/metabolism
- Rats
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Cytoplasmic and Nuclear/genetics
- Gastrointestinal Microbiome/drug effects
- Amidohydrolases/metabolism
- Amidohydrolases/genetics
- Humans
- Rats, Sprague-Dawley
- Infant, Newborn
- Disease Models, Animal
- Male
- Female
- Probiotics/administration & dosage
- Probiotics/pharmacology
- Infant, Premature
- Dysbiosis/microbiology
Collapse
Affiliation(s)
- Zhenhui Chen
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Huijuan Chen
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Wanwen Huang
- Experimental Teaching Center of Preventive Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xiaotong Guo
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Lu Yu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiamin Shan
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xiaoshi Deng
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jiaxin Liu
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Wendan Li
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Wei Shen
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongying Fan
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| |
Collapse
|
26
|
Manohar K, Mesfin FM, Liu J, Shelley WC, Brokaw JP, Markel TA. Effect of Oral Chondroitin Sulfate Supplementation on Acute Brain Injury in a Murine Necrotizing Enterocolitis Model. J Am Coll Surg 2024; 238:82-98. [PMID: 37870229 DOI: 10.1097/xcs.0000000000000896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is a devastating condition where inflammatory changes and necrosis in the gut results in activation of brain microglia and subsequent neurodevelopmental impairment. Chondroitin sulfate (CS) is a glycosaminoglycan in human breast milk that is absent in conventional formulas. We hypothesized that oral formula supplementation with CS during a murine model of experimental NEC would not only attenuate intestinal injury, but also brain injury. STUDY DESIGN NEC was induced in mouse pups on postnatal days (PNDs) 5 to 8. Three conditions were studied: (1) breastfed controls, (2) NEC, and (3) NEC+enteral CS (formula+200 mg/kg/d of CS). Pups were euthanized on PND 9 or reunited with dams by the evening of PND 8. Intestinal segments were H&E stained, and immunohistochemistry was performed on brain tissue for Iba-1 to assess for microglial morphology and cortical changes. Neurodevelopmental assays were performed on mice reunited with foster dams on PND 9. Single-cell RNA-sequencing analysis was performed on human intestinal epithelial cells exposed to (1) nothing, (2) hydrogen peroxide (H 2 O 2 ) alone, or (3) H 2 O 2 + CS to look at the differential gene expression between groups. Groups were compared with ANOVA or Kruskal-Wallis tests as appropriate with p < 0.05 considered significant. RESULTS Compared with NEC, mice treated with oral CS showed improved clinical outcomes, decreased intestinal injury, and attenuated microglial activation and deleterious cortical change. Mice with CS performed better on early neurodevelopmental assays when compared with NEC alone. Single-cell analysis of HIEC-6 cells demonstrated that CS treatment down regulated several inflammatory pathways including nuclear factor κB-suggesting an explanation for the improved Th17 intestinal cytokine profile. CONCLUSIONS Oral CS supplementation improved both physiological, clinical, and developmental outcomes. These data suggest that CS is a safe compound for formula supplementation for the prevention of NEC.
Collapse
Affiliation(s)
- Krishna Manohar
- From the Department of Surgery, Indiana University School of Medicine, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
- Riley Hospital for Children at Indiana University Health, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
| | - Fikir M Mesfin
- From the Department of Surgery, Indiana University School of Medicine, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
- Riley Hospital for Children at Indiana University Health, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
| | - Jianyun Liu
- From the Department of Surgery, Indiana University School of Medicine, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
- Riley Hospital for Children at Indiana University Health, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
| | - W Christopher Shelley
- From the Department of Surgery, Indiana University School of Medicine, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
- Riley Hospital for Children at Indiana University Health, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
| | - John P Brokaw
- From the Department of Surgery, Indiana University School of Medicine, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
- Riley Hospital for Children at Indiana University Health, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
| | - Troy A Markel
- From the Department of Surgery, Indiana University School of Medicine, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
- Riley Hospital for Children at Indiana University Health, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
| |
Collapse
|
27
|
Cui Z, Amevor FK, Zhao X, Mou C, Pang J, Peng X, Liu A, Lan X, Liu L. Potential therapeutic effects of milk-derived exosomes on intestinal diseases. J Nanobiotechnology 2023; 21:496. [PMID: 38115131 PMCID: PMC10731872 DOI: 10.1186/s12951-023-02176-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/25/2023] [Indexed: 12/21/2023] Open
Abstract
Exosomes are extracellular vesicles with the diameter of 30 ~ 150 nm, and are widely involved in intercellular communication, disease diagnosis and drug delivery carriers for targeted disease therapy. Therapeutic application of exosomes as drug carriers is limited due to the lack of sources and methods for obtaining adequate exosomes. Milk contains abundant exosomes, several studies have shown that milk-derived exosomes play crucial roles in preventing and treating intestinal diseases. In this review, we summarized the biogenesis, secretion and structure, current novel methods used for the extraction and identification of exosomes, as well as discussed the role of milk-derived exosomes in treating intestinal diseases, such as inflammatory bowel disease, necrotizing enterocolitis, colorectal cancer, and intestinal ischemia and reperfusion injury by regulating intestinal immune homeostasis, restoring gut microbiota composition and improving intestinal structure and integrity, alleviating conditions such as oxidative stress, cell apoptosis and inflammation, and reducing mitochondrial reactive oxygen species (ROS) and lysosome accumulation in both humans and animals. In addition, we discussed future prospects for the standardization of milk exosome production platform to obtain higher concentration and purity, and complete exosomes derived from milk. Several in vivo clinical studies are needed to establish milk-derived exosomes as an effective and efficient drug delivery system, and promote its application in the treatment of various diseases in both humans and animals.
Collapse
Affiliation(s)
- Zhifu Cui
- College of Animal Science and Technology, Southwest University, Chongqing, P. R. China
| | - Felix Kwame Amevor
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, P. R. China
| | - Xingtao Zhao
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Sichuan, P. R. China
| | - Chunyan Mou
- College of Animal Science and Technology, Southwest University, Chongqing, P. R. China
| | - Jiaman Pang
- College of Animal Science and Technology, Southwest University, Chongqing, P. R. China
| | - Xie Peng
- College of Animal Science and Technology, Southwest University, Chongqing, P. R. China
| | - Anfang Liu
- College of Animal Science and Technology, Southwest University, Chongqing, P. R. China
| | - Xi Lan
- College of Animal Science and Technology, Southwest University, Chongqing, P. R. China.
| | - Lingbin Liu
- College of Animal Science and Technology, Southwest University, Chongqing, P. R. China.
- College of Animal Science and Technology, Chongqing Key Laboratory of Forage & Herbivore, Chongqing Engineering Research Center for Herbivores Resource Protection and Utilization, Southwest University, Beibei, Chongqing, 400715, P. R. China.
| |
Collapse
|
28
|
Zeng X, Liu MH, Xiong Y, Zheng LX, Guo KE, Zhao HM, Yin YT, Liu DY, Zhou BG. Pien Tze Huang alleviates Concanavalin A-induced autoimmune hepatitis by regulating intestinal microbiota and memory regulatory T cells. World J Gastroenterol 2023; 29:5988-6016. [PMID: 38130997 PMCID: PMC10731150 DOI: 10.3748/wjg.v29.i45.5988] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 09/26/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND Traditional Chinese medicine has used the drug Pien Tze Huang (PTH), a classic prescription, to treat autoimmune hepatitis (AIH). However, the precise mode of action is still unknown. AIM To investigate the mechanism of PTH in an AIH mouse model by determining the changes in gut microbiota structure and memory regulatory T (mTreg) cells functional levels. METHODS Following induction of the AIH mouse model induced by Concanavalin A (Con A), prophylactic administration of PTH was given for 10 d. The levels of mTreg cells were measured by flow cytometry, and intestinal microbiota was analyzed by 16S rRNA analysis, while western blotting was used to identify activation of the toll-like receptor (TLR)2, TLR4/nuclear factor-κB (NF-κB), and CXCL16/CXCR6 signaling pathways. RESULTS In the liver of mice with AIH, PTH relieved the pathological damage and reduced the numbers of T helper type 17 cells and interferon-γ, tumor necrosis factor-alpha, interleukin (IL)-1β, IL-2, IL-6, and IL-21 expression. Simultaneously, PTH stimulated the abundance of helpful bacteria, promoted activation of the TLR2 signal, which may enhance Treg/mTreg cells quantity to produce IL-10, and suppressed activation of the TLR4/NF-κB and CXCL16/CXCR6 signaling pathways. CONCLUSION PTH regulates intestinal microbiota balance and restores mTreg cells to alleviate experimental AIH, which is closely related to the TLR/CXCL16/CXCR6/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Xin Zeng
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Miao-Hua Liu
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Yi Xiong
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Lin-Xin Zheng
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Kai-En Guo
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Hai-Mei Zhao
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Yu-Ting Yin
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Duan-Yong Liu
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Bu-Gao Zhou
- Office of Academic Research, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| |
Collapse
|
29
|
Kolba N, Tako E. Effective alternatives for dietary interventions for necrotizing enterocolitis: a systematic review of in vivo studies. Crit Rev Food Sci Nutr 2023; 65:811-831. [PMID: 37971890 DOI: 10.1080/10408398.2023.2281623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
Necrotizing enterocolitis (NEC) is a significant cause of morbidity and mortality among neonates and low birth weight children in the United States. Current treatment options, such as antibiotics and intestinal resections, often result in complications related to pediatric nutrition and development. This systematic review aimed to identify alternative dietary bioactive compounds that have shown promising outcomes in ameliorating NEC in vivo studies conducted within the past six years. Following PRISMA guidelines and registering in PROSPERO (CRD42023330617), we conducted a comprehensive search of PubMed, Scopus, and Web of Science. Our analysis included 19 studies, predominantly involving in vivo models of rats (Rattus norvegicus) and mice (Mus musculus). The findings revealed that various types of compounds have demonstrated successful amelioration of NEC symptoms. Specifically, six studies employed plant phenolics, seven utilized plant metabolites/cytotoxic chemicals, three explored the efficacy of vitamins, and three investigated the potential of whole food extracts. Importantly, all administered compounds exhibited positive effects in mitigating the disease. These results highlight the potential of natural cytotoxic chemicals derived from medicinal plants in identifying and implementing powerful alternative drugs and therapies for NEC. Such approaches have the capacity to impact multiple pathways involved in the development and progression of NEC symptoms.
Collapse
Affiliation(s)
- Nikolai Kolba
- Department of Food Science, Cornell University, Ithaca, New York, USA
| | - Elad Tako
- Department of Food Science, Cornell University, Ithaca, New York, USA
| |
Collapse
|
30
|
Frazer LC, Yamaguchi Y, Jania CM, Lanik WE, Gong Q, Singh DK, Mackay S, Akopyants NS, Good M. Microfluidic Model of Necrotizing Enterocolitis Incorporating Human Neonatal Intestinal Enteroids and a Dysbiotic Microbiome. J Vis Exp 2023:10.3791/65605. [PMID: 37590536 PMCID: PMC11003451 DOI: 10.3791/65605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/19/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a severe and potentially fatal intestinal disease that has been difficult to study due to its complex pathogenesis, which remains incompletely understood. The pathophysiology of NEC includes disruption of intestinal tight junctions, increased gut barrier permeability, epithelial cell death, microbial dysbiosis, and dysregulated inflammation. Traditional tools to study NEC include animal models, cell lines, and human or mouse intestinal organoids. While studies using those model systems have improved the field's understanding of disease pathophysiology, their ability to recapitulate the complexity of human NEC is limited. An improved in vitro model of NEC using microfluidic technology, named NEC-on-a-chip, has now been developed. The NEC-on-a-chip model consists of a microfluidic device seeded with intestinal enteroids derived from a preterm neonate, co-cultured with human endothelial cells and the microbiome from an infant with severe NEC. This model is a valuable tool for mechanistic studies into the pathophysiology of NEC and a new resource for drug discovery testing for neonatal intestinal diseases. In this manuscript, a detailed description of the NEC-on-a-chip model will be provided.
Collapse
Affiliation(s)
- Lauren C Frazer
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill
| | - Yukihiro Yamaguchi
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill
| | - Corey M Jania
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill
| | | | - Qingqing Gong
- Department of Surgery, Washington University School of Medicine
| | - Dhirendra K Singh
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill
| | - Stephen Mackay
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill
| | - Natalia S Akopyants
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill
| | - Misty Good
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill;
| |
Collapse
|
31
|
Sodhi CP, Ahmad R, Fulton WB, Lopez CM, Eke BO, Scheese D, Duess JW, Steinway SN, Raouf Z, Moore H, Tsuboi K, Sampah ME, Jang HS, Buck RH, Hill DR, Niemiro GM, Prindle T, Wang S, Wang M, Jia H, Catazaro J, Lu P, Hackam DJ. Human milk oligosaccharides reduce necrotizing enterocolitis-induced neuroinflammation and cognitive impairment in mice. Am J Physiol Gastrointest Liver Physiol 2023; 325:G23-G41. [PMID: 37120853 PMCID: PMC10259852 DOI: 10.1152/ajpgi.00233.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 04/20/2023] [Accepted: 04/23/2023] [Indexed: 05/02/2023]
Abstract
Necrotizing enterocolitis (NEC) is the leading cause of morbidity and mortality in premature infants. One of the most devastating complications of NEC is the development of NEC-induced brain injury, which manifests as impaired cognition that persists beyond infancy and which represents a proinflammatory activation of the gut-brain axis. Given that oral administration of the human milk oligosaccharides (HMOs) 2'-fucosyllactose (2'-FL) and 6'-sialyslactose (6'-SL) significantly reduced intestinal inflammation in mice, we hypothesized that oral administration of these HMOs would reduce NEC-induced brain injury and sought to determine the mechanisms involved. We now show that the administration of either 2'-FL or 6'-SL significantly attenuated NEC-induced brain injury, reversed myelin loss in the corpus callosum and midbrain of newborn mice, and prevented the impaired cognition observed in mice with NEC-induced brain injury. In seeking to define the mechanisms involved, 2'-FL or 6'-SL administration resulted in a restoration of the blood-brain barrier in newborn mice and also had a direct anti-inflammatory effect on the brain as revealed through the study of brain organoids. Metabolites of 2'-FL were detected in the infant mouse brain by nuclear magnetic resonance (NMR), whereas intact 2'-FL was not. Strikingly, the beneficial effects of 2'-FL or 6'-SL against NEC-induced brain injury required the release of the neurotrophic factor brain-derived neurotrophic factor (BDNF), as mice lacking BDNF were not protected by these HMOs from the development of NEC-induced brain injury. Taken in aggregate, these findings reveal that the HMOs 2'-FL and 6'-SL interrupt the gut-brain inflammatory axis and reduce the risk of NEC-induced brain injury.NEW & NOTEWORTHY This study reveals that the administration of human milk oligosaccharides, which are present in human breast milk, can interfere with the proinflammatory gut-brain axis and prevent neuroinflammation in the setting of necrotizing enterocolitis, a major intestinal disorder seen in premature infants.
Collapse
Affiliation(s)
- Chhinder P Sodhi
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Raheel Ahmad
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - William B Fulton
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Carla M Lopez
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Benjamin O Eke
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Daniel Scheese
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Johannes W Duess
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Steve N Steinway
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Zachariah Raouf
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Hannah Moore
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Koichi Tsuboi
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Maame Efua Sampah
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Hee-Seong Jang
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Rachael H Buck
- Nutrition Division, Abbott, Columbus, Ohio, United States
| | - David R Hill
- Nutrition Division, Abbott, Columbus, Ohio, United States
| | | | - Thomas Prindle
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Sanxia Wang
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Menghan Wang
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Hongpeng Jia
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Jonathan Catazaro
- Department of Chemistry, Johns Hopkins University, Baltimore, Maryland, United States
| | - Peng Lu
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - David J Hackam
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| |
Collapse
|
32
|
Sami AS, Frazer LC, Miller CM, Singh DK, Clodfelter LG, Orgel KA, Good M. The role of human milk nutrients in preventing necrotizing enterocolitis. Front Pediatr 2023; 11:1188050. [PMID: 37334221 PMCID: PMC10272619 DOI: 10.3389/fped.2023.1188050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/22/2023] [Indexed: 06/20/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is an intestinal disease that primarily impacts preterm infants. The pathophysiology of NEC involves a complex interplay of factors that result in a deleterious immune response, injury to the intestinal mucosa, and in its most severe form, irreversible intestinal necrosis. Treatments for NEC remain limited, but one of the most effective preventative strategies for NEC is the provision of breast milk feeds. In this review, we discuss mechanisms by which bioactive nutrients in breast milk impact neonatal intestinal physiology and the development of NEC. We also review experimental models of NEC that have been used to study the role of breast milk components in disease pathophysiology. These models are necessary to accelerate mechanistic research and improve outcomes for neonates with NEC.
Collapse
Affiliation(s)
- Ahmad S. Sami
- Division of Pediatric Gastroenterology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Lauren C. Frazer
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Claire M. Miller
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Dhirendra K. Singh
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Lynda G. Clodfelter
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kelly A. Orgel
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Misty Good
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
33
|
Scheese DJ, Sodhi CP, Hackam DJ. New insights into the pathogenesis of necrotizing enterocolitis and the dawn of potential therapeutics. Semin Pediatr Surg 2023; 32:151309. [PMID: 37290338 PMCID: PMC10330774 DOI: 10.1016/j.sempedsurg.2023.151309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Necrotizing enterocolitis (NEC) is a devastating gastrointestinal disorder in premature infants that causes significant morbidity and mortality. Research efforts into the pathogenesis of NEC have discovered a pivotal role for the gram-negative bacterial receptor, Toll-like receptor 4 (TLR4), in its development. TLR4 is activated by dysbiotic microbes within the intestinal lumen, which leads to an exaggerated inflammatory response within the developing intestine, resulting in mucosal injury. More recently, studies have identified that the impaired intestinal motility that occurs early in NEC has a causative role in disease development, as strategies to enhance intestinal motility can reverse NEC in preclinical models. There has also been broad appreciation that NEC also contributes to significant neuroinflammation, which we have linked to the effects of gut-derived pro-inflammatory molecules and immune cells which activate microglia in the developing brain, resulting in white matter injury. These findings suggest that the management of the intestinal inflammation may secondarily be neuroprotective. Importantly, despite the significant burden of NEC on premature infants, these and other studies have provided a strong rationale for the development of small molecules with the capability of reducing NEC severity in pre-clinical models, thus guiding the development of specific anti-NEC therapies. This review summarizes the roles of TLR4 signaling in the premature gut in the pathogenesis of NEC, and provides insights into optimal clinical management strategies based upon findings from laboratory studies.
Collapse
Affiliation(s)
- Daniel J Scheese
- Division of Pediatric Surgery, Johns Hopkins University School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Chhinder P Sodhi
- Division of Pediatric Surgery, Johns Hopkins University School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA
| | - David J Hackam
- Division of Pediatric Surgery, Johns Hopkins University School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA.
| |
Collapse
|
34
|
Coleman S, Unterhauser K, Rezaul K, Ledala N, Lesmes S, Caimano MJ, Zhou Y, Jackson E, Gratalo D, Driscoll MD, Matson AP. High-resolution microbiome analysis reveals exclusionary Klebsiella species competition in preterm infants at risk for necrotizing enterocolitis. Sci Rep 2023; 13:7893. [PMID: 37193703 DOI: 10.1038/s41598-023-34735-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 05/06/2023] [Indexed: 05/18/2023] Open
Abstract
Intestinal colonization with Klebsiella has been linked to necrotizing enterocolitis (NEC), but methods of analysis usually failed to discriminate Klebsiella species or strains. A novel ~ 2500-base amplicon (StrainID) that spans the 16S and 23S rRNA genes was used to generate amplicon sequence variant (ASV) fingerprints for Klebsiella oxytoca and Klebsiella pneumoniae species complexes (KoSC and KpSC, respectively) and co-occurring fecal bacterial strains from 10 preterm infants with NEC and 20 matched controls. Complementary approaches were used to identify cytotoxin-producing isolates of KoSC. Klebsiella species colonized most preterm infants, were more prevalent in NEC subjects versus controls, and replaced Escherichia in NEC subjects. Single KoSC or KpSC ASV fingerprinted strains dominated the gut microbiota, suggesting exclusionary Klebsiella competition for luminal resources. Enterococcus faecalis was co-dominant with KoSC but present infrequently with KpSC. Cytotoxin-producing KoSC members were identified in most NEC subjects and were less frequent in controls. Few Klebsiella strains were shared between subjects. We conclude that inter-species Klebsiella competition, within an environment of KoSC and E. faecalis cooperation, appears to be an important factor for the development of NEC. Preterm infants seem to acquire Klebsiella primarily through routes other than patient-to-patient transmission.
Collapse
Affiliation(s)
- Spencer Coleman
- Department of Pediatrics, UConn Health, Farmington, CT, USA
- University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, USA
| | | | - Karim Rezaul
- Department of Pediatrics, UConn Health, Farmington, CT, USA
| | | | - Stephanie Lesmes
- Department of Research, Connecticut Children's Medical Center, Hartford, CT, USA
| | - Melissa J Caimano
- Department of Pediatrics, UConn Health, Farmington, CT, USA
- Department of Medicine, UConn Health, Farmington, CT, USA
- Department of Molecular Biology and Biophysics, UConn Health, Farmington, CT, USA
| | - Yanjiao Zhou
- Department of Medicine, UConn Health, Farmington, CT, USA
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | | | | | - Adam P Matson
- Department of Pediatrics, UConn Health, Farmington, CT, USA.
- Division of Neonatology, Connecticut Children's Medical Center, Hartford, CT, USA.
- Department of Immunology, UConn Health, Farmington, CT, USA.
| |
Collapse
|
35
|
Egozi A, Olaloye O, Werner L, Silva T, McCourt B, Pierce RW, An X, Wang F, Chen K, Pober JS, Shouval D, Itzkovitz S, Konnikova L. Single-cell atlas of the human neonatal small intestine affected by necrotizing enterocolitis. PLoS Biol 2023; 21:e3002124. [PMID: 37205711 PMCID: PMC10234541 DOI: 10.1371/journal.pbio.3002124] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 06/01/2023] [Accepted: 04/13/2023] [Indexed: 05/21/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a gastrointestinal complication of premature infants with high rates of morbidity and mortality. A comprehensive view of the cellular changes and aberrant interactions that underlie NEC is lacking. This study aimed at filling in this gap. We combine single-cell RNA sequencing (scRNAseq), T-cell receptor beta (TCRβ) analysis, bulk transcriptomics, and imaging to characterize cell identities, interactions, and zonal changes in NEC. We find an abundance of proinflammatory macrophages, fibroblasts, endothelial cells as well as T cells that exhibit increased TCRβ clonal expansion. Villus tip epithelial cells are reduced in NEC and the remaining epithelial cells up-regulate proinflammatory genes. We establish a detailed map of aberrant epithelial-mesenchymal-immune interactions that are associated with inflammation in NEC mucosa. Our analyses highlight the cellular dysregulations of NEC-associated intestinal tissue and identify potential targets for biomarker discovery and therapeutics.
Collapse
Affiliation(s)
- Adi Egozi
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Oluwabunmi Olaloye
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Lael Werner
- Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children’s Medical Center of Israel, Petah Tikva, Israel, affiliated to the Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tatiana Silva
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Blake McCourt
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Richard W. Pierce
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut, United States of America
- Program in Human and Translational Immunology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Xiaojing An
- Department of Medicine, University of Pittsburgh Medical Center Montefiore Hospital, Pittsburgh, Pennsylvania, United States of America
| | - Fujing Wang
- Department of Medicine, University of Pittsburgh Medical Center Montefiore Hospital, Pittsburgh, Pennsylvania, United States of America
| | - Kong Chen
- Department of Medicine, University of Pittsburgh Medical Center Montefiore Hospital, Pittsburgh, Pennsylvania, United States of America
| | - Jordan S. Pober
- Program in Human and Translational Immunology, Yale School of Medicine, New Haven, Connecticut, United States of America
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Dror Shouval
- Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children’s Medical Center of Israel, Petah Tikva, Israel, affiliated to the Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shalev Itzkovitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Liza Konnikova
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut, United States of America
- Program in Human and Translational Immunology, Yale School of Medicine, New Haven, Connecticut, United States of America
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, United States of America
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, United States of America
| |
Collapse
|
36
|
Oshima K, Hinoki A, Uchida H, Tanaka Y, Okuno Y, Go Y, Shirota C, Tainaka T, Sumida W, Yokota K, Makita S, Takimoto A, Kano Y, Sawa S. Single-cell RNA sequencing of intestinal immune cells in neonatal necrotizing enterocolitis. Pediatr Surg Int 2023; 39:179. [PMID: 37041419 DOI: 10.1007/s00383-023-05461-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/03/2023] [Indexed: 04/13/2023]
Abstract
PURPOSE Necrotizing enterocolitis (NEC) causes fatal intestinal necrosis in neonates, but its etiology is unknown. We analyzed the intestinal immune response to NEC. METHODS Using single-cell RNA sequencing (scRNA-seq), we analyzed the gene expression profiles of intestinal immune cells from four neonates with intestinal perforation (two with NEC and two without NEC). Target mononuclear cells were extracted from the lamina propria of the resected intestines. RESULTS In all four cases, major immune cells, such as T cells (15.1-47.7%), B cells (3.1-19.0%), monocytes (16.5-31.2%), macrophages (1.6-17.4%), dendritic cells (2.4-12.2%), and natural killer cells (7.5-12.8%), were present in similar proportions to those in the neonatal cord blood. Gene set enrichment analysis showed that the MTOR, TNF-α, and MYC signaling pathways were enriched in T cells of the NEC patients, suggesting upregulated immune responses related to inflammation and cell proliferation. In addition, all four cases exhibited a bias toward cell-mediated inflammation, based on the predominance of T helper 1 cells. CONCLUSION Intestinal immunity in NEC subjects exhibited stronger inflammatory responses compared to non-NEC subjects. Further scRNA-seq and cellular analysis may improve our understanding of the pathogenesis of NEC.
Collapse
Affiliation(s)
- Kazuo Oshima
- Department of Pediatric Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Pediatric Surgery, Saitama Medical University, Saitama, Japan
| | - Akinari Hinoki
- Department of Pediatric Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroo Uchida
- Department of Pediatric Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yujiro Tanaka
- Department of Pediatric Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Pediatric Surgery, Saitama Medical University, Saitama, Japan
| | - Yusuke Okuno
- Department of Virology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yasuhiro Go
- Cognitive Genomics Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Japan
- Department of System Neuroscience, National Institute for Physiological Science, Okazaki, Japan
- Department of Physiological Science, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Japan
| | - Chiyoe Shirota
- Department of Pediatric Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takahisa Tainaka
- Department of Pediatric Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Wataru Sumida
- Department of Pediatric Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuki Yokota
- Department of Pediatric Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Satoshi Makita
- Department of Pediatric Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Aitaro Takimoto
- Department of Pediatric Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoko Kano
- Department of Pediatric Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinichiro Sawa
- Division of Mucosal Immunology, Research Center for Systems Immunology, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
37
|
Li W, Zhang C, Li W, Qin F, Gao X, Xu F. Nomogram for predicting fulminant necrotizing enterocolitis. Pediatr Surg Int 2023; 39:154. [PMID: 36939896 PMCID: PMC10027821 DOI: 10.1007/s00383-023-05435-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/22/2023] [Indexed: 03/21/2023]
Abstract
BACKGROUND Fulminant necrotizing enterocolitis (FNEC) is the most serious subtype of NEC and has a high mortality rate and a high incidence of sequelae. Onset prediction can help in the establishment of a customized treatment strategy. This study aimed to develop and evaluate a predictive nomogram for FNEC. METHODS We conducted a retrospective observation to study the clinical data of neonates diagnosed with NEC (Bell stage ≥ IIB). Neonates were divided into the FNEC and NEC groups. A multivariate logistic regression model was used to construct the nomogram model. The performance of the nomogram was assessed using area under the curve, calibration analysis, and decision curve analysis. RESULTS A total of 206 neonate cases were included, among which 40 (19.4%) fulfilled the definition of FNEC. The identified predictors were assisted ventilation after NEC onset; shock at NEC onset; feeding volumes before NEC onset; neutrophil counts on the day of NEC onset; and neutrophil, lymphocyte, and monocyte counts on day 1 after NEC onset. The nomogram exhibited good discrimination, with an area under the receiver operating characteristic curve of 0.884 (95% CI 0.825-0.943). The predictive model was well calibrated. Decision curve analysis confirmed the clinical usefulness of this nomogram. CONCLUSION A nomogram with a potentially effective application was developed to facilitate the individualized prediction of FNEC, with the hope of providing further direction for the early diagnosis of FNEC and timing of intervention.
Collapse
Affiliation(s)
- Weibo Li
- Department of Neonatology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Branch Center of the Third Affiliated Hospital of Advanced Medical Research Center of Zhengzhou University, Zhengzhou, Henan, China
| | - Chen Zhang
- Department of Neonatology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Branch Center of the Third Affiliated Hospital of Advanced Medical Research Center of Zhengzhou University, Zhengzhou, Henan, China
| | - Wenli Li
- Department of Neonatology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Branch Center of the Third Affiliated Hospital of Advanced Medical Research Center of Zhengzhou University, Zhengzhou, Henan, China
| | - Fanyue Qin
- Department of Neonatology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Branch Center of the Third Affiliated Hospital of Advanced Medical Research Center of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiang Gao
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Falin Xu
- Department of Neonatology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Branch Center of the Third Affiliated Hospital of Advanced Medical Research Center of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
38
|
Molecular Mechanisms of Hyperoxia-Induced Neonatal Intestinal Injury. Int J Mol Sci 2023; 24:ijms24054366. [PMID: 36901800 PMCID: PMC10002283 DOI: 10.3390/ijms24054366] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/15/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Oxygen therapy is important for newborns. However, hyperoxia can cause intestinal inflammation and injury. Hyperoxia-induced oxidative stress is mediated by multiple molecular factors and leads to intestinal damage. Histological changes include ileal mucosal thickness, intestinal barrier damage, and fewer Paneth cells, goblet cells, and villi, effects which decrease the protection from pathogens and increase the risk of necrotizing enterocolitis (NEC). It also causes vascular changes with microbiota influence. Hyperoxia-induced intestinal injuries are influenced by several molecular factors, including excessive nitric oxide, the nuclear factor-κB (NF-κB) pathway, reactive oxygen species, toll-like receptor-4, CXC motif ligand-1, and interleukin-6. Nuclear factor erythroid 2-related factor 2 (Nrf2) pathways and some antioxidant cytokines or molecules including interleukin-17D, n-acetylcysteine, arginyl-glutamine, deoxyribonucleic acid, cathelicidin, and health microbiota play a role in preventing cell apoptosis and tissue inflammation from oxidative stress. NF-κB and Nrf2 pathways are essential to maintain the balance of oxidative stress and antioxidants and prevent cell apoptosis and tissue inflammation. Intestinal inflammation can lead to intestinal damage and death of the intestinal tissue, such as in NEC. This review focuses on histologic changes and molecular pathways of hyperoxia-induced intestinal injuries to establish a framework for potential interventions.
Collapse
|
39
|
Abstract
Necrotizing enterocolitis (NEC) is the leading cause of death and disability from gastrointestinal disease in premature infants. The mortality of patients with NEC is approximately 30%, a figure that has not changed in many decades, reflecting the need for a greater understanding of its pathogenesis. Progress towards understanding the cellular and molecular mechanisms underlying NEC requires the study of highly translational animal models. Such animal models must mimic the biology and physiology of premature infants, while still allowing for safe experimental manipulation of environmental and microbial factors thought to be associated with the risk and severity of NEC. Findings from animal models have yielded insights into the interactions between the host, the colonizing microbes, and the innate immune receptor Toll-like Receptor 4 (TLR4) in driving disease development. This review discusses the relative strengths and weaknesses of available in vivo, in vitro, and NEC-in-a-dish models of this disease. We also highlight the unique contributions that each model has made to our understanding of the complex interactions between enterocytes, microbiota, and immune cells in the pathogenesis of NEC. The overall purpose of this review is to provide a menu of options regarding currently available animal models of NEC, while in parallel hopefully reducing the potential uncertainty and confusion regarding NEC models to assist those who wish to enter this field from other disciplines.
Collapse
Affiliation(s)
- Carla M Lopez
- Division of Pediatric Surgery and the Department of Surgery at the Johns Hopkins University, Bloomberg Children's Center, The Johns Hopkins Hospital, Room 7323, 1800 Orleans Street, Baltimore, MD 21287, USA
| | - Maame Efua S Sampah
- Division of Pediatric Surgery and the Department of Surgery at the Johns Hopkins University, Bloomberg Children's Center, The Johns Hopkins Hospital, Room 7323, 1800 Orleans Street, Baltimore, MD 21287, USA
| | - Johannes W Duess
- Division of Pediatric Surgery and the Department of Surgery at the Johns Hopkins University, Bloomberg Children's Center, The Johns Hopkins Hospital, Room 7323, 1800 Orleans Street, Baltimore, MD 21287, USA
| | - Asuka Ishiyama
- Division of Pediatric Surgery and the Department of Surgery at the Johns Hopkins University, Bloomberg Children's Center, The Johns Hopkins Hospital, Room 7323, 1800 Orleans Street, Baltimore, MD 21287, USA
| | - Raheel Ahmad
- Division of Pediatric Surgery and the Department of Surgery at the Johns Hopkins University, Bloomberg Children's Center, The Johns Hopkins Hospital, Room 7323, 1800 Orleans Street, Baltimore, MD 21287, USA
| | - Chhinder P Sodhi
- Division of Pediatric Surgery and the Department of Surgery at the Johns Hopkins University, Bloomberg Children's Center, The Johns Hopkins Hospital, Room 7323, 1800 Orleans Street, Baltimore, MD 21287, USA
| | - David J Hackam
- Division of Pediatric Surgery and the Department of Surgery at the Johns Hopkins University, Bloomberg Children's Center, The Johns Hopkins Hospital, Room 7323, 1800 Orleans Street, Baltimore, MD 21287, USA.
| |
Collapse
|
40
|
Lu J, Martin CR, Claud EC. Neurodevelopmental outcome of infants who develop necrotizing enterocolitis: The gut-brain axis. Semin Perinatol 2023; 47:151694. [PMID: 36572620 PMCID: PMC9974904 DOI: 10.1016/j.semperi.2022.151694] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Necrotizing enterocolitis (NEC) poses a significant risk for neurodevelopmental impairment in extremely preterm infants. The gut microbiota shapes the development of the gut, immune system, and the brain; and dysbiosis drive neonatal morbidities including NEC. In this chapter, we delineate a gut-brain axis linking gut microbiota to the adverse neurological outcomes in NEC patients. We propose that in NEC, immaturity of the microbiome along with aberrant gut microbiota-driven immaturity of the gut barrier and immune system can lead to effects including systemic inflammation and circulating microbial mediators. This nexus of gut microbiota-driven systemic effects further interacts with a likewise underdeveloped blood-brain barrier to regulate neuroinflammation and neurodevelopment. Targeting deviant gut-brain axis signaling presents an opportunity to improve the neurodevelopmental outcomes of NEC patients.
Collapse
Affiliation(s)
- Jing Lu
- Department of Pediatrics, Division of Biological Sciences, University of Chicago, Pritzker School of Medicine, Chicago, Illinois 60637, United States
| | - Camilia R Martin
- Department of Pediatrics, Division of Newborn Medicine, Weill Cornell Medicine, New York, New York 10021, United States
| | - Erika C Claud
- Department of Pediatrics, Division of Biological Sciences, University of Chicago, Pritzker School of Medicine, Chicago, Illinois 60637, United States.
| |
Collapse
|
41
|
Sadeghpour Heravi F, Hu H. Bifidobacterium: Host-Microbiome Interaction and Mechanism of Action in Preventing Common Gut-Microbiota-Associated Complications in Preterm Infants: A Narrative Review. Nutrients 2023; 15:709. [PMID: 36771414 PMCID: PMC9919561 DOI: 10.3390/nu15030709] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 01/23/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
The development and health of infants are intertwined with the protective and regulatory functions of different microorganisms in the gut known as the gut microbiota. Preterm infants born with an imbalanced gut microbiota are at substantial risk of several diseases including inflammatory intestinal diseases, necrotizing enterocolitis, late-onset sepsis, neurodevelopmental disorders, and allergies which can potentially persist throughout adulthood. In this review, we have evaluated the role of Bifidobacterium as commonly used probiotics in the development of gut microbiota and prevention of common diseases in preterm infants which is not fully understood yet. The application of Bifidobacterium as a therapeutical approach in the re-programming of the gut microbiota in preterm infants, the mechanisms of host-microbiome interaction, and the mechanism of action of this bacterium have also been investigated, aiming to provide new insights and opportunities in microbiome-targeted interventions in personalized medicine.
Collapse
Affiliation(s)
| | - Honghua Hu
- Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia
- Innovation Center of Translational Pharmacy, Jinhua Institute of Zhejiang University, Jinhua 321016, China
| |
Collapse
|
42
|
Singh DK, Miller CM, Orgel KA, Dave M, Mackay S, Good M. Necrotizing enterocolitis: Bench to bedside approaches and advancing our understanding of disease pathogenesis. Front Pediatr 2023; 10:1107404. [PMID: 36714655 PMCID: PMC9874231 DOI: 10.3389/fped.2022.1107404] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 12/20/2022] [Indexed: 01/13/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a devastating, multifactorial disease mainly affecting the intestine of premature infants. Recent discoveries have significantly enhanced our understanding of risk factors, as well as, cellular and genetic mechanisms of this complex disease. Despite these advancements, no essential, single risk factor, nor the mechanism by which each risk factor affects NEC has been elucidated. Nonetheless, recent research indicates that maternal factors, antibiotic exposure, feeding, hypoxia, and altered gut microbiota pose a threat to the underdeveloped immunity of preterm infants. Here we review predisposing factors, status of unwarranted immune responses, and microbial pathogenesis in NEC based on currently available scientific evidence. We additionally discuss novel techniques and models used to study NEC and how this research translates from the bench to the bedside into potential treatment strategies.
Collapse
Affiliation(s)
- Dhirendra K. Singh
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Claire M. Miller
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kelly A. Orgel
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Mili Dave
- University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, United States
| | - Stephen Mackay
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Misty Good
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
43
|
CCL3 aggravates intestinal damage in NEC by promoting macrophage chemotaxis and M1 macrophage polarization. Pediatr Res 2022:10.1038/s41390-022-02409-w. [PMID: 36550354 DOI: 10.1038/s41390-022-02409-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 11/04/2022] [Accepted: 11/17/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND NEC is a life-threatening gastrointestinal disease in neonates, the pathogenesis of which remains poorly understood. METHODS CCL3 levels in intestinal tissue of mice were measured and analyzed. HE staining was used to assess pathological changes in intestinal tissue. FCM was used to detect the proportion and phenotype of macrophages. RNA-seq and RT-PCR were used to evaluate the effect of CCL3 on macrophages. RESULTS CCL3 was highly expressed in the intestinal tissues of mice with NEC and induced macrophage infiltration. Transcriptome data showed that CCL3 strongly induced a transition in the phenotype of macrophages into a proinflammatory one. Mechanistically, in vivo experiments confirmed that CCL3 induced M1 macrophage polarization in NEC intestinal tissue, thereby aggravating inflammatory injury of intestinal tissue, which was alleviated by anti-CCL3 treatment. In addition, in vitro experiments showed that CCL3 significantly enhances the expression of M1-related genes in both PMφ and BMDM while inhibiting the expression of M2-related genes, which was also alleviated by anti-CCl3 treatment. CONCLUSIONS Our data elucidated the involvement of CCL3 in the pathogenesis of NEC, in which upregulated CCL3 expression exacerbated inflammatory intestinal damage by regulating macrophage chemotaxis and M1 phenotype polarization, suggesting that blocking CCL3 may be a potential strategy for effective intervention in NEC. IMPACT Our study represents an important conceptual advancement that CCL3 may be one of the key culprits of intestinal tissue damage in patients with NEC. CCL3 aggravates inflammatory intestinal injury and intestinal mucosal barrier imbalance by regulating the chemotaxis, polarization, and function of macrophages. Blocking CCL3 significantly reduced NEC-mediated intestinal injury, suggesting a new potential therapeutic strategy.
Collapse
|
44
|
Akduman H, Tayman C, Korkmaz V, Akduman F, Fettah ND, Gürsoy BK, Turkmenoglu TT, Çağlayan M. Astaxanthin Reduces the Severity of Intestinal Damage in a Neonatal Rat Model of Necrotizing Enterocolitis. Am J Perinatol 2022; 39:1820-1827. [PMID: 33853144 DOI: 10.1055/s-0041-1727156] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
OBJECTIVE This study aimed to ascertain the effects of astaxanthin (ASX) in an experimental necrotizing enterocolitis (NEC) model using rat pups. STUDY DESIGN Forty-two pups born from five Wistar albino rats were randomly divided into three groups as the control group, NEC + placebo (saline), and NEC + ASX. Pups in the NEC + ASX group were given 100 mg/kg/day oral ASX from day 1 to day 4 of the study. Saline of 2 mL/kg was given to the NEC + placebo group. Histopathological, immunohistochemical (caspase-3), and biochemical evaluations including the total antioxidant status (TAS), total oxidant status (TOS), superoxide dismutase (SOD), glutathione (GSH), lipid hydroperoxide (LPO), 8-hydroxydeoxyguanosine (8-OHdG), advanced oxidation protein products (AOPP), myeloperoxidase (MPO), tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and nuclear factor erythroid 2-related factor 2 (Nfr-2) activities were all performed. RESULTS A better survival rate and weight gain were demonstrated in the NEC + ASX group (p < 0.05). In the histopathological evaluation, the severity of intestinal damage was significantly reduced in the NEC + ASX group, as well as decreased apoptosis (enzyme-linked immunosorbent assay [ELISA] for caspase-3; p = 0.001). The biochemical analyses of intestinal tissue TOS, oxidative stress index (OSI; TOS/TAS), IL-1β, LPO, 8-OHdG, AOPP, caspase-3 (p < 0.001 for all), and TNF-α and MPO (p = 0.001 for both parameters) levels were lower in the NEC + ASX group than in the NEC + placebo group. Nrf-2, TAS, GSH, and SOD levels were higher in the NEC + ASX group than in the NEC + placebo group (p = 0.001, 0.001, <0.001, and 0.01, respectively). CONCLUSION ASX treatment has been shown to effectively reduce the severity of intestinal damage in NEC due to its antioxidant, anti-inflammatory, and antiapoptotic properties. KEY POINTS · NEC causes extremely high morbidity and mortality, as well as many complications.. · We investigated the effectiveness of ASX in the experimental NEC model created in rat pups.. · First study examining the effect of ASX on the experimental NEC rat model..
Collapse
Affiliation(s)
- Hasan Akduman
- Division of Neonatology, Department of Pediatrics, SBU Ankara Dr. Sami Ulus Maternity Child Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Cuneyt Tayman
- Department of Neonatology, Ankara City Hospital, Cankaya, Ankara, Turkey
| | - Veli Korkmaz
- Department of Pediatrics, Lokman Hekim University, Ankara, Turkey
| | - Filiz Akduman
- Department of Pediatrics, Beypazarı State Hospital, Ankara, Turkey
| | - Nurdan D Fettah
- Department of Neonatology, SBU Ankara Dr. Sami Ulus Maternity Child Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Başak K Gürsoy
- Department of Neonatology, SBU Ankara Dr. Sami Ulus Maternity Child Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Tugba T Turkmenoglu
- Department of Pathology, Ankara Diskapi Yildirim Beyzat Training and Research Hospital, Ankara, Turkey
| | - Murat Çağlayan
- Department of Medicinal Biochemistry, University of Health Sciences Gülhane Health Sciences Institute, Ankara, Turkey
| |
Collapse
|
45
|
Huang D, Wang P, Chen J, Li Y, Zhu M, Tang Y, Zhou W. Selective targeting of MD2 attenuates intestinal inflammation and prevents neonatal necrotizing enterocolitis by suppressing TLR4 signaling. Front Immunol 2022; 13:995791. [PMID: 36389716 PMCID: PMC9663461 DOI: 10.3389/fimmu.2022.995791] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/14/2022] [Indexed: 10/17/2023] Open
Abstract
Neonatal necrotizing enterocolitis (NEC) is an inflammatory disease that occurs in premature infants and has a high mortality rate; however, the mechanisms behind this disease remain unclear. The TLR4 signaling pathway in intestinal epithelial cells, mediated by TLR4, is important for the activation of the inflammatory storm in NEC infants. Myeloid differentiation protein 2 (MD2) is a key auxiliary component of the TLR4 signaling pathway. In this study, MD2 was found to be significantly increased in intestinal tissues of NEC patients at the acute stage. We further confirmed that MD2 was upregulated in NEC rats. MD2 inhibitor (MI) pretreatment reduced the occurrence and severity of NEC in neonatal rats, inhibited the activation of NF-κB and the release of inflammatory molecules (TNF-α and IL-6), and reduced the severity of intestinal injury. MI pretreatment significantly reduced enterocyte apoptosis while also maintaining tight junction proteins, including occludin and claudin-1, and protecting intestinal mucosal permeability in NEC rats. In addition, an NEC in vitro model was established by stimulating IEC-6 enterocytes with LPS. MD2 overexpression in IEC-6 enterocytes significantly activated NF-κB. Further, both MD2 silencing and MI pretreatment inhibited the inflammatory response. Overexpression of MD2 increased damage to the IEC-6 monolayer cell barrier, while both MD2 silencing and MI pretreatment played a protective role. In conclusion, MD2 triggers an inflammatory response through the TLR4 signaling pathway, leading to intestinal mucosal injury in NEC. In addition, MI alleviates inflammation and reduces intestinal mucosal injury caused by the inflammatory response by blocking the TLR4-MD2/NF-κB signaling axis. These results suggest that inhibiting MD2 may be an important way to prevent NEC.
Collapse
Affiliation(s)
- Dabin Huang
- Department of Neonatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
- Department of Pediatrics, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Ping Wang
- Department of Neonatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Juncao Chen
- Department of Neonatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yanbin Li
- Department of Neonatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Mingwei Zhu
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yaping Tang
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Wei Zhou
- Department of Neonatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
46
|
Han Y, Liu X, Kang L, Chen D, Li Y, Zhang H, Sun M, Gao H, Gai Z, Li X. A potential pathogenic hypoxia-related gene HK2 in necrotizing enterocolitis (NEC) of newborns. BMC Pediatr 2022; 22:617. [PMID: 36289463 PMCID: PMC9597967 DOI: 10.1186/s12887-022-03664-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 09/30/2022] [Indexed: 12/05/2022] Open
Abstract
Background Necrotizing enterocolitis (NEC) is a disastrous gastrointestinal disease of newborns, and the mortality rate of infants with NEC is approximately 20%-30%. The exploration of pathogenic targets of NEC will be conducive to timely diagnosis of NEC. Methods The whole transcriptome RNA sequencing was performed on NEC samples to reveal the expression of lncRNAs, circRNAs, miRNAs and mRNAs. Using differential expression analysis, cross analysis, target prediction, enrichment analysis, the pathogenic ceRNA network and target was found. Results Preliminarily, 281 DEmRNAs, 21 DEmiRNAs, 253 DElncRNAs and 207 DEcircRNAs were identified in NEC samples compared with controls. After target prediction and cross analyses, a key ceRNA regulatory network was built including 2 lncRNAs, 4 circRNAs, 2 miRNAs and 20 mRNAs. These 20 mRNAs were significantly enriched in many carbohydrate metabolism related pathways. After cross analysis of hypoxia-, carbohydrate metabolism-related genes, and 20 core genes, one gene HK2 was finally obtained. Dendritic cells activated were significantly differentially infiltrated and negatively correlated with HK2 expression in NEC samples. Conclusions The promising pathogenic hypoxia-related gene HK2 has been firstly identified in NEC, which might also involve in the carbohydrate metabolism in NEC. Supplementary Information The online version contains supplementary material available at 10.1186/s12887-022-03664-w.
Collapse
Affiliation(s)
- Yujie Han
- grid.27255.370000 0004 1761 1174Department of Neonatal, Children’s Hospital Affiliated to Shandong University/Jinan Children’s Hospital, No. 23976 Huaiyin District, Jinan, 250022 Shandong, People’s Republic of China
| | - Xianghong Liu
- grid.27255.370000 0004 1761 1174Department of Neonatal, Children’s Hospital Affiliated to Shandong University/Jinan Children’s Hospital, No. 23976 Huaiyin District, Jinan, 250022 Shandong, People’s Republic of China
| | - Lili Kang
- grid.27255.370000 0004 1761 1174Department of Neonatal, Children’s Hospital Affiliated to Shandong University/Jinan Children’s Hospital, No. 23976 Huaiyin District, Jinan, 250022 Shandong, People’s Republic of China
| | - Dong Chen
- grid.27255.370000 0004 1761 1174Department of Neonatal, Children’s Hospital Affiliated to Shandong University/Jinan Children’s Hospital, No. 23976 Huaiyin District, Jinan, 250022 Shandong, People’s Republic of China
| | - Yongqing Li
- Department of Neonatal, LaoLing Maternity and Child Health Care Hospital, 118 Anju Road, Laoling County, Dezhou, Shandong Province, 253600 People’s Republic of China
| | - Huiping Zhang
- grid.27255.370000 0004 1761 1174Department of Neonatal, Children’s Hospital Affiliated to Shandong University/Jinan Children’s Hospital, No. 23976 Huaiyin District, Jinan, 250022 Shandong, People’s Republic of China
| | - Mingying Sun
- grid.27255.370000 0004 1761 1174Department of Neonatal, Children’s Hospital Affiliated to Shandong University/Jinan Children’s Hospital, No. 23976 Huaiyin District, Jinan, 250022 Shandong, People’s Republic of China
| | - Hui Gao
- grid.27255.370000 0004 1761 1174Department of Neonatal, Children’s Hospital Affiliated to Shandong University/Jinan Children’s Hospital, No. 23976 Huaiyin District, Jinan, 250022 Shandong, People’s Republic of China
| | - Zhongtao Gai
- grid.27255.370000 0004 1761 1174Department of Neonatal, Children’s Hospital Affiliated to Shandong University/Jinan Children’s Hospital, No. 23976 Huaiyin District, Jinan, 250022 Shandong, People’s Republic of China
| | - Xiaoying Li
- grid.27255.370000 0004 1761 1174Department of Neonatal, Children’s Hospital Affiliated to Shandong University/Jinan Children’s Hospital, No. 23976 Huaiyin District, Jinan, 250022 Shandong, People’s Republic of China
| |
Collapse
|
47
|
Yan X, Cao Y, Chen W, Yu Q, Chen Y, Yao S, Jiang C, Chen X, Han S. Peptide Tat(48-60) YVEEL protects against necrotizing enterocolitis through inhibition of toll-like receptor 4-mediated signaling in a phosphatidylinositol 3-kinase/AKT dependent manner. Front Nutr 2022; 9:992145. [PMID: 36299988 PMCID: PMC9590307 DOI: 10.3389/fnut.2022.992145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a catastrophic disease largely occurring in preterm infants, and toll-like receptor 4 (TLR4) has been implicated in its pathogenesis. The current therapeutic strategies for NEC are, however, far from optimal. In the present study, a whey-derived antioxidative peptide conjugated with a cell-penetrating TAT [Tat (48-60) YVEEL] was prepared to endow it with enhanced cell uptake capability and bioavailability. The protective effect of Tat (48-60) YVEEL on experimental NEC was evaluated both in vitro and in vivo. Inhibition of TLR4-mediated signaling by Tat (48-60) YVEEL was assessed in FHC and IEC-6 enterocytes, neonatal rat model of NEC, and the mechanism underlying this effect was determined. Tat (48-60) YVEEL significantly inhibited TLR4-mediated expression of pro-inflammatory cytokines, p65 nuclear translocation and restored the impaired enterocyte migration in cultured enterocytes. In addition, Tat (48-60) YVEEL administration strikingly increased the survival rate, and reduced the severity of NEC in rats through inhibition of TLR4-mediated signaling. These protective effects of Tat (48-60) YVEEL occurred in a PI3K/AKT dependent manner, as administration of PI3K activator Ys49 abrogated its protective effects. Combined with liposomes, Tat (48-60) YVEEL demonstrated longer retention in the intestines that better for potential clinical applications. These data demonstrate that Tat (48-60) YVEEL protects against NEC through inhibition of TLR4-mediated signaling in a PI3K/AKT dependent manner, and offer a potential therapeutic approach to this disease.
Collapse
Affiliation(s)
- Xiangyun Yan
- Department of Pediatrics, Nanjing Maternity and Child Health Care Hospital, Women’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yan Cao
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Women’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wenjuan Chen
- Department of Pediatrics, Nanjing Maternity and Child Health Care Hospital, Women’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qinlei Yu
- Department of Pediatrics, Nanjing Maternity and Child Health Care Hospital, Women’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yanjie Chen
- Department of Pediatrics, Nanjing Maternity and Child Health Care Hospital, Women’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shuwen Yao
- Department of Pediatrics, Nanjing Maternity and Child Health Care Hospital, Women’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chengyao Jiang
- Department of Pediatrics, Nanjing Maternity and Child Health Care Hospital, Women’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaohui Chen
- Department of Pediatrics, Nanjing Maternity and Child Health Care Hospital, Women’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China,Xiaohui Chen,
| | - Shuping Han
- Department of Pediatrics, Nanjing Maternity and Child Health Care Hospital, Women’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China,*Correspondence: Shuping Han,
| |
Collapse
|
48
|
Zuiderwijk MO, van der Burg M, Bekker V, Schoenaker MHD. Regulatory T Cells in Development and Prediction of Necrotizing Enterocolitis in Preterm Neonates: A Scoping Review. Int J Mol Sci 2022; 23:10903. [PMID: 36142816 PMCID: PMC9504949 DOI: 10.3390/ijms231810903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/11/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a leading cause of mortality in premature infants. However, the pathophysiology and influence of regulatory T cells (Tregs) have not been sufficiently elucidated. We performed a scoping review to investigate current knowledge on the influence of Tregs in NEC, and to investigate the predictive value of Treg number in NEC development. Pubmed, Embase, Prospero and Cochrane Library were searched during December 2020. Primary research articles discussing Tregs and NEC development written in English were selected. Two reviewers screened title and abstract for relevance, after which full-text screening was performed. A total of 20 articles were selected-13 of the articles discussed studies performed in animal models, while 8 used human neonate data. One study discussed both animal and human data. It was shown that after NEC diagnosis or induction, Treg levels were decreased while Th17 levels were increased. No studies were found which investigated the predictive value of Treg number in NEC development. A reduced Treg level is found in animals and neonates with NEC. The question remains whether this effect is a factor on the causal pathway of NEC development or a bystander effect. Future research focusing on the pathophysiological timeline of NEC and the involvement of Tregs is required for better understanding of this disease.
Collapse
Affiliation(s)
- Mara O. Zuiderwijk
- Willem Alexander Children’s Hospital, Division of Neonatology, Department of Pediatrics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Mirjam van der Burg
- Willem Alexander Children’s Hospital, Laboratory for Pediatric Immunology, Department of Pediatrics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Vincent Bekker
- Willem Alexander Children’s Hospital, Division of Neonatology, Department of Pediatrics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Michiel H. D. Schoenaker
- Willem Alexander Children’s Hospital, Division of Neonatology, Department of Pediatrics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Willem Alexander Children’s Hospital, Laboratory for Pediatric Immunology, Department of Pediatrics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
49
|
Tian J, Yan C, Jiang Y, Zhou H, Li L, Shen J, Wang J, Sun H, Yang G, Sun W. Peripheral and intestinal mucosal-associated invariant T cells in premature infants with necrotizing enterocolitis. Front Pharmacol 2022; 13:1008080. [PMID: 36188574 PMCID: PMC9515899 DOI: 10.3389/fphar.2022.1008080] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 08/18/2022] [Indexed: 12/04/2022] Open
Abstract
Background: Necrotizing enterocolitis (NEC) is a potentially fatal inflammatory gastrointestinal disease in preterm infants with unknown pathogenesis. Mucosal-associated invariant T (MAIT) cells primarily accumulate at sites where exposure to microbes is ubiquitous and regulate immunological responses. As the implications of these cells in NEC development in premature infants remain unknown, we investigated the role and characteristics of MAIT cells in NEC pathogenesis. Methods: The percentage of different MAIT cell subsets in peripheral blood samples of 30 preterm infants with NEC and 22 control subjects was estimated using flow cytometry. The frequency of MAIT cells in the intestinal tissues of five NEC patients and five control subjects was also examined. The level of serum cytokines was estimated using cytometric bead array. Potential associations between the different measurements were analyzed using the Spearman’s correlation test. Results: Compared with controls, the NEC patients were found to have significantly reduced percentages of circulating CD161+ CD3+ CD8αα+ T cells and CD161+ CD3+ TCRγδ-TCRVa7.2+ MAIT cells. In the intestinal tissues, the percentage of MAIT cells was significantly higher in samples from the NEC patients than the controls. Furthermore, the percentage of circulating MAIT cells in the peripheral blood samples was inversely correlated with that in the intestinal tissues of the NEC patients. The percentage of CD8αα+ MAIT cells was found to be significantly reduced in both peripheral blood and intestinal tissues of NEC patients. Following treatment, the frequency of circulating MAIT cells significantly increased in NEC patients and reached a level similar to that in the control subjects. However, there was no difference in the percentage of circulating CD8αα+ MAIT cells before and after treatment in the NEC patients. Conclusion: Our results suggested that during the development of NEC MAIT cells accumulate in the inflammatory intestinal tissues, while the percentage of CD8aa+ MAIT cells is significantly decreased, which may lead to the dysfunction of MAIT cells in gut immunity.
Collapse
Affiliation(s)
- Jiayi Tian
- Center for Reproductive Medicine and Center for Prenatal Diagnosis, First Hospital, Jilin University, Changchun, China
| | - Chaoying Yan
- Department of Neonatology, The First Hospital of Jilin University, Changchun, China
| | - Yanfang Jiang
- Department of Center of Gene Diagnosis, The First Hospital of Jilin University, Changchun, China
| | - Haohan Zhou
- Department of Orthopaedic Oncology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Liyuan Li
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Jingjing Shen
- School of Civil Engineering and Architecture, Taizhou University, Taizhou, China
| | - Jian Wang
- Department of Neonatology, The First Hospital of Jilin University, Changchun, China
| | - Hongyu Sun
- Department of Orthopaedic Oncology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Guang Yang
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Wei Sun
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, China
- *Correspondence: Wei Sun,
| |
Collapse
|
50
|
DNA Methylome Mapping Identifies Epigenetic Abnormalities in Intestinal Lymphocyte Regulation in Human Necrotizing Enterocolitis. Dig Dis Sci 2022; 67:4434-4443. [PMID: 34846677 DOI: 10.1007/s10620-021-07314-6] [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: 04/13/2021] [Accepted: 11/02/2021] [Indexed: 12/09/2022]
Abstract
BACKGROUND Epigenetic changes occur in response to environmental factors during the pathogenesis of necrotizing enterocolitis (NEC) in animal models, but the DNA methylation signature in human patients with NEC has not been examined. AIM To illustrate the signature and function of DNA methylation in the intestine of human NEC. METHODS DNA methyltransferases (DNMTs) were compared between intestinal tissue with NEC and control. Genome-wide DNA methylation was analyzed by reduced representation bisulfite sequencing (RRBS). The biological functions of the potential methylation regulated genes were analyzed by Gene Ontology. Gene methylation and expression were confirmed by bisulfite genomic sequencing (BGS) and RT-qPCR. RESULTS By screening the expression of DNMTs, we identified a marked reduction in DNMT3A at both the mRNA and protein levels in NEC. Genome-wide variation of DNA methylation was detected in NEC lesions. The CG methylation level in almost all unique regions except CpG islands (CGIs) was lower in NEC compared with control. A total of 287 differentially methylated regions (DMRs) were identified across the whole genome in NEC, 123 of them are located on the CGI in the promoter. The DMR-associated genes were linked to intestinal epithelial permeability, platelet aggregation, and lymphocyte proliferation. Four genes (ZNF335, MPL, RASAL3, and KDM6A) with roles in the regulation of lymphocytes that may predispose the intestine to imbalanced immune processes were further confirmed to be hypermethylated and transcriptionally downregulated. CONCLUSIONS These findings underscore the novel relationship between epigenetic changes and lymphocyte regulation in human NEC, which may have potential diagnostic and therapeutic relevance for NEC.
Collapse
|