1
|
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
|
2
|
Lu D, Yao D, Hu G, Zhou J, Shen X, Qian L. Maternal docosahexaenoic acid supplementation during lactation improves exercise performance, enhances intestinal glucose absorption and modulates gut microbiota in weaning offspring mice. Front Nutr 2024; 11:1423576. [PMID: 39036494 PMCID: PMC11258037 DOI: 10.3389/fnut.2024.1423576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 06/19/2024] [Indexed: 07/23/2024] Open
Abstract
Introduction Intestinal dysfunction induced by weaning stress is common during breastfeeding period. Docosahexaenoic acid (DHA) is well known for promoting visual and brain development, but its effects on early intestinal development remain unknown. This study investigated the impact of maternal DHA supplementation during lactation on intestinal glucose absorption and gut microbiota in weaning offspring mice. Materials and methods Dams were supplemented with vehicle (control), 150 mg/(kg body weight · day) DHA (L-DHA), or 450 mg/(kg body weight · day) DHA (H-DHA) throughout lactation by oral administration. After weaning, pups were randomly divided into three groups for athletic analysis, microbial and proteomic analysis, biochemical analysis, 4-deoxy-4-fluoro-D-glucose (4-FDG) absorption test, and gene expression quantitation of glucose transport-associated proteins and mTOR signaling components. Results The H-DHA group exhibited enhanced grip strength and prolonged swimming duration compared to the control group. Additionally, there were significant increases in jejunal and ileal villus height, and expanded surface area of jejunal villi in the H-DHA group. Microbial analyses revealed that maternal DHA intake increased the abundance of beneficial gut bacteria and promoted metabolic pathways linked to carbohydrate and energy metabolism. Proteomic studies indicated an increased abundance of nutrient transport proteins and enrichment of pathways involved in absorption and digestion in the H-DHA group. This group also showed higher concentrations of glucose in the jejunum and ileum, as well as elevated glycogen levels in the liver and muscles, in contrast to lower glucose levels in the intestinal contents and feces compared to the control group. The 4-FDG absorption test showed more efficient absorption after oral 4-FDG gavage in the H-DHA group. Moreover, the expressions of glucose transport-associated proteins, GLUT2 and SGLT1, and the activation of mTOR pathway were enhanced in the H-DHA group compared to the control group. The L-DHA group also showed similar but less pronounced improvements in these aspects relative to the H-DHA group. Conclusion Our findings suggested that maternal DHA supplementation during lactation improves the exercise performance, enhances the intestinal glucose absorption by increasing the expressions of glucose transporters, and beneficially alters the structure of gut microbiome in weaning offspring mice.
Collapse
Affiliation(s)
- Dalu Lu
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute for Pediatric Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Die Yao
- Department of Clinical Nutrition, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Gaoli Hu
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute for Pediatric Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiefei Zhou
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute for Pediatric Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiuhua Shen
- Department of Clinical Nutrition, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Linxi Qian
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute for Pediatric Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Clinical Nutrition, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Wilson AP, Moshal KS, Franca AP, Ramani S, Gallucci R, Chaaban H, Burge KY. Analyzing efficiency of a lentiviral shRNA knockdown system in human enteroids using western blot and flow cytometry. STAR Protoc 2024; 5:103082. [PMID: 38781076 PMCID: PMC11145376 DOI: 10.1016/j.xpro.2024.103082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/11/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024] Open
Abstract
Enteroids are in vitro models to study gastrointestinal pathologies and test personalized therapeutics; however, the inherent complexity of enteroids often renders standard gene editing approaches ineffective. Here, we introduce a refined lentiviral transfection protocol, ensuring sufficient lentiviral engagement with enteroids while considering spatiotemporal growth variability throughout the extracellular matrix. Additionally, we highlight a selection process for transduced cells, introduce a protocol to accurately measure transduction efficiency, and explore methodologies to gauge effects of gene knockdown on biological processes.
Collapse
Affiliation(s)
- Adam P Wilson
- Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| | - Karni S Moshal
- Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| | - Addison P Franca
- Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Sasirekha Ramani
- Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Randle Gallucci
- Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Hala Chaaban
- Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| | - Kathryn Y Burge
- Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
4
|
Ramos R, Swedlund B, Ganesan AK, Morsut L, Maini PK, Monuki ES, Lander AD, Chuong CM, Plikus MV. Parsing patterns: Emerging roles of tissue self-organization in health and disease. Cell 2024; 187:3165-3186. [PMID: 38906093 PMCID: PMC11299420 DOI: 10.1016/j.cell.2024.05.016] [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: 09/12/2023] [Revised: 02/22/2024] [Accepted: 05/08/2024] [Indexed: 06/23/2024]
Abstract
Patterned morphologies, such as segments, spirals, stripes, and spots, frequently emerge during embryogenesis through self-organized coordination between cells. Yet, complex patterns also emerge in adults, suggesting that the capacity for spontaneous self-organization is a ubiquitous property of biological tissues. We review current knowledge on the principles and mechanisms of self-organized patterning in embryonic tissues and explore how these principles and mechanisms apply to adult tissues that exhibit features of patterning. We discuss how and why spontaneous pattern generation is integral to homeostasis and healing of tissues, illustrating it with examples from regenerative biology. We examine how aberrant self-organization underlies diverse pathological states, including inflammatory skin disorders and tumors. Lastly, we posit that based on such blueprints, targeted engineering of pattern-driving molecular circuits can be leveraged for synthetic biology and the generation of organoids with intricate patterns.
Collapse
Affiliation(s)
- Raul Ramos
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
| | - Benjamin Swedlund
- Eli and Edythe Broad CIRM Center, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Anand K Ganesan
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, USA; Department of Dermatology, University of California, Irvine, Irvine, CA, USA
| | - Leonardo Morsut
- Eli and Edythe Broad CIRM Center, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Alfred E. Mann Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Philip K Maini
- Mathematical Institute, University of Oxford, Oxford, UK
| | - Edwin S Monuki
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA; Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, USA
| | - Arthur D Lander
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, USA.
| | - Cheng-Ming Chuong
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
5
|
Bao L, Fu L, Su Y, Chen Z, Peng Z, Sun L, Gonzalez FJ, Wu C, Zhang H, Shi B, Shi YB. Amino acid transporter SLC7A5 regulates cell proliferation and secretary cell differentiation and distribution in the mouse intestine. Int J Biol Sci 2024; 20:2187-2201. [PMID: 38617535 PMCID: PMC11008275 DOI: 10.7150/ijbs.94297] [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: 01/15/2024] [Accepted: 02/16/2024] [Indexed: 04/16/2024] Open
Abstract
The intestine is critical for not only processing nutrients but also protecting the organism from the environment. These functions are mainly carried out by the epithelium, which is constantly being self-renewed. Many genes and pathways can influence intestinal epithelial cell proliferation. Among them is mTORC1, whose activation increases cell proliferation. Here, we report the first intestinal epithelial cell (IEC)-specific knockout (ΔIEC) of an amino acid transporter capable of activating mTORC1. We show that the transporter, SLC7A5, is highly expressed in mouse intestinal crypt and Slc7a5ΔIEC reduces mTORC1 signaling. Surprisingly, adult Slc7a5ΔIEC intestinal crypts have increased cell proliferation but reduced mature Paneth cells. Goblet cells, the other major secretory cell type in the small intestine, are increased in the crypts but reduced in the villi. Analyses with scRNA-seq and electron microscopy have revealed dedifferentiation of Paneth cells in Slc7a5ΔIEC mice, leading to markedly reduced secretory granules with little effect on Paneth cell number. Thus, SLC7A5 likely regulates secretory cell differentiation to affect stem cell niche and indirectly regulate cell proliferation.
Collapse
Affiliation(s)
- Lingyu Bao
- Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, MD, USA
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine. No.277, Yanta West Road, Xi'an, Shaanxi, 710061, P.R. China
| | - Liezhen Fu
- Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, MD, USA
| | - Yijun Su
- Laboratory of High Resolution Optical Imaging and Advanced Imaging and Microscopy Resource, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
- Janelia Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA
| | - Zuojia Chen
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zhaoyi Peng
- Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, MD, USA
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine. No.277, Yanta West Road, Xi'an, Shaanxi, 710061, P.R. China
| | - Lulu Sun
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Frank J. Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chuan Wu
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hongen Zhang
- Bioinformatics and Scientific Programming Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Bingyin Shi
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine. No.277, Yanta West Road, Xi'an, Shaanxi, 710061, P.R. China
| | - Yun-Bo Shi
- Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, MD, USA
| |
Collapse
|
6
|
Huang GX, Hallen NR, Lee M, Zheng K, Wang X, Mandanas MV, Djeddi S, Fernandez D, Hacker J, Ryan T, Bergmark RW, Bhattacharyya N, Lee S, Maxfield AZ, Roditi RE, Buchheit KM, Laidlaw TM, Gern JE, Hallstrand TS, Ray A, Wenzel SE, Boyce JA, Gutierrez-Arcelus M, Barrett NA. Increased epithelial mTORC1 activity in chronic rhinosinusitis with nasal polyps. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.13.562288. [PMID: 37904989 PMCID: PMC10614789 DOI: 10.1101/2023.10.13.562288] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Background The airway epithelium plays a central role in the pathogenesis of chronic respiratory diseases such as asthma and chronic rhinosinusitis with nasal polyps (CRSwNP), but the mechanisms by which airway epithelial cells (EpCs) maintain inflammation are poorly understood. Objective We hypothesized that transcriptomic assessment of sorted airway EpCs across the spectrum of differentiation would allow us to define mechanisms by which EpCs perpetuate airway inflammation. Methods Ethmoid sinus EpCs from adult patients with CRS were sorted into 3 subsets, bulk RNA sequenced, and analyzed for differentially expressed genes and pathways. Single cell RNA-seq (scRNA-seq) datasets from eosinophilic and non-eosinophilic CRSwNP and bulk RNA-seq of EpCs from mild/moderate and severe asthma were assessed. Immunofluorescent staining and ex vivo functional analysis of sinus EpCs were used to validate our findings. Results Analysis within and across purified EpC subsets revealed an enrichment in glycolytic programming in CRSwNP vs CRSsNP. Correlation analysis identified mammalian target of rapamycin complex 1 (mTORC1) as a potential regulator of the glycolytic program and identified EpC expression of cytokines and wound healing genes as potential sequelae. mTORC1 activity was upregulated in CRSwNP, and ex vivo inhibition demonstrated that mTOR is critical for EpC generation of CXCL8, IL-33, and CXCL2. Across patient samples, the degree of glycolytic activity was associated with T2 inflammation in CRSwNP, and with both T2 and non-T2 inflammation in severe asthma. Conclusions Together, these findings highlight a metabolic axis required to support epithelial generation of cytokines critical to both chronic T2 and non-T2 inflammation in CRSwNP and asthma.
Collapse
Affiliation(s)
- George X. Huang
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Nils R. Hallen
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Minkyu Lee
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Kelly Zheng
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Xin Wang
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | | | - Sarah Djeddi
- Division of Immunology, Boston Children’s Hospital; Boston, MA
| | | | - Jonathan Hacker
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Tessa Ryan
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Regan W. Bergmark
- Department of Otolaryngology, Head and Neck Surgery, Brigham and Women’s Hospital; Boston, MA
| | - Neil Bhattacharyya
- Department of Otolaryngology, Head and Neck Surgery, Massachusetts Eye and Ear Infirmary; Boston, MA
| | - Stella Lee
- Department of Otolaryngology, Head and Neck Surgery, Brigham and Women’s Hospital; Boston, MA
| | - Alice Z. Maxfield
- Department of Otolaryngology, Head and Neck Surgery, Brigham and Women’s Hospital; Boston, MA
| | - Rachel E. Roditi
- Department of Otolaryngology, Head and Neck Surgery, Brigham and Women’s Hospital; Boston, MA
| | - Kathleen M. Buchheit
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Tanya M. Laidlaw
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - James E. Gern
- Division of Allergy, Immunology, and Rheumatology, University of Wisconsin School of Medicine and Public Health; Madison, WI
| | - Teal S. Hallstrand
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington Medical Center; Seattle, WA
| | - Anuradha Ray
- Department of Immunology, University of Pittsburgh; Pittsburgh, PA
| | - Sally E. Wenzel
- Department of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh Medical Center; Pittsburgh, PA
| | - Joshua A. Boyce
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Maria Gutierrez-Arcelus
- Division of Immunology, Boston Children’s Hospital; Boston, MA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard; Cambridge, MA
| | - Nora A. Barrett
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| |
Collapse
|
7
|
Park SY, Kang JH, Jung HJ, Hwang JH, Chun HS, Yoon YS, Oh SH. Okadaic Acid Is at Least as Toxic as Dinophysistoxin-1 after Repeated Administration to Mice by Gavage. Toxins (Basel) 2023; 15:587. [PMID: 37888618 PMCID: PMC10611360 DOI: 10.3390/toxins15100587] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/07/2023] [Accepted: 09/21/2023] [Indexed: 10/28/2023] Open
Abstract
Okadaic acid (OA) and its analogues cause diarrhetic shellfish poisoning (DSP) in humans, and risk assessments of these toxins require toxicity equivalency factors (TEFs), which represent the relative toxicities of analogues. However, no human death by DSP toxin has been reported, and its current TEF value is based on acute lethality. To properly reflect the symptoms of DSP, such as diarrhea without death, the chronic toxicity of DSP toxins at sublethal doses should be considered. In this study, we obtained acute oral LD50 values for OA and dinophysistoxin-1 (DTX-1) (1069 and 897 μg/kg, respectively) to set sublethal doses. Mice were treated with sublethal doses of OA and DTX-1 for 7 days. The mice lost body weight, and the disease activity index and intestinal crypt depths increased. Furthermore, these changes were more severe in OA-treated mice than in the DTX-1-treated mice. Strikingly, ascites was observed, and its severity was greater in mice treated with OA. Our findings suggest that OA is at least as toxic as DTX-1 after repeated oral administration at a low dose. This is the first study to compare repeated oral dosing of DSP toxins. Further sub-chronic and chronic studies are warranted to determine appropriate TEF values for DSP toxins.
Collapse
Affiliation(s)
- Se Yong Park
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea;
| | - Ju-Hee Kang
- College of Pharmacy, Gachon University, Incheon 21963, Republic of Korea; (J.-H.K.); (H.J.J.); (J.H.H.)
| | - Hyun Jin Jung
- College of Pharmacy, Gachon University, Incheon 21963, Republic of Korea; (J.-H.K.); (H.J.J.); (J.H.H.)
| | - Jung Ho Hwang
- College of Pharmacy, Gachon University, Incheon 21963, Republic of Korea; (J.-H.K.); (H.J.J.); (J.H.H.)
| | - Hyang Sook Chun
- Food Toxicology Laboratory, School of Food Science and Technology, Chung-Ang University, Anseong 17546, Republic of Korea;
| | - Yeo Sung Yoon
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea;
| | - Seung Hyun Oh
- College of Pharmacy, Gachon University, Incheon 21963, Republic of Korea; (J.-H.K.); (H.J.J.); (J.H.H.)
| |
Collapse
|
8
|
Brubaker PL. A New Role for Endocrine Cells in the Intestinal Crypt. Cell Mol Gastroenterol Hepatol 2023; 15:1525-1526. [PMID: 36805429 DOI: 10.1016/j.jcmgh.2023.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 01/25/2023] [Accepted: 01/25/2023] [Indexed: 02/23/2023]
|
9
|
Bao L, Fu L, Su Y, Chen Z, Peng Z, Sun L, Gonzalez FJ, Wu C, Zhang H, Shi B, Shi YB. Amino acid transporter SLC7A5 regulates Paneth cell function to affect the intestinal inflammatory response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.24.524966. [PMID: 36789439 PMCID: PMC9928054 DOI: 10.1101/2023.01.24.524966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The intestine is critical for not only processing and resorbing nutrients but also protecting the organism from the environment. These functions are mainly carried out by the epithelium, which is constantly being self-renewed. Many genes and pathways can influence intestinal epithelial cell proliferation. Among them is mTORC1, whose activation increases cell proliferation. Here, we report the first intestinal epithelial cell-specific knockout ( ΔIEC ) of an amino acid transporter capable of activating mTORC1. We show that the transporter, SLC7A5, is highly expressed in mouse intestinal crypt and Slc7a5 ΔIEC reduces mTORC1 signaling. Surprisingly, Slc7a5 ΔIEC mice have increased cell proliferation but reduced secretory cells, particularly mature Paneth cells. scRNA-seq and electron microscopic analyses revealed dedifferentiation of Paneth cells in Slc7a5 ΔIEC mice, leading to markedly reduced secretory granules with little effect on Paneth cell number. We further show that Slc7a5 ΔIEC mice are prone to experimental colitis. Thus, SLC7A5 regulates secretory cell differentiation to affect stem cell niche and/or inflammatory response to regulate cell proliferation.
Collapse
|
10
|
Kaur H, Moreau R. Raptor knockdown concurrently increases the electrical resistance and paracellular permeability of Caco-2 cell monolayers. Life Sci 2022; 308:120989. [PMID: 36152680 DOI: 10.1016/j.lfs.2022.120989] [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: 07/13/2022] [Revised: 09/12/2022] [Accepted: 09/19/2022] [Indexed: 11/18/2022]
Abstract
AIMS As a critical regulatory point of nutrient sensing, growth and metabolism, the mechanistic target of rapamycin complex 1 (mTORC1) is poised to influence intestinal homeostasis under basal conditions and in disease state. Intestinal barrier integrity ensures tissue homeostasis by closely regulating the permeability of the epithelium to lumenal contents. The role of mTORC1 in the regulation of intestinal barrier function and permeability remains to be fully elucidated. MATERIALS AND METHODS In this study, we employed lentivirus-mediated knockdown of mTORC1 signaling-associated proteins Raptor (regulatory-associated protein of mTOR) and TSC2 (tuberin) to ascertain the effects of constitutive activation or repression of mTORC1 activity on barrier function in Caco-2 cell monolayers. KEY FINDINGS Results showed that the loss of Raptor concomitantly raised the transepithelial electrical resistance (TEER) and para/transcellular permeability leading to a cell monolayer that is leaky for dextran yet electrically resistant to the movement of ions. Paracellular permeability was linked to the downregulation of tight junction protein expression and enhanced autophagy. Raptor-depleted cells had the highest abundance of myosin binding subunit MYPT1 concomitantly with the lowest abundance of p-MYPT1 (Thr696) and phosphorylated myosin light chain (p-MLC, Ser19) implying that MLC phosphatase activity was increased resulting in MLC relaxation. Although rapamycin suppressed mTORC1 activity and decreased the abundance of tight junction proteins in control cells, rapamycin caused a modest increase of TEER compared to Raptor knockdown. SIGNIFICANCE The study showed that epithelium paracellular permeability of small molecular weight dextran is dissociated from TEER.
Collapse
Affiliation(s)
- Harleen Kaur
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Régis Moreau
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA.
| |
Collapse
|
11
|
Steinberger AE, Tecos ME, Phelps HM, Rubin DC, Davidson NO, Guo J, Warner BW. A novel maladaptive unfolded protein response as a mechanism for small bowel resection-induced liver injury. Am J Physiol Gastrointest Liver Physiol 2022; 323:G165-G176. [PMID: 35727920 PMCID: PMC9377788 DOI: 10.1152/ajpgi.00302.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 05/10/2022] [Accepted: 05/10/2022] [Indexed: 01/31/2023]
Abstract
The unfolded protein response (UPR) is a complex adaptive signaling pathway activated by the accumulation of misfolded proteins in the endoplasmic reticulum (ER). ER stress (ERS) triggers a cascade of responses that converge upon C/EBP homologous protein (CHOP) to drive inflammation and apoptosis. Herein, we sought to determine whether liver injury and fibrosis after small bowel resection (SBR) were mediated by a maladaptive hepatic ERS/UPR. C57BL/6 mice underwent 50% proximal SBR or sham operation. Markers of liver injury and UPR/ERS pathways were analyzed. These were compared with experimental groups including dietary fat manipulation, tauroursodeoxycholic acid (TUDCA) treatment, distal SBR, and global CHOP knockout (KO). At 10 wk, proximal SBR had elevated alanine aminotransferase/aspartate aminotransferase (ALT/AST) (P < 0.005) and greater hepatic tumor necrosis factor-α (TNFα) (P = 0.001) and collagen type 1 α1 (COL1A1) (P = 0.02) than shams. SBR livers had increased CHOP and p-eIF2α, but were absent in activating transcription factor 4 (ATF4) protein expression. Low-fat diet (LFD), TUDCA, and distal SBR groups had decreased liver enzymes, inflammation, and fibrosis (P < 0.05). Importantly, they demonstrated reversal of hepatic UPR with diminished CHOP and robust ATF4 signal. CHOP KO-SBR had decreased ALT but not AST compared with wild-type (WT)-SBR (P = 0.01, P = 0.12). There were no differences in TNFα and COL1A1 (P = 0.09, P = 0.50). SBR-induced liver injury, fibrosis is associated with a novel hepatic UPR/ERS response characterized by increased CHOP and decreased ATF4. LFD, TUDCA, and ileocecal resection rescued the hepatic phenotype and reversed the UPR pattern. Global CHOP KO only partially attenuated liver injury. This underscores the significance of disruptions to the gut/liver axis after SBR and potentiates targets to mitigate the progression of intestinal failure-associated liver disease.NEW & NOTEWORTHY The unfolded protein response (UPR) is a complex signaling cascade that converges upon C/EBP-homologous protein (CHOP). Under conditions of chronic cellular stress, the UPR shifts from homeostatic to proapoptotic leading to inflammation and cell death. Here, we provide evidence that small bowel resection-induced liver injury and fibrosis are mediated by a maladaptive hepatic UPR. Low-fat diet, TUDCA treatment, and ileocecal resection rescued the hepatic phenotype and reversed the UPR pattern.
Collapse
Affiliation(s)
- Allie E Steinberger
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Maria E Tecos
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska
| | - Hannah M Phelps
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Deborah C Rubin
- Division of Gastroenterology, Department of Medicine, Washington University, St. Louis, Missouri
| | - Nicholas O Davidson
- Division of Gastroenterology, Department of Medicine, Washington University, St. Louis, Missouri
| | - Jun Guo
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Brad W Warner
- Division of Pediatric Surgery, Department of Surgery, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, Missouri
| |
Collapse
|
12
|
Araujo A, Safronova A, Burger E, López-Yglesias A, Giri S, Camanzo ET, Martin AT, Grivennikov S, Yarovinsky F. IFN-γ mediates Paneth cell death via suppression of mTOR. eLife 2021; 10:e60478. [PMID: 34633285 PMCID: PMC8570691 DOI: 10.7554/elife.60478] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 10/10/2021] [Indexed: 12/12/2022] Open
Abstract
Paneth cells constitutively produce antimicrobial peptides and growth factors that allow for intestinal homeostasis, host protection, and intestinal stem cell replication. Paneth cells rely heavily on the glycolytic metabolic program, which is in part controlled by the kinase complex Mechanistic target of rapamycin (mTORC1). Yet, little is known about mTOR importance in Paneth cell integrity under steady-state and inflammatory conditions. Our results demonstrate that IFN-γ, a crucial mediator of the intestinal inflammation, acts directly on murine Paneth cells to alter their mitochondrial integrity and membrane potential, resulting in an TORC1-dependent cell death mechanism distinct from canonical cell death pathways including apoptosis, necroptosis, and pyroptosis. These results were established with the purified cytokine and a physiologically relevant common Th1-inducing human parasite Toxoplasma gondii. Given the crucial role for IFN-γ, which is a cytokine frequently associated with the development of inflammatory bowel disease and compromised Paneth cell functions, the identified mechanisms underlying mTORC1-dependent Paneth cell death downstream of IFN-γ may provide promising novel approaches for treating intestinal inflammation.
Collapse
Affiliation(s)
- Alessandra Araujo
- Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical CenterNew YorkUnited States
| | - Alexandra Safronova
- Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical CenterNew YorkUnited States
| | - Elise Burger
- Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical CenterNew YorkUnited States
| | - Américo López-Yglesias
- Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical CenterNew YorkUnited States
| | - Shilpi Giri
- Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical CenterNew YorkUnited States
| | - Ellie T Camanzo
- Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical CenterNew YorkUnited States
| | - Andrew T Martin
- Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical CenterNew YorkUnited States
| | - Sergei Grivennikov
- Department of Medicine and Department of Biomedical Sciences, Cedars-Sinai Medical CenterLos AngelesUnited States
- Cancer Prevention and Control Program, Fox Chase Cancer CenterPhiladelphiaUnited States
| | - Felix Yarovinsky
- Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical CenterNew YorkUnited States
| |
Collapse
|
13
|
Chaaban H, Burge K, Eckert J, Trammell M, Dyer D, Keshari RS, Silasi R, Regmi G, Lupu C, Good M, McElroy SJ, Lupu F. Acceleration of Small Intestine Development and Remodeling of the Microbiome Following Hyaluronan 35 kDa Treatment in Neonatal Mice. Nutrients 2021; 13:2030. [PMID: 34204790 PMCID: PMC8231646 DOI: 10.3390/nu13062030] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/07/2021] [Accepted: 06/09/2021] [Indexed: 12/26/2022] Open
Abstract
The beneficial effects of human milk suppressing the development of intestinal pathologies such as necrotizing enterocolitis in preterm infants are widely known. Human milk (HM) is rich in a multitude of bioactive factors that play major roles in promoting postnatal maturation, differentiation, and the development of the microbiome. Previous studies showed that HM is rich in hyaluronan (HA) especially in colostrum and early milk. This study aims to determine the role of HA 35 KDa, a HM HA mimic, on intestinal proliferation, differentiation, and the development of the intestinal microbiome. We show that oral HA 35 KDa supplementation for 7 days in mouse pups leads to increased villus length and crypt depth, and increased goblet and Paneth cells, compared to controls. We also show that HA 35 KDa leads to an increased predominance of Clostridiales Ruminococcaceae, Lactobacillales Lactobacillaceae, and Clostridiales Lachnospiraceae. In seeking the mechanisms involved in the changes, bulk RNA seq was performed on samples from the terminal ileum and identified upregulation in several genes essential for cellular growth, proliferation, and survival. Taken together, this study shows that HA 35 KDa supplemented to mouse pups promotes intestinal epithelial cell proliferation, as well as the development of Paneth cells and goblet cell subsets. HA 35 KDa also impacted the intestinal microbiota; the implications of these responses need to be determined.
Collapse
Affiliation(s)
- Hala Chaaban
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.B.); (J.E.)
| | - Kathryn Burge
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.B.); (J.E.)
| | - Jeffrey Eckert
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.B.); (J.E.)
| | - MaJoi Trammell
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (M.T.); (D.D.)
| | - David Dyer
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (M.T.); (D.D.)
| | - Ravi S. Keshari
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (R.S.K.); (R.S.); (G.R.); (C.L.); (F.L.)
| | - Robert Silasi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (R.S.K.); (R.S.); (G.R.); (C.L.); (F.L.)
| | - Girija Regmi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (R.S.K.); (R.S.); (G.R.); (C.L.); (F.L.)
| | - Cristina Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (R.S.K.); (R.S.); (G.R.); (C.L.); (F.L.)
| | - Misty Good
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA;
| | - Steven J. McElroy
- Department of Microbiology and Immunology, Stead Family Department of Pediatrics, University of Iowa, Iowa City, IA 52242, USA;
| | - Florea Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (R.S.K.); (R.S.); (G.R.); (C.L.); (F.L.)
| |
Collapse
|
14
|
Xie Y, Zhao Y, Shi L, Li W, Chen K, Li M, Chen X, Zhang H, Li T, Matsuzawa-Ishimoto Y, Yao X, Shao D, Ke Z, Li J, Chen Y, Zhang X, Cui J, Cui S, Leng Q, Cadwell K, Li X, Wei H, Zhang H, Li H, Xiao H. Gut epithelial TSC1/mTOR controls RIPK3-dependent necroptosis in intestinal inflammation and cancer. J Clin Invest 2020; 130:2111-2128. [PMID: 31961824 DOI: 10.1172/jci133264] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 01/14/2020] [Indexed: 02/06/2023] Open
Abstract
Although Western diet and dysbiosis are the most prominent environmental factors associated with inflammatory bowel diseases (IBDs), the corresponding host factors and cellular mechanisms remain poorly defined. Here we report that the TSC1/mTOR pathway in the gut epithelium represents a metabolic and innate immune checkpoint for intestinal dysfunction and inflammation. mTOR hyperactivation triggered by Western diet or Tsc1 ablation led to epithelium necroptosis, barrier disruption, and predisposition to dextran sulfate sodium-induced colitis and inflammation-associated colon cancer. Mechanistically, our results uncovered a critical role for TSC1/mTOR in restraining the expression and activation of RIPK3 in the gut epithelium through TRIM11-mediated ubiquitination and autophagy-dependent degradation. Notably, microbiota depletion by antibiotics or gnotobiotics attenuated RIPK3 expression and activation, thereby alleviating epithelial necroptosis and colitis driven by mTOR hyperactivation. mTOR primarily impinged on RIPK3 to potentiate necroptosis induced by TNF and by microbial pathogen-associated molecular patterns (PAMPs), and hyperactive mTOR and aberrant necroptosis were intertwined in human IBDs. Together, our data reveal a previously unsuspected link between the Western diet, microbiota, and necroptosis and identify the mTOR/RIPK3/necroptosis axis as a driving force for intestinal inflammation and cancer.
Collapse
Affiliation(s)
- Yadong Xie
- The Center for Microbes, Development and Health, Chinese Academy of Sciences (CAS) Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai; CAS Center for Excellence in Molecular Cell Science; University of Chinese Academy of Sciences, CAS, Shanghai, China.,Center for Allergic and Inflammatory Diseases & Department of Otolaryngology, Head and Neck Surgery, Affiliated Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China
| | - Yifan Zhao
- The Center for Microbes, Development and Health, Chinese Academy of Sciences (CAS) Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai; CAS Center for Excellence in Molecular Cell Science; University of Chinese Academy of Sciences, CAS, Shanghai, China
| | - Lei Shi
- The Center for Microbes, Development and Health, Chinese Academy of Sciences (CAS) Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai; CAS Center for Excellence in Molecular Cell Science; University of Chinese Academy of Sciences, CAS, Shanghai, China
| | - Wei Li
- The Center for Microbes, Development and Health, Chinese Academy of Sciences (CAS) Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai; CAS Center for Excellence in Molecular Cell Science; University of Chinese Academy of Sciences, CAS, Shanghai, China
| | - Kun Chen
- Center for Allergic and Inflammatory Diseases & Department of Otolaryngology, Head and Neck Surgery, Affiliated Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China
| | - Min Li
- Center for Allergic and Inflammatory Diseases & Department of Otolaryngology, Head and Neck Surgery, Affiliated Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China
| | - Xia Chen
- The Center for Microbes, Development and Health, Chinese Academy of Sciences (CAS) Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai; CAS Center for Excellence in Molecular Cell Science; University of Chinese Academy of Sciences, CAS, Shanghai, China
| | - Haiwei Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, CAS, Shanghai, China
| | - Tiantian Li
- The Center for Microbes, Development and Health, Chinese Academy of Sciences (CAS) Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai; CAS Center for Excellence in Molecular Cell Science; University of Chinese Academy of Sciences, CAS, Shanghai, China
| | - Yu Matsuzawa-Ishimoto
- Kimmel Center for Biology and Medicine, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, New York, USA
| | - Xiaomin Yao
- Kimmel Center for Biology and Medicine, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, New York, USA
| | - Dianhui Shao
- The Center for Microbes, Development and Health, Chinese Academy of Sciences (CAS) Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai; CAS Center for Excellence in Molecular Cell Science; University of Chinese Academy of Sciences, CAS, Shanghai, China
| | - Zunfu Ke
- Department of Pathology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jian Li
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Yan Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, CAS, Shanghai, China
| | - Xiaoming Zhang
- The Center for Microbes, Development and Health, Chinese Academy of Sciences (CAS) Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai; CAS Center for Excellence in Molecular Cell Science; University of Chinese Academy of Sciences, CAS, Shanghai, China
| | - Jun Cui
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shuzhong Cui
- State Key Laboratory of Respiratory Diseases, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Qibin Leng
- State Key Laboratory of Respiratory Diseases, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ken Cadwell
- Kimmel Center for Biology and Medicine, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, New York, USA
| | - Xiaoxia Li
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Hong Wei
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Haibing Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, CAS, Shanghai, China
| | - Huabin Li
- Center for Allergic and Inflammatory Diseases & Department of Otolaryngology, Head and Neck Surgery, Affiliated Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China.,Department of Pathology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hui Xiao
- The Center for Microbes, Development and Health, Chinese Academy of Sciences (CAS) Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai; CAS Center for Excellence in Molecular Cell Science; University of Chinese Academy of Sciences, CAS, Shanghai, China
| |
Collapse
|
15
|
Onufer EJ, Aladegbami B, Imai T, Seiler K, Bajinting A, Courtney C, Sutton S, Bustos A, Yao J, Yeh CH, Sescleifer A, Wang LV, Guo J, Warner BW. EGFR in enterocytes & endothelium and HIF1α in enterocytes are dispensable for massive small bowel resection induced angiogenesis. PLoS One 2020; 15:e0236964. [PMID: 32931498 PMCID: PMC7491746 DOI: 10.1371/journal.pone.0236964] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 07/16/2020] [Indexed: 12/15/2022] Open
Abstract
Background Short bowel syndrome (SBS) results from significant loss of small intestinal length. In response to this loss, adaptation occurs, with Epidermal Growth Factor Receptor (EGFR) being a key driver. Besides enhanced enterocyte proliferation, we have revealed that adaptation is associated with angiogenesis. Further, we have found that small bowel resection (SBR) is associated with diminished oxygen delivery and elevated levels of hypoxia-inducible factor 1-alpha (HIF1α). Methods We ablated EGFR in the epithelium and endothelium as well as HIF1α in the epithelium, ostensibly the most hypoxic element. Using these mice, we determined the effects of these genetic manipulations on intestinal blood flow after SBR using photoacoustic microscopy (PAM), intestinal adaptation and angiogenic responses. Then, given that endothelial cells require a stromal support cell for efficient vascularization, we ablated EGFR expression in intestinal subepithelial myofibroblasts (ISEMFs) to determine its effects on angiogenesis in a microfluidic model of human small intestine. Results Despite immediate increased demand in oxygen extraction fraction measured by PAM in all mouse lines, were no differences in enterocyte and endothelial cell EGFR knockouts or enterocyte HIF1α knockouts by POD3. Submucosal capillary density was also unchanged by POD7 in all mouse lines. Additionally, EGFR silencing in ISEMFs did not impact vascular network development in a microfluidic device of human small intestine. Conclusions Overall, despite the importance of EGFR in facilitating intestinal adaptation after SBR, it had no impact on angiogenesis in three cell types–enterocytes, endothelial cells, and ISEMFs. Epithelial ablation of HIF1α also had no impact on angiogenesis in the setting of SBS.
Collapse
Affiliation(s)
- Emily J. Onufer
- Division of Pediatric Surgery, Department of Surgery, St. Louis Children’s Hospital, Washington University in St. Louis School of Medicine, St. Louis, MO, United States of America
| | - Bola Aladegbami
- Division of Pediatric Surgery, Department of Surgery, St. Louis Children’s Hospital, Washington University in St. Louis School of Medicine, St. Louis, MO, United States of America
| | - Toru Imai
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, United States of America
- Department of Electrical Engineering, Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, United States of America
| | - Kristen Seiler
- Division of Pediatric Surgery, Department of Surgery, St. Louis Children’s Hospital, Washington University in St. Louis School of Medicine, St. Louis, MO, United States of America
| | - Adam Bajinting
- Saint Louis University School of Medicine, St. Louis, MO, United States of America
| | - Cathleen Courtney
- Division of Pediatric Surgery, Department of Surgery, St. Louis Children’s Hospital, Washington University in St. Louis School of Medicine, St. Louis, MO, United States of America
| | - Stephanie Sutton
- Division of Pediatric Surgery, Department of Surgery, St. Louis Children’s Hospital, Washington University in St. Louis School of Medicine, St. Louis, MO, United States of America
| | - Aiza Bustos
- Division of Pediatric Surgery, Department of Surgery, St. Louis Children’s Hospital, Washington University in St. Louis School of Medicine, St. Louis, MO, United States of America
| | - Junjie Yao
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, United States of America
| | - Cheng-Hung Yeh
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, United States of America
| | - Anne Sescleifer
- Saint Louis University School of Medicine, St. Louis, MO, United States of America
| | - Lihong V. Wang
- Department of Electrical Engineering, Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, United States of America
| | - Jun Guo
- Division of Pediatric Surgery, Department of Surgery, St. Louis Children’s Hospital, Washington University in St. Louis School of Medicine, St. Louis, MO, United States of America
| | - Brad W. Warner
- Division of Pediatric Surgery, Department of Surgery, St. Louis Children’s Hospital, Washington University in St. Louis School of Medicine, St. Louis, MO, United States of America
- * E-mail:
| |
Collapse
|
16
|
Bohin N, McGowan KP, Keeley TM, Carlson EA, Yan KS, Samuelson LC. Insulin-like Growth Factor-1 and mTORC1 Signaling Promote the Intestinal Regenerative Response After Irradiation Injury. Cell Mol Gastroenterol Hepatol 2020; 10:797-810. [PMID: 32502530 PMCID: PMC7502577 DOI: 10.1016/j.jcmgh.2020.05.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 05/28/2020] [Accepted: 05/29/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND & AIMS Intestinal crypts have a remarkable capacity to regenerate after injury from loss of crypt base columnar (CBC) stem cells. After injury, facultative stem cells (FSCs) are activated to replenish the epithelium and replace lost CBCs. Our aim was to assess the role of insulin-like growth factor-1 (IGF-1) to activate FSCs for crypt repair. METHODS The intestinal regenerative response was measured after whole body 12-Gy γ-irradiation of adult mice. IGF-1 signaling or its downstream effector mammalian target of rapamycin complex 1 (mTORC1) was inhibited by administering BMS-754807 or rapamycin, respectively. Mice with inducible Rptor gene deletion were studied to test the role of mTORC1 signaling in the intestinal epithelium. FSC activation post-irradiation was measured by lineage tracing. RESULTS We observed a coordinate increase in growth factor expression, including IGF-1, at 2 days post-irradiation, followed by a surge in mTORC1 activity during the regenerative phase of crypt repair at day 4. IGF-1 was localized to pericryptal mesenchymal cells, and IGF-1 receptor was broadly expressed in crypt progenitor cells. Inhibition of IGF-1 signaling via BMS-754807 treatment impaired crypt regeneration after 12-Gy irradiation, with no effect on homeostasis. Similarly, rapamycin inhibition of mTORC1 during the growth factor surge blunted the regenerative response. Analysis of Villin-CreERT2;Rptorfl/fl mice showed that epithelial mTORC1 signaling was essential for crypt regeneration. Lineage tracing from Bmi1-marked cells showed that rapamycin blocked FSC activation post-irradiation. CONCLUSIONS Our study shows that IGF-1 signaling through mTORC1 drives crypt regeneration. We propose that IGF-1 release from pericryptal cells stimulates mTORC1 in FSCs to regenerate lost CBCs.
Collapse
Affiliation(s)
- Natacha Bohin
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan; Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, Michigan
| | - Kevin P McGowan
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Theresa M Keeley
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Elizabeth A Carlson
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Kelley S Yan
- Columbia Center for Human Development, Columbia Stem Cell Initiative, Departments of Medicine and Genetics and Development, Columbia University Irving Medical Center, New York, New York
| | - Linda C Samuelson
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan; Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
17
|
Seiler KM, Goo WH, Zhang Q, Courtney C, Bajinting A, Guo J, Warner BW. Adaptation of extracellular matrix to massive small bowel resection in mice. J Pediatr Surg 2020; 55:1107-1112. [PMID: 32164986 PMCID: PMC7299777 DOI: 10.1016/j.jpedsurg.2020.02.038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 02/20/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Extracellular matrix (ECM) affects cell behavior, and vice versa. How ECM changes after small bowel resection (SBR) to support adaptive cellular processes has not been described. Here we characterize changes in ECM following SBR and integrate this with concomitant transcriptional perturbations. METHODS A 50% proximal SBR or sham surgery was performed on mice. On postoperative day 7, ileal tissue was sequentially depleted of protein components to generate an ECM-enriched fraction. ECM was analyzed for protein composition using mass spectrometry with subsequent Ingenuity Pathway Analysis (IPA) to identify predicted pathways and upstream regulators. qPCR and RNA-sequencing (RNA-Seq) were performed to corroborate these predicted pathways. RESULTS 3034 proteins were differentially regulated between sham and SBR, of which 95 were significant (P < 0.05). IPA analysis predicted PPARα agonism to be an upstream regulator of the observed proteomic changes (P < 0.001). qPCR and RNA-Seq with KEGG analysis confirmed significant engagement of the PPAR pathway (P < 0.05). CONCLUSION Transcriptional signatures of adapting bowel predict subsequent ECM changes after SBR. How ECM communicates with surrounding cells to drive adaptation and vice versa merits further investigation. Our findings thus far suggest ECM supports tissue hyperplasia and altered metabolic demand following SBR.
Collapse
Affiliation(s)
- Kristen M. Seiler
- Division of Pediatric Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO
| | | | - Qiang Zhang
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, MO
| | - Cathleen Courtney
- Division of Pediatric Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO
| | - Adam Bajinting
- Division of Pediatric Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO
| | - Jun Guo
- Division of Pediatric Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO
| | - Brad W. Warner
- Division of Pediatric Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
18
|
Epidermal growth factor promotes intestinal secretory cell differentiation in weaning piglets via Wnt/ β-catenin signalling. Animal 2019; 14:790-798. [PMID: 31650938 DOI: 10.1017/s1751731119002581] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Small intestinal epithelium homeostasis involves four principal cell types: enterocytes, goblet, enteroendocrine and Paneth cells. Epidermal growth factor (EGF) has been shown to affect enterocyte differentiation. This study determined the effect of dietary EGF on goblet, enteroendocrine and Paneth cell differentiation in piglet small intestine and potential mechanisms. Forty-two weaned piglets were used in a 2 × 3 factorial design; the major factors were time post-weaning (days 7 and 14) and dietary treatment (0, 200 or 400 µg/kg EGF supplementation). The numbers of goblet and enteroendocrine cells were generally greater with the increase in time post-weaning. Moreover, the supplementation of 200 µg/kg EGF increased (P < 0.01) the number of goblet and enteroendocrine cells in villus and crypt of the piglet small intestine as compared with the control. Dietary supplementation with 200 µg/kg EGF enhanced (P < 0.05) abundances of differentiation-related genes atonal homologue 1, mucin 2 and intestinal trefoil factor 3 messenger RNA (mRNA) as compared with the control. Piglets fed 200 or 400 µg/kg EGF diet had increased (P < 0.05) abundances of growth factor-independent 1, SAM pointed domain containing ETS transcription factor and pancreatic and duodenal homeobox 1 mRNA, but decreased the abundance (P < 0.01) of E74 like ETS transcription factor 3 mRNA as compared with the control. Animals receiving 400 µg/kg EGF diets had enhanced (P < 0.05) abundances of neurogenin3 and SRY-box containing gene 9 mRNA as compared with the control. The mRNA abundance and protein expression of lysozyme, a marker of Paneth cell, were also increased (P < 0.05) in those animals. As compared with the control, dietary supplementation with 200 µg/kg EGF increased the abundance of EGF receptor mRNA and the ratio of non-phospho(p)-β-catenin/β-catenin (P < 0.05) in villus epithelial cells at days 7 and 14. This ratio in crypt epithelial cells was higher (P < 0.05) on the both 200 and 400 µg/kg EGF groups during the same period. Our results demonstrated that dietary EGF stimulated goblet, enteroendocrine and Paneth cell differentiation in piglets during the post-weaning period, partly through EGFR and Wnt/β-catenin signalling.
Collapse
|
19
|
Kaur H, Moreau R. Role of mTORC1 in intestinal epithelial repair and tumorigenesis. Cell Mol Life Sci 2019; 76:2525-2546. [PMID: 30944973 PMCID: PMC11105546 DOI: 10.1007/s00018-019-03085-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 03/08/2019] [Accepted: 03/25/2019] [Indexed: 12/15/2022]
Abstract
mTORC1 signaling is the prototypical pathway regulating protein synthesis and cell proliferation. mTORC1 is active in stem cells located at the base of intestinal crypts but silenced as transit-amplifying cells differentiate into enterocytes or secretory cells along the epithelium. After an insult or injury, self-limiting and controlled activation of mTORC1 is critical for the renewal and repair of intestinal epithelium. mTORC1 promotes epithelial cell renewal by driving cryptic stem cell division, and epithelial cell repair by supporting the dedifferentiation and proliferation of enterocytes or secretory cells. Under repeated insult or injury, mTORC1 becomes constitutively active, triggering an irreversible return to stemness, cell division, proliferation, and inflammation among dedifferentiated epithelial cells. Epithelium-derived cytokines promulgate inflammation within the lamina propria, which in turn releases inflammatory factors that act back on the epithelium where undamaged intestinal epithelial cells participate in the pervading state of inflammation and become susceptible to tumorigenesis.
Collapse
Affiliation(s)
- Harleen Kaur
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | - Régis Moreau
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA.
| |
Collapse
|
20
|
Seiler KM, Waye SE, Kong W, Kamimoto K, Bajinting A, Goo WH, Onufer EJ, Courtney C, Guo J, Warner BW, Morris SA. Single-Cell Analysis Reveals Regional Reprogramming During Adaptation to Massive Small Bowel Resection in Mice. Cell Mol Gastroenterol Hepatol 2019; 8:407-426. [PMID: 31195149 PMCID: PMC6718927 DOI: 10.1016/j.jcmgh.2019.06.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 05/29/2019] [Accepted: 06/03/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS The small intestine (SI) displays regionality in nutrient and immunological function. Following SI tissue loss (as occurs in short gut syndrome, or SGS), remaining SI must compensate, or "adapt"; the capacity of SI epithelium to reprogram its regional identity has not been described. Here, we apply single-cell resolution analyses to characterize molecular changes underpinning adaptation to SGS. METHODS Single-cell RNA sequencing was performed on epithelial cells isolated from distal SI of mice following 50% proximal small bowel resection (SBR) vs sham surgery. Single-cell profiles were clustered based on transcriptional similarity, reconstructing differentiation events from intestinal stem cells (ISCs) through to mature enterocytes. An unsupervised computational approach to score cell identity was used to quantify changes in regional (proximal vs distal) SI identity, validated using immunofluorescence, immunohistochemistry, qPCR, western blotting, and RNA-FISH. RESULTS Uniform Manifold Approximation and Projection-based clustering and visualization revealed differentiation trajectories from ISCs to mature enterocytes in sham and SBR. Cell identity scoring demonstrated segregation of enterocytes by regional SI identity: SBR enterocytes assumed more mature proximal identities. This was associated with significant upregulation of lipid metabolism and oxidative stress gene expression, which was validated via orthogonal analyses. Observed upstream transcriptional changes suggest retinoid metabolism and proximal transcription factor Creb3l3 drive proximalization of cell identity in response to SBR. CONCLUSIONS Adaptation to proximal SBR involves regional reprogramming of ileal enterocytes toward a proximal identity. Interventions bolstering the endogenous reprogramming capacity of SI enterocytes-conceivably by engaging the retinoid metabolism pathway-merit further investigation, as they may increase enteral feeding tolerance, and obviate intestinal failure, in SGS.
Collapse
Affiliation(s)
- Kristen M Seiler
- Division of Pediatric Surgery, Department of Surgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - Sarah E Waye
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, Missouri; Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, Missouri; Center of Regenerative Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - Wenjun Kong
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, Missouri; Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, Missouri; Center of Regenerative Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - Kenji Kamimoto
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, Missouri; Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, Missouri; Center of Regenerative Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - Adam Bajinting
- Division of Pediatric Surgery, Department of Surgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - William H Goo
- Division of Pediatric Surgery, Department of Surgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - Emily J Onufer
- Division of Pediatric Surgery, Department of Surgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - Cathleen Courtney
- Division of Pediatric Surgery, Department of Surgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - Jun Guo
- Division of Pediatric Surgery, Department of Surgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - Brad W Warner
- Division of Pediatric Surgery, Department of Surgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - Samantha A Morris
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, Missouri; Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, Missouri; Center of Regenerative Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri.
| |
Collapse
|
21
|
SETIAWAN JAJAR, KOTANI TAKENORI, KONNO TASUKU, SAITO YASUYUKI, MURATA YOJI, NODA TETSUO, MATOZAKI TAKASHI. Regulation of Small Intestinal Epithelial Homeostasis by Tsc2-mTORC1 Signaling. THE KOBE JOURNAL OF MEDICAL SCIENCES 2019; 64:E200-E209. [PMID: 31327863 PMCID: PMC6668652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 12/27/2018] [Indexed: 06/10/2023]
Abstract
Mammalian target of rapamycin complex 1 (mTORC1), a protein complex containing the serine/threonine kinase mTOR, integrates various growth stimulating signals. mTORC1 is expressed in intestinal epithelial cells (IECs), whereas the physiological roles of this protein complex in homeostasis of IECs remain virtually unknown. We here generated mice, in which tuberous sclerosis complex 2 (Tsc2), a negative regulator of mTORC1, was specifically ablated in IECs (Tsc2 CKO mice). Ablation of Tsc2 enhanced the phosphorylation of mTORC1 downstream molecules such as ribosomal S6 protein and 4E-BP1 in IECs. Tsc2 CKO mice manifested the enhanced proliferative activity of IECs in intestinal crypts as well as the promoted migration of these cells along the crypt-villus axis. The mutant mice also manifested the increased apoptotic rate of IECs as well as the increased ectopic Paneth cells, which are one of the major differentiated IECs. In addition, in vitro study showed that ablation of Tsc2 promoted the development of intestinal organoids without epidermal growth factor, while mTORC1 inhibitor, rapamycin, diminished this phenotype. Our results thus suggest that Tsc2-mTORC1 signaling regulates the proliferation, migration, and positioning of IECs, and thereby contributes to the proper regulation of intestinal homeostasis.
Collapse
Affiliation(s)
- JAJAR SETIAWAN
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
- Department of Physiology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - TAKENORI KOTANI
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - TASUKU KONNO
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - YASUYUKI SAITO
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - YOJI MURATA
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - TETSUO NODA
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - TAKASHI MATOZAKI
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
22
|
Onufer EJ, Tay S, Barron LK, Courtney CM, Warner BW, Guo J. Intestinal epithelial cell-specific Raptor is essential for high fat diet-induced weight gain in mice. Biochem Biophys Res Commun 2018; 505:1174-1179. [PMID: 30318117 DOI: 10.1016/j.bbrc.2018.10.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 10/05/2018] [Indexed: 11/30/2022]
Abstract
Mammalian target of rapamycin complex 1 (mTORC1) is a major regulator of cell growth and proliferation through fuel sensing. Systemic inhibition of mTOR as well as manipulation of its downstream products prevent diet-induced obesity. The purpose of this study was to determine the consequences of intestine-targeted mTORC1 inhibition. To attenuate intestinal mTORC1 activity, Villin-CreER mice were crossed with Raptorflox/flox mice, creating an intestinal-specific Raptor null line (i-Raptor -/-). Mice were fed a high fat diet (HFD) and compositional changes as well as food intake levels were assessed. Over a five-week time course, i-Raptor -/- mice consistently gained less body weight on a HFD compared to wildtype (WT) mice secondary to significantly reduced food intake. Importantly, the i-Raptor -/- mice did not appear to be malnourished, demonstrated by their preservation of lean body mass. i-Raptor -/- mice also maintained a normal metabolic profile without significant changes in triglyceride or fasting glucose levels. Further investigation revealed that GDF-15 mRNA expression was significantly enhanced in i-Raptor -/- enterocytes when refed with HFD after overnight starvation. In summary, our study establishes that loss of intestinal specific-mTORC1 is protective of the development of diet-induced obesity by reducing food intake without altering the metabolic profile.
Collapse
Affiliation(s)
- Emily J Onufer
- Division of Pediatric Surgery, Department of Surgery, St. Louis Children's Hospital, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Shirli Tay
- Division of Pediatric Surgery, Department of Surgery, St. Louis Children's Hospital, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Lauren K Barron
- Division of Pediatric Surgery, Department of Surgery, St. Louis Children's Hospital, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Cathleen M Courtney
- Division of Pediatric Surgery, Department of Surgery, St. Louis Children's Hospital, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Brad W Warner
- Division of Pediatric Surgery, Department of Surgery, St. Louis Children's Hospital, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Jun Guo
- Division of Pediatric Surgery, Department of Surgery, St. Louis Children's Hospital, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
23
|
Acharya P, Kutum R, Pandey R, Mishra A, Saha R, Munjal A, Ahuja V, Mukerji M, Makharia GK. First Degree Relatives of Patients with Celiac Disease Harbour an Intestinal Transcriptomic Signature that Might Protect them from Enterocyte Damage. Clin Transl Gastroenterol 2018; 9:195. [PMID: 30293993 PMCID: PMC6174158 DOI: 10.1038/s41424-018-0059-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 08/02/2018] [Accepted: 09/08/2018] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION Celiac disease (CeD) is an autoimmune enteropathy which affects approximately 0.7% of the global population. While first-degree relatives (FDR) of patients with CeD have a 7.5% risk of developing enteropathy, many remain protected. Therefore, intestinal mucosa of FDR might have protective compensatory mechanisms against immunological injury. We have explored the protective mechanisms that may be active in intestinal mucosa of FDR. METHODS Intestinal mucosal biopsies (4-5 pieces) from treatment naïve patients with CeD (n = 12), FDR (n = 12) (anti-tTG negative) and controls (n = 12) (anti-tTG negative) were obtained from each individual and subjected to microarray analysis using HT-12-v4 Human Expression BeadChips (Illumina). Differential gene expression analysis was carried out among CeD, FDR and controls; and resulting gene lists were analyzed using gene ontology and pathway enrichment tools. RESULTS Patients with CeD, FDR and control groups displayed significant differential gene expression. Thirty seven genes were upregulated and 372 were downregulated in the intestinal mucosa of FDR in comparison to CeD and controls. Pseudogenes constituted about 18% (315/1751) of FDR differentially expressed genes, and formed "clusters" that associated uniquely with individual study groups. The three study groups segregated into distinct clusters in unsupervised (PCA) and supervised (random forests) modelling approaches. Pathways analysis revealed an emphasis on crypt-villous maintenance and immune regulation in the intestinal mucosa of FDR. CONCLUSIONS Our analysis suggests that the intestinal mucosa of celiac FDR consist of a unique molecular phenotype that is distinct from CeD and controls. The transcriptomic landscape of FDR promotes maintenance of crypt-villous axis and modulation of immune mechanisms. These differences clearly demonstrate the existence of compensatory protective mechanisms in the FDR intestinal mucosa.
Collapse
Affiliation(s)
- Pragyan Acharya
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Rintu Kutum
- Genomics and Molecular Medicine and CSIR-TRISUTRA Ayurgenomics Unit, Council for Scientific and Industrial Research-Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research, CSIR-IGIB, Delhi, India
| | - Rajesh Pandey
- Genomics and Molecular Medicine and CSIR-TRISUTRA Ayurgenomics Unit, Council for Scientific and Industrial Research-Institute of Genomics and Integrative Biology, New Delhi, India
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Science and Innovation Campus, Oxfordshire, OX11 0RD, United Kingdom
| | - Asha Mishra
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Rohini Saha
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Akshay Munjal
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Vineet Ahuja
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Mitali Mukerji
- Genomics and Molecular Medicine and CSIR-TRISUTRA Ayurgenomics Unit, Council for Scientific and Industrial Research-Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research, CSIR-IGIB, Delhi, India
| | - Govind K Makharia
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi, 110029, India.
| |
Collapse
|
24
|
Aladegbami B, Barron L, Bao J, Colasanti J, Erwin CR, Warner BW, Guo J. Epithelial cell specific Raptor is required for initiation of type 2 mucosal immunity in small intestine. Sci Rep 2017; 7:5580. [PMID: 28717211 PMCID: PMC5514129 DOI: 10.1038/s41598-017-06070-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 06/07/2017] [Indexed: 12/13/2022] Open
Abstract
Intestinal tuft cells are one of 4 secretory cell linages in the small intestine and the source of IL-25, a critical initiator of the type 2 immune response to parasite infection. When Raptor, a critical scaffold protein for mammalian target of rapamycin complex 1 (mTORC1), was acutely deleted in intestinal epithelium via Tamoxifen injection in Tritrichomonas muris (Tm) infected mice, tuft cells, IL-25 in epithelium and IL-13 in the mesenchyme were significantly reduced, but Tm burden was not affected. When Tm infected mice were treated with rapamycin, DCLK1 and IL-25 expression in enterocytes and IL-13 expression in mesenchyme were diminished. After massive small bowel resection, tuft cells and Tm were diminished due to the diet used postoperatively. The elimination of Tm and subsequent re-infection of mice with Tm led to type 2 immune response only in WT, but Tm colonization in both WT and Raptor deficient mice. When intestinal organoids were stimulated with IL-4, tuft cells and IL-25 were induced in both WT and Raptor deficient organoids. In summary, our study reveals that enterocyte specific Raptor is required for initiating a type 2 immune response which appears to function through the regulation of mTORC1 activity.
Collapse
Affiliation(s)
- Bola Aladegbami
- Division of Pediatric Surgery, St Louis Children's Hospital, Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Lauren Barron
- Division of Pediatric Surgery, St Louis Children's Hospital, Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - James Bao
- Department of Biology, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Jason Colasanti
- Fischell Department of Bioengineering in the A. James Clark School of Engineering at the University of Maryland, College Park, MD, 20742, USA
| | - Christopher R Erwin
- Division of Pediatric Surgery, St Louis Children's Hospital, Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Brad W Warner
- Division of Pediatric Surgery, St Louis Children's Hospital, Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jun Guo
- Division of Pediatric Surgery, St Louis Children's Hospital, Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|