1
|
Neurath MF, Artis D, Becker C. The intestinal barrier: a pivotal role in health, inflammation, and cancer. Lancet Gastroenterol Hepatol 2025; 10:573-592. [PMID: 40086468 DOI: 10.1016/s2468-1253(24)00390-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/08/2024] [Accepted: 11/15/2024] [Indexed: 03/16/2025]
Abstract
The intestinal barrier serves as a boundary between the mucosal immune system in the lamina propria and the external environment of the intestinal lumen, which contains a diverse array of microorganisms and ingested environmental factors, including pathogens, food antigens, toxins, and other foreign substances. This barrier has a central role in regulating the controlled interaction between luminal factors and the intestinal immune system. Disruptions of intestinal epithelial cells, which serve as a physical barrier, or the antimicrobial peptides and mucins they produce, which act as a chemical barrier, can lead to a leaky gut. In this state, the intestinal wall is unable to efficiently separate the intestinal flora and luminal contents from the intestinal immune system. The subsequent activation of the immune system has an important role in the pathogenesis of inflammatory bowel disease, as well as in metabolic dysfunction-associated steatohepatitis, primary sclerosing cholangitis, and colorectal cancer. Dysregulated intestinal barrier integrity has also been described in patients with chronic inflammatory diseases outside the gastrointestinal tract, including rheumatoid arthritis and neurodegenerative disorders. Mechanistic studies of barrier dysfunction have revealed that the subsequent local activation and systemic circulation of activated immune cells and the cytokines they secrete, as well as extracellular vesicles, promote proinflammatory processes within and outside the gastrointestinal tract. In this Review, we summarise these findings and highlight several new therapeutic concepts currently being developed that attempt to control inflammatory processes via direct or indirect modulation of intestinal barrier function.
Collapse
Affiliation(s)
- Markus F Neurath
- Medical Clinic 1, Department of Gastroenterology, Ludwig Demling Endoscopy Center of Excellence, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.
| | - David Artis
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA; Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY, USA; Joan and Sanford I Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA; Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA; Allen Discovery Center for Neuroimmune Interactions, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Christoph Becker
- Medical Clinic 1, Department of Gastroenterology, Ludwig Demling Endoscopy Center of Excellence, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
2
|
Wu F, Liu Y, Zhang M, Yuan X, Jin Y, Li Y, Wang R, Hao Y, Fang B. Effects of 1,3-Dioleoyl-2-palmitoylglycerol on Intestine Structural and Functional Development in Early Life. Mol Nutr Food Res 2025; 69:e70051. [PMID: 40129020 DOI: 10.1002/mnfr.70051] [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: 11/06/2024] [Revised: 03/03/2025] [Accepted: 03/12/2025] [Indexed: 03/26/2025]
Abstract
1,3-Dioleoyl-2-palmitoyl-glycerol (OPO) is a specific triglyceride in human breast milk, and it has been added to infant formula to mimic human breast milk fat. Existing studies only focused on its effects on fatty acid and calcium absorption, as well as the intestinal microbial composition; however, effects of OPO on the early-life development of intestine were still unclear. Our study explored the effects of OPO on intestinal epithelial structure and barrier construction in neonatal mice and the involvement of intestinal microorganisms. OPO supplementation significantly increased the number of intestinal stem cells, which in turn promoted villus and crypt, and promoted goblet cell and Paneth cell differentiation. OPO also promotes epithelial barrier integrity by increasing the expression of mucin 2, lysozyme 1, and tight junction proteins. Furthermore, the benefits of OPO were associated with the higher abundance of beneficial bacteria (unclassified_f_Muribaculaceae, Akkermansia, Bifidobacterium, and Blautia) and elevated butyrate levels. This study demonstrates the efficacy of OPO on intestinal health in neonatal mice beyond defecation, expands the understanding of the biological functions of OPO, and expands its application in intestinal health products targeting special populations, such as the elderly or individuals with intestinal fragility or injury.
Collapse
Affiliation(s)
- Fang Wu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yaqiong Liu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Ming Zhang
- School of Food Science and Chemical Engineering, Beijing Technology and Business University, Beijing, China
| | - Xinlei Yuan
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
- College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Yutong Jin
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yixuan Li
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Ran Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yanling Hao
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Bing Fang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| |
Collapse
|
3
|
Bertulfo K, Perez-Duran P, Miller H, Ma C, Ambesi-Impiombato A, Samon J, Mackey A, Lin WHW, Ferrando AA, Palomero T. Therapeutic targeting of the NOTCH1 and neddylation pathways in T cell acute lymphoblastic leukemia. Proc Natl Acad Sci U S A 2025; 122:e2426742122. [PMID: 40163723 PMCID: PMC12002235 DOI: 10.1073/pnas.2426742122] [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: 12/30/2024] [Accepted: 01/30/2025] [Indexed: 04/02/2025] Open
Abstract
Gamma Secretase Inhibitors (GSIs) effectively block oncogenic Notch homolog-1 (NOTCH1), a characteristic feature of T cell acute lymphoblastic leukemias (T-ALL). However, their clinical application has been stalled by the induction of severe gastrointestinal toxicity resulting from the inhibition of NOTCH signaling in the gut, which translates into increased goblet cell differentiation. Genome-wide CRISPR loss-of-function screen in the colon cancer cell line LS174T identified the neddylation pathway as a main regulator of goblet cell differentiation upon NOTCH1 inhibition. Consistently, pharmacologic inhibition of the neddylation pathway with the small molecule inhibitor MLN4924, rescued GSI-induced differentiation in LS174T cells. Mechanistically, neddylation inhibition by MLN4924 increases the protein stability of Hairy and enhancer of split-1, a direct NOTCH1 transcriptional target and key regulator of absorptive and secretory cell fate decisions. Combined treatment with GSI and MLN4924 in a murine Notch1-dependent model of T-ALL led to leukemia regression and improved overall survival in the absence of gut toxicity. Overall, these results support the combined targeting of the NOTCH1 and neddylation pathways for the treatment of NOTCH1-induced T-ALL.
Collapse
Affiliation(s)
- Kalay Bertulfo
- Institute for Cancer Genetics, Columbia University, New York, NY10032
- Department of Biological Sciences, Columbia University, New York, NY10027
| | - Pablo Perez-Duran
- Institute for Cancer Genetics, Columbia University, New York, NY10032
| | - Hannah Miller
- Institute for Cancer Genetics, Columbia University, New York, NY10032
| | - Cindy Ma
- Institute for Cancer Genetics, Columbia University, New York, NY10032
| | | | - Jeremy Samon
- Institute for Cancer Genetics, Columbia University, New York, NY10032
| | - Adam Mackey
- Institute for Cancer Genetics, Columbia University, New York, NY10032
| | - Wen-Hsuan Wendy Lin
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY10032
| | - Adolfo A. Ferrando
- Institute for Cancer Genetics, Columbia University, New York, NY10032
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY10032
- Department of Pediatrics, Columbia University Medical Center, New York, NY10032
| | - Teresa Palomero
- Institute for Cancer Genetics, Columbia University, New York, NY10032
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY10032
| |
Collapse
|
4
|
Bhattacharya S, Tie G, Singh PNP, Malagola E, Eskiocak O, He R, Kraiczy J, Gu W, Perlov Y, Alici-Garipcan A, Beyaz S, Wang TC, Zhou Q, Shivdasani RA. Intestinal secretory differentiation reflects niche-driven phenotypic and epigenetic plasticity of a common signal-responsive terminal cell. Cell Stem Cell 2025:S1934-5909(25)00095-5. [PMID: 40203837 DOI: 10.1016/j.stem.2025.03.005] [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: 09/19/2024] [Revised: 12/27/2024] [Accepted: 03/10/2025] [Indexed: 04/11/2025]
Abstract
Enterocytes and four classic secretory cell types derive from intestinal epithelial stem cells. Based on morphology, location, and canonical markers, goblet and Paneth cells are considered distinct secretory types. Here, we report high overlap in their transcripts and sites of accessible chromatin, in marked contrast to those of their enteroendocrine or tuft cell siblings. Mouse and human goblet and Paneth cells express extraordinary fractions of few antimicrobial genes, which reflect specific responses to local niches. Wnt signaling retains some ATOH1+ secretory cells in crypt bottoms, where the absence of BMP signaling potently induces Paneth features. Cells that migrate away from crypt bottoms encounter BMPs and thereby acquire goblet properties. These phenotypes and underlying accessible cis-elements interconvert in post-mitotic cells. Thus, goblet and Paneth properties represent alternative phenotypic manifestations of a common signal-responsive terminal cell type. These findings reveal exquisite niche-dependent cell plasticity and cis-regulatory dynamics in likely response to antimicrobial needs.
Collapse
Affiliation(s)
- Swarnabh Bhattacharya
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Guodong Tie
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Pratik N P Singh
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Ermanno Malagola
- Division of Digestive and Liver Diseases, Department of Medicine and Irving Cancer Research Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Onur Eskiocak
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Graduate Program in Genetics, State University of New York, Stony Brook, NY 11794, USA
| | - Ruiyang He
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Judith Kraiczy
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Wei Gu
- Division of Regenerative Medicine & Hartman Institute for Therapeutic Organ Regeneration, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Yakov Perlov
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | | | - Semir Beyaz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Timothy C Wang
- Division of Digestive and Liver Diseases, Department of Medicine and Irving Cancer Research Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Qiao Zhou
- Division of Regenerative Medicine & Hartman Institute for Therapeutic Organ Regeneration, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ramesh A Shivdasani
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| |
Collapse
|
5
|
Guerin DJ, Gutierrez B, Zhang B, Tseng KAS. Notch Is Required for Neural Progenitor Proliferation During Embryonic Eye Regrowth. Int J Mol Sci 2025; 26:2637. [PMID: 40141279 PMCID: PMC11942531 DOI: 10.3390/ijms26062637] [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: 01/20/2025] [Revised: 02/27/2025] [Accepted: 03/03/2025] [Indexed: 03/28/2025] Open
Abstract
The ability of an organism to regrow tissues is regulated by various signaling pathways. One such pathway that has been studied widely both in the context of regeneration and development is the Notch signaling pathway. Notch is required for the development of the eye and regeneration of tissues in multiple organisms, but it is unknown if Notch plays a role in the regulation of Xenopus laevis embryonic eye regrowth. We found that Notch1 is required for eye regrowth and regulates retinal progenitor cell proliferation. Chemical and molecular inhibition of Notch1 significantly decreased eye regrowth by reducing retinal progenitor cell proliferation without affecting retinal differentiation. Temporal inhibition studies showed that Notch function is required during the first day of regrowth. Interestingly, Notch1 loss-of-function phenocopied the effects of the inhibition of the proton pump, vacuolar-type ATPase (V-ATPase), where retinal proliferation but not differentiation was blocked during eye regrowth. Overexpression of a form of activated Notch1, the Notch intracellular domain (NICD) rescued the loss of eye regrowth due to V-ATPase inhibition. These findings highlight the importance of the Notch signaling pathway in eye regeneration and its role in inducing retinal progenitor cell proliferation in response to injury.
Collapse
|
6
|
Kreimeyer H, Llorente C, Schnabl B. Influence of Alcohol on the Intestinal Immune System. Alcohol Res 2025; 45:03. [PMID: 40151622 PMCID: PMC11913448 DOI: 10.35946/arcr.v45.1.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025] Open
Abstract
PURPOSE Alcohol misuse is associated with disruption of the microbial homeostasis (dysbiosis) and microbial overgrowth in the gut, gut barrier disruption, and translocation of microbes into the systemic circulation. It also induces changes in regulatory mechanisms of the gut, which is the largest peripheral immune organ. The gut-liver axis is important for health and disease, and alterations in the intestinal immune system contribute to alcohol-associated liver disease (ALD). Understanding these changes might help discover new targets for drugs and therapeutic approaches. SEARCH METHODS A systematic literature search was conducted in PubMed, Medline, and Embase of manuscripts published between January 2000 and November 2023 using the terms ("alcohol" or "ethanol") AND ("immune" or "immunol") AND ("intestine," "colon," or "gut"). Eligible manuscripts included studies and reviews that discussed the effects of ethanol on immune cells in the intestine. SEARCH RESULTS A total of 506 publications were found in the databases on November 20, 2023. After excluding duplicates and research not covering ALD (415 articles), 91 studies were reviewed. Also included were manuscripts covering specific immune cells in the context of ALD. DISCUSSION AND CONCLUSIONS Balancing immune tolerance vs. initiating an immune response challenges the intestinal immune system. Alcohol induces disruption of the intestinal barrier, which is accompanied by a thicker mucus layer and reduced anti-microbial peptides. This leads to longer attachment of bacteria to epithelial cells and consequently greater translocation into the circulation. Bacterial translocation activates the immune system, reducing the activity of regulatory T cells and inducing T helper 17 response via a variety of pathways. The role of innate immune cells, especially Type 3 innate lymphoid cells, and of specific B- and T-cell subsets in ALD remains elusive. Gut dysbiosis, translocation of viable bacteria and bacterial products into the circulation, and changes in the intestinal barrier have been linked to immune deficiency and infections in patients with cirrhosis. Modifying the intestinal immune system could reduce intestinal inflammation and alcohol-induced liver injury. Understanding the underlying pathophysiology can help to detect new targets for drugs and design therapeutic strategies.
Collapse
Affiliation(s)
- Henriette Kreimeyer
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Cristina Llorente
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, California
- Department of Medicine, U.S. Department of Veterans Affairs San Diego Healthcare System, San Diego, California
| |
Collapse
|
7
|
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
|
8
|
Peng H, Song X, Chen J, Xiong X, Yang L, Yu C, Qiu M, Zhang Z, Hu C, Zhu S, Xia B, Wang J, Xiong Z, Du L, Yang C. Soybean bioactive peptide supplementation improves gut health and metabolism in broiler chickens. Poult Sci 2025; 104:104727. [PMID: 39729732 PMCID: PMC11741984 DOI: 10.1016/j.psj.2024.104727] [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: 09/18/2024] [Revised: 12/19/2024] [Accepted: 12/21/2024] [Indexed: 12/29/2024] Open
Abstract
This study aimed to investigate the effects of soybean bioactive peptide (SBP) on the growth performance and intestinal health of yellow-feathered broilers and to further elucidate the regulatory mechanisms of intestinal health using multi-omics analysis. A total of 320 1-day-old yellow-feathered broilers were randomly divided into two groups, with 10 replicates per group and 16 birds per replicate. Broilers in the control group received the basal diet, and those in the experimental group (SBPG) received the basal diet with 0.2 % SBP replacing the same amount of soybean meal. The experiment lasted for 70 d. The results showed that, compared with those in the control group, the final body weight and average daily gain of SBPG broilers were significantly higher (P < 0.05), and the feed conversion ratio was significantly lower (P < 0.05). Notably, SBP significantly improved gut health in chickens, including increased intestinal villus height, decreased levels of proinflammatory factors, such as IL-1β and interferon-γ, and upregulated expression of tight junction proteins, such as ZO-1 and occludin. In addition, transcriptome sequencing results revealed that broilers in the SBP group exhibited significant enrichment in multiple metabolic pathways, including fatty acid metabolism, fatty acid degradation, and the biosynthesis of unsaturated fatty acids (P < 0.05). Cecal 16S rRNA sequencing showed that SBPG increased the abundance of the butyrate-producing beneficial bacteria Muribaculaceae. Subsequent cecal metabolome analysis also revealed that SBPG enhanced lipid-related metabolic pathways, such as alpha-linolenic acid metabolism and GPI-anchor biosynthesis. In conclusion, SBP is a potential feed additive that can improve intestinal morphology, enhance intestinal immunity and barrier function, optimize the structure of the intestinal microbiota, and enhance metabolic function.
Collapse
Affiliation(s)
- Han Peng
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China
| | - Xiaoyan Song
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China
| | - Jialei Chen
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China
| | - Xia Xiong
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China
| | - Li Yang
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China
| | - Chunlin Yu
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China
| | - Mohan Qiu
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China
| | - Zengrong Zhang
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China
| | - Chenming Hu
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China
| | - Shiliang Zhu
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China
| | - Bo Xia
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China
| | - Jiangxian Wang
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China
| | - Zhuxiang Xiong
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China
| | - Longhuan Du
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China
| | - Chaowu Yang
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China.
| |
Collapse
|
9
|
Poghosyan S, Frenkel N, van den Bent L, Raats D, Spaapen T, Laoukili J, Borel Rinkes I, Kranenburg O, Hagendoorn J. VEGF-C propagates 'onward' colorectal cancer metastasis from liver to lung. Br J Cancer 2025; 132:69-80. [PMID: 39521880 PMCID: PMC11724081 DOI: 10.1038/s41416-024-02892-4] [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: 07/23/2023] [Revised: 10/09/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND The formation of lung metastasis as part of the progression of colon cancer is a poorly understood process. Theoretically, liver metastases could seed lung metastases. METHODS To assess the contribution of the liver lymphatic vasculature to metastatic spread to the lungs, we generated murine liver-metastasis-derived organoids overexpressing vascular endothelial growth factor (VEGF)-C. The organoids were reimplanted into the mouse liver for tumour generation and onward metastasis. RESULTS Liver metastases from patients with concomitant lung metastases showed higher expression of VEGF-C, lymphatic vessel hyperplasia, and tumour cell invasion into lymphatic vessels when compared to those without lung metastases. Reimplantation of VEGF-C overexpressing organoids into the mouse liver showed that VEGF-C caused peritumoral lymphatic vessel hyperplasia, lymphatic tumour cell invasion, and lung metastasis formation. This change in metastatic organotropism was accompanied by reduced expression of WNT-driven adult stem cell markers, and increased expression of fetal stem cell markers and NOTCH pathway genes. Further NOTCH pathway inhibition with γ-secretase inhibitor (DAPT) in vivo results in a slight reduction in lung metastases and a decrease in lymphatic hyperplasia and invasion in VEGF-C-overexpressing tumours. CONCLUSION Collectively, these data indicate that VEGF-C can drive onward metastasis from the liver to the lung and suggest that targeting VEGF-C/NOTCH pathways may impair the progression of colorectal cancer.
Collapse
Affiliation(s)
- Susanna Poghosyan
- Laboratory for Translational Oncology and Department of Surgical Oncology, Division of Imaging and Cancer, University Medical Center Utrecht and Utrecht University, Heidelberglaan 100, 3584CX, Utrecht, The Netherlands.
| | - Nicola Frenkel
- Laboratory for Translational Oncology and Department of Surgical Oncology, Division of Imaging and Cancer, University Medical Center Utrecht and Utrecht University, Heidelberglaan 100, 3584CX, Utrecht, The Netherlands
| | - Lotte van den Bent
- Laboratory for Translational Oncology and Department of Surgical Oncology, Division of Imaging and Cancer, University Medical Center Utrecht and Utrecht University, Heidelberglaan 100, 3584CX, Utrecht, The Netherlands
| | - Danielle Raats
- Laboratory for Translational Oncology and Department of Surgical Oncology, Division of Imaging and Cancer, University Medical Center Utrecht and Utrecht University, Heidelberglaan 100, 3584CX, Utrecht, The Netherlands
| | - Tessa Spaapen
- Laboratory for Translational Oncology and Department of Surgical Oncology, Division of Imaging and Cancer, University Medical Center Utrecht and Utrecht University, Heidelberglaan 100, 3584CX, Utrecht, The Netherlands
| | - Jamila Laoukili
- Laboratory for Translational Oncology and Department of Surgical Oncology, Division of Imaging and Cancer, University Medical Center Utrecht and Utrecht University, Heidelberglaan 100, 3584CX, Utrecht, The Netherlands
| | - Inne Borel Rinkes
- Laboratory for Translational Oncology and Department of Surgical Oncology, Division of Imaging and Cancer, University Medical Center Utrecht and Utrecht University, Heidelberglaan 100, 3584CX, Utrecht, The Netherlands
| | - Onno Kranenburg
- Laboratory for Translational Oncology and Department of Surgical Oncology, Division of Imaging and Cancer, University Medical Center Utrecht and Utrecht University, Heidelberglaan 100, 3584CX, Utrecht, The Netherlands
| | - Jeroen Hagendoorn
- Laboratory for Translational Oncology and Department of Surgical Oncology, Division of Imaging and Cancer, University Medical Center Utrecht and Utrecht University, Heidelberglaan 100, 3584CX, Utrecht, The Netherlands
| |
Collapse
|
10
|
Quintero M, Samuelson LC. Paneth Cells: Dispensable yet Irreplaceable for the Intestinal Stem Cell Niche. Cell Mol Gastroenterol Hepatol 2024; 19:101443. [PMID: 39708920 PMCID: PMC11847746 DOI: 10.1016/j.jcmgh.2024.101443] [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: 09/06/2024] [Revised: 12/09/2024] [Accepted: 12/09/2024] [Indexed: 12/23/2024]
Abstract
Intestinal stem cells replenish the epithelium throughout life by continuously generating intestinal epithelial cell types, including absorptive enterocytes, and secretory goblet, endocrine, and Paneth cells. This process is orchestrated by a symphony of niche factors required to maintain intestinal stem cells and to direct their proliferation and differentiation. Among the various mature intestinal epithelial cell types, Paneth cells are unique in their location in the stem cell zone, directly adjacent to intestinal stem cells. Although Paneth cells were first described as an epithelial cell component of the innate immune system due to their expression of anti-microbial peptides, they have been proposed to be niche cells due to their close proximity to intestinal stem cells and expression of niche factors. However, function as a niche cell has been debated since mice lacking Paneth cells retain functional stem cells that continue to replenish the intestinal epithelium. In this review, we summarize the intestinal stem cell niche, including the Notch, Wnt, growth factor, mechanical, and metabolic niche, and discuss how Paneth cells might contribute to these various components. We also present a nuanced view of the Paneth cell as a niche cell. Although not required, Paneth cells enhance stem cell function, particularly during intestinal development and regeneration. Furthermore, we suggest that Paneth cell loss induces intestinal stem cell remodeling to adjust their niche demands.
Collapse
Affiliation(s)
- Michaela Quintero
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Linda C Samuelson
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan; Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
11
|
Yu LE, Yang WC, Liang YC. Crosstalk Within the Intestinal Epithelium: Aspects of Intestinal Absorption, Homeostasis, and Immunity. Biomedicines 2024; 12:2771. [PMID: 39767678 PMCID: PMC11673925 DOI: 10.3390/biomedicines12122771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
Gut health is crucial in many ways, such as in improving human health in general and enhancing production in agricultural animals. To maximize the effect of a healthy gastrointestinal tract (GIT), an understanding of the regulation of intestinal functions is needed. Proper intestinal functions depend on the activity, composition, and behavior of intestinal epithelial cells (IECs). There are various types of IECs, including enterocytes, Paneth cells, enteroendocrine cells (EECs), goblet cells, tuft cells, M cells, and intestinal epithelial stem cells (IESCs), each with unique 3D structures and IEC distributions. Although the communication between IECs and other cell types, such as immune cells and neurons, has been intensively reviewed, communication between different IECs has rarely been addressed. The present paper overviews the networks among IECs that influence intestinal functions. Intestinal absorption is regulated by incretins derived from EECs that induce nutrient transporter activity in enterocytes. EECs, Paneth cells, tuft cells, and enterocytes release signals to activate Notch signaling, which modulates IESC activity and intestinal homeostasis, including proliferation and differentiation. Intestinal immunity can be altered via EECs, goblet cells, tuft cells, and cytokines derived from IECs. Finally, tools for investigating IEC communication have been discussed, including the novel 3D intestinal cell model utilizing enteroids that can be considered a powerful tool for IEC communication research. Overall, the importance of IEC communication, especially EECs and Paneth cells, which cover most intestinal functional regulating pathways, are overviewed in this paper. Such a compilation will be helpful in developing strategies for maintaining gut health.
Collapse
Affiliation(s)
| | | | - Yu-Chaun Liang
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 115201, Taiwan; (L.-E.Y.); (W.-C.Y.)
| |
Collapse
|
12
|
Feng X, Flüchter P, De Tenorio JC, Schneider C. Tuft cells in the intestine, immunity and beyond. Nat Rev Gastroenterol Hepatol 2024; 21:852-868. [PMID: 39327439 DOI: 10.1038/s41575-024-00978-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/02/2024] [Indexed: 09/28/2024]
Abstract
Tuft cells have gained substantial attention over the past 10 years due to numerous reports linking them with type 2 immunity and microorganism-sensing capacity in many mucosal tissues. This heightened interest is fuelled by their unique ability to produce an array of biological effector molecules, including IL-25, allergy-related eicosanoids, and the neurotransmitter acetylcholine, enabling downstream responses in diverse cell types. Operating through G protein-coupled receptor-mediated signalling pathways reminiscent of type II taste cells in oral taste buds, tuft cells emerge as chemosensory sentinels that integrate luminal conditions, eliciting appropriate responses in immune, epithelial and neuronal populations. How tuft cells promote tissue alterations and adaptation to the variety of stimuli at mucosal surfaces has been explored in multiple studies in the past few years. Since the initial recognition of the role of tuft cells, the discovery of diverse tuft cell effector functions and associated feedback loops have also revealed the complexity of tuft cell biology. Although earlier work largely focused on extraintestinal tissues, novel genetic tools and recent mechanistic studies on intestinal tuft cells established fundamental concepts of tuft cell activation and functions. This Review is an overview of intestinal tuft cells, providing insights into their development, signalling and interaction modules in immunity and other states.
Collapse
Affiliation(s)
- Xiaogang Feng
- Department of Physiology, University of Zurich, Zurich, Switzerland
| | - Pascal Flüchter
- Department of Physiology, University of Zurich, Zurich, Switzerland
| | | | | |
Collapse
|
13
|
Momoh M, Rathan-Kumar S, Burman A, Brown ME, Adeniran F, Ramos C, Goldenring JR, Roland JT, Kaji I. Alterations in cellular metabolic pathway and epithelial cell maturation induced by MYO5B defects are partially reversible by LPAR5 activation. Am J Physiol Gastrointest Liver Physiol 2024; 327:G877-G899. [PMID: 39404772 PMCID: PMC11684887 DOI: 10.1152/ajpgi.00091.2024] [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: 03/25/2024] [Revised: 09/12/2024] [Accepted: 09/24/2024] [Indexed: 10/23/2024]
Abstract
Functional loss of the motor protein myosin Vb (MYO5B) induces various defects in intestinal epithelial function and causes a congenital diarrheal disorder, namely, microvillus inclusion disease (MVID). Utilizing the MVID model mice Vil1-CreERT2;Myo5bflox/flox (MYO5BΔIEC) and Vil1-CreERT2;Myo5bflox/G519R [MYO5B(G519R)], we previously reported that functional MYO5B loss disrupts progenitor cell differentiation and enterocyte maturation that result in villus blunting and deadly malabsorption symptoms. In this study, we determined that both absence and a point mutation of MYO5B impair lipid metabolism and alter mitochondrial structure, which may underlie the progenitor cell malfunction observed in the MVID intestine. Along with a decrease in fatty acid oxidation, the lipogenesis pathway was enhanced in the MYO5BΔIEC small intestine. Consistent with these observations in vivo, RNA sequencing of enteroids generated from the two MVID mouse strains showed similar downregulation of energy metabolic enzymes, including mitochondrial oxidative phosphorylation genes. In our previous studies, we reported that lysophosphatidic acid (LPA) signaling ameliorated epithelial cell defects in MYO5BΔIEC tissues and enteroids. The present study demonstrated that the highly soluble LPA receptor (LPAR)5-preferred agonist Compound-1 improved sodium transporter localization and absorptive function and tuft cell differentiation in patient-modeled MVID animals that carry independent mutations in MYO5B. Body weight loss in male MYO5B(G519R) mice was ameliorated by Compound-1. These observations suggest that Compound-1 treatment has a trophic effect on the intestine with MYO5B functional loss through epithelial cell-autonomous pathways that can accelerate the differentiation of progenitor cells and the maturation of enterocytes. Targeting LPAR5 may represent an effective therapeutic approach for the treatment of MVID symptoms induced by different point mutations in MYO5B.NEW & NOTEWORTHY This study demonstrates the importance of MYO5B for cellular lipid metabolism and mitochondria in intestinal epithelial cells, previously unexplored functions of MYO5B. The alterations may underlie the progenitor cell malfunction observed in microvillus inclusion disease (MVID) intestines. To examine the therapeutic potential of progenitor-targeted treatments, the effects of the LPAR5-preferred agonist Compound-1 were investigated utilizing several MVID model mice and enteroids. Our observations suggest that Compound-1 may provide a therapeutic approach for treating MVID.
Collapse
Affiliation(s)
- Michael Momoh
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States
| | - Sudiksha Rathan-Kumar
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States
| | - Andreanna Burman
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States
| | - Monica E Brown
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States
| | - Francisca Adeniran
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States
| | - Cynthia Ramos
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States
| | - James R Goldenring
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States
- Nashville VA Medical Center, Nashville, Tennessee, United States
| | - Joseph T Roland
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States
| | - Izumi Kaji
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States
| |
Collapse
|
14
|
Dai Q, Preusse K, Yu D, Kovall RA, Thorner K, Lin X, Kopan R. Loss of Notch dimerization perturbs intestinal homeostasis by a mechanism involving HDAC activity. PLoS Genet 2024; 20:e1011486. [PMID: 39666740 DOI: 10.1371/journal.pgen.1011486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 12/26/2024] [Accepted: 11/04/2024] [Indexed: 12/14/2024] Open
Abstract
A tri-protein complex containing NICD, RBPj and MAML1 binds DNA as monomer or as cooperative dimers to regulate transcription. Mice expressing Notch dimerization-deficient alleles (NDD) of Notch1 and Notch2 are sensitized to environmental insults but otherwise develop and age normally. Transcriptomic analysis of colonic spheroids uncovered no evidence of dimer-dependent target gene miss-regulation, confirmed impaired stem cell maintenance in-vitro, and discovered an elevated signature of epithelial innate immune response to symbionts, a likely underlying cause for heightened sensitivity in NDD mice. TurboID followed by quantitative nano-spray MS/MS mass-spectrometry analyses in a human colon carcinoma cell line expressing either NOTCH2DD or NOTCH2 revealed an unbalanced interactome, with reduced interaction of NOTCH2DD with the transcription machinery but relatively preserved interaction with the HDAC2 interactome suggesting modulation via cooperativity. To ask if HDAC2 activity contributes to Notch loss-of-function phenotypes, we used the HDAC2 inhibitor Valproic acid (VPA) and discovered it could prevent the intestinal consequences of NDD and gamma secretase inhibitors (DBZ or DAPT) treatment in mice and spheroids, suggesting synergy between HDAC activity and pro-differentiation program in intestinal stem cells.
Collapse
Affiliation(s)
- Quanhui Dai
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Greater Bay Area Institute of Precision Medicine (Guangzhou), Zhongshan Hospital, Fudan University, Shanghai, China
- Division of Developmental Biology, Department of Pediatrics, University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Kristina Preusse
- Division of Developmental Biology, Department of Pediatrics, University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Danni Yu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Greater Bay Area Institute of Precision Medicine (Guangzhou), Zhongshan Hospital, Fudan University, Shanghai, China
| | - Rhett A Kovall
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Konrad Thorner
- Division of Developmental Biology, Department of Pediatrics, University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Xinhua Lin
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Greater Bay Area Institute of Precision Medicine (Guangzhou), Zhongshan Hospital, Fudan University, Shanghai, China
| | - Raphael Kopan
- Division of Developmental Biology, Department of Pediatrics, University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| |
Collapse
|
15
|
Ma Q, Meng M, Zhou X, Guo W, Feng K, Huang T, Cai YD. Identification of Key Genes in Fetal Gut Development at Single-Cell Level by Exploiting Machine Learning Techniques. Proteomics 2024; 24:e202400104. [PMID: 39324223 DOI: 10.1002/pmic.202400104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 08/29/2024] [Accepted: 09/11/2024] [Indexed: 09/27/2024]
Abstract
The study of fetal gut development is critical due to its substantial influence on immediate neonatal and long-term adult health. Current research largely focuses on microbiome colonization, gut immunity, and barrier function, alongside the impact of external factors on these phenomena. Limited research has been dedicated to the categorization of developing fetal gut cells. Our study aimed to enhance our understanding of fetal gut development by employing advanced machine-learning techniques on single-cell sequencing data. This dataset consisted of 62,849 samples, each characterized by 33,694 distinct gene features. Four feature ranking algorithms were utilized to sort features according to their significance, resulting in four feature lists. Then, these lists were fed into an incremental feature selection method to extract essential genes, classification rules, and build efficient classifiers. Several important genes were recognized by multiple feature ranking algorithms, such as FGG, MDK, RBP1, RBP2, IGFBP7, and SPON2. These features were key in differentiating specific developing intestinal cells, including epithelial, immune, mesenchymal, and vasculature cells of the colon, duo jejunum, and ileum cells. The classification rules showed special gene expression patterns on some intestinal cell types and the efficient classifiers can be useful tools for identifying intestinal cells.
Collapse
Affiliation(s)
- QingLan Ma
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Mei Meng
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - XianChao Zhou
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Guo
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, China
| | - KaiYan Feng
- Department of Computer Science, Guangdong AIB Polytechnic College, Guangzhou, China
| | - Tao Huang
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai, China
| |
Collapse
|
16
|
Baghdadi MB, Houtekamer RM, Perrin L, Rao-Bhatia A, Whelen M, Decker L, Bergert M, Pérez-Gonzàlez C, Bouras R, Gropplero G, Loe AKH, Afkhami-Poostchi A, Chen X, Huang X, Descroix S, Wrana JL, Diz-Muñoz A, Gloerich M, Ayyaz A, Matic Vignjevic D, Kim TH. PIEZO-dependent mechanosensing is essential for intestinal stem cell fate decision and maintenance. Science 2024; 386:eadj7615. [PMID: 39607940 DOI: 10.1126/science.adj7615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 07/27/2024] [Accepted: 10/07/2024] [Indexed: 11/30/2024]
Abstract
Stem cells perceive and respond to biochemical and physical signals to maintain homeostasis. Yet, it remains unclear how stem cells sense mechanical signals from their niche in vivo. In this work, we investigated the roles of PIEZO mechanosensitive channels in the intestinal stem cell (ISC) niche. We used mouse genetics and single-cell RNA sequencing analysis to assess the requirement for PIEZO channels in ISC maintenance. In vivo measurement of basement membrane stiffness showed that ISCs reside in a more rigid microenvironment at the bottom of the crypt. Three-dimensional and two-dimensional organoid systems combined with bioengineered substrates and a stretching device revealed that PIEZO channels sense extracellular mechanical stimuli to modulate ISC function. This study delineates the mechanistic cascade of PIEZO activation that coordinates ISC fate decision and maintenance.
Collapse
Affiliation(s)
- Meryem B Baghdadi
- Institut Curie, PSL Research University, CNRS UMR 144, Paris, France
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Ronja M Houtekamer
- Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - Louisiane Perrin
- Institut Curie, PSL Research University, CNRS UMR 144, Paris, France
| | - Abilasha Rao-Bhatia
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Myles Whelen
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, AB, Canada
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Linda Decker
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Martin Bergert
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | | | - Réda Bouras
- Institut Curie, PSL Research University, CNRS UMR 144, Paris, France
| | - Giacomo Gropplero
- Institut Curie, IPGG, PSL Research University, CNRS UMR 168, Paris, France
| | - Adrian K H Loe
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Amin Afkhami-Poostchi
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Xin Chen
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Xi Huang
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Stephanie Descroix
- Institut Curie, IPGG, PSL Research University, CNRS UMR 168, Paris, France
| | - Jeffrey L Wrana
- Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Laboratory Medicine, St. Michael's Hospital, Toronto, ON, Canada
| | - Alba Diz-Muñoz
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Martijn Gloerich
- Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - Arshad Ayyaz
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, AB, Canada
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | | | - Tae-Hee Kim
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
17
|
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
|
18
|
Gámez-Belmonte R, Wagner Y, Mahapatro M, Wang R, Erkert L, González-Acera M, Cineus R, Hainbuch S, Patankar JV, Voehringer D, Hegazy AN, Neurath MF, Wirtz S, Becker C. Intestinal epithelial Gasdermin C is induced by IL-4R/STAT6 signaling but is dispensable for gut immune homeostasis. Sci Rep 2024; 14:26522. [PMID: 39489845 PMCID: PMC11532336 DOI: 10.1038/s41598-024-78336-z] [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: 06/19/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024] Open
Abstract
Gasdermin C is one of the least studied members of the gasdermin family of proteins, known for their critical involvement in pyroptosis and host defense. Furthermore, evidence for the role of Gasdermin C in the intestine is scarce and partly controversial. Here, we tested the functional role of Gasdermin C in intestinal homeostasis, inflammation and tumorigenesis. : We studied Gasdermin C in response to cytokines in intestinal organoids. We evaluated epithelial differentiation, cell death and immune infiltration under steady state conditions in a new mouse line deficient in Gasdermin C. The role of Gasdermin C was analyzed in acute colitis, infection and colitis-associated cancer. Gasdemin C is highly expressed in the intestinal epithelium and strongly induced by the type 2 cytokines IL-4 and IL-13 in a STAT6-dependent manner. Gasdermin C-deficient mice show no changes in tissue architecture and epithelial homeostasis. Epithelial organoids deficient in Gasdermin C develop normally and show no alterations in proliferation or cell death. No changes were found in models of acute colitis, type 2 intestinal infection and colitis-associated cancer. Gasdermin C genes are upregulated by type 2 immunity, yet appear dispensable for the development of intestinal inflammation, infection and colitis-associated cancer.
Collapse
Affiliation(s)
- Reyes Gámez-Belmonte
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Yara Wagner
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Mousumi Mahapatro
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Ru Wang
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Lena Erkert
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Miguel González-Acera
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Roodline Cineus
- Department of Gastroenterology, Infectiology and Rheumatology, Charité Universitätsmedizin, Berlin, Germany
- Deutsches Rheumaforschungszentrum Berlin (DRFZ), An Institute of the Leibniz Association, Berlin, Germany
| | - Saskia Hainbuch
- Deutsches Rheumaforschungszentrum Berlin (DRFZ), An Institute of the Leibniz Association, Berlin, Germany
| | - Jay V Patankar
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - David Voehringer
- Institute of Immunology, Ludwig-Maximilians-Universität München, 80336, München, Germany
- Department of Infection Biology, University of Erlangen, 91054, Erlangen, Germany
| | - Ahmed N Hegazy
- Department of Gastroenterology, Infectiology and Rheumatology, Charité Universitätsmedizin, Berlin, Germany
- Deutsches Rheumaforschungszentrum Berlin (DRFZ), An Institute of the Leibniz Association, Berlin, Germany
- The Transregio 241 IBDome Consortium, Berlin, Germany
| | - Markus F Neurath
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- The Transregio 241 IBDome Consortium, Berlin, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Stefan Wirtz
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- The Transregio 241 IBDome Consortium, Berlin, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Christoph Becker
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.
- The Transregio 241 IBDome Consortium, Berlin, Germany.
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany.
- Department of Medicine 1, University Medical Centre Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
19
|
Zhang R, Perekatt A, Chen L. Metabolic regulation of intestinal homeostasis: molecular and cellular mechanisms and diseases. MedComm (Beijing) 2024; 5:e776. [PMID: 39465140 PMCID: PMC11502721 DOI: 10.1002/mco2.776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/21/2024] [Accepted: 09/22/2024] [Indexed: 10/29/2024] Open
Abstract
Metabolism serves not only as the organism's energy source but also yields metabolites crucial for maintaining tissue homeostasis and overall health. Intestinal stem cells (ISCs) maintain intestinal homeostasis through continuous self-renewal and differentiation divisions. The intricate relationship between metabolic pathways and intestinal homeostasis underscores their crucial interplay. Metabolic pathways have been shown to directly regulate ISC self-renewal and influence ISC fate decisions under homeostatic conditions, but the cellular and molecular mechanisms remain incompletely understood. Understanding the intricate involvement of various pathways in maintaining intestinal homeostasis holds promise for devising innovative strategies to address intestinal diseases. Here, we provide a comprehensive review of recent advances in the regulation of intestinal homeostasis. We describe the regulation of intestinal homeostasis from multiple perspectives, including the regulation of intestinal epithelial cells, the regulation of the tissue microenvironment, and the key role of nutrient metabolism. We highlight the regulation of intestinal homeostasis and ISC by nutrient metabolism. This review provides a multifaceted perspective on how intestinal homeostasis is regulated and provides ideas for intestinal diseases and repair of intestinal damage.
Collapse
Affiliation(s)
- Ruolan Zhang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human DiseaseSoutheast UniversityNanjingChina
| | - Ansu Perekatt
- Department of Chemistry and Chemical BiologyStevens Institute of TechnologyHobokenNew JerseyUSA
| | - Lei Chen
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human DiseaseSoutheast UniversityNanjingChina
- Institute of Microphysiological SystemsSoutheast UniversityNanjingChina
| |
Collapse
|
20
|
Lin Y, Lu Y, Wang Y, Lv C, Chen J, Luo Y, Quan H, Yu W, Chen L, Huang Z, Hao Y, Wang Q, Luo Q, Yan J, Li Y, Zhang W, Du M, He J, Ren F, Guo H. The Regeneration of Intestinal Stem Cells Is Driven by miR-29-Induced Metabolic Reprogramming. ENGINEERING 2024; 42:39-58. [DOI: 10.1016/j.eng.2024.08.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
|
21
|
Anbazhagan M, Sharma G, Murthy S, Maddipatla SC, Kolachala VL, Dodd A, Randunne A, Cutler DJ, Kugathasan S, Matthews JD. PTGER4 signaling regulates class IIa HDAC function and SPINK4 mRNA levels in rectal epithelial cells. Cell Commun Signal 2024; 22:493. [PMID: 39396982 PMCID: PMC11472582 DOI: 10.1186/s12964-024-01879-1] [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: 07/24/2024] [Accepted: 10/06/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND The prostaglandin receptor PTGER4 facilitates homeostasis in the gut. Previous reports indicate that goblet cells, marked by SPINK4 expression, might be affected by PTGER4 activity. Current evidence suggests that prostaglandin E2 (PGE2) produced by mesenchymal stromal cells (MSC) stimulates PTGER4 in epithelial cells during inflammatory conditions. Here, we investigate the subcellular mechanisms and mRNA levels downstream of PTGER4 activity in epithelial cells. METHODS Mucosal cells, organoids, and MSC were obtained from patient biopsies harvested by endoscopy. Using independent and co-cultures, we manipulated the activity of PTGER4, the downstream enzymes, and mRNA levels, by using PGE2, in combination with chemical inhibitors, L-161982, H89, LB100, DAPT, LMK-235, or with butyrate. Immunofluorescence, single cell sequencing, RNAscope, ELISA, real time PCR, and Western blotting were used to examine these samples. RESULTS SPINK4 mRNA levels were increased in organoids by co-culture with MSC or exogenous stimulation with PGE2 that could be blocked by L-161982 or LMK-235, PTGER4 or HDAC4 inhibitors, respectively. Expression of PTGER4 was co-localized with JAM-A in the basolateral surfaces in rectal epithelial cells grown as organoids. PGE2 treatment of rectal organoids decreased HDAC4, 5, and 7 phosphorylation levels that could be blocked by L-161982 treatment. Butyrate treatment, or addition of L-161982, increased the phosphorylated levels of HDAC4, 5, and 7. CONCLUSIONS These findings suggest a mechanism during mucosal injury whereby MSC production of PGE2 increases HDAC4, 5, and 7 activities in epithelial cells by upregulating PTGER4 signaling, ultimately increasing SPINK4 mRNA levels and extracellular release of SPINK4.
Collapse
Affiliation(s)
- Murugadas Anbazhagan
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Emory University School of Medicine, Emory University, Atlanta, GA, USA
| | - Garima Sharma
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Emory University School of Medicine, Emory University, Atlanta, GA, USA
| | - Shanta Murthy
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Emory University School of Medicine, Emory University, Atlanta, GA, USA
| | - Sushma Chowdary Maddipatla
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Emory University School of Medicine, Emory University, Atlanta, GA, USA
| | - Vasantha L Kolachala
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Emory University School of Medicine, Emory University, Atlanta, GA, USA
| | - Anne Dodd
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Emory University School of Medicine, Emory University, Atlanta, GA, USA
| | - Amanda Randunne
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Emory University School of Medicine, Emory University, Atlanta, GA, USA
| | - David J Cutler
- Department of Human Genetics, Emory University, Atlanta, GA, USA
| | - Subra Kugathasan
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Emory University School of Medicine, Emory University, Atlanta, GA, USA
- Department of Pediatrics and Pediatric Research Institute, Division of Pediatric Gastroenterology, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, USA
| | - Jason D Matthews
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Emory University School of Medicine, Emory University, Atlanta, GA, USA.
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Health Science Research Building, 1760 Haygood Dr, E-246, Atlanta, GA, 30322, USA.
| |
Collapse
|
22
|
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
|
23
|
Jiang L, Tian J, Yang J, Luo R, Zhang Y, Shao C, Guo B, Wu X, Dan J, Luo Y. p21 Regulates Wnt-Notch balance via DREAM/MMB/Rb-E2F1 and maintains intestinal stem cell homeostasis. Cell Death Discov 2024; 10:413. [PMID: 39341834 PMCID: PMC11438959 DOI: 10.1038/s41420-024-02192-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024] Open
Abstract
The crosstalk and balance regulation of Wnt-Notch have been known to be essential for cell fate decision and tissue regeneration, however, how this balance is maintained and how the Wnt-Notch pathways are connected with cell cycle regulation is still not clear. By analyzing the molecular alterations in mouse model with accelerated aging phenotypes due to loss of p21 function in a Werner syndrome background, we observed that Wnt3 and β-Catenin were down-regulated, while Notch1 and Hes1 were up-regulated. This disruption in Wnt-Notch signaling was accompanied by the loss of intestinal stem cell compartment, increase in Bmi1 positive cells, loss of Olfm4/Lgr5 positive cells, and reduced secretory Paneth cells and goblet cells in the intestinal crypts of p21TKO mice. BrdU incorporation, cleaved caspase 3, and Tunel assay results revealed the fast turnover of intestinal epithelia, which may result in abnormal stem cell mobilization and exhaustion of the stem cell reservoir in the intestinal crypts. We further identified shift of DREAM complex towards MMB complex due to the loss of p21 as the cause for faster turnover of intestinal epithelia. Importantly, we identified the E2F1 as the transcriptional regulator for Notch1, which linked the p21-DREAM/MMB/Rb-E2F1 pathway with Wnt-Notch pathway. The overexpression of p21 rescued the DREAM pathway, as well as the imbalance of Wnt-Notch pathway. In summary, our data identify p21 as an important factor in maintaining sequential mobilization, proliferation, and homeostasis of intestinal stem cells.
Collapse
Affiliation(s)
- Liangxia Jiang
- Department of Pathophysiology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Jie Tian
- Department of Pathophysiology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Jun Yang
- Department of Pathophysiology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Ronggang Luo
- Department of Pathophysiology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yongjin Zhang
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Chihao Shao
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Bing Guo
- Department of Pathophysiology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Xiaoming Wu
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Juhua Dan
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Ying Luo
- Department of Pathophysiology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China.
| |
Collapse
|
24
|
Kayama H, Takeda K. Regulation of intestinal epithelial homeostasis by mesenchymal cells. Inflamm Regen 2024; 44:42. [PMID: 39327633 PMCID: PMC11426228 DOI: 10.1186/s41232-024-00355-0] [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: 03/18/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024] Open
Abstract
The gastrointestinal tract harbors diverse microorganisms in the lumen. Epithelial cells segregate the luminal microorganisms from immune cells in the lamina propria by constructing chemical and physical barriers through the production of various factors to prevent excessive immune responses against microbes. Therefore, perturbations of epithelial integrity are linked to the development of gastrointestinal disorders. Several mesenchymal stromal cell populations, including fibroblasts, myofibroblasts, pericytes, and myocytes, contribute to the establishment and maintenance of epithelial homeostasis in the gut through regulation of the self-renewal, proliferation, and differentiation of intestinal stem cells. Recent studies have revealed alterations in the composition of intestinal mesenchymal stromal cells in patients with inflammatory bowel disease and colorectal cancer. A better understanding of the interplay between mesenchymal stromal cells and epithelial cells associated with intestinal health and diseases will facilitate identification of novel biomarkers and therapeutic targets for gastrointestinal disorders. This review summarizes the key findings obtained to date on the mechanisms by which functionally distinct mesenchymal stromal cells regulate epithelial integrity in intestinal health and diseases at different developmental stages.
Collapse
Affiliation(s)
- Hisako Kayama
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
- WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.
- Institute for Advanced Co-Creation Studies, Osaka University, Suita, Osaka, 565-0871, Japan.
| | - Kiyoshi Takeda
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
25
|
Pujar AA, Barua A, Dey PS, Singh D, Roy U, Jolly MK, Hatzikirou H. Microenvironmental entropy dynamics analysis reveals novel insights into Notch-Delta-Jagged decision-making mechanism. iScience 2024; 27:110569. [PMID: 39318535 PMCID: PMC11420447 DOI: 10.1016/j.isci.2024.110569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/31/2024] [Accepted: 07/19/2024] [Indexed: 09/26/2024] Open
Abstract
Notch-Delta-Jagged (NDJ) signaling among neighboring cells contributes crucially to spatiotemporal pattern formation and developmental decision-making. Despite numerous detailed mathematical models, their high-dimensionality parametric space limits analytical treatment, especially regarding local microenvironmental fluctuations. Using the low-dimensional dynamics of the recently postulated least microenvironmental uncertainty principle (LEUP) framework, we showcase how the LEUP formalism recapitulates a noisy NDJ spatial patterning. Our LEUP simulations show that local phenotypic entropy increases for lateral inhibition but decreases for lateral induction. This distinction allows us to identify a critical parameter that captures the transition from a Notch-Delta-driven lateral inhibition to a Notch-Jagged-driven lateral induction phenomenon and suggests random phenotypic patterning in the case of lack of dominance of either Notch-Delta or Notch-Jagged signaling. Our results enable an analytical treatment to map the high-dimensional dynamics of NDJ signaling on tissue-level patterning and can possibly be generalized to decode operating principles of collective cellular decision-making.
Collapse
Affiliation(s)
- Aditi Ajith Pujar
- Department of Bioengineering, Indian Institute of Science, Bangalore 560012, India
- Undergraduate Program, Indian Institute of Science, Bangalore 560012, India
| | - Arnab Barua
- Tata Institute of Fundamental Research, Hyderabad 500046, India
| | - Partha Sarathi Dey
- Department of Bioengineering, Indian Institute of Science, Bangalore 560012, India
| | - Divyoj Singh
- Department of Bioengineering, Indian Institute of Science, Bangalore 560012, India
- Undergraduate Program, Indian Institute of Science, Bangalore 560012, India
| | - Ushasi Roy
- Department of Bioengineering, Indian Institute of Science, Bangalore 560012, India
| | - Mohit Kumar Jolly
- Department of Bioengineering, Indian Institute of Science, Bangalore 560012, India
| | - Haralampos Hatzikirou
- Mathematics Department, Khalifa University, P.O. Box: 127788, Abu Dhabi, UAE
- Technische Univesität Dresden, Center for Information Services and High Performance Computing, Nöthnitzer Straße 46, P.O. Box: 01062, Dresden, Germany
| |
Collapse
|
26
|
Momoh M, Rathan-Kumar S, Burman A, Brown ME, Adeniran F, Ramos C, Goldenring JR, Roland JT, Kaji I. Altered cellular metabolic pathway and epithelial cell maturation induced by MYO5B defects are partially reversible by LPAR5 activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.03.610579. [PMID: 39282272 PMCID: PMC11398351 DOI: 10.1101/2024.09.03.610579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Functional loss of the motor protein, Myosin Vb (MYO5B), induces various defects in intestinal epithelial function and causes a congenital diarrheal disorder, microvillus inclusion disease (MVID). Utilizing the MVID model mice, Vil1-Cre ERT2 ;Myo5b flox/flox (MYO5BΔIEC) and Vil1-Cre ERT2 ;Myo5b flox/G519R (MYO5B(G519R)), we previously reported that functional MYO5B loss disrupts progenitor cell differentiation and enterocyte maturation that result in villus blunting and deadly malabsorption symptoms. In this study, we determined that both absence and a point mutation of MYO5B impair lipid metabolism and alter mitochondrial structure, which may underlie the progenitor cell malfunction observed in MVID intestine. Along with a decrease in fatty acid oxidation, the lipogenesis pathway was enhanced in the MYO5BΔIEC small intestine. Consistent with these observations in vivo , RNA-sequencing of enteroids generated from two MVID mouse strains showed similar downregulation of energy metabolic enzymes, including mitochondrial oxidative phosphorylation genes. In our previous studies, lysophosphatidic acid (LPA) signaling ameliorates epithelial cell defects in MYO5BΔIEC tissues and enteroids. The present study demonstrates that the highly soluble LPAR5-preferred agonist, Compound-1, improved sodium transporter localization and absorptive function, and tuft cell differentiation in patient-modeled MVID animals that carry independent mutations in MYO5B. Body weight loss in male MYO5B(G519R) mice was ameliorated by Compound-1. These observations suggest that Compound-1 treatment has a trophic effect on intestine with MYO5B functional loss through epithelial cell-autonomous pathways that may improve the differentiation of progenitor cells and the maturation of enterocytes. Targeting LPAR5 may represent an effective therapeutic approach for treatment of MVID symptoms induced by different point mutations in MYO5B. NEW & NOTEWOTHY This study demonstrates the importance of MYO5B for cellular lipid metabolism and mitochondria in intestinal epithelial cells, a previously unexplored function of MYO5B. Alterations in cellular metabolism may underlie the progenitor cell malfunction observed in microvillus inclusion disease (MVID). To examine the therapeutic potential of progenitor-targeted treatments, the effects of LPAR5-preferred agonist, Compound-1, was investigated utilizing several MVID model mice and enteroids. Our observations suggests that Compound-1 may provide a therapeutic approach for treating MVID. Graphic Abstract
Collapse
|
27
|
Encarnacion-Garcia MR, De la Torre-Baez R, Hernandez-Cueto MA, Velázquez-Villegas LA, Candelario-Martinez A, Sánchez-Argáez AB, Horta-López PH, Montoya-García A, Jaimes-Ortega GA, Lopez-Bailon L, Piedra-Quintero Z, Carrasco-Torres G, De Ita M, Figueroa-Corona MDP, Muñoz-Medina JE, Sánchez-Uribe M, Ortiz-Fernández A, Meraz-Ríos MA, Silva-Olivares A, Betanzos A, Baay-Guzman GJ, Navarro-Garcia F, Villa-Treviño S, Garcia-Sierra F, Cisneros B, Schnoor M, Ortíz-Navarrete VF, Villegas-Sepúlveda N, Valle-Rios R, Medina-Contreras O, Noriega LG, Nava P. IFN-γ stimulates Paneth cell secretion through necroptosis mTORC1 dependent. Eur J Immunol 2024; 54:e2350716. [PMID: 38837757 DOI: 10.1002/eji.202350716] [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: 08/14/2023] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/07/2024]
Abstract
Immune mediators affect multiple biological functions of intestinal epithelial cells (IECs) and, like Paneth and Paneth-like cells, play an important role in intestinal epithelial homeostasis. IFN-γ a prototypical proinflammatory cytokine disrupts intestinal epithelial homeostasis. However, the mechanism underlying the process remains unknown. In this study, using in vivo and in vitro models we demonstrate that IFN-γ is spontaneously secreted in the small intestine. Furthermore, we observed that this cytokine stimulates mitochondrial activity, ROS production, and Paneth and Paneth-like cell secretion. Paneth and Paneth-like secretion downstream of IFN-γ, as identified here, is mTORC1 and necroptosis-dependent. Thus, our findings revealed that the pleiotropic function of IFN-γ also includes the regulation of Paneth cell function in the homeostatic gut.
Collapse
Affiliation(s)
- Maria R Encarnacion-Garcia
- Departament of Physiology, Biophysics, and Neurosciences, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Raúl De la Torre-Baez
- Departament of Physiology, Biophysics, and Neurosciences, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - María A Hernandez-Cueto
- Clinical Laboratory of Infectology, National Hospital "La Raza" Medical Center, IMSS, Mexico City, Mexico
| | - Laura A Velázquez-Villegas
- Physiology of Nutrition Department, The National Institute of Health Sciences and Nutrition "Salvador Zubirán", Mexico City, Mexico
| | - Aurora Candelario-Martinez
- Departament of Physiology, Biophysics, and Neurosciences, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Ana Beatriz Sánchez-Argáez
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Perla H Horta-López
- Departament of Physiology, Biophysics, and Neurosciences, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Armando Montoya-García
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Gustavo Alberto Jaimes-Ortega
- Immunology and Proteomics Research Laboratory, Children's Hospital of Mexico "Federico Gómez" (HIMFG), Mexico City, Mexico
- Experimental Biology Postgraduate Program, Department of Biological and Health Sciences, Metropolitan Autonomous University (UAM), Mexico City, Mexico
| | - Luis Lopez-Bailon
- Immunology Department and Immunology Postgraduate Program, National School of Biological Sciences of the National Polytechnic Institute (ENCB-IPN), Mexico City, Mexico
| | - Zayda Piedra-Quintero
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Gabriela Carrasco-Torres
- CICATA Unidad Morelos, Instituto Politécnico Nacional, Boulevard de la Tecnología, 1036 Z-1, P 2/2, Atlacholoaya, 62790, México
| | - Marlon De Ita
- Department of Genetics and Molecular Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
- Medical Research Unit in Human Genetics, UMAE Children's Hospital, National Medical Center "Siglo XXI", IMSS, Ciudad de México, 06720, Mexico
| | - María Del Pilar Figueroa-Corona
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - José Esteban Muñoz-Medina
- Clinical Laboratory of Infectology, National Hospital "La Raza" Medical Center, IMSS, Mexico City, Mexico
| | - Magdalena Sánchez-Uribe
- Pathological Anatomy, Specialized hospital "Dr. Antonio Fraga Mouret", National Hospital "La Raza" Medical Center, IMSS, Ciudad de México, México
| | - Arturo Ortiz-Fernández
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Marco Antonio Meraz-Ríos
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Angélica Silva-Olivares
- Departament of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Abigail Betanzos
- Departament of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | | | - Fernando Navarro-Garcia
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Saúl Villa-Treviño
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Francisco Garcia-Sierra
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Bulmaro Cisneros
- Department of Genetics and Molecular Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Michael Schnoor
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Vianney F Ortíz-Navarrete
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Nicolás Villegas-Sepúlveda
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Ricardo Valle-Rios
- Immunology and Proteomics Research Laboratory, Children's Hospital of Mexico "Federico Gómez" (HIMFG), Mexico City, Mexico
- University Research Unit, Research Division, Faculty of Medicine, National Autonomous University of Mexico-Children's Hospital of Mexico "Federico Gomez" (UNAM-HIMFG), Mexico City, Mexico
| | - Oscar Medina-Contreras
- Epidemiology, Endocrinology & Nutrition Research Unit, Children's Hospital of Mexico "Federico Gomez", Mexico City, Mexico
| | - Lilia G Noriega
- Physiology of Nutrition Department, The National Institute of Health Sciences and Nutrition "Salvador Zubirán", Mexico City, Mexico
| | - Porfirio Nava
- Departament of Physiology, Biophysics, and Neurosciences, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| |
Collapse
|
28
|
Wathieu C, Lavergne A, Xu X, Rolot M, Nemazanyy I, Shostak K, El Hachem N, Maurizy C, Leemans C, Close P, Nguyen L, Desmet C, Tielens S, Dewals BG, Chariot A. Loss of Elp3 blocks intestinal tuft cell differentiation via an mTORC1-Atf4 axis. EMBO J 2024; 43:3916-3947. [PMID: 39085648 PMCID: PMC11405396 DOI: 10.1038/s44318-024-00184-4] [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: 09/19/2023] [Revised: 06/25/2024] [Accepted: 07/12/2024] [Indexed: 08/02/2024] Open
Abstract
Intestinal tuft cells are critical for anti-helminth parasite immunity because they produce IL-25, which triggers IL-13 secretion by activated group 2 innate lymphoid cells (ILC2s) to expand both goblet and tuft cells. We show that epithelial Elp3, a tRNA-modifying enzyme, promotes tuft cell differentiation and is consequently critical for IL-25 production, ILC2 activation, goblet cell expansion and control of Nippostrongylus brasiliensis helminth infection in mice. Elp3 is essential for the generation of intestinal immature tuft cells and for the IL-13-dependent induction of glycolytic enzymes such as Hexokinase 1 and Aldolase A. Importantly, loss of epithelial Elp3 in the intestine blocks the codon-dependent translation of the Gator1 subunit Nprl2, an mTORC1 inhibitor, which consequently enhances mTORC1 activation and stabilizes Atf4 in progenitor cells. Likewise, Atf4 overexpression in mouse intestinal epithelium blocks tuft cell differentiation in response to intestinal helminth infection. Collectively, our data define Atf4 as a negative regulator of tuft cells and provide insights into promotion of intestinal type 2 immune response to parasites through tRNA modifications.
Collapse
Affiliation(s)
- Caroline Wathieu
- Interdisciplinary Cluster for Applied Genoproteomics, Liege, Belgium
- Laboratory of Cancer Biology, GIGA, University of Liege, Liege, Belgium
| | | | - Xinyi Xu
- Interdisciplinary Cluster for Applied Genoproteomics, Liege, Belgium
- Laboratory of Cancer Biology, GIGA, University of Liege, Liege, Belgium
| | - Marion Rolot
- Laboratory of Immunology-Vaccinology, Fundamental and Applied Research in Animals and Health (FARAH), University of Liege, Liege, Belgium
| | - Ivan Nemazanyy
- Platform for Metabolic Analyses, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS 3633, Paris, France
| | - Kateryna Shostak
- Interdisciplinary Cluster for Applied Genoproteomics, Liege, Belgium
- Laboratory of Cancer Biology, GIGA, University of Liege, Liege, Belgium
| | - Najla El Hachem
- Interdisciplinary Cluster for Applied Genoproteomics, Liege, Belgium
- Laboratory of Cancer Signaling, GIGA, University of Liege, Liege, Belgium
| | - Chloé Maurizy
- Interdisciplinary Cluster for Applied Genoproteomics, Liege, Belgium
- Laboratory of Cancer Biology, GIGA, University of Liege, Liege, Belgium
| | - Charlotte Leemans
- Interdisciplinary Cluster for Applied Genoproteomics, Liege, Belgium
- Laboratory of Cancer Signaling, GIGA, University of Liege, Liege, Belgium
| | - Pierre Close
- Interdisciplinary Cluster for Applied Genoproteomics, Liege, Belgium
- Laboratory of Cancer Signaling, GIGA, University of Liege, Liege, Belgium
- WELBIO department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium
| | - Laurent Nguyen
- Interdisciplinary Cluster for Applied Genoproteomics, Liege, Belgium
- WELBIO department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium
- Laboratory of Molecular Regulation of Neurogenesis, University of Liege, Liege, Belgium
| | - Christophe Desmet
- Interdisciplinary Cluster for Applied Genoproteomics, Liege, Belgium
- Laboratory of Cellular and Molecular Immunology, University of Liege, Liege, GIGA-I3, Belgium
| | - Sylvia Tielens
- Interdisciplinary Cluster for Applied Genoproteomics, Liege, Belgium
- Laboratory of Cancer Biology, GIGA, University of Liege, Liege, Belgium
| | - Benjamin G Dewals
- Laboratory of Immunology-Vaccinology, Fundamental and Applied Research in Animals and Health (FARAH), University of Liege, Liege, Belgium
| | - Alain Chariot
- Interdisciplinary Cluster for Applied Genoproteomics, Liege, Belgium.
- Laboratory of Cancer Biology, GIGA, University of Liege, Liege, Belgium.
- WELBIO department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium.
| |
Collapse
|
29
|
Urciuolo F, Imparato G, Netti PA. Engineering Cell Instructive Microenvironments for In Vitro Replication of Functional Barrier Organs. Adv Healthc Mater 2024; 13:e2400357. [PMID: 38695274 DOI: 10.1002/adhm.202400357] [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: 01/29/2024] [Revised: 04/02/2024] [Indexed: 05/14/2024]
Abstract
Multicellular organisms exhibit synergistic effects among their components, giving rise to emergent properties crucial for their genesis and overall functionality and survival. Morphogenesis involves and relies upon intricate and biunivocal interactions among cells and their environment, that is, the extracellular matrix (ECM). Cells secrete their own ECM, which in turn, regulates their morphogenetic program by controlling time and space presentation of matricellular signals. The ECM, once considered passive, is now recognized as an informative space where both biochemical and biophysical signals are tightly orchestrated. Replicating this sophisticated and highly interconnected informative media in a synthetic scaffold for tissue engineering is unattainable with current technology and this limits the capability to engineer functional human organs in vitro and in vivo. This review explores current limitations to in vitro organ morphogenesis, emphasizing the interplay of gene regulatory networks, mechanical factors, and tissue microenvironment cues. In vitro efforts to replicate biological processes for barrier organs such as the lung and intestine, are examined. The importance of maintaining cells within their native microenvironmental context is highlighted to accurately replicate organ-specific properties. The review underscores the necessity for microphysiological systems that faithfully reproduce cell-native interactions, for advancing the understanding of developmental disorders and disease progression.
Collapse
Affiliation(s)
- Francesco Urciuolo
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI) and Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Piazzale Tecchio 80, Napoli, 80125, Italy
| | - Giorgia Imparato
- Centre for Advanced Biomaterials for Health Care (IIT@CRIB), Istituto Italiano di Tecnologia, L.go Barsanti e Matteucci, Napoli, 80125, Italy
| | - Paolo Antonio Netti
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI) and Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Piazzale Tecchio 80, Napoli, 80125, Italy
- Centre for Advanced Biomaterials for Health Care (IIT@CRIB), Istituto Italiano di Tecnologia, L.go Barsanti e Matteucci, Napoli, 80125, Italy
| |
Collapse
|
30
|
Cui C, Wang X, Zheng Y, Wu L, Li L, Wei H, Peng J. Nur77 as a novel regulator of Paneth cell differentiation and function. Mucosal Immunol 2024; 17:752-767. [PMID: 37683828 DOI: 10.1016/j.mucimm.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/11/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023]
Abstract
Serving as a part of intestinal innate immunity, Paneth cells play an important role in intestinal homeostasis maintenance via their multiple functions. However, the regulation of Paneth cells has been proven to be complex and diverse. Here, we identified nuclear receptor Nur77 as a novel regulator of Paneth cell differentiation and function. Nur77 deficiency led to the loss of Paneth cells in murine ileal crypts. Intestinal tissues or organoids with Nur77 deficiency exhibited the impaired intestinal stem cell niche and failed to enhance antimicrobial peptide expression after Paneth cell degranulation. The defects in Paneth cells and antimicrobial peptides in Nur7-/- mice led to intestinal microbiota disorders. Nur77 deficiency rendered postnatal mice susceptible to necrotizing enterocolitis. Mechanistically, Nur77 transcriptionally inhibited Dact1 expression to activate Wnt signaling activity, thus promoting Paneth cell differentiation and function. Taken together, our data suggest the regulatory role of Nur77 in Paneth cell differentiation and function and reveal a novel Dact1-mediated Wnt inhibition mechanism in Paneth cell development.
Collapse
Affiliation(s)
- Chenbin Cui
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China; Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, China
| | - Xinru Wang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China; Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, China
| | - Yao Zheng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China; Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, China
| | - Lin Wu
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China; Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, China
| | - Lindeng Li
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China; Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, China
| | - Hongkui Wei
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China; Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, China
| | - Jian Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China; Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.
| |
Collapse
|
31
|
Li Y, Guo Y, Geng C, Song S, Yang W, Li X, Wang C. Vitamin D/vitamin D receptor protects intestinal barrier against colitis by positively regulating Notch pathway. Front Pharmacol 2024; 15:1421577. [PMID: 39130644 PMCID: PMC11310051 DOI: 10.3389/fphar.2024.1421577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/08/2024] [Indexed: 08/13/2024] Open
Abstract
Objective Vitamin D/Vitamin D receptor (VD/VDR) signaling and the Notch pathway are involved in intestinal barrier restoration in colitis; however, their relationship and underlying mechanism are largely unknown. Therefore, this study aimed to investigate the role and mechanism of VD/VDR and the Notch pathways in intestinal barrier protection. Methods Genetic Vdr knockout (VDR KO) and VD deficient (VDd) mice were established, and colitis was induced by feeding 2.5% dextran sodium sulfate (DSS) water. Mechanistic studies, including real-time PCR, immunofluorescence, Western blotting and dual-luciferase reporter assays, were performed on cultured Caco-2 cells and intestinal organoids. Results VD deficiency and VDR genetical KO increased the severity of DSS-induced colitis in mice, which presented a higher disease activity index score, increased intestinal permeability, and more severe intestinal histological damage than controls, accompanied by decreased and disrupted claudin-1 and claudin-3. Moreover, inhibition of Notch pathway by LY411,575 aggravated the severity of DSS-induced colitis and intestinal injury. In Caco-2 cells and intestinal organoids, the expression of Notch-1, N1ICD and Hes1 decreased upon downregulation or KO of VDR but increased upon paricalcitol (PAR, a VDR agonist) treatment. Meanwhile, PAR rescued claudin-1 and claudin-3 impairments that resulted from TNF-α exposure but failed to restore claudin-3 upon Notch inhibition. The dual-luciferase reporter assay further suggested that VD/VDR positively regulated the Notch signaling pathway by modulating Notch-1 transcription. Conclusion VD/VDR positively modulates Notch activation by promoting Notch-1 transcription to maintain intestinal tight junction integrity and barrier function. This highlights the VD/VDR-Notch pathway as a potential new therapeutic target for protecting the intestinal barrier against ulcerative colitis.
Collapse
Affiliation(s)
- Yanni Li
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, China
| | - Yaoyu Guo
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, China
| | - Chong Geng
- Laboratory of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
| | - Shuailing Song
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, China
| | - Wenjuan Yang
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, China
| | - Xiao Li
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, China
| | - Chunhui Wang
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
32
|
van Luyk ME, Krotenberg Garcia A, Lamprou M, Suijkerbuijk SJE. Cell competition in primary and metastatic colorectal cancer. Oncogenesis 2024; 13:28. [PMID: 39060237 PMCID: PMC11282291 DOI: 10.1038/s41389-024-00530-5] [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: 01/19/2024] [Revised: 07/05/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Adult tissues set the scene for a continuous battle between cells, where a comparison of cellular fitness results in the elimination of weaker "loser" cells. This phenomenon, named cell competition, is beneficial for tissue integrity and homeostasis. In fact, cell competition plays a crucial role in tumor suppression, through elimination of early malignant cells, as part of Epithelial Defense Against Cancer. However, it is increasingly apparent that cell competition doubles as a tumor-promoting mechanism. The comparative nature of cell competition means that mutational background, proliferation rate and polarity all factor in to determine the outcome of these processes. In this review, we explore the intricate and context-dependent involvement of cell competition in homeostasis and regeneration, as well as during initiation and progression of primary and metastasized colorectal cancer. We provide a comprehensive overview of molecular and cellular mechanisms governing cell competition and its parallels with regeneration.
Collapse
Affiliation(s)
- Merel Elise van Luyk
- Division of Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Ana Krotenberg Garcia
- Division of Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Maria Lamprou
- Division of Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Saskia Jacoba Elisabeth Suijkerbuijk
- Division of Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
33
|
Zutshi N, Mohapatra BC, Mondal P, An W, Goetz BT, Wang S, Li S, Storck MD, Mercer DF, Black AR, Thayer SP, Black JD, Lin C, Band V, Band H. Cbl and Cbl-b ubiquitin ligases are essential for intestinal epithelial stem cell maintenance. iScience 2024; 27:109912. [PMID: 38974465 PMCID: PMC11225835 DOI: 10.1016/j.isci.2024.109912] [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: 05/23/2023] [Revised: 02/29/2024] [Accepted: 05/03/2024] [Indexed: 07/09/2024] Open
Abstract
Receptor tyrosine kinases (RTKs) control stem cell maintenance vs. differentiation decisions. Casitas B-lineage lymphoma (CBL) family ubiquitin ligases are negative regulators of RTKs, but their stem cell regulatory roles remain unclear. Here, we show that Lgr5+ intestinal stem cell (ISC)-specific inducible Cbl-knockout (KO) on a Cblb null mouse background (iDKO) induced rapid loss of the Lgr5 Hi ISCs with transient expansion of the Lgr5 Lo transit-amplifying population. LacZ-based lineage tracing revealed increased ISC commitment toward enterocyte and goblet cell fate at the expense of Paneth cells. Functionally, Cbl/Cblb iDKO impaired the recovery from radiation-induced intestinal epithelial injury. In vitro, Cbl/Cblb iDKO led to inability to maintain intestinal organoids. Single-cell RNA sequencing in organoids identified Akt-mTOR (mammalian target of rapamycin) pathway hyperactivation upon iDKO, and pharmacological Akt-mTOR axis inhibition rescued the iDKO defects. Our results demonstrate a requirement for Cbl/Cblb in the maintenance of ISCs by fine-tuning the Akt-mTOR axis to balance stem cell maintenance vs. commitment to differentiation.
Collapse
Affiliation(s)
- Neha Zutshi
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Pathology & Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Bhopal C. Mohapatra
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Genetics, Cell Biology & Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Pinaki Mondal
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Wei An
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Benjamin T. Goetz
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Shuo Wang
- Department of Radiation Oncology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Sicong Li
- Department of Radiation Oncology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Matthew D. Storck
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - David F. Mercer
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Adrian R. Black
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Sarah P. Thayer
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jennifer D. Black
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Chi Lin
- Department of Radiation Oncology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Vimla Band
- Department of Genetics, Cell Biology & Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Hamid Band
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Pathology & Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Genetics, Cell Biology & Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
34
|
Taracena-Agarwal ML, Walter-Nuno AB, Bottino-Rojas V, Mejia APG, Xu K, Segal S, Dotson EM, Oliveira PL, Paiva-Silva GO. Juvenile Hormone as a contributing factor in establishing midgut microbiota for fecundity and fitness enhancement in adult female Aedes aegypti. Commun Biol 2024; 7:687. [PMID: 38839829 PMCID: PMC11153597 DOI: 10.1038/s42003-024-06334-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: 07/13/2023] [Accepted: 05/15/2024] [Indexed: 06/07/2024] Open
Abstract
Understanding the factors influencing mosquitoes' fecundity and longevity is important for designing better and more sustainable vector control strategies, as these parameters can impact their vectorial capacity. Here, we address how mating affects midgut growth in Aedes aegypti, what role Juvenile Hormone (JH) plays in this process, and how it impacts the mosquito's immune response and microbiota. Our findings reveal that mating and JH induce midgut growth. Additionally, the establishment of a native bacterial population in the midgut due to JH-dependent suppression of the immune response has important reproductive outcomes. Specific downregulation of AMPs with an increase in bacteria abundance in the gut results in increased egg counts and longer lifespans. Overall, these findings provide evidence of a cross-talk between JH response, gut epithelial tissue, cell cycle regulation, and the mechanisms governing the trade-offs between nutrition, immunity, and reproduction at the cellular level in the mosquito gut.
Collapse
Affiliation(s)
- Mabel L Taracena-Agarwal
- Programa de Biologia Molecular e Biotecnologia, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil.
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brasil.
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA.
- Entomology Department, Cornell University, College of Agriculture and Life Sciences, Ithaca, NY, USA.
| | - Ana Beatriz Walter-Nuno
- Programa de Biologia Molecular e Biotecnologia, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brasil
| | - Vanessa Bottino-Rojas
- Programa de Biologia Molecular e Biotecnologia, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brasil
| | | | - Kelsey Xu
- Entomology Department, Cornell University, College of Agriculture and Life Sciences, Ithaca, NY, USA
| | - Steven Segal
- Entomology Department, Cornell University, College of Agriculture and Life Sciences, Ithaca, NY, USA
| | - Ellen M Dotson
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Pedro L Oliveira
- Programa de Biologia Molecular e Biotecnologia, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brasil
| | - Gabriela O Paiva-Silva
- Programa de Biologia Molecular e Biotecnologia, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil.
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brasil.
| |
Collapse
|
35
|
Novak S, Tanigawa H, Singh V, Root SH, Schmidt TA, Hankenson KD, Kalajzic I. Endothelial to mesenchymal Notch signaling regulates skeletal repair. JCI Insight 2024; 9:e181073. [PMID: 38781018 PMCID: PMC11383173 DOI: 10.1172/jci.insight.181073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
We present a transcriptomic analysis that provides a better understanding of regulatory mechanisms within the healthy and injured periosteum. The focus of this work is on characterizing early events controlling bone healing during formation of periosteal callus on day 3 after fracture. Building on our previous findings showing that induced Notch1 signaling in osteoprogenitors leads to better healing, we compared samples in which the Notch 1 intracellular domain is overexpressed by periosteal stem/progenitor cells, with control intact and fractured periosteum. Molecular mechanisms and changes in skeletal stem/progenitor cells (SSPCs) and other cell populations within the callus, including hematopoietic lineages, were determined. Notably, Notch ligands were differentially expressed in endothelial and mesenchymal populations, with Dll4 restricted to endothelial cells, whereas Jag1 was expressed by mesenchymal populations. Targeted deletion of Dll4 in endothelial cells using Cdh5CreER resulted in negative effects on early fracture healing, while deletion in SSPCs using α-smooth muscle actin-CreER did not impact bone healing. Translating these observations into a clinically relevant model of bone healing revealed the beneficial effects of delivering Notch ligands alongside the osteogenic inducer, BMP2. These findings provide insights into the regulatory mechanisms within the healthy and injured periosteum, paving the way for novel translational approaches to bone healing.
Collapse
Affiliation(s)
- Sanja Novak
- Center for Regenerative Medicine and Skeletal Development, School of Dental Medicine, UConn Health, Farmington, Connecticut, USA
| | - Hitoshi Tanigawa
- Center for Regenerative Medicine and Skeletal Development, School of Dental Medicine, UConn Health, Farmington, Connecticut, USA
| | - Vijender Singh
- Institute for Systems Genomics, Computational Biology Core, UConn, Storrs, Connecticut, USA
| | - Sierra H Root
- Center for Regenerative Medicine and Skeletal Development, School of Dental Medicine, UConn Health, Farmington, Connecticut, USA
| | - Tannin A Schmidt
- Biomedical Engineering Department, School of Dental Medicine, UConn Health, Farmington, Connecticut, USA
| | - Kurt D Hankenson
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Ivo Kalajzic
- Center for Regenerative Medicine and Skeletal Development, School of Dental Medicine, UConn Health, Farmington, Connecticut, USA
| |
Collapse
|
36
|
Liang Y, Luo C, Sun L, Feng T, Yin W, Zhang Y, Mulholland MW, Zhang W, Yin Y. Reduction of specific enterocytes from loss of intestinal LGR4 improves lipid metabolism in mice. Nat Commun 2024; 15:4393. [PMID: 38782937 PMCID: PMC11116434 DOI: 10.1038/s41467-024-48622-5] [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: 01/26/2023] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
Whether intestinal Leucine-rich repeat containing G-protein-coupled receptor 4 (LGR4) impacts nutrition absorption and energy homeostasis remains unknown. Here, we report that deficiency of Lgr4 (Lgr4iKO) in intestinal epithelium decreased the proportion of enterocytes selective for long-chain fatty acid absorption, leading to reduction in lipid absorption and subsequent improvement in lipid and glucose metabolism. Single-cell RNA sequencing demonstrates the heterogeneity of absorptive enterocytes, with a decrease in enterocytes selective for long-chain fatty acid-absorption and an increase in enterocytes selective for carbohydrate absorption in Lgr4iKO mice. Activation of Notch signaling and concurrent inhibition of Wnt signaling are observed in the transgenes. Associated with these alterations is the substantial reduction in lipid absorption. Decrement in lipid absorption renders Lgr4iKO mice resistant to high fat diet-induced obesity relevant to wild type littermates. Our study thus suggests that targeting intestinal LGR4 is a potential strategy for the intervention of obesity and liver steatosis.
Collapse
Affiliation(s)
- Yuan Liang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, 100191, Beijing, China
| | - Chao Luo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, 100191, Beijing, China
| | - Lijun Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, 100191, Beijing, China
| | - Tiange Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, 100191, Beijing, China
| | - Wenzhen Yin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, 100191, Beijing, China
| | - Yunhua Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, 100191, Beijing, China
| | - Michael W Mulholland
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, 48109-0346, USA
| | - Weizhen Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, 100191, Beijing, China.
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, 48109-0346, USA.
| | - Yue Yin
- Department of Pharmacology, School of Basic Medical Sciences, and State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, 100191, Beijing, China.
| |
Collapse
|
37
|
Naser AN, Xing T, Tatum R, Lu Q, Boyer PJ, Chen YH. Colonic crypt stem cell functions are controlled by tight junction protein claudin-7 through Notch/Hippo signaling. Ann N Y Acad Sci 2024; 1535:92-108. [PMID: 38598500 PMCID: PMC11111361 DOI: 10.1111/nyas.15137] [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: 07/13/2023] [Revised: 02/26/2024] [Accepted: 03/13/2024] [Indexed: 04/12/2024]
Abstract
The tight junction protein claudin-7 is essential for tight junction function and intestinal homeostasis. Cldn7 deletion in mice leads to an inflammatory bowel disease-like phenotype exhibiting severe intestinal epithelial damage, weight loss, inflammation, mucosal ulcerations, and epithelial hyperplasia. Claudin-7 has also been shown to be involved in cancer metastasis and invasion. Here, we test our hypothesis that claudin-7 plays an important role in regulating colonic intestinal stem cell function. Conditional knockout of Cldn7 in the colon led to impaired epithelial cell differentiation, hyperproliferative epithelium, a decrease in active stem cells, and dramatically altered gene expression profiles. In 3D colonoid culture, claudin-7-deficient crypts were unable to survive and form spheroids, emphasizing the importance of claudin-7 in stem cell survival. Inhibition of the Hippo pathway or activation of Notch signaling partially rescued the defective stem cell behavior. Concurrent Notch activation and Hippo inhibition resulted in restored colonoid survival, growth, and differentiation to the level comparable to those of wild-type derived crypts. In this study, we highlight the essential role of claudin-7 in regulating Notch and Hippo signaling-dependent colonic stem cell functions, including survival, self-renewal, and differentiation. These new findings may shed light on potential avenues to explore for drug development in colorectal cancer.
Collapse
Affiliation(s)
- Amna N. Naser
- Department of Anatomy and Cell Biology, University of South Carolina, Columbia, South Carolina, USA
| | - Tiaosi Xing
- Department of Anatomy and Cell Biology, University of South Carolina, Columbia, South Carolina, USA
- Neural and Behavioral Science Department, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Rodney Tatum
- Department of Anatomy and Cell Biology, University of South Carolina, Columbia, South Carolina, USA
| | - Qun Lu
- Department of Anatomy and Cell Biology, University of South Carolina, Columbia, South Carolina, USA
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina, USA
| | - Philip J. Boyer
- Department of Pathology and Laboratory Medicine, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | - Yan-Hua Chen
- Department of Anatomy and Cell Biology, University of South Carolina, Columbia, South Carolina, USA
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina, USA
| |
Collapse
|
38
|
Bustamante-Madrid P, Barbáchano A, Albandea-Rodríguez D, Rodríguez-Cobos J, Rodríguez-Salas N, Prieto I, Burgos A, Martínez de Villarreal J, Real FX, González-Sancho JM, Larriba MJ, Lafarga M, Muñoz A, Fernández-Barral A. Vitamin D opposes multilineage cell differentiation induced by Notch inhibition and BMP4 pathway activation in human colon organoids. Cell Death Dis 2024; 15:301. [PMID: 38684650 PMCID: PMC11058856 DOI: 10.1038/s41419-024-06680-z] [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: 12/11/2023] [Revised: 04/08/2024] [Accepted: 04/12/2024] [Indexed: 05/02/2024]
Abstract
Understanding the mechanisms involved in colonic epithelial differentiation is key to unraveling the alterations causing inflammatory conditions and cancer. Organoid cultures provide an unique tool to address these questions but studies are scarce. We report a differentiation system toward enterocytes and goblet cells, the two major colonic epithelial cell lineages, using colon organoids generated from healthy tissue of colorectal cancer patients. Culture of these organoids in medium lacking stemness agents resulted in a modest ultrastructural differentiation phenotype with low-level expression of enterocyte (KLF4, KRT20, CA1, FABP2) and goblet cell (TFF2, TFF3, AGR2) lineage markers. BMP pathway activation through depletion of Noggin and addition of BMP4 resulted in enterocyte-biased differentiation. Contrarily, blockade of the Notch pathway using the γ-secretase inhibitor dibenzazepine (DBZ) favored goblet cell differentiation. Combination treatment with BMP4 and DBZ caused a balanced strong induction of both lineages. In contrast, colon tumor organoids responded poorly to BMP4 showing only weak signals of cell differentiation, and were unresponsive to DBZ. We also investigated the effects of 1α,25-dihydroxyvitamin D3 (calcitriol) on differentiation. Calcitriol attenuated the effects of BMP4 and DBZ on colon normal organoids, with reduced expression of differentiation genes and phenotype. Consistently, in normal organoids, calcitriol inhibited early signaling by BMP4 as assessed by reduction of the level of phospho-SMAD1/5/8. Our results show that BMP and Notch signaling play key roles in human colon stem cell differentiation to the enterocytic and goblet cell lineages and that calcitriol modulates these processes favoring stemness features.
Collapse
Affiliation(s)
- Pilar Bustamante-Madrid
- Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM, 28029, Madrid, Spain
- Centro de Investigación Biomédica en Red-Cáncer (CIBERONC), 28029, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Universitario La Paz (IdiPAZ), 28046, Madrid, Spain
| | - Antonio Barbáchano
- Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM, 28029, Madrid, Spain
- Centro de Investigación Biomédica en Red-Cáncer (CIBERONC), 28029, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Universitario La Paz (IdiPAZ), 28046, Madrid, Spain
| | - David Albandea-Rodríguez
- Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM, 28029, Madrid, Spain
- Centro de Investigación Biomédica en Red-Cáncer (CIBERONC), 28029, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Universitario La Paz (IdiPAZ), 28046, Madrid, Spain
| | - Javier Rodríguez-Cobos
- Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM, 28029, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Universitario La Paz (IdiPAZ), 28046, Madrid, Spain
| | - Nuria Rodríguez-Salas
- Centro de Investigación Biomédica en Red-Cáncer (CIBERONC), 28029, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Universitario La Paz (IdiPAZ), 28046, Madrid, Spain
- Servicio de Oncología Médica, Hospital Universitario La Paz, 28046, Madrid, Spain
| | - Isabel Prieto
- Instituto de Investigación Sanitaria Hospital Universitario La Paz (IdiPAZ), 28046, Madrid, Spain
- Servicio de Cirugía General, Hospital Universitario La Paz, 28046, Madrid, Spain
| | - Aurora Burgos
- Servicio de Digestivo, Hospital Universitario La Paz, 28046, Madrid, Spain
| | - Jaime Martínez de Villarreal
- Centro de Investigación Biomédica en Red-Cáncer (CIBERONC), 28029, Madrid, Spain
- Centro Nacional de Investigaciones Oncológicas (CNIO), 28029, Madrid, Spain
| | - Francisco X Real
- Centro de Investigación Biomédica en Red-Cáncer (CIBERONC), 28029, Madrid, Spain
- Centro Nacional de Investigaciones Oncológicas (CNIO), 28029, Madrid, Spain
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003, Barcelona, Spain
| | - José Manuel González-Sancho
- Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM, 28029, Madrid, Spain
- Centro de Investigación Biomédica en Red-Cáncer (CIBERONC), 28029, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Universitario La Paz (IdiPAZ), 28046, Madrid, Spain
| | - María Jesús Larriba
- Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM, 28029, Madrid, Spain
- Centro de Investigación Biomédica en Red-Cáncer (CIBERONC), 28029, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Universitario La Paz (IdiPAZ), 28046, Madrid, Spain
| | - Miguel Lafarga
- Departamento de Anatomía y Biología Celular, Universidad de Cantabria-IDIVAL, 39008, Santander, Spain
| | - Alberto Muñoz
- Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM, 28029, Madrid, Spain.
- Centro de Investigación Biomédica en Red-Cáncer (CIBERONC), 28029, Madrid, Spain.
- Instituto de Investigación Sanitaria Hospital Universitario La Paz (IdiPAZ), 28046, Madrid, Spain.
| | - Asunción Fernández-Barral
- Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM, 28029, Madrid, Spain.
- Centro de Investigación Biomédica en Red-Cáncer (CIBERONC), 28029, Madrid, Spain.
- Instituto de Investigación Sanitaria Hospital Universitario La Paz (IdiPAZ), 28046, Madrid, Spain.
| |
Collapse
|
39
|
Xu Y, Mao S, Fan H, Wan J, Wang L, Zhang M, Zhu S, Yuan J, Lu Y, Wang Z, Yu B, Jiang Z, Huang Y. LINC MIR503HG Controls SC-β Cell Differentiation and Insulin Production by Targeting CDH1 and HES1. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305631. [PMID: 38243869 PMCID: PMC10987150 DOI: 10.1002/advs.202305631] [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: 08/12/2023] [Revised: 01/03/2024] [Indexed: 01/22/2024]
Abstract
Stem cell-derived pancreatic progenitors (SC-PPs), as an unlimited source of SC-derived β (SC-β) cells, offers a robust tool for diabetes treatment in stem cell-based transplantation, disease modeling, and drug screening. Whereas, PDX1+/NKX6.1+ PPs enhances the subsequent endocrine lineage specification and gives rise to glucose-responsive SC-β cells in vivo and in vitro. To identify the regulators that promote induction efficiency and cellular function maturation, single-cell RNA-sequencing is performed to decipher the transcriptional landscape during PPs differentiation. The comprehensive evaluation of functionality demonstrated that manipulating LINC MIR503HG using CRISPR in PP cell fate decision can improve insulin synthesis and secretion in mature SC-β cells, without effects on liver lineage specification. Importantly, transplantation of MIR503HG-/- SC-β cells in recipients significantly restored blood glucose homeostasis, accompanied by serum C-peptide release and an increase in body weight. Mechanistically, by releasing CtBP1 occupying the CDH1 and HES1 promoters, the decrease in MIR503HG expression levels provided an excellent extracellular niche and appropriate Notch signaling activation for PPs following differentiation. Furthermore, this exhibited higher crucial transcription factors and mature epithelial markers in CDH1High expressed clusters. Altogether, these findings highlighted MIR503HG as an essential and exclusive PP cell fate specification regulator with promising therapeutic potential for patients with diabetes.
Collapse
Affiliation(s)
- Yang Xu
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
- Center of Gallbladder DiseaseShanghai East HospitalInstitute of Gallstone DiseaseSchool of MedicineTongji UniversityShanghai200092China
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
| | - Susu Mao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of EducationNMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology ProductsCo‐innovation Center of NeuroregenerationNantong UniversityNantong226001China
| | - Haowen Fan
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
| | - Jian Wan
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
| | - Lin Wang
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
- Department of Graduate SchoolDalian Medical UniversityDalianLiaoning116000China
| | - Mingyu Zhang
- Department of Nuclear MedicineBeijing Friendship HospitalAffiliated to Capital Medical UniversityBeijing100050China
| | - Shajun Zhu
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
| | - Jin Yuan
- Department of Endocrinology and MetabolismAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
| | - Yuhua Lu
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
| | - Zhiwei Wang
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
| | - Bin Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of EducationNMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology ProductsCo‐innovation Center of NeuroregenerationNantong UniversityNantong226001China
| | - Zhaoyan Jiang
- Center of Gallbladder DiseaseShanghai East HospitalInstitute of Gallstone DiseaseSchool of MedicineTongji UniversityShanghai200092China
| | - Yan Huang
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of EducationNMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology ProductsCo‐innovation Center of NeuroregenerationNantong UniversityNantong226001China
| |
Collapse
|
40
|
Pordel S, McCloskey AP, Almahmeed W, Sahebkar A. The protective effects of statins in traumatic brain injury. Pharmacol Rep 2024; 76:235-250. [PMID: 38448729 DOI: 10.1007/s43440-024-00582-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 03/08/2024]
Abstract
Traumatic brain injury (TBI), often referred to as the "silent epidemic", is the most common cause of mortality and morbidity worldwide among all trauma-related injuries. It is associated with considerable personal, medical, and economic consequences. Although remarkable advances in therapeutic approaches have been made, current treatments and clinical management for TBI recovery still remain to be improved. One of the factors that may contribute to this gap is that existing therapies target only a single event or pathology. However, brain injury after TBI involves various pathological mechanisms, including inflammation, oxidative stress, blood-brain barrier (BBB) disruption, ionic disturbance, excitotoxicity, mitochondrial dysfunction, neuronal necrosis, and apoptosis. Statins have several beneficial pleiotropic effects (anti-excitotoxicity, anti-inflammatory, anti-oxidant, anti-thrombotic, immunomodulatory activity, endothelial and vasoactive properties) in addition to promoting angiogenesis, neurogenesis, and synaptogenesis in TBI. Supposedly, using agents such as statins that target numerous and diverse pathological mechanisms, may be more effective than a single-target approach in TBI management. The current review was undertaken to investigate and summarize the protective mechanisms of statins against TBI. The limitations of conducted studies and directions for future research on this potential therapeutic application of statins are also discussed.
Collapse
Affiliation(s)
- Safoora Pordel
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alice P McCloskey
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Wael Almahmeed
- Heart and Vascular Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
41
|
Bravo Iniguez A, Du M, Zhu MJ. α-Ketoglutarate for Preventing and Managing Intestinal Epithelial Dysfunction. Adv Nutr 2024; 15:100200. [PMID: 38438107 PMCID: PMC11016550 DOI: 10.1016/j.advnut.2024.100200] [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: 11/22/2023] [Revised: 02/16/2024] [Accepted: 02/29/2024] [Indexed: 03/06/2024] Open
Abstract
The epithelium lining the intestinal tract serves a multifaceted role. It plays a crucial role in nutrient absorption and immune regulation and also acts as a protective barrier, separating underlying tissues from the gut lumen content. Disruptions in the delicate balance of the gut epithelium trigger inflammatory responses, aggravate conditions such as inflammatory bowel disease, and potentially lead to more severe complications such as colorectal cancer. Maintaining intestinal epithelial homeostasis is vital for overall health, and there is growing interest in identifying nutraceuticals that can strengthen the intestinal epithelium. α-Ketoglutarate, a metabolite of the tricarboxylic acid cycle, displays a variety of bioactive effects, including functioning as an antioxidant, a necessary cofactor for epigenetic modification, and exerting anti-inflammatory effects. This article presents a comprehensive overview of studies investigating the potential of α-ketoglutarate supplementation in preventing dysfunction of the intestinal epithelium.
Collapse
Affiliation(s)
| | - Min Du
- Department of Animal Sciences, Washington State University, Pullman, WA, United States
| | - Mei-Jun Zhu
- School of Food Science, Washington State University, Pullman, WA, United States.
| |
Collapse
|
42
|
Robertson R, Li S, Filippelli RL, Chang NC. Muscle stem cell dysfunction in rhabdomyosarcoma and muscular dystrophy. Curr Top Dev Biol 2024; 158:83-121. [PMID: 38670717 DOI: 10.1016/bs.ctdb.2024.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Muscle stem cells (MuSCs) are crucial to the repair and homeostasis of mature skeletal muscle. MuSC dysfunction and dysregulation of the myogenic program can contribute to the development of pathology ranging from cancers like rhabdomyosarcoma (RMS) or muscle degenerative diseases such as Duchenne muscular dystrophy (DMD). Both diseases exhibit dysregulation at nearly all steps of myogenesis. For instance, MuSC self-renewal processes are altered. In RMS, this leads to the creation of tumor propagating cells. In DMD, impaired asymmetric stem cell division creates a bias towards producing self-renewing stem cells instead of committing to differentiation. Hyperproliferation of these cells contribute to tumorigenesis in RMS and symmetric expansion of the self-renewing MuSC population in DMD. Both diseases also exhibit a repression of factors involved in terminal differentiation, halting RMS cells in the proliferative stage and thus driving tumor growth. Conversely, the MuSCs in DMD exhibit impaired differentiation and fuse prematurely, affecting myonuclei maturation and the integrity of the dystrophic muscle fiber. Finally, both disease states cause alterations to the MuSC niche. Various elements of the niche such as inflammatory and migratory signaling that impact MuSC behavior are dysregulated. Here we show how these seemingly distantly related diseases indeed have similarities in MuSC dysfunction, underlying the importance of considering MuSCs when studying the pathophysiology of muscle diseases.
Collapse
Affiliation(s)
- Rebecca Robertson
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada
| | - Shulei Li
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada; Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada
| | - Romina L Filippelli
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada
| | - Natasha C Chang
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada; Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada.
| |
Collapse
|
43
|
Del Vecchio A, Mulé P, Fernández-Pérez D, Amato S, Lattanzi G, Zanotti M, Rustichelli S, Pivetti S, Oldani P, Mariani A, Iommazzo F, Koseki H, Facciotti F, Tamburri S, Ferrari KJ, Pasini D. PCGF6 controls murine Tuft cell differentiation via H3K9me2 modification independently of Polycomb repression. Dev Cell 2024; 59:368-383.e7. [PMID: 38228142 DOI: 10.1016/j.devcel.2023.12.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 10/01/2023] [Accepted: 12/21/2023] [Indexed: 01/18/2024]
Abstract
Cell fate is determined by specific transcription programs that are essential for tissue homeostasis and regeneration. The E3-ligases RING1A and B represent the core activity of the Polycomb repressive complex 1 (PRC1) that deposits repressive histone H2AK119 mono-ubiquitination (H2AK119ub1), which is essential for mouse intestinal homeostasis by preserving stem cell functions. However, the specific role of different PRC1 forms, which are defined by the six distinct PCGF1-6 paralogs, remains largely unexplored in vivo. We report that PCGF6 regulates mouse intestinal Tuft cell differentiation independently of H2AK119ub1 deposition. We show that PCGF6 chromatin occupancy expands outside Polycomb repressive domains, associating with unique promoter and distal regulatory elements. This occurs in the absence of RING1A/B and involves MGA-mediated E-BOX recognition and specific H3K9me2 promoter deposition. PCGF6 inactivation induces an epithelial autonomous accumulation of Tuft cells that was not phenocopied by RING1A/B loss. This involves direct PCGF6 association with a Tuft cell differentiation program that identified Polycomb-independent properties of PCGF6 in adult tissues homeostasis.
Collapse
Affiliation(s)
- Annachiara Del Vecchio
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Patrizia Mulé
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Daniel Fernández-Pérez
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Simona Amato
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Georgia Lattanzi
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Marika Zanotti
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Samantha Rustichelli
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Silvia Pivetti
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Paola Oldani
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Andrea Mariani
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Fabiola Iommazzo
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Haruhiko Koseki
- RIKEN Centre for Integrative Medical Sciences, Laboratory for Developmental Genetics, 1-7-22 Suehiuro-cho, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Federica Facciotti
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy; University of Milano-Bicocca, Department of Biotechnology and Biosciences, Piazza della Scienza, 2, 20126 Milan, Italy
| | - Simone Tamburri
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy; University of Milan, Department of Health Sciences, Via A. di Rudinì 8, 20142 Milan, Italy
| | - Karin J Ferrari
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Diego Pasini
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Via Adamello 16, 20139 Milan, Italy; University of Milan, Department of Health Sciences, Via A. di Rudinì 8, 20142 Milan, Italy.
| |
Collapse
|
44
|
Ning H, Liu J, Tan J, Yi M, Lin X. The role of the Notch signalling pathway in the pathogenesis of ulcerative colitis: from the perspective of intestinal mucosal barrier. Front Med (Lausanne) 2024; 10:1333531. [PMID: 38249980 PMCID: PMC10796567 DOI: 10.3389/fmed.2023.1333531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 12/12/2023] [Indexed: 01/23/2024] Open
Abstract
Ulcerative colitis is a common digestive disorder worldwide, with increasing incidence in recent years. It is an urgent problem to be solved, as it seriously affects and threatens the health and life of the global population. Studies have shown that dysfunction of the intestinal mucosal barrier is a critical pathogenic factor and molecular basis of ulcerative colitis, and some scholars have described it as a "barrier organ disease." While the Notch signalling pathway affects a series of cellular processes, including proliferation, differentiation, development, migration, and apoptosis. Therefore, it can regulate intestinal stem cells, CD4+ T cells, innate lymphoid cells, macrophages, and intestinal microbiota and intervene in the chemical, physical, immune, and biological mucosal barriers in cases of ulcerative colitis. The Notch signalling pathway associated with the pathogenesis of ulcerative colitis has distinct characteristics, with good regulatory effects on the mucosal barrier. However, research on ulcerative colitis has mainly focused on immune regulation, anti-inflammatory activity, and antioxidant stress; therefore, the study of the Notch signalling pathway suggests the possibility of understanding the pathogenesis of ulcerative colitis from another perspective. In this article we explore the role and mechanism of the Notch signalling pathway in the pathogenesis of ulcerative colitis from the perspective of the intestinal mucosal barrier to provide new targets and theoretical support for further research on the pathogenesis and clinical treatment of ulcerative colitis.
Collapse
Affiliation(s)
- Hang Ning
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Jiemin Liu
- Guizhou Provincial People’s Hospital, Guiyang, China
| | - Jiaqian Tan
- Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Mengni Yi
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Xiaoyuan Lin
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
45
|
Li C, Sun Y, He T, Lu Y, Szeto IMY, Duan S, Zhang Y, Liu B, Zhang Y, Zhang W, He J, Li Y. Synergistic effect of lactoferrin and osteopontin on intestinal barrier injury. Int J Biol Macromol 2023; 253:127416. [PMID: 37838132 DOI: 10.1016/j.ijbiomac.2023.127416] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 10/10/2023] [Accepted: 10/10/2023] [Indexed: 10/16/2023]
Abstract
Several studies indicate that the disruption of the intestinal epithelial barrier can lead to inflammatory bowel disease (IBD). Recent evidence has increasingly demonstrated that lactoferrin (LF) and osteopontin (OPN) can alleviate intestinal barrier injury. However, the potential synergistic effects of these two proteins and the mechanisms underlying their effects remain unclear. To address this question, we developed a lipopolysaccharide-induced intestinal barrier injury model in C57BL/6 N mice. Our findings demonstrated that the combination of LF and OPN at a 1:5 ratio exerts the strongest protective effect on the intestinal barrier, and it is more effective than LF or OPN alone. This protection is evidenced by the decrease in serum diamine oxidase (DAO) activity (1.66-fold decrease) and D-lactic content (1.51-fold decrease) and the reduced rate of FITC-labeled glucan transport across the jejunum (3.18-fold decrease). Moreover, the protein combination significantly promoted villi length (1.66-fold increase) and crypt depth (1.57-fold increase), improved tight junction protein structure and expression, and boosted the number of absorptive cells (4.34-fold increase) in the intestinal epithelium. Furthermore, the combination promoted crypt cell proliferation and differentiation via Notch signaling. In summary, our findings provide scientific evidence supporting the use of dietary intervention strategies for preventing IBD.
Collapse
Affiliation(s)
- Chuangang Li
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Functional Daily, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Yanan Sun
- Key Laboratory of Functional Daily, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Tingchao He
- Inner Mongolia Dairy Technology Research Institute Co. Ltd., Hohhot 010110, China; Inner Mongolia Yili Industrial Group, Co. Ltd., Yili Maternal and Infant Nutrition Institute (YMINI), Beijing 100070, China
| | - Yao Lu
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Ignatius Man-Yau Szeto
- Inner Mongolia Yili Industrial Group, Co. Ltd., Yili Maternal and Infant Nutrition Institute (YMINI), Beijing 100070, China; National Center of Technology Innovation for Dairy, Hohhot 010110, China
| | - Sufang Duan
- Inner Mongolia Dairy Technology Research Institute Co. Ltd., Hohhot 010110, China; Inner Mongolia Yili Industrial Group, Co. Ltd., Yili Maternal and Infant Nutrition Institute (YMINI), Beijing 100070, China
| | - Yifan Zhang
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Biao Liu
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; Inner Mongolia Yili Industrial Group, Co. Ltd., Yili Maternal and Infant Nutrition Institute (YMINI), Beijing 100070, China
| | - Yiran Zhang
- Key Laboratory of Functional Daily, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Wen Zhang
- Key Laboratory of Functional Daily, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Jian He
- National Center of Technology Innovation for Dairy, Hohhot 010110, China
| | - Yixuan Li
- Key Laboratory of Functional Daily, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
46
|
Parham LR, Williams PA, Katada K, Nettleford SK, Chatterji P, Acheampong KK, Danan CH, Ma X, Simon LA, Naughton KE, Mizuno R, Karakasheva T, McMillan EA, Whelan KA, Brady DC, Shaffer SM, Hamilton KE. IGF2BP1/IMP1 Deletion Enhances a Facultative Stem Cell State via Regulation of MAP1LC3B. Cell Mol Gastroenterol Hepatol 2023; 17:439-451. [PMID: 38081361 PMCID: PMC10835461 DOI: 10.1016/j.jcmgh.2023.12.001] [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: 10/14/2022] [Revised: 11/30/2023] [Accepted: 12/04/2023] [Indexed: 01/02/2024]
Abstract
BACKGROUND & AIMS The intestinal epithelium interfaces with a diverse milieu of luminal contents while maintaining robust digestive and barrier functions. Facultative intestinal stem cells are cells that survive tissue injury and divide to re-establish the epithelium. Prior studies have shown autophagic state as functional marker of facultative intestinal stem cells, but regulatory mechanisms are not known. The current study evaluated a post-transcriptional regulation of autophagy as an important factor for facultative stem cell state and tissue regeneration. METHODS We evaluated stem cell composition, autophagic vesicle content, organoid formation, and in vivo regeneration in mice with intestinal epithelial deletion of the RNA binding protein IGF2 messenger RNA binding protein 1 (IMP1). The contribution of autophagy to resulting in vitro and in vivo phenotypes was evaluated via genetic inactivation of Atg7. Molecular analyses of IMP1 modulation of autophagy at the protein and transcript localization levels were performed using IMP1 mutant studies and single-molecule fluorescent in situ hybridization. RESULTS Epithelial Imp1 deletion reduced leucine rich repeat containing G protein coupled receptor 5 cell frequency but enhanced both organoid formation efficiency and in vivo regeneration after irradiation. We confirmed prior studies showing increased autophagy with IMP1 deletion. Deletion of Atg7 reversed the enhanced regeneration observed with Imp1 deletion. IMP1 deletion or mutation of IMP1 phosphorylation sites enhanced expression of essential autophagy protein microtubule-associated protein 1 light chain 3β. Furthermore, immunofluorescence imaging coupled with single-molecule fluorescent in situ hybridization showed IMP1 colocalization with MAP1LC3B transcripts at homeostasis. Stress induction led to decreased colocalization. CONCLUSIONS Depletion of IMP1 enhances autophagy, which promotes intestinal regeneration via expansion of facultative intestinal stem cells.
Collapse
Affiliation(s)
- Louis R Parham
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Patrick A Williams
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Kay Katada
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Shaneice K Nettleford
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Priya Chatterji
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Kofi K Acheampong
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Charles H Danan
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Xianghui Ma
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Lauren A Simon
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Kaitlyn E Naughton
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Rei Mizuno
- Department of Surgery, Uji-Tokushukai Medical Center, Uji, Kyoto, Japan
| | - Tatiana Karakasheva
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Emily A McMillan
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Kelly A Whelan
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania; Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Donita C Brady
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sydney M Shaffer
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania; Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kathryn E Hamilton
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania.
| |
Collapse
|
47
|
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
|
48
|
Schumacher MA. The emerging roles of deep crypt secretory cells in colonic physiology. Am J Physiol Gastrointest Liver Physiol 2023; 325:G493-G500. [PMID: 37697924 PMCID: PMC10887841 DOI: 10.1152/ajpgi.00093.2023] [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: 05/02/2023] [Revised: 08/18/2023] [Accepted: 09/03/2023] [Indexed: 09/13/2023]
Abstract
Deep crypt secretory (DCS) cells are a population of epithelial cells located at the colonic crypt base that share some similarities to Paneth and goblet cells. They were initially defined as c-Kit expressing cells, though subsequent work showed that they are more specifically marked by Reg4 in the murine colon. The best-understood function of DCS cells at present is supporting the stem cell niche by generating Notch and EGF ligands. However, as these cells also express immunoregulatory (e.g., Ccl6) and host defense (e.g., Retnlb) genes, it is likely they have additional functions in maintaining colonic health outside of maintenance of the stem niche. Recent advances in single-cell transcriptomic profiling hint at additional epithelial and immune roles that may exist for these cells and have aided in elucidating their developmental lineage. This review highlights the emerging evidence supporting a crucial role for DCS cells in intestinal physiology, the current understanding of how these cells are regulated, and their potential role(s) in colonic disease.
Collapse
Affiliation(s)
- Michael A Schumacher
- Department of Pediatrics, University of Southern California Keck School of Medicine, Los Angeles, California, United States
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California, United States
| |
Collapse
|
49
|
Hibdon ES, Keeley TM, Merchant JL, Samuelson LC. The bHLH transcription factor ASCL1 promotes differentiation of endocrine cells in the stomach and is regulated by Notch signaling. Am J Physiol Gastrointest Liver Physiol 2023; 325:G458-G470. [PMID: 37698169 PMCID: PMC10887855 DOI: 10.1152/ajpgi.00043.2023] [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: 03/06/2023] [Revised: 09/05/2023] [Accepted: 09/05/2023] [Indexed: 09/13/2023]
Abstract
Notch signaling regulates gastrointestinal stem cell proliferation and differentiation yet Notch-regulated transcriptional effectors of gastric epithelial cell differentiation are poorly understood. Here we tested the role of the bHLH transcription factor Achaete-Scute homolog 1 (ASCL1) in gastric epithelial cell differentiation, and its regulation by Notch. Newborn Ascl1 null mice showed a loss of expression of markers of neurogenin-3-dependent enteroendocrine cells, with normal expression of enterochromaffin-like cells, mucous cells, chief cells, and parietal cells. In adult mice, Ascl1 gene expression was observed in the stomach, but not the intestine, with higher expression in antral than corpus epithelium. Lineage tracing in Ascl1-CreERT2; Rosa26-LSL-tdTomato mice revealed single, scattered ASCL1+ cells in the gastric epithelium, demonstrating expression in antral gastrin- and serotonin-producing endocrine cells. ASCL1-expressing endocrine cells persisted for several weeks posttamoxifen labeling with a half-life of approximately 2 months. Lineage tracing in Gastrin-CreERT2 mice demonstrated a similar lifespan for gastrin-producing cells, confirming that gastric endocrine cells are long-lived. Finally, treatment of Ascl1-CreERT2; Rosa26-LSL-tdTomato mice with the pan-Notch inhibitor dibenzazepine increased the number of lineage-labeled cells in the gastric antrum, suggesting that Notch signaling normally inhibits Ascl1 expression. Notch regulation of Ascl1 was also demonstrated in a genetic mouse model of Notch activation, as well as Notch-manipulated antral organoid cultures, thus suggesting that ASCL1 is a key downstream Notch pathway effector promoting endocrine cell differentiation in the gastric epithelium.NEW & NOTEWORTHY Although Notch signaling is known to regulate cellular differentiation in the stomach, downstream effectors are poorly described. Here we demonstrate that the bHLH transcription factor ASCL1 is expressed in endocrine cells in the stomach and is required for formation of neurogenin-3-dependent enteroendocrine cells but not enterochromaffin-like cells. We also demonstrate that Ascl1 expression is inhibited by Notch signaling, suggesting that ASCL1 is a Notch-regulated transcriptional effector directing enteroendocrine cell fate in the mouse stomach.
Collapse
Affiliation(s)
- Elise S Hibdon
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Theresa M Keeley
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Juanita L Merchant
- Department of Medicine, University of Arizona, Tucson, Arizona, United States
| | - Linda C Samuelson
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States
| |
Collapse
|
50
|
Boers R, Boers J, Tan B, van Leeuwen ME, Wassenaar E, Sanchez EG, Sleddens E, Tenhagen Y, Mulugeta E, Laven J, Creyghton M, Baarends W, van IJcken WFJ, Gribnau J. Retrospective analysis of enhancer activity and transcriptome history. Nat Biotechnol 2023; 41:1582-1592. [PMID: 36823354 PMCID: PMC10635829 DOI: 10.1038/s41587-023-01683-1] [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: 07/01/2021] [Accepted: 01/20/2023] [Indexed: 02/25/2023]
Abstract
Cell state changes in development and disease are controlled by gene regulatory networks, the dynamics of which are difficult to track in real time. In this study, we used an inducible DCM-RNA polymerase subunit b fusion protein which labels active genes and enhancers with a bacterial methylation mark that does not affect gene transcription and is propagated in S-phase. This DCM-RNA polymerase fusion protein enables transcribed genes and active enhancers to be tagged and then examined at later stages of development or differentiation. We apply this DCM-time machine (DCM-TM) technology to study intestinal homeostasis, revealing rapid and coordinated activation of enhancers and nearby genes during enterocyte differentiation. We provide new insights in absorptive-secretory lineage decision-making in intestinal stem cell (ISC) differentiation and show that ISCs retain a unique chromatin landscape required to maintain ISC identity and delineate future expression of differentiation-associated genes. DCM-TM has wide applicability in tracking cell states, providing new insights in the regulatory networks underlying cell state changes.
Collapse
Affiliation(s)
- Ruben Boers
- Department of Developmental Biology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Joachim Boers
- Department of Developmental Biology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Beatrice Tan
- Department of Developmental Biology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Marieke E van Leeuwen
- Department of Developmental Biology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Evelyne Wassenaar
- Department of Developmental Biology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Erlantz Gonzalez Sanchez
- Department of Developmental Biology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Esther Sleddens
- Department of Developmental Biology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Yasha Tenhagen
- Department of Developmental Biology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Eskeatnaf Mulugeta
- Department of Cell Biology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Joop Laven
- Department of Obstetrics and Gynaecology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Menno Creyghton
- Department of Developmental Biology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Willy Baarends
- Department of Developmental Biology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Wilfred F J van IJcken
- Erasmus Center for Biomics, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Joost Gribnau
- Department of Developmental Biology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands.
| |
Collapse
|