1
|
Zhao Y, Liu MJ, Zhang L, Yang Q, Sun QH, Guo JR, Lei XY, He KY, Li JQ, Yang JY, Jian YP, Xu ZX. High mobility group A1 (HMGA1) promotes the tumorigenesis of colorectal cancer by increasing lipid synthesis. Nat Commun 2024; 15:9909. [PMID: 39548107 DOI: 10.1038/s41467-024-54400-0] [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: 11/05/2023] [Accepted: 11/07/2024] [Indexed: 11/17/2024] Open
Abstract
Metabolic reprogramming is a hallmark of cancer, enabling tumor cells to meet the high energy and biosynthetic demands required for their proliferation. High mobility group A1 (HMGA1) is a structural transcription factor and frequently overexpressed in human colorectal cancer (CRC). Here, we show that HMGA1 promotes CRC progression by driving lipid synthesis in a AOM/DSS-induced CRC mouse model. Using conditional knockout (Hmga1△IEC) and knock-in (Hmga1IEC-OE/+) mouse models, we demonstrate that HMGA1 enhances CRC cell proliferation and accelerates tumor development by upregulating fatty acid synthase (FASN). Mechanistically, HMGA1 increases the transcriptional activity of sterol regulatory element-binding protein 1 (SREBP1) on the FASN promoter, leading to increased lipid accumulation in intestinal epithelial cells. Moreover, a high-fat diet exacerbates CRC progression in Hmga1△IEC mice, while pharmacological inhibition of FASN by orlistat reduces tumor growth in Hmga1IEC-OE/+ mice. Our findings suggest that targeting lipid metabolism could offer a promising therapeutic strategy for CRC.
Collapse
Affiliation(s)
- Yuan Zhao
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Meng-Jie Liu
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Lei Zhang
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Qi Yang
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Qian-Hui Sun
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Jin-Rong Guo
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Xin-Yuan Lei
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Kai-Yue He
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Jun-Qi Li
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Jing-Yu Yang
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Yong-Ping Jian
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China.
| | - Zhi-Xiang Xu
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China.
| |
Collapse
|
2
|
Zhang R, Perekatt A, Chen L. Metabolic regulation of intestinal homeostasis: molecular and cellular mechanisms and diseases. MedComm (Beijing) 2024; 5:e776. [PMID: 39465140 PMCID: PMC11502721 DOI: 10.1002/mco2.776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/21/2024] [Accepted: 09/22/2024] [Indexed: 10/29/2024] Open
Abstract
Metabolism serves not only as the organism's energy source but also yields metabolites crucial for maintaining tissue homeostasis and overall health. Intestinal stem cells (ISCs) maintain intestinal homeostasis through continuous self-renewal and differentiation divisions. The intricate relationship between metabolic pathways and intestinal homeostasis underscores their crucial interplay. Metabolic pathways have been shown to directly regulate ISC self-renewal and influence ISC fate decisions under homeostatic conditions, but the cellular and molecular mechanisms remain incompletely understood. Understanding the intricate involvement of various pathways in maintaining intestinal homeostasis holds promise for devising innovative strategies to address intestinal diseases. Here, we provide a comprehensive review of recent advances in the regulation of intestinal homeostasis. We describe the regulation of intestinal homeostasis from multiple perspectives, including the regulation of intestinal epithelial cells, the regulation of the tissue microenvironment, and the key role of nutrient metabolism. We highlight the regulation of intestinal homeostasis and ISC by nutrient metabolism. This review provides a multifaceted perspective on how intestinal homeostasis is regulated and provides ideas for intestinal diseases and repair of intestinal damage.
Collapse
Affiliation(s)
- Ruolan Zhang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human DiseaseSoutheast UniversityNanjingChina
| | - Ansu Perekatt
- Department of Chemistry and Chemical BiologyStevens Institute of TechnologyHobokenNew JerseyUSA
| | - Lei Chen
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human DiseaseSoutheast UniversityNanjingChina
- Institute of Microphysiological SystemsSoutheast UniversityNanjingChina
| |
Collapse
|
3
|
Du J, Fang L, Wang Y, Zhao J, Feng Z, Yu Y, Fang D, Huang D, Zhai X, Cheng Y, Min R, Gao F, Liu C. Gelsolin regulates intestinal stem cell regeneration and Th17 cellular function. Cell Commun Signal 2024; 22:524. [PMID: 39472865 PMCID: PMC11520831 DOI: 10.1186/s12964-024-01902-5] [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: 05/24/2024] [Accepted: 10/20/2024] [Indexed: 11/02/2024] Open
Abstract
Intestinal stem cells (ISCs) are responsible for intestinal homeostasis and are important for the regeneration of damaged intestine. We established an ionizing radiation (IR)-induced intestinal injury model and observed that Gelsolin KO mice had increased radiosensitivity. The deletion of Gelsolin aggravated intestinal damage and reduced the number of ISCs after lethal IR. The intestinal organoid experiments showed that Gelsolin deletion inhibited ISCs function after IR. Notably, RNA sequencing and RT-PCR results showed IL-17 signaling pathway was down-regulated and Th17 cells differentiation was inhibited in Gelsolin KO mice. Moreover, recombinant IL-17 A ameliorated IR-induced intestinal injury and promoted ISCs regeneration. To figure out the role of Gelsolin in Th17 cells differentiation, flow cytometry was used and we found that Gelsolin targets Th17 cells functionality via the p-STAT3/RORγt axis. By establishing the co-culture system, we proved that Th17 cells promoted self-renewal and budding abilities in Gelsolin-deficient organoids. Finally, we found that Gelsolin was protective against DSS-induced colitis and that this protective effect was not specific or limited to the IR induced intestinal injury model. Based on these results, we proved Gelsolin maintained the regeneration of ISCs by sustaining Th17 cells functions via the p-STAT3/RORγt axis.
Collapse
Affiliation(s)
- Jicong Du
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, P.R. China
| | - Lan Fang
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, P.R. China
| | - Yuedong Wang
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, P.R. China
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, 325000, P.R. China
| | - Jianpeng Zhao
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, P.R. China
| | - Zhenlan Feng
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, 325000, P.R. China
| | - Yike Yu
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, P.R. China
| | - Duo Fang
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, P.R. China
| | - Daqian Huang
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, P.R. China
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, 325000, P.R. China
| | - Xuanlu Zhai
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, P.R. China
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, 325000, P.R. China
| | - Ying Cheng
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, P.R. China
| | - Rui Min
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, P.R. China
| | - Fu Gao
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, P.R. China.
| | - Cong Liu
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, P.R. China.
| |
Collapse
|
4
|
Zhao L, Zhao C, Miao Y, Lei S, Li Y, Gong J, Peng C. Theabrownin from Pu-erh Tea Improves DSS-Induced Colitis via Restoring Gut Homeostasis and Inhibiting TLR2&4 Signaling Pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155852. [PMID: 39029137 DOI: 10.1016/j.phymed.2024.155852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 06/07/2024] [Accepted: 06/27/2024] [Indexed: 07/21/2024]
Abstract
BACKGROUND Theabrownin (TB) is a dark brown pigment from Pu-erh tea or other dark teas. It is formed by further oxidization of theaflavins and thearubigins, in combination with proteins, polysaccharides, and caffeine etc. TB is a characteristic ingredient and bioactive substance of Pu-erh tea. However, the effects of TB on ulcerative colitis (UC) remains unclear. PURPOSE This study aims to elucidate the mechanism of TB on UC in terms of recovery of intestinal homeostasis and regulation of toll-like receptor (TLR) 2&4 signaling pathway. METHODS The colitis models were established by administering 5% dextran sulfate sodium (DSS) to C57BL/6 mice for 5 days to evaluate the therapeutic and preventive effects of TB on UC. Mesalazine was used as a positive control. H&E staining, complete blood count, enzyme-linked immunosorbent assay, immunohistochemistry, flow cytometry, and 16S rRNA sequencing were employed to assess histological changes, blood cells analysis, content of cytokines, expression and distribution of mucin (MUC)2 and TLR2&4, differentiation of CD4+T cells in lamina propria, and changes in intestinal microbiota, respectively. Western blot was utilized to study the relative expression of tight junction proteins and the key proteins in TLR2&4-mediated MyD88-dependent MAPK, NF-κB, and AKT signaling pathways. RESULTS TB outstanding alleviated colitis, inhibited the release of pro-inflammatory cytokines, reduced white blood cells while increasing red blood cells, hemoglobin, and platelets. TB increased the expression of occludin, claudin-1 and MUC2, effectively restored intestinal barrier function. TB also suppressed differentiation of Th1 and Th17 cells in the colon's lamina propria, increased the fraction of Treg cells, and promoted the balance of Treg/Th17 to tilt towards Tregs. Moreover, TB increased the Firmicutes to Bacteroides (F/B) ratio, as well as the abundance of Akkermansia, Muribaculaceae, and Eubacterium_coprostanoligenes_group at the genus level. In addition, TB inhibited the activation of TLR2&4-mediated MAPK, NF-κB, and AKT signaling pathways in intestinal epithelial cells of DSS-induced mice. CONCLUSION TB acts in restoring intestinal homeostasis and anti-inflammatory in DSS-induced UC, and exhibiting a preventive effect after long-term use. In a word, TB is a promising beverage, health product and food additive for UC.
Collapse
Affiliation(s)
- Lei Zhao
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, China; College of Science, Yunnan Agricultural University, Kunming, 650201, China
| | - Chunyan Zhao
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Yue Miao
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, China; Medicinal Plants Research Institute, Yunnan Academy of Agricultural Sciences, Kunming, 650223, China
| | - Shuwen Lei
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Yujing Li
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Jiashun Gong
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, China; Agro-products Processing Research Institute, Yunnan Academy of Agricultural Sciences, Kunming, 650223, China.
| | - Chunxiu Peng
- College of Horticulture and Landscape, Yunnan Agricultural University, Kunming, 650201, China.
| |
Collapse
|
5
|
Urbauer E, Aguanno D, Mindermann N, Omer H, Metwaly A, Krammel T, Faro T, Remke M, Reitmeier S, Bärthel S, Kersting J, Huang Z, Xian F, Schmidt M, Saur D, Huber S, Stecher B, List M, Gómez-Varela D, Steiger K, Allez M, Rath E, Haller D. Mitochondrial perturbation in the intestine causes microbiota-dependent injury and gene signatures discriminative of inflammatory disease. Cell Host Microbe 2024; 32:1347-1364.e10. [PMID: 39013472 DOI: 10.1016/j.chom.2024.06.013] [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: 10/12/2023] [Revised: 05/13/2024] [Accepted: 06/20/2024] [Indexed: 07/18/2024]
Abstract
Mitochondrial dysfunction is associated with inflammatory bowel diseases (IBDs). To understand how microbial-metabolic circuits contribute to intestinal injury, we disrupt mitochondrial function in the epithelium by deleting the mitochondrial chaperone, heat shock protein 60 (Hsp60Δ/ΔIEC). This metabolic perturbation causes self-resolving tissue injury. Regeneration is disrupted in the absence of the aryl hydrocarbon receptor (Hsp60Δ/ΔIEC;AhR-/-) involved in intestinal homeostasis or inflammatory regulator interleukin (IL)-10 (Hsp60Δ/ΔIEC;Il10-/-), causing IBD-like pathology. Injury is absent in the distal colon of germ-free (GF) Hsp60Δ/ΔIEC mice, highlighting bacterial control of metabolic injury. Colonizing GF Hsp60Δ/ΔIEC mice with the synthetic community OMM12 reveals expansion of metabolically flexible Bacteroides, and B. caecimuris mono-colonization recapitulates the injury. Transcriptional profiling of the metabolically impaired epithelium reveals gene signatures involved in oxidative stress (Ido1, Nos2, Duox2). These signatures are observed in samples from Crohn's disease patients, distinguishing active from inactive inflammation. Thus, mitochondrial perturbation of the epithelium causes microbiota-dependent injury with discriminative inflammatory gene profiles relevant for IBD.
Collapse
Affiliation(s)
- Elisabeth Urbauer
- Chair of Nutrition and Immunology, Technical University of Munich, Gregor-Mendel-Strasse 2, 85354 Freising, Germany
| | - Doriane Aguanno
- Chair of Nutrition and Immunology, Technical University of Munich, Gregor-Mendel-Strasse 2, 85354 Freising, Germany
| | - Nora Mindermann
- Chair of Nutrition and Immunology, Technical University of Munich, Gregor-Mendel-Strasse 2, 85354 Freising, Germany
| | - Hélène Omer
- Chair of Nutrition and Immunology, Technical University of Munich, Gregor-Mendel-Strasse 2, 85354 Freising, Germany
| | - Amira Metwaly
- Chair of Nutrition and Immunology, Technical University of Munich, Gregor-Mendel-Strasse 2, 85354 Freising, Germany
| | - Tina Krammel
- Chair of Nutrition and Immunology, Technical University of Munich, Gregor-Mendel-Strasse 2, 85354 Freising, Germany
| | - Tim Faro
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, 80337 Munich, Germany
| | - Marianne Remke
- Institute of Pathology, Technical University of Munich, 81675 Munich, Germany
| | - Sandra Reitmeier
- Chair of Nutrition and Immunology, Technical University of Munich, Gregor-Mendel-Strasse 2, 85354 Freising, Germany
| | - Stefanie Bärthel
- Division of Translational Cancer Research, German Cancer Research Center and German Cancer Consortium, 69120 Heidelberg, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; Institute of Experimental Cancer Therapy, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Johannes Kersting
- Data Science in Systems Biology, TUM School of Life Sciences, Technical University of Munich, Maximus-von-Imhof Forum 3, 85354 Freising, Germany
| | - Zihua Huang
- Data Science in Systems Biology, TUM School of Life Sciences, Technical University of Munich, Maximus-von-Imhof Forum 3, 85354 Freising, Germany
| | - Feng Xian
- Systems Biology of Pain, Division of Pharmacology & Toxicology, Department of Pharmaceutical Sciences, Faculty of Life Sciences, University of Vienna, 1090 Vienna, Austria
| | - Manuela Schmidt
- Systems Biology of Pain, Division of Pharmacology & Toxicology, Department of Pharmaceutical Sciences, Faculty of Life Sciences, University of Vienna, 1090 Vienna, Austria
| | - Dieter Saur
- Division of Translational Cancer Research, German Cancer Research Center and German Cancer Consortium, 69120 Heidelberg, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; Institute of Experimental Cancer Therapy, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Samuel Huber
- Section of Molecular Immunology and Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Bärbel Stecher
- Max von Pettenkofer-Institute for Hygiene and Clinical Microbiology, Ludwig-Maximilians University of Munich, 80336 Munich, Germany; German Center for Infection Research, Partner site LMU Munich, 80336 Munich, Germany
| | - Markus List
- Data Science in Systems Biology, TUM School of Life Sciences, Technical University of Munich, Maximus-von-Imhof Forum 3, 85354 Freising, Germany; Munich Data Science Institute (MDSI), Technical University of Munich, 85748 Garching, Germany
| | - David Gómez-Varela
- Systems Biology of Pain, Division of Pharmacology & Toxicology, Department of Pharmaceutical Sciences, Faculty of Life Sciences, University of Vienna, 1090 Vienna, Austria
| | - Katja Steiger
- Institute of Pathology, Technical University of Munich, 81675 Munich, Germany
| | - Matthieu Allez
- Department of Gastroenterology, Hôpital Saint-Louis, APHP, INSERM UMRS 1160, Paris Diderot, Sorbonne Paris-Cité University, 75010 Paris, France
| | - Eva Rath
- Chair of Nutrition and Immunology, Technical University of Munich, Gregor-Mendel-Strasse 2, 85354 Freising, Germany
| | - Dirk Haller
- Chair of Nutrition and Immunology, Technical University of Munich, Gregor-Mendel-Strasse 2, 85354 Freising, Germany; ZIEL - Institute for Food & Health, Technical University of Munich, 85354 Freising, Germany.
| |
Collapse
|
6
|
Chen X, Xuan Y, Chen Y, Yang F, Zhu M, Xu J, Chen J. Polystyrene nanoplastics induce intestinal and hepatic inflammation through activation of NF-κB/NLRP3 pathways and related gut-liver axis in mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 935:173458. [PMID: 38796000 DOI: 10.1016/j.scitotenv.2024.173458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/01/2024] [Accepted: 05/20/2024] [Indexed: 05/28/2024]
Abstract
Nanoplastics (NPs) present in food and water poses a genuine risk of their accumulation in humans through the diet. Preferential contact between ingested NPs and the intestine as well as the liver has the potential to induce enteritis and hepatitis. However, there is still a lack of comprehensive understanding regarding the inter-organ crosstalk between the intestine and liver when exposed to NPs, as well as the underlying signaling pathways involved. In this study, we employed a 21-day mice exposure model to investigate the accumulation profile of PS-NPs and elucidate the mechanism of intestinal and hepatic inflammation induced by NPs. After exposure, notable fluorescent signals originating from PS-NPs were detected not only in the stomach and intestine but also in other organs such as liver, lung, kidney, brain, and testes. Histopathological analysis along with routine blood tests both revealed an acute inflammatory reaction in mice. Further mechanistic investigations demonstrated that PS-NPs activated inflammatory NF-κB/NLRP3 pathways and induced the expression of cytokines IL-1β and IL-18 in the intestine, which recruited macrophages and neutrophils into the intestine. Concurrently, a significant decrease in the expression levels of intestinal tight junction proteins (Claudin-1, Occludin, and ZO-1) was observed, resulting in an increase in intestinal permeability and elevated endotoxin (LPS) levels. The high levels of LPS further activated TLR4/NF-κB/NLRP3/GSDMD pathways in the liver, inducing liver inflammation and hepatocyte pyroptosis. The impairment of liver function was positively correlated with intestinal inflammation and barrier disruption. These findings underscore that exposure to NPs can instigate enteritis and hepatitis while emphasizing the crucial role played by the indirect gut-liver axis in elucidating the potential mechanism underlying NP-induced liver pathogenesis.
Collapse
Affiliation(s)
- Xuanwei Chen
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hang Zhou 310053, China
| | - Yang Xuan
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hang Zhou 310053, China
| | - Yawen Chen
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hang Zhou 310053, China
| | - Fanfan Yang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hang Zhou 310053, China
| | - Mengying Zhu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hang Zhou 310053, China
| | - Jian Xu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hang Zhou 310053, China.
| | - Jin Chen
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hang Zhou 310053, China.
| |
Collapse
|
7
|
Huang Y, Wang X, Sun L. The gut-liver axis calibrates PEDF production for ISC homeostasis. BIOPHYSICS REPORTS 2024; 10:175-177. [PMID: 39027319 PMCID: PMC11252237 DOI: 10.52601/bpr.2024.240904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 04/03/2024] [Indexed: 07/20/2024] Open
Affiliation(s)
- Ying Huang
- State Key Laboratory of Female Fertility Promotion, Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing 100191, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
| | - Xinran Wang
- State Key Laboratory of Female Fertility Promotion, Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing 100191, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
| | - Lulu Sun
- State Key Laboratory of Female Fertility Promotion, Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing 100191, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
| |
Collapse
|
8
|
Li C, Zhang P, Xie Y, Wang S, Guo M, Wei X, Zhang K, Cao D, Zhou R, Wang S, Song X, Zhu S, Pan W. Enterococcus-derived tyramine hijacks α 2A-adrenergic receptor in intestinal stem cells to exacerbate colitis. Cell Host Microbe 2024; 32:950-963.e8. [PMID: 38788722 DOI: 10.1016/j.chom.2024.04.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/28/2024] [Accepted: 04/30/2024] [Indexed: 05/26/2024]
Abstract
Inflammatory bowel disease (IBD) is characterized by dysbiosis of the gut microbiota and dysfunction of intestinal stem cells (ISCs). However, the direct interactions between IBD microbial factors and ISCs are undescribed. Here, we identify α2A-adrenergic receptor (ADRA2A) as a highly expressed GPCR in ISCs. Through PRESTO-Tango screening, we demonstrate that tyramine, primarily produced by Enterococcus via tyrosine decarboxylase (tyrDC), serves as a microbial ligand for ADRA2A. Using an engineered tyrDC-deficient Enterococcus faecalis strain and intestinal epithelial cell-specific Adra2a knockout mice, we show that Enterococcus-derived tyramine suppresses ISC proliferation, thereby impairing epithelial regeneration and exacerbating DSS-induced colitis through ADRA2A. Importantly, blocking the axis with an ADRA2A antagonist, yohimbine, disrupts tyramine-mediated suppression on ISCs and alleviates colitis. Our findings highlight a microbial ligand-GPCR pair in ISCs, revealing a causal link between microbial regulation of ISCs and colitis exacerbation and yielding a targeted therapeutic approach to restore ISC function in colitis.
Collapse
Affiliation(s)
- Chaoliang Li
- Department of Digestive Disease, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Key Laboratory of immune response and immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Panrui Zhang
- Department of Digestive Disease, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Key Laboratory of immune response and immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Yadong Xie
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Shishan Wang
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Meng Guo
- Department of Digestive Disease, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Key Laboratory of immune response and immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Xiaowei Wei
- Department of Digestive Disease, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Key Laboratory of immune response and immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Kaiguang Zhang
- Department of Digestive Disease, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Dan Cao
- Department of Digestive Disease, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Key Laboratory of immune response and immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Rongbin Zhou
- Key Laboratory of immune response and immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Sheng Wang
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Xinyang Song
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.
| | - Shu Zhu
- Department of Digestive Disease, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Key Laboratory of immune response and immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Wen Pan
- Department of Digestive Disease, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Key Laboratory of immune response and immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| |
Collapse
|
9
|
Liu YY, Wu DK, Chen JB, Tang YM, Jiang F. Advances in the study of gastric organoids as disease models. World J Gastrointest Oncol 2024; 16:1725-1736. [PMID: 38764838 PMCID: PMC11099456 DOI: 10.4251/wjgo.v16.i5.1725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/23/2024] [Accepted: 03/25/2024] [Indexed: 05/09/2024] Open
Abstract
Gastric organoids are models created in the laboratory using stem cells and sophisticated three-dimensional cell culture techniques. These models have shown great promise in providing valuable insights into gastric physiology and advanced disease research. This review comprehensively summarizes and analyzes the research advances in culture methods and techniques for adult stem cells and induced pluripotent stem cell-derived organoids, and patient-derived organoids. The potential value of gastric organoids in studying the pathogenesis of stomach-related diseases and facilitating drug screening is initially discussed. The construction of gastric organoids involves several key steps, including cell extraction and culture, three-dimensional structure formation, and functional expression. Simulating the structure and function of the human stomach by disease modeling with gastric organoids provides a platform to study the mechanism of gastric cancer induction by Helicobacter pylori. In addition, in drug screening and development, gastric organoids can be used as a key tool to evaluate drug efficacy and toxicity in preclinical trials. They can also be used for precision medicine according to the specific conditions of patients with gastric cancer, to assess drug resistance, and to predict the possibility of adverse reactions. However, despite the impressive progress in the field of gastric organoids, there are still many unknowns that need to be addressed, especially in the field of regenerative medicine. Meanwhile, the reproducibility and consistency of organoid cultures are major challenges that must be overcome. These challenges have had a significant impact on the development of gastric organoids. Nonetheless, as technology continues to advance, we can foresee more comprehensive research in the construction of gastric organoids. Such research will provide better solutions for the treatment of stomach-related diseases and personalized medicine.
Collapse
Affiliation(s)
- Yi-Yang Liu
- Graduate School, Guangxi University of Chinese Medicine, Nanning 530011, Guangxi Zhuang Autonomous Region, China
| | - De-Kun Wu
- Teaching Experiment and Training Center, Guangxi University of Chinese Medicine, Nanning 530011, Guangxi Zhuang Autonomous Region, China
| | - Ji-Bing Chen
- Central Laboratory, Ruikang Hospital Affiliated to Guangxi University of Traditional Chinese Medicine, Nanning 530011, Guangxi Zhuang Autonomous Region, China
| | - You-Ming Tang
- Department of Digestive Disease, Ruikang Hospital Affiliated to Guangxi University of Traditional Chinese Medicine, Nanning 530011, Guangxi Zhuang Autonomous Region, China
| | - Feng Jiang
- AIDS Research Center, Ruikang Hospital Affiliated to Guangxi University of Traditional Chinese Medicine, Nanning 530011, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
10
|
Chen Y, Sun H, Luo Z, Mei Y, Xu Z, Tan J, Xie Y, Li M, Xia J, Yang B, Su B. Crosstalk between CD8 + T cells and mesenchymal stromal cells in intestine homeostasis and immunity. Adv Immunol 2024; 162:23-58. [PMID: 38866438 DOI: 10.1016/bs.ai.2024.02.001] [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] [Indexed: 06/14/2024]
Abstract
The intestine represents the most complex cellular network in the whole body. It is constantly faced with multiple types of immunostimulatory agents encompassing from food antigen, gut microbiome, metabolic waste products, and dead cell debris. Within the intestine, most T cells are found in three primary compartments: the organized gut-associated lymphoid tissue, the lamina propria, and the epithelium. The well-orchestrated epithelial-immune-microbial interaction is critically important for the precise immune response. The main role of intestinal mesenchymal stromal cells is to support a structural framework within the gut wall. However, recent evidence from stromal cell studies indicates that they also possess significant immunomodulatory functions, such as maintaining intestinal tolerance via the expression of PDL1/2 and MHC-II molecules, and promoting the development of CD103+ dendritic cells, and IgA+ plasma cells, thereby enhancing intestinal homeostasis. In this review, we will summarize the current understanding of CD8+ T cells and stromal cells alongside the intestinal tract and discuss the reciprocal interactions between T subsets and mesenchymal stromal cell populations. We will focus on how the tissue residency, migration, and function of CD8+ T cells could be potentially regulated by mesenchymal stromal cell populations and explore the molecular mediators, such as TGF-β, IL-33, and MHC-II molecules that might influence these processes. Finally, we discuss the potential pathophysiological impact of such interaction in intestine hemostasis as well as diseases of inflammation, infection, and malignancies.
Collapse
Affiliation(s)
- Yao Chen
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongxiang Sun
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhengnan Luo
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yisong Mei
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziyang Xu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianmei Tan
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiting Xie
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengda Li
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaqi Xia
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Beichun Yang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bing Su
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Key Laboratory of Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
11
|
Zhang L, Luo X, Tang R, Wu Y, Liang Z, Liu J, Pi J, Zhang H. MiR-106a-5p by Targeting MAP3K2 Promotes Repair of Oxidative Stress Damage to the Intestinal Barrier in Prelaying Ducks. Animals (Basel) 2024; 14:1037. [PMID: 38612276 PMCID: PMC11010895 DOI: 10.3390/ani14071037] [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: 01/24/2024] [Revised: 03/08/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Under caged stress conditions, severe disruptions in duck intestinal barrier function, which adversely affect economic performance, have been observed. MiRNAs play a crucial role in cellular processes, but the mechanisms underlying their involvement in repairing oxidative stress-induced damage to duck intestinal barriers have not been elucidated. We performed miRNA-seq and protein tandem mass tagging (TMT) sequencing and identified differentially expressed miRNAs and proteins in oxidative stress-treated ducks. Dual-luciferase reporter vector experiments, RT-qPCR, and Western blotting revealed the regulatory role of apla-miR-106a-5p/MAP3K2 in intestinal barrier damage repair. The results showed that oxidative stress led to shortened villi and deepened crypts, impairing intestinal immune function. Significant downregulation of apla-miR-106a-5p was revealed by miRNA-seq, and the inhibition of its expression not only enhanced cell viability but also improved intestinal barrier function. TMT protein sequencing revealed MAP3K2 upregulation in caged-stressed duck intestines, and software analysis confirmed MAP3K2 as the target gene of apla-miR-106a-5p. Dual-fluorescence reporter gene experiments demonstrated direct targeting of MAP3K2 by apla-miR-106a-5p. RT-qPCR showed no effect on MAP3K2 expression, while Western blot analysis indicated that MAP3K2 protein expression was suppressed. In summary, apla-miR-106a-5p targets MAP3K2, regulating gene expression at the transcriptional level and facilitating effective repair of intestinal barrier damage. This discovery provides new insights into the molecular mechanisms of physiological damage in ducks under caged stress, offering valuable guidance for related research.
Collapse
Affiliation(s)
- Li Zhang
- Hubei Key Laboratory of Animal Embryo Engineering and Molecular Breeding, Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; (L.Z.); (X.L.); (R.T.); (Y.W.); (Z.L.); (J.P.)
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, China;
| | - Xiang Luo
- Hubei Key Laboratory of Animal Embryo Engineering and Molecular Breeding, Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; (L.Z.); (X.L.); (R.T.); (Y.W.); (Z.L.); (J.P.)
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, China;
| | - Rui Tang
- Hubei Key Laboratory of Animal Embryo Engineering and Molecular Breeding, Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; (L.Z.); (X.L.); (R.T.); (Y.W.); (Z.L.); (J.P.)
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, China;
| | - Yan Wu
- Hubei Key Laboratory of Animal Embryo Engineering and Molecular Breeding, Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; (L.Z.); (X.L.); (R.T.); (Y.W.); (Z.L.); (J.P.)
| | - Zhenhua Liang
- Hubei Key Laboratory of Animal Embryo Engineering and Molecular Breeding, Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; (L.Z.); (X.L.); (R.T.); (Y.W.); (Z.L.); (J.P.)
| | - Jingbo Liu
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, China;
| | - Jinsong Pi
- Hubei Key Laboratory of Animal Embryo Engineering and Molecular Breeding, Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; (L.Z.); (X.L.); (R.T.); (Y.W.); (Z.L.); (J.P.)
| | - Hao Zhang
- Hubei Key Laboratory of Animal Embryo Engineering and Molecular Breeding, Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; (L.Z.); (X.L.); (R.T.); (Y.W.); (Z.L.); (J.P.)
| |
Collapse
|
12
|
Ventrello SW, McMurry NR, Edwards NM, Bain LJ. Chronic arsenic exposure affects stromal cells and signaling in the small intestine in a sex-specific manner. Toxicol Sci 2024; 198:303-315. [PMID: 38310360 DOI: 10.1093/toxsci/kfae016] [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: 02/05/2024] Open
Abstract
Arsenic is a toxicant that is ingested through drinking water and food, exposing nearly 140 million people to levels above the 10 ppb guideline concentration. Studies have shown that arsenic affects intestinal stem cells (ISCs), but the mechanisms by which arsenic alters the formation of adult cells in the small intestine are not well understood. Signals derived from intestinal stromal cells initiate and maintain differentiation. The goal of this study is to evaluate arsenic's effect on intestinal stromal cells, including PdgfrαLo trophocytes, located proximal to the ISCs, and PdgfrαHi telocytes, located proximal to the transit-amplifying region and up the villi. Adult Sox9tm2Crm-EGFP mice were exposed to 0, 33, and 100 ppb sodium arsenite in their drinking water for 13 weeks, and sections of duodenum were examined. Flow cytometry indicated that arsenic exposure dose-responsively reduced Sox9+ epithelial cells and trended toward increased Pdgfrα+ cells. The trophocyte marker, CD81, was reduced by 10-fold and 9.0-fold in the 100 ppb exposure group in male and female mice, respectively. Additionally, a significant 2.2- to 3.1-fold increase in PdgfrαLo expression was found in male mice in trophocytes and Igfbp5+ cells. PdgfrαHi protein expression, a telocyte marker, was more prevalent along the villus/crypt structure in females, whereas Gli1 expression (telocytes) was reduced in male mice exposed to arsenic. Principle coordinate analysis confirmed the sex-dependent response to arsenic exposure, with an increase in trophocyte and decrease in telocyte marker expression observed in male mice. These results imply that arsenic alters intestinal mesenchymal cells in a sex-dependent manner.
Collapse
Affiliation(s)
- Scott W Ventrello
- Department of Biological Sciences, Clemson University, Clemson, South Carolina 29634, USA
| | - Nicholas R McMurry
- Department of Biological Sciences, Clemson University, Clemson, South Carolina 29634, USA
| | - Nicholas M Edwards
- Department of Biological Sciences, Clemson University, Clemson, South Carolina 29634, USA
| | - Lisa J Bain
- Department of Biological Sciences, Clemson University, Clemson, South Carolina 29634, USA
| |
Collapse
|
13
|
Wei W, Liu Y, Hou Y, Cao S, Chen Z, Zhang Y, Cai X, Yan Q, Li Z, Yuan Y, Wang G, Zheng X, Hao H. Psychological stress-induced microbial metabolite indole-3-acetate disrupts intestinal cell lineage commitment. Cell Metab 2024; 36:466-483.e7. [PMID: 38266651 DOI: 10.1016/j.cmet.2023.12.026] [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: 06/23/2023] [Revised: 11/12/2023] [Accepted: 12/21/2023] [Indexed: 01/26/2024]
Abstract
The brain and gut are intricately connected and respond to various stimuli. Stress-induced brain-gut communication is implicated in the pathogenesis and relapse of gut disorders. The mechanism that relays psychological stress to the intestinal epithelium, resulting in maladaptation, remains poorly understood. Here, we describe a stress-responsive brain-to-gut metabolic axis that impairs intestinal stem cell (ISC) lineage commitment. Psychological stress-triggered sympathetic output enriches gut commensal Lactobacillus murinus, increasing the production of indole-3-acetate (IAA), which contributes to a transferrable loss of intestinal secretory cells. Bacterial IAA disrupts ISC mitochondrial bioenergetics and thereby prevents secretory lineage commitment in a cell-intrinsic manner. Oral α-ketoglutarate supplementation bolsters ISC differentiation and confers resilience to stress-triggered intestinal epithelial injury. We confirm that fecal IAA is higher in patients with mental distress and is correlated with gut dysfunction. These findings uncover a microbe-mediated brain-gut pathway that could be therapeutically targeted for stress-driven gut-brain comorbidities.
Collapse
Affiliation(s)
- Wei Wei
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yali Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China; State Key Laboratory of Digestive Diseases, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Yuanlong Hou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmacy, Shenzhen Luohu People's Hospital, Shenzhen 518005, China
| | - Shuqi Cao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zhuo Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Youying Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaoying Cai
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qingyuan Yan
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ziguang Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yonggui Yuan
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Guangji Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Xiao Zheng
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Haiping Hao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
14
|
Chen W, Cui Y, Li C, He C, Du L, Liu W, He Z. KLF2 controls proliferation and apoptosis of human spermatogonial stem cells via targeting GJA1. iScience 2024; 27:109024. [PMID: 38352225 PMCID: PMC10863320 DOI: 10.1016/j.isci.2024.109024] [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: 07/25/2023] [Revised: 11/14/2023] [Accepted: 01/22/2024] [Indexed: 02/16/2024] Open
Abstract
Human spermatogonial stem cells (SSCs) are essential for spermatogenesis and male fertility. However, molecular mechanisms regulating fate determinations of human SSCs remain elusive. In this study, we revealed that KLF2 decreased the proliferation, DNA synthesis, and colonization of human SSCs as well as increased apoptosis of these cells. We identified and demonstrated that GJA1 was a target gene for KLF2 in human SSCs. Notably, KLF2 overexpression rescued the reduction of proliferation of human SSCs caused by GJA1 silencing as well as the enhancement of apoptosis of human SSCs. Abnormalities in the higher level of KLF2 and/or KIF2 mutations might lead to male infertility. Collectively, these results implicate that KLF2 inhibits proliferation of human SSCs and enhances their apoptosis by targeting GJA1. This study thus provides novel genetic mechanisms underlying human spermatogenesis and azoospermia, and it offers new endogenous targets for treating male infertility.
Collapse
Affiliation(s)
- Wei Chen
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Manufacture-Based Learning and Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha, China
| | - Yinghong Cui
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Manufacture-Based Learning and Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha, China
| | - Chunyun Li
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Manufacture-Based Learning and Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha, China
| | - Caimei He
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Manufacture-Based Learning and Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha, China
| | - Li Du
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Manufacture-Based Learning and Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha, China
| | - Wei Liu
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Manufacture-Based Learning and Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha, China
| | - Zuping He
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Manufacture-Based Learning and Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha, China
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
15
|
Chai T, Shen J, Sheng Y, Huang Y, Liang W, Zhang Z, Zhao R, Shang H, Cheng W, Zhang H, Chen X, Huang X, Zhang Y, Liu J, Yang H, Wang L, Pan S, Chen Y, Han L, Qiu Q, Gao A, Wei H, Fang X. Effects of flora deficiency on the structure and function of the large intestine. iScience 2024; 27:108941. [PMID: 38333708 PMCID: PMC10850757 DOI: 10.1016/j.isci.2024.108941] [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: 06/27/2023] [Revised: 11/03/2023] [Accepted: 01/15/2024] [Indexed: 02/10/2024] Open
Abstract
The significant anatomical changes in large intestine of germ-free (GF) mice provide excellent material for understanding microbe-host crosstalk. We observed significant differences of GF mice in anatomical and physiological involving in enlarged cecum, thinned mucosal layer and enriched water in cecal content. Furthermore, integration analysis of multi-omics data revealed the associations between the structure of large intestinal mesenchymal cells and the thinning of the mucosal layer. Increased Aqp8 expression in GF mice may contribute to enhanced water secretion or altered hydrodynamics in the cecum. In addition, the proportion of epithelial cells, nutrient absorption capacity, immune function and the metabolome of cecum contents of large intestine were also significantly altered. Together, this is the first systematic study of the transcriptome and metabolome of the cecum and colon of GF mice, and these findings contribute to our understanding of the intricate interactions between microbes and the large intestine.
Collapse
Affiliation(s)
- Tailiang Chai
- University of the Chinese Academy of Sciences, College of Life Sciences, Beijing, Beijing, China
- BGI, Shenzhen, Guangdong, China
| | | | - Yifei Sheng
- University of the Chinese Academy of Sciences, College of Life Sciences, Beijing, Beijing, China
- BGI, Shenzhen, Guangdong, China
| | | | | | - Zhao Zhang
- University of the Chinese Academy of Sciences, College of Life Sciences, Beijing, Beijing, China
- BGI, Shenzhen, Guangdong, China
| | - Ruizhen Zhao
- University of the Chinese Academy of Sciences, College of Life Sciences, Beijing, Beijing, China
- BGI, Shenzhen, Guangdong, China
| | - Haitao Shang
- Sun Yat-sen University First Affiliated Hospital, Precision Medicine Institute, Guangzhou, Guangdong, China
| | - Wei Cheng
- Huazhong Agricultural University, College of Animal Sciences and Technology, Wuhan, Hubei, China
| | - Hang Zhang
- Huazhong Agricultural University, College of Animal Sciences and Technology, Wuhan, Hubei, China
| | - Xueting Chen
- University of the Chinese Academy of Sciences, College of Life Sciences, Beijing, Beijing, China
- BGI, Shenzhen, Guangdong, China
| | - Xiang Huang
- University of the Chinese Academy of Sciences, College of Life Sciences, Beijing, Beijing, China
| | - Yin Zhang
- University of the Chinese Academy of Sciences, College of Life Sciences, Beijing, Beijing, China
- BGI, Shenzhen, Guangdong, China
| | | | | | | | | | - Yang Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lijuan Han
- Department of Scientific Research, Kangmeihuada GeneTech Co., Ltd. (KMHD), Shenzhen, China
| | - Qinwei Qiu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Aibo Gao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hong Wei
- Sun Yat-sen University First Affiliated Hospital, Precision Medicine Institute, Guangzhou, Guangdong, China
| | - Xiaodong Fang
- BGI, Shenzhen, Guangdong, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
16
|
Wang X, Ding C, Li HB. The crosstalk between enteric nervous system and immune system in intestinal development, homeostasis and diseases. SCIENCE CHINA. LIFE SCIENCES 2024; 67:41-50. [PMID: 37672184 DOI: 10.1007/s11427-023-2376-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/19/2023] [Indexed: 09/07/2023]
Abstract
The gut is the largest digestive and absorptive organ, which is essential for induction of mucosal and systemic immune responses, and maintenance of metabolic-immune homeostasis. The intestinal components contain the epithelium, stromal cells, immune cells, and enteric nervous system (ENS), as well as the outers, such as gut microbiota, metabolites, and nutrients. The dyshomeostasis of intestinal microenvironment induces abnormal intestinal development and functions, even colon diseases including dysplasia, inflammation and tumor. Several recent studies have identified that ENS plays a crucial role in maintaining the immune homeostasis of gastrointestinal (GI) microenvironment. The crosstalk between ENS and immune cells, mainly macrophages, T cells, and innate lymphoid cells (ILCs), has been found to exert important regulatory roles in intestinal tissue programming, homeostasis, function, and inflammation. In this review, we mainly summarize the critical roles of the interactions between ENS and immune cells in intestinal homeostasis during intestinal development and diseases progression, to provide theoretical bases and ideas for the exploration of immunotherapy for gastrointestinal diseases with the ENS as potential novel targets.
Collapse
Affiliation(s)
- Xindi Wang
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chenbo Ding
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Hua-Bing Li
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
17
|
Teshima T, Hashimoto D. Separation of GVL from GVHD -location, location, location. Front Immunol 2023; 14:1296663. [PMID: 38116007 PMCID: PMC10728488 DOI: 10.3389/fimmu.2023.1296663] [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: 09/18/2023] [Accepted: 11/21/2023] [Indexed: 12/21/2023] Open
Abstract
Allogeneic hematopoietic cell transplantation (HCT) is a curative therapy for various hematologic malignancies. However, alloimmune response is a double-edged sword that mediates both beneficial graft-versus-leukemia (GVL) effects and harmful graft-versus-host disease (GVHD). Separation of GVL effects from GVHD has been a topic of intense research to improve transplant outcomes, but reliable clinical strategies have not yet been established. Target tissues of acute GVHD are the skin, liver, and intestine, while leukemic stem cells reside in the bone marrow. Tissue specific effector T-cell migration is determined by a combination of inflammatory and chemotactic signals that interact with specific receptors on T cells. Specific inhibition of donor T cell migration to GVHD target tissues while preserving migration to the bone marrow may represent a novel strategy to separate GVL from GVHD. Furthermore, tissue specific GVHD therapy, promoting tissue tolerance, and targeting of the tumor immune microenvironment may also help to separate GVHD and GVL.
Collapse
Affiliation(s)
- Takanori Teshima
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | | |
Collapse
|
18
|
Minegishi M, Kuchimaru T, Nishikawa K, Isagawa T, Iwano S, Iida K, Hara H, Miura S, Sato M, Watanabe S, Shiomi A, Mabuchi Y, Hamana H, Kishi H, Sato T, Sawaki D, Sato S, Hanazono Y, Suzuki A, Kohro T, Kadonosono T, Shimogori T, Miyawaki A, Takeda N, Shintaku H, Kizaka-Kondoh S, Nishimura S. Secretory GFP reconstitution labeling of neighboring cells interrogates cell-cell interactions in metastatic niches. Nat Commun 2023; 14:8031. [PMID: 38052804 DOI: 10.1038/s41467-023-43855-2] [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: 09/02/2022] [Accepted: 11/17/2023] [Indexed: 12/07/2023] Open
Abstract
Cancer cells inevitably interact with neighboring host tissue-resident cells during the process of metastatic colonization, establishing a metastatic niche to fuel their survival, growth, and invasion. However, the underlying mechanisms in the metastatic niche are yet to be fully elucidated owing to the lack of methodologies for comprehensively studying the mechanisms of cell-cell interactions in the niche. Here, we improve a split green fluorescent protein (GFP)-based genetically encoded system to develop secretory glycosylphosphatidylinositol-anchored reconstitution-activated proteins to highlight intercellular connections (sGRAPHIC) for efficient fluorescent labeling of tissue-resident cells that neighbor on and putatively interact with cancer cells in deep tissues. The sGRAPHIC system enables the isolation of metastatic niche-associated tissue-resident cells for their characterization using a single-cell RNA sequencing platform. We use this sGRAPHIC-leveraged transcriptomic platform to uncover gene expression patterns in metastatic niche-associated hepatocytes in a murine model of liver metastasis. Among the marker genes of metastatic niche-associated hepatocytes, we identify Lgals3, encoding galectin-3, as a potential pro-metastatic factor that accelerates metastatic growth and invasion.
Collapse
Affiliation(s)
- Misa Minegishi
- School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa, Japan
- RIKEN Cluster for Pioneering Research, Saitama, Japan
| | - Takahiro Kuchimaru
- RIKEN Cluster for Pioneering Research, Saitama, Japan.
- Graduate School of Medicine, Jichi Medical University, Tochigi, Japan.
- Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan.
- Data Science Center, Jichi Medical University, Tochigi, Japan.
| | | | - Takayuki Isagawa
- Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
- Data Science Center, Jichi Medical University, Tochigi, Japan
| | - Satoshi Iwano
- RIKEN Center for Brain Science, Saitama, Japan
- Institute for Tenure Track Promotion, University of Miyazaki, Miyazaki, Japan
| | - Kei Iida
- Faculty of Science and Engineering, Kindai University, Osaka, Japan
| | - Hiromasa Hara
- Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Shizuka Miura
- Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Marika Sato
- MediGear International Corporation, Kanagawa, Japan
| | | | | | - Yo Mabuchi
- Graduate School of Medicine, Juntendo University, Tokyo, Japan
- School of Medicine, Fujita Health University, Aichi, Japan
| | - Hiroshi Hamana
- Department of Immunology, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, Japan
| | - Hiroyuki Kishi
- Department of Immunology, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, Japan
| | - Tatsuyuki Sato
- Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Daigo Sawaki
- Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
- Clinical Pharmacology, Jichi Medical University, Tochigi, Japan
| | - Shigeru Sato
- Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Yutaka Hanazono
- Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Atsushi Suzuki
- Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Takahide Kohro
- Data Science Center, Jichi Medical University, Tochigi, Japan
| | - Tetsuya Kadonosono
- School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa, Japan
| | | | | | - Norihiko Takeda
- Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | | | - Shinae Kizaka-Kondoh
- School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa, Japan
| | - Satoshi Nishimura
- Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
19
|
Guan J, Fan Y, Wang S, Zhou F. Functions of MAP3Ks in antiviral immunity. Immunol Res 2023; 71:814-832. [PMID: 37286768 PMCID: PMC10247270 DOI: 10.1007/s12026-023-09401-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 06/01/2023] [Indexed: 06/09/2023]
Abstract
Immune signal transduction is crucial to the body's defense against viral infection. Recognition of pathogen-associated molecular patterns by pattern recognition receptors (PRRs) activates the transcription of interferon regulators and nuclear factor-κB (NF-κB); this promotes the release of interferons and inflammatory factors. Efficient regulation of type I interferon and NF-κB signaling by members of the mitogen-activated protein (MAP) kinase kinase kinase (MAP3K) family plays an important role in antiviral immunity. Elucidating the specific roles of MAP3K activation during viral infection is essential to develop effective antiviral therapies. In this review, we outline the specific regulatory mechanisms of MAP3Ks in antiviral immunity and discuss the feasibility of targeting MAP3Ks for the treatment of virus-induced diseases.
Collapse
Affiliation(s)
- Jizhong Guan
- Institutes of Biology and Medical Science, Soochow University, Suzhou, 215123, China
| | - Yao Fan
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China
| | - Shuai Wang
- Institutes of Biology and Medical Science, Soochow University, Suzhou, 215123, China
| | - Fangfang Zhou
- Institutes of Biology and Medical Science, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
20
|
Zhao X, Li L, Sun H, Jiang X, Bai W, Wu N, Wang J, Su B. A novel reporter mouse line for studying alveolar macrophages. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2527-2542. [PMID: 37428305 DOI: 10.1007/s11427-022-2325-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 03/13/2023] [Indexed: 07/11/2023]
Abstract
Alveolar macrophages (AMs) are self-maintained immune cells that play vital roles in lung homeostasis and immunity. Although reporter mice and culture systems have been established for studying macrophages, an accurate and specific reporter line for alveolar macrophage study is still not available. Here we reported a novel Rspo1-tdTomato gene reporter mouse line that could specifically label mouse AMs in a cell-intrinsic manner. Using this reporter system, we visualized the dynamics of alveolar macrophages intravitally under steady state and characterized the alveolar macrophage differentiation under in vitro condition. By performing ATAC-seq, we found that insertion of the tdTomato cassette in the Rspo1 locus increased the accessibility of a PPARE motif within the Rspo1 locus and revealed a potential regulation by key transcription factor PPAR-γ for alveolar macrophage differentiation in vitro and in vivo. Consistently, perturbation of PPAR-γ by its agonist rosiglitazone or inhibitor GW9662 resulted in corresponding alteration of tdTomato expression in alveolar macrophages together with the transcription of PPAR-γ downstream target genes. Furthermore, global transcriptomic analyses of AMs from the wild type mice and the Rspo1-tdTomato mice showed comparable gene expression profiles, especially those AM-specific genes, confirming that the insertion of the tdTomato cassette in the Rspo1 locus does not impact the cell identity and biological function of AMs under normal condition. Taken together, our study provides an alternative tool for in vivo and in vitro labeling of alveolar macrophages with high specificity which could also be utilized as an indicator of PPAR-γ activity for future development of PPAR-γ specific targeting drugs.
Collapse
Affiliation(s)
- Xiaoyun Zhao
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Liang Li
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hongxiang Sun
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaoli Jiang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wenjuan Bai
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ningbo Wu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jing Wang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Bing Su
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Key Laboratory of Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
21
|
Sun M, Tan Z, Lin K, Li X, Zhu J, Zhan L, Zheng H. Advanced Progression for the Heterogeneity and Homeostasis of Intestinal Stem Cells. Stem Cell Rev Rep 2023; 19:2109-2119. [PMID: 37351833 DOI: 10.1007/s12015-023-10578-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2023] [Indexed: 06/24/2023]
Abstract
Current understanding of the leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5) in intestinal stem cells (ISCs) is well established, however, the implications of ISC heterogeneity and homeostasis are poorly understood. Prior studies have provided important evidence for the association between heterogeneity of ISC pools with pathogenesis and therapeutic response of malignant disease. Leveraging the advantages of organoids and single cell RNA sequencing (scRNA-seq), glandular development has been simulated and cell heterogeneity has been clarified. Based on this research, several potential ISCs were identified, such as LGR5 + p27 + quiescent ISCs, LGR5 + Mex3a + slowly proliferating stem cells, and CLU + reverse stem cells. We also illustrated major factors responsible for ISC homeostasis including metabolism-related (LKB1, TGR5, HMGCS2), inflammation-related (IFB-b, IFN2, TNF), and Wnt signaling-related (CREPT, Mex3a, MTG16) factors. ISCs play complex roles in intestinal tumorigenesis, chemoresistance and occasional relapse of colon cancer, which bear discussion. In this review, we focus on novel technical challenges in ISCs fate drawing upon recent research with the goals of clarifying our understanding of complex ISCs, elucidating the integrated intestinal crypt niche, and creating new opportunities for therapeutic development.
Collapse
Affiliation(s)
- Minqiong Sun
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui, China
| | - Zhenya Tan
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui, China
| | - Keqiong Lin
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui, China
| | - Xiaofei Li
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui, China
| | - Jicheng Zhu
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui, China
| | - Li Zhan
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui, China
| | - Hong Zheng
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
22
|
Wiggins BG, Wang YF, Burke A, Grunberg N, Vlachaki Walker JM, Dore M, Chahrour C, Pennycook BR, Sanchez-Garrido J, Vernia S, Barr AR, Frankel G, Birdsey GM, Randi AM, Schiering C. Endothelial sensing of AHR ligands regulates intestinal homeostasis. Nature 2023; 621:821-829. [PMID: 37586410 PMCID: PMC10533400 DOI: 10.1038/s41586-023-06508-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/02/2023] [Indexed: 08/18/2023]
Abstract
Endothelial cells line the blood and lymphatic vasculature, and act as an essential physical barrier, control nutrient transport, facilitate tissue immunosurveillance and coordinate angiogenesis and lymphangiogenesis1,2. In the intestine, dietary and microbial cues are particularly important in the regulation of organ homeostasis. However, whether enteric endothelial cells actively sense and integrate such signals is currently unknown. Here we show that the aryl hydrocarbon receptor (AHR) acts as a critical node for endothelial cell sensing of dietary metabolites in adult mice and human primary endothelial cells. We first established a comprehensive single-cell endothelial atlas of the mouse small intestine, uncovering the cellular complexity and functional heterogeneity of blood and lymphatic endothelial cells. Analyses of AHR-mediated responses at single-cell resolution identified tissue-protective transcriptional signatures and regulatory networks promoting cellular quiescence and vascular normalcy at steady state. Endothelial AHR deficiency in adult mice resulted in dysregulated inflammatory responses and the initiation of proliferative pathways. Furthermore, endothelial sensing of dietary AHR ligands was required for optimal protection against enteric infection. In human endothelial cells, AHR signalling promoted quiescence and restrained activation by inflammatory mediators. Together, our data provide a comprehensive dissection of the effect of environmental sensing across the spectrum of enteric endothelia, demonstrating that endothelial AHR signalling integrates dietary cues to maintain tissue homeostasis by promoting endothelial cell quiescence and vascular normalcy.
Collapse
Affiliation(s)
- Benjamin G Wiggins
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK.
- MRC London Institute of Medical Sciences, London, UK.
| | - Yi-Fang Wang
- MRC London Institute of Medical Sciences, London, UK
| | - Alice Burke
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | - Nil Grunberg
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | - Julia M Vlachaki Walker
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | - Marian Dore
- MRC London Institute of Medical Sciences, London, UK
| | | | - Betheney R Pennycook
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | | | - Santiago Vernia
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | - Alexis R Barr
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | - Gad Frankel
- Department of Life Sciences, Imperial College London, London, UK
| | - Graeme M Birdsey
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Anna M Randi
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Chris Schiering
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK.
- MRC London Institute of Medical Sciences, London, UK.
| |
Collapse
|
23
|
Parker JB, Valencia C, Akras D, DiIorio SE, Griffin MF, Longaker MT, Wan DC. Understanding Fibroblast Heterogeneity in Form and Function. Biomedicines 2023; 11:2264. [PMID: 37626760 PMCID: PMC10452440 DOI: 10.3390/biomedicines11082264] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/09/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Historically believed to be a homogeneous cell type that is often overlooked, fibroblasts are more and more understood to be heterogeneous in nature. Though the mechanisms behind how fibroblasts participate in homeostasis and pathology are just beginning to be understood, these cells are believed to be highly dynamic and play key roles in fibrosis and remodeling. Focusing primarily on fibroblasts within the skin and during wound healing, we describe the field's current understanding of fibroblast heterogeneity in form and function. From differences due to embryonic origins to anatomical variations, we explore the diverse contributions that fibroblasts have in fibrosis and plasticity. Following this, we describe molecular techniques used in the field to provide deeper insights into subpopulations of fibroblasts and their varied roles in complex processes such as wound healing. Limitations to current work are also discussed, with a focus on future directions that investigators are recommended to take in order to gain a deeper understanding of fibroblast biology and to develop potential targets for translational applications in a clinical setting.
Collapse
Affiliation(s)
- Jennifer B. Parker
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA (M.F.G.)
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Caleb Valencia
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA (M.F.G.)
| | - Deena Akras
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA (M.F.G.)
| | - Sarah E. DiIorio
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA (M.F.G.)
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michelle F. Griffin
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA (M.F.G.)
| | - Michael T. Longaker
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA (M.F.G.)
| | - Derrick C. Wan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA (M.F.G.)
| |
Collapse
|
24
|
Ding C, Yu Z, Sefik E, Zhou J, Kaffe E, Wang G, Li B, Flavell RA, Hu W, Ye Y, Li HB. A T reg-specific long noncoding RNA maintains immune-metabolic homeostasis in aging liver. NATURE AGING 2023; 3:813-828. [PMID: 37277640 DOI: 10.1038/s43587-023-00428-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 04/28/2023] [Indexed: 06/07/2023]
Abstract
Regulatory T (Treg) cells modulate several aging-related liver diseases. However, the molecular mechanisms regulating Treg function in this context are unknown. Here we identified a long noncoding RNA, Altre (aging liver Treg-expressed non-protein-coding RNA), which was specifically expressed in the nucleus of Treg cells and increased with aging. Treg-specific deletion of Altre did not affect Treg homeostasis and function in young mice but caused Treg metabolic dysfunction, inflammatory liver microenvironment, liver fibrosis and liver cancer in aged mice. Depletion of Altre reduced Treg mitochondrial integrity and respiratory capacity, and induced reactive oxygen species accumulation, thus increasing intrahepatic Treg apoptosis in aged mice. Moreover, lipidomic analysis identified a specific lipid species driving Treg aging and apoptosis in the aging liver microenvironment. Mechanistically, Altre interacts with Yin Yang 1 to orchestrate its occupation on chromatin, thereby regulating the expression of a group of mitochondrial genes, and maintaining optimal mitochondrial function and Treg fitness in the liver of aged mice. In conclusion, the Treg-specific nuclear long noncoding RNA Altre maintains the immune-metabolic homeostasis of the aged liver through Yin Yang 1-regulated optimal mitochondrial function and the Treg-sustained liver immune microenvironment. Thus, Altre is a potential therapeutic target for the treatment of liver diseases affecting older adults.
Collapse
Affiliation(s)
- Chenbo Ding
- Medical Center on Aging, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhibin Yu
- Medical Center on Aging, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Esen Sefik
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
| | - Jing Zhou
- Medical Center on Aging, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Eleanna Kaffe
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Gaoyang Wang
- Medical Center on Aging, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Li
- Medical Center on Aging, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
| | - Weiguo Hu
- Department of Geriatrics, Medical Center on Aging of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Youqiong Ye
- Medical Center on Aging, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hua-Bing Li
- Medical Center on Aging, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Department of Geriatrics, Medical Center on Aging of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Chongqing International Institute for Immunology, Chongqing, China.
| |
Collapse
|
25
|
Bai X, Mei L, Shi Y, Huang H, Guo Y, Liang C, Yang M, Wu R, Zhang Y, Chen Q. The Cellular Mechanism of Acupuncture for Ulcerative Colitis based on the Communication of Telocytes. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2023; 29:1190-1204. [PMID: 37749671 DOI: 10.1093/micmic/ozad028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/16/2023] [Accepted: 02/21/2023] [Indexed: 09/27/2023]
Abstract
Acupuncture can ameliorate or treat diseases according to the meridian theory in traditional Chinese medicine (TCM); however, its mechanism has not been scientifically clarified. On the other hand, telocytes (TCs) are morphologically in accordance with the meridian system, which needs further cytological investigations and acupuncture confirmation. The present study showed that acupuncture could activate TCs in several ways, alleviating rabbit ulcerative colitis. TCs could cytologically communicate the acupoints, the acupuncture sites in skin with their corresponding large intestine by TC homo-cellular junctions, exosomes around TCs, and TC-mediated nerves or blood vessels. TCs expressed transient receptor potential vanilloid type 4, the mechanosensitive channel protein that can transduce the mechanical stimulation of acupuncture into biochemical signals transferring along the extremely thin and long TCs. Collectively, a cellular mechanism diagram of acupuncture was concluded based on TC characteristics. Those results also confirmed the viewpoint that TCs were the key cells of meridian essence in TCM.
Collapse
Affiliation(s)
- Xuebing Bai
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Lu Mei
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Yonghong Shi
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- Chinese Academy of Agricultural Sciences, Shanghai Veterinary Institute, Shanghai 200241, China
| | - Haixiang Huang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Yanna Guo
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Chunhua Liang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Min Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Ruizhi Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Yingxin Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Qiusheng Chen
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| |
Collapse
|
26
|
Pærregaard SI, Wulff L, Schussek S, Niss K, Mörbe U, Jendholm J, Wendland K, Andrusaite AT, Brulois KF, Nibbs RJB, Sitnik K, Mowat AM, Butcher EC, Brunak S, Agace WW. The small and large intestine contain related mesenchymal subsets that derive from embryonic Gli1 + precursors. Nat Commun 2023; 14:2307. [PMID: 37085516 PMCID: PMC10121680 DOI: 10.1038/s41467-023-37952-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 04/06/2023] [Indexed: 04/23/2023] Open
Abstract
The intestinal lamina propria contains a diverse network of fibroblasts that provide key support functions to cells within their local environment. Despite this, our understanding of the diversity, location and ontogeny of fibroblasts within and along the length of the intestine remains incomplete. Here we show that the small and large intestinal lamina propria contain similar fibroblast subsets that locate in specific anatomical niches. Nevertheless, we find that the transcriptional profile of similar fibroblast subsets differs markedly between the small intestine and colon suggesting region specific functions. We perform in vivo transplantation and lineage-tracing experiments to demonstrate that adult intestinal fibroblast subsets, smooth muscle cells and pericytes derive from Gli1-expressing precursors present in embryonic day 12.5 intestine. Trajectory analysis of single cell RNA-seq datasets of E12.5 and adult mesenchymal cells suggest that adult smooth muscle cells and fibroblasts derive from distinct embryonic intermediates and that adult fibroblast subsets develop in a linear trajectory from CD81+ fibroblasts. Finally, we provide evidence that colonic subepithelial PDGFRαhi fibroblasts comprise several functionally distinct populations that originate from an Fgfr2-expressing fibroblast intermediate. Our results provide insights into intestinal stromal cell diversity, location, function, and ontogeny, with implications for intestinal development and homeostasis.
Collapse
Affiliation(s)
- Simone Isling Pærregaard
- Department of Health Technology, Technical University of Denmark, Kemitorvet, 2800 Kgs, Lyngby, Denmark
| | - Line Wulff
- Department of Health Technology, Technical University of Denmark, Kemitorvet, 2800 Kgs, Lyngby, Denmark
| | - Sophie Schussek
- Department of Health Technology, Technical University of Denmark, Kemitorvet, 2800 Kgs, Lyngby, Denmark
| | - Kristoffer Niss
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Urs Mörbe
- Department of Health Technology, Technical University of Denmark, Kemitorvet, 2800 Kgs, Lyngby, Denmark
| | - Johan Jendholm
- Department of Health Technology, Technical University of Denmark, Kemitorvet, 2800 Kgs, Lyngby, Denmark
| | | | - Anna T Andrusaite
- Institute of Infection, immunity and Inflammation, University of Glasgow, Glasgow, Scotland, UK
| | - Kevin F Brulois
- Laboratory of Immunology and Vascular Biology, Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Robert J B Nibbs
- Institute of Infection, immunity and Inflammation, University of Glasgow, Glasgow, Scotland, UK
| | - Katarzyna Sitnik
- Department of Health Technology, Technical University of Denmark, Kemitorvet, 2800 Kgs, Lyngby, Denmark
| | - Allan McI Mowat
- Institute of Infection, immunity and Inflammation, University of Glasgow, Glasgow, Scotland, UK
| | - Eugene C Butcher
- Laboratory of Immunology and Vascular Biology, Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA
- The Center for Molecular Biology and Medicine, Veterans Affairs Palo Alto Health Care System and the Palo Alto Veterans Institute for Research (PAVIR), Palo Alto, CA, USA
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - William W Agace
- Department of Health Technology, Technical University of Denmark, Kemitorvet, 2800 Kgs, Lyngby, Denmark.
- Immunology Section, Lund University, Lund, 221 84, Sweden.
| |
Collapse
|
27
|
Tusa I, Menconi A, Tubita A, Rovida E. Pathophysiological Impact of the MEK5/ERK5 Pathway in Oxidative Stress. Cells 2023; 12:cells12081154. [PMID: 37190064 DOI: 10.3390/cells12081154] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/22/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Oxidative stress regulates many physiological and pathological processes. Indeed, a low increase in the basal level of reactive oxygen species (ROS) is essential for various cellular functions, including signal transduction, gene expression, cell survival or death, as well as antioxidant capacity. However, if the amount of generated ROS overcomes the antioxidant capacity, excessive ROS results in cellular dysfunctions as a consequence of damage to cellular components, including DNA, lipids and proteins, and may eventually lead to cell death or carcinogenesis. Both in vitro and in vivo investigations have shown that activation of the mitogen-activated protein kinase kinase 5/extracellular signal-regulated kinase 5 (MEK5/ERK5) pathway is frequently involved in oxidative stress-elicited effects. In particular, accumulating evidence identified a prominent role of this pathway in the anti-oxidative response. In this respect, activation of krüppel-like factor 2/4 and nuclear factor erythroid 2-related factor 2 emerged among the most frequent events in ERK5-mediated response to oxidative stress. This review summarizes what is known about the role of the MEK5/ERK5 pathway in the response to oxidative stress in pathophysiological contexts within the cardiovascular, respiratory, lymphohematopoietic, urinary and central nervous systems. The possible beneficial or detrimental effects exerted by the MEK5/ERK5 pathway in the above systems are also discussed.
Collapse
Affiliation(s)
- Ignazia Tusa
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Alessio Menconi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Alessandro Tubita
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Elisabetta Rovida
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| |
Collapse
|
28
|
Hsu NY, Nayar S, Gettler K, Talware S, Giri M, Alter I, Argmann C, Sabic K, Thin TH, Ko HBM, Werner R, Tastad C, Stappenbeck T, Azabdaftari A, Uhlig HH, Chuang LS, Cho JH. NOX1 is essential for TNFα-induced intestinal epithelial ROS secretion and inhibits M cell signatures. Gut 2023; 72:654-662. [PMID: 36191961 PMCID: PMC9998338 DOI: 10.1136/gutjnl-2021-326305] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 08/20/2022] [Indexed: 12/08/2022]
Abstract
OBJECTIVE Loss-of-function mutations in genes generating reactive oxygen species (ROS), such as NOX1, are associated with IBD. Mechanisms whereby loss of ROS drive IBD are incompletely defined. DESIGN ROS measurements and single-cell transcriptomics were performed on colonoids stratified by NOX1 genotype and TNFα stimulation. Clustering of epithelial cells from human UC (inflamed and uninflamed) scRNASeq was performed. Validation of M cell induction was performed by immunohistochemistry using UEA1 (ulex europaeus agglutin-1 lectin) and in vivo with DSS injury. RESULTS TNFα induces ROS production more in NOX1-WT versus NOX1-deficient murine colonoids under a range of Wnt-mediated and Notch-mediated conditions. scRNASeq from inflamed and uninflamed human colitis versus TNFα stimulated, in vitro colonoids defines substantially shared, induced transcription factors; NOX1-deficient colonoids express substantially lower levels of STAT3 (signal transducer and activator of transcription 3), CEBPD (CCAAT enhancer-binding protein delta), DNMT1 (DNA methyltransferase) and HIF1A (hypoxia-inducible factor) baseline. Subclustering unexpectedly showed marked TNFα-mediated induction of M cells (sentinel cells overlying lymphoid aggregates) in NOX1-deficient colonoids. M cell induction by UEA1 staining is rescued with H2O2 and paraquat, defining extra- and intracellular ROS roles in maintenance of LGR5+ stem cells. DSS injury demonstrated GP2 (glycoprotein-2), basal lymphoplasmacytosis and UEA1 induction in NOX1-deficiency. Principal components analyses of M cell genes and decreased DNMT1 RNA velocity correlate with UC inflammation. CONCLUSIONS NOX1 deficiency plus TNFα stimulation contribute to colitis through dysregulation of the stem cell niche and altered cell differentiation, enhancing basal lymphoplasmacytosis. Our findings prioritise ROS modulation for future therapies.
Collapse
Affiliation(s)
- Nai-Yun Hsu
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Shikha Nayar
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kyle Gettler
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Sayali Talware
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai Department of Medicine, New York, New York, USA
- The Icahn Genomic Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Mamta Giri
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Isaac Alter
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Carmen Argmann
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ksenija Sabic
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Tin Htwe Thin
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Huai-Bin Mabel Ko
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, USA
| | - Robert Werner
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Christopher Tastad
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Thaddeus Stappenbeck
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland, Ohio, USA
| | - Aline Azabdaftari
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Holm H Uhlig
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Ling-Shiang Chuang
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Judy H Cho
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
29
|
Li Z, Dong S, Huang S, Sun Y, Sun Y, Zhao B, Qi Q, Xiong L, Hong F, Jiang Y. Role of CD34 in inflammatory bowel disease. Front Physiol 2023; 14:1144980. [PMID: 37051017 PMCID: PMC10083274 DOI: 10.3389/fphys.2023.1144980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 03/15/2023] [Indexed: 03/29/2023] Open
Abstract
Inflammatory bowel disease (IBD) is caused by a variety of pathogenic factors, including chronic recurrent inflammation of the ileum, rectum, and colon. Immune cells and adhesion molecules play an important role in the course of the disease, which is actually an autoimmune disease. During IBD, CD34 is involved in mediating the migration of a variety of immune cells (neutrophils, eosinophils, and mast cells) to the inflammatory site, and its interaction with various adhesion molecules is involved in the occurrence and development of IBD. Although the function of CD34 as a partial cell marker is well known, little is known on its role in IBD. Therefore, this article describes the structure and biological function of CD34, as well as on its potential mechanism in the development of IBD.
Collapse
Affiliation(s)
- Zhiyuan Li
- Jiaxing Key Laboratory of Virus-Related Infectious Diseases, The Affiliated Hospital of Jiaxing University, Jiaxing University College of Medicine, Jiaxing, Zhejiang, China
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui, China
| | - Shuyan Dong
- Jiaxing Key Laboratory of Virus-Related Infectious Diseases, The Affiliated Hospital of Jiaxing University, Jiaxing University College of Medicine, Jiaxing, Zhejiang, China
| | - Shichen Huang
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui, China
| | - Yuhan Sun
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui, China
| | - Yingzhi Sun
- Jiaxing Key Laboratory of Virus-Related Infectious Diseases, The Affiliated Hospital of Jiaxing University, Jiaxing University College of Medicine, Jiaxing, Zhejiang, China
| | - Beibei Zhao
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui, China
| | - Qiulan Qi
- Jiaxing Key Laboratory of Virus-Related Infectious Diseases, The Affiliated Hospital of Jiaxing University, Jiaxing University College of Medicine, Jiaxing, Zhejiang, China
| | - Lei Xiong
- Department of Biochemistry and Molecular Biology, Wannan Medical College, Wuhu, Anhui, China
- *Correspondence: Yuxin Jiang, ; Feng Hong, ; Lei Xiong,
| | - Feng Hong
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- *Correspondence: Yuxin Jiang, ; Feng Hong, ; Lei Xiong,
| | - Yuxin Jiang
- Jiaxing Key Laboratory of Virus-Related Infectious Diseases, The Affiliated Hospital of Jiaxing University, Jiaxing University College of Medicine, Jiaxing, Zhejiang, China
- *Correspondence: Yuxin Jiang, ; Feng Hong, ; Lei Xiong,
| |
Collapse
|
30
|
Wang G, Lin Z, Li Y, Chen L, Reddy SK, Hu Z, Garza LA. Colonizing microbiota is associated with clinical outcomes in diabetic wound healing. Adv Drug Deliv Rev 2023; 194:114727. [PMID: 36758858 PMCID: PMC10163681 DOI: 10.1016/j.addr.2023.114727] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 02/02/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023]
Abstract
With the development of society and the improvement of life quality, more than 500 million people are affected by diabetes. More than 10 % of people with diabetes will suffer from diabetic wounds, and 80 % of diabetic wounds will reoccur, so the development of new diabetic wound treatments is of great importance. The development of skin microbe research technology has gradually drawn people's attention to the complex relationship between microbes and diabetic wounds. Many studies have shown that skin microbes are associated with the outcome of diabetic wounds and can even be used as one of the indicators of wound prognosis. Skin microbes have also been found to have the potential to treat diabetic wounds. The wound colonization of different bacteria can exert opposing therapeutic effects. It is necessary to fully understand the skin microbes in diabetic wounds, which can provide valuable guidance for clinical diabetic wound treatment.
Collapse
Affiliation(s)
- Gaofeng Wang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China; Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA.
| | - Zhen Lin
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Yue Li
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Lu Chen
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Sashank K Reddy
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA; Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Zhiqi Hu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - L A Garza
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA; Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA.
| |
Collapse
|
31
|
Qi-Long-Tian capsule alleviates pulmonary fibrosis development by modulating inflammatory response and gut microbiota. Funct Integr Genomics 2023; 23:64. [PMID: 36810971 DOI: 10.1007/s10142-023-00988-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/24/2023]
Abstract
Pulmonary fibrosis (PF) is a chronic, progressive, and fibrotic interstitial lung disease with a high mortality rate. Qi-Long-Tian (QLT) capsule is an herbal formula with great potential for antifibrotic effects, consisting of San Qi (Notoginseng Radix et Rhizoma), Di Long [Pheretima aspergillum (E. Perrier)], and Hong Jingtian (Rhodiolae Crenulatae Radix et Rhizoma), and has been used in clinical practice for many years. To explore the relationship between the effects of Qi-Long-Tian capsule and gut microbiota of PF mice, pulmonary fibrosis model were established by tracheal drip injection of bleomycin. Thirty-six mice were randomly divided into 6 groups: control group (control), model group (model), QLT capsule low dose group (QL), QLT capsule medium dose group (QM), QLT capsule high dose group (QH), and pirfenidone group (PFD). After 21 days of treatment, after pulmonary function tests, the lung tissues, serums, and enterobacterial samples were collected for further analysis. HE staining and Masson's staining were used to detect changes as the main indicators of PF in each group, and the expression of hydroxyproline (HYP) related to collagen metabolism was detected by and alkaline hydrolysis method. qRT-PCR and ELISA were used to detect the mRNA and protein expressions of pro-inflammatory factors include interleukin 1β (IL-1β), interleukin 6 (IL-6), transforming growth factor β1 (TGF-β1), tumor necrosis factor α (TNF-α) in lung tissues and serums, and the inflammation-mediating factors include tight junction protein (ZO-1, Claudin, Occludin). ELISA was used to detect the protein expressions of secretory immunoglobulin A (sIgA), short-chain fatty acids (SCFAs), and lipopolysaccharide (LPS) in colonic tissues. 16sRNA gene sequencing was used to detect changes in the abundance and diversity of intestinal flora in the control, model, and QM groups, to search for differential genera, and analyze the correlation with inflammatory factors. QLT capsule effectively improved the status of pulmonary fibrosis and reduced HYP. In addition, QLT capsule significantly reduced the abnormal levels of pro-inflammatory factors, including IL-1β, IL-6, TNF-α, and TGF-β in lung tissue and serum, while improving the levels of pro-inflammatory related factors ZO-1, Claudin, Occludin, sIgA, SCFAs, and reducing LPS in the colon. The comparison between the alpha diversity and beta diversity in enterobacteria suggested that the composition of the gut flora in the control, model, and QLT capsule groups were different. QLT capsule significantly increased the relative abundance of Bacteroidia (which might limit the onset of inflammation) and decreased the relative abundance of Clostridia (which might promote inflammation). In addition, these two enterobacteria were closely associated with pro-inflammatory-related indicators and pro-inflammatory factors in PF. All these results suggest that QLT capsule intervenes in pulmonary fibrosis by regulating the differential genera of intestinal flora, increasing immunoglobulin secretion, repairing the intestinal mucosal barrier, reducing LPS entry into the blood, and decreasing inflammatory factor secretion in the serum, which in turn alleviates pulmonary inflammation. This study clarifies the therapeutic mechanism of QLT capsule in PF and provides a theoretical basis for it. It provides a theoretical basis for its further clinical application.
Collapse
|
32
|
Stromal regulation of the intestinal barrier. Mucosal Immunol 2023; 16:221-231. [PMID: 36708806 DOI: 10.1016/j.mucimm.2023.01.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/27/2022] [Accepted: 01/12/2023] [Indexed: 01/26/2023]
Abstract
The intestinal barrier is a complex structure that allows the absorption of nutrients while ensuring protection against intestinal pathogens and balanced immunity. The development and maintenance of a functional intestinal barrier is a multifactorial process that is only partially understood. Here we review novel findings on the emerging role of mesenchymal cells in this process using insights gained from lineage tracing approaches, Cre-based gene deletion, and single-cell transcriptomics. The current evidence points toward a key organizer role for distinct mesenchymal lineages in intestinal development and homeostasis, regulating both epithelial and immune components of the intestinal barrier. We further discuss recent findings on functional mesenchymal heterogeneity and implications for intestinal regeneration and inflammatory intestinal pathologies.
Collapse
|
33
|
Valatas V, Kitamura K, Ward SG, Kolios G. Editorial: Stromal and immune cell interactions in intestinal inflammation and fibrosis. Front Immunol 2023; 14:1152140. [PMID: 36875109 PMCID: PMC9978737 DOI: 10.3389/fimmu.2023.1152140] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 02/09/2023] [Indexed: 02/18/2023] Open
Affiliation(s)
- Vassilis Valatas
- Department of Gastroenterology, University Hospital of Heraklion, Heraklion, Greece
| | - Kazuya Kitamura
- Department of Gastroenterology, Tonami General Hospital, Tonami, Japan
| | - Stephen G Ward
- Department of Life Sciences, University of Bath, Bath, United Kingdom
| | - George Kolios
- Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| |
Collapse
|
34
|
Cheng L, Wu H, Chen Z, Hao H, Zheng X. Gut microbiome at the crossroad of genetic variants and behavior disorders. Gut Microbes 2023; 15:2201156. [PMID: 37089016 PMCID: PMC10128504 DOI: 10.1080/19490976.2023.2201156] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 04/03/2023] [Indexed: 04/25/2023] Open
Abstract
Genetic variants are traditionally known to shape the susceptibility to neuropsychiatric disorders. An increasing number of studies indicate that remodeling of the gut microbiome by genetic variance serves as a versatile regulator of gut-brain crosstalk and behavior. Evidence also emerges that certain behavioral symptoms are specifically attributed to gut microbial remodeling and gut-to-brain signals, which necessitates rethinking of neuropsychiatric disease etiology and treatment from a systems perspective of reciprocal gene-microbe interactions. Here, we present an emerging picture of how gut microbes and host genetics interactively shape complex psychiatric phenotypes. We illustrate the growing understanding of how the gut microbiome is shaped by genetic changes and its connection to behavioral outcome. We also discuss working strategies and open questions in translating associative gene-microbiome-behavior findings into causal links and novel targets for neurobehavioral disorders. Dual targeting of the genetic and microbial factors may expand the space of drug discovery for neuropsychiatric diseases.
Collapse
Affiliation(s)
- Lingsha Cheng
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
- Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, Jiangsu Province Key Laboratory of Drug Metabolism, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Haoqian Wu
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
- Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, Jiangsu Province Key Laboratory of Drug Metabolism, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Zhuo Chen
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
- Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, Jiangsu Province Key Laboratory of Drug Metabolism, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Haiping Hao
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
- Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, Jiangsu Province Key Laboratory of Drug Metabolism, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Xiao Zheng
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
- Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, Jiangsu Province Key Laboratory of Drug Metabolism, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
35
|
Sun H, Tan J, Chen H, Wu N, Su B. Immune niches orchestrated by intestinal mesenchymal stromal cells lining the crypt-villus. Front Immunol 2022; 13:1057932. [PMID: 36405734 PMCID: PMC9669707 DOI: 10.3389/fimmu.2022.1057932] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 10/20/2022] [Indexed: 07/22/2023] Open
Abstract
The mammalian intestine is an organ that can be spatially defined by two axes: longitudinal and vertical. Such anatomical structure ensures the maintenance of a relatively immuno-quiescent and proliferation-promoting crypt for intestinal stem cell differentiation while actively warding off the invading intestinal microbes at the villus tip during digestion and nutrient absorption. Such behavior is achieved by the fine coordination among intestinal epithelial cells, intestinal mesenchymal stromal cells and tissue-resident immune cells like myeloid cells and lymphocytes. Among these cell types resided in the colon, intestinal mesenchymal stromal cells are considered to be the essential link between epithelium, vasculature, neuronal system, and hematopoietic compartment. Recent advancement of single cell and spatial transcriptomics has enabled us to characterize the spatial and functional heterogeneity of intestinal mesenchymal stromal cells. These studies reveal distinctive intestinal mesenchymal stromal cells localized in different regions of the intestine with diverse functions including but not limited to providing cytokines and growth factors essential for different immune cells and epithelial cells which predict niche formation for immune function from the villus tip to the crypt bottom. In this review, we aim to provide an overall view of the heterogeneity of intestinal mesenchymal stromal cells, the spatial distribution of these cells along with their interaction with immune cells and the potential regulatory cytokine profile of these cell types. Summarization of such information may enrich our current understanding of the immuno-regulatory functions of the newly identified mesenchymal stromal cell subsets beyond their epithelial regulatory function.
Collapse
Affiliation(s)
- Hongxiang Sun
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Gastroenterology, Center for Immune-Related Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Jiao Tong University School of Medicine–Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianmei Tan
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Gastroenterology, Center for Immune-Related Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Jiao Tong University School of Medicine–Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongqian Chen
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Gastroenterology, Center for Immune-Related Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Jiao Tong University School of Medicine–Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ningbo Wu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Gastroenterology, Center for Immune-Related Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Jiao Tong University School of Medicine–Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bing Su
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Gastroenterology, Center for Immune-Related Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Jiao Tong University School of Medicine–Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
36
|
Li H, Wang N, Jiang Y, Wang H, Xin Z, An H, Pan H, Ma W, Zhang T, Wang X, Lin W. E3
ubiquitin ligase
NEDD4L
negatively regulates inflammation by promoting ubiquitination of
MEKK2. EMBO Rep 2022; 23:e54603. [PMID: 36161689 PMCID: PMC9638856 DOI: 10.15252/embr.202254603] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 07/25/2022] [Accepted: 08/19/2022] [Indexed: 11/26/2022] Open
Abstract
Aberrant activation of inflammation signaling triggered by tumor necrosis factor α (TNF‐α), interleukin‐1 (IL‐1), and interleukin‐17 (IL‐17) is associated with immunopathology. Here, we identify neural precursor cells expressed developmentally down‐regulated gene 4‐like (NEDD4L), a HECT type E3 ligase, as a common negative regulator of signaling induced by TNF‐α, IL‐1, and IL‐17. NEDD4L modulates the degradation of mitogen‐activated protein kinase kinase kinase 2 (MEKK2) via constitutively and directly binding to MEKK2 and promotes its poly‐ubiquitination. In interleukin‐17 receptor (IL‐17R) signaling, Nedd4l knockdown or deficiency enhances IL‐17‐induced p38 and NF‐κB activation and the production of proinflammatory cytokines and chemokines in a MEKK2‐dependent manner. We further show that IL‐17‐induced MEKK2 Ser520 phosphorylation is required not only for downstream p38 and NF‐κB activation but also for NEDD4L‐mediated MEKK2 degradation and the subsequent shutdown of IL‐17R signaling. Importantly, Nedd4l‐deficient mice show increased susceptibility to IL‐17‐induced inflammation and aggravated symptoms of experimental autoimmune encephalomyelitis (EAE) in IL‐17R signaling‐dependent manner. These data suggest that NEDD4L acts as an inhibitor of IL‐17R signaling, which ameliorates the pathogenesis of IL‐17‐mediated autoimmune diseases.
Collapse
Affiliation(s)
- Hui Li
- Institute of Immunology and Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine Zhejiang China
- Department of Medical Oncology The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital) Hangzhou China
- Institute of Basic Medicine and Cancer (IBMC) Chinese Academy of Sciences Hangzhou China
| | - Ning Wang
- Institute of Immunology and Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine Zhejiang China
| | - Yu Jiang
- Institute of Immunology and Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine Zhejiang China
| | - Haofei Wang
- Institute of Immunology and Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine Zhejiang China
| | - Zengfeng Xin
- Institute of Immunology and Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine Zhejiang China
| | - Huazhang An
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital Jinan China
| | - Hao Pan
- Department of Urology, The First Affiliated Hospital, College of Medicine Zhejiang University Hangzhou China
| | - Wangqian Ma
- Department of Gastroenterology, The Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou China
| | - Ting Zhang
- Department of Radiation Oncology, The Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou China
| | - Xiaojian Wang
- Institute of Immunology and Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine Zhejiang China
| | - Wenlong Lin
- Institute of Immunology and Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine Zhejiang China
| |
Collapse
|
37
|
Colozza G, Park SY, Koo BK. Clone wars: From molecules to cell competition in intestinal stem cell homeostasis and disease. Exp Mol Med 2022; 54:1367-1378. [PMID: 36117218 PMCID: PMC9534868 DOI: 10.1038/s12276-022-00854-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/27/2022] [Accepted: 07/19/2022] [Indexed: 11/14/2022] Open
Abstract
The small intestine is among the fastest self-renewing tissues in adult mammals. This rapid turnover is fueled by the intestinal stem cells residing in the intestinal crypt. Wnt signaling plays a pivotal role in regulating intestinal stem cell renewal and differentiation, and the dysregulation of this pathway leads to cancer formation. Several studies demonstrate that intestinal stem cells follow neutral drift dynamics, as they divide symmetrically to generate other equipotent stem cells. Competition for niche space and extrinsic signals in the intestinal crypt is the governing mechanism that regulates stemness versus cell differentiation, but the underlying molecular mechanisms are still poorly understood, and it is not yet clear how this process changes during disease. In this review, we highlight the mechanisms that regulate stem cell homeostasis in the small intestine, focusing on Wnt signaling and its regulation by RNF43 and ZNRF3, key inhibitors of the Wnt pathway. Furthermore, we summarize the evidence supporting the current model of intestinal stem cell regulation, highlighting the principles of neutral drift at the basis of intestinal stem cell homeostasis. Finally, we discuss recent studies showing how cancer cells bypass this mechanism to gain a competitive advantage against neighboring normal cells. Stem cells in the gut rapidly renew themselves through processes that cancer cells co-opt to trigger tumor development. Gabriele Colozza from the Institute of Molecular Biotechnology in Vienna, Austria, and colleagues review how a network of critical molecular signals and competition for limited space help to regulate the dynamics of stem cells in the intestines. The correct balance between self-renewal and differentiation is tightly controlled by the so-called Wnt signaling pathway and its inhibitors. Competition between dividing cells in the intestinal crypts, the locations between finger-like protrusions in the gut where stem cells are found, provides another protective mechanism against runaway stem cell growth. However, intestinal cancer cells, thanks to their activating mutations, bypass these safeguards to gain a survival advantage. Drugs that target these ‘super-competitive’ behaviors could therefore help combat tumor proliferation.
Collapse
|
38
|
Wang Y, Song W, Yu S, Liu Y, Chen YG. Intestinal cellular heterogeneity and disease development revealed by single-cell technology. CELL REGENERATION 2022; 11:26. [PMID: 36045190 PMCID: PMC9433512 DOI: 10.1186/s13619-022-00127-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 07/15/2022] [Indexed: 11/10/2022]
Abstract
The intestinal epithelium is responsible for food digestion and nutrient absorption and plays a critical role in hormone secretion, microorganism defense, and immune response. These functions depend on the integral single-layered intestinal epithelium, which shows diversified cell constitution and rapid self-renewal and presents powerful regeneration plasticity after injury. Derailment of homeostasis of the intestine epithelium leads to the development of diseases, most commonly including enteritis and colorectal cancer. Therefore, it is important to understand the cellular characterization of the intestinal epithelium at the molecular level and the mechanisms underlying its homeostatic maintenance. Single-cell technologies allow us to gain molecular insights at the single-cell level. In this review, we summarize the single-cell RNA sequencing applications to understand intestinal cell characteristics, spatiotemporal evolution, and intestinal disease development.
Collapse
|
39
|
Chalkidi N, Paraskeva C, Koliaraki V. Fibroblasts in intestinal homeostasis, damage, and repair. Front Immunol 2022; 13:924866. [PMID: 36032088 PMCID: PMC9399414 DOI: 10.3389/fimmu.2022.924866] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 07/20/2022] [Indexed: 12/02/2022] Open
Abstract
The mammalian intestine is a self-renewing tissue that ensures nutrient absorption while acting as a barrier against environmental insults. This is achieved by mature intestinal epithelial cells, the renewing capacity of intestinal stem cells at the base of the crypts, the development of immune tolerance, and the regulatory functions of stromal cells. Upon intestinal injury or inflammation, this tightly regulated mucosal homeostasis is disrupted and is followed by a series of events that lead to tissue repair and the restoration of organ function. It is now well established that fibroblasts play significant roles both in the maintenance of epithelial and immune homeostasis in the intestine and the response to tissue damage mainly through the secretion of a variety of soluble mediators and ligands and the remodeling of the extracellular matrix. In addition, recent advances in single-cell transcriptomics have revealed an unexpected heterogeneity of fibroblasts that comprise distinct cell subsets in normal and inflammatory conditions, indicative of diverse functions. However, there is still little consensus on the number, terminology, and functional properties of these subsets. Moreover, it is still unclear how individual fibroblast subsets can regulate intestinal repair processes and what is their impact on the pathogenesis of inflammatory bowel disease. In this mini-review, we aim to provide a concise overview of recent advances in the field, that we believe will help clarify current concepts on fibroblast heterogeneity and functions and advance our understanding of the contribution of fibroblasts in intestinal damage and repair.
Collapse
|
40
|
Arconada-Luque E, Jiménez-Suarez J, Pascual-Serra R, Nam-Cha SH, Moline T, Cimas FJ, Fliquete G, Ortega-Muelas M, Roche O, Fernández-Aroca DM, Muñoz Velasco R, García-Flores N, Garnés-García C, Sánchez-Fdez A, Matilla-Almazán S, Sánchez-Arévalo Lobo VJ, Hernández-Losa J, Belandia B, Pandiella A, Esparís-Ogando A, Ramón y Cajal S, del Peso L, Sánchez-Prieto R, Ruiz-Hidalgo MJ. ERK5 Is a Major Determinant of Chemical Sarcomagenesis: Implications in Human Pathology. Cancers (Basel) 2022; 14:cancers14143509. [PMID: 35884568 PMCID: PMC9316148 DOI: 10.3390/cancers14143509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/11/2022] [Accepted: 07/16/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Sarcoma is a heterogeneous group of tumors poorly studied with few therapeutic opportunities. Interestingly, the role of MAPKs still remains unclear in sarcomatous pathology. Here, we describe for the first time the critical role of ERK5 in the biology of soft tissue sarcoma by using in vitro and in vivo approaches in a murine experimental model of chemical sarcomagenesis. Indeed, our observations were extrapolated to a short series of human leiomyosarcoma and rhabdomyosarcomas. Furthermore, transcriptome analysis allows us to demonstrate the critical role of KLF2 in the biological effects of ERK5. Therefore, the data presented here open new windows in the diagnosis and therapy of soft tissue sarcomas. Abstract Sarcomas are a heterogeneous group of tumors in which the role of ERK5 is poorly studied. To clarify the role of this MAPK in sarcomatous pathology, we used a murine 3-methyl-cholanthrene (3MC)-induced sarcoma model. Our data show that 3MC induces pleomorphic sarcomas with muscle differentiation, showing an increased expression of ERK5. Indeed, this upregulation was also observed in human sarcomas of muscular origin, such as leiomyosarcoma or rhabdomyosarcoma. Moreover, in cell lines derived from these 3MC-induced tumors, abrogation of Mapk7 expression by using specific shRNAs decreased in vitro growth and colony-forming capacity and led to a marked loss of tumor growth in vivo. In fact, transcriptomic profiling in ERK5 abrogated cell lines by RNAseq showed a deregulated gene expression pattern for key biological processes such as angiogenesis, migration, motility, etc., correlating with a better prognostic in human pathology. Finally, among the various differentially expressed genes, Klf2 is a key mediator of the biological effects of ERK5 as indicated by its specific interference, demonstrating that the ERK5–KLF2 axis is an important determinant of sarcoma biology that should be further studied in human pathology.
Collapse
Affiliation(s)
- Elena Arconada-Luque
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (E.A.-L.); (J.J.-S.); (R.P.-S.); (M.O.-M.); (O.R.); (D.M.F.-A.); (N.G.-F.); (C.G.-G.); (M.J.R.-H.)
| | - Jaime Jiménez-Suarez
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (E.A.-L.); (J.J.-S.); (R.P.-S.); (M.O.-M.); (O.R.); (D.M.F.-A.); (N.G.-F.); (C.G.-G.); (M.J.R.-H.)
| | - Raquel Pascual-Serra
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (E.A.-L.); (J.J.-S.); (R.P.-S.); (M.O.-M.); (O.R.); (D.M.F.-A.); (N.G.-F.); (C.G.-G.); (M.J.R.-H.)
| | - Syong Hyun Nam-Cha
- Servicio de Anatomía Patológica, Hospital General de Albacete, 02008 Albacete, Spain;
| | - Teresa Moline
- Grupo de Patología Molecular Traslacional, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona Centro de Investigación Biomédica en RED de Cancer CIBERONC, 08035 Barcelona, Spain; (T.M.); (G.F.); (J.H.-L.); (S.R.y.C.)
| | - Francisco J. Cimas
- Unidad de Bioquímica y Biología Molecular, Servicio de Instrumentación Biomédica, Universidad de Castilla-La Mancha, 02008 Albacete, Spain;
| | - Germán Fliquete
- Grupo de Patología Molecular Traslacional, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona Centro de Investigación Biomédica en RED de Cancer CIBERONC, 08035 Barcelona, Spain; (T.M.); (G.F.); (J.H.-L.); (S.R.y.C.)
| | - Marta Ortega-Muelas
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (E.A.-L.); (J.J.-S.); (R.P.-S.); (M.O.-M.); (O.R.); (D.M.F.-A.); (N.G.-F.); (C.G.-G.); (M.J.R.-H.)
| | - Olga Roche
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (E.A.-L.); (J.J.-S.); (R.P.-S.); (M.O.-M.); (O.R.); (D.M.F.-A.); (N.G.-F.); (C.G.-G.); (M.J.R.-H.)
- Departamento de Ciencias Médicas, Facultad de Medicina, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
| | - Diego M. Fernández-Aroca
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (E.A.-L.); (J.J.-S.); (R.P.-S.); (M.O.-M.); (O.R.); (D.M.F.-A.); (N.G.-F.); (C.G.-G.); (M.J.R.-H.)
| | - Raúl Muñoz Velasco
- Grupo de Oncología Molecular, Facultad de Ciencias Experimentales, Instituto de Investigación Biosanitaria, Universidad Francisco de Vitoria, Pozuelo de Alarcón, 28223 Madrid, Spain; (R.M.V.); (V.J.S.-A.L.)
- Departamento de Anatomía Patológica, Instituto de Investigación Hospital 12 de Octubre, Av. Córdoba, s/n, 28041 Madrid, Spain
| | - Natalia García-Flores
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (E.A.-L.); (J.J.-S.); (R.P.-S.); (M.O.-M.); (O.R.); (D.M.F.-A.); (N.G.-F.); (C.G.-G.); (M.J.R.-H.)
| | - Cristina Garnés-García
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (E.A.-L.); (J.J.-S.); (R.P.-S.); (M.O.-M.); (O.R.); (D.M.F.-A.); (N.G.-F.); (C.G.-G.); (M.J.R.-H.)
| | - Adrián Sánchez-Fdez
- Instituto de Biología Molecular y Celular del Cáncer-CSIC, 37007 Salamanca, Spain; (A.S.-F.); (S.M.-A.); (A.P.); (A.E.-O.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Universidad de Salamanca, CSIC, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en RED de Cancer CIBERONC, 37007 Salamanca, Spain
| | - Sofía Matilla-Almazán
- Instituto de Biología Molecular y Celular del Cáncer-CSIC, 37007 Salamanca, Spain; (A.S.-F.); (S.M.-A.); (A.P.); (A.E.-O.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Universidad de Salamanca, CSIC, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en RED de Cancer CIBERONC, 37007 Salamanca, Spain
| | - Víctor J. Sánchez-Arévalo Lobo
- Grupo de Oncología Molecular, Facultad de Ciencias Experimentales, Instituto de Investigación Biosanitaria, Universidad Francisco de Vitoria, Pozuelo de Alarcón, 28223 Madrid, Spain; (R.M.V.); (V.J.S.-A.L.)
- Departamento de Anatomía Patológica, Instituto de Investigación Hospital 12 de Octubre, Av. Córdoba, s/n, 28041 Madrid, Spain
| | - Javier Hernández-Losa
- Grupo de Patología Molecular Traslacional, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona Centro de Investigación Biomédica en RED de Cancer CIBERONC, 08035 Barcelona, Spain; (T.M.); (G.F.); (J.H.-L.); (S.R.y.C.)
| | - Borja Belandia
- Departamento de Biología del Cáncer, Instituto de Investigaciones Biomédicas ‘Alberto Sols’ (CSIC-UAM), Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, 28029 Madrid, Spain;
| | - Atanasio Pandiella
- Instituto de Biología Molecular y Celular del Cáncer-CSIC, 37007 Salamanca, Spain; (A.S.-F.); (S.M.-A.); (A.P.); (A.E.-O.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Universidad de Salamanca, CSIC, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en RED de Cancer CIBERONC, 37007 Salamanca, Spain
| | - Azucena Esparís-Ogando
- Instituto de Biología Molecular y Celular del Cáncer-CSIC, 37007 Salamanca, Spain; (A.S.-F.); (S.M.-A.); (A.P.); (A.E.-O.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Universidad de Salamanca, CSIC, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en RED de Cancer CIBERONC, 37007 Salamanca, Spain
| | - Santiago Ramón y Cajal
- Grupo de Patología Molecular Traslacional, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona Centro de Investigación Biomédica en RED de Cancer CIBERONC, 08035 Barcelona, Spain; (T.M.); (G.F.); (J.H.-L.); (S.R.y.C.)
| | - Luis del Peso
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM) and Instituto de Investigaciones Biomédicas ‘Alberto Sols’ (CSIC-UAM), 28029 Madrid, Spain;
- Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias CIBERES, 28029 Madrid, Spain
| | - Ricardo Sánchez-Prieto
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (E.A.-L.); (J.J.-S.); (R.P.-S.); (M.O.-M.); (O.R.); (D.M.F.-A.); (N.G.-F.); (C.G.-G.); (M.J.R.-H.)
- Departamento de Ciencias Médicas, Facultad de Medicina, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
- Departamento de Biología del Cáncer, Instituto de Investigaciones Biomédicas ‘Alberto Sols’ (CSIC-UAM), Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, 28029 Madrid, Spain;
- Instituto de Investigaciones Biomédicas ‘Alberto Sols’, Consejo Superior de Investigaciones Científicas (IIBM-CSIC)-Universidad de Castilla-La Mancha, 02008 Albacete, Spain
- Correspondence:
| | - María José Ruiz-Hidalgo
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (E.A.-L.); (J.J.-S.); (R.P.-S.); (M.O.-M.); (O.R.); (D.M.F.-A.); (N.G.-F.); (C.G.-G.); (M.J.R.-H.)
- Departamento de Química Inorgánica, Orgánica y Bioquímica, Área de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
| |
Collapse
|
41
|
Bernier-Latmani J, Mauri C, Marcone R, Renevey F, Durot S, He L, Vanlandewijck M, Maclachlan C, Davanture S, Zamboni N, Knott GW, Luther SA, Betsholtz C, Delorenzi M, Brisken C, Petrova TV. ADAMTS18 + villus tip telocytes maintain a polarized VEGFA signaling domain and fenestrations in nutrient-absorbing intestinal blood vessels. Nat Commun 2022; 13:3983. [PMID: 35810168 PMCID: PMC9271081 DOI: 10.1038/s41467-022-31571-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 06/21/2022] [Indexed: 12/17/2022] Open
Abstract
The small intestinal villus tip is the first point of contact for lumen-derived substances including nutrients and microbial products. Electron microscopy studies from the early 1970s uncovered unusual spatial organization of small intestinal villus tip blood vessels: their exterior, epithelial-facing side is fenestrated, while the side facing the villus stroma is non-fenestrated, covered by pericytes and harbors endothelial nuclei. Such organization optimizes the absorption process, however the molecular mechanisms maintaining this highly specialized structure remain unclear. Here we report that perivascular LGR5+ villus tip telocytes (VTTs) are necessary for maintenance of villus tip endothelial cell polarization and fenestration by sequestering VEGFA signaling. Mechanistically, unique VTT expression of the protease ADAMTS18 is necessary for VEGFA signaling sequestration through limiting fibronectin accumulation. Therefore, we propose a model in which LGR5+ ADAMTS18+ telocytes are necessary to maintain a “just-right” level and location of VEGFA signaling in intestinal villus blood vasculature to ensure on one hand the presence of sufficient endothelial fenestrae, while avoiding excessive leakiness of the vessels and destabilization of villus tip epithelial structures. The molecular mechanisms ensuring the specialized structure of small intestinal villus tip blood vessels are incompletely understood. Here the authors show that ADAMTS18+ telocytes maintain a “just-right” level and location of VEGFA signaling on intestinal villus blood vessels, thereby ensuring the presence of endothelial fenestrae for nutrient absorption, while avoiding excessive leakiness and destabilization of villus tip epithelial structures.
Collapse
Affiliation(s)
- Jeremiah Bernier-Latmani
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne and University of Lausanne, Lausanne, Switzerland.
| | - Cristina Mauri
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne and University of Lausanne, Lausanne, Switzerland
| | - Rachel Marcone
- Bioinformatics Core Facility, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - François Renevey
- Department of Immunobiology, University of Lausanne, Lausanne, Switzerland
| | - Stephan Durot
- Institute of Molecular Systems Biology ETH, Zurich, Switzerland
| | - Liqun He
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Michael Vanlandewijck
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden.,Department of Medicine-Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Catherine Maclachlan
- Bio Electron Microscopy Laboratory, School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Suzel Davanture
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne and University of Lausanne, Lausanne, Switzerland
| | - Nicola Zamboni
- Institute of Molecular Systems Biology ETH, Zurich, Switzerland
| | - Graham W Knott
- Bio Electron Microscopy Laboratory, School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Sanjiv A Luther
- Department of Immunobiology, University of Lausanne, Lausanne, Switzerland
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden.,Department of Medicine-Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Mauro Delorenzi
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne and University of Lausanne, Lausanne, Switzerland.,Bioinformatics Core Facility, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Cathrin Brisken
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Tatiana V Petrova
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne and University of Lausanne, Lausanne, Switzerland. .,Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, EPFL, Lausanne, Switzerland.
| |
Collapse
|
42
|
Fu S, Tan R, Feng Y, Yu P, Mo Y, Xiao W, Wang S, Zhang J. N-methyl-N-nitrosourea induces zebrafish anomalous angiogenesis through Wnt/β-catenin pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 239:113674. [PMID: 35623148 DOI: 10.1016/j.ecoenv.2022.113674] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 05/05/2022] [Accepted: 05/18/2022] [Indexed: 06/15/2023]
Abstract
N-methyl-N-nitrosourea (MNU) is a prevalent environmental carcinogen, which leads to tumors in various organs in animal models, while the mechanisms involved were still not fully understood. It is well known that anomalous angiogenesis is a key step in tumorigenesis and progression. In this study, we found that MNU induced abnormal angiogenesis which was accompanied by upregulation of rspo1, p53 and vegfaa in zebrafish embryos. Moreover, it revealed that MNU-induced ectopic sprouting of blood vessels was significantly reduced in rspo1-knockdown but not p53-knockdown embryos, indicating that rspo1 was necessary for MNU-induced abnormal angiogenesis. Additionally, pharmaceutical activation or inhibition of Wnt/β-catenin signaling pathway using (2'Z,3'E)- 6-bromoindirubin-3'-oxime or CCT036477 significantly increased or inhibited the pro-angiogenic effect of MNU on developing zebrafish embryos, which was confirmed by the effect of proliferation and migration in MNU-treated bEnd.3 cells. These data together indicated that rspo1/Wnt/β-catenin/vegfaa axis is involved in the modulation of MNU-induced anomalous angiogenesis.
Collapse
Affiliation(s)
- Saifang Fu
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical University, Zhanjiang 524001, China
| | - Rongbang Tan
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical University, Zhanjiang 524001, China
| | - Yufei Feng
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical University, Zhanjiang 524001, China
| | - Ping Yu
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical University, Zhanjiang 524001, China
| | - Yuqian Mo
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical University, Zhanjiang 524001, China
| | - Wei Xiao
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical University, Zhanjiang 524001, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China
| | - Shouyu Wang
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Jingjing Zhang
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical University, Zhanjiang 524001, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China.
| |
Collapse
|
43
|
Liu J, Tang G, Liu W, Zhou Y, Fan C, Zhang W. MiR-20a-5p facilitates cartilage repair in osteoarthritis via suppressing mitogen-activated protein kinase kinase kinase 2. Bioengineered 2022; 13:13801-13814. [PMID: 35707845 PMCID: PMC9276018 DOI: 10.1080/21655979.2022.2084270] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Bone marrow mesenchymal stem cell (BMSC) chondrogenic differentiation contributes to the treatment of osteoarthritis (OA). Numerous studies have indicated that microRNAs (miRNAs) regulate the pathogenesis and development of multiple disorders, including OA. Nevertheless, the role of miR-20a-5p in OA remains obscure. Forty male C57BL/6 mice were divided into four groups and were surgically induced OA or underwent sham surgery in the presence or absence of miR-20a-5p. Flow cytometry was implemented to detect surface markers of BMSCs. Reverse transcription quantitative polymerase chain reaction revealed the upregulation of miR-20a-5p during BMSC chondrogenic differentiation. Western blotting displayed that miR-20a-5p inhibition decreased protein levels of cartilage matrix markers but enhanced those of catabolic and hypertrophic chondrocyte markers in BMSCs. Alcian blue staining, hematoxylin‑eosin staining and micro-CT revealed that miR-20a-5p inhibition restrained chondrogenic differentiation and miR-20a-5p overexpression promoted the repair of damaged cartilage and subchondral bone, respectively. Luciferase reporter assay identified that mitogen activated protein kinase kinase kinase 2 (Map3k2) was a target of miR-20a-5p in BMSCs. Moreover, the expression of miR-20a-5p and Map3k2 was negatively correlated in murine cartilage tissues. Knocking down Map3k2 could rescue the suppressive influence of miR-20a-5p inhibition on chondrogenic differentiation of BMSCs. In conclusion, miR-20a-5p facilitates BMSC chondrogenic differentiation and contributes to cartilage repair in OA by suppressing Map3k2.
Collapse
Affiliation(s)
- Jiazhi Liu
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Guo Tang
- Department of Orthopaedics, Shanghai Songjiang District Central Hospital, Shanghai, China
| | - Wenjun Liu
- Department of Orthopaedics, South Hospital of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yi Zhou
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Cunyi Fan
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Wei Zhang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
44
|
Gut microbiota drives macrophage-dependent self-renewal of intestinal stem cells via niche enteric serotonergic neurons. Cell Res 2022; 32:555-569. [PMID: 35379903 DOI: 10.1038/s41422-022-00645-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 02/08/2022] [Indexed: 11/08/2022] Open
Abstract
Lgr5+ intestinal stem cells (ISCs) reside within specialized niches at the crypt base and harbor self-renewal and differentiation capacities. ISCs in the crypt base are sustained by their surrounding niche for precise modulation of self-renewal and differentiation. However, how intestinal cells in the crypt niche and microbiota in enteric cavity coordinately regulate ISC stemness remains unclear. Here, we show that ISCs are regulated by microbiota and niche enteric serotonergic neurons. The gut microbiota metabolite valeric acid promotes Tph2 expression in enteric serotonergic neurons via blocking the recruitment of the NuRD complex onto Tph2 promoter. 5-hydroxytryptamine (5-HT) in turn activates PGE2 production in a PGE2+ macrophage subset through its receptors HTR2A/3 A; and PGE2 via binding its receptors EP1/EP4, promotes Wnt/β-catenin signaling in ISCs to promote their self-renewal. Our findings illustrate a complex crosstalk among microbiota, intestinal nerve cells, intestinal immune cells and ISCs, revealing a new layer of ISC regulation by niche cells and microbiota.
Collapse
|
45
|
Wan Y, Huang J, Song Y, Gu C, Kong J, Zuo L, Chen J. hsa-miR-340-5p inhibits epithelial–mesenchymal transition in endometriosis by targeting MAP3K2 and inactivating MAPK/ERK signaling. Open Med (Wars) 2022; 17:566-576. [PMID: 35415247 PMCID: PMC8932397 DOI: 10.1515/med-2022-0448] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/20/2022] [Accepted: 02/07/2022] [Indexed: 11/17/2022] Open
Abstract
Increasing evidence has verified the indispensable effect of microRNAs (miRNAs) in the biological processes of human diseases, including endometriosis. hsa-miR-340-5p was reported to display a low level in patients with endometriosis, but the detailed function of miR-340-5p in endometriosis is unclarified. RT-qPCR was used for the assessment of RNA levels of miR-340-5p and its downstream target genes in endometrial stromal cells (ESCs). Western blotting and Transwell assays revealed that upregulation of miR-340-5p suppressed the migration, invasiveness, and epithelial–mesenchymal transition (EMT) in ESCs. Bioinformatics tools were used to predict miR-340-5p downstream genes. Luciferase reporter assay displayed that miR-340-5p could bind to messenger RNA mitogen-activated protein kinase kinase kinase 2 (MAP3K2). MAP3K2 was targeted by miR-349-5p and could reverse the influence of miR-340-5p. miR-340-5p exerted its impact on the invasive characters of ESCs by inactivating the MAP3K2-mediated MAPK/ERK signaling. In conclusion, miR-340-5p restrains cell migration, invasiveness, and EMT in ESCs by targeting MAP3K2 and inactivating MAPK/ERK signaling.
Collapse
Affiliation(s)
- Yiting Wan
- Department of Gynecology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine , Shanghai 200071 , China
| | - Jiami Huang
- Department of Gynecology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine , Shanghai 200071 , China
| | - Yanhua Song
- Department of Gynecology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine , Shanghai 200071 , China
| | - Cancan Gu
- Department of Gynecology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine , Shanghai 200071 , China
| | - Jueying Kong
- Department of Gynecology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine , Shanghai 200071 , China
| | - Ling Zuo
- Department of Gynecology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine , Shanghai 200071 , China
| | - Jing Chen
- Department of Gynecology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine , No. 274 Middle Zhijiang Road , Shanghai 200071 , China
| |
Collapse
|
46
|
Lepr + mesenchymal cells sense diet to modulate intestinal stem/progenitor cells via Leptin-Igf1 axis. Cell Res 2022; 32:670-686. [PMID: 35296796 DOI: 10.1038/s41422-022-00643-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 02/25/2022] [Indexed: 02/07/2023] Open
Abstract
Diet can impact on gut health and disease by modulating intestinal stem cells (ISCs). However, it is largely unknown if and how the ISC niche responds to diet and influences ISC function. Here, we demonstrate that Lepr+ mesenchymal cells (MCs) surrounding intestinal crypts sense diet change and provide a novel niche signal to maintain ISC and progenitor cell proliferation. The abundance of these MCs increases upon administration of a high-fat diet (HFD) but dramatically decreases upon fasting. Depletion of Lepr+ MCs resulted in fewer intestinal stem/progenitor cells, compromised the architecture of crypt-villus axis and impaired intestinal regeneration. Furthermore, we showed that IGF1 secreted by Lepr+ MCs is an important effector that promotes proliferation of ISCs and progenitor cells in the intestinal crypt. We conclude that Lepr+ MCs sense diet alterations and, in turn, modulate intestinal stem/progenitor cell function via a stromal IGF1-epithelial IGF1R axis. These findings reveal that Lepr+ MCs are important mediators linking systemic diet changes to local ISC function and might serve as a novel therapeutic target for gut diseases.
Collapse
|
47
|
Challenges and opportunities targeting mechanisms of epithelial injury and recovery in acute intestinal graft-versus-host disease. Mucosal Immunol 2022; 15:605-619. [PMID: 35654837 PMCID: PMC9259481 DOI: 10.1038/s41385-022-00527-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/21/2022] [Accepted: 05/16/2022] [Indexed: 02/04/2023]
Abstract
Despite advances in immunosuppressive prophylaxis and overall supportive care, gastrointestinal (GI) graft-versus-host disease (GVHD) remains a major, lethal side effect after allogeneic hematopoietic stem cell transplantation (allo-HSCT). It has become increasingly clear that the intestinal epithelium, in addition to being a target of transplant-related toxicity and GVHD, plays an important role in the onset of GVHD. Over the last two decades, increased understanding of the epithelial constituents and their microenvironment has led to the development of novel prophylactic and therapeutic interventions, with the potential to protect the intestinal epithelium from GVHD-associated damage and promote its recovery following insult. In this review, we will discuss intestinal epithelial injury and the role of the intestinal epithelium in GVHD pathogenesis. In addition, we will highlight possible approaches to protect the GI tract from damage posttransplant and to stimulate epithelial regeneration, in order to promote intestinal recovery. Combined treatment modalities integrating immunomodulation, epithelial protection, and induction of regeneration may hold the key to unlocking mucosal recovery and optimizing therapy for acute intestinal GVHD.
Collapse
|
48
|
Zhang Y, Wang J, Yu C, Xia K, Yang B, Zhang Y, Ying L, Wang C, Huang X, Chen Q, Shen L, Li F, Liang C. Advances in single-cell sequencing and its application to musculoskeletal system research. Cell Prolif 2022; 55:e13161. [PMID: 34888976 PMCID: PMC8780907 DOI: 10.1111/cpr.13161] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 10/30/2021] [Accepted: 11/12/2021] [Indexed: 11/30/2022] Open
Abstract
In recent years, single-cell sequencing (SCS) technologies have continued to advance with improved operating procedures and reduced cost, leading to increasing practical adoption among researchers. These emerging technologies have superior abilities to analyse cell heterogeneity at a single-cell level, which have elevated multi-omics research to a higher level. In some fields of research, application of SCS has enabled many valuable discoveries, and musculoskeletal system offers typical examples. This article reviews some major scientific issues and recent advances in musculoskeletal system. In addition, combined with SCS technologies, the research of cell or tissue heterogeneity in limb development and various musculoskeletal system clinical diseases also provides new possibilities for treatment strategies. Finally, this article discusses the challenges and future development potential of SCS and recommends the direction of future applications of SCS to musculoskeletal medicine.
Collapse
Affiliation(s)
- Yongxiang Zhang
- Department of Orthopedics SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Zhejiang Key Laboratory of Bone and Joint Precision and Department of OrthopedicsResearch Institute of Zhejiang UniversityHangzhouZhejiangChina
| | - Jingkai Wang
- Department of Orthopedics SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Zhejiang Key Laboratory of Bone and Joint Precision and Department of OrthopedicsResearch Institute of Zhejiang UniversityHangzhouZhejiangChina
| | - Chao Yu
- Department of Orthopedics SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Zhejiang Key Laboratory of Bone and Joint Precision and Department of OrthopedicsResearch Institute of Zhejiang UniversityHangzhouZhejiangChina
| | - Kaishun Xia
- Department of Orthopedics SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Zhejiang Key Laboratory of Bone and Joint Precision and Department of OrthopedicsResearch Institute of Zhejiang UniversityHangzhouZhejiangChina
| | - Biao Yang
- Department of Orthopedics SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Zhejiang Key Laboratory of Bone and Joint Precision and Department of OrthopedicsResearch Institute of Zhejiang UniversityHangzhouZhejiangChina
| | - Yuang Zhang
- Department of Orthopedics SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Zhejiang Key Laboratory of Bone and Joint Precision and Department of OrthopedicsResearch Institute of Zhejiang UniversityHangzhouZhejiangChina
| | - Liwei Ying
- Department of Orthopedics SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Zhejiang Key Laboratory of Bone and Joint Precision and Department of OrthopedicsResearch Institute of Zhejiang UniversityHangzhouZhejiangChina
| | - Chenggui Wang
- Department of Orthopedics SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Zhejiang Key Laboratory of Bone and Joint Precision and Department of OrthopedicsResearch Institute of Zhejiang UniversityHangzhouZhejiangChina
| | - Xianpeng Huang
- Department of Orthopedics SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Zhejiang Key Laboratory of Bone and Joint Precision and Department of OrthopedicsResearch Institute of Zhejiang UniversityHangzhouZhejiangChina
| | - Qixin Chen
- Department of Orthopedics SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Zhejiang Key Laboratory of Bone and Joint Precision and Department of OrthopedicsResearch Institute of Zhejiang UniversityHangzhouZhejiangChina
| | - Li Shen
- Department of Orthopedics SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell BiologyLife Sciences InstituteZhejiang UniversityHangzhouChina
- Hangzhou Innovation CenterZhejiang UniversityHangzhouChina
| | - Fangcai Li
- Department of Orthopedics SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Zhejiang Key Laboratory of Bone and Joint Precision and Department of OrthopedicsResearch Institute of Zhejiang UniversityHangzhouZhejiangChina
| | - Chengzhen Liang
- Department of Orthopedics SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Zhejiang Key Laboratory of Bone and Joint Precision and Department of OrthopedicsResearch Institute of Zhejiang UniversityHangzhouZhejiangChina
| |
Collapse
|
49
|
García García CJ, Acevedo Diaz AC, Kumari N, Govindaraju S, de la Cruz Bonilla M, San Lucas FA, Nguyen ND, Jiménez Sacarello I, Piwnica-Worms H, Maitra A, Taniguchi CM. HIF2 Regulates Intestinal Wnt5a Expression. Front Oncol 2021; 11:769385. [PMID: 34900719 PMCID: PMC8656274 DOI: 10.3389/fonc.2021.769385] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/29/2021] [Indexed: 12/11/2022] Open
Abstract
Radiation therapy for abdominal tumors is challenging because the small intestine is exquisitely radiosensitive. Unfortunately, there are no FDA-approved therapies to prevent or mitigate GI radiotoxicity. The EGLN protein family are oxygen sensors that regulate cell survival and metabolism through the degradation of hypoxia-inducible factors (HIFs). Our group has previously shown that stabilization of HIF2 through genetic deletion or pharmacologic inhibition of the EGLNs mitigates and protects against GI radiotoxicity in mice by improving intestinal crypt stem cell survival. Here we aimed to elucidate the molecular mechanisms by which HIF2 confers GI radioprotection. We developed duodenal organoids from mice, transiently overexpressed non-degradable HIF2, and performed bulk RNA sequencing. Interestingly, HIF2 upregulated known radiation modulators and genes involved in GI homeostasis, including Wnt5a. Non-canonical Wnt5a signaling has been shown by other groups to improve intestinal crypt regeneration in response to injury. Here we show that HIF2 drives Wnt5a expression in multiple duodenal organoid models. Luciferase reporter assays performed in human cells showed that HIF2 directly activates the WNT5A promoter via a hypoxia response element. We then evaluated crypt regeneration using spheroid formation assays. Duodenal organoids that were pre-treated with recombinant Wnt5a had a higher cryptogenic capacity after irradiation, compared to vehicle-treated organoids. Conversely, we found that Wnt5a knockout decreased the cryptogenic potential of intestinal stem cells following irradiation. Treatment with recombinant Wnt5a prior to irradiation rescued the cryptogenic capacity of Wnt5a knockout organoids, indicating that Wnt5a is necessary and sufficient for duodenal radioprotection. Taken together, our results suggest that HIF2 radioprotects the GI tract by inducing Wnt5a expression.
Collapse
Affiliation(s)
- Carolina J. García García
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- School of Medicine, University of Puerto Rico, Rio Piedras, PR, United States
| | | | - Neeraj Kumari
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Suman Govindaraju
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Marimar de la Cruz Bonilla
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- School of Medicine, University of Puerto Rico, Rio Piedras, PR, United States
| | - F. Anthony San Lucas
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nicholas D. Nguyen
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | - Helen Piwnica-Worms
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Anirban Maitra
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Cullen M. Taniguchi
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
50
|
Ara T, Hashimoto D. Novel Insights Into the Mechanism of GVHD-Induced Tissue Damage. Front Immunol 2021; 12:713631. [PMID: 34512636 PMCID: PMC8429834 DOI: 10.3389/fimmu.2021.713631] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 08/10/2021] [Indexed: 12/22/2022] Open
Abstract
Prophylaxis for and treatment of graft-versus-host disease (GVHD) are essential for successful allogeneic hematopoietic stem cell transplantation (allo-SCT) and mainly consist of immunosuppressants such as calcineurin inhibitors. However, profound immunosuppression can lead to tumor relapse and infectious complications, which emphasizes the necessity of developing novel management strategies for GVHD. Emerging evidence has revealed that tissue-specific mechanisms maintaining tissue homeostasis and promoting tissue tolerance to combat GVHD are damaged after allo-SCT, resulting in exacerbation and treatment refractoriness of GVHD. In the gastrointestinal tract, epithelial regeneration derived from intestinal stem cells (ISCs), a microenvironment that maintains healthy gut microbiota, and physical and chemical mucosal barrier functions against pathogens are damaged by conditioning regimens and/or GVHD. The administration of growth factors for cells that maintain intestinal homeostasis, such as interleukin-22 (IL-22) for ISCs, R-spondin 1 (R-Spo1) for ISCs and Paneth cells, and interleukin-25 (IL-25) for goblet cells, mitigates murine GVHD. In this review, we summarize recent advances in the understanding of GVHD-induced tissue damage and emerging strategies for the management of GVHD.
Collapse
Affiliation(s)
- Takahide Ara
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Daigo Hashimoto
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| |
Collapse
|