1
|
Prochera A, Muppirala AN, Kuziel GA, Soualhi S, Shepherd A, Sun L, Issac B, Rosenberg HJ, Karim F, Perez K, Smith KH, Archibald TH, Rakoff-Nahoum S, Hagen SJ, Rao M. Enteric glia regulate Paneth cell secretion and intestinal microbial ecology. eLife 2025; 13:RP97144. [PMID: 40227232 PMCID: PMC11996175 DOI: 10.7554/elife.97144] [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: 04/15/2025] Open
Abstract
Glial cells of the enteric nervous system (ENS) interact closely with the intestinal epithelium and secrete signals that influence epithelial cell proliferation and barrier formation in vitro. Whether these interactions are important in vivo, however, is unclear because previous studies reached conflicting conclusions (Prochera and Rao, 2023). To better define the roles of enteric glia in steady state regulation of the intestinal epithelium, we characterized the glia in closest proximity to epithelial cells and found that the majority express the gene Proteolipid protein 1 (PLP1) in both mice and humans. To test their functions using an unbiased approach, we genetically depleted PLP1+ cells in mice and transcriptionally profiled the small and large intestines. Surprisingly, glial loss had minimal effects on transcriptional programs and the few identified changes varied along the gastrointestinal tract. In the ileum, where enteric glia had been considered most essential for epithelial integrity, glial depletion did not drastically alter epithelial gene expression but caused a modest enrichment in signatures of Paneth cells, a secretory cell type important for innate immunity. In the absence of PLP1+ glia, Paneth cell number was intact, but a subset appeared abnormal with irregular and heterogenous cytoplasmic granules, suggesting a secretory deficit. Consistent with this possibility, ileal explants from glial-depleted mice secreted less functional lysozyme than controls with corresponding effects on fecal microbial composition. Collectively, these data suggest that enteric glia do not exert broad effects on the intestinal epithelium but have an essential role in regulating Paneth cell function and gut microbial ecology.
Collapse
Affiliation(s)
- Aleksandra Prochera
- Division of Gastroenterology, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical SchoolBostonUnited States
| | - Anoohya N Muppirala
- Division of Gastroenterology, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical SchoolBostonUnited States
| | - Gavin A Kuziel
- Division of Gastroenterology, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical SchoolBostonUnited States
- Division of Infectious Diseases, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical SchoolBostonUnited States
- Department of Microbiology, Harvard Medical SchoolBostonUnited States
| | - Salima Soualhi
- Division of Gastroenterology, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical SchoolBostonUnited States
| | - Amy Shepherd
- Division of Gastroenterology, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical SchoolBostonUnited States
| | - Liang Sun
- Research Computing, Department of Information Technology, Boston Children’s HospitalBostonUnited States
| | - Biju Issac
- Research Computing, Department of Information Technology, Boston Children’s HospitalBostonUnited States
| | - Harry J Rosenberg
- Division of Gastroenterology, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical SchoolBostonUnited States
- Department of Pathology, Beth Israel Deaconess Medical CenterBostonUnited States
| | - Farah Karim
- Institute of Human Nutrition, Columbia University Irving Medical CenterNew YorkUnited States
| | - Kristina Perez
- Division of Gastroenterology, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical SchoolBostonUnited States
| | - Kyle H Smith
- Department of Surgery, Beth Israel Deaconess Medical CenterBostonUnited States
| | - Tonora H Archibald
- Research Computing, Department of Information Technology, Boston Children’s HospitalBostonUnited States
| | - Seth Rakoff-Nahoum
- Division of Gastroenterology, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical SchoolBostonUnited States
- Division of Infectious Diseases, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical SchoolBostonUnited States
- Department of Microbiology, Harvard Medical SchoolBostonUnited States
| | - Susan J Hagen
- Department of Surgery, Beth Israel Deaconess Medical CenterBostonUnited States
| | - Meenakshi Rao
- Division of Gastroenterology, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical SchoolBostonUnited States
| |
Collapse
|
2
|
Lori L, Neuranter V, Lebreton C, Berthelet J, Parlato M, Michail C, Khiat A, Berrebi D, Bruneau J, Terris B, Malamut G, Hanein S, Schmitt Y, Banal C, Lima FR, Azouguene E, Ruemmele F, Talbotec C, Lambe C, Cerf-Bensussan N, Charbit-Henrion F. Loss of WNT2B Results in Epithelial Defects and Predisposes to Gastrointestinal Dysplasia in Humans. Cell Mol Gastroenterol Hepatol 2025:101514. [PMID: 40221090 DOI: 10.1016/j.jcmgh.2025.101514] [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: 07/23/2024] [Revised: 03/28/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025]
Affiliation(s)
- Leslie Lori
- Université Paris-Cité, Institut Imagine, Laboratory of Intestinal Immunity , INSERM U1163, Paris, France
| | - Valentin Neuranter
- Université Paris-Cité, Institut Imagine, Laboratory of Intestinal Immunity , INSERM U1163, Paris, France
| | - Corinne Lebreton
- Université Paris-Cité, Institut Imagine, Laboratory of Intestinal Immunity , INSERM U1163, Paris, France
| | - Jérémy Berthelet
- Université Paris Cité, CNRS, Epigenetics and Cell Fate, Paris, France
| | - Marianna Parlato
- Université Paris-Cité, Institut Imagine, Laboratory of Intestinal Immunity, INSERM U1163, Paris, France
| | - Christina Michail
- Université Paris Cité, CNRS, Epigenetics and Cell Fate, Paris, France
| | - Anis Khiat
- Université Paris-Cité, Institut Imagine, Laboratory of Intestinal Immunity, INSERM U1163, Paris, France
| | - Dominique Berrebi
- Department of Pathology, AP-HP. Centre-Université Paris Cité, Hôpital Necker-Enfants Malades, Paris, France
| | - Julie Bruneau
- Department of Pathology, AP-HP. Centre-Université Paris Cité, Hôpital Necker-Enfants Malades, Paris, France
| | - Benoit Terris
- Department of Pathology, AP-HP. Centre-Université Paris Cité, Hôpital Cochin, Paris, France
| | - Georgia Malamut
- Université Paris-Cité, Institut Imagine, Laboratory of Intestinal Immunity, INSERM U1163, Paris, France; Department of Gastroenterology, AP-HP. Centre-Université Paris Cité, Hôpital Cochin, Paris, France
| | - Sylvain Hanein
- Bioinformatic Platform, Institute of Genetic Diseases, INSERM UMR1163, Imagine, Université Paris-Cité and Structure Fédérative de Recherche Necker, Paris, France
| | - Yohann Schmitt
- Genomics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163 et INSERM US24/CNRS UAR3633, Paris Cite University, Paris, France
| | - Céline Banal
- Université Paris-Cité, iPSC Core Facility, Institut Imagine, INSERM U1163, Paris, France
| | | | - Emilie Azouguene
- Department of Genomic Medecine of Rare Diseases, AP-HP. Centre-Université Paris Cité, Hôpital Necker-Enfants Malades, Paris, France
| | - Frank Ruemmele
- Université Paris-Cité, Institut Imagine, Laboratory of Intestinal Immunity, INSERM U1163, Paris, France; Service de Gastro-entérologie et Nutrition Pédiatrique, AP-HP. Centre-Université Paris Cité, Hôpital Necker-Enfants Malades, Paris, France
| | - Cecile Talbotec
- Service de Gastro-entérologie et Nutrition Pédiatrique, AP-HP. Centre-Université Paris Cité, Hôpital Necker-Enfants Malades, Paris, France
| | - Cecile Lambe
- Université Paris-Cité, Institut Imagine, Laboratory of Intestinal Immunity, INSERM U1163, Paris, France; Service de Gastro-entérologie et Nutrition Pédiatrique, AP-HP. Centre-Université Paris Cité, Hôpital Necker-Enfants Malades, Paris, France
| | - Nadine Cerf-Bensussan
- Université Paris-Cité, Institut Imagine, Laboratory of Intestinal Immunity, INSERM U1163, Paris, France
| | - Fabienne Charbit-Henrion
- Université Paris-Cité, Institut Imagine, Laboratory of Intestinal Immunity, INSERM U1163, Paris, France; Department of Genomic Medecine of Rare Diseases , AP-HP. Centre-Université Paris Cité, Hôpital Necker-Enfants Malades, Paris, France.
| |
Collapse
|
3
|
Schwarzmueller LJ, Adam RS, Moreno LF, Nijman LE, Logiantara A, Eleonora S, Bril O, Vromans S, de Groot NE, Giugliano FP, Stepanova E, Muncan V, Elbers CC, Lenos KJ, Zwijnenburg DA, van Eijndhoven MAJ, Pegtel DM, van Neerven SM, Loayza-Puch F, Dadali T, Broom WJ, Maier MA, Koster J, Vermeulen L, Léveillé N. Identifying colorectal cancer-specific vulnerabilities in the Wnt-driven long non-coding transcriptome. Gut 2025; 74:571-585. [PMID: 39562049 PMCID: PMC12013597 DOI: 10.1136/gutjnl-2024-332752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 10/31/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND Aberrant Wnt pathway activation is a key driver of colorectal cancer (CRC) and is essential to sustain tumour growth and progression. Although the downstream protein-coding target genes of the Wnt cascade are well known, the long non-coding transcriptome has not yet been fully resolved. OBJECTIVE In this study, we aim to comprehensively reveal the Wnt-regulated long non-coding transcriptome and exploit essential molecules as novel therapeutic targets. DESIGN We used global run-on sequencing to define β-catenin-regulated long non-coding RNAs (lncRNAs) in CRC. CRISPRi dropout screens were subsequently used to establish the functional relevance of a subset of these lncRNAs for long-term expansion of CRC. RESULTS We uncovered that LINC02418 is essential for cancer cell clonogenic outgrowth. Mechanistically, LINC02418 regulates MYC expression levels to promote CRC stem cell functionality and prevent terminal differentiation. Furthermore, we developed effective small interfering RNA (siRNA)-based therapeutics to target LINC02418 RNA in vivo. CONCLUSION We propose that cancer-specific Wnt-regulated lncRNAs provide novel therapeutic opportunities to interfere with the Wnt pathway, which has so far defied effective pharmacological inhibition.
Collapse
Affiliation(s)
- Laura J Schwarzmueller
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Ronja S Adam
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Leandro F Moreno
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Lisanne E Nijman
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Adrian Logiantara
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Steven Eleonora
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Oscar Bril
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Sophie Vromans
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Nina E de Groot
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Francesca Paola Giugliano
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ekaterina Stepanova
- Translational Control and Metabolism, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Vanesa Muncan
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Clara C Elbers
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Kristiaan J Lenos
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Danny A Zwijnenburg
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | | | - Dirk Michiel Pegtel
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Sanne M van Neerven
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Fabricio Loayza-Puch
- Translational Control and Metabolism, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tulin Dadali
- Alnylam Pharmaceuticals Inc, Cambridge, Massachusetts, USA
| | - Wendy J Broom
- Alnylam Pharmaceuticals Inc, Cambridge, Massachusetts, USA
| | - Martin A Maier
- Alnylam Pharmaceuticals Inc, Cambridge, Massachusetts, USA
| | - Jan Koster
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Louis Vermeulen
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Nicolas Léveillé
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| |
Collapse
|
4
|
Nojima Y, Yao R, Suzuki T. Single-cell RNA sequencing and machine learning provide candidate drugs against drug-tolerant persister cells in colorectal cancer. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167693. [PMID: 39870146 DOI: 10.1016/j.bbadis.2025.167693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/24/2024] [Accepted: 01/22/2025] [Indexed: 01/29/2025]
Abstract
Drug resistance often stems from drug-tolerant persister (DTP) cells in cancer. These cells arise from various lineages and exhibit complex dynamics. However, effectively targeting DTP cells remains challenging. We used single-cell RNA sequencing (scRNA-Seq) data and machine learning (ML) models to identify DTP cells in patient-derived organoids (PDOs) and computationally screened candidate drugs targeting these cells in familial adenomatous polyposis (FAP), associated with a high risk of colorectal cancer. Three PDOs (benign and malignant tumor organoids and a normal organoid) were evaluated using scRNA-Seq. ML models constructed based on public scRNA-Seq data classified DTP versus non-DTP cells. Candidate drugs for DTP cells in a malignant tumor organoid were identified from public drug sensitivity data. From FAP scRNA-Seq data, a specific TC1 cell cluster in tumor organoids was identified. The ML model identified up to 36 % of TC1 cells as DTP cells, a higher proportion than those for other clusters. A viability assay using a malignant tumor organoid demonstrated that YM-155 and THZ2 exert synergistic effects with trametinib. The constructed ML model is effective for DTP cell identification based on scRNA-Seq data for FAP and provides candidate treatments. This approach may improve DTP cell targeting in the treatment of colorectal and other cancers.
Collapse
Affiliation(s)
- Yosui Nojima
- Center for Mathematical Modeling and Data Science, Osaka University, 1-3 Machikaneyama, Toyonaka, Osaka 560-8531, Japan.
| | - Ryoji Yao
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan.
| | - Takashi Suzuki
- Center for Mathematical Modeling and Data Science, Osaka University, 1-3 Machikaneyama, Toyonaka, Osaka 560-8531, Japan.
| |
Collapse
|
5
|
Hwang S, Sung SI, Kim YE, Yang M, Koh A, Ahn SY, Chang YS. Thrombin-preconditioned mesenchymal stromal cell-derived extracellular vesicles attenuate experimental necrotizing enterocolitis. Stem Cell Res Ther 2025; 16:101. [PMID: 40022236 PMCID: PMC11871789 DOI: 10.1186/s13287-025-04243-3] [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: 10/26/2024] [Accepted: 02/19/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is a critical gastrointestinal disease in preterm infants, for which no specific treatment is established. We previously demonstrated that thrombin-preconditioned mesenchymal stromal cell-derived extracellular vesicles (thMSC-EVs) enhance protection against other neonatal tissue injuries. Therefore, this study aimed to evaluate the therapeutic potential of thMSC-EVs in modified in vitro, in vivo, and organoid models of NEC. METHODS In vitro, the effects of thMSC-EVs and naïveMSC-EVs were compared in hyperosmotic, ischemic, and hypothermic (HIT)-stressed IEC-6 cells and LPS-treated peritoneal macrophages. In vivo, NEC was induced in P4 mouse pups by three cycles of formula feeding, oral LPS administration, hypoxia, and hypothermia, followed by overnight dam care. 2 × 109 thMSC-EVs were intraperitoneally administered daily for three days, and the therapeutic effects were assessed macroscopically, histologically, and biochemically. NEC mouse-derived organoids were established to evaluate the thMSC-EVs' effect in mature enterocytes. LC-MS/MS was performed to analyze the EV proteomics. RESULTS In vitro, compared with naïveMSC-EVs, thMSC-EVs significantly improved cellular viability in HIT-induced IEC-6 cells and reduced pro-inflammatory (IL-1α, IL-1β, TNF-α) but increased anti-inflammatory (TGF-b) cytokine levels in LPS-treated peritoneal macrophages. In vivo, thMSC-EVs significantly attenuated clinical symptoms, reduced intestinal damage, and retained intestinal stem cell markers, showing more significant localization in NEC-induced intestines than in healthy intestines. In NEC mouse-derived organoids, thMSC-EVs significantly increased OLFM4 and claudin-4 expression and reduced stress-related markers such as sucrase-isomaltase, defensin, and chromogranin A. Proteomic analysis revealed that thMSC-EVs were greater enriched in anti-apoptotic, anti-inflammatory, cell adhesion, and Wnt signaling pathways than naïveMSC-EVs. CONCLUSION thMSC-EVs improved cellular viability, reduced apoptosis, attenuated inflammation, and upregulated key intestinal stem cell markers, collectively suggesting their tissue-protective effects and highlighting their potential as a treatment for NEC.
Collapse
Affiliation(s)
- Sein Hwang
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06355, Republic of Korea
- Cell and Gene Therapy Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, Republic of Korea
| | - Se In Sung
- Cell and Gene Therapy Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, Republic of Korea
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Republic of Korea
| | - Young Eun Kim
- Cell and Gene Therapy Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, Republic of Korea
| | - Misun Yang
- Cell and Gene Therapy Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, Republic of Korea
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Republic of Korea
| | - Ara Koh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, South Korea
| | - So Yoon Ahn
- Cell and Gene Therapy Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, Republic of Korea
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Republic of Korea
| | - Yun Sil Chang
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06355, Republic of Korea.
- Cell and Gene Therapy Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, Republic of Korea.
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Republic of Korea.
| |
Collapse
|
6
|
Deng L, He XC, Chen S, Zhang N, Deng F, Scott A, He Y, Tsuchiya D, Smith SE, Epp M, Malloy S, Liu F, Hembree M, Mu Q, Haug JS, Malagola E, Hassan H, Petentler K, Egidy R, Maddera L, Russell J, Wang Y, Li H, Zhao C, Perera A, Wang TC, Kuo CJ, Li L. Frizzled5 controls murine intestinal epithelial cell plasticity through organization of chromatin accessibility. Dev Cell 2025; 60:352-363.e6. [PMID: 39579769 PMCID: PMC11794035 DOI: 10.1016/j.devcel.2024.10.021] [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: 05/12/2023] [Revised: 04/15/2024] [Accepted: 10/29/2024] [Indexed: 11/25/2024]
Abstract
The homeostasis of the intestinal epithelium relies on intricate yet insufficiently understood mechanisms of intestinal epithelial plasticity. Here, we elucidate the pivotal role of Frizzled5 (Fzd5), a Wnt pathway receptor, as a determinant of murine intestinal epithelial cell fate. Deletion of Fzd5 in Lgr5+ intestinal stem cells (ISCs) impairs their self-renewal, whereas its deletion in Krt19+ cells disrupts lineage generation, without affecting crypt integrity in either case. However, a broader deletion of Fzd5 across the epithelium leads to substantial crypt deterioration. Integrated analysis of single-cell RNA sequencing (scRNA-seq) and single-cell ATAC-seq (scATAC-seq) identifies that Fzd5 governs chromatin accessibility, orchestrating the regulation of stem- and lineage-related gene expression mainly in ISCs and progenitor cells. In summary, our findings provide insights into the regulatory role of Fzd5 in governing intestinal epithelial plasticity.
Collapse
Affiliation(s)
- Lu Deng
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Xi C He
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Shiyuan Chen
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Ning Zhang
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Fengyan Deng
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Allison Scott
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Yanfeng He
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Dai Tsuchiya
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Sarah E Smith
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Michael Epp
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Seth Malloy
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Fang Liu
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Mark Hembree
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Qinghui Mu
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Jeffrey S Haug
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Ermanno Malagola
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, College of Physicians and Surgeons, New York, NY 10032, USA
| | - Huzaifa Hassan
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | | | - Rhonda Egidy
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Lucinda Maddera
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Jonathon Russell
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Yan Wang
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Hua Li
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Chongbei Zhao
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Anoja Perera
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Timothy C Wang
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, College of Physicians and Surgeons, New York, NY 10032, USA
| | - Calvin J Kuo
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Linheng Li
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA; Department of Pathology and Laboratory Medicine and Division of Medical Oncology, Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
7
|
Mehra L, Bhowmik S, Makharia GK, Das P. Intestinal stem cell niche: An upcoming area of immense importance in gastrointestinal disorders. Indian J Gastroenterol 2025; 44:8-23. [PMID: 39514159 DOI: 10.1007/s12664-024-01699-8] [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: 07/17/2024] [Accepted: 09/29/2024] [Indexed: 11/16/2024]
Abstract
The intestinal stem cell (ISC) niche is vital for maintaining the integrity and function of the intestinal epithelium. ISC populations, characterized by their high proliferation and multipotency, reside within a specialized microenvironment at the base of crypts. Crypt base columnar (CBC) cells at the deepest part of crypts serve as replicating ISCs, while position 4 label-retaining cells (LRCs) located higher up in the crypts are also important for ISC maintenance during experiments. The interplay between CBCs, position 4 LRCs, transient amplifying (TA) cells and other niche components, including the pericrypt stromal cells, ensures a continuous supply of differentiated epithelial cells. Recent advancements in ISC biomarker studies have provided valuable insights into their molecular signatures, regulatory pathways and roles in the pathogenesis of intestinal disorders. Understanding the ISC niche has significant therapeutic implications, as manipulating ISC behaviors and regenerating damaged or diseased intestinal tissue show promise for novel therapeutic approaches. ISC organoids have also provided a platform for studying intestinal diseases and testing personalized therapies. This comprehensive review covers the anatomical composition, physiological regulation, ISC biomarker studies, contribution to intestinal disorder pathogenesis and potential therapeutic implications of the ISC niche.
Collapse
Affiliation(s)
- Lalita Mehra
- Department of Pathology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110 029, India
| | - Subham Bhowmik
- Department of Pathology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110 029, India
| | - Govind K Makharia
- Department of Gastroenterology and Human Nutritions, All India Institute of Medical Sciences, New Delhi, 110 029, India
| | - Prasenjit Das
- Department of Pathology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110 029, India.
| |
Collapse
|
8
|
Li C, Zhou Y, Jiang Y, Yin Z, Weiss HL, Wang Q, Evers BM. miR-27a-3p regulates intestinal cell proliferation and differentiation through Wnt/β-catenin signalling. Cell Prolif 2025; 58:e13757. [PMID: 39329245 PMCID: PMC11839187 DOI: 10.1111/cpr.13757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/04/2024] [Accepted: 09/14/2024] [Indexed: 09/28/2024] Open
Abstract
Intestinal stem cells differentiate into absorptive enterocytes, characterised by increased brush border enzymes such as intestinal alkaline phosphatase (IAP), making up the majority (95%) of the terminally differentiated cells in the villus. Loss of integrity of the intestinal epithelium plays a key role in inflammatory diseases and gastrointestinal infection. Here, we show that the intestinal microRNA (miR)-27a-3p is an important regulator of intestinal epithelial cell proliferation and enterocyte differentiation. Repression of endogenous miR-27a-3p leads to increased enterocyte differentiation and decreased intestinal epithelial cell proliferation in mouse and human small intestinal organoids. Mechanistically, miR-27a-3p regulates intestinal cell differentiation and proliferation at least in part through the regulation of retinoic acid receptor α (RXRα), a modulator of Wnt/β-catenin signalling. Repression of miR-27a-3p increases the expression of RXRα and concomitantly, decreases the expression of active β-catenin and cyclin D1. In contrast, overexpression of miR-27a-3p mimic decreases the expression of RXRα and increases the expression of active β-catenin and cyclin D1. Moreover, overexpression of the miR-27a-3p mimic results in impaired enterocyte differentiation and increases intestinal epithelial cell proliferation. These alterations were attenuated or blocked by Wnt inhibition. Our study demonstrates an miR-27a-3p/RXRα/Wnt/β-catenin pathway that is important for the maintenance of enterocyte homeostasis in the small intestine.
Collapse
Affiliation(s)
- Chang Li
- Markey Cancer Center, University of KentuckyLexingtonKentuckyUSA
| | - Yuning Zhou
- Markey Cancer Center, University of KentuckyLexingtonKentuckyUSA
| | - Yinping Jiang
- Markey Cancer Center, University of KentuckyLexingtonKentuckyUSA
| | - Zhijie Yin
- Markey Cancer Center, University of KentuckyLexingtonKentuckyUSA
| | - Heidi L. Weiss
- Markey Cancer Center, University of KentuckyLexingtonKentuckyUSA
| | - Qingding Wang
- Markey Cancer Center, University of KentuckyLexingtonKentuckyUSA
- Department of SurgeryUniversity of KentuckyLexingtonKentuckyUSA
| | - B. Mark Evers
- Markey Cancer Center, University of KentuckyLexingtonKentuckyUSA
- Department of SurgeryUniversity of KentuckyLexingtonKentuckyUSA
| |
Collapse
|
9
|
Wang W, Lokman NA, Barry SC, Oehler MK, Ricciardelli C. LGR5: An emerging therapeutic target for cancer metastasis and chemotherapy resistance. Cancer Metastasis Rev 2025; 44:23. [PMID: 39821694 PMCID: PMC11742290 DOI: 10.1007/s10555-024-10239-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 12/24/2024] [Indexed: 01/19/2025]
Abstract
Cancer stem cells play an important role in tumor progression and chemotherapy resistance. Leucine-rich G repeat-containing protein-coupled receptor 5 (LGR5) has been identified as a cancer stem cell marker in several cancer types. LGR5 is involved in cancer development and progression via several pathways including WNT/β-catenin signaling pathway. LGR5 plays a role in tumor progression by promoting cancer cell migration, invasion, metastasis, and angiogenesis in many cancers including colorectal, brain, gastric, and ovarian cancer. This review summarises the current knowledge on the expression and functional role of LGR5 in cancers, the molecular mechanisms regulated by LGR5, and the relationship between LGR5 and chemotherapy resistance. The review also includes highlights potential strategies to inhibit LGR5 expression and function. The majority of functional studies have shown that LGR5 plays an important role in promoting cancer progression, metastasis and chemotherapy resistance however, in some contexts LGR5 can also activate tumor-suppressive pathways and LGR5 negative cells can also promote cancer progression. The review highlights that targeting LGR5 is a promising anti-cancer treatment but the functional effect of LGR5 on tumor cells is complex may be dependent on cancer type, tumor microenvironment and cross-talk with other molecules in the LGR5 signaling pathway.
Collapse
Affiliation(s)
- Wanqi Wang
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, The University of Adelaide, Adelaide, 5005, Australia
| | - Noor A Lokman
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, The University of Adelaide, Adelaide, 5005, Australia
| | - Simon C Barry
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, The University of Adelaide, Adelaide, 5005, Australia
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, 5005, Australia
| | - Martin K Oehler
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, The University of Adelaide, Adelaide, 5005, Australia
- Department of Gynaecological Oncology, Royal Adelaide Hospital, Adelaide, 5000, Australia
| | - Carmela Ricciardelli
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, The University of Adelaide, Adelaide, 5005, Australia.
| |
Collapse
|
10
|
Yilmaz O, Arora K, Lee SH, Hosseini S, Chen F, Padmanabha N, Eng G, Kantekure K, Yilmaz O, Deshpande V. LGR5 as a diagnostic marker for dysplasia in serrated polyps. J Clin Pathol 2025:jcp-2024-209856. [PMID: 39788729 DOI: 10.1136/jcp-2024-209856] [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: 09/12/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025]
Abstract
AIMS WNT signalling pathway dysregulation is often a critical early component in colorectal neoplasia, particularly the chromosomal instability pathway. Using two WNT reporters, LGR5 and AXIN2, we sought to assess whether these polyps demonstrate predictable expression patterns and if these patterns show diagnostic value. METHODS We evaluated 23 adenomas (TA), 23 sessile serrated lesions (SSLs), 14 SSL with dysplasia and 38 traditional serrated adenomas (TSA). Chromogenic in situ hybridisation stains (ISH) for LGR5 and AXIN2 were performed. Reactivity was defined as strong, intermediate or weak. Upper third crypt reactivity was defined as full-thickness staining. Accentuation within ectopic crypts (ECF) was recorded. RESULTS TAs (91%) showed strong reactivity and full-thickness staining with LGR5. TSAs showed full-thickness and weak to intermediate LGR5 reactivity (79%) and ECF with LGR5 accentuation was exclusively seen in TSA. SSL showed weak LGR5 reactivity confined to the basal crypt region (100%). SSL with dysplasia also showed weak or intermediate (100%) LGR5 reactivity, but the reactivity pattern was full thickness (88%). AXIN2 expression parallels LGR5 expression (Pearson coefficient=0.63) regarding signal intensity for the examined polyp groups. CONCLUSIONS Qualitative and quantitative differences in AXIN2 and LGR5 expression assist in the diagnosis of SSL with dysplasia.
Collapse
Affiliation(s)
- Osman Yilmaz
- Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Kshtij Arora
- Massachusetts General Hospital, Wuincy, Massachusetts, USA
| | - Soo Hyun Lee
- Boston Medical Center, Boston, Massachusetts, USA
| | | | - Feidi Chen
- Harvard Medical School, Boston, Massachusetts, USA
| | - Nandan Padmanabha
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - George Eng
- Harvard Medical School, Boston, Massachusetts, USA
| | | | - Omer Yilmaz
- Massachusetts General Hospital, Boston, Massachusetts, USA
| | | |
Collapse
|
11
|
Cheng J, Wu H, Cui Y. WNT4 promotes the symmetric fission of crypt in radiation-induced intestinal epithelial regeneration. Cell Mol Biol Lett 2024; 29:158. [PMID: 39725925 DOI: 10.1186/s11658-024-00677-4] [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: 06/20/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Radiotherapy for pelvic malignant tumors inevitably causes intestinal tissue damage. The regeneration of intestinal epithelium after radiation injury relies mainly on crypt fission. However, little is known about the regulatory mechanisms of crypt fission events. METHODS The effects of WNT4 on crypt regeneration and the symmetry of crypt fission were examined using a mouse small intestinal organoid culture model. Three-dimensional (3D) reconstructed images of organoids were applied to assess the symmetry of crypt fission and Paneth cell localization upon manipulation of WNT4 expression. The effect of WNT4 on the expression of β-catenin target genes was analyzed by real-time quantitative polymerase chain reaction (RT-qPCR). The in vivo effect of WNT4 overexpression mediated by adeno-associated virus (AAV) on symmetric fission of crypt was investigated using a radiation-injured mouse model. RESULTS WNT4 has a special function of promoting symmetric fission of small intestinal crypts, although it inhibits budding, stemness, and cell proliferation on organoids. WNT4 promotes the correct localization of Paneth cells in the crypt base by regulating the expression of EphB3, thereby promoting the symmetric fission of small intestinal crypts. WNT4 negatively regulates the canonical WNT/β-catenin signaling pathway, and it promotes symmetric crypt fission in a ROR2 receptor-dependent manner. Moreover, in patients and animal models of radiation-induced intestinal injury, we found that the regenerated crypts are irregular in size and shape, Paneth cells are mislocalized, and the expression of WNT4 is decreased while EphB3 is increased. Importantly, restoration of WNT4 expression mediated by AAV effectively promotes symmetric crypt fission and thus improves the regularity of regenerating crypts in mice with radiation-induced injury. CONCLUSIONS Our study highlights the critical role of WNT4 in the regulation of crypt fission and provides WNT4 as a potential therapeutic target for radiation enteritis.
Collapse
Affiliation(s)
- Jingyang Cheng
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Haiyong Wu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Yanmei Cui
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China.
| |
Collapse
|
12
|
Reif S, Birimberg-Schwartz L, Grunewald M, Duran D, Sebbag-Sznajder N, Toledano T, Musseri M, Golan-Gerstl R. The Effect of Milk-Derived Extracellular Vesicles on Intestinal Epithelial Cell Proliferation. Int J Mol Sci 2024; 25:13519. [PMID: 39769282 PMCID: PMC11678886 DOI: 10.3390/ijms252413519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic, relapsing inflammation disorder of the gastrointestinal tract characterized by disrupted intestinal epithelial barrier function. Despite advances in treatment, including biological agents, achieving sustained remission remains challenging for many patients with IBD. This highlights the urgent need for novel therapeutic strategies. Milk-derived extracellular vesicles (MDEs) have emerged as a promising therapeutic option. In this study, we isolated and characterized MDEs and evaluated their effects on the function of intestinal epithelial cells (IECs). Using a murine model of Dextran Sulfate Sodium (DSS)-induced colitis, we observed that MDEs significantly ameliorated disease symptoms. The upregulation of β-catenin, a crucial mediator of Wnt signaling, in colonic tissues suggests that MDEs may facilitate epithelial regeneration and restore barrier function. In patient-derived colon organoids (PDCOs), MDEs were internalized and modulated the expression of key signaling molecules, such as the upregulation of β-catenin, cyclin D1, and the proliferation marker Ki67, indicating their potential to promote IEC proliferation and intestinal barrier repair. Importantly, MDEs demonstrated selective activity by downregulating β-catenin and cyclin D1 in colon cancer cells, leading to reduced proliferation. This selectivity indicates a dual therapeutic potential of MDEs for promoting healthy IEC proliferation while potentially mitigating malignancy risks.
Collapse
Affiliation(s)
- Shimon Reif
- Department of Pediatrics, Hadassah-Hebrew University Medical Center, Jerusalem 9166100, Israel; (S.R.)
| | - Liron Birimberg-Schwartz
- Hadassah Organoid Center, The Hadassah Medical Organization, Jerusalem 9166100, Israel; (L.B.-S.)
- Department of Pediatric Gastroenterology, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9166100, Israel
| | - Myriam Grunewald
- Hadassah Organoid Center, The Hadassah Medical Organization, Jerusalem 9166100, Israel; (L.B.-S.)
- Department of Developmental Biology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9166100, Israel
| | - Deborah Duran
- Hadassah Organoid Center, The Hadassah Medical Organization, Jerusalem 9166100, Israel; (L.B.-S.)
| | - Naama Sebbag-Sznajder
- Hadassah Organoid Center, The Hadassah Medical Organization, Jerusalem 9166100, Israel; (L.B.-S.)
| | - Tirtsa Toledano
- Hadassah Organoid Center, The Hadassah Medical Organization, Jerusalem 9166100, Israel; (L.B.-S.)
| | - Mirit Musseri
- Department of Pediatrics, Hadassah-Hebrew University Medical Center, Jerusalem 9166100, Israel; (S.R.)
| | - Regina Golan-Gerstl
- Department of Pediatrics, Hadassah-Hebrew University Medical Center, Jerusalem 9166100, Israel; (S.R.)
| |
Collapse
|
13
|
Yao W, Zhao Y, Yan S, Zhang H, Bao T, Bao S, Li X, Song Y. Alterations in the Microbiomes and Metabolic Profiles of the Ileal Between the Hu Sheep and East Friesian Sheep. Int J Mol Sci 2024; 25:13267. [PMID: 39769032 PMCID: PMC11675978 DOI: 10.3390/ijms252413267] [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: 11/07/2024] [Revised: 12/07/2024] [Accepted: 12/09/2024] [Indexed: 01/03/2025] Open
Abstract
The East Friesian sheep is a dairy breed known for its high fertility and high milk production and is currently one of the best dairy sheep breeds in the world. This breed is known to have a poor disease-resistant phenotype compared to Hu sheep. Gut microbiota and metabolites play a role in host disease resistance. The intestinal bacterial microbiota is essential for maintaining the health of sheep and ensuring their productive potential, and it may also help explain disease-resistant phenotypic differences related to breeds. However, the ileum microbiota and metabolite profiles of Hu sheep and East Friesian sheep have remained poorly characterized. The ileal is a significant organ in the intestinal tract, and most nutrients and minerals in food are absorbed through the small intestine. It is necessary to understand the composition of both species' ileal microbiota and metabolites using the same feeding conditions. Therefore, studying the differences in the ileal microorganisms between breeds is essential to decipher the mechanisms behind these differences and identify microorganisms that influence the disease-resistant phenotype drive of ruminants. Due to the poor disease-resistant phenotype in sheep during the weaning period, with diarrhea and other diseases most likely to occur, we selected dairy sheep that were just two months old and had recently been weaned. This study comprehensively examined differences between the ileal microbiota in a large cohort of two breeds of sheep, including six Hu sheep and six East Friesian sheep. Using 16S rRNA and non-targeted metabolomics analysis, we determined that the Hu sheep had more microorganisms, including Lactobacillus, Bifidobacterium, Streptococcus, and Limmosilactobacillus, and more metabolites, including 2,7-Dihydroxy-5-methyl-1-naphthoic acid, Leu-Pro-Glu-Phe-Tyr, dodecanoic acid, Ala-Gln-Phe-Ile-Met, and Ala-Gln-Glu-Val-His, compared to the EF sheep group. Moreover, the Hu sheep were significantly enriched in amino acid biosynthesis, fatty acid metabolites, and bile secretion compared to the EF sheep groups, which may have been the main driver of the observed differences in disease-resistant phenotypes between the Hu sheep and East Friesian sheep. In addition, we hypothesized that there may be multiple beneficial microbes and metabolites that modulate the immune response and ultimately affect disease resistance. Therefore, these findings provide insights into the mechanisms underlying disease-resistant phenotype in sheep and may provide useful information for optimizing the composition of the ileal bacterial microbiota in sheep.
Collapse
Affiliation(s)
- Wenna Yao
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot 010020, China; (W.Y.); (Y.Z.); (S.Y.); (H.Z.); (T.B.); (S.B.)
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot 010020, China
| | - Yue Zhao
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot 010020, China; (W.Y.); (Y.Z.); (S.Y.); (H.Z.); (T.B.); (S.B.)
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot 010020, China
| | - Shuo Yan
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot 010020, China; (W.Y.); (Y.Z.); (S.Y.); (H.Z.); (T.B.); (S.B.)
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot 010020, China
| | - Huimin Zhang
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot 010020, China; (W.Y.); (Y.Z.); (S.Y.); (H.Z.); (T.B.); (S.B.)
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot 010020, China
| | - Teligun Bao
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot 010020, China; (W.Y.); (Y.Z.); (S.Y.); (H.Z.); (T.B.); (S.B.)
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot 010020, China
| | - Siqin Bao
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot 010020, China; (W.Y.); (Y.Z.); (S.Y.); (H.Z.); (T.B.); (S.B.)
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot 010020, China
| | - Xihe Li
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot 010020, China; (W.Y.); (Y.Z.); (S.Y.); (H.Z.); (T.B.); (S.B.)
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot 010020, China
| | - Yongli Song
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot 010020, China; (W.Y.); (Y.Z.); (S.Y.); (H.Z.); (T.B.); (S.B.)
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot 010020, China
| |
Collapse
|
14
|
Li Y, Wang X, Huang M, Wang X, Li C, Li S, Tang Y, Yu S, Wang Y, Song W, Wu W, Liu Y, Chen YG. BMP suppresses Wnt signaling via the Bcl11b-regulated NuRD complex to maintain intestinal stem cells. EMBO J 2024; 43:6032-6051. [PMID: 39433900 PMCID: PMC11612440 DOI: 10.1038/s44318-024-00276-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 09/22/2024] [Accepted: 10/02/2024] [Indexed: 10/23/2024] Open
Abstract
Lgr5+ intestinal stem cells (ISCs) are crucial for the intestinal epithelium renewal and regeneration after injury. However, the mechanism underlying the interplay between Wnt and BMP signaling in this process is not fully understood. Here we report that Bcl11b, which is downregulated by BMP signaling, enhances Wnt signaling to maintain Lgr5+ ISCs and thus promotes the regeneration of the intestinal epithelium upon injury. Loss of Bcl11b function leads to a significant decrease of Lgr5+ ISCs in both intestinal crypts and cultured organoids. Mechanistically, BMP suppresses the expression of Bcl11b, which can positively regulate Wnt target genes by inhibiting the function of the Nucleosome Remodeling and Deacetylase (NuRD) complex and facilitating the β-catenin-TCF4 interaction. Bcl11b can also promote intestinal epithelium repair after injuries elicited by both irradiation and DSS-induced inflammation. Furthermore, Bcl11b deletion prevents proliferation and tumorigenesis of colorectal cancer cells. Together, our findings suggest that BMP suppresses Wnt signaling via Bcl11b regulation, thus balancing homeostasis and regeneration in the intestinal epithelium.
Collapse
Affiliation(s)
- Yehua Li
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xiaodan Wang
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Meimei Huang
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xu Wang
- Guangzhou National Laboratory, Guangzhou, 510700, China
| | - Chunlin Li
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Siqi Li
- Guangzhou National Laboratory, Guangzhou, 510700, China
| | - Yuhui Tang
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Shicheng Yu
- Guangzhou National Laboratory, Guangzhou, 510700, China
| | - Yalong Wang
- Guangzhou National Laboratory, Guangzhou, 510700, China
| | - Wanglu Song
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Wei Wu
- MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yuan Liu
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- Guangzhou National Laboratory, Guangzhou, 510700, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
| |
Collapse
|
15
|
Liu G, Fang Y, Zhang Y, Zhu M. Dihydroquercetin improves the proliferation of porcine intestinal epithelial cells via the Wnt/β-catenin pathway. Biochem Biophys Res Commun 2024; 734:150460. [PMID: 39083968 DOI: 10.1016/j.bbrc.2024.150460] [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: 07/19/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
Dihydroquercetin (DHQ), also known as Taxifolin (TA), is a flavanonol with various biological activities, such as anticancer, anti-inflammatory, and antioxidative properties. It has been found to effectively increase the viability of porcine intestinal epithelial cells (IPEC-J2). However, the precise mechanism by which DHQ increases the proliferation of IPEC-J2 cells is not entirely understood. This study aimed to explore the potential pathways through which DHQ encourages the proliferation of IPEC-J2 cells. The findings indicated that DHQ significantly improved the protein expression of tight junction proteins (ZO-1, Occludin, and Claudin1) and a molecular biomarker of proliferation (PCNA) in IPEC-J2 cells. Furthermore, DHQ was found to increase the Wnt/β-catenin pathway-associated β-catenin, c-Myc, and cyclin D1 mRNA expression, and promote the protein expression of β-catenin and TCF4. To confirm the involvement of the Wnt/β-catenin signaling pathway in the DHQ-promoted proliferation of IPEC-J2 cells, the inhibitor LF3, which targets β-catenin/TCF4 interaction, was used. It was found that LF3 inhibited the protein expressions upregulated by DHQ and blocked the promotion of cell proliferation. These results indicate that DHQ positively regulates IPEC-J2 cell proliferation through the Wnt/β-catenin pathway, providing constructive insights into the role of DHQ in regulating intestine development.
Collapse
Affiliation(s)
- Guowei Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, 550025, China; Institute of Animal Nutrition and Feed Science, Guizhou University, Guiyang, 550025, China
| | - Yongxia Fang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, 550025, China; Institute of Animal Nutrition and Feed Science, Guizhou University, Guiyang, 550025, China
| | - Yiyu Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, 550025, China; Institute of Animal Nutrition and Feed Science, Guizhou University, Guiyang, 550025, China
| | - Min Zhu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, 550025, China; Institute of Animal Nutrition and Feed Science, Guizhou University, Guiyang, 550025, China.
| |
Collapse
|
16
|
Kim M, Park Y, Kim YS, Ko S. Cellular Plasticity in Gut and Liver Regeneration. Gut Liver 2024; 18:949-960. [PMID: 39081200 PMCID: PMC11565004 DOI: 10.5009/gnl240005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 06/07/2024] [Accepted: 06/21/2024] [Indexed: 11/16/2024] Open
Abstract
The intestine and liver share a unique regenerative property that sets them apart from other mammalian visceral organs. The intestinal epithelium exhibits rapid renewal, making it one of the fastest renewing tissues in humans. Under physiological conditions, intestinal stem cells within each intestinal crypt continuously differentiate into the different types of intestinal epithelial cells to maintain intestinal homeostasis. However, when exposed to tissue damage or stressful conditions such as inflammation, intestinal epithelial cells in the gastrointestinal tract exhibit plasticity, allowing fully differentiated cells to regain their stem cell properties. Likewise, hepatic epithelial cells possess a remarkable regenerative capacity to restore lost liver mass through proliferation-mediated liver regeneration. When the proliferation-mediated regenerative capacity is impaired, hepatocytes and biliary epithelial cells (BECs) can undergo plasticity-mediated regeneration and replenish each other. The transition of mammalian liver progenitor cells to hepatocytes/BECs can be observed under tightly controlled experimental conditions such as severe hepatocyte injury accompanied by the loss of regenerative capacity. In this review, we will discuss the mechanism by which cellular plasticity contributes to the regeneration process and the potential therapeutic implications of understanding and harnessing cellular plasticity in the gut and liver.
Collapse
Affiliation(s)
- Minwook Kim
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yoojeong Park
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - You Sun Kim
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Internal Medicine, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea
| | - Sungjin Ko
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
17
|
Chen SM, Guo BJ, Feng AQ, Wang XL, Zhang SL, Miao CY. Pathways regulating intestinal stem cells and potential therapeutic targets for radiation enteropathy. MOLECULAR BIOMEDICINE 2024; 5:46. [PMID: 39388072 PMCID: PMC11467144 DOI: 10.1186/s43556-024-00211-0] [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: 07/22/2024] [Accepted: 09/19/2024] [Indexed: 10/12/2024] Open
Abstract
Radiotherapy is a pivotal intervention for cancer patients, significantly impacting their treatment outcomes and survival prospects. Nevertheless, in the course of treating those with abdominal, pelvic, or retroperitoneal malignant tumors, the procedure inadvertently exposes adjacent intestinal tissues to radiation, posing risks of radiation-induced enteropathy upon reaching threshold doses. Stem cells within the intestinal crypts, through their controlled proliferation and differentiation, support the critical functions of the intestinal epithelium, ensuring efficient nutrient absorption while upholding its protective barrier properties. Intestinal stem cells (ISCs) regulation is intricately orchestrated by diverse signaling pathways, among which are the WNT, BMP, NOTCH, EGF, Hippo, Hedgehog and NF-κB, each contributing to the complex control of these cells' behavior. Complementing these pathways are additional regulators such as nutrient metabolic states, and the intestinal microbiota, all of which contribute to the fine-tuning of ISCs behavior in the intestinal crypts. It is the harmonious interplay among these signaling cascades and modulating elements that preserves the homeostasis of intestinal epithelial cells (IECs), thereby ensuring the gut's overall health and function. This review delves into the molecular underpinnings of how stem cells respond in the context of radiation enteropathy, aiming to illuminate potential biological targets for therapeutic intervention. Furthermore, we have compiled a summary of several current treatment methodologies. By unraveling these mechanisms and treatment methods, we aspire to furnish a roadmap for the development of novel therapeutics, advancing our capabilities in mitigating radiation-induced intestinal damage.
Collapse
Affiliation(s)
- Si-Min Chen
- Department of Pharmacology, Second Military Medical University/Naval Medical University, 325 Guo He Road, Shanghai, 200433, China
| | - Bing-Jie Guo
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - An-Qiang Feng
- Department of Digestive Disease, Xuzhou Central Hospital, Xuzhou, China
| | - Xue-Lian Wang
- School of Medicine, Shanghai University, Shanghai, China
| | - Sai-Long Zhang
- Department of Pharmacology, Second Military Medical University/Naval Medical University, 325 Guo He Road, Shanghai, 200433, China.
| | - Chao-Yu Miao
- Department of Pharmacology, Second Military Medical University/Naval Medical University, 325 Guo He Road, Shanghai, 200433, China.
| |
Collapse
|
18
|
Bugacov H, Der B, Briantseva BM, Guo Q, Kim S, Lindström NO, McMahon AP. Dose-dependent responses to canonical Wnt transcriptional complexes in the regulation of mammalian nephron progenitors. Development 2024; 151:dev202279. [PMID: 39250420 PMCID: PMC11463962 DOI: 10.1242/dev.202279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 09/02/2024] [Indexed: 09/11/2024]
Abstract
In vivo and in vitro studies argue that concentration-dependent Wnt signaling regulates mammalian nephron progenitor cell (NPC) programs. Canonical Wnt signaling is regulated through the stabilization of β-catenin, a transcriptional co-activator when complexed with Lef/Tcf DNA-binding partners. Using the GSK3β inhibitor CHIR99021 (CHIR) to block GSK3β-dependent destruction of β-catenin, we examined dose-dependent responses to β-catenin in mouse NPCs, using mRNA transduction to modify gene expression. Low CHIR-dependent proliferation of NPCs was blocked on β-catenin removal, with evidence of NPCs arresting at the G2-M transition. While NPC identity was maintained following β-catenin removal, mRNA-seq identified low CHIR and β-catenin dependent genes. High CHIR activated nephrogenesis. Nephrogenic programming was dependent on Lef/Tcf factors and β-catenin transcriptional activity. Molecular and cellular features of early nephrogenesis were driven in the absence of CHIR by a mutated stabilized form of β-catenin. Chromatin association studies indicate low and high CHIR response genes are likely direct targets of canonical Wnt transcriptional complexes. Together, these studies provide evidence for concentration-dependent Wnt signaling in the regulation of NPCs and provide new insight into Wnt targets initiating mammalian nephrogenesis.
Collapse
Affiliation(s)
- Helena Bugacov
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Balint Der
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
- Department of Urology, Faculty of Medicine, Semmelweis University, Budapest 1082, Hungary
- Institute of Translational Medicine, Faculty of Medicine, Semmelweis University, Budapest 1094, Hungary
| | - Bohdana-Myroslava Briantseva
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Qiuyu Guo
- Discovery Biomarkers, Amgen Research, 1 Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Sunghyun Kim
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Nils O. Lindström
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Andrew P. McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
19
|
Wang Y, Ge H, Chen P, Wang Y. Wnt/β-catenin signaling in corneal epithelium development, homeostasis, and pathobiology. Exp Eye Res 2024; 246:110022. [PMID: 39117134 DOI: 10.1016/j.exer.2024.110022] [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: 04/18/2024] [Revised: 07/07/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
The corneal epithelium is located on the most anterior surface of the eyeball and protects against external stimuli. The development of the corneal epithelium and the maintenance of corneal homeostasis are essential for the maintenance of visual acuity. It has been discovered recently via the in-depth investigation of ocular surface illnesses that the Wnt/β-catenin signaling pathway is necessary for the growth and stratification of corneal epithelial cells as well as the control of endothelial cell stability. In addition, the Wnt/β-catenin signaling pathway is directly linked to the development of common corneal illnesses such as keratoconus, fungal keratitis, and corneal neovascularization. This review mainly summarizes the role of the Wnt/β-catenin signaling pathway in the development, homeostasis, and pathobiology of cornea, hoping to provide new insights into the study of corneal epithelium and the treatment of related diseases.
Collapse
Affiliation(s)
- Yihui Wang
- School of Basic Medicine, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Huanhuan Ge
- School of Basic Medicine, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Peng Chen
- School of Basic Medicine, Qingdao University, Qingdao 266071, Shandong Province, China; Institute of Stem Cell Regeneration Medicine, School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
| | - Ye Wang
- Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, Shandong 266042, China.
| |
Collapse
|
20
|
Wu H, Mu C, Li X, Fan W, Shen L, Zhu W. Breed-Driven Microbiome Heterogeneity Regulates Intestinal Stem Cell Proliferation via Lactobacillus-Lactate-GPR81 Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400058. [PMID: 38937989 PMCID: PMC11434115 DOI: 10.1002/advs.202400058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/20/2024] [Indexed: 06/29/2024]
Abstract
Genetically lean and obese individuals have distinct intestinal microbiota and function. However, the underlying mechanisms of the microbiome heterogeneity and its regulation on epithelial function such as intestinal stem cell (ISC) fate remain unclear. Employing pigs of genetically distinct breeds (obese Meishan and lean Yorkshire), this study reveals transcriptome-wide variations in microbial ecology of the jejunum, characterized by enrichment of active Lactobacillus species, notably the predominant Lactobacillus amylovorus (L. amylovorus), and lactate metabolism network in obese breeds. The L. amylovorus-dominant heterogeneity is paralleled with epithelial functionality difference as reflected by highly expressed GPR81, more proliferative ISCs and activated Wnt/β-catenin signaling. Experiments using in-house developed porcine jejunal organoids prove that live L. amylovorus and its metabolite lactate promote intestinal organoid growth. Mechanistically, L. amylovorus and lactate activate Wnt/β-catenin signaling in a GPR81-dependent manner to promote ISC-mediated epithelial proliferation. However, heat-killed L. amylovorus fail to cause these changes. These findings uncover a previously underrepresented role of L. amylovorus in regulating jejunal stem cells via Lactobacillus-lactate-GPR81 axis, a key mechanism bridging breed-driven intestinal microbiome heterogeneity with ISC fate. Thus, results from this study provide new insights into the role of gut microbiome and stem cell interactions in maintaining intestinal homeostasis.
Collapse
Affiliation(s)
- Haiqin Wu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China
| | - Chunlong Mu
- Food Informatics, AgResearch, Te Ohu Rangahau Kai, Palmerston North, 4474, New Zealand
| | - Xuan Li
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China
| | - Wenlu Fan
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China
| | - Le Shen
- Department of Surgery, The University of Chicago, Maryland Ave, 60637, USA
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China
| |
Collapse
|
21
|
Chen C, Cao Z, Lei H, Zhang C, Wu M, Huang S, Li X, Xie D, Liu M, Zhang L, Chen G. Microbial Tryptophan Metabolites Ameliorate Ovariectomy-Induced Bone Loss by Repairing Intestinal AhR-Mediated Gut-Bone Signaling Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404545. [PMID: 39041942 PMCID: PMC11423200 DOI: 10.1002/advs.202404545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 06/03/2024] [Indexed: 07/24/2024]
Abstract
Microbial tryptophan (Trp) metabolites acting as aryl hydrocarbon receptor (AhR) ligands are shown to effectively improve metabolic diseases via regulating microbial community. However, the underlying mechanisms by which Trp metabolites ameliorate bone loss via gut-bone crosstalk are largely unknown. In this study, supplementation with Trp metabolites, indole acetic acid (IAA), and indole-3-propionic acid (IPA), markedly ameliorate bone loss by repairing intestinal barrier integrity in ovariectomy (OVX)-induced postmenopausal osteoporosis mice in an AhR-dependent manner. Mechanistically, intestinal AhR activation by Trp metabolites, especially IAA, effectively repairs intestinal barrier function by stimulating Wnt/β-catenin signaling pathway. Consequently, enhanced M2 macrophage by supplementation with IAA and IPA secrete large amount of IL-10 that expands from intestinal lamina propria to bone marrow, thereby simultaneously promoting osteoblastogenesis and inhibiting osteoclastogenesis in vivo and in vitro. Interestingly, supplementation with Trp metabolites exhibit negligible ameliorative effects on both gut homeostasis and bone loss of OVX mice with intestinal AhR knockout (VillinCreAhrfl/fl). These findings suggest that microbial Trp metabolites may be potential therapeutic candidates against osteoporosis via regulating AhR-mediated gut-bone axis.
Collapse
Affiliation(s)
- Chuan Chen
- State Key Laboratory of Magnetic Resonance and ImagingNational Centre for Magnetic Resonance in WuhanInnovation Academy of Precision Measurement Science and TechnologyCASWuhan430071China
- University of Chinese Academy of SciencesBeijing100049China
| | - Zheng Cao
- State Key Laboratory of Magnetic Resonance and ImagingNational Centre for Magnetic Resonance in WuhanInnovation Academy of Precision Measurement Science and TechnologyCASWuhan430071China
- University of Chinese Academy of SciencesBeijing100049China
| | - Hehua Lei
- State Key Laboratory of Magnetic Resonance and ImagingNational Centre for Magnetic Resonance in WuhanInnovation Academy of Precision Measurement Science and TechnologyCASWuhan430071China
- University of Chinese Academy of SciencesBeijing100049China
| | - Cui Zhang
- State Key Laboratory of Magnetic Resonance and ImagingNational Centre for Magnetic Resonance in WuhanInnovation Academy of Precision Measurement Science and TechnologyCASWuhan430071China
- University of Chinese Academy of SciencesBeijing100049China
| | - Mengjing Wu
- State Key Laboratory of Magnetic Resonance and ImagingNational Centre for Magnetic Resonance in WuhanInnovation Academy of Precision Measurement Science and TechnologyCASWuhan430071China
- University of Chinese Academy of SciencesBeijing100049China
| | - Shaohua Huang
- Institute of Drug Discovery and TechnologyNingbo UniversityNingbo315211China
| | - Xinzhi Li
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyMacau999078China
| | - Denghui Xie
- Department of Joint SurgeryCenter for Orthopaedic SurgeryThe Third Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Maili Liu
- State Key Laboratory of Magnetic Resonance and ImagingNational Centre for Magnetic Resonance in WuhanInnovation Academy of Precision Measurement Science and TechnologyCASWuhan430071China
- University of Chinese Academy of SciencesBeijing100049China
| | - Limin Zhang
- State Key Laboratory of Magnetic Resonance and ImagingNational Centre for Magnetic Resonance in WuhanInnovation Academy of Precision Measurement Science and TechnologyCASWuhan430071China
- University of Chinese Academy of SciencesBeijing100049China
| | - Gang Chen
- Department of GeriatricsHubei Provincial Hospital of Traditional Chinese Medicine (Affiliated Hospital of Hubei University of Chinese Medicine)Wuhan430060China
| |
Collapse
|
22
|
Xie Z, Yun Y, Yu G, Zhang X, Zhang H, Wang T, Zhang L. Bacillus subtilis alleviates excessive apoptosis of intestinal epithelial cells in intrauterine growth restriction suckling piglets via the members of Bcl-2 and caspase families. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:6924-6932. [PMID: 38597265 DOI: 10.1002/jsfa.13525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 02/15/2024] [Accepted: 04/10/2024] [Indexed: 04/11/2024]
Abstract
BACKGROUND The intestine is a barrier resisting various stress responses. Intrauterine growth restriction (IUGR) can cause damage to the intestinal barrier via destroying the balance of intestinal epithelial cells' proliferation and apoptosis. Bacillus subtilis has been reported to regulate intestinal epithelial cells' proliferation and apoptosis. Thus, the purpose of this study was to determine if B. subtilis could regulate intestinal epithelial cells' proliferation and apoptosis in intrauterine growth restriction suckling piglets. RESULTS Compared with the normal birth weight group, the IUGR group showed greater mean optical density values of Ki-67-positive cells in the ileal crypt (P < 0.05). IUGR resulted in higher ability of proliferation and apoptosis of intestinal epithelial cells, by upregulation of the messenger RNA (mRNA) or proteins expression of leucine rich repeat containing G protein coupled receptor 5, Caspase-3, Caspase-7, β-catenin, cyclinD1, B-cell lymphoma-2 associated agonist of cell death, and BCL2 associated X (P < 0.05), and downregulation of the mRNA or protein expression of B-cell lymphoma-2 and B-cell lymphoma-2-like 1 (P < 0.05). However, B. subtilis supplementation decreased the mRNA or proteins expression of leucine rich repeat containing G protein coupled receptor 5, SPARC related modular calcium binding 2, tumor necrosis factor receptor superfamily member 19, cyclinD1, Caspase-7, β-catenin, B-cell lymphoma-2 associated agonist of cell death, and Caspase-3 (P < 0.05), and increased the mRNA expression of B-cell lymphoma-2 (P < 0.05). CONCLUSION IUGR led to excessive apoptosis of intestinal epithelial cells, which induced compensatory proliferation. However, B. subtilis treatment prevented intestinal epithelial cells of IUGR suckling piglets from excessive apoptosis. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Zechen Xie
- College of Animal Science and Technology, Nanjing Agriculture University, Nanjing, China
| | - Yang Yun
- College of Animal Science and Technology, Nanjing Agriculture University, Nanjing, China
| | - Ge Yu
- College of Animal Science and Technology, Nanjing Agriculture University, Nanjing, China
| | - Xin Zhang
- College of Animal Science and Technology, Nanjing Agriculture University, Nanjing, China
| | - Hao Zhang
- College of Animal Science and Technology, Nanjing Agriculture University, Nanjing, China
| | - Tian Wang
- College of Animal Science and Technology, Nanjing Agriculture University, Nanjing, China
| | - Lili Zhang
- College of Animal Science and Technology, Nanjing Agriculture University, Nanjing, China
| |
Collapse
|
23
|
Sun H, Shami Shah A, Chiu DC, Bonfini A, Buchon N, Baskin JM. Wnt/β-catenin signaling within multiple cell types dependent upon kramer regulates Drosophila intestinal stem cell proliferation. iScience 2024; 27:110113. [PMID: 38952681 PMCID: PMC11215309 DOI: 10.1016/j.isci.2024.110113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/08/2024] [Accepted: 05/23/2024] [Indexed: 07/03/2024] Open
Abstract
The gut epithelium is subject to constant renewal, a process reliant upon intestinal stem cell (ISC) proliferation that is driven by Wnt/β-catenin signaling. Despite the importance of Wnt signaling within ISCs, the relevance of Wnt signaling within other gut cell types and the underlying mechanisms that modulate Wnt signaling in these contexts remain incompletely understood. Using challenge of the Drosophila midgut with a non-lethal enteric pathogen, we examine the cellular determinants of ISC proliferation, harnessing kramer, a recently identified regulator of Wnt signaling pathways, as a mechanistic tool. We find that Wnt signaling within Prospero-positive cells supports ISC proliferation and that kramer regulates Wnt signaling in this context by antagonizing kelch, a Cullin-3 E3 ligase adaptor that mediates Dishevelled polyubiquitination. This work establishes kramer as a physiological regulator of Wnt/β-catenin signaling in vivo and suggests enteroendocrine cells as a new cell type that regulates ISC proliferation via Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Hongyan Sun
- Weill Institute for Cell & Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | - Adnan Shami Shah
- Weill Institute for Cell & Molecular Biology, Cornell University, Ithaca, NY 14853, USA
- Department of Chemistry & Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Din-Chi Chiu
- Weill Institute for Cell & Molecular Biology, Cornell University, Ithaca, NY 14853, USA
- Department of Chemistry & Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Alessandro Bonfini
- Cornell Institute of Host Microbe Interactions and Disease, Department of Entomology, Cornell University, Ithaca, NY 14853, USA
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining 314400, P.R. China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Nicolas Buchon
- Cornell Institute of Host Microbe Interactions and Disease, Department of Entomology, Cornell University, Ithaca, NY 14853, USA
| | - Jeremy M. Baskin
- Weill Institute for Cell & Molecular Biology, Cornell University, Ithaca, NY 14853, USA
- Department of Chemistry & Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
24
|
Ando T, Takazawa I, Spencer ZT, Ito R, Tomimori Y, Mikulski Z, Matsumoto K, Ishitani T, Denson LA, Kawakami Y, Kawakami Y, Kitaura J, Ahmed Y, Kawakami T. Ileal Crohn's Disease Exhibits Reduced Activity of Phospholipase C-β3-Dependent Wnt/β-Catenin Signaling Pathway. Cells 2024; 13:986. [PMID: 38891118 PMCID: PMC11171731 DOI: 10.3390/cells13110986] [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: 05/06/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Crohn's disease is a chronic, debilitating, inflammatory bowel disease. Here, we report a critical role of phospholipase C-β3 (PLC-β3) in intestinal homeostasis. In PLC-β3-deficient mice, exposure to oral dextran sodium sulfate induced lethality and severe inflammation in the small intestine. The lethality was due to PLC-β3 deficiency in multiple non-hematopoietic cell types. PLC-β3 deficiency resulted in reduced Wnt/β-catenin signaling, which is essential for homeostasis and the regeneration of the intestinal epithelium. PLC-β3 regulated the Wnt/β-catenin pathway in small intestinal epithelial cells (IECs) at transcriptional, epigenetic, and, potentially, protein-protein interaction levels. PLC-β3-deficient IECs were unable to respond to stimulation by R-spondin 1, an enhancer of Wnt/β-catenin signaling. Reduced expression of PLC-β3 and its signature genes was found in biopsies of patients with ileal Crohn's disease. PLC-β regulation of Wnt signaling was evolutionally conserved in Drosophila. Our data indicate that a reduction in PLC-β3-mediated Wnt/β-catenin signaling contributes to the pathogenesis of ileal Crohn's disease.
Collapse
Affiliation(s)
- Tomoaki Ando
- Laboratory of Allergic Diseases, Center for Autoimmunity and Inflammation, La Jolla, CA 92037, USA; (T.A.)
- Atopy Research Center, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Ikuo Takazawa
- Laboratory of Allergic Diseases, Center for Autoimmunity and Inflammation, La Jolla, CA 92037, USA; (T.A.)
| | - Zachary T. Spencer
- Department of Molecular and Systems Biology and the Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth College, Hanover, NH 03755, USA; (Z.T.S.)
| | - Ryoji Ito
- Laboratory of Allergic Diseases, Center for Autoimmunity and Inflammation, La Jolla, CA 92037, USA; (T.A.)
- Central Institute for Experimental Animals, Kawasaki 210-0821, Kanagawa, Japan
| | - Yoshiaki Tomimori
- Laboratory of Allergic Diseases, Center for Autoimmunity and Inflammation, La Jolla, CA 92037, USA; (T.A.)
| | - Zbigniew Mikulski
- Imaging Facility, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Kenji Matsumoto
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Tohru Ishitani
- Institute for Molecular and Cellular Regulation, Gunma University, Maebashi 371-0044, Gunma, Japan
| | - Lee A. Denson
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Yu Kawakami
- Laboratory of Allergic Diseases, Center for Autoimmunity and Inflammation, La Jolla, CA 92037, USA; (T.A.)
| | - Yuko Kawakami
- Laboratory of Allergic Diseases, Center for Autoimmunity and Inflammation, La Jolla, CA 92037, USA; (T.A.)
| | - Jiro Kitaura
- Atopy Research Center, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Yashi Ahmed
- Department of Molecular and Systems Biology and the Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth College, Hanover, NH 03755, USA; (Z.T.S.)
| | - Toshiaki Kawakami
- Laboratory of Allergic Diseases, Center for Autoimmunity and Inflammation, La Jolla, CA 92037, USA; (T.A.)
| |
Collapse
|
25
|
Verma M, Garg M, Khan AS, Yadav P, Rahman SS, Ali A, Kamthan M. Cadmium modulates intestinal Wnt/β-catenin signaling ensuing intestinal barrier disruption and systemic inflammation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 277:116337. [PMID: 38640798 DOI: 10.1016/j.ecoenv.2024.116337] [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: 01/18/2024] [Revised: 03/14/2024] [Accepted: 04/12/2024] [Indexed: 04/21/2024]
Abstract
The intricate architecture of the intestinal epithelium, crucial for nutrient absorption, is constantly threatened by environmental factors. The epithelium undergoes rapid turnover, which is essential for maintaining homeostasis, under the control of intestinal stem cells (ISCs). The central regulator, Wnt/β-catenin signaling plays a key role in intestinal integrity and turnover. Despite its significance, the impact of environmental factors on this pathway has been largely overlooked. This study, for the first time, investigates the influence of Cd on the intestinal Wnt signaling pathway using a mouse model. In this study, male BALB/c mice were administered an environmentally relevant Cd dose (0.98 mg/kg) through oral gavage to investigate the intestinal disruption and Wnt signaling pathway. Various studies, including histopathology, immunohistochemistry, RT-PCR, western blotting, ELISA, intestinal permeability assay, and flow cytometry, were conducted to study Cd-induced changes in the intestine. The canonical Wnt signaling pathway experienced significant downregulation as a result of sub-chronic Cd exposure, which caused extensive damage throughout the small intestine. Increased intestinal permeability and a skewed immune response were also observed. To confirm that Wnt signaling downregulation is the key driver of Cd-induced gastrointestinal toxicity, mice were co-exposed to LiCl (a recognized Wnt activator) and Cd. The results clearly showed that the harmful effects of Cd could be reversed, which is strong evidence that Cd mostly damages the intestine through the Wnt/β-catenin signalling axis. In conclusion, this research advances the current understanding of the role of Wnt/β catenin signaling in gastrointestinal toxicity caused by diverse environmental pollutants.
Collapse
Affiliation(s)
- Muskan Verma
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, Hamdard Nagar, New Delhi 110062, India
| | - Manika Garg
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, Hamdard Nagar, New Delhi 110062, India
| | - Aiysha Siddiq Khan
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, Hamdard Nagar, New Delhi 110062, India
| | - Pawan Yadav
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, Hamdard Nagar, New Delhi 110062, India
| | - Saman Saim Rahman
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, Hamdard Nagar, New Delhi 110062, India
| | - Asghar Ali
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, Hamdard Nagar, New Delhi 110062, India
| | - Mohan Kamthan
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, Hamdard Nagar, New Delhi 110062, India.
| |
Collapse
|
26
|
Cox CM, Wu MH, Padilla-Rodriguez M, Blum I, Momtaz S, Mitchell SAT, Wilson JM. Regulation of YAP and Wnt signaling by the endosomal protein MAMDC4. PLoS One 2024; 19:e0296003. [PMID: 38787854 PMCID: PMC11125477 DOI: 10.1371/journal.pone.0296003] [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: 03/27/2023] [Accepted: 12/04/2023] [Indexed: 05/26/2024] Open
Abstract
Maintenance of the intestinal epithelium requires constant self-renewal and regeneration. Tight regulation of proliferation and differentiation of intestinal stem cells within the crypt region is critical to maintaining homeostasis. The transcriptional co-factors β-catenin and YAP are required for proliferation during normal homeostasis as well as intestinal regeneration after injury: aberrant signaling activity results in over proliferation and tumorigenesis. Although both YAP and β-catenin activity are controlled along canonical pathways, it is becoming increasingly clear that non-canonical regulation of these transcriptional regulators plays a role in fine tuning their activity. We have shown previously that MAMDC4 (Endotubin, AEGP), an integral membrane protein present in endosomes, regulates both YAP and β-catenin activity in kidney epithelial cells and in the developing intestinal epithelium. Here we show that MAMDC4 interacts with members of the signalosome and mediates cross-talk between YAP and β-catenin. Interestingly, this cross-talk occurs through a non-canonical pathway involving interactions between AMOT:YAP and AMOT:β-catenin.
Collapse
Affiliation(s)
- Christopher M. Cox
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States of America
| | - Meng-Han Wu
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States of America
| | - Marco Padilla-Rodriguez
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States of America
| | - Isabella Blum
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States of America
| | - Samina Momtaz
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States of America
| | - Stefanie A. T. Mitchell
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States of America
| | - Jean M. Wilson
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States of America
- The University of Arizona Cancer Center, University of Arizona, Tucson, AZ, United States of America
- Bio5 Institute, University of Arizona, Tucson, AZ, United States of America
| |
Collapse
|
27
|
Hu X, Yuan X, Zhang G, Song H, Ji P, Guo Y, Liu Z, Tian Y, Shen R, Wang D. The intestinal epithelial-macrophage-crypt stem cell axis plays a crucial role in regulating and maintaining intestinal homeostasis. Life Sci 2024; 344:122452. [PMID: 38462226 DOI: 10.1016/j.lfs.2024.122452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/18/2024] [Accepted: 01/18/2024] [Indexed: 03/12/2024]
Abstract
The intestinal tract plays a vital role in both digestion and immunity, making its equilibrium crucial for overall health. This equilibrium relies on the dynamic interplay among intestinal epithelial cells, macrophages, and crypt stem cells. Intestinal epithelial cells play a pivotal role in protecting and regulating the gut. They form vital barriers, modulate immune responses, and engage in pathogen defense and cytokine secretion. Moreover, they supervise the regulation of intestinal stem cells. Macrophages, serving as immune cells, actively influence the immune response through the phagocytosis of pathogens and the release of cytokines. They also contribute to regulating intestinal stem cells. Stem cells, known for their self-renewal and differentiation abilities, play a vital role in repairing damaged intestinal epithelium and maintaining homeostasis. Although research has primarily concentrated on the connections between epithelial and stem cells, interactions with macrophages have been less explored. This review aims to fill this gap by exploring the roles of the intestinal epithelial-macrophage-crypt stem cell axis in maintaining intestinal balance. It seeks to unravel the intricate dynamics and regulatory mechanisms among these essential players. A comprehensive understanding of these cell types' functions and interactions promises insights into intestinal homeostasis regulation. Moreover, it holds potential for innovative approaches to manage conditions like radiation-induced intestinal injury, inflammatory bowel disease, and related diseases.
Collapse
Affiliation(s)
- Xiaohui Hu
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province 73000, China.
| | - Xinyi Yuan
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province 73000, China.
| | - Guokun Zhang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province 73000, China.
| | - Haoyun Song
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province 73000, China.
| | - Pengfei Ji
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province 73000, China.
| | - Yanan Guo
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province 73000, China.
| | - Zihua Liu
- Lanzhou University Second Hospital, Lanzhou University, Lanzhou, Gansu Province 73000, China
| | - Yixiao Tian
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province 73000, China.
| | - Rong Shen
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province 73000, China.
| | - Degui Wang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province 73000, China; NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Lanzhou, Gansu Province 730000, China.
| |
Collapse
|
28
|
Bowes MM, Casares-Marfil D, Sawalha AH. Intestinal permeability correlates with disease activity and DNA methylation changes in lupus patients. Clin Immunol 2024; 262:110173. [PMID: 38460891 PMCID: PMC11009052 DOI: 10.1016/j.clim.2024.110173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/30/2024] [Accepted: 02/16/2024] [Indexed: 03/11/2024]
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE or lupus) is a chronic autoimmune disease that can involve various organ systems. Several studies have suggested that increased intestinal permeability may play a role in the pathogenesis of lupus. The aim of this study was to elucidate the relationship between intestinal permeability, disease activity, and epigenetic changes in lupus patients. METHODS A total of 25 female lupus patients were included in this study. Systemic Lupus Erythematosus Disease Activity Index (SLEDAI) scores were used as indicator of disease activity. Plasma zonulin levels were measured, using an ELISA, as a marker of intestinal permeability. Genome-wide DNA methylation patterns were assessed in neutrophils for 19 of the lupus patients using the Infinium MethylationEPIC array. Linear regression and Pearson's correlation were used to evaluate the correlation between zonulin concentrations and SLEDAI scores. The relationship between DNA methylation levels and zonulin concentrations was assessed using beta regression, linear regression, and Pearson's correlation, adjusting for age and race. RESULTS Intestinal permeability positively correlated with disease activity in lupus patients (p-value = 7.60 × 10-3, r = 0.53). DNA methylation levels in 926 CpG sites significantly correlated with intestinal permeability. The highest correlation was identified in LRIG1 (cg14159396, FDR-adjusted p-value = 1.35 × 10-12, adjusted r2 = 0.92), which plays a role in intestinal homeostasis. Gene Ontologies related to cell-cell adhesion were enriched among the genes that were hypomethylated with increased intestinal permeability in lupus. CONCLUSION Our data suggest a correlation between increased intestinal permeability and disease activity in lupus patients. Further, increased intestinal permeability might be associated with epigenetic changes that could play a role in the pathogenesis of lupus.
Collapse
Affiliation(s)
- Mckenna M Bowes
- Division of Rheumatology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Desiré Casares-Marfil
- Division of Rheumatology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Amr H Sawalha
- Division of Rheumatology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA; Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Lupus Center of Excellence, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
29
|
Kang EJ, Kim JH, Kim YE, Lee H, Jung KB, Chang DH, Lee Y, Park S, Lee EY, Lee EJ, Kang HB, Rhyoo MY, Seo S, Park S, Huh Y, Go J, Choi JH, Choi YK, Lee IB, Choi DH, Seo YJ, Noh JR, Kim KS, Hwang JH, Jeong JS, Kwon HJ, Yoo HM, Son MY, Kim YG, Lee DH, Kim TY, Kwon HJ, Kim MH, Kim BC, Kim YH, Kang D, Lee CH. The secreted protein Amuc_1409 from Akkermansia muciniphila improves gut health through intestinal stem cell regulation. Nat Commun 2024; 15:2983. [PMID: 38582860 PMCID: PMC10998920 DOI: 10.1038/s41467-024-47275-8] [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: 10/03/2023] [Accepted: 03/26/2024] [Indexed: 04/08/2024] Open
Abstract
Akkermansia muciniphila has received great attention because of its beneficial roles in gut health by regulating gut immunity, promoting intestinal epithelial development, and improving barrier integrity. However, A. muciniphila-derived functional molecules regulating gut health are not well understood. Microbiome-secreted proteins act as key arbitrators of host-microbiome crosstalk through interactions with host cells in the gut and are important for understanding host-microbiome relationships. Herein, we report the biological function of Amuc_1409, a previously uncharacterised A. muciniphila-secreted protein. Amuc_1409 increased intestinal stem cell (ISC) proliferation and regeneration in ex vivo intestinal organoids and in vivo models of radiation- or chemotherapeutic drug-induced intestinal injury and natural aging with male mice. Mechanistically, Amuc_1409 promoted E-cadherin/β-catenin complex dissociation via interaction with E-cadherin, resulting in the activation of Wnt/β-catenin signaling. Our results demonstrate that Amuc_1409 plays a crucial role in intestinal homeostasis by regulating ISC activity in an E-cadherin-dependent manner and is a promising biomolecule for improving and maintaining gut health.
Collapse
Affiliation(s)
- Eun-Jung Kang
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Department of Veterinary Pathology, College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Jae-Hoon Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Livestock Products Analysis Division, Division of Animal health, Daejeon Metropolitan City Institute of Health and Environment, Daejeon, 34146, Republic of Korea
| | - Young Eun Kim
- Group for Biometrology, Korea Research Institute of Standards and Science (KRISS), Daejeon, 34113, Republic of Korea
- School of Earth Sciences & Environmental Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Hana Lee
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Kwang Bo Jung
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Dong-Ho Chang
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Youngjin Lee
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Shinhye Park
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Eun-Young Lee
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Eun-Ji Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Ho Bum Kang
- Synthetic Biology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Moon-Young Rhyoo
- Laboratory Animal Resource Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Seungwoo Seo
- School of Earth Sciences & Environmental Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Sohee Park
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Department of Bio-Molecular Science, Korea Research Institute of Bioscience and Biotechnology (KRIBB) School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34141, Republic of Korea
| | - Yubin Huh
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Department of Bio-Molecular Science, Korea Research Institute of Bioscience and Biotechnology (KRIBB) School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34141, Republic of Korea
| | - Jun Go
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Jung Hyeon Choi
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Young-Keun Choi
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - In-Bok Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Dong-Hee Choi
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Yun Jeong Seo
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Jung-Ran Noh
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Kyoung-Shim Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Department of Functional Genomics, Korea Research Institute of Bioscience and Biotechnology (KRIBB) School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34141, Republic of Korea
| | - Jung Hwan Hwang
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Department of Functional Genomics, Korea Research Institute of Bioscience and Biotechnology (KRIBB) School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34141, Republic of Korea
| | - Ji-Seon Jeong
- Group for Biometrology, Korea Research Institute of Standards and Science (KRISS), Daejeon, 34113, Republic of Korea
- Department of Measurement Science, Korea Research Institute of Standards and Science (KRISS) School of Precision Measurement, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Ha-Jeong Kwon
- Group for Biometrology, Korea Research Institute of Standards and Science (KRISS), Daejeon, 34113, Republic of Korea
| | - Hee Min Yoo
- Group for Biometrology, Korea Research Institute of Standards and Science (KRISS), Daejeon, 34113, Republic of Korea
- Department of Measurement Science, Korea Research Institute of Standards and Science (KRISS) School of Precision Measurement, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Mi-Young Son
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Department of Bio-Molecular Science, Korea Research Institute of Bioscience and Biotechnology (KRIBB) School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34141, Republic of Korea
| | - Yeon-Gu Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Department of Applied Biological Engineering, Korea Research Institute of Bioscience and Biotechnology (KRIBB) School of Biotechnology, University of Science and Technology (UST), Daejeon, 34141, Republic of Korea
| | - Dae-Hee Lee
- Synthetic Biology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Department of Biosystems and Bioengineering, Korea Research Institute of Bioscience and Biotechnology (KRIBB) School of Biotechnology, University of Science and Technology (UST), Daejeon, 34141, Republic of Korea
| | - Tae-Young Kim
- School of Earth Sciences & Environmental Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Hyo-Jung Kwon
- Department of Veterinary Pathology, College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Myung Hee Kim
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Byoung-Chan Kim
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- HealthBiome Inc., Daejeon, 34141, Republic of Korea
| | - Yong-Hoon Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea.
- Department of Functional Genomics, Korea Research Institute of Bioscience and Biotechnology (KRIBB) School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34141, Republic of Korea.
| | - Dukjin Kang
- Group for Biometrology, Korea Research Institute of Standards and Science (KRISS), Daejeon, 34113, Republic of Korea.
| | - Chul-Ho Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea.
- Department of Functional Genomics, Korea Research Institute of Bioscience and Biotechnology (KRIBB) School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34141, Republic of Korea.
| |
Collapse
|
30
|
Czowski BJ, White KA. Intracellular pH regulates β-catenin with low pHi increasing adhesion and signaling functions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.22.586349. [PMID: 38585883 PMCID: PMC10996556 DOI: 10.1101/2024.03.22.586349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Intracellular pH (pHi) dynamics are linked to cell processes including proliferation, migration, and differentiation. The adherens junction (AJ) and signaling protein β-catenin has decreased abundance at high pHi due to increased proteasomal-mediated degradation. However, the effects of low pHi on β-catenin abundance and functions have not been characterized. Here, we show that low pHi stabilizes β-catenin in epithelial cells using population-level and single-cell assays. β-catenin abundance is increased at low pHi and decreased at high pHi. We also assay single-cell protein degradation rates to show that β-catenin half-life is longer at low compared to high pHi. Importantly, we show that AJs are not disrupted by β-catenin loss at high pHi due to rescue by plakoglobin. Finally, we show that low pHi increases β-catenin transcriptional activity in single cells and is indistinguishable from a Wnt-on state. This work characterizes pHi as a rheostat regulating β-catenin abundance, stability, and function and implicates β-catenin as a molecular mediator of pHi-dependent cell processes.
Collapse
Affiliation(s)
- Brandon J Czowski
- Department of Chemistry and Biochemistry, University of Notre Dame
- Harper Cancer Research Institute, University of Notre Dame
| | - Katharine A White
- Department of Chemistry and Biochemistry, University of Notre Dame
- Harper Cancer Research Institute, University of Notre Dame
| |
Collapse
|
31
|
Kang M, Armenteros JJA, Gulati GS, Gleyzer R, Avagyan S, Brown EL, Zhang W, Usmani A, Earland N, Wu Z, Zou J, Fields RC, Chen DY, Chaudhuri AA, Newman AM. Mapping single-cell developmental potential in health and disease with interpretable deep learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.19.585637. [PMID: 38562882 PMCID: PMC10983880 DOI: 10.1101/2024.03.19.585637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Single-cell RNA sequencing (scRNA-seq) has transformed our understanding of cell fate in developmental systems. However, identifying the molecular hallmarks of potency - the capacity of a cell to differentiate into other cell types - has remained challenging. Here, we introduce CytoTRACE 2, an interpretable deep learning framework for characterizing potency and differentiation states on an absolute scale from scRNA-seq data. Across 31 human and mouse scRNA-seq datasets encompassing 28 tissue types, CytoTRACE 2 outperformed existing methods for recovering experimentally determined potency levels and differentiation states covering the entire range of cellular ontogeny. Moreover, it reconstructed the temporal hierarchy of mouse embryogenesis across 62 timepoints; identified pan-tissue expression programs that discriminate major potency levels; and facilitated discovery of cellular phenotypes in cancer linked to survival and immunotherapy resistance. Our results illuminate a fundamental feature of cell biology and provide a broadly applicable platform for delineating single-cell differentiation landscapes in health and disease.
Collapse
|
32
|
Riva F, Draghi S, Inglesi A, Filipe J, Cremonesi P, Lavazza A, Cavadini P, Vigo D, Agradi S, Menchetti L, Di Giancamillo A, Aidos L, Modina SC, Fehri NE, Pastorelli G, Serra V, Balzaretti CM, Castrica M, Severgnini M, Brecchia G, Curone G. Bovine Colostrum Supplementation in Rabbit Diet Modulates Gene Expression of Cytokines, Gut-Vascular Barrier, and Red-Ox-Related Molecules in the Gut Wall. Animals (Basel) 2024; 14:800. [PMID: 38473185 DOI: 10.3390/ani14050800] [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: 01/03/2024] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Rabbits, pivotal in the EU as livestock, pets, and experimental animals, face bacterial infection challenges, prompting a quest for alternatives to curb antibiotic resistance. Bovine colostrum (BC), rich in immunoregulatory compounds, antimicrobial peptides, and growth factors, is explored for disease treatment and prevention. This study assesses BC diet supplementation effects on rabbit intestines, examining gene expression. Thirty female New Zealand White rabbits at weaning (35 days) were divided into three experimental groups: control (commercial feed), 2.5% BC, and 5% BC. The diets were administered until slaughtering (81 days). BC-upregulated genes in the jejunum included IL-8, TGF-β, and CTNN-β1 at 5% BC, while PLVAP at 2.5% BC. Antioxidant-related genes (SOD1, GSR) were downregulated in the cecum and colon with 2.5% BC. BC 5% promoted IL-8 in the jejunum, fostering inflammation and immune cell migration. It also induced genes regulating inflammatory responses (TGF-β) and gastrointestinal permeability (CTNN-β1). BC 5% enhanced antioxidant activity in the cecum and colon, but no significant impact on anti-myxo antibody production was observed. These results suggest that BC has significant effects on the rabbit gastrointestinal tract's inflammatory and antioxidant response, but further research is required to fully understand its histological and physiological impact.
Collapse
Affiliation(s)
- Federica Riva
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via dell'Università 6, 26900 Lodi, Italy
| | - Susanna Draghi
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via dell'Università 6, 26900 Lodi, Italy
| | - Alessia Inglesi
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via dell'Università 6, 26900 Lodi, Italy
| | - Joel Filipe
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via dell'Università 6, 26900 Lodi, Italy
| | - Paola Cremonesi
- Istituto di Biologia e Biotecnologia Agraria (IBBA), National Research Council (CNR), Via Einstein, 26900 Lodi, Italy
| | - Antonio Lavazza
- Virology Laboratory, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna (IZSLER), Via Bianchi 9, 25124 Brescia, Italy
| | - Patrizia Cavadini
- Virology Laboratory, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna (IZSLER), Via Bianchi 9, 25124 Brescia, Italy
| | - Daniele Vigo
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via dell'Università 6, 26900 Lodi, Italy
| | - Stella Agradi
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via dell'Università 6, 26900 Lodi, Italy
| | - Laura Menchetti
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Circonvallazione 93-95, 62024 Matelica, Italy
| | - Alessia Di Giancamillo
- Department of Biomedical Sciences for Health, University of Milan, Via Mangiagalli 31, 20133 Milan, Italy
| | - Lucia Aidos
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via dell'Università 6, 26900 Lodi, Italy
| | - Silvia Clotilde Modina
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via dell'Università 6, 26900 Lodi, Italy
| | - Nour Elhouda Fehri
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via dell'Università 6, 26900 Lodi, Italy
| | - Grazia Pastorelli
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via dell'Università 6, 26900 Lodi, Italy
| | - Valentina Serra
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via dell'Università 6, 26900 Lodi, Italy
| | - Claudia Maria Balzaretti
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via dell'Università 6, 26900 Lodi, Italy
| | - Marta Castrica
- Dipartimento di Biomedicina Comparata e Alimentazione-BCA, University of Padua, Viale dell'Università 16, 35020 Legnaro, Italy
| | - Marco Severgnini
- Institute of Biomedical Technologies (ITB), National Research Council (CNR), Via Fratelli Cervi 93, 20090 Segrate, Italy
| | - Gabriele Brecchia
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via dell'Università 6, 26900 Lodi, Italy
| | - Giulio Curone
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via dell'Università 6, 26900 Lodi, Italy
| |
Collapse
|
33
|
Jones J, Shi Q, Nath RR, Brito IL. Keystone pathobionts associated with colorectal cancer promote oncogenic reprograming. PLoS One 2024; 19:e0297897. [PMID: 38363784 PMCID: PMC10871517 DOI: 10.1371/journal.pone.0297897] [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: 06/07/2023] [Accepted: 01/12/2024] [Indexed: 02/18/2024] Open
Abstract
Fusobacterium nucleatum (Fn) and enterotoxigenic Bacteroides fragilis (ETBF) are two pathobionts consistently enriched in the gut microbiomes of patients with colorectal cancer (CRC) compared to healthy counterparts and frequently observed for their direct association within tumors. Although several molecular mechanisms have been identified that directly link these organisms to features of CRC in specific cell types, their specific effects on the epithelium and local immune compartment are not well-understood. To fill this gap, we leveraged single-cell RNA sequencing (scRNA-seq) on wildtype mice and mouse model of CRC. We find that Fn and ETBF exacerbate cancer-like transcriptional phenotypes in transit-amplifying and mature enterocytes in a mouse model of CRC. We also observed increased T cells in the pathobiont-exposed mice, but these pathobiont-specific differences observed in wildtype mice were abrogated in the mouse model of CRC. Although there are similarities in the responses provoked by each organism, we find pathobiont-specific effects in Myc-signaling and fatty acid metabolism. These findings support a role for Fn and ETBF in potentiating tumorigenesis via the induction of a cancer stem cell-like transit-amplifying and enterocyte population and the disruption of CTL cytotoxic function.
Collapse
Affiliation(s)
- Josh Jones
- Meinig School for Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
| | - Qiaojuan Shi
- Meinig School for Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
| | - Rahul R. Nath
- Meinig School for Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
| | - Ilana L. Brito
- Meinig School for Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
| |
Collapse
|
34
|
Kim G, Chen Z, Li J, Luo J, Castro-Martinez F, Wisniewski J, Cui K, Wang Y, Sun J, Ren X, Crawford SE, Becerra SP, Zhu J, Liu T, Wang S, Zhao K, Wu C. Gut-liver axis calibrates intestinal stem cell fitness. Cell 2024; 187:914-930.e20. [PMID: 38280375 PMCID: PMC10923069 DOI: 10.1016/j.cell.2024.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 10/25/2023] [Accepted: 01/02/2024] [Indexed: 01/29/2024]
Abstract
The gut and liver are recognized to mutually communicate through the biliary tract, portal vein, and systemic circulation. However, it remains unclear how this gut-liver axis regulates intestinal physiology. Through hepatectomy and transcriptomic and proteomic profiling, we identified pigment epithelium-derived factor (PEDF), a liver-derived soluble Wnt inhibitor, which restrains intestinal stem cell (ISC) hyperproliferation to maintain gut homeostasis by suppressing the Wnt/β-catenin signaling pathway. Furthermore, we found that microbial danger signals resulting from intestinal inflammation can be sensed by the liver, leading to the repression of PEDF production through peroxisome proliferator-activated receptor-α (PPARα). This repression liberates ISC proliferation to accelerate tissue repair in the gut. Additionally, treating mice with fenofibrate, a clinical PPARα agonist used for hypolipidemia, enhances colitis susceptibility due to PEDF activity. Therefore, we have identified a distinct role for PEDF in calibrating ISC expansion for intestinal homeostasis through reciprocal interactions between the gut and liver.
Collapse
Affiliation(s)
- Girak Kim
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zuojia Chen
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jian Li
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jialie Luo
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Felipe Castro-Martinez
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jan Wisniewski
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kairong Cui
- Laboratory of Epigenome Biology, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yan Wang
- Mass Spectrometry Facility, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jialei Sun
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiaobai Ren
- Department of Ophthalmology, Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA 94304, USA
| | - Susan E Crawford
- Department of Surgery, North Shore University Research Institute, University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
| | - S Patricia Becerra
- Section of Protein Structure and Function, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jimin Zhu
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Taotao Liu
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Sui Wang
- Department of Ophthalmology, Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA 94304, USA
| | - Keji Zhao
- Laboratory of Epigenome Biology, Systems Biology Center, National Heart, Lung, and Blood 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.
| |
Collapse
|
35
|
Abud HE, Amarasinghe SL, Micati D, Jardé T. Stromal Niche Signals That Orchestrate Intestinal Regeneration. Cell Mol Gastroenterol Hepatol 2024; 17:679-685. [PMID: 38342301 PMCID: PMC10957453 DOI: 10.1016/j.jcmgh.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/01/2024] [Accepted: 02/01/2024] [Indexed: 02/13/2024]
Abstract
Stromal cell populations have a central role in providing signals that support the maintenance, differentiation, and function of the intestinal epithelium. The behavior and fate of epithelial cells is directed by the spatial organization of stromal cells that either sustain stem and progenitor cell identity or drive differentiation. A combination of single-cell analyses, mouse models, and organoid coculture assays have provided insight into the diversity of signals delivered by stromal cells. Signaling gradients are established and fine-tuned by the expression of signaling agonists and antagonists along the crypt-villus axis. On epithelial injury, there are disruptions to the abundance and organization of stromal populations. There are also distinct changes in the signals originating from these cells that impact remodeling of the epithelium. How these signals coordinate to mediate epithelial repair or sustain tissue injury in inflammatory bowel diseases is beginning to emerge. Understanding of these processes may lead to opportunities to target stromal cell populations as a strategy to modify disease states.
Collapse
Affiliation(s)
- Helen E Abud
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.
| | - Shanika L Amarasinghe
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Diana Micati
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Thierry Jardé
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia; Cancer Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
36
|
Gudi R, Johnson BM, Gaudreau MC, Sun W, Ball L, Vasu C. Intestinal permeability and inflammatory features of juvenile age correlate with the eventual systemic autoimmunity in lupus-prone female SWR × NZB F1 (SNF1) mice. Immunology 2024; 171:235-249. [PMID: 37947218 PMCID: PMC10842200 DOI: 10.1111/imm.13713] [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: 04/24/2023] [Accepted: 10/29/2023] [Indexed: 11/12/2023] Open
Abstract
The incidence of systemic lupus erythematosus (SLE) is about nine times higher in women than in men, and the underlying mechanisms that contribute to this gender bias are not fully understood. Previously, using lupus-prone (SWR × NZB)F1 (SNF1) mice, we have shown that the intestinal immune system could play a role in the initiation and progression of disease in SLE, and depletion of gut microbiota produces more pronounced disease protection in females than in males. Here, we show that the gut permeability features of lupus-prone female SNF1 mice at juvenile ages directly correlate with the expression levels of pro-inflammatory factors, faecal IgA abundance and nAg reactivity and the eventual systemic autoantibody levels and proteinuria onset. Furthermore, we observed that the disease protection achieved in female SNF1 mice upon depletion of gut microbiota correlates with the diminished gut inflammatory protein levels, intestinal permeability and circulating microbial DNA levels. However, faecal microbiota transplant from juvenile male and females did not result in modulation of gut inflammatory features or permeability. Overall, these observations suggest that the early onset of intestinal inflammation, systemic autoantibody production and clinical stage disease in lupus-prone females is linked to higher gut permeability in them starting at as early as juvenile age. While the higher gut permeability in juvenile lupus-prone females is dependent on the presence of gut microbes, it appears to be independent of the composition of gut microbiota.
Collapse
Affiliation(s)
- Radhika Gudi
- Department of Microbiology and Immunology, College of Medicine, Medical University of South Carolina, Charleston, SC-29425
| | - Benjamin M. Johnson
- Department of Microbiology and Immunology, College of Medicine, Medical University of South Carolina, Charleston, SC-29425
| | - Marie-Claude Gaudreau
- Department of Microbiology and Immunology, College of Medicine, Medical University of South Carolina, Charleston, SC-29425
| | - Wei Sun
- Department of Microbiology and Immunology, College of Medicine, Medical University of South Carolina, Charleston, SC-29425
| | - Lauren Ball
- Department of Pharmacology, College of Medicine, Medical University of South Carolina, Charleston, SC-29425
| | - Chenthamarakshan Vasu
- Department of Microbiology and Immunology, College of Medicine, Medical University of South Carolina, Charleston, SC-29425
| |
Collapse
|
37
|
Janubová M, Žitňanová I. The effects of vitamin D on different types of cells. Steroids 2024; 202:109350. [PMID: 38096964 DOI: 10.1016/j.steroids.2023.109350] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 11/24/2023] [Accepted: 12/07/2023] [Indexed: 12/25/2023]
Abstract
Vitamin D is neccessary for regulation of calcium and phosphorus metabolism in bones, affects imunity, the cardiovascular system, muscles, skin, epithelium, extracellular matrix, the central nervous system, and plays arole in prevention of aging-associated diseases. Vitamin D receptor is expressed in almost all types of cells and its activation leads to modulation of different signaling pathways. In this review, we have analysed the current knowledge of 1,25-dihydroxyvitamin D3 or 25-hydroxyvitamin D3 effects on metabolism of cells important for the function of the cardiovascular system (endothelial cells, vascular smooth muscle cells, cardiac cells and pericytes), tissue healing (fibroblasts), epithelium (various types of epithelial cells) and the central nervous system (neurons, astrocytes and microglia). The goal of this review was to compare the effects of vitamin D on the above mentioned cells in in vitro conditions and to summarize what is known in this field of research.
Collapse
Affiliation(s)
- Mária Janubová
- Institute of Medical Chemistry, Biochemistry and Clinical Biochemistry, Comenius University, 813 72 Bratislava, Slovakia.
| | - Ingrid Žitňanová
- Institute of Medical Chemistry, Biochemistry and Clinical Biochemistry, Comenius University, 813 72 Bratislava, Slovakia
| |
Collapse
|
38
|
Yang L, Ruan Z, Lin X, Wang H, Xin Y, Tang H, Hu Z, Zhou Y, Wu Y, Wang J, Qin D, Lu G, Loomes KM, Chan WY, Liu X. NAD + dependent UPR mt activation underlies intestinal aging caused by mitochondrial DNA mutations. Nat Commun 2024; 15:546. [PMID: 38228611 PMCID: PMC10791663 DOI: 10.1038/s41467-024-44808-z] [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: 12/20/2022] [Accepted: 01/03/2024] [Indexed: 01/18/2024] Open
Abstract
Aging in mammals is accompanied by an imbalance of intestinal homeostasis and accumulation of mitochondrial DNA (mtDNA) mutations. However, little is known about how accumulated mtDNA mutations modulate intestinal homeostasis. We observe the accumulation of mtDNA mutations in the small intestine of aged male mice, suggesting an association with physiological intestinal aging. Using polymerase gamma (POLG) mutator mice and wild-type mice, we generate male mice with progressive mtDNA mutation burdens. Investigation utilizing organoid technology and in vivo intestinal stem cell labeling reveals decreased colony formation efficiency of intestinal crypts and LGR5-expressing intestinal stem cells in response to a threshold mtDNA mutation burden. Mechanistically, increased mtDNA mutation burden exacerbates the aging phenotype of the small intestine through ATF5 dependent mitochondrial unfolded protein response (UPRmt) activation. This aging phenotype is reversed by supplementation with the NAD+ precursor, NMN. Thus, we uncover a NAD+ dependent UPRmt triggered by mtDNA mutations that regulates the intestinal aging.
Collapse
Affiliation(s)
- Liang Yang
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zifeng Ruan
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaobing Lin
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Hao Wang
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yanmin Xin
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Haite Tang
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zhijuan Hu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yunhao Zhou
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yi Wu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Junwei Wang
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Dajiang Qin
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Gang Lu
- CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, CUHK-Jinan University Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kerry M Loomes
- School of Biological Sciences and Institute for Innovation in Biotechnology, University of Auckland, Auckland, 1010, New Zealand
| | - Wai-Yee Chan
- CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, CUHK-Jinan University Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xingguo Liu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China.
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China.
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.
| |
Collapse
|
39
|
Kulkarni PP, Ekhlak M, Dash D. Non-canonical non-genomic morphogen signaling in anucleate platelets: a critical determinant of prothrombotic function in circulation. Cell Commun Signal 2024; 22:13. [PMID: 38172855 PMCID: PMC10763172 DOI: 10.1186/s12964-023-01448-y] [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: 11/08/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024] Open
Abstract
Circulating platelets derived from bone marrow megakaryocytes play a central role in thrombosis and hemostasis. Despite being anucleate, platelets express several proteins known to have nuclear niche. These include transcription factors and steroid receptors whose non-genomic functions are being elucidated in platelets. Quite remarkably, components of some of the best-studied morphogen pathways, namely Notch, Sonic Hedgehog (Shh), and Wnt have also been described in recent years in platelets, which regulate platelet function in the context of thrombosis as well as influence their survival. Shh and Notch pathways in stimulated platelets establish feed-forward loops of autocrine/juxtacrine/paracrine non-canonical signaling that helps perpetuate thrombosis. On the other hand, non-canonical Wnt signaling is part of a negative feedback loop for restricting platelet activation and possibly limiting thrombus growth. The present review will provide an overview of these signaling pathways in general. We will then briefly discuss the non-genomic roles of transcription factors and steroid receptors in platelet activation. This will be followed by an elaborate description of morphogen signaling in platelets with a focus on their bearing on platelet activation leading to hemostasis and thrombosis as well as their potential for therapeutic targeting in thrombotic disorders.
Collapse
Affiliation(s)
- Paresh P Kulkarni
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| | - Mohammad Ekhlak
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Debabrata Dash
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| |
Collapse
|
40
|
Peng Z, Bao L, Shi B, Shi YB. Protein arginine methyltransferase 1 is required for the maintenance of adult small intestinal and colonic epithelial cell homeostasis. Int J Biol Sci 2024; 20:554-568. [PMID: 38169732 PMCID: PMC10758107 DOI: 10.7150/ijbs.89958] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 11/25/2023] [Indexed: 01/05/2024] Open
Abstract
The vertebrate adult intestinal epithelium has a high self-renewal rate driven by intestinal stem cells (ISCs) in the crypts, which play central roles in maintaining intestinal integrity and homeostasis. However, the underlying mechanisms remain elusive. Here we showed that protein arginine methyltransferase 1 (PRMT1), a major arginine methyltransferase that can also function as a transcription co-activator, was highly expressed in the proliferating cells of adult mouse intestinal crypts. Intestinal epithelium-specific knockout of PRMT1, which ablates PRMT1 gene starting during embryogenesis, caused distinct, region-specific effects on small intestine and colon: increasing and decreasing the goblet cell number in the small intestinal and colonic crypts, respectively, leading to elongation of the crypts in small intestine but not colon, while increasing crypt cell proliferation in both regions. We further generated a tamoxifen-inducible intestinal epithelium-specific PRMT1 knockout mouse model and found that tamoxifen-induced knockout of PRMT1 in the adult mice resulted in the same region-specific intestinal phenotypes. Thus, our studies have for the first time revealed that the epigenetic enzyme PRMT1 has distinct, region-specific roles in the maintenance of intestinal epithelial architecture and homeostasis, although PRMT1 may influence intestinal development.
Collapse
Affiliation(s)
- Zhaoyi Peng
- Department of Endocrinology, The First Affiliated Hospital of Xi'an JiaoTong University, No. 277, West Yanta Road, Xi'an, Shaanxi 710061, P.R. China
- Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, MD, USA
| | - Lingyu Bao
- Department of Endocrinology, The First Affiliated Hospital of Xi'an JiaoTong University, No. 277, West Yanta Road, Xi'an, Shaanxi 710061, P.R. China
- Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, MD, USA
| | - Bingyin Shi
- Department of Endocrinology, The First Affiliated Hospital of Xi'an JiaoTong University, No. 277, West Yanta 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
|
41
|
Wang J, Ma Y, Li T, Li J, Yang X, Hua G, Cai G, Zhang H, Liu Z, Wu K, Deng X. MiR-199a-3p Regulates the PTPRF/β-Catenin Axis in Hair Follicle Development: Insights into the Pathogenic Mechanism of Alopecia Areata. Int J Mol Sci 2023; 24:17632. [PMID: 38139460 PMCID: PMC10743674 DOI: 10.3390/ijms242417632] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/10/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Alopecia areata is an autoimmune disease characterized by the immune system attacking self hair follicles, mainly in the scalp. There is no complete cure, and the pathogenesis is still not fully understood. Here, sequencing of skin tissues collected from 1-month-old coarse- and fine-wool lambs identified miR-199a-3p as the only small RNA significantly overexpressed in the fine-wool group, suggesting a role in hair follicle development. MiR-199a-3p expression was concentrated in the dermal papillae cells of sheep hair follicles, along with enhanced β-catenin expression and the inhibition of PTPRF protein expression. We also successfully constructed a mouse model of alopecia areata by intracutaneous injection with an miR-199a-3p antagomir. Injection of the miR-199a-3p agomir resulted in hair growth and earlier anagen entry. Conversely, local injection with the miR-199a-3p antagomir resulted in suppressed hair growth at the injection site, upregulation of immune system-related genes, and downregulation of hair follicle development-related genes. In vivo and in vitro analyses demonstrated that miR-199a-3p regulates hair follicle development through the PTPRF/β-catenin axis. In conclusion, a mouse model of alopecia areata was successfully established by downregulation of a small RNA, suggesting the potential value of miR-199a-3p in the study of alopecia diseases. The regulatory role of miR-199a-3p in the PTPRF/β-catenin axis was confirmed, further demonstrating the link between alopecia areata and the Wnt-signaling pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Xuemei Deng
- Beijing Key Laboratory for Animal Genetic Improvement & State Key Laboratory of Animal Biotech Breeding & Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, China Agricultural University, Beijing 100193, China; (J.W.); (Y.M.); (T.L.); (J.L.); (X.Y.); (G.H.); (G.C.); (H.Z.); (Z.L.); (K.W.)
| |
Collapse
|
42
|
Gall L, Duckworth C, Jardi F, Lammens L, Parker A, Bianco A, Kimko H, Pritchard DM, Pin C. Homeostasis, injury, and recovery dynamics at multiple scales in a self-organizing mouse intestinal crypt. eLife 2023; 12:e85478. [PMID: 38063302 PMCID: PMC10789491 DOI: 10.7554/elife.85478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 12/07/2023] [Indexed: 01/16/2024] Open
Abstract
The maintenance of the functional integrity of the intestinal epithelium requires a tight coordination between cell production, migration, and shedding along the crypt-villus axis. Dysregulation of these processes may result in loss of the intestinal barrier and disease. With the aim of generating a more complete and integrated understanding of how the epithelium maintains homeostasis and recovers after injury, we have built a multi-scale agent-based model (ABM) of the mouse intestinal epithelium. We demonstrate that stable, self-organizing behaviour in the crypt emerges from the dynamic interaction of multiple signalling pathways, such as Wnt, Notch, BMP, ZNRF3/RNF43, and YAP-Hippo pathways, which regulate proliferation and differentiation, respond to environmental mechanical cues, form feedback mechanisms, and modulate the dynamics of the cell cycle protein network. The model recapitulates the crypt phenotype reported after persistent stem cell ablation and after the inhibition of the CDK1 cycle protein. Moreover, we simulated 5-fluorouracil (5-FU)-induced toxicity at multiple scales starting from DNA and RNA damage, which disrupts the cell cycle, cell signalling, proliferation, differentiation, and migration and leads to loss of barrier integrity. During recovery, our in silico crypt regenerates its structure in a self-organizing, dynamic fashion driven by dedifferentiation and enhanced by negative feedback loops. Thus, the model enables the simulation of xenobiotic-, in particular chemotherapy-, induced mechanisms of intestinal toxicity and epithelial recovery. Overall, we present a systems model able to simulate the disruption of molecular events and its impact across multiple levels of epithelial organization and demonstrate its application to epithelial research and drug development.
Collapse
Affiliation(s)
- Louis Gall
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZenecaCambridgeUnited Kingdom
| | - Carrie Duckworth
- Institute of Systems, Molecular and Integrative Biology, University of LiverpoolLiverpoolUnited Kingdom
| | - Ferran Jardi
- Preclinical Sciences and Translational Safety, JanssenBeerseBelgium
| | - Lieve Lammens
- Preclinical Sciences and Translational Safety, JanssenBeerseBelgium
| | - Aimee Parker
- Gut Microbes and Health Programme, Quadram InstituteNorwichUnited Kingdom
| | - Ambra Bianco
- Clinical Pharmacology and Safety Sciences, AstraZenecaCambridgeUnited Kingdom
| | - Holly Kimko
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZenecaCambridgeUnited Kingdom
| | - David Mark Pritchard
- Institute of Systems, Molecular and Integrative Biology, University of LiverpoolLiverpoolUnited Kingdom
| | - Carmen Pin
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZenecaCambridgeUnited Kingdom
| |
Collapse
|
43
|
Jones LO, Willms RJ, Xu X, Graham RDV, Eklund M, Shin M, Foley E. Single-cell resolution of the adult zebrafish intestine under conventional conditions and in response to an acute Vibrio cholerae infection. Cell Rep 2023; 42:113407. [PMID: 37948182 DOI: 10.1016/j.celrep.2023.113407] [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: 04/18/2023] [Revised: 10/08/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023] Open
Abstract
Vibrio cholerae is an aquatic bacterium that causes severe and potentially deadly diarrheal disease. Despite the impact on global health, our understanding of host mucosal responses to Vibrio remains limited, highlighting a knowledge gap critical for the development of effective prevention and treatment strategies. Using a natural infection model, we combine physiological and single-cell transcriptomic studies to characterize conventionally reared adult zebrafish guts and guts challenged with Vibrio. We demonstrate that Vibrio causes a mild mucosal immune response characterized by T cell activation and enhanced antigen capture; Vibrio suppresses host interferon signaling; and ectopic activation of interferon alters the course of infection. We show that the adult zebrafish gut shares similarities with mammalian counterparts, including the presence of Best4+ cells, tuft cells, and a population of basal cycling cells. These findings provide important insights into host-pathogen interactions and emphasize the utility of zebrafish as a natural model of Vibrio infection.
Collapse
Affiliation(s)
- Lena O Jones
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Reegan J Willms
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Xinyue Xu
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Ralph Derrick V Graham
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Mckenna Eklund
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Minjeong Shin
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Edan Foley
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada; Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
44
|
Tang C, Ke M, Yu X, Sun S, Luo X, Liu X, Zhou Y, Wang Z, Cui X, Gu C, Yang Y. GART Functions as a Novel Methyltransferase in the RUVBL1/β-Catenin Signaling Pathway to Promote Tumor Stemness in Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301264. [PMID: 37439412 PMCID: PMC10477903 DOI: 10.1002/advs.202301264] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/30/2023] [Indexed: 07/14/2023]
Abstract
Tumor stemness is associated with the recurrence and incurability of colorectal cancer (CRC), which lacks effective therapeutic targets and drugs. Glycinamide ribonucleotide transformylase (GART) fulfills an important role in numerous types of malignancies. The present study aims to identify the underlying mechanism through which GART may promote CRC stemness, as to developing novel therapeutic methods. An elevated level of GART is associated with poor outcomes in CRC patients and promotes the proliferation and migration of CRC cells. CD133+ cells with increased GART expression possess higher tumorigenic and proliferative capabilities both in vitro and in vivo. GART is identified to have a novel methyltransferase function, whose enzymatic activity center is located at the E948 site. GART also enhances the stability of RuvB-like AAA ATPase 1 (RUVBL1) through methylating its K7 site, which consequently aberrantly activates the Wnt/β-catenin signaling pathway to induce tumor stemness. Pemetrexed (PEM), a compound targeting GART, combined with other chemotherapy drugs greatly suppresses tumor growth both in a PDX model and in CRC patients. The present study demonstrates a novel methyltransferase function of GART and the role of the GART/RUVBL1/β-catenin signaling axis in promoting CRC stemness. PEM may be a promising therapeutic agent for the treatment of CRC.
Collapse
Affiliation(s)
- Chao Tang
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western MedicineNanjing University of Chinese MedicineNanjing210008China
- School of Medicine & Holistic Integrative MedicineNanjing University of Chinese MedicineNanjing210046China
| | - Mengying Ke
- School of Medicine & Holistic Integrative MedicineNanjing University of Chinese MedicineNanjing210046China
| | - Xichao Yu
- School of Medicine & Holistic Integrative MedicineNanjing University of Chinese MedicineNanjing210046China
| | - Shanliang Sun
- School of PharmacyNanjing University of Chinese MedicineNanjing210046China
| | - Xian Luo
- School of Medicine & Holistic Integrative MedicineNanjing University of Chinese MedicineNanjing210046China
| | - Xin Liu
- School of Medicine & Holistic Integrative MedicineNanjing University of Chinese MedicineNanjing210046China
| | - Yanyan Zhou
- School of Medicine & Holistic Integrative MedicineNanjing University of Chinese MedicineNanjing210046China
| | - Ze Wang
- School of Medicine & Holistic Integrative MedicineNanjing University of Chinese MedicineNanjing210046China
| | - Xing Cui
- Department of Hematology and OncologyThe Second Affiliated Hospital of Shandong University of Traditional Chinese MedicineJinan250001China
| | - Chunyan Gu
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western MedicineNanjing University of Chinese MedicineNanjing210008China
- School of Medicine & Holistic Integrative MedicineNanjing University of Chinese MedicineNanjing210046China
| | - Ye Yang
- School of Medicine & Holistic Integrative MedicineNanjing University of Chinese MedicineNanjing210046China
| |
Collapse
|
45
|
Bugacov H, Der B, Kim S, Lindström NO, McMahon AP. Canonical Wnt transcriptional complexes are essential for induction of nephrogenesis but not maintenance or proliferation of nephron progenitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.20.554044. [PMID: 37662369 PMCID: PMC10473675 DOI: 10.1101/2023.08.20.554044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Wnt regulated transcriptional programs are associated with both the maintenance of mammalian nephron progenitor cells (NPC) and their induction, initiating the process of nephrogenesis. How opposing transcriptional roles are regulated remain unclear. Using an in vitro model replicating in vivo events, we examined the requirement for canonical Wnt transcriptional complexes in NPC regulation. In canonical transcription, Lef/Tcf DNA binding proteins associate the transcriptional co-activator β-catenin. Wnt signaling is readily substituted by CHIR99021, a small molecule antagonist of glycogen synthase kinase-3β (GSK3β). GSK3β inhibition blocks Gskβ-dependent turnover of β-catenin, enabling formation of Lef/Tcf/β-catenin transcriptional complexes, and enhancer-mediated transcriptional activation. Removal of β-catenin activity from NPCs under cell expansion conditions (low CHIR) demonstrated a non-transcriptional role for β-catenin in the CHIR-dependent proliferation of NPCs. In contrast, CHIR-mediated induction of nephrogenesis, on switching from low to high CHIR, was dependent on Lef/Tcf and β-catenin transcriptional activity. These studies point to a non-transcriptional mechanism for β-catenin in regulation of NPCs, and potentially other stem progenitor cell types. Further, analysis of the β-catenin-directed transcriptional response provides new insight into induction of nephrogenesis. Summary Statement The study provides a mechanistic understanding of Wnt/ β-catenin activity in self-renewal and differentiation of mammalian nephron progenitors.
Collapse
|
46
|
Zheng X, Betjes MA, Ender P, Goos YJ, Huelsz-Prince G, Clevers H, van Zon JS, Tans SJ. Organoid cell fate dynamics in space and time. SCIENCE ADVANCES 2023; 9:eadd6480. [PMID: 37595032 PMCID: PMC10438469 DOI: 10.1126/sciadv.add6480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 07/20/2023] [Indexed: 08/20/2023]
Abstract
Organoids are a major new tool to study tissue renewal. However, characterizing the underlying differentiation dynamics remains challenging. Here, we developed TypeTracker, which identifies cell fates by AI-enabled cell tracking and propagating end point fates back along the branched lineage trees. Cells that ultimately migrate to the villus commit to their new type early, when still deep inside the crypt, with important consequences: (i) Secretory cells commit before terminal division, with secretory fates emerging symmetrically in sister cells. (ii) Different secretory types descend from distinct stem cell lineages rather than an omnipotent secretory progenitor. (iii) The ratio between secretory and absorptive cells is strongly affected by proliferation after commitment. (iv) Spatial patterning occurs after commitment through type-dependent cell rearrangements. This "commit-then-sort" model contrasts with the conventional conveyor belt picture, where cells differentiate by moving up the crypt-villus axis and hence raises new questions about the underlying commitment and sorting mechanisms.
Collapse
Affiliation(s)
| | | | | | | | | | - Hans Clevers
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, Uppsalalaan 8, Utrecht 3584 CT, Netherlands
| | | | - Sander J Tans
- Bionanoscience Department, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, Netherlands
- AMOLF, Amsterdam, Netherlands.
| |
Collapse
|
47
|
Suzuki T, Kilbey A, Casa-Rodríguez N, Lawlor A, Georgakopoulou A, Hayman H, Yin Swe KL, Nordin A, Cantù C, Vantourout P, Ridgway RA, Byrne RM, Chen L, Verzi MP, Gay DM, Gil Vázquez E, Belnoue-Davis HL, Gilroy K, Køstner AH, Kersten C, Thuwajit C, Andersen DK, Wiesheu R, Jandke A, Blyth K, Roseweir AK, Leedham SJ, Dunne PD, Edwards J, Hayday A, Sansom OJ, Coffelt SB. β-Catenin Drives Butyrophilin-like Molecule Loss and γδ T-cell Exclusion in Colon Cancer. Cancer Immunol Res 2023; 11:1137-1155. [PMID: 37309673 PMCID: PMC10398359 DOI: 10.1158/2326-6066.cir-22-0644] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 02/20/2023] [Accepted: 06/09/2023] [Indexed: 06/14/2023]
Abstract
Intraepithelial lymphocytes (IEL) expressing γδ T-cell receptors (γδTCR) play key roles in elimination of colon cancer. However, the precise mechanisms by which progressing cancer cells evade immunosurveillance by these innate T cells are unknown. Here, we investigated how loss of the Apc tumor suppressor in gut tissue could enable nascent cancer cells to escape immunosurveillance by cytotoxic γδIELs. In contrast with healthy intestinal or colonic tissue, we found that γδIELs were largely absent from the microenvironment of both mouse and human tumors, and that butyrophilin-like (BTNL) molecules, which can critically regulate γδIEL through direct γδTCR interactions, were also downregulated in tumors. We then demonstrated that β-catenin activation through loss of Apc rapidly suppressed expression of the mRNA encoding the HNF4A and HNF4G transcription factors, preventing their binding to promoter regions of Btnl genes. Reexpression of BTNL1 and BTNL6 in cancer cells increased γδIEL survival and activation in coculture assays but failed to augment their cancer-killing ability in vitro or their recruitment to orthotopic tumors. However, inhibition of β-catenin signaling via genetic deletion of Bcl9/Bcl9L in either Apc-deficient or mutant β-catenin mouse models restored Hnf4a, Hnf4g, and Btnl gene expression and γδ T-cell infiltration into tumors. These observations highlight an immune-evasion mechanism specific to WNT-driven colon cancer cells that disrupts γδIEL immunosurveillance and furthers cancer progression.
Collapse
Affiliation(s)
- Toshiyasu Suzuki
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Anna Kilbey
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Nuria Casa-Rodríguez
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Amy Lawlor
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Anastasia Georgakopoulou
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Hannah Hayman
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Kyi Lai Yin Swe
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Anna Nordin
- Wallenberg Centre for Molecular Medicine, Linköping University, Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Claudio Cantù
- Wallenberg Centre for Molecular Medicine, Linköping University, Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Pierre Vantourout
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
- The Francis Crick Institute, London, United Kingdom
| | | | - Ryan M. Byrne
- School of Medicine, Dentistry and Biomedical Sciences, Queen's University, Belfast, United Kingdom
| | - Lei Chen
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, New Jersey
| | - Michael P. Verzi
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, New Jersey
| | - David M. Gay
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Ester Gil Vázquez
- Nuffield Department of Medicine, Oxford University, Oxford, United Kingdom
| | | | - Kathryn Gilroy
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | | | - Christian Kersten
- Department of Research, Southern Hospital Trust, Kristiansand, Norway
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway
| | - Chanitra Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Nakhon Pathom, Thailand
| | | | - Robert Wiesheu
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Anett Jandke
- The Francis Crick Institute, London, United Kingdom
| | - Karen Blyth
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Antonia K. Roseweir
- School of Medicine, Dentistry & Nursing, University of Glasgow, Glasgow, United Kingdom
| | - Simon J. Leedham
- Nuffield Department of Medicine, Oxford University, Oxford, United Kingdom
| | - Philip D. Dunne
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
- School of Medicine, Dentistry and Biomedical Sciences, Queen's University, Belfast, United Kingdom
| | - Joanne Edwards
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Adrian Hayday
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
- The Francis Crick Institute, London, United Kingdom
| | - Owen J. Sansom
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Seth B. Coffelt
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
48
|
Song H, Sontz RA, Vance MJ, Morris JM, Sheriff S, Zhu S, Duan S, Zeng J, Koeppe E, Pandey R, Thorne CA, Stoffel EM, Merchant JL. High-fat diet plus HNF1A variant promotes polyps by activating β-catenin in early-onset colorectal cancer. JCI Insight 2023; 8:e167163. [PMID: 37219942 PMCID: PMC10371337 DOI: 10.1172/jci.insight.167163] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 05/19/2023] [Indexed: 05/24/2023] Open
Abstract
The incidence of early-onset colorectal cancer (EO-CRC) is rising and is poorly understood. Lifestyle factors and altered genetic background possibly contribute. Here, we performed targeted exon sequencing of archived leukocyte DNA from 158 EO-CRC participants, which identified a missense mutation at p.A98V within the proximal DNA binding domain of Hepatic Nuclear Factor 1 α (HNF1AA98V, rs1800574). The HNF1AA98V exhibited reduced DNA binding. To test function, the HNF1A variant was introduced into the mouse genome by CRISPR/Cas9, and the mice were placed on either a high-fat diet (HFD) or high-sugar diet (HSD). Only 1% of the HNF1A mutant mice developed polyps on normal chow; however, 19% and 3% developed polyps on the HFD and HSD, respectively. RNA-Seq revealed an increase in metabolic, immune, lipid biogenesis genes, and Wnt/β-catenin signaling components in the HNF1A mutant relative to the WT mice. Mouse polyps and colon cancers from participants carrying the HNF1AA98V variant exhibited reduced CDX2 and elevated β-catenin proteins. We further demonstrated decreased occupancy of HNF1AA98V at the Cdx2 locus and reduced Cdx2 promoter activity compared with WT HNF1A. Collectively, our study shows that the HNF1AA98V variant plus a HFD promotes the formation of colonic polyps by activating β-catenin via decreasing Cdx2 expression.
Collapse
Affiliation(s)
- Heyu Song
- Department of Medicine, Division of Gastroenterology and Hepatology, Arizona Comprehensive Cancer Center, and
| | - Ricky A. Sontz
- Department of Medicine, Division of Gastroenterology and Hepatology, Arizona Comprehensive Cancer Center, and
| | - Matthew J. Vance
- Department of Medicine, Division of Gastroenterology and Hepatology, Arizona Comprehensive Cancer Center, and
| | - Julia M. Morris
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Sulaiman Sheriff
- Department of Medicine, Division of Gastroenterology and Hepatology, Arizona Comprehensive Cancer Center, and
| | - Songli Zhu
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Suzann Duan
- Department of Medicine, Division of Gastroenterology and Hepatology, Arizona Comprehensive Cancer Center, and
| | - Jiping Zeng
- Department of Urology, University of Arizona College of Medicine, Tucson, Arizona, USA
| | | | - Ritu Pandey
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Curtis A. Thorne
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Elena M. Stoffel
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Juanita L. Merchant
- Department of Medicine, Division of Gastroenterology and Hepatology, Arizona Comprehensive Cancer Center, and
| |
Collapse
|
49
|
Paul EN, Carpenter TJ, Fitch S, Sheridan R, Lau KH, Arora R, Teixeira JM. Cysteine-rich intestinal protein 1 is a novel surface marker for human myometrial stem/progenitor cells. Commun Biol 2023; 6:686. [PMID: 37400623 DOI: 10.1038/s42003-023-05061-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/21/2023] [Indexed: 07/05/2023] Open
Abstract
Myometrial stem/progenitor cells (MyoSPCs) have been proposed as the cells of origin for uterine fibroids, but the identity of the MyoSPC has not been well established. We previously identified SUSD2 as a possible MyoSPC marker, but the relatively poor enrichment in stem cell characteristics of SUSD2+ over SUSD2- cells compelled us to find better markers. We combined bulk RNA-seq of SUSD2+/- cells with single cell RNA-seq to identify markers for MyoSPCs. We observed seven distinct cell clusters within the myometrium, with the vascular myocyte cluster most highly enriched for MyoSPC characteristics and markers. CRIP1 expression was found highly upregulated by both techniques and was used as a marker to sort CRIP1+/PECAM1- cells that were both enriched for colony forming potential and able to differentiate into mesenchymal lineages, suggesting that CRIP1+/PECAM1- cells could be used to better study the etiology of uterine fibroids.
Collapse
Affiliation(s)
- Emmanuel N Paul
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503, USA
| | - Tyler J Carpenter
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503, USA
| | - Sarah Fitch
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503, USA
- Institute for Quantitative Health Science and Engineering, East Lansing, MI, 48824, USA
| | - Rachael Sheridan
- Flow Cytometry Core, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Kin H Lau
- Bioinformatics and Biostatistics Core, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Ripla Arora
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503, USA
- Institute for Quantitative Health Science and Engineering, East Lansing, MI, 48824, USA
| | - Jose M Teixeira
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503, USA.
| |
Collapse
|
50
|
Chittimalli K, Jahan J, Sakamuri A, McAdams ZL, Ericsson AC, Jarajapu YP. Restoration of the gut barrier integrity and restructuring of the gut microbiome in aging by angiotensin-(1-7). Clin Sci (Lond) 2023; 137:913-930. [PMID: 37254732 PMCID: PMC10881191 DOI: 10.1042/cs20220904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 05/16/2023] [Accepted: 05/30/2023] [Indexed: 06/01/2023]
Abstract
Compromised barrier function of colon epithelium with aging is largely due to gut microbial dysbiosis. Recent studies implicate an important role for angiotensin converting enzymes, ACE and ACE2, angiotensins, and the receptors, AT1 receptor (AT1R) and Mas receptor (MasR), in the regulation of colon functions. The present study tested the hypothesis that leaky gut in aging is associated with an imbalance in ACE2/ACE and that the treatment with angiotenisn-(1-7) (Ang-(1-7)) will restore gut barrier integrity and microbiome. Studies were carried out in Young (3-4 months) and old (20-24 months) male mice. Ang-(1-7) was administered by using osmotic pumps. Outcome measures included expressions of ACE, ACE2, AT1R, and MasR, intestinal permeability by using FITC-dextran, and immunohistochemistry of claudin 1 and occludin, and intestinal stem cells (ISCs). ACE2 protein and activity were decreased in Old group while that of ACE were unchanged. Increased intestinal permeability and plasma levels of zonulin-1 in the Old group were normalized by Ang-(1-7). Epithelial disintegrity, reduced number of goblet cells and ISCs in the old group were restored by Ang-(1-7). Expression of claudin 1 and occludin in the aging colon was increased by Ang-(1-7). Infiltration of CD11b+ or F4/80+ inflammatory cells in the old colons were decreased by Ang-(1-7). Gut microbial dysbiosis in aging was evident by decreased richness and altered beta diversity that were reversed by Ang-(1-7) with increased abundance of Lactobacillus or Lachnospiraceae. The present study shows that Ang-(1-7) restores gut barrier integrity and reduces inflammation in the aging colon by restoring the layer of ISCs and by restructuring the gut microbiome.
Collapse
Affiliation(s)
- Kishore Chittimalli
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND, U.S.A
| | - Jesmin Jahan
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND, U.S.A
| | - Anil Sakamuri
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND, U.S.A
| | - Zachary L. McAdams
- Missouri Metagenomics Center, Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, U.S.A
| | - Aaron C. Ericsson
- Missouri Metagenomics Center, Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, U.S.A
| | - Yagna P.R. Jarajapu
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND, U.S.A
| |
Collapse
|