1
|
Daniele T, Cury J, Morin MC, Ahier A, Isaia D, Jarriault S. Essential and dual effects of Notch activity on a natural transdifferentiation event. Nat Commun 2025; 16:75. [PMID: 39746948 PMCID: PMC11697417 DOI: 10.1038/s41467-024-55286-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 12/06/2024] [Indexed: 01/04/2025] Open
Abstract
Cell identity can be reprogrammed, naturally or experimentally, albeit with low frequency. Why some cells, but not their neighbours, undergo a cell identity conversion remains unclear. We find that Notch signalling plays a key role to promote natural transdifferentiation in C. elegans hermaphrodites. Endogenous Notch signalling endows a cell with the competence to transdifferentiate by promoting plasticity factors expression (hlh-16/Olig and sem-4/Sall). Strikingly, ectopic Notch can trigger additional transdifferentiation in vivo. However, Notch signalling can both promote and block transdifferentiation depending on its activation timing. Notch only promotes transdifferentiation during an early precise window of opportunity and signal duration must be tightly controlled in time. Our findings emphasise the importance of temporality and dynamics of the underlying molecular events preceding the initiation of natural cell reprogramming. Finally, our results support a model where both an extrinsic signal and the intrinsic cellular context combine to empower a cell with the competence to transdifferentiate.
Collapse
Affiliation(s)
- Thomas Daniele
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR 7104, INSERM U1298, Université de Strasbourg, Illkirch, France
- Vertex Pharmaceuticals (CH) GmbH, Zug, Switzerland
| | - Jeanne Cury
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR 7104, INSERM U1298, Université de Strasbourg, Illkirch, France
| | - Marie-Charlotte Morin
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR 7104, INSERM U1298, Université de Strasbourg, Illkirch, France
| | - Arnaud Ahier
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR 7104, INSERM U1298, Université de Strasbourg, Illkirch, France
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Davide Isaia
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR 7104, INSERM U1298, Université de Strasbourg, Illkirch, France
- Skyhawk Therapeutics, Basel, Switzerland
| | - Sophie Jarriault
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR 7104, INSERM U1298, Université de Strasbourg, Illkirch, France.
| |
Collapse
|
2
|
Yu X, Li C, Tao Y, Xia T, Jia Z. Lacidipine Inhibits NF-κB and Notch Pathways and Mitigates DSS-Induced Colitis. Dig Dis Sci 2024; 69:3753-3759. [PMID: 39261383 DOI: 10.1007/s10620-024-08618-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 08/23/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Ulcerative colitis (UC) is a chronic inflammatory condition affecting the colon, with a global incidence that is rising. Despite the increasing prevalence, effective treatment options for UC remain limited. METHODS We utilized an NF-κB promoter dual fluorescence reporter system to screen for compounds that could inhibit p65 and IκBα phosphorylation. The anti-hypertension drug lacidipine was identified as a candidate. Its efficacy was further evaluated in a murine model of dextran sulfate sodium (DSS)-induced colitis. The analysis included the assessment of colon lesions, inflammation markers, and signal pathway activation, with a focus on NF-κB and Notch signaling. RESULTS Lacidipine effectively inhibited p65 and IκBα phosphorylation in the reporter system. In the DSS-induced colitis murine model, lacidipine treatment led to a reduction in colon lesions and inflammatory markers. Target analysis showed significant enrichment of the Notch signaling pathway. Additionally, lacidipine inhibited both NF-κB and Notch activation in DSS-stimulated colons. CONCLUSION Lacidipine demonstrated a protective effect in UC, reducing inflammation and modulating key signaling pathways. These findings suggest that lacidipine could be a promising candidate for the treatment of UC.
Collapse
Affiliation(s)
- Xuezhao Yu
- Department of Pharmacy, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430016, China
| | - Cheng Li
- Department of Rehabilitation Medicine, Wuhan Ninth Hospital, Wuhan, 430081, China
| | - Yu Tao
- Department of Pharmacy, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430016, China
| | - Tingting Xia
- Department of Gastroenterology and Digestive Diseases, The First Affiliated Hospital of Soochow University, 188 Shizi St., Suzhou, 215006, China
| | - Zhenyu Jia
- Department of Gastroenterology and Digestive Diseases, The First Affiliated Hospital of Soochow University, 188 Shizi St., Suzhou, 215006, China.
| |
Collapse
|
3
|
Sfeir N, Kajdan M, Jalaguier S, Bonnet S, Teyssier C, Pyrdziak S, Yuan R, Bousquet E, Maraver A, Bernex F, Pirot N, Boissière‐Michot F, Castet‐Nicolas A, Lapierre M, Cavaillès V. RIP140 regulates transcription factor HES1 oscillatory expression and mitogenic activity in colon cancer cells. Mol Oncol 2024; 18:1510-1530. [PMID: 38459621 PMCID: PMC11161732 DOI: 10.1002/1878-0261.13626] [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: 06/28/2023] [Revised: 01/17/2024] [Accepted: 02/23/2024] [Indexed: 03/10/2024] Open
Abstract
The transcription factor receptor-interacting protein 140 (RIP140) regulates intestinal homeostasis and tumorigenesis through Wnt signaling. In this study, we investigated its effect on the Notch/HES1 signaling pathway. In colorectal cancer (CRC) cell lines, RIP140 positively regulated HES1 gene expression at the transcriptional level via a recombining binding protein suppressor of hairless (RBPJ)/neurogenic locus notch homolog protein 1 (NICD)-mediated mechanism. In support of these in vitro data, RIP140 and HES1 expression significantly correlated in mouse intestine and in a cohort of CRC samples, thus supporting the positive regulation of HES1 gene expression by RIP140. Interestingly, when the Notch pathway is fully activated, RIP140 exerted a strong inhibition of HES1 gene transcription controlled by the level of HES1 itself. Moreover, RIP140 directly interacts with HES1 and reversed its mitogenic activity in human CRC cells. In line with this observation, HES1 levels were associated with a better patient survival only when tumors expressed high levels of RIP140. Our data identify RIP140 as a key regulator of the Notch/HES1 signaling pathway, with a dual effect on HES1 gene expression at the transcriptional level and a strong impact on colon cancer cell proliferation.
Collapse
Affiliation(s)
- Nour Sfeir
- IRCM, Institut de Recherche en Cancérologie de MontpellierFrance
- INSERM, U1194France
- Université de MontpellierFrance
- Institut régional du Cancer de MontpellierFrance
| | - Marilyn Kajdan
- IRCM, Institut de Recherche en Cancérologie de MontpellierFrance
- INSERM, U1194France
- Université de MontpellierFrance
- Institut régional du Cancer de MontpellierFrance
| | - Stéphan Jalaguier
- IRCM, Institut de Recherche en Cancérologie de MontpellierFrance
- INSERM, U1194France
- Université de MontpellierFrance
- Institut régional du Cancer de MontpellierFrance
| | - Sandrine Bonnet
- IRCM, Institut de Recherche en Cancérologie de MontpellierFrance
- INSERM, U1194France
- Université de MontpellierFrance
- Institut régional du Cancer de MontpellierFrance
| | - Catherine Teyssier
- IRCM, Institut de Recherche en Cancérologie de MontpellierFrance
- INSERM, U1194France
- Université de MontpellierFrance
- Institut régional du Cancer de MontpellierFrance
| | - Samuel Pyrdziak
- IRCM, Institut de Recherche en Cancérologie de MontpellierFrance
- INSERM, U1194France
- Université de MontpellierFrance
- Institut régional du Cancer de MontpellierFrance
| | - Rong Yuan
- Department of Medical Microbiology, Immunology and Cell Biology, School of MedicineSouthern Illinois UniversitySpringfieldILUSA
| | - Emilie Bousquet
- IRCM, Institut de Recherche en Cancérologie de MontpellierFrance
- INSERM, U1194France
- Université de MontpellierFrance
- Institut régional du Cancer de MontpellierFrance
| | - Antonio Maraver
- IRCM, Institut de Recherche en Cancérologie de MontpellierFrance
- INSERM, U1194France
- Université de MontpellierFrance
- Institut régional du Cancer de MontpellierFrance
| | - Florence Bernex
- IRCM, Institut de Recherche en Cancérologie de MontpellierFrance
- INSERM, U1194France
- Université de MontpellierFrance
- Institut régional du Cancer de MontpellierFrance
| | - Nelly Pirot
- IRCM, Institut de Recherche en Cancérologie de MontpellierFrance
- INSERM, U1194France
- Université de MontpellierFrance
- Institut régional du Cancer de MontpellierFrance
| | - Florence Boissière‐Michot
- IRCM, Institut de Recherche en Cancérologie de MontpellierFrance
- INSERM, U1194France
- Université de MontpellierFrance
- Institut régional du Cancer de MontpellierFrance
- Translational Research UnitMontpellier Cancer Institute Val d'AurelleFrance
| | - Audrey Castet‐Nicolas
- IRCM, Institut de Recherche en Cancérologie de MontpellierFrance
- INSERM, U1194France
- Université de MontpellierFrance
- Institut régional du Cancer de MontpellierFrance
| | - Marion Lapierre
- IRCM, Institut de Recherche en Cancérologie de MontpellierFrance
- INSERM, U1194France
- Université de MontpellierFrance
- Institut régional du Cancer de MontpellierFrance
| | - Vincent Cavaillès
- IRCM, Institut de Recherche en Cancérologie de MontpellierFrance
- INSERM, U1194France
- Université de MontpellierFrance
- Institut régional du Cancer de MontpellierFrance
| |
Collapse
|
4
|
Xiu M, Wang Y, Yang D, Zhang X, Dai Y, Liu Y, Lin X, Li B, He J. Using Drosophila melanogaster as a suitable platform for drug discovery from natural products in inflammatory bowel disease. Front Pharmacol 2022; 13:1072715. [PMID: 36545307 PMCID: PMC9760693 DOI: 10.3389/fphar.2022.1072715] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/24/2022] [Indexed: 12/07/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic and life-treating inflammatory disease that can occur in multiple parts of the human intestine and has become a worldwide problem with a continually increasing incidence. Because of its mild early symptoms, most of them will not attract people's attention and may cause more serious consequences. There is an urgent need for new therapeutics to prevent disease progression. Natural products have a variety of active ingredients, diverse biological activities, and low toxicity or side effects, which are the new options for preventing and treating the intestinal inflammatory diseases. Because of multiple genetic models, less ethical concerns, conserved signaling pathways with mammals, and low maintenance costs, the fruit fly Drosophila melanogaster has become a suitable model for studying mechanism and treatment strategy of IBD. Here, we review the advantages of fly model as screening platform in drug discovery, describe the conserved molecular pathways as therapetic targets for IBD between mammals and flies, dissect the feasibility of Drosophila model in IBD research, and summarize the natural products for IBD treatment using flies. This review comprehensively elaborates that the benefit of flies as a perfact model to evaluate the therapeutic potential of phytochemicals against IBD.
Collapse
Affiliation(s)
- Minghui Xiu
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, China,Provincial-level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China,Key Laboratory of Dunhuang Medicine, Ministry of Education, Lanzhou, China
| | - Yixuan Wang
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
| | - Dan Yang
- College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Xueyan Zhang
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yuting Dai
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yongqi Liu
- Provincial-level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China,Key Laboratory of Dunhuang Medicine, Ministry of Education, Lanzhou, China
| | - Xingyao Lin
- Key Laboratory of Dunhuang Medicine, Ministry of Education, Lanzhou, China
| | - Botong Li
- College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Jianzheng He
- Provincial-level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China,Key Laboratory of Dunhuang Medicine, Ministry of Education, Lanzhou, China,College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China,*Correspondence: Jianzheng He,
| |
Collapse
|
5
|
Bacillus subtilis programs the differentiation of intestinal secretory lineages to inhibit Salmonella infection. Cell Rep 2022; 40:111416. [PMID: 36170821 DOI: 10.1016/j.celrep.2022.111416] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 08/17/2022] [Accepted: 09/01/2022] [Indexed: 11/20/2022] Open
Abstract
The role of intestinal microbiota on fate determination of intestinal epithelial cells has not been extensively examined. In this study, we explore the effect of Bacillus subtilis on programmed intestinal epithelial differentiation. We find that B. subtilis stimulates the differentiation of intestinal secretory cells. Moreover, B. subtilis inhibits the Notch pathway to reduce the expression of hairy and enhancer of split 1, thereby shifting intestinal stem cell differentiation toward a secretory cell fate. Moreover, we demonstrate that the programming effect of B. subtilis on intestinal differentiation is Toll-like receptor 2 pathway dependent. B. subtilis is associated with increased numbers of Paneth and goblet cells in the intestine. This results in the production of antimicrobial peptides to protect the intestinal mucosal barrier against Salmonella typhimurium. This study demonstrates that B. subtilis contributes to the differentiation of secretory cells by affecting Notch pathway signaling to maintain the intestinal barrier.
Collapse
|
6
|
Ochi S, Manabe S, Kikkawa T, Osumi N. Thirty Years' History since the Discovery of Pax6: From Central Nervous System Development to Neurodevelopmental Disorders. Int J Mol Sci 2022; 23:6115. [PMID: 35682795 PMCID: PMC9181425 DOI: 10.3390/ijms23116115] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/19/2022] [Accepted: 05/27/2022] [Indexed: 12/23/2022] Open
Abstract
Pax6 is a sequence-specific DNA binding transcription factor that positively and negatively regulates transcription and is expressed in multiple cell types in the developing and adult central nervous system (CNS). As indicated by the morphological and functional abnormalities in spontaneous Pax6 mutant rodents, Pax6 plays pivotal roles in various biological processes in the CNS. At the initial stage of CNS development, Pax6 is responsible for brain patterning along the anteroposterior and dorsoventral axes of the telencephalon. Regarding the anteroposterior axis, Pax6 is expressed inversely to Emx2 and Coup-TF1, and Pax6 mutant mice exhibit a rostral shift, resulting in an alteration of the size of certain cortical areas. Pax6 and its downstream genes play important roles in balancing the proliferation and differentiation of neural stem cells. The Pax6 gene was originally identified in mice and humans 30 years ago via genetic analyses of the eye phenotypes. The human PAX6 gene was discovered in patients who suffer from WAGR syndrome (i.e., Wilms tumor, aniridia, genital ridge defects, mental retardation). Mutations of the human PAX6 gene have also been reported to be associated with autism spectrum disorder (ASD) and intellectual disability. Rodents that lack the Pax6 gene exhibit diverse neural phenotypes, which might lead to a better understanding of human pathology and neurodevelopmental disorders. This review describes the expression and function of Pax6 during brain development, and their implications for neuropathology.
Collapse
Affiliation(s)
| | | | | | - Noriko Osumi
- Department of Developmental Neuroscience, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; (S.O.); (S.M.); (T.K.)
| |
Collapse
|
7
|
Salim S, Banu A, Alwa A, Gowda SBM, Mohammad F. The gut-microbiota-brain axis in autism: what Drosophila models can offer? J Neurodev Disord 2021; 13:37. [PMID: 34525941 PMCID: PMC8442445 DOI: 10.1186/s11689-021-09378-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/06/2021] [Indexed: 12/28/2022] Open
Abstract
The idea that alterations in gut-microbiome-brain axis (GUMBA)-mediated communication play a crucial role in human brain disorders like autism remains a topic of intensive research in various labs. Gastrointestinal issues are a common comorbidity in patients with autism spectrum disorder (ASD). Although gut microbiome and microbial metabolites have been implicated in the etiology of ASD, the underlying molecular mechanism remains largely unknown. In this review, we have summarized recent findings in human and animal models highlighting the role of the gut-brain axis in ASD. We have discussed genetic and neurobehavioral characteristics of Drosophila as an animal model to study the role of GUMBA in ASD. The utility of Drosophila fruit flies as an amenable genetic tool, combined with axenic and gnotobiotic approaches, and availability of transgenic flies may reveal mechanistic insight into gut-microbiota-brain interactions and the impact of its alteration on behaviors relevant to neurological disorders like ASD.
Collapse
Affiliation(s)
- Safa Salim
- Division of Biological and Biomedical Sciences (BBS), College of Health & Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Doha, 34110, Qatar
| | - Ayesha Banu
- Division of Biological and Biomedical Sciences (BBS), College of Health & Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Doha, 34110, Qatar
| | - Amira Alwa
- Division of Biological and Biomedical Sciences (BBS), College of Health & Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Doha, 34110, Qatar
| | - Swetha B M Gowda
- Division of Biological and Biomedical Sciences (BBS), College of Health & Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Doha, 34110, Qatar
| | - Farhan Mohammad
- Division of Biological and Biomedical Sciences (BBS), College of Health & Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Doha, 34110, Qatar.
| |
Collapse
|
8
|
Collins M, Michot JM, Bellanger C, Mussini C, Benhadji K, Massard C, Carbonnel F. Notch inhibitors induce diarrhea, hypercrinia and secretory cell metaplasia in the human colon. EXCLI JOURNAL 2021; 20:819-827. [PMID: 34121974 PMCID: PMC8192874 DOI: 10.17179/excli2021-3572] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/21/2021] [Indexed: 11/20/2022]
Abstract
In humans, inhibition of Notch oncogenic signaling leads to tumor regression. Preclinical studies indicate that Notch signaling contributes to the maintenance of intestinal homeostasis. Here, we sought to describe the intestinal effects of a first-in-human Notch inhibitor in an indication of refractory cancer. Between 2014 and 2017, adult patients treated for refractory cancer with the novel Notch inhibitor LY3039478 and who had grade ≥ 2 diarrhea were referred to the gastroenterology department of a tertiary hospital in the Paris region of France. Eleven patients (median (range) age: 72 (29-83)) were included in the study. All patients had advanced cancer: adenoid cystic carcinoma (n=3, 27 %), sarcoma (n=3, 27 %), and other types (n=5, 46 %). In all cases, digestive tract endoscopy revealed abundant mucus in the intestinal lumen, and digestive tract biopsies showed an abnormally low proportion of enterocytes and marked elevation of the proportion of pseudostratified goblet cells. Microscopic inflammation was seen in colon biopsies from 2 of the 11 patients (18 %). The clinical, endoscopic and histological abnormalities were dependent on the dose of Notch inhibitor. All patients resolved their digestive signs or symptoms after discontinuing the dose and the median (range) time interval between discontinuation of the Notch inhibitor and resolution of all the gastrointestinal signs and symptoms was 7 days (4-24). Likewise, the median time interval between discontinuation and resolution of the histological abnormalities was 7 days (1-10). Blocking Notch signaling induces secretory cell metaplasia of the intestinal epithelium, which in turn leads to transient diarrhea. Our results confirm the role of Notch signaling in intestinal homeostasis in humans.
Collapse
Affiliation(s)
- Michael Collins
- Department of Gastroenterology, Kremlin Bicêtre Hospital, Assistance Publique-Hopitaux de Paris, Le Kremlin Bicêtre, France.,Paris Sud University, Le Kremlin Bicêtre, France.,INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, France; University Paris-Sud, Université Paris-Saclay, Faculté de Médecine Le Kremlin-Bicêtre, France; Assistance Publique-Hôpitaux de Paris (AP-HP), Pôle de Biologie Médicale, Paul-Brousse University Hospital, Villejuif, France
| | - Jean-Marie Michot
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Christophe Bellanger
- Department of Gastroenterology, Kremlin Bicêtre Hospital, Assistance Publique-Hopitaux de Paris, Le Kremlin Bicêtre, France.,Paris Sud University, Le Kremlin Bicêtre, France
| | - Charlotte Mussini
- Department of Pathology, Kremlin Bicêtre Hospital, Assistance Publique-Hopitaux de Paris, Le Kremlin Bicêtre, France
| | | | - Christophe Massard
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Franck Carbonnel
- Department of Gastroenterology, Kremlin Bicêtre Hospital, Assistance Publique-Hopitaux de Paris, Le Kremlin Bicêtre, France.,Paris Sud University, Le Kremlin Bicêtre, France
| |
Collapse
|
9
|
Sun J, Wang X, Xu R, Mao D, Shen D, Wang X, Qiu Y, Han Y, Lu X, Li Y, Che Q, Zheng L, Peng P, Kang X, Zhu R, Jia Y, Wang Y, Liu L, Chang Z, Ji J, Wang Z, Liu Q, Li S, Sun F, Ni J. HP1c regulates development and gut homeostasis by suppressing Notch signaling through Su(H). EMBO Rep 2021; 22:e51298. [PMID: 33594776 PMCID: PMC8024896 DOI: 10.15252/embr.202051298] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 01/01/2021] [Accepted: 01/13/2021] [Indexed: 12/30/2022] Open
Abstract
Notch signaling and epigenetic factors are known to play critical roles in regulating tissue homeostasis in most multicellular organisms, but how Notch signaling coordinates with epigenetic modulators to control differentiation remains poorly understood. Here, we identify heterochromatin protein 1c (HP1c) as an essential epigenetic regulator of gut homeostasis in Drosophila. Specifically, we observe that HP1c loss-of-function phenotypes resemble those observed after Notch signaling perturbation and that HP1c interacts genetically with components of the Notch pathway. HP1c represses the transcription of Notch target genes by directly interacting with Suppressor of Hairless (Su(H)), the key transcription factor of Notch signaling. Moreover, phenotypes caused by depletion of HP1c in Drosophila can be rescued by expressing human HP1γ, suggesting that HP1γ functions similar to HP1c in Drosophila. Taken together, our findings reveal an essential role of HP1c in normal development and gut homeostasis by suppressing Notch signaling.
Collapse
Affiliation(s)
- Jin Sun
- Gene Regulatory LabSchool of MedicineTsinghua UniversityBeijingChina
- Shandong First Medical University & Shandong Academy of Medical SciencesJinanChina
| | - Xia Wang
- Gene Regulatory LabSchool of MedicineTsinghua UniversityBeijingChina
- School of Life SciencesPeking UniversityBeijingChina
| | - Rong‐Gang Xu
- Gene Regulatory LabSchool of MedicineTsinghua UniversityBeijingChina
| | - Decai Mao
- Gene Regulatory LabSchool of MedicineTsinghua UniversityBeijingChina
- Sichuan Academy of Grassland ScienceChengduChina
| | - Da Shen
- Gene Regulatory LabSchool of MedicineTsinghua UniversityBeijingChina
| | - Xin Wang
- Institute for TCM‐XMOE Key Laboratory of Bioinformatics/Bioinformatics DivisionBNRISTDepartment of AutomationTsinghua UniversityBeijingChina
| | - Yuhao Qiu
- Gene Regulatory LabSchool of MedicineTsinghua UniversityBeijingChina
- Tsinghua University‐Peking University Joint Center for Life SciencesBeijingChina
| | - Yuting Han
- Gene Regulatory LabSchool of MedicineTsinghua UniversityBeijingChina
| | - Xinyi Lu
- Gene Regulatory LabSchool of MedicineTsinghua UniversityBeijingChina
| | - Yutong Li
- Gene Regulatory LabSchool of MedicineTsinghua UniversityBeijingChina
| | - Qinyun Che
- Gene Regulatory LabSchool of MedicineTsinghua UniversityBeijingChina
| | - Li Zheng
- Gene Regulatory LabSchool of MedicineTsinghua UniversityBeijingChina
| | - Ping Peng
- Gene Regulatory LabSchool of MedicineTsinghua UniversityBeijingChina
- Tsinghua University‐Peking University Joint Center for Life SciencesBeijingChina
| | - Xuan Kang
- Research Center for Translational Medicine at East HospitalSchool of Life Sciences and TechnologyAdvanced Institute of Translational MedicineTongji UniversityShanghaiChina
| | - Ruibao Zhu
- Gene Regulatory LabSchool of MedicineTsinghua UniversityBeijingChina
- Tsinghua University‐Peking University Joint Center for Life SciencesBeijingChina
| | - Yu Jia
- Gene Regulatory LabSchool of MedicineTsinghua UniversityBeijingChina
- Tsinghua University‐Peking University Joint Center for Life SciencesBeijingChina
| | - Yinyin Wang
- State Key Laboratory of Membrane BiologySchool of Medicine and the School of Life SciencesTsinghua UniversityBeijingChina
| | - Lu‐Ping Liu
- Gene Regulatory LabSchool of MedicineTsinghua UniversityBeijingChina
| | - Zhijie Chang
- State Key Laboratory of Membrane BiologySchool of Medicine and the School of Life SciencesTsinghua UniversityBeijingChina
| | - Jun‐Yuan Ji
- Department of Molecular and Cellular MedicineCollege of MedicineTexas A&M Health Science CenterCollege StationTXUSA
| | - Zhao Wang
- School of Pharmaceutical SciencesTsinghua UniversityBeijingChina
| | - Qingfei Liu
- School of Pharmaceutical SciencesTsinghua UniversityBeijingChina
| | - Shao Li
- Institute for TCM‐XMOE Key Laboratory of Bioinformatics/Bioinformatics DivisionBNRISTDepartment of AutomationTsinghua UniversityBeijingChina
| | - Fang‐Lin Sun
- Research Center for Translational Medicine at East HospitalSchool of Life Sciences and TechnologyAdvanced Institute of Translational MedicineTongji UniversityShanghaiChina
| | - Jian‐Quan Ni
- Gene Regulatory LabSchool of MedicineTsinghua UniversityBeijingChina
- Tsingdao Advanced Research InstituteTongji UniversityQingdaoChina
| |
Collapse
|
10
|
Park JH, Jung IK, Lee Y, Jin S, Yun HJ, Kim BW, Kwon HJ. Alcohol stimulates the proliferation of mouse small intestinal epithelial cells via Wnt signaling. Biochem Biophys Res Commun 2020; 534:639-645. [PMID: 33220923 DOI: 10.1016/j.bbrc.2020.11.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 11/08/2020] [Indexed: 02/07/2023]
Abstract
The intestinal epithelium is one of the fastest renewing tissues in mammals and is a barrier against toxic substances such as alcohol. Excessive alcohol can induce intestinal damage leading to intestinal bowel diseases. Thus, the control of small intestinal epithelial cell (IEC) regeneration is thought to be important for homeostasis in response to epithelium damage. However, reports on how epithelial cells respond to small intestinal damage are scarce. We investigated the effects of alcohol consumption on small intestinal epithelial cells of mice. To verify that alcohol altered the small intestinal epithelium, we used 8-10 weeks old male C57BL/6J mice for models of chronic and binge alcohol consumption (the NIAAA model) in addition to an organoid model. Alcohol promoted the proliferative activity of IECs and intestinal stem cells (ISCs) in intestinal crypts. Alcohol consumption increased expression of the proliferation marker cyclin D1 and activated the p44/42 MAPK (Erk1/2) signaling pathway in small intestinal epithelial cells. The Wnt target genes were markedly increased in IECs from alcohol-treated mice. In the small intestinal organoid model exposed to alcohol, the organoid area and numbers of buds increased with alcohol concentrations up to 0.5% similar to in vivo observations. These results suggest that alcohol consumption stimulates the proliferation of small intestinal epithelial cells via Wnt signaling.
Collapse
Affiliation(s)
- Jung-Ha Park
- Biopharmaceutical Engineering Major, Division of Applied Bioengineering, Dong-eui University, Busan, 47340, South Korea; Core-Facility Center for Tissue Regeneration, Dong-eui University, South Korea.
| | - In Kyo Jung
- Biopharmaceutical Engineering Major, Division of Applied Bioengineering, Dong-eui University, Busan, 47340, South Korea; Department of Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, 28159, South Korea
| | - Yongjun Lee
- Hongcheon Institute of Medicinal Herb, Hongcheon, 25142, South Korea
| | - Soojung Jin
- Core-Facility Center for Tissue Regeneration, Dong-eui University, South Korea
| | - Hee Jung Yun
- Biopharmaceutical Engineering Major, Division of Applied Bioengineering, Dong-eui University, Busan, 47340, South Korea
| | - Byung Woo Kim
- Biopharmaceutical Engineering Major, Division of Applied Bioengineering, Dong-eui University, Busan, 47340, South Korea; Blue-Bio Industry Regional Innovation Center, Dong-eui University, South Korea
| | - Hyun Ju Kwon
- Biopharmaceutical Engineering Major, Division of Applied Bioengineering, Dong-eui University, Busan, 47340, South Korea; Core-Facility Center for Tissue Regeneration, Dong-eui University, South Korea; Blue-Bio Industry Regional Innovation Center, Dong-eui University, South Korea.
| |
Collapse
|
11
|
Swamynathan SK, Wells A. Conjunctival goblet cells: Ocular surface functions, disorders that affect them, and the potential for their regeneration. Ocul Surf 2020; 18:19-26. [PMID: 31734511 PMCID: PMC7004882 DOI: 10.1016/j.jtos.2019.11.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/15/2019] [Accepted: 11/13/2019] [Indexed: 02/06/2023]
Abstract
Conjunctival goblet cells (CGCs) are specialized cells that produce and secrete soluble mucins to the tear film that bathes the ocular surface. CGC numbers and functions are affected in various ocular surface diseases including dry eye disease with diverse etiologies. In this review we will (i) summarize the important functions of CGCs in ocular surface health, (ii) describe the ocular surface diseases that affect CGC numbers and function, (iii) provide an update on recent research outcomes that elucidate CGC differentiation, gene expression and functions, and (iv) present evidence in support of the prediction that restoring CGC numbers and/or functions is a viable strategy for alleviating ocular surface disorders that impact the CGCs.
Collapse
Affiliation(s)
- Shivalingappa K Swamynathan
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Fox Center for Vision Restoration, University of Pittsburgh, Pittsburgh, PA, USA; Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Alan Wells
- McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA; Pittsburgh Veterans Affairs Medical Center, Pittsburgh, PA, USA.
| |
Collapse
|
12
|
Lloyd-Lewis B, Mourikis P, Fre S. Notch signalling: sensor and instructor of the microenvironment to coordinate cell fate and organ morphogenesis. Curr Opin Cell Biol 2019; 61:16-23. [PMID: 31323467 DOI: 10.1016/j.ceb.2019.06.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/01/2019] [Accepted: 06/13/2019] [Indexed: 12/17/2022]
Abstract
During development, stem cells give rise to specialised cell types in a tightly regulated, spatiotemporal manner to drive the formation of complex three-dimensional tissues. While mechanistic insights into the gene regulatory pathways that guide cell fate choices are emerging, how morphogenetic changes are coordinated with cell fate specification remains a fundamental question in organogenesis and adult tissue homeostasis. The requirement of cell contacts for Notch signalling makes it a central pathway capable of linking dynamic cellular rearrangements during tissue morphogenesis with stem cell function. Here, we highlight recent studies that support a critical role for the Notch pathway in translating microenvironmental cues into cell fate decisions, guiding the development of diverse organ systems.
Collapse
Affiliation(s)
- Bethan Lloyd-Lewis
- Institut Curie, PSL Research University, Inserm, CNRS, Paris, France; Sorbonne University, UPMC University of Paris VI, Paris, France
| | - Philippos Mourikis
- Université Paris Est Créteil, IMRB U955-E10, Inserm, CNRS, Créteil, France
| | - Silvia Fre
- Institut Curie, PSL Research University, Inserm, CNRS, Paris, France; Sorbonne University, UPMC University of Paris VI, Paris, France.
| |
Collapse
|
13
|
Downregulation of Notch Signaling in Kras-Induced Gastric Metaplasia. Neoplasia 2019; 21:810-821. [PMID: 31276933 PMCID: PMC6611983 DOI: 10.1016/j.neo.2019.06.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/28/2019] [Accepted: 06/04/2019] [Indexed: 11/21/2022] Open
Abstract
Activating mutations and amplification of Kras and, more frequently, signatures for Kras activation are noted in stomach cancer. Expression of mutant KrasG12D in the mouse gastric mucosa has been shown to induce hyperplasia and metaplasia. However, the mechanisms by which Kras activation leads to gastric metaplasia are not fully understood. Here we report that KrasLSL-G12D/+;Pdx1-cre, a mouse model known for pancreatic cancer, also mediates KrasG12D expression in the stomach, causing gastric hyperplasia and metaplasia prior to the pathologic changes in the pancreas. These mice exhibit ectopic cell proliferation at the base of gastric glands, whereas wild-type mice contain proliferating cells primarily at the isthmus/neck of the gastric glands. Notch signaling is decreased in the KrasLSL-G12D/+;Pdx1-cre gastric mucosa, as shown by lower levels of cleaved Notch intracellular domains and downregulation of Notch downstream target genes. Expression of a Notch ligand Jagged1 is downregulated at the base of the mutant gland, accompanied by loss of chief cell marker Mist1. We demonstrate that exogenous Jagged1 or overexpression of Notch intracellular domain stimulates Mist1 expression in gastric cancer cell lines, suggesting positive regulation of Mist1 by Notch signaling. Finally, deletion of Jagged1 or Notch3 in KrasLSL-G12D/+;Pdx1-cre mice promoted development of squamous cell carcinoma in the forestomach, albeit short of invasive adenocarcinoma in the glandular stomach. Taken together, these results reveal downregulation of Notch signaling and Mist1 expression during the initiation of Kras-driven gastric tumorigenesis and suggest a tumor-suppressive role for Notch in this context.
Collapse
|
14
|
Abstract
Purpose of Review The trillions of microbes collectively referred to as the human microbiota, inhabit the human body and establish a beneficial relationship with the host. It is clear however that dysbiosis impacting microbial diversity in the gut, may lead to development of inflammatory and malignant gastrointestinal diseases including colorectal cancer (CRC). We provide a literature review of the recent influx of information related to the alterations in gut microbiota composition that influences CRC incidence and progression. Recent Findings A growing body of evidence implicates altered gut microbiota in the development of CRC. Profiles of CRC associated microbiota have been shown to differ from those in healthy subjects and bacterial phylotypes vary depending on the primary tumor location. The compositional variation in the microbial profile is not restricted to cancerous tissue however and is different between cancers of the proximal and distal colons, respectively. More recently, studies have shed light on the "driver-passenger" model for CRC wherein, driver bacteria cause inflammation, increased cell proliferation and production of genotoxic substances to contribute towards mutational acquisition associated with adenoma-carcinoma sequence. These changes facilitate gradual replacement of driver bacteria by passengers that either promote or suppress tumor progression. Significant advances have also been made in associating individual bacterial species to consensus molecular subtypes (CMS) of CRC and this remarkable development is expected to galvanize scientific community into advancing therapeutic strategies for CRC. Summary Increasing evidence suggests a link between the intestinal microbiota and CRC development although the mechanisms through which the bacterial constituents of the microbiome contribute towards CRC are complex and yet to be fully fathomed. Thus, more exhaustive and mechanistic studies are needed to identify key interactions amongst diet, microbial community and metabolites that help facilitate the adenoma-carcinoma sequence evolution in CRC. It is expected that development of therapeutics based on microbial association with CMS will likely facilitate the translation of molecular subtypes into the clinic for CRCs and potentially other malignancies.
Collapse
|
15
|
Troll JV, Hamilton MK, Abel ML, Ganz J, Bates JM, Stephens WZ, Melancon E, van der Vaart M, Meijer AH, Distel M, Eisen JS, Guillemin K. Microbiota promote secretory cell determination in the intestinal epithelium by modulating host Notch signaling. Development 2018; 145:145/4/dev155317. [PMID: 29475973 DOI: 10.1242/dev.155317] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 01/19/2018] [Indexed: 12/15/2022]
Abstract
Resident microbes promote many aspects of host development, although the mechanisms by which microbiota influence host tissues remain unclear. We showed previously that the microbiota is required for allocation of appropriate numbers of secretory cells in the zebrafish intestinal epithelium. Because Notch signaling is crucial for secretory fate determination, we conducted epistasis experiments to establish whether the microbiota modulates host Notch signaling. We also investigated whether innate immune signaling transduces microbiota cues via the Myd88 adaptor protein. We provide the first evidence that microbiota-induced, Myd88-dependent signaling inhibits host Notch signaling in the intestinal epithelium, thereby promoting secretory cell fate determination. These results connect microbiota activity via innate immune signaling to the Notch pathway, which also plays crucial roles in intestinal homeostasis throughout life and when impaired can result in chronic inflammation and cancer.
Collapse
Affiliation(s)
- Joshua V Troll
- Institute of Molecular Biology, Department of Biology, 1229 University of Oregon, Eugene, OR 97403, USA
| | - M Kristina Hamilton
- Institute of Neuroscience, Department of Biology, 1254 University of Oregon, Eugene, OR 97403, USA
| | - Melissa L Abel
- Institute of Molecular Biology, Department of Biology, 1229 University of Oregon, Eugene, OR 97403, USA
| | - Julia Ganz
- Institute of Neuroscience, Department of Biology, 1254 University of Oregon, Eugene, OR 97403, USA
| | - Jennifer M Bates
- Institute of Molecular Biology, Department of Biology, 1229 University of Oregon, Eugene, OR 97403, USA
| | - W Zac Stephens
- Institute of Molecular Biology, Department of Biology, 1229 University of Oregon, Eugene, OR 97403, USA
| | - Ellie Melancon
- Institute of Neuroscience, Department of Biology, 1254 University of Oregon, Eugene, OR 97403, USA
| | | | - Annemarie H Meijer
- Institute of Biology, Leiden University, 2300 RA Leiden, The Netherlands
| | - Martin Distel
- Children's Cancer Research Institute, 1090 Vienna, Austria
| | - Judith S Eisen
- Institute of Neuroscience, Department of Biology, 1254 University of Oregon, Eugene, OR 97403, USA
| | - Karen Guillemin
- Institute of Molecular Biology, Department of Biology, 1229 University of Oregon, Eugene, OR 97403, USA .,Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, Ontario, Canada M5G 1Z8
| |
Collapse
|
16
|
Frau C, Godart M, Plateroti M. Thyroid hormone regulation of intestinal epithelial stem cell biology. Mol Cell Endocrinol 2017; 459:90-97. [PMID: 28288904 DOI: 10.1016/j.mce.2017.03.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 02/28/2017] [Accepted: 03/02/2017] [Indexed: 01/08/2023]
Abstract
The gastrointestinal tract is a well-characterized target of thyroid hormones and thyroid hormone nuclear receptors TRs, as extensively described in the literature. The paradigm is its important remodelling in amphibians during thyroid hormone-dependent metamorphosis. Interestingly, several studies have described the conservation of this hormonal signal during intestinal development in mammals. Additional data suggested that it may also play a role in intestinal homeostasis, stem cell physiology and progenitor commitment as well as in tumour development. It is worth underlining that in the mammalian intestine the functionality of the TRα1 receptor is coordinated and integrated with other signalling pathways, such as Wnt and Notch, specifically at the level of stem/progenitor cell populations. Here, we summarize these data and concepts and discuss this new role for thyroid hormones and the TRα1 receptor in the biology of intestinal epithelial precursor cells.
Collapse
Affiliation(s)
- Carla Frau
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université de Lyon, Université Lyon 1, Centre Léon Bérard, Département de La Recherche, 69000 Lyon, France
| | - Matthias Godart
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université de Lyon, Université Lyon 1, Centre Léon Bérard, Département de La Recherche, 69000 Lyon, France
| | - Michelina Plateroti
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université de Lyon, Université Lyon 1, Centre Léon Bérard, Département de La Recherche, 69000 Lyon, France.
| |
Collapse
|
17
|
Sallé J, Gervais L, Boumard B, Stefanutti M, Siudeja K, Bardin AJ. Intrinsic regulation of enteroendocrine fate by Numb. EMBO J 2017; 36:1928-1945. [PMID: 28533229 DOI: 10.15252/embj.201695622] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 04/10/2017] [Accepted: 04/11/2017] [Indexed: 12/25/2022] Open
Abstract
How terminal cell fates are specified in dynamically renewing adult tissues is not well understood. Here we explore terminal cell fate establishment during homeostasis using the enteroendocrine cells (EEs) of the adult Drosophila midgut as a paradigm. Our data argue against the existence of local feedback signals, and we identify Numb as an intrinsic regulator of EE fate. Our data further indicate that Numb, with alpha-adaptin, acts upstream or in parallel of known regulators of EE fate to limit Notch signaling, thereby facilitating EE fate acquisition. We find that Numb is regulated in part through its asymmetric and symmetric distribution during stem cell divisions; however, its de novo synthesis is also required during the differentiation of the EE cell. Thus, this work identifies Numb as a crucial factor for cell fate choice in the adult Drosophila intestine. Furthermore, our findings demonstrate that cell-intrinsic control mechanisms of terminal cell fate acquisition can result in a balanced tissue-wide production of terminally differentiated cell types.
Collapse
Affiliation(s)
- Jérémy Sallé
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, Stem Cells and Tissue Homeostasis Group, Paris, France.,Sorbonne Universités, UPMC Univ Paris 6, Paris, France
| | - Louis Gervais
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, Stem Cells and Tissue Homeostasis Group, Paris, France.,Sorbonne Universités, UPMC Univ Paris 6, Paris, France
| | - Benjamin Boumard
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, Stem Cells and Tissue Homeostasis Group, Paris, France.,Sorbonne Universités, UPMC Univ Paris 6, Paris, France.,Département de Biologie, École Normale Supérieure de Lyon, Lyon, France
| | - Marine Stefanutti
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, Stem Cells and Tissue Homeostasis Group, Paris, France.,Sorbonne Universités, UPMC Univ Paris 6, Paris, France
| | - Katarzyna Siudeja
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, Stem Cells and Tissue Homeostasis Group, Paris, France.,Sorbonne Universités, UPMC Univ Paris 6, Paris, France
| | - Allison J Bardin
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, Stem Cells and Tissue Homeostasis Group, Paris, France .,Sorbonne Universités, UPMC Univ Paris 6, Paris, France
| |
Collapse
|
18
|
The thyroid hormone nuclear receptors and the Wnt/β-catenin pathway: An intriguing liaison. Dev Biol 2017; 422:71-82. [DOI: 10.1016/j.ydbio.2017.01.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 12/26/2016] [Accepted: 01/04/2017] [Indexed: 12/11/2022]
|
19
|
Origin and dynamic lineage characteristics of the developing Drosophila midgut stem cells. Dev Biol 2016; 416:347-60. [PMID: 27321560 DOI: 10.1016/j.ydbio.2016.06.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 06/14/2016] [Accepted: 06/15/2016] [Indexed: 01/01/2023]
Abstract
Proliferating intestinal stem cells (ISCs) generate all cell types of the Drosophila midgut, including enterocytes, endocrine cells, and gland cells (e.g., copper cells), throughout the lifetime of the animal. Among the signaling mechanisms controlling the balance between ISC self-renewal and the production of different cell types, Notch (N) plays a pivotal role. In this paper we investigated the emergence of ISCs during metamorphosis and the role of N in this process. Precursors of the Drosophila adult intestinal stem cells (pISCs) can be first detected within the pupal midgut during the first hours after onset of metamorphosis as motile mesenchymal cells. pISCs perform 2-3 rounds of parasynchronous divisions. The first mitosis yields only an increase in pISC number. During the following rounds of mitosis, dividing pISCs give rise to more pISCs, as well as the endocrine cells that populate the midgut of the eclosing fly. Enterocytes do not appear among the pISC progeny until around the time of eclosion. The "proendocrine" gene prospero (pros), expressed from mid-pupal stages onward in pISCs, is responsible to advance the endocrine fate in these cells; following removal of pros, pISCs continue to proliferate, but endocrine cells do not form. Conversely, the onset of N activity that occurs around the stage when pros comes on restricts pros expression among pISCs. Loss of N abrogates proliferation and switches on an endocrine fate among all pISCs. Our results suggest that a switch depending on the activity of N and pros acts at the level of the pISC to decide between continued proliferation and endocrine differentiation.
Collapse
|
20
|
Fender AW, Nutter JM, Fitzgerald TL, Bertrand FE, Sigounas G. Notch-1 promotes stemness and epithelial to mesenchymal transition in colorectal cancer. J Cell Biochem 2016; 116:2517-27. [PMID: 25914224 DOI: 10.1002/jcb.25196] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 04/14/2015] [Indexed: 12/16/2022]
Abstract
Colorectal cancer (CRC) is the third leading cause of cancer death in the United States, resulting in an average of 50,000 deaths per year. Surgery and combination chemotherapy comprise current treatment strategies. However, curative options are limited if surgery and chemotherapy are unsuccessful. Several studies have indicated that CRC aggressiveness and potential for metastatic spread are associated with the acquisition of stem cell like properties. The Notch-1 receptor and its cognate signaling pathway is well known for controlling cell fate decisions and stem-cell phenotypes. Alterations in Notch receptors and Notch signaling has been reported for some colon cancers. Herein, we examine a potential role for Notch-1 signaling in CRC. In CRC patient samples, Notch-1 expression was increased in colon tumor tissue as compared with normal colon tissue. Retroviral transduction of constitutively active Notch-1 (ICN1) into the colon tumor cell line HCT-116 resulted in increased expression of the EMT/stemness associated proteins CD44, Slug, Smad-3, and induction of Jagged-1 expression. These changes in ICN1 expressing cells were accompanied by increased migration and increased anchorage independent growth by 2.5-fold and 23%, respectively. Experiments with the pan-Notch inhibitor DAPT, and soluble Jagged-1-Fc protein provided evidence that Notch-1 signaling activates CD44, Slug, and Smad-3 via a cascade of other Notch-receptors through induction of Jagged-1 expression. These data indicate a key role for Notch signaling in the phenotype of CRC and suggest that targeting of Notch signaling may be of therapeutic value in colon cancers.
Collapse
Affiliation(s)
- Alexander W Fender
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, North Carolina.,Department of Oncology, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - Jennifer M Nutter
- Department of Oncology, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - Timothy L Fitzgerald
- Department of Surgery, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - Fred E Bertrand
- Department of Oncology, Brody School of Medicine, East Carolina University, Greenville, North Carolina.,Department of Clinical and Diagnostic Sciences, Department of Nutrition Sciences, School of Health Professions, University of Alabama, Birmingham, Alabama
| | - George Sigounas
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, North Carolina.,Department of Oncology, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| |
Collapse
|
21
|
|
22
|
Sirakov M, Boussouar A, Kress E, Frau C, Lone IN, Nadjar J, Angelov D, Plateroti M. The thyroid hormone nuclear receptor TRα1 controls the Notch signaling pathway and cell fate in murine intestine. Development 2015; 142:2764-74. [PMID: 26286942 DOI: 10.1242/dev.121962] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Thyroid hormones control various aspects of gut development and homeostasis. The best-known example is in gastrointestinal tract remodeling during amphibian metamorphosis. It is well documented that these hormones act via the TR nuclear receptors, which are hormone-modulated transcription factors. Several studies have shown that thyroid hormones regulate the expression of several genes in the Notch signaling pathway, indicating a possible means by which they participate in the control of gut physiology. However, the mechanisms and biological significance of this control have remained unexplored. Using multiple in vivo and in vitro approaches, we show that thyroid hormones positively regulate Notch activity through the TRα1 receptor. From a molecular point of view, TRα1 indirectly controls Notch1, Dll1, Dll4 and Hes1 expression but acts as a direct transcriptional regulator of the Jag1 gene by binding to a responsive element in the Jag1 promoter. Our findings show that the TRα1 nuclear receptor plays a key role in intestinal crypt progenitor/stem cell biology by controlling the Notch pathway and hence the balance between cell proliferation and cell differentiation.
Collapse
Affiliation(s)
- Maria Sirakov
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Claude Bernard Lyon 1, 16 Rue Raphael Dubois, Villeurbanne 69622, France
| | - Amina Boussouar
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Claude Bernard Lyon 1, 16 Rue Raphael Dubois, Villeurbanne 69622, France
| | - Elsa Kress
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Claude Bernard Lyon 1, 16 Rue Raphael Dubois, Villeurbanne 69622, France
| | - Carla Frau
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Claude Bernard Lyon 1, 16 Rue Raphael Dubois, Villeurbanne 69622, France
| | - Imtiaz Nisar Lone
- Laboratoire de Biologie Moléculaire de la cellule, Ecole Normale Supérieure de Lyon, 46 Allée d'Italie, Lyon 69007, France
| | - Julien Nadjar
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Claude Bernard Lyon 1, 16 Rue Raphael Dubois, Villeurbanne 69622, France
| | - Dimitar Angelov
- Laboratoire de Biologie Moléculaire de la cellule, Ecole Normale Supérieure de Lyon, 46 Allée d'Italie, Lyon 69007, France
| | - Michelina Plateroti
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Claude Bernard Lyon 1, 16 Rue Raphael Dubois, Villeurbanne 69622, France
| |
Collapse
|
23
|
Le Guelte A, Macara IG. Plasmolipin--a new player in endocytosis and epithelial development. EMBO J 2015; 34:1147-8. [PMID: 25825384 DOI: 10.15252/embj.201591448] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
- Armelle Le Guelte
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Ian G Macara
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
24
|
Developmental regulation of apical endocytosis controls epithelial patterning in vertebrate tubular organs. Nat Cell Biol 2015; 17:241-50. [PMID: 25706235 DOI: 10.1038/ncb3106] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 01/09/2015] [Indexed: 02/07/2023]
Abstract
Epithelial organs develop through tightly coordinated events of cell proliferation and differentiation in which endocytosis plays a major role. Despite recent advances, how endocytosis regulates the development of vertebrate organs is still unknown. Here we describe a mechanism that facilitates the apical availability of endosomal SNARE receptors for epithelial morphogenesis through the developmental upregulation of plasmolipin (pllp) in a highly endocytic segment of the zebrafish posterior midgut. The protein PLLP (Pllp in fish) recruits the clathrin adaptor EpsinR to sort the SNARE machinery of the endolysosomal pathway into the subapical compartment, which is a switch for polarized endocytosis. Furthermore, PLLP expression induces apical Crumbs internalization and the activation of the Notch signalling pathway, both crucial steps in the acquisition of cell polarity and differentiation of epithelial cells. We thus postulate that differential apical endosomal SNARE sorting is a mechanism that regulates epithelial patterning.
Collapse
|
25
|
De Mey JR, Freund JN. Understanding epithelial homeostasis in the intestine: An old battlefield of ideas, recent breakthroughs and remaining controversies. Tissue Barriers 2014; 1:e24965. [PMID: 24665395 PMCID: PMC3879175 DOI: 10.4161/tisb.24965] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 05/04/2013] [Accepted: 05/07/2013] [Indexed: 12/14/2022] Open
Abstract
The intestinal epithelium constitutes the barrier between the gut lumen and the rest of the body and a very actively renewing cell population. The crypt/villus and crypt/cuff units of the mouse small intestine and colon are its basic functional units. The field is confronted with competing concepts with regard to the nature of the cells that are responsible for all the day-to day cell replacement and those that act to regenerate the tissue upon injury and with two diametrically opposed models for lineage specification. The review revisits groundbreaking pioneering studies to provide non expert readers and crypt watchers with a factual analysis of the origins of the current models deduced from the latest spectacular advances. It also discusses recent progress made by addressing these issues in the crypts of the colon, which need to be better understood, since they are the preferred sites of major pathologies.
Collapse
Affiliation(s)
- Jan R De Mey
- CNRS, UMR 7213; Laboratoire de Biophotonique et Pharmacologie; Illkirch, France ; Université de Strasbourg; Strasbourg, France
| | - Jean-Noël Freund
- Université de Strasbourg; Strasbourg, France ; INSERM_U113; Strasbourg, France ; Fédération de Médecine Translationnelle; Strasbourg, France
| |
Collapse
|
26
|
Castellanos-Rivera RM, Pentz ES, Lin E, Gross KW, Medrano S, Yu J, Sequeira-Lopez MLS, Gomez RA. Recombination signal binding protein for Ig-κJ region regulates juxtaglomerular cell phenotype by activating the myo-endocrine program and suppressing ectopic gene expression. J Am Soc Nephrol 2014; 26:67-80. [PMID: 24904090 DOI: 10.1681/asn.2013101045] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Recombination signal binding protein for Ig-κJ region (RBP-J), the major downstream effector of Notch signaling, is necessary to maintain the number of renin-positive juxtaglomerular cells and the plasticity of arteriolar smooth muscle cells to re-express renin when homeostasis is threatened. We hypothesized that RBP-J controls a repertoire of genes that defines the phenotype of the renin cell. Mice bearing a bacterial artificial chromosome reporter with a mutated RBP-J binding site in the renin promoter had markedly reduced reporter expression at the basal state and in response to a homeostatic challenge. Mice with conditional deletion of RBP-J in renin cells had decreased expression of endocrine (renin and Akr1b7) and smooth muscle (Acta2, Myh11, Cnn1, and Smtn) genes and regulators of smooth muscle expression (miR-145, SRF, Nfatc4, and Crip1). To determine whether RBP-J deletion decreased the endowment of renin cells, we traced the fate of these cells in RBP-J conditional deletion mice. Notably, the lineage staining patterns in mutant and control kidneys were identical, although mutant kidneys had fewer or no renin-expressing cells in the juxtaglomerular apparatus. Microarray analysis of mutant arterioles revealed upregulation of genes usually expressed in hematopoietic cells. Thus, these results suggest that RBP-J maintains the identity of the renin cell by not only activating genes characteristic of the myo-endocrine phenotype but also, preventing ectopic gene expression and adoption of an aberrant phenotype, which could have severe consequences for the control of homeostasis.
Collapse
Affiliation(s)
- Ruth M Castellanos-Rivera
- Department of Pediatrics, School of Medicine, Department of Biology, Graduate School of Arts and Sciences, and
| | | | - Eugene Lin
- Department of Pediatrics, School of Medicine, Department of Biology, Graduate School of Arts and Sciences, and
| | - Kenneth W Gross
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, New York
| | | | - Jing Yu
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia; and
| | | | - R Ariel Gomez
- Department of Pediatrics, School of Medicine, Department of Biology, Graduate School of Arts and Sciences, and
| |
Collapse
|
27
|
Okumura T, Takeda K, Taniguchi K, Adachi-Yamada T. βν integrin inhibits chronic and high level activation of JNK to repress senescence phenotypes in Drosophila adult midgut. PLoS One 2014; 9:e89387. [PMID: 24586740 PMCID: PMC3930726 DOI: 10.1371/journal.pone.0089387] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 01/20/2014] [Indexed: 01/24/2023] Open
Abstract
Proper control of adult stem cells including their proliferation and differentiation is crucial in maintaining homeostasis of well-organized tissues/organs throughout an organism's life. The Drosophila adult midgut has intestinal stem cells (ISCs), which have been exploited as a simple model system to investigate mechanisms controlling adult tissue homeostasis. Here, we found that a viable mutant of βν integrin (βint-ν), encoding one of two Drosophila integrin β subunits, showed a short midgut and abnormal multilayered epithelia accompanied by an increase in ISC proliferation and misdifferentiation defects. The increase in ISC proliferation and misdifferentiation was due to frequent ISC duplication expanding a pool of ISCs, which was caused by depression of the Notch signalling, and up-regulation of unpaired (upd), a gene encoding an extracellular ligand in the JAK/STAT signalling pathway. In addition, we observed that abnormally high accumulation of filamentous actin (F-actin) was caused in the βint-ν mutant enterocytes. Furthermore, the defects were rescued by suppressing c-Jun N-terminal kinase (JNK) signalling, which was up-regulated in a manner correlated with the defect levels in the above-mentioned βint-ν mutant phenotype. These symptoms observed in young βint-ν mutant midgut were very similar to those in the aged midgut in wild type. Our results suggested that βint-ν has a novel function for the Drosophila adult midgut homeostasis under normal conditions and provided a new insight into possible age-related diseases caused by latent abnormality of an integrin function.
Collapse
Affiliation(s)
- Takashi Okumura
- Department of Life Science, Faculty of Science, Gakushuin University, Tokyo, Japan
| | - Koji Takeda
- Department of Life Science, Faculty of Science, Gakushuin University, Tokyo, Japan
- Graduate Course in Life Science, Graduate School of Science, Gakushuin University, Tokyo, Japan
| | - Kiichiro Taniguchi
- Department of Life Science, Faculty of Science, Gakushuin University, Tokyo, Japan
| | - Takashi Adachi-Yamada
- Department of Life Science, Faculty of Science, Gakushuin University, Tokyo, Japan
- Graduate Course in Life Science, Graduate School of Science, Gakushuin University, Tokyo, Japan
- Institute for Biomolecular Science, Gakushuin University, Tokyo, Japan
- * E-mail:
| |
Collapse
|
28
|
Abstract
Cell-cell interactions define a quintessential aspect of multicellular development. Metazoan morphogenesis depends on a handful of fundamental, conserved cellular interaction mechanisms, one of which is defined by the Notch signaling pathway. Signals transmitted through the Notch surface receptor have a unique developmental role: Notch signaling links the fate of one cell with that of a cellular neighbor through physical interactions between the Notch receptor and the membrane-bound ligands that are expressed in an apposing cell. The developmental outcome of Notch signals is strictly dependent on the cellular context and can influence differentiation, proliferation and apoptotic cell fates. The Notch pathway is conserved across species (Artavanis-Tsakonas et al., 1999; Bray, 2006; Kopan and Ilagan, 2009). In humans, Notch malfunction has been associated with a diverse range of diseases linked to changes in cell fate and cell proliferation including cancer (Louvi and Artavanis-Tsakonas, 2012). In this Cell Science at a Glance article and the accompanying poster we summarize the molecular biology of Notch signaling, its role in development and its relevance to disease.
Collapse
Affiliation(s)
- Kazuya Hori
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, LHRRB-418, Boston, MA 02115, USA
| | | | | |
Collapse
|
29
|
Panayidou S, Apidianakis Y. Regenerative inflammation: lessons from Drosophila intestinal epithelium in health and disease. Pathogens 2013; 2:209-31. [PMID: 25437036 PMCID: PMC4235722 DOI: 10.3390/pathogens2020209] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 03/18/2013] [Accepted: 03/22/2013] [Indexed: 01/04/2023] Open
Abstract
Intestinal inflammation is widely recognized as a pivotal player in health and disease. Defined cytologically as the infiltration of leukocytes in the lamina propria layer of the intestine, it can damage the epithelium and, on a chronic basis, induce inflammatory bowel disease and potentially cancer. The current view thus dictates that blood cell infiltration is the instigator of intestinal inflammation and tumor-promoting inflammation. This is based partially on work in humans and mice showing that intestinal damage during microbially mediated inflammation activates phagocytic cells and lymphocytes that secrete inflammatory signals promoting tissue damage and tumorigenesis. Nevertheless, extensive parallel work in the Drosophila midgut shows that intestinal epithelium damage induces inflammatory signals and growth factors acting mainly in a paracrine manner to induce intestinal stem cell proliferation and tumor formation when genetically predisposed. This is accomplished without any apparent need to involve Drosophila hemocytes. Therefore, recent work on Drosophila host defense to infection by expanding its main focus on systemic immunity signaling pathways to include the study of organ homeostasis in health and disease shapes a new notion that epithelially emanating cytokines and growth factors can directly act on the intestinal stem cell niche to promote “regenerative inflammation” and potentially cancer.
Collapse
Affiliation(s)
- Stavria Panayidou
- Department of Biological Sciences, University of Cyprus, Nicosia 1678, Cyprus.
| | - Yiorgos Apidianakis
- Department of Biological Sciences, University of Cyprus, Nicosia 1678, Cyprus.
| |
Collapse
|
30
|
Takashima S, Gold D, Hartenstein V. Stem cells and lineages of the intestine: a developmental and evolutionary perspective. Dev Genes Evol 2013; 223:85-102. [PMID: 23179635 PMCID: PMC3873164 DOI: 10.1007/s00427-012-0422-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Accepted: 10/12/2012] [Indexed: 12/19/2022]
Abstract
The intestine consists of epithelial cells that secrete digestive enzymes and mucus (gland cells), absorb food particles (enterocytes), and produce hormones (endocrine cells). Intestinal cells are rapidly turned over and need to be replaced. In cnidarians, mitosis of differentiated intestinal cells accounts for much of the replacement; in addition, migratory, multipotent stem cells (interstitial cells) contribute to the production of intestinal cells. In other phyla, intestinal cell replacement is solely the function of stem cells entering the gut from the outside (such as in case of the neoblasts of platyhelminths) or intestinal stem cells located within the midgut epithelium (as in both vertebrates or arthropods). We will attempt in the following to review important aspects of midgut stem cells in different animal groups: where are they located, what types of lineages do they produce, and how do they develop. We will start out with a comparative survey of midgut cell types found across the animal kingdom; then briefly look at the specification of these cells during embryonic development; and finally focus on the stem cells that regenerate midgut cells during adult life. In a number of model systems, including mouse, zebrafish and Drosophila, the molecular pathways controlling intestinal stem cells proliferation and the specification of intestinal cell types are under intensive investigation. We will highlight findings of the recent literature, focusing on aspects that are shared between the different models and that point at evolutionary ancient mechanisms of intestinal cell formation.
Collapse
Affiliation(s)
- Shigeo Takashima
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | | | | |
Collapse
|
31
|
Andriatsilavo M, Gervais L, Fons C, Bardin AJ. [The Drosophila midgut as a model to study adult stem cells]. Med Sci (Paris) 2013; 29:75-81. [PMID: 23351697 DOI: 10.1051/medsci/2013291016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Constant renewal of cells occurs in most tissues throughout the adult lifetime and is insured by the activity of resident stem cells. Recent work has demonstrated the presence of adult stem cells in the Drosophila intestine and consequently, the Drosophila intestine has become a powerful model to understand adult stem cells in vivo. In this review, we summarize our current understanding of the mechanisms controlling cell fate decisions of the intestinal stem cells with a particular focus on the role of the Notch pathway in this process. We also summarize what is known about proliferation control of the intestinal stem cells, which is crucial to maintain tissue homeostasis during normal and environmentally stressful conditions.
Collapse
Affiliation(s)
- Mahéva Andriatsilavo
- Génétique et biologie du développement, Institut Curie, UMR 3215 CNRS, Inserm U934, 26, rue d'Ulm, 75248 Paris Cedex 05, France
| | | | | | | |
Collapse
|
32
|
Roach G, Heath Wallace R, Cameron A, Emrah Ozel R, Hongay CF, Baral R, Andreescu S, Wallace KN. Loss of ascl1a prevents secretory cell differentiation within the zebrafish intestinal epithelium resulting in a loss of distal intestinal motility. Dev Biol 2013; 376:171-86. [PMID: 23353550 DOI: 10.1016/j.ydbio.2013.01.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Revised: 01/05/2013] [Accepted: 01/08/2013] [Indexed: 02/07/2023]
Abstract
The vertebrate intestinal epithelium is renewed continuously from stem cells at the base of the crypt in mammals or base of the fold in fish over the life of the organism. As stem cells divide, newly formed epithelial cells make an initial choice between a secretory or enterocyte fate. This choice has previously been demonstrated to involve Notch signaling as well as Atonal and Her transcription factors in both embryogenesis and adults. Here, we demonstrate that in contrast to the atoh1 in mammals, ascl1a is responsible for formation of secretory cells in zebrafish. ascl1a-/- embryos lack all intestinal epithelial secretory cells and instead differentiate into enterocytes. ascl1a-/- embryos also fail to induce intestinal epithelial expression of deltaD suggesting that ascl1a plays a role in initiation of Notch signaling. Inhibition of Notch signaling increases the number of ascl1a and deltaD expressing intestinal epithelial cells as well as the number of developing secretory cells during two specific time periods: between 30 and 34hpf and again between 64 and 74hpf. Loss of enteroendocrine products results in loss of anterograde motility in ascl1a-/- embryos. 5HT produced by enterochromaffin cells is critical in motility and secretion within the intestine. We find that addition of exogenous 5HT to ascl1a-/- embryos at near physiological levels (measured by differential pulse voltammetry) induce anterograde motility at similar levels to wild type velocity, distance, and frequency. Removal or doubling the concentration of 5HT in WT embryos does not significantly affect anterograde motility, suggesting that the loss of additional enteroendocrine products in ascl1a-/- embryos also contributes to intestinal motility. Thus, zebrafish intestinal epithelial cells appear to have a common secretory progenitor from which all subtypes form. Loss of enteroendocrine cells reveals the critical need for enteroendocrine products in maintenance of normal intestinal motility.
Collapse
Affiliation(s)
- Gillian Roach
- Department of Biology, Clarkson University, 8 Clarkson Ave., Potsdam, NY 13699, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Flasse LC, Stern DG, Pirson JL, Manfroid I, Peers B, Voz ML. The bHLH transcription factor Ascl1a is essential for the specification of the intestinal secretory cells and mediates Notch signaling in the zebrafish intestine. Dev Biol 2013; 376:187-97. [PMID: 23352790 DOI: 10.1016/j.ydbio.2013.01.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 01/09/2013] [Accepted: 01/11/2013] [Indexed: 11/24/2022]
Abstract
Notch signaling has a fundamental role in stem cell maintenance and in cell fate choice in the intestine of different species. Canonically, Notch signaling represses the expression of transcription factors of the achaete-scute like (ASCL) or atonal related protein (ARP) families. Identifying the ARP/ASCL genes expressed in the gastrointestinal tract is essential to build the regulatory cascade controlling the differentiation of gastrointestinal progenitors into the different intestinal cell types. The expression of the ARP/ASCL factors was analyzed in zebrafish to identify, among all the ARP/ASCL factors found in the zebrafish genome, those expressed in the gastrointestinal tract. ascl1a was found to be the earliest factor detected in the intestine. Loss-of-function analyses using the pia/ascl1a mutant, revealed that ascl1a is crucial for the differentiation of all secretory cells. Furthermore, we identify a battery of transcription factors expressed during secretory cell differentiation and downstream of ascl1a. Finally, we show that the repression of secretory cell fate by Notch signaling is mediated by the inhibition of ascl1a expression. In conclusion, this work identifies Ascl1a as a key regulator of the secretory cell lineage in the zebrafish intestine, playing the same role as Atoh1 in the mouse intestine. This highlights the diversity in the ARP/ASCL family members acting as cell fate determinants downstream from Notch signaling.
Collapse
Affiliation(s)
- Lydie C Flasse
- Unit of Molecular Biology and Genetic Engineering, Giga-Research, University of Liège, 1 avenue de l'Hôpital B34, B-4000 Sart-Tilman (Liège), Belgium
| | | | | | | | | | | |
Collapse
|
34
|
Abstract
The small and large intestines are tubular organs composed of several tissue types. The columnar epithelium that lines the inner surface of the intestines distinguishes the digestive physiology of each region of the intestine and consists of several distinct cell types that are rapidly and continually renewed by intestinal stem cells that reside near the base of the crypts of Lieberkühn. Notch signaling controls the fate of intestinal stem cells by regulating the expression of Hes genes and by repressing Atoh1. Alternate models of Notch pathway control of cell fate determination are presented. Roles for Notch signaling in development of the intestine, including mesenchymal and neural cells, are discussed. The oncogenic activities of Notch in colorectal cancer, as well as the tumor suppressive activities of Atoh1, are reviewed. Therapeutic targeting of the Notch pathway in colorectal cancers is discussed, along with potential caveats.
Collapse
Affiliation(s)
- Taeko K Noah
- Division of Gastroenterology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio 45229, USA
| | | |
Collapse
|
35
|
Bertrand FE, Angus CW, Partis WJ, Sigounas G. Developmental pathways in colon cancer: crosstalk between WNT, BMP, Hedgehog and Notch. Cell Cycle 2012; 11:4344-51. [PMID: 23032367 DOI: 10.4161/cc.22134] [Citation(s) in RCA: 135] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A hallmark of cancer is reactivation/alteration of pathways that control cellular differentiation during developmental processes. Evidence indicates that WNT, Notch, BMP and Hedgehog pathways have a role in normal epithelial cell differentiation, and that alterations in these pathways accompany establishment of the tumorigenic state. Interestingly, there is recent evidence that these pathways are intertwined at the molecular level, and these nodes of intersection may provide opportunities for effective targeted therapies. This review will highlight the role of the WNT, Notch, BMP and Hedgehog pathways in colon cancer.
Collapse
Affiliation(s)
- Fred E Bertrand
- Division of Cancer Biology, Department of Oncology, Brody School of Medicine at East Carolina University, Greenville, NC USA.
| | | | | | | |
Collapse
|
36
|
Vanuytsel T, Senger S, Fasano A, Shea-Donohue T. Major signaling pathways in intestinal stem cells. Biochim Biophys Acta Gen Subj 2012; 1830:2410-26. [PMID: 22922290 DOI: 10.1016/j.bbagen.2012.08.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 07/05/2012] [Accepted: 08/07/2012] [Indexed: 12/12/2022]
Abstract
BACKGROUND The discovery of markers to identify the intestinal stem cell population and the generation of powerful transgenic mouse models to study stem cell physiology have led to seminal discoveries in stem cell biology. SCOPE OF REVIEW In this review we give an overview of the current knowledge in the field of intestinal stem cells (ISCs) highlighting the most recent progress on markers defining the ISC population and pathways governing intestinal stem cell maintenance and differentiation. Furthermore we review their interaction with other stem cell related pathways. Finally we give an overview of alteration of these pathways in human inflammatory gastrointestinal diseases. MAJOR CONCLUSIONS We highlight the complex network of interactions occurring among different pathways and put in perspective the many layers of regulation that occur in maintaining the intestinal homeostasis. GENERAL SIGNIFICANCE Understanding the involvement of ISCs in inflammatory diseases can potentially lead to new therapeutic approaches to treat inflammatory GI pathologies such as IBD and celiac disease and could reveal the molecular mechanisms leading to the pathogenesis of dysplasia and cancer in inflammatory chronic conditions. This article is part of a Special Issue entitled Biochemistry of Stem Cells.
Collapse
Affiliation(s)
- Tim Vanuytsel
- Mucosal Biology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | |
Collapse
|
37
|
Evaluation of phosphatidylinositol-4-kinase IIIα as a hepatitis C virus drug target. J Virol 2012; 86:11595-607. [PMID: 22896614 DOI: 10.1128/jvi.01320-12] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Phosphatidylinositol-4-kinase IIIα (PI4KIIIα) is an essential host cell factor for hepatitis C virus (HCV) replication. An N-terminally truncated 130-kDa form was used to reconstitute an in vitro biochemical lipid kinase assay that was optimized for small-molecule compound screening and identified potent and specific inhibitors. Cell culture studies with PI4KIIIα inhibitors demonstrated that the kinase activity was essential for HCV RNA replication. Two PI4KIIIα inhibitors were used to select cell lines harboring HCV replicon mutants with a 20-fold loss in sensitivity to the compounds. Reverse genetic mapping isolated an NS4B-NS5A segment that rescued HCV RNA replication in PIK4IIIα-deficient cells. HCV RNA replication occurs on specialized membranous webs, and this study with PIK4IIIα inhibitor-resistant mutants provides a genetic link between NS4B/NS5A functions and PI4-phosphate lipid metabolism. A comprehensive assessment of PI4KIIIα as a drug target included its evaluation for pharmacologic intervention in vivo through conditional transgenic murine lines that mimic target-specific inhibition in adult mice. Homozygotes that induce a knockout of the kinase domain or knock in a single amino acid substitution, kinase-defective PI4KIIIα, displayed a lethal phenotype with a fairly widespread mucosal epithelial degeneration of the gastrointestinal tract. This essential host physiologic role raises doubt about the pursuit of PI4KIIIα inhibitors for treatment of chronic HCV infection.
Collapse
|
38
|
Bellis J, Duluc I, Romagnolo B, Perret C, Faux MC, Dujardin D, Formstone C, Lightowler S, Ramsay RG, Freund JN, De Mey JR. The tumor suppressor Apc controls planar cell polarities central to gut homeostasis. ACTA ACUST UNITED AC 2012; 198:331-41. [PMID: 22851318 PMCID: PMC3413367 DOI: 10.1083/jcb.201204086] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Asymmetric stem cell divisions controlled by Apc in the intestinal crypt result in regulated, anisotropic movement of daughter cells away from the niche. The stem cells (SCs) at the bottom of intestinal crypts tightly contact niche-supporting cells and fuel the extraordinary tissue renewal of intestinal epithelia. Their fate is regulated stochastically by populational asymmetry, yet whether asymmetrical fate as a mode of SC division is relevant and whether the SC niche contains committed progenitors of the specialized cell types are under debate. We demonstrate spindle alignments and planar cell polarities, which form a novel functional unit that, in SCs, can yield daughter cell anisotropic movement away from niche-supporting cells. We propose that this contributes to SC homeostasis. Importantly, we demonstrate that some SC divisions are asymmetric with respect to cell fate and provide data suggesting that, in some SCs, mNumb displays asymmetric segregation. Some of these processes were altered in apparently normal crypts and microadenomas of mice carrying germline Apc mutations, shedding new light on the first stages of progression toward colorectal cancer.
Collapse
Affiliation(s)
- Julien Bellis
- Laboratoire de Biophotonique et Pharmacologie, Unité Mixte de Recherche 7213, Centre National de la Recherche Scientifique, 67401 Illkirch, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Takashima S, Hartenstein V. Genetic control of intestinal stem cell specification and development: a comparative view. Stem Cell Rev Rep 2012; 8:597-608. [PMID: 22529012 PMCID: PMC3950647 DOI: 10.1007/s12015-012-9351-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Stem cells of the adult vertebrate intestine (ISCs) are responsible for the continuous replacement of intestinal cells, but also serve as site of origin of intestinal neoplasms. The interaction between multiple signaling pathways, including Wnt/Wg, Shh/Hh, BMP, and Notch, orchestrate mitosis, motility, and differentiation of ISCs. Many fundamental questions of how these pathways carry out their function remain unanswered. One approach to gain more insight is to look at the development of stem cells, to analyze the "programming" process which these cells undergo as they emerge from the large populations of embryonic progenitors. This review intends to summarize pertinent data on vertebrate intestinal stem cell biology, to then take a closer look at recent studies of intestinal stem cell development in Drosophila. Here, stem cell pools and their niche environment consist of relatively small numbers of cells, and questions concerning the pattern of cell division, niche-stem cell contacts, or differentiation can be addressed at the single cell level. Likewise, it is possible to analyze the emergence of stem cells during development more easily than in vertebrate systems: where in the embryo do stem cells arise, what structures in their environment do they interact with, and what signaling pathways are active sequentially as a result of these interactions. Given the high degree of conservation among genetic mechanisms controlling stem cell behavior in all animals, findings in Drosophila will provide answers that inform research in the vertebrate stem cell field.
Collapse
Affiliation(s)
- Shigeo Takashima
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | | |
Collapse
|
40
|
Jiang H, Edgar BA. Intestinal stem cell function in Drosophila and mice. Curr Opin Genet Dev 2012; 22:354-60. [PMID: 22608824 DOI: 10.1016/j.gde.2012.04.002] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 03/29/2012] [Accepted: 04/11/2012] [Indexed: 11/17/2022]
Abstract
Epithelial cells of the digestive tracts of most animals are short-lived, and are constantly replenished by the progeny of long-lived, resident intestinal stem cells. Proper regulation of intestinal stem cell maintenance, proliferation and differentiation is critical for maintaining gut homeostasis. Here we review recent genetic studies of stem cell-mediated homeostatic growth in the Drosophila midgut and the mouse small intestine, highlighting similarities and differences in the mechanisms that control stem cell proliferation and differentiation.
Collapse
Affiliation(s)
- Huaqi Jiang
- Department of Developmental Biology, UT Southwestern Medical Center, Dallas, TX 75235-9133, USA.
| | | |
Collapse
|
41
|
Drosophila midgut homeostasis involves neutral competition between symmetrically dividing intestinal stem cells. EMBO J 2012; 31:2473-85. [PMID: 22522699 PMCID: PMC3365418 DOI: 10.1038/emboj.2012.106] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2011] [Accepted: 03/23/2012] [Indexed: 12/13/2022] Open
Abstract
The Drosophila adult posterior midgut has been identified as a powerful system in which to study mechanisms that control intestinal maintenance, in normal conditions as well as during injury or infection. Early work on this system has established a model of tissue turnover based on the asymmetric division of intestinal stem cells. From the quantitative analysis of clonal fate data, we show that tissue turnover involves the neutral competition of symmetrically dividing stem cells. This competition leads to stem-cell loss and replacement, resulting in neutral drift dynamics of the clonal population. As well as providing new insight into the mechanisms regulating tissue self-renewal, these findings establish intriguing parallels with the mammalian system, and confirm Drosophila as a useful model for studying adult intestinal maintenance.
Collapse
|
42
|
Nakamura K, Ayabe T. Paneth cells and stem cells in the intestinal stem cell niche and their association with inflammatory bowel disease. Inflamm Regen 2012. [DOI: 10.2492/inflammregen.32.053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|