1
|
Zhang P, Li Y, Xu P, Zou P, Sheng S, Xiao R, Xu P, Chen Y, Du Y, Ma L, Wang Y. Identification and Exploration of Pyroptosis-Related Genes in Macrophage Cells Reveal Necrotizing Enterocolitis Heterogeneity Through Single-Cell and Bulk-Sequencing. Int J Mol Sci 2025; 26:4036. [PMID: 40362275 PMCID: PMC12071306 DOI: 10.3390/ijms26094036] [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: 02/16/2025] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
Necrotizing enterocolitis (NEC) is an acute intestine dysfunction intestinal disorder characterized by inflammation and cell death, including pyroptosis. Previous studies have implicated pyroptosis, particularly via NLRP3 inflammatory activation, and contribute to the development of NEC. However, the genetic and molecular mechanisms underlying pyroptosis in NEC pathogenesis and sequelae remain unclear. Our study aimed to identify the pyroptosis-related cell populations and genes and explore potential therapeutic targets. Single-cell RNA sequencing (scRNA-seq) data were analyzed to identify the cell populations related to NEC and pyroptosis. Weighted gene correlation network analysis (WGCNA) of bulk RNA-seq was performed to identify gene modules associate with pyroptosis. Cell-cell communication was employed to investigate intercellular signaling networks. Gene Set Enrichment Analysis (GSEA) was conducted to compare the pathways enriched in the high and low TREM1-expressing subgroups. Immunofluorescence staining was performed to detect the TREM1+CD163+ macrophages in the intestines. PCR and Western blot were performed to detect the expression of mRNA and proteins in the intestine tissues and cells. scRNA-seq analysis revealed increased macrophage abundance in NEC, with one macrophage cluster (cluster 4) exhibiting a markedly elevated pyroptosis score. WGCNA identified a gene module (MEbrown) that positively correlated with pyroptosis. Five genes (TREM1, TLN1, NOTCH2, MPZL1, and ADA) within this module were identified as potential diagnostic markers of pyroptosis. Furthermore, we identified a novel macrophage subpopulation, TREM1+CD163+, in NEC. Cell-cell communication analysis suggested that TREM1+CD163+ macrophages interact with other cells primarily through the NAMPT/ITGA5/ITGB1 and CCL3/CCR1 pathways. GSEA revealed a significant association between high TREM1 expression and pathways related to pyroptosis, cell proliferation, and inflammation. In vivo and in vitro experiments confirmed an increase in TREM1+CD163+ macrophages in NEC-affected intestines. TREM1 inhibition in THP-1 cells significantly reduced the expression of pro-inflammatory cytokines and pyroptosis-related genes and proteins. We identified the TREM1+CD163+ macrophage population that plays a crucial role in pyroptosis during NEC progression. Our findings elucidate the biological functions and molecular mechanisms of TREM1, demonstrating its upregulation in vivo and pro-pyroptosis effects in vitro. These insights advance our understanding of the role of pyroptosis in NEC pathogenesis and suggest TREM1 is a potential therapeutic target for NEC.
Collapse
Affiliation(s)
- Peipei Zhang
- Capital Institute of Pediatrics, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; (P.Z.); (P.X.); (P.Z.); (S.S.); (P.X.)
| | - Ying Li
- Department of Neonatology, Capital Center for Children’s Health, Capital Medical University, No. 2 Yabao Road, Chaoyang District, Beijing 100020, China; (Y.L.); (Y.C.); (Y.D.)
| | - Panpan Xu
- Capital Institute of Pediatrics, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; (P.Z.); (P.X.); (P.Z.); (S.S.); (P.X.)
| | - Peicen Zou
- Capital Institute of Pediatrics, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; (P.Z.); (P.X.); (P.Z.); (S.S.); (P.X.)
| | - Sihan Sheng
- Capital Institute of Pediatrics, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; (P.Z.); (P.X.); (P.Z.); (S.S.); (P.X.)
| | - Ruiqi Xiao
- Capital Institute of Pediatrics, Beijing 100020, China;
| | - Pu Xu
- Capital Institute of Pediatrics, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; (P.Z.); (P.X.); (P.Z.); (S.S.); (P.X.)
| | - Ying Chen
- Department of Neonatology, Capital Center for Children’s Health, Capital Medical University, No. 2 Yabao Road, Chaoyang District, Beijing 100020, China; (Y.L.); (Y.C.); (Y.D.)
| | - Yue Du
- Department of Neonatology, Capital Center for Children’s Health, Capital Medical University, No. 2 Yabao Road, Chaoyang District, Beijing 100020, China; (Y.L.); (Y.C.); (Y.D.)
| | - Lishuang Ma
- Department of Neonatology Surgery, Capital Center for Children’s Health, Capital Medical University, No. 2 Yabao Road, Chaoyang District, Beijing 100020, China
| | - Yajuan Wang
- Department of Neonatology, Capital Center for Children’s Health, Capital Medical University, No. 2 Yabao Road, Chaoyang District, Beijing 100020, China; (Y.L.); (Y.C.); (Y.D.)
| |
Collapse
|
2
|
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] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [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
|
3
|
Jin X, Sun W, Li Y, Zhu X. Mitophagy and immune cell interaction: insights into pathogenesis and potential targets for necrotizing enterocolitis. Transl Pediatr 2025; 14:171-186. [PMID: 40115463 PMCID: PMC11921342 DOI: 10.21037/tp-24-441] [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: 10/17/2024] [Accepted: 01/21/2025] [Indexed: 03/23/2025] Open
Abstract
Background Neonatal necrotizing enterocolitis (NEC) is a fatal disease in early life characterized by an inflammatory response or even necrosis of the bowel wall. NEC is one of the leading causes of preterm infant mortality. The pathogenesis of NEC is intricate and involves mitochondrial damage to intestinal cells and infiltration of immune cells. However, the specific functions of mitophagy and its association with immune cells in NEC remain unclear. The aim of this study was to explore the pivotal roles of mitophagy and the immune microenvironment in NEC and their potential interactions. Methods Microarray data (GSE46619) associated with NEC were obtained from the Gene Expression Omnibus (GEO) at the National Center for Biotechnology Information (NCBI). Differentially expressed genes (DEGs) were screened by GEO2R. Mitophagy gene data were downloaded from the Pathway Unification database and subjected to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. Consequently, mitophagy-related differentially expressed genes (MRDEGs) were obtained. To identify hub MRDEGs that are closely associated with NEC, we used CytoHubba, Molecular Complex Detection (MCODE) and Comparative Toxicogenomics Database (CTD) scores. Cytoscape and miRWalk databases were used to predict the transcription factors (TFs) and target microRNAs (miRNAs) of hub MRDEGs, respectively, and a regulatory network was established. The ImmuCellAI was used to analyze the pattern of immune infiltration, and the Spearman correlation was used to investigate the relationship between the hub MRDEGs and the abundance of infiltrating immune cells. Finally, the expression levels of the hub MRDEGs were verified by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting in NEC animal model. Results A total of 14 up-regulated and 22 down-regulated MRDEGs were identified, these genes exhibited enrichment in mitophagy, and inflammation-related pathways. Furthermore, 13 hub MRDEGs closely related to NEC were identified. The increased presence of immune cells such as neutrophils, M1 macrophages, and activated mast cells were observed while adaptive immune cells including B cells and various T-cell subsets exhibited reduced infiltration. Furthermore, up-regulated MRDEGs were positively correlated with the proinflammatory immune cell infiltration, and the down-regulated MRDEGs were positively correlated with the anti-inflammatory immune cell infiltration. In vivo experiments demonstrated that the expressions of four genes Hif-1a, Acsl4, Pck2, and Aifm1 were consistent with the bioinformatics analysis results. Conclusions The potential interplay of mitophagy and immune cells is crucial in the onset and progression of NEC. This perspective opens the door for deeper investigations into NEC pathogenesis, presenting a possible target for disease intervention.
Collapse
Affiliation(s)
- Xinyun Jin
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, China
- Clinical Pediatrics School, Soochow University, Suzhou, China
| | - Wenqiang Sun
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, China
- Clinical Pediatrics School, Soochow University, Suzhou, China
| | - Yihui Li
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, China
- Clinical Pediatrics School, Soochow University, Suzhou, China
| | - Xueping Zhu
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, China
| |
Collapse
|
4
|
Tian B, Xu X, Li L, Tian Y, Liu Y, Mu Y, Lu J, Song K, Lv J, He Q, Zhong W, Xia H, Lan C. Epigenetic Insights Into Necrotizing Enterocolitis: Unraveling Methylation-Regulated Biomarkers. Inflammation 2025; 48:236-253. [PMID: 38814387 PMCID: PMC11807086 DOI: 10.1007/s10753-024-02054-x] [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: 05/11/2024] [Accepted: 05/14/2024] [Indexed: 05/31/2024]
Abstract
Necrotizing enterocolitis (NEC) is a multifactorial gastrointestinal disease with high morbidity and mortality among premature infants. This study aimed to identify novel methylation-regulated biomarkers in NEC intestinal tissue through multiomics analysis. We analyzed DNA methylation and transcriptome datasets from ileum and colon tissues of patients with NEC. We identify methylation-related differential genes (MrDEGs) based on the rule that the degree of methylation in the promoter region is inversely proportional to RNA transcription. These MrDEGs included ADAP1, GUCA2A, BCL2L14, FUT3, MISP, USH1C, ITGA3, UNC93A and IL22RA1. Single-cell data revealed that MrDEGs were mainly located in the intestinal epithelial part of intestinal tissue. These MrDEGs were verified through Target gene bisulfite sequencing and RT-qPCR. We successfully identified and verified the ADAP1, GUCA2A, IL22RA1 and MISP, primarily expressed in intestinal epithelial villus cells through single-cell data. Through single-gene gene set enrichment analysis, we found that these genes participate mainly in the pathological process of T-cell differentiation and the suppression of intestinal inflammation in NEC. This study enhances our understanding of the pathogenesis of NEC and may promote the development of new precision medicine methods for NEC prediction and diagnosis.
Collapse
Affiliation(s)
- Bowen Tian
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaogang Xu
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, China
| | - Lin Li
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, China
| | - Yan Tian
- Department of Anesthesiology, Jiangxi Provincial Children's Hospital, Nanchang, Jiangxi, China
| | - Yanqing Liu
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, China
| | - Yide Mu
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, China
| | - Jieting Lu
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Kai Song
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, China
| | - Junjian Lv
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, China
| | - Qiuming He
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, China
| | - Wei Zhong
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, China.
| | - Huimin Xia
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China.
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, China.
| | - Chaoting Lan
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, China.
| |
Collapse
|
5
|
Kollmann C, Niklas C, Ernestus K, Hiller GG, Hörner M, Burkard N, Germer CT, Härtel C, Meyer T, Bartfeld S, Glaser K, Schlegel N. The phenotype of necrotizing enterocolitis correlates with distinct changes of intestinal junctional proteins. Pathol Res Pract 2025; 265:155731. [PMID: 39579526 DOI: 10.1016/j.prp.2024.155731] [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: 09/14/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 11/25/2024]
Abstract
Necrotizing enterocolitis (NEC) is a major cause of mortality in preterm infants. Its pathophysiology remains poorly understood but intestinal epithelial barrier dysfunction contributes to the disease. We characterized junctional proteins in intestinal specimens from preterm infants. Samples from 27 patients with NEC and 20 patients with focal intestinal perforation (FIP) from the center of the specimens (affected) or the macroscopically healthy resection margins whenever available (non-affected) were collected. NEC patients displayed higher mortality and more commonly occurrence of impaired glucose homeostasis, patent ductus arteriosus, anemia and antibiotic treatment compared to FIP patients. Discrimination between NEC and FIP was not possible in affected areas based on H.E. staining using a newly developed scoring system. Immunofluorescence revealed reduced Claudin-3 in affected NEC samples and decreased Claudin-4 in affected FIP and all NEC samples. E-cadherin and Desmoglein-2 were reduced in a subgroup of the affected NEC samples. Plakophilin-2 was decreased in intestine affected by FIP and unaffected intestine in patients with NEC. In affected areas of NEC, Plakophilin-2 was completely lost. Plakoglobin reduction in affected NEC samples correlated with poor survival. This study provides novel insights into changes of junctional proteins in NEC, suggesting Claudin-3 and Plakophilin-2 as diagnostic markers to differentiate FIP from NEC and reduced Plakoglobin as a prognostic marker.
Collapse
Affiliation(s)
- Catherine Kollmann
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, Oberdürrbacher Straße 6, Würzburg 97080, Germany
| | - Carolin Niklas
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, Oberdürrbacher Straße 6, Würzburg 97080, Germany
| | - Karen Ernestus
- Institute of Pathology, Julius-Maximilians-University of Würzburg, Josef-Schneider-Straße 2, Würzburg 97080, Germany
| | - Grit Gr Hiller
- Institute of Pathology, University of Leipzig, Liebigstraße 26, Leipzig 04103, Germany
| | - Marius Hörner
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, Oberdürrbacher Straße 6, Würzburg 97080, Germany
| | - Natalie Burkard
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, Oberdürrbacher Straße 6, Würzburg 97080, Germany
| | - Christoph-Thomas Germer
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, Oberdürrbacher Straße 6, Würzburg 97080, Germany
| | - Christoph Härtel
- Department of Pediatrics, University Hospital Würzburg, Josef-Schneider-Straße 2, Würzburg 97080, Germany
| | - Thomas Meyer
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, Oberdürrbacher Straße 6, Würzburg 97080, Germany
| | - Sina Bartfeld
- Institute of Biotechnology, Technical University Berlin, Straße des 17. Juni 135, Berlin 10623, Germany
| | - Kirsten Glaser
- Division of Neonatology, Department of Women's and Children's Health, University of Leipzig Medical Center, Liebigstraße 20A, Leipzig 04103, Germany
| | - Nicolas Schlegel
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, Oberdürrbacher Straße 6, Würzburg 97080, Germany.
| |
Collapse
|
6
|
Yuan F, Han X, Huang M, Su Y, Zhang Y, Hu M, Yu X, Jin W, Li Y, Zhang L. The Human Milk-derived Peptide Drives Rapid Regulation of Macrophage Inflammation Responses in the Neonatal Intestine. Cell Mol Gastroenterol Hepatol 2024; 19:101420. [PMID: 39414025 PMCID: PMC11652890 DOI: 10.1016/j.jcmgh.2024.101420] [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: 01/14/2024] [Revised: 09/25/2024] [Accepted: 10/03/2024] [Indexed: 10/18/2024]
Abstract
BACKGROUND & AIMS The interactions between human milk and the regulation of innate immune homeostasis in newborns, and their impact on intestinal health, are not fully understood. This study aimed to explore the role of peptides in human milk extracellular vesicles (EVs) in this process. METHODS A comprehensive screening of peptides within human milk EVs was performed, leading to the identification of a beta-casein-derived peptide (CASB135-150). The effects of CASB135-150 on intestinal injury were evaluated in a rat necrotizing enterocolitis (NEC) model. Immunofluorescence analysis was used to determine its distribution, and its impact on NF-κB signaling and inflammation was studied in bone marrow-derived macrophages (BMDMs) and intestinal macrophages. Protein-protein interaction (PPI) analysis, single-cell RNA-seq (scRNA-seq), and co-immunoprecipitation (co-IP) experiments were conducted to explore the mechanism underlying CASB135-150 function. RESULTS CASB135-150 significantly mitigated intestinal injury in the rat NEC model. Immunofluorescence analysis revealed that CASB135-150 could target intestinal macrophages and rapidly inhibited NF-κB signaling and reduced inflammation. ScRNA-seq analyses indicated a strong association between FHL2 and NEC development, and co-IP confirmed the interaction between CASB135-150 and FHL2. CASB135-150 disrupted the FHL2/TRAF6 complex, reducing TRAF6 protein levels. Mutation of key amino acids in CASB135-150 disrupted its interaction with FHL2 and abolished its ability to inhibit NF-κB signaling, which also prevented its protective effect in vivo. RNA-seq of intestinal tissue further highlighted the impact of CASB135-150 on the NF-κB signaling pathway. CONCLUSIONS Our study identifies CASB135-150, a novel peptide in human milk EVs, that rapidly regulates macrophage inflammatory responses and protects against NEC-induced intestinal injury. These findings provide new insights into the role of human milk in modulating the infant immune system and intestinal health.
Collapse
Affiliation(s)
- Fuqiang Yuan
- Department of Neonatology, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, China; Department of Pediatric Laboratory, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, China
| | - Xu Han
- Department of Neonatology, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, China
| | - Masha Huang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yinglin Su
- Department of Neonatology, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, China
| | - Yiting Zhang
- Department of Neonatology, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, China
| | - Mengyuan Hu
- Department of Neonatology, Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, China
| | - Xiang Yu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weilai Jin
- Department of Neonatology, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, China.
| | - Yun Li
- Department of Pediatric Laboratory, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, China.
| | - Le Zhang
- Department of Neonatology, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, China; Department of Pediatric Laboratory, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, China.
| |
Collapse
|
7
|
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
|
8
|
Gu W, Eke C, Gonzalez Santiago E, Olaloye O, Konnikova L. Single-cell atlas of the small intestine throughout the human lifespan demonstrates unique features of fetal immune cells. Mucosal Immunol 2024; 17:599-617. [PMID: 38555026 PMCID: PMC11384551 DOI: 10.1016/j.mucimm.2024.03.011] [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: 11/02/2023] [Revised: 03/15/2024] [Accepted: 03/23/2024] [Indexed: 04/02/2024]
Abstract
Proper development of mucosal immunity is critical for human health. Over the past decade, it has become evident that in humans, this process begins in utero. However, there are limited data on the unique features and functions of fetal mucosal immune cells. To address this gap, we integrated several single-cell ribonucleic acid sequencing datasets of the human small intestine (SI) to create an SI transcriptional atlas throughout the human life span, ranging from the first trimester to adulthood, with a focus on immune cells. Fetal SI displayed a complex immune landscape comprising innate and adaptive immune cells that exhibited distinct transcriptional programs from postnatal samples, especially compared with pediatric and adult samples. We identified shifts in myeloid populations across gestation and progression of memory T-cell states throughout the human lifespan. In particular, there was a marked shift of memory T cells from those with stem-like properties in the fetal samples to fully differentiated cells with a high expression of activation and effector function genes in adult samples, with neonatal samples containing both features. Finally, we demonstrate that the SI developmental atlas can be used to elucidate improper trajectories linked to mucosal diseases by implicating developmental abnormalities underlying necrotizing enterocolitis, a severe intestinal complication of prematurity. Collectively, our data provide valuable resources and important insights into intestinal immunity that will facilitate regenerative medicine and disease understanding.
Collapse
Affiliation(s)
- Weihong Gu
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Chino Eke
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | | | - Oluwabunmi Olaloye
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Liza Konnikova
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA; Department of Obstetrics, Gynecology and Reproductive Science, Yale University School of Medicine, New Haven, CT, USA; Program in Translational Biomedicine, Yale University School of Medicine, New Haven, CT, USA; Program in Human Translational Immunology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
9
|
Harnik Y, Yakubovsky O, Hoefflin R, Novoselsky R, Bahar Halpern K, Barkai T, Korem Kohanim Y, Egozi A, Golani O, Addadi Y, Kedmi M, Keidar Haran T, Levin Y, Savidor A, Keren-Shaul H, Mayer C, Pencovich N, Pery R, Shouval DS, Tirosh I, Nachmany I, Itzkovitz S. A spatial expression atlas of the adult human proximal small intestine. Nature 2024; 632:1101-1109. [PMID: 39112711 DOI: 10.1038/s41586-024-07793-3] [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: 10/11/2023] [Accepted: 07/05/2024] [Indexed: 08/17/2024]
Abstract
The mouse small intestine shows profound variability in gene expression along the crypt-villus axis1,2. Whether similar spatial heterogeneity exists in the adult human gut remains unclear. Here we use spatial transcriptomics, spatial proteomics and single-molecule fluorescence in situ hybridization to reconstruct a comprehensive spatial expression atlas of the adult human proximal small intestine. We describe zonated expression and cell type representation for epithelial, mesenchymal and immune cell types. We find that migrating enterocytes switch from lipid droplet assembly and iron uptake at the villus bottom to chylomicron biosynthesis and iron release at the tip. Villus tip cells are pro-immunogenic, recruiting γδ T cells and macrophages to the tip, in contrast to their immunosuppressive roles in mouse. We also show that the human small intestine contains abundant serrated and branched villi that are enriched at the tops of circular folds. Our study presents a detailed resource for understanding the biology of the adult human small intestine.
Collapse
Affiliation(s)
- Yotam Harnik
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Oran Yakubovsky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
- Department of General Surgery and Transplantation, Sheba Medical Center, Ramat Gan, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Rouven Hoefflin
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Roy Novoselsky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Keren Bahar Halpern
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Tal Barkai
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
- Sheba Medical Center, Ramat Gan, Israel
| | - Yael Korem Kohanim
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Adi Egozi
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ofra Golani
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Yoseph Addadi
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Merav Kedmi
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Tal Keidar Haran
- Department of Pathology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Yishai Levin
- The De Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Alon Savidor
- The De Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Hadas Keren-Shaul
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Chen Mayer
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Institute of Pathology, Sheba Medical Center, Ramat Gan, Israel
| | - Niv Pencovich
- Department of General Surgery and Transplantation, Sheba Medical Center, Ramat Gan, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ron Pery
- Department of General Surgery and Transplantation, Sheba Medical Center, Ramat Gan, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dror S Shouval
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Itay Tirosh
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ido Nachmany
- Department of General Surgery and Transplantation, Sheba Medical Center, Ramat Gan, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shalev Itzkovitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
10
|
Smith L, Santiago EG, Eke C, Gu W, Wang W, Llivichuzhca-Loja D, Kehoe T, St Denis K, Strine M, Taylor S, Tseng G, Konnikova L. Human Milk Supports Robust Intestinal Organoid Growth, Differentiation, and Homeostatic Cytokine Production. GASTRO HEP ADVANCES 2024; 3:1030-1042. [PMID: 39529649 PMCID: PMC11550179 DOI: 10.1016/j.gastha.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 07/11/2024] [Indexed: 11/16/2024]
Abstract
Background and Aims Necrotizing enterocolitis is a severe gastrointestinal complication of prematurity. Using small intestinal organoids derived from fetal tissue of a gestational age similar to an extremely preterm infant, this study aims to assess the effect of diet on intestinal epithelial growth and differentiation to elucidate the role nutrition type plays in intestinal development and modifies the risk for necrotizing enterocolitis. Methods Organoids were cultured for 5 days in growth media and 5 days in differentiation media supplemented 1:40 with 4 different diets: parental milk, donor human milk, standard formula, or extensively hydrolyzed formula. Images were captured daily and organoids were quantified. Organoids were preserved for RNA sequencing and immunofluorescence staining with Ki67, cleaved caspase 3, and chromogranin-A. Media was saved for cytokine/chemokine and growth factor analysis. Results Human milk supplementation improved growth and differentiation of intestinal organoids generating larger organoids during the growth phase and organoids with longer and wider buds during differentiation compared to formula. Ki67 staining confirmed the proliferative nature of milk-supplemented organoids and chromogranin A staining proved that MM-supplemented organoids induced highest enteroendocrine differentiation. Human milk supplementation also upregulated genes involved in Wnt signaling and fatty acid metabolism pathways and promoted a homeostatic immune landscape, including via increased secretion of tumor necrosis factor-related apoptosis-inducing ligand among other cytokines. Conversely, organoids supplemented with formula had a downregulation of cell-cycle-promoting genes and a more inflammatory immune signature, including a reduced level of leukemia inhibitory factor. Conclusion Our results demonstrate that parental milk, and to a lesser extent donor human milk, support robust intestinal epithelial proliferation, differentiation, and homeostatic cytokine production, suggesting a critical role for factors enriched in human milk in intestinal epithelial health.
Collapse
Affiliation(s)
- Lauren Smith
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
| | | | - Chino Eke
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
| | - Weihong Gu
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
| | - Wenjia Wang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Tessa Kehoe
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
| | - Kerri St Denis
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
| | - Madison Strine
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
| | - Sarah Taylor
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
| | - George Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Liza Konnikova
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut
- Program in Human and Translational Immunology, Yale School of Medicine, New Haven, Connecticut
- Program in Translational Biomedicine, Yale School of Medicine, New Haven, Connecticut
- Center for Systems and Engineering Immunology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
11
|
Chen J, Yan Z, Lin Z, Fan Y, Bao X, Chen X, Zheng A. I-FABP protein/mRNA and IL-6 as biomarkers of intestinal barrier dysfunction in neonates with necrotizing enterocolitis and SPF BALB/c mouse models. J Int Med Res 2024; 52:3000605241254788. [PMID: 38867509 PMCID: PMC11179468 DOI: 10.1177/03000605241254788] [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: 01/12/2024] [Accepted: 04/26/2024] [Indexed: 06/14/2024] Open
Abstract
OBJECTIVE Neonatal necrotizing enterocolitis (NEC) is a serious intestinal inflammatory disease. We investigated intestinal fatty acid binding protein (I-FABP), I-FABP mRNA, and interleukin-6 (IL-6) as potential diagnostic biomarkers in NEC. METHODS Forty mice were subjected to hypoxic-ischemic intestinal injury, and then serum I-FABP protein and mRNA levels were quantified. Ileal tissue pathological scores were determined by hematoxylin and eosin staining. I-FABP expression levels and translocation in these tissues were detected using western blotting and immunofluorescence, respectively. Samples from 30 human neonates with NEC and 30 healthy neonates had serum I-FABP protein/mRNA and IL-6 levels measured. RESULTS The mouse ileal tissue pathological score and I-FABP levels, as well as serum I-FABP and I-FABP mRNA levels, were significantly higher in the model group than in the control group. Serum I-FABP, I-FABP mRNA, and IL-6 levels were significantly higher in human neonates with NEC than in the healthy group. Logistic regression and receiver operating curve analyses revealed that I-FABP protein/mRNA and IL-6 levels could be diagnostic biomarkers for NEC. CONCLUSIONS I-FABP protein/mRNA and IL-6 levels are useful biomarkers of intestinal ischemic injury in neonates with NEC. The combined detection of I-FABP protein/mRNA and IL-6 is recommended rather than using a single biomarker.
Collapse
MESH Headings
- Enterocolitis, Necrotizing/metabolism
- Enterocolitis, Necrotizing/blood
- Enterocolitis, Necrotizing/pathology
- Enterocolitis, Necrotizing/genetics
- Enterocolitis, Necrotizing/diagnosis
- Animals
- Fatty Acid-Binding Proteins/blood
- Fatty Acid-Binding Proteins/genetics
- Fatty Acid-Binding Proteins/metabolism
- Interleukin-6/blood
- Interleukin-6/genetics
- Infant, Newborn
- Humans
- Biomarkers/blood
- Biomarkers/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Messenger/blood
- Disease Models, Animal
- Mice
- Male
- Female
- Mice, Inbred BALB C
- Animals, Newborn
- Intestinal Mucosa/metabolism
- Intestinal Mucosa/pathology
- Ileum/metabolism
- Ileum/pathology
- Case-Control Studies
- ROC Curve
Collapse
Affiliation(s)
- Jun Chen
- Department of Paediatrics, Fuzhou First General Hospital Affiliated with Fujian Medical University, No. 190 Dadao Road, Fuzhou, Fujian Province, China
| | - Zheng Yan
- Department of Paediatrics, Fuzhou First General Hospital Affiliated with Fujian Medical University, No. 190 Dadao Road, Fuzhou, Fujian Province, China
| | - Zhibing Lin
- Department of Clinical Laboratory, Fuzhou First General Hospital Affiliated with Fujian Medical University, No. 190 Dadao Road, Fuzhou, Fujian Province, China
| | - Yong Fan
- Department of Clinical Laboratory, Fuzhou First General Hospital Affiliated with Fujian Medical University, No. 190 Dadao Road, Fuzhou, Fujian Province, China
| | - Xuan Bao
- Department of Paediatrics, Fuzhou First General Hospital Affiliated with Fujian Medical University, No. 190 Dadao Road, Fuzhou, Fujian Province, China
| | - Xiaolin Chen
- Department of Paediatrics, Fuzhou First General Hospital Affiliated with Fujian Medical University, No. 190 Dadao Road, Fuzhou, Fujian Province, China
| | - Airu Zheng
- Department of Paediatrics, Fuzhou First General Hospital Affiliated with Fujian Medical University, No. 190 Dadao Road, Fuzhou, Fujian Province, China
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Liu Y, Zhou J, Chen B, Liu X, Cai Y, Liu W, Hao H, Li S. High-dimensional mass cytometry reveals systemic and local immune signatures in necrotizing enterocolitis. Front Immunol 2023; 14:1292987. [PMID: 38045686 PMCID: PMC10690805 DOI: 10.3389/fimmu.2023.1292987] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 10/30/2023] [Indexed: 12/05/2023] Open
Abstract
Objective Patients with necrotizing enterocolitis display severe gastrointestinal complications of prematurity, but the mechanism driving this clinical profile remains unknown. We used mass cytometry time-of-flight to characterize and compare immune cell populations in the blood and intestine tissue from patients with and without (controls) necrotizing enterocolitis at single-cell resolution. Methods We completed a deep mapping of the immune system of the peripheral blood mononuclear cells and intestinal mucosa tissue using mass cytometry to evaluate immune cell types, which revealed global immune dysregulation characteristics underlying necrotizing enterocolitis. Results Compared with controls, natural killer cells display signs of heightened activation and increased cytotoxic potential in the peripheral blood and mucosa of patients with necrotizing enterocolitis. Furthermore, CD4+ T effector memory cells, non-classical monocytes, active dendritic cells, and neutrophils were specifically enriched in the mucosa, suggesting trafficking from the periphery to areas of inflammation. Moreover, we mapped the systemic and local distinct immune signatures suggesting patterns of cell localization in necrotizing enterocolitis. Conclusion We used mass cytometry time-of-flight technology to identify immune cell populations specific to the peripheral blood and intestinal mucosa tissue from patients with necrotizing enterocolitis and controls. This information might be used to develop precise diagnosis and therapies that target specific cell populations in patients with necrotizing enterocolitis.
Collapse
Affiliation(s)
- Yufeng Liu
- Center for Medical Research on Innovation and Translation, Guangzhou First People's Hospital, Guangzhou, China
| | - Jialiang Zhou
- Department of Neonatal Surgery, Guangdong Women and Children Hospital, Guangzhou, China
| | - Baozhu Chen
- 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
| | - Xiao Liu
- 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
| | - Yao Cai
- 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
| | - Wei Liu
- 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
| | - 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
| | - Sitao 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
| |
Collapse
|
15
|
Schill EM, Joyce EL, Floyd AN, Udayan S, Rusconi B, Gaddipati S, Barrios BE, John V, Kaye ME, Kulkarni DH, Pauta JT, McDonald KG, Newberry RD. Vancomycin-induced gut microbial dysbiosis alters enteric neuron-macrophage interactions during a critical period of postnatal development. Front Immunol 2023; 14:1268909. [PMID: 37901245 PMCID: PMC10602895 DOI: 10.3389/fimmu.2023.1268909] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 09/20/2023] [Indexed: 10/31/2023] Open
Abstract
Vancomycin is a broad-spectrum antibiotic widely used in cases of suspected sepsis in premature neonates. While appropriate and potentially lifesaving in this setting, early-life antibiotic exposure alters the developing microbiome and is associated with an increased risk of deadly complications, including late-onset sepsis (LOS) and necrotizing enterocolitis (NEC). Recent studies show that neonatal vancomycin treatment disrupts postnatal enteric nervous system (ENS) development in mouse pups, which is in part dependent upon neuroimmune interactions. This suggests that early-life antibiotic exposure could disrupt these interactions in the neonatal gut. Notably, a subset of tissue-resident intestinal macrophages, muscularis macrophages, has been identified as important contributors to the development of postnatal ENS. We hypothesized that vancomycin-induced neonatal dysbiosis impacts postnatal ENS development through its effects on macrophages. Using a mouse model, we found that exposure to vancomycin in the first 10 days of life, but not in adult mice, resulted in an expansion of pro-inflammatory colonic macrophages by increasing the recruitment of bone-marrow-derived macrophages. Single-cell RNA sequencing of neonatal colonic macrophages revealed that early-life vancomycin exposure was associated with an increase in immature and inflammatory macrophages, consistent with an influx of circulating monocytes differentiating into macrophages. Lineage tracing confirmed that vancomycin significantly increased the non-yolk-sac-derived macrophage population. Consistent with these results, early-life vancomycin exposure did not expand the colonic macrophage population nor decrease enteric neuron density in CCR2-deficient mice. Collectively, these findings demonstrate that early-life vancomycin exposure alters macrophage number and phenotypes in distinct ways compared with vancomycin exposure in adult mice and results in altered ENS development.
Collapse
Affiliation(s)
- Ellen Merrick Schill
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Elisabeth L. Joyce
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Alexandria N. Floyd
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Sreeram Udayan
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Brigida Rusconi
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Shreya Gaddipati
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Bibiana E. Barrios
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Vini John
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Mitchell E. Kaye
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Devesha H. Kulkarni
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Jocelyn T. Pauta
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Keely G. McDonald
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Rodney D. Newberry
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|