1
|
Miura Y, Fujii S, Ichinohe T. Cell-based and extracellular vesicle-based MSC therapies for acute radiation syndrome affecting organ systems. JOURNAL OF RADIATION RESEARCH 2024; 65:i80-i87. [PMID: 39679884 DOI: 10.1093/jrr/rrae009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/28/2024] [Indexed: 12/17/2024]
Abstract
Exposure to ionizing radiation can induce harmful biological effects on the human body, particularly in cases of high-dose γ-irradiation affecting the gastrointestinal tract, bone marrow, skin and lung. Such exposures lead to lethal outcomes as individuals experience a breakdown in their immune system's ability to defend against pathogens, predisposing them to sepsis-induced multiple organ failures. Mesenchymal stromal/stem cells (MSCs) possess diverse biological characteristics, including immunomodulation, anti-inflammation and tissue regeneration. Off-the-shelf culture-expanded human bone marrow- or adipose tissue-derived MSCs are clinically available to treat graft-versus-host disease following hematopoietic cell transplantation and perianal fistulas in Crohn's disease in Japan. While preclinical studies showcase encouraging outcomes in radiation-induced injuries, the effectiveness of MSC transplantation in addressing acute radiation syndrome affecting organs in irradiated individuals is limited. Recent studies have highlighted MSC-releasing extracellular vesicles as nanoparticle substances responsible for outlining the mechanism of action and have identified various components, including proteins and microRNA, that serve as functional molecules. MSC-releasing extracellular vesicle-based therapy emerges as a promising avenue, offering a potential solution to the challenges posed by radiation-induced injuries. However, further investigation is required, especially regarding whether MSC-releasing extracellular vesicles have regenerative effects on tissue-resident stem cells. These unresolved issues represent key aspects that need to be addressed to optimize the therapeutic potential of cell-based and extracellular vesicle-based MSC therapies for interventions in the context of radiation-induced injuries.
Collapse
Affiliation(s)
- Yasuo Miura
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
- Department of Transfusion Medicine and Cell Therapy, Fujita Health University School of Medicine, 1-93 Dengakugakubo, Kutsukakecho, Toyoake, Aichi 470-1192, Japan
| | - Sumie Fujii
- Department of Transfusion Medicine and Cell Therapy, Fujita Health University School of Medicine, 1-93 Dengakugakubo, Kutsukakecho, Toyoake, Aichi 470-1192, Japan
| | - Tatsuo Ichinohe
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| |
Collapse
|
2
|
Baquero J, Tang XH, Ferrotta A, Zhang T, DiKun KM, Gudas LJ. The transcription factor BMI1 increases hypoxic signaling in oral cavity epithelia. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167161. [PMID: 38599260 PMCID: PMC11370312 DOI: 10.1016/j.bbadis.2024.167161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/07/2024] [Accepted: 04/04/2024] [Indexed: 04/12/2024]
Abstract
The tongue epithelium is maintained by a proliferative basal layer. This layer contains long-lived stem cells (SCs), which produce progeny cells that move up to the surface as they differentiate. B-lymphoma Mo-MLV insertion region 1 (BMI1), a protein in mammalian Polycomb Repressive Complex 1 (PRC1) and a biomarker of oral squamous cell carcinoma, is expressed in almost all basal epithelial SCs of the tongue, and single, Bmi1-labelled SCs give rise to cells in all epithelial layers. We previously developed a transgenic mouse model (KrTB) containing a doxycycline- (dox) controlled, Tet-responsive element system to selectively overexpress Bmi1 in the tongue basal epithelial SCs. Here, we used this model to assess BMI1 actions in tongue epithelia. Genome-wide transcriptomics revealed increased levels of transcripts involved in the cellular response to hypoxia in Bmi1-overexpressing (KrTB+DOX) oral epithelia even though these mice were not subjected to hypoxia conditions. Ectopic Bmi1 expression in tongue epithelia increased the levels of hypoxia inducible factor-1 alpha (HIF1α) and HIF1α targets linked to metabolic reprogramming during hypoxia. We used chromatin immunoprecipitation (ChIP) to demonstrate that Bmi1 associates with the promoters of HIF1A and HIF1A-activator RELA (p65) in tongue epithelia. We also detected increased SC proliferation and oxidative stress in Bmi1-overexpressing tongue epithelia. Finally, using a human oral keratinocyte line (OKF6-TERT1R), we showed that ectopic BMI1 overexpression decreases the oxygen consumption rate while increasing the extracellular acidification rate, indicative of elevated glycolysis. Thus, our data demonstrate that high BMI1 expression drives hypoxic signaling, including metabolic reprogramming, in normal oral cavity epithelia.
Collapse
Affiliation(s)
- Jorge Baquero
- Department of Pharmacology, Weill Cornell Medical College, New York, NY, USA; Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Xiao-Han Tang
- Department of Pharmacology, Weill Cornell Medical College, New York, NY, USA; Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Annalisa Ferrotta
- Department of Pharmacology, Weill Cornell Medical College, New York, NY, USA; Department of Pharmacology, Weill Cornell Graduate School of Biomedical Sciences, New York, NY, USA
| | - Tuo Zhang
- Weill Cornell Genomics Core Facility, Weill Cornell Medical College, New York, NY, USA
| | - Krysta M DiKun
- Department of Pharmacology, Weill Cornell Medical College, New York, NY, USA; Department of Pharmacology, Weill Cornell Graduate School of Biomedical Sciences, New York, NY, USA
| | - Lorraine J Gudas
- Department of Pharmacology, Weill Cornell Medical College, New York, NY, USA; Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Department of Pharmacology, Weill Cornell Graduate School of Biomedical Sciences, New York, NY, USA.
| |
Collapse
|
3
|
Krzysiek-Maczka G, Brzozowski T, Ptak-Belowska A. Helicobacter pylori-activated fibroblasts as a silent partner in gastric cancer development. Cancer Metastasis Rev 2023; 42:1219-1256. [PMID: 37460910 PMCID: PMC10713772 DOI: 10.1007/s10555-023-10122-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/20/2023] [Indexed: 12/18/2023]
Abstract
The discovery of Helicobacter pylori (Hp) infection of gastric mucosa leading to active chronic gastritis, gastroduodenal ulcers, and MALT lymphoma laid the groundwork for understanding of the general relationship between chronic infection, inflammation, and cancer. Nevertheless, this sequence of events is still far from full understanding with new players and mediators being constantly identified. Originally, the Hp virulence factors affecting mainly gastric epithelium were proposed to contribute considerably to gastric inflammation, ulceration, and cancer. Furthermore, it has been shown that Hp possesses the ability to penetrate the mucus layer and directly interact with stroma components including fibroblasts and myofibroblasts. These cells, which are the source of biophysical and biochemical signals providing the proper balance between cell proliferation and differentiation within gastric epithelial stem cell compartment, when exposed to Hp, can convert into cancer-associated fibroblast (CAF) phenotype. The crosstalk between fibroblasts and myofibroblasts with gastric epithelial cells including stem/progenitor cell niche involves several pathways mediated by non-coding RNAs, Wnt, BMP, TGF-β, and Notch signaling ligands. The current review concentrates on the consequences of Hp-induced increase in gastric fibroblast and myofibroblast number, and their activation towards CAFs with the emphasis to the altered communication between mesenchymal and epithelial cell compartment, which may lead to inflammation, epithelial stem cell overproliferation, disturbed differentiation, and gradual gastric cancer development. Thus, Hp-activated fibroblasts may constitute the target for anti-cancer treatment and, importantly, for the pharmacotherapies diminishing their activation particularly at the early stages of Hp infection.
Collapse
Affiliation(s)
- Gracjana Krzysiek-Maczka
- Department of Physiology, the Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531, Kraków, Poland.
| | - Tomasz Brzozowski
- Department of Physiology, the Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531, Kraków, Poland.
| | - Agata Ptak-Belowska
- Department of Physiology, the Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531, Kraków, Poland
| |
Collapse
|
4
|
Chen G, Yu Z, Zhang Y, Liu S, Chen C, Zhang S. Radiation-induced gastric injury during radiotherapy: molecular mechanisms and clinical treatment. JOURNAL OF RADIATION RESEARCH 2023; 64:870-879. [PMID: 37788485 PMCID: PMC10665304 DOI: 10.1093/jrr/rrad071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/08/2023] [Indexed: 10/05/2023]
Abstract
Radiotherapy (RT) has been the standard of care for treating a multitude of cancer types. Radiation-induced gastric injury (RIGI) is a common complication of RT for thoracic and abdominal tumors. It manifests acutely as radiation gastritis or gastric ulcers, and chronically as chronic atrophic gastritis or intestinal metaplasia. In recent years, studies have shown that intracellular signals such as oxidative stress response, p38/MAPK pathway and transforming growth factor-β signaling pathway are involved in the progression of RIGI. This review also summarized the risk factors, diagnosis and treatment of this disease. However, the root of therapeutic challenges lies in the incomplete understanding of the mechanisms. Here, we also highlight the potential mechanistic, diagnostic and therapeutic directions of RIGI.
Collapse
Affiliation(s)
- Guangxia Chen
- Department of Gastroenterology, The First People’s Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou 221200, China
| | - Zuxiang Yu
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Yuehua Zhang
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Shiyu Liu
- Department of Gastroenterology, The First People’s Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou 221200, China
| | - Chong Chen
- Department of Gastroenterology, The First People’s Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou 221200, China
| | - Shuyu Zhang
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital , Chengdu 610051, China
- NHC Key Laboratory of Nuclear Technology Medical Transformation (Mianyang Central Hospital), Mianyang 621099, China
| |
Collapse
|
5
|
Shiokawa D, Sakai H, Koizumi M, Okimoto Y, Mori Y, Kanda Y, Ohata H, Honda H, Okamoto K. Elevated stress response marks deeply quiescent reserve cells of gastric chief cells. Commun Biol 2023; 6:1183. [PMID: 37985874 PMCID: PMC10662433 DOI: 10.1038/s42003-023-05550-2] [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: 02/11/2023] [Accepted: 11/07/2023] [Indexed: 11/22/2023] Open
Abstract
Gastrointestinal tract organs harbor reserve cells, which are endowed with cellular plasticity and regenerate functional units in response to tissue damage. However, whether the reserve cells in gastrointestinal tract exist as long-term quiescent cells remain incompletely understood. In the present study, we systematically examine H2b-GFP label-retaining cells and identify a long-term slow-cycling population in the gastric corpus but not in other gastrointestinal organs. The label-retaining cells, which reside near the basal layers of the corpus, comprise a subpopulation of chief cells. The identified quiescent cells exhibit induction of Atf4 and its target genes including Atf3, a marker of paligenosis, and activation of the unfolded protein response, but do not show elevated expression of Troy, Lgr5, or Mist. External damage to the gastric mucosa induced by indomethacin treatment triggers proliferation of the quiescent Atf4+ population, indicating that the gastric corpus harbors a specific cell population that is primed to facilitate stomach regeneration.
Collapse
Affiliation(s)
- Daisuke Shiokawa
- Division of Molecular Pharmacology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
- Ehime University Hospital Translational Research Center, Shitsukawa, Toon, 791-0295, Ehime, Japan
| | - Hiroaki Sakai
- Advanced Comprehensive Research Organization, Teikyo University, 2-21-1 Kaga, Itabashi-ku, Tokyo, 173-0003, Japan
| | - Miho Koizumi
- Field of Human Disease Models, Major in Advanced Life Sciences and Medicine, Tokyo Women's Medical University, 81- Kawada-cho, Shinjuku-ku, 162-8666, Tokyo, Japan
| | - Yoshie Okimoto
- Advanced Comprehensive Research Organization, Teikyo University, 2-21-1 Kaga, Itabashi-ku, Tokyo, 173-0003, Japan
| | - Yutaro Mori
- Advanced Comprehensive Research Organization, Teikyo University, 2-21-1 Kaga, Itabashi-ku, Tokyo, 173-0003, Japan
| | - Yusuke Kanda
- Advanced Comprehensive Research Organization, Teikyo University, 2-21-1 Kaga, Itabashi-ku, Tokyo, 173-0003, Japan
| | - Hirokazu Ohata
- Advanced Comprehensive Research Organization, Teikyo University, 2-21-1 Kaga, Itabashi-ku, Tokyo, 173-0003, Japan
| | - Hiroaki Honda
- Field of Human Disease Models, Major in Advanced Life Sciences and Medicine, Tokyo Women's Medical University, 81- Kawada-cho, Shinjuku-ku, 162-8666, Tokyo, Japan.
| | - Koji Okamoto
- Advanced Comprehensive Research Organization, Teikyo University, 2-21-1 Kaga, Itabashi-ku, Tokyo, 173-0003, Japan.
| |
Collapse
|
6
|
Alvina FB, Chen TCY, Lim HYG, Barker N. Gastric epithelial stem cells in development, homeostasis and regeneration. Development 2023; 150:dev201494. [PMID: 37746871 DOI: 10.1242/dev.201494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
The stem/progenitor cell pool is indispensable for the development, homeostasis and regeneration of the gastric epithelium, owing to its defining ability to self-renew whilst supplying the various functional epithelial lineages needed to digest food efficiently. A detailed understanding of the intricacies and complexities surrounding the behaviours and roles of these stem cells offers insights, not only into the physiology of gastric epithelial development and maintenance, but also into the pathological consequences following aberrations in stem cell regulation. Here, we provide an insightful synthesis of the existing knowledge on gastric epithelial stem cell biology, including the in vitro and in vivo experimental techniques that have advanced such studies. We highlight the contributions of stem/progenitor cells towards patterning the developing stomach, specification of the differentiated cell lineages and maintenance of the mature epithelium during homeostasis and following injury. Finally, we discuss gaps in our understanding and identify key research areas for future work.
Collapse
Affiliation(s)
- Fidelia B Alvina
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Tanysha Chi-Ying Chen
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Hui Yi Grace Lim
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Nick Barker
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore 117593, Republic of Singapore
| |
Collapse
|
7
|
Chen Q, Weng K, Lin M, Jiang M, Fang Y, Chung SSW, Huang X, Zhong Q, Liu Z, Huang Z, Lin J, Li P, El-Rifai W, Zaika A, Li H, Rustgi AK, Nakagawa H, Abrams JA, Wang TC, Lu C, Huang C, Que J. SOX9 Modulates the Transformation of Gastric Stem Cells Through Biased Symmetric Cell Division. Gastroenterology 2023; 164:1119-1136.e12. [PMID: 36740200 PMCID: PMC10200757 DOI: 10.1053/j.gastro.2023.01.037] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 01/24/2023] [Accepted: 01/31/2023] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Transformation of stem/progenitor cells has been associated with tumorigenesis in multiple tissues, but stem cells in the stomach have been hard to localize. We therefore aimed to use a combination of several markers to better target oncogenes to gastric stem cells and understand their behavior in the initial stages of gastric tumorigenesis. METHODS Mouse models of gastric metaplasia and cancer by targeting stem/progenitor cells were generated and analyzed with techniques including reanalysis of single-cell RNA sequencing and immunostaining. Gastric cancer cell organoids were genetically manipulated with clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) for functional studies. Cell division was determined by bromodeoxyuridine-chasing assay and the assessment of the orientation of the mitotic spindles. Gastric tissues from patients were examined by histopathology and immunostaining. RESULTS Oncogenic insults lead to expansion of SOX9+ progenitor cells in the mouse stomach. Genetic lineage tracing and organoid culture studies show that SOX9+ gastric epithelial cells overlap with SOX2+ progenitors and include stem cells that can self-renew and differentiate to generate all gastric epithelial cells. Moreover, oncogenic targeting of SOX9+SOX2+ cells leads to invasive gastric cancer in our novel mouse model (Sox2-CreERT;Sox9-loxp(66)-rtTA-T2A-Flpo-IRES-loxp(71);Kras(Frt-STOP-Frt-G12D);P53R172H), which combines Cre-loxp and Flippase-Frt genetic recombination systems. Sox9 deletion impedes the expansion of gastric progenitor cells and blocks neoplasia after Kras activation. Although Sox9 is not required for maintaining tissue homeostasis where asymmetric division predominates, loss of Sox9 in the setting of Kras activation leads to reduced symmetric cell division and effectively attenuates the Kras-dependent expansion of stem/progenitor cells. Similarly, Sox9 deletion in gastric cancer organoids reduces symmetric cell division, organoid number, and organoid size. In patients with gastric cancer, high levels of SOX9 are associated with recurrence and poor prognosis. CONCLUSION SOX9 marks gastric stem cells and modulates biased symmetric cell division, which appears to be required for the malignant transformation of gastric stem cells.
Collapse
Affiliation(s)
- Qiyue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China; Columbia Center for Human Development, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, People's Republic of China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, People's Republic of China; Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York
| | - Kai Weng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, People's Republic of China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Mi Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China; Columbia Center for Human Development, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, People's Republic of China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, People's Republic of China; Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York
| | - Ming Jiang
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Yinshan Fang
- Columbia Center for Human Development, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York
| | - Sanny S W Chung
- Columbia Center for Human Development, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York
| | - Xiaobo Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, People's Republic of China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Qing Zhong
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, People's Republic of China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Zhiyu Liu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, People's Republic of China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Zening Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, People's Republic of China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Jianxian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, People's Republic of China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, People's Republic of China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Wael El-Rifai
- Department of Surgery, University of Miami, Miami, Florida; Department of Veterans Affairs, Miami Healthcare System, Miami, Florida
| | - Alexander Zaika
- Department of Surgery, University of Miami, Miami, Florida; Department of Veterans Affairs, Miami Healthcare System, Miami, Florida
| | - Haiyan Li
- Department of Pathology & Laboratory Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Anil K Rustgi
- Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York
| | - Hiroshi Nakagawa
- Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York
| | - Julian A Abrams
- Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York
| | - Timothy C Wang
- Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York
| | - Chao Lu
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, New York
| | - Changming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, People's Republic of China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, People's Republic of China.
| | - Jianwen Que
- Columbia Center for Human Development, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York; Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York.
| |
Collapse
|
8
|
Huebner AJ, Gorelov RA, Deviatiiarov R, Demharter S, Kull T, Walsh RM, Taylor MS, Steiger S, Mullen JT, Kharchenko PV, Hochedlinger K. Dissection of gastric homeostasis in vivo facilitates permanent capture of isthmus-like stem cells in vitro. Nat Cell Biol 2023; 25:390-403. [PMID: 36717627 DOI: 10.1038/s41556-022-01079-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 12/12/2022] [Indexed: 02/01/2023]
Abstract
The glandular stomach is composed of two regenerative compartments termed corpus and antrum, and our understanding of the transcriptional networks that maintain these tissues is incomplete. Here we show that cell types with equivalent functional roles in the corpus and antrum share similar transcriptional states including the poorly characterized stem cells of the isthmus region. To further study the isthmus, we developed a monolayer two-dimensional (2D) culture system that is continually maintained by Wnt-responsive isthmus-like cells capable of differentiating into several gastric cell types. Importantly, 2D cultures can be converted into conventional three-dimensional organoids, modelling the plasticity of gastric epithelial cells in vivo. Finally, we utilized the 2D culture system to show that Sox2 is both necessary and sufficient to generate enterochromaffin cells. Together, our data provide important insights into gastric homeostasis, establish a tractable culture system to capture isthmus cells and uncover a role for Sox2 in enterochromaffin cells.
Collapse
Affiliation(s)
- Aaron J Huebner
- Massachusetts General Hospital Department of Molecular Biology, Boston, MA, USA
- Massachusetts General Hospital Cancer Center and Center for Regenerative Medicine, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Rebecca A Gorelov
- Massachusetts General Hospital Department of Molecular Biology, Boston, MA, USA
- Massachusetts General Hospital Cancer Center and Center for Regenerative Medicine, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Ruslan Deviatiiarov
- Institute of Fundamental Medicine and Biology, Kazan Feberal University, Kazan, Russia
| | - Samuel Demharter
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Tobias Kull
- Massachusetts General Hospital Department of Molecular Biology, Boston, MA, USA
- Massachusetts General Hospital Cancer Center and Center for Regenerative Medicine, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Ryan M Walsh
- Massachusetts General Hospital Department of Molecular Biology, Boston, MA, USA
- Massachusetts General Hospital Cancer Center and Center for Regenerative Medicine, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Marty S Taylor
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Simon Steiger
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - John T Mullen
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Peter V Kharchenko
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
- San Diego Institute, Altos Labs, San Diego, CA, USA.
| | - Konrad Hochedlinger
- Massachusetts General Hospital Department of Molecular Biology, Boston, MA, USA.
- Massachusetts General Hospital Cancer Center and Center for Regenerative Medicine, Boston, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
9
|
Peng M, Wu J, Wang W, Liao T, Xu S, Xiao D, He Z, Yang X. Alpha-tocopherol enhances spermatogonial stem cell proliferation and restores mouse spermatogenesis by up-regulating BMI1. Front Nutr 2023; 10:1141964. [PMID: 37139440 PMCID: PMC10150882 DOI: 10.3389/fnut.2023.1141964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/31/2023] [Indexed: 05/05/2023] Open
Abstract
Purpose Spermatogonial stem cells (SSCs) are essential for maintaining reproductive function in males. B-lymphoma Mo-MLV insertion region 1 (BMI1) is a vital transcription repressor that regulates cell proliferation and differentiation. However, little is known about the role of BMI1 in mediating the fate of mammalian SSCs and in male reproduction. This study investigated whether BMI1 is essential for male reproduction and the role of alpha-tocopherol (α-tocopherol), a protective agent for male fertility, as a modulator of BMI1 both in vitro and in vivo. Methods Methyl thiazolyl tetrazolium (MTT) and 5-ethynyl-2'-deoxyuridine (EDU) assays were used to assess the effect of BMI1 on the proliferative ability of the mouse SSC line C18-4. Real-time polymerase chain reaction (PCR), western blotting, and immunofluorescence were applied to investigate changes in the mRNA and protein expression levels of BMI1. Male mice were used to investigate the effect of α-tocopherol and a BMI1 inhibitor on reproduction-associated functionality in vivo. Results Analysis revealed that BMI1 was expressed at high levels in testicular tissues and spermatogonia in mice. The silencing of BMI1 inhibited the proliferation of SSCs and DNA synthesis and enhanced the levels of γ-H2AX. α-tocopherol enhanced the proliferation and DNA synthesis of C18-4 cells, and increased the levels of BMI1. Notably, α-tocopherol rescued the inhibition of cell proliferation and DNA damage in C18-4 cells caused by the silencing of BMI1. Furthermore, α-tocopherol restored sperm count (Ctrl vs. PTC-209, p = 0.0034; Ctrl vs. PTC-209 + α-tocopherol, p = 0.7293) and normalized sperm malformation such as broken heads, irregular heads, lost and curled tails in vivo, as demonstrated by its antagonism with the BMI1 inhibitor PTC-209. Conclusion Analysis demonstrated that α-tocopherol is a potent in vitro and in vivo modulator of BMI1, a transcription factor that plays an important role in in SSC proliferation and spermatogenesis. Our findings identify a new target and strategy for treating male infertility that deserves further pre-clinical investigation.
Collapse
|
10
|
Wizenty J, Sigal M. Gastric Stem Cell Biology and Helicobacter pylori Infection. Curr Top Microbiol Immunol 2023; 444:1-24. [PMID: 38231213 DOI: 10.1007/978-3-031-47331-9_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Helicobacter pylori colonizes the human gastric mucosa and persists lifelong. An interactive network between the bacteria and host cells shapes a unique microbial niche within gastric glands that alters epithelial behavior, leading to pathologies such as chronic gastritis and eventually gastric cancer. Gland colonization by the bacterium initiates aberrant trajectories by inducing long-term inflammatory and regenerative gland responses, which involve various specialized epithelial and stromal cells. Recent studies using cell lineage tracing, organoids and scRNA-seq techniques have significantly advanced our knowledge of the molecular "identity" of epithelial and stromal cell subtypes during normal homeostasis and upon infection, and revealed the principles that underly stem cell (niche) behavior under homeostatic conditions as well as upon H. pylori infection. The activation of long-lived stem cells deep in the gastric glands has emerged as a key prerequisite of H. pylori-associated gastric site-specific pathologies such as hyperplasia in the antrum, and atrophy or metaplasia in the corpus, that are considered premalignant lesions. In addition to altering the behaviour of bona fide stem cells, injury-driven de-differentiation and trans-differentation programs, such as "paligenosis", subsequently allow highly specialized secretory cells to re-acquire stem cell functions, driving gland regeneration. This plastic regenerative capacity of gastric glands is required to maintain homeostasis and repair mucosal injuries. However, these processes are co-opted in the context of stepwise malignant transformation in chronic H. pylori infection, causing the emergence, selection and expansion of cancer-promoting stem cells.
Collapse
Affiliation(s)
- Jonas Wizenty
- Division of Gastroenterology and Hepatology, Medical Department, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Sigal
- Division of Gastroenterology and Hepatology, Medical Department, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
11
|
Asano N, Takeuchi A, Imatani A, Saito M, Jin X, Hatta W, Uno K, Koike T, Masamune A. Wnt Signaling and Aging of the Gastrointestinal Tract. Int J Mol Sci 2022; 23:12210. [PMID: 36293064 PMCID: PMC9603545 DOI: 10.3390/ijms232012210] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/10/2022] [Accepted: 10/10/2022] [Indexed: 11/30/2022] Open
Abstract
Aging is considered a risk factor for various diseases including cancers. In this aging society, there is an urgent need to clarify the molecular mechanisms involved in aging. Wnt signaling has been shown to play a crucial role in the maintenance and differentiation of tissue stem cells, and intensive studies have elucidated its pivotal role in the aging of neural and muscle stem cells. However, until recently, such studies on the gastrointestinal tract have been limited. In this review, we discuss recent advances in the study of the role of Wnt signaling in the aging of the gastrointestinal tract and aging-related carcinogenesis.
Collapse
Affiliation(s)
- Naoki Asano
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Wuputra K, Ku CC, Pan JB, Liu CJ, Liu YC, Saito S, Kato K, Lin YC, Kuo KK, Chan TF, Chong IW, Lin CS, Wu DC, Yokoyama KK. Stem Cell Biomarkers and Tumorigenesis in Gastric Cancer. J Pers Med 2022; 12:jpm12060929. [PMID: 35743714 PMCID: PMC9224738 DOI: 10.3390/jpm12060929] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/25/2022] [Accepted: 05/31/2022] [Indexed: 02/01/2023] Open
Abstract
Stomach cancer has a high mortality, which is partially caused by an absence of suitable biomarkers to allow detection of the initiation stages of cancer progression. Thus, identification of critical biomarkers associated with gastric cancer (GC) is required to advance its clinical diagnoses and treatment. Recent studies using tracing models for lineage analysis of GC stem cells indicate that the cell fate decision of the gastric stem cells might be an important issue for stem cell plasticity. They include leucine-rich repeat-containing G-protein-coupled receptor 5 (Lgr5+), Cholecystokinin receptor 2 (Cckr2+), and axis inhibition protein 2 (Axin2+) as the stem cell markers in the antrum, Trefoil Factor 2 (TFF2+), Mist1+ stem cells, and Troy+ chief cells in the corpus. By contrast, Estrogen receptor 1 (eR1), Leucine-rich repeats and immunoglobulin-like domains 1 (Lrig1), SRY (sex determining region Y)-box 2 (Sox2), and B lymphoma Mo-MLV insertion region 1 homolog (Bmi1) are rich in both the antrum and corpus regions. These markers might help to identify the cell-lineage identity and analyze the plasticity of each stem cell population. Thus, identification of marker genes for the development of GC and its environment is critical for the clinical application of cancer stem cells in the prevention of stomach cancers.
Collapse
Affiliation(s)
- Kenly Wuputra
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (K.W.); (C.-C.K.); (J.-B.P.); (C.-S.L.)
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-J.L.); (K.-K.K.); (D.-C.W.)
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan;
| | - Chia-Chen Ku
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (K.W.); (C.-C.K.); (J.-B.P.); (C.-S.L.)
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-J.L.); (K.-K.K.); (D.-C.W.)
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan;
| | - Jia-Bin Pan
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (K.W.); (C.-C.K.); (J.-B.P.); (C.-S.L.)
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-J.L.); (K.-K.K.); (D.-C.W.)
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan;
| | - Chung-Jung Liu
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-J.L.); (K.-K.K.); (D.-C.W.)
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan;
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan
- Department of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yi-Chang Liu
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan;
| | - Shigeo Saito
- Saito Laboratory of Cell Technology, Yaita 329-2192, Japan;
- Horus Co., Ltd., Nakano, Tokyo 164-0001, Japan
| | - Kohsuke Kato
- Department of Infection Biology, Graduate School of Comprehensive Human Sciences, The University of Tsukuba, Tsukuba 305-8577, Japan;
| | - Ying-Chu Lin
- School of Dentistry, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Kung-Kai Kuo
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-J.L.); (K.-K.K.); (D.-C.W.)
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan;
- Division of General & Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan
| | - Te-Fu Chan
- Department of Obstetrics and Genecology, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan;
| | - Inn-Wen Chong
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan;
| | - Chang-Shen Lin
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (K.W.); (C.-C.K.); (J.-B.P.); (C.-S.L.)
| | - Deng-Chyang Wu
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-J.L.); (K.-K.K.); (D.-C.W.)
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan;
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan
- Department of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Obstetrics and Genecology, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan;
| | - Kazunari K. Yokoyama
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (K.W.); (C.-C.K.); (J.-B.P.); (C.-S.L.)
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-J.L.); (K.-K.K.); (D.-C.W.)
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan;
- Correspondence: ; Tel.: +886-7312-1101 (ext. 2729); Fax: +886-7313-3849
| |
Collapse
|
13
|
Hoffmann W. Self-Renewal and Cancers of the Gastric Epithelium: An Update and the Role of the Lectin TFF1 as an Antral Tumor Suppressor. Int J Mol Sci 2022; 23:ijms23105377. [PMID: 35628183 PMCID: PMC9141172 DOI: 10.3390/ijms23105377] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/09/2022] [Accepted: 05/10/2022] [Indexed: 11/16/2022] Open
Abstract
In 2020, gastric cancer was the fourth leading cause of cancer deaths globally. About 90% of gastric cancers are sporadic and the vast majority are correlated with Helicobacter pylori infection; whereas familial clustering is observed in about 10% of cases. Gastric cancer is now considered to be a disease originating from dysregulated self-renewal of the gastric glands in the setting of an inflammatory environment. The human stomach contains two types of gastric units, which show bi-directional self-renewal from a complex variety of stem cells. This review focuses on recent progress concerning the characterization of the different stem cell populations and the mainly mesenchymal signals triggering their stepwise differentiation as well as the genesis of pre-cancerous lesions and carcinogenesis. Furthermore, a model is presented (Lectin-triggered Receptor Blocking Hypothesis) explaining the role of the lectin TFF1 as an antral tumor suppressor possibly regulating Lgr5+ antral stem cells in a paracrine or maybe autocrine fashion, with neighboring antral gland cells having a role as niche cells.
Collapse
Affiliation(s)
- Werner Hoffmann
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| |
Collapse
|
14
|
Liabeuf D, Oshima M, Stange DE, Sigal M. Stem Cells, Helicobacter pylori, and Mutational Landscape: Utility of Preclinical Models to Understand Carcinogenesis and to Direct Management of Gastric Cancer. Gastroenterology 2022; 162:1067-1087. [PMID: 34942172 DOI: 10.1053/j.gastro.2021.12.252] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 12/20/2022]
Abstract
Several genetic and environmental factors increase gastric cancer (GC) risk, with Helicobacter pylori being the main environmental agent. GC is thought to emerge through a sequence of morphological changes that have been elucidated on the molecular level. New technologies have shed light onto pathways that are altered in GC, involving mutational and epigenetic changes and altered signaling pathways. Using various new model systems and innovative approaches, the relevance of such alterations for the emergence and progression of GC has been validated. Here, we highlight the key strategies and the resulting achievements. A major step is the characterization of epithelial stem cell behavior in the healthy stomach. These data, obtained through new reporter mouse lines and lineage tracing, enabled insights into the processes that control cellular proliferation, self-renewal, and differentiation of gastric stem cells. It has become evident that these cells and pathways are often deregulated in carcinogenesis. Second, insights into how H pylori colonizes gastric glands, directly interacts with stem cells, and alters cellular and genomic integrity, as well as the characterization of tissue responses to infection, provide a comprehensive picture of how this bacterium contributes to gastric carcinogenesis. Third, the development of stem cell- and tissue-specific reporter mice have driven our understanding of the signals and mutations that promote different types of GC and now also enable the study of more advanced, metastasized stages. Finally, organoids from human tissue have allowed insights into gastric carcinogenesis by validating mutational and signaling alterations in human primary cells and opening a route to predicting responses to personalized treatment.
Collapse
Affiliation(s)
- Dylan Liabeuf
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Masanobu Oshima
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Daniel E Stange
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany; National Center for Tumor Diseases (NCT/UCC), Dresden, Germany, German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Michael Sigal
- Department of Internal Medicine, Division of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Germany; Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
| |
Collapse
|
15
|
Morphogen Signals Shaping the Gastric Glands in Health and Disease. Int J Mol Sci 2022; 23:ijms23073632. [PMID: 35408991 PMCID: PMC8998987 DOI: 10.3390/ijms23073632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 12/17/2022] Open
Abstract
The adult gastric mucosa is characterised by deep invaginations of the epithelium called glands. These tissue architectural elements are maintained with the contribution of morphogen signals. Morphogens are expressed in specific areas of the tissue, and their diffusion generates gradients in the microenvironment. Cells at different positions in the gland sense a specific combination of signals that instruct them to differentiate, proliferate, regenerate, or migrate. Differentiated cells perform specific functions involved in digestion, such as the production of protective mucus and the secretion of digestive enzymes or gastric acid. Biopsies from gastric precancerous conditions usually display tissue aberrations and change the shape of the glands. Alteration of the morphogen signalling microenvironment is likely to underlie those conditions. Furthermore, genes involved in morphogen signalling pathways are found to be frequently mutated in gastric cancer. We summarise the most recent findings regarding alterations of morphogen signalling during gastric carcinogenesis, and we highlight the new stem cell technologies that are improving our understanding of the regulation of human tissue shape.
Collapse
|
16
|
Wang R, Wu Y, Zhu Y, Yao S, Zhu Y. ANKRD22 is a novel therapeutic target for gastric mucosal injury. Pharmacotherapy 2022; 147:112649. [PMID: 35051858 DOI: 10.1016/j.biopha.2022.112649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 11/28/2022]
|
17
|
Goldenring JR, Mills JC. Cellular Plasticity, Reprogramming, and Regeneration: Metaplasia in the Stomach and Beyond. Gastroenterology 2022; 162:415-430. [PMID: 34728185 PMCID: PMC8792220 DOI: 10.1053/j.gastro.2021.10.036] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 10/21/2021] [Accepted: 10/24/2021] [Indexed: 02/03/2023]
Abstract
The mucosa of the body of the stomach (ie, the gastric corpus) uses 2 overlapping, depth-dependent mechanisms to respond to injury. Superficial injury heals via surface cells with histopathologic changes like foveolar hyperplasia. Deeper, usually chronic, injury/inflammation, most frequently induced by the carcinogenic bacteria Helicobacter pylori, elicits glandular histopathologic alterations, initially manifesting as pyloric (also known as pseudopyloric) metaplasia. In this pyloric metaplasia, corpus glands become antrum (pylorus)-like with loss of acid-secreting parietal cells (atrophic gastritis), expansion of foveolar cells, and reprogramming of digestive enzyme-secreting chief cells into deep antral gland-like mucous cells. After acute parietal cell loss, chief cells can reprogram through an orderly stepwise progression (paligenosis) initiated by interleukin-13-secreting innate lymphoid cells (ILC2s). First, massive lysosomal activation helps mitigate reactive oxygen species and remove damaged organelles. Second, mucus and wound-healing proteins (eg, TFF2) and other transcriptional alterations are induced, at which point the reprogrammed chief cells are recognized as mucus-secreting spasmolytic polypeptide-expressing metaplasia cells. In chronic severe injury, glands with pyloric metaplasia can harbor both actively proliferating spasmolytic polypeptide-expressing metaplasia cells and eventually intestine-like cells. Gastric glands with such lineage confusion (mixed incomplete intestinal metaplasia and proliferative spasmolytic polypeptide-expressing metaplasia) may be at particular risk for progression to dysplasia and cancer. A pyloric-like pattern of metaplasia after injury also occurs in other gastrointestinal organs including esophagus, pancreas, and intestines, and the paligenosis program itself seems broadly conserved across tissues and species. Here we discuss aspects of metaplasia in stomach, incorporating data derived from animal models and work on human cells and tissues in correlation with diagnostic and clinical implications.
Collapse
Affiliation(s)
- James R Goldenring
- Nashville Veterans Affairs Medical Center, Vanderbilt University School of Medicine, Nashville, Tennessee; Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee; Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee.
| | - Jason C Mills
- Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas; Department of Medicine, Baylor College of Medicine, Houston, Texas; Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
18
|
Generation of Human Stomach Cancer iPSC-Derived Organoids Induced by Helicobacter pylori Infection and Their Application to Gastric Cancer Research. Cells 2022; 11:cells11020184. [PMID: 35053302 PMCID: PMC8773924 DOI: 10.3390/cells11020184] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/30/2021] [Accepted: 01/02/2022] [Indexed: 12/13/2022] Open
Abstract
There is considerable cellular diversity in the human stomach, which has helped to clarify cell plasticity in normal development and tumorigenesis. Thus, the stomach is an interesting model for understanding cellular plasticity and for developing prospective anticancer therapeutic agents. However, many questions remain regarding the development of cancers in vivo and in vitro in two- or three-dimensional (2D/3D) cultures, as well as the role of Helicobacter pylori (H. p.) infection. Here, we focus on the characteristics of cancer stem cells and their derived 3D organoids in culture, including the formation of stem cell niches. We define the conditions required for such organoid culture in vitro and examine the ability of such models for testing the use of anticancer agents. We also summarize the signaling cascades and the specific markers of stomach-cancer-derived organoids induced by H. p. infection, and their stem cell niches.
Collapse
|
19
|
Matsuo J, Douchi D, Myint K, Mon NN, Yamamura A, Kohu K, Heng DL, Chen S, Mawan NA, Nuttonmanit N, Li Y, Srivastava S, Ho SWT, Lee NYS, Lee HK, Adachi M, Tamura A, Chen J, Yang H, Teh M, So JBY, Yong WP, Tan P, Yeoh KG, Chuang LSH, Tsukita S, Ito Y. Iqgap3-Ras axis drives stem cell proliferation in the stomach corpus during homoeostasis and repair. Gut 2021; 70:1833-1846. [PMID: 33293280 PMCID: PMC8458072 DOI: 10.1136/gutjnl-2020-322779] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/13/2020] [Accepted: 11/14/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Tissue stem cells are central regulators of organ homoeostasis. We looked for a protein that is exclusively expressed and functionally involved in stem cell activity in rapidly proliferating isthmus stem cells in the stomach corpus. DESIGN We uncovered the specific expression of Iqgap3 in proliferating isthmus stem cells through immunofluorescence and in situ hybridisation. We performed lineage tracing and transcriptomic analysis of Iqgap3 +isthmus stem cells with the Iqgap3-2A-tdTomato mouse model. Depletion of Iqgap3 revealed its functional importance in maintenance and proliferation of stem cells. We further studied Iqgap3 expression and the associated gene expression changes during tissue repair after tamoxifen-induced damage. Immunohistochemistry revealed elevated expression of Iqgap3 in proliferating regions of gastric tumours from patient samples. RESULTS Iqgap3 is a highly specific marker of proliferating isthmus stem cells during homoeostasis. Iqgap3+isthmus stem cells give rise to major cell types of the corpus unit. Iqgap3 expression is essential for the maintenance of stem potential. The Ras pathway is a critical partner of Iqgap3 in promoting strong proliferation in isthmus stem cells. The robust induction of Iqgap3 expression following tissue damage indicates an active role for Iqgap3 in tissue regeneration. CONCLUSION IQGAP3 is a major regulator of stomach epithelial tissue homoeostasis and repair. The upregulation of IQGAP3 in gastric cancer suggests that IQGAP3 plays an important role in cancer cell proliferation.
Collapse
Affiliation(s)
- Junichi Matsuo
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Daisuke Douchi
- Cancer Science Institute of Singapore, National University of Singapore, Singapore,Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Khine Myint
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Naing Naing Mon
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Akihiro Yamamura
- Cancer Science Institute of Singapore, National University of Singapore, Singapore,Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuyoshi Kohu
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Dede Liana Heng
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Sabirah Chen
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Nur Astiana Mawan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Napat Nuttonmanit
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Ying Li
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | | | - Shamaine Wei Ting Ho
- Cancer Science Institute of Singapore, National University of Singapore, Singapore,Cancer and Stem Cell Biology Program, Duke-NUS Graduate Medical School, Singapore
| | - Nicole Yee Shin Lee
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Hong Kai Lee
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Makoto Adachi
- Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Atsushi Tamura
- Department of Pharmacology, School of Medicine, Teikyo University, Tokyo, Japan,Strategic Innovation and Research Center, Teikyo University, Tokyo, Japan,Laboratory of Barriology and Cell Biology, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Jinmiao Chen
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Henry Yang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Ming Teh
- Department of Pathology, National University of Singapore, Singapore
| | - Jimmy Bok-Yan So
- Department of Surgery, National University Health System, National University of Singapore, Singapore
| | - Wei Peng Yong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore,Department of Hematology-Oncology, National University Cancer Institute, Singapore
| | - Patrick Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore,Cancer and Stem Cell Biology Program, Duke-NUS Graduate Medical School, Singapore,Genome Institute of Singapore, Singapore
| | - Khay Guan Yeoh
- Department of Medicine, National University of Singapore, Singapore,Department of Gastroenterology and Hepatology, National University Hospital, Singapore
| | | | - Sachiko Tsukita
- Strategic Innovation and Research Center, Teikyo University, Tokyo, Japan .,Laboratory of Barriology and Cell Biology, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Yoshiaki Ito
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| |
Collapse
|
20
|
Mesquita da Silva K, Rattes IC, Pereira GMA, Gama P. Lifelong Adaptation of Gastric Cell Proliferation and Mucosa Structure to Early Weaning-Induced Effects. Front Physiol 2021; 12:721242. [PMID: 34588994 PMCID: PMC8475651 DOI: 10.3389/fphys.2021.721242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/12/2021] [Indexed: 11/13/2022] Open
Abstract
The gastric mucosa is disturbed when breastfeeding is interrupted, and such early weaning (EW) condition permanently affects the differentiation of zymogenic cells. The aim of the study was to evaluate the immediate and long-term effects of EW on gastric cell proliferation, considering the molecular markers for cell cycle, inflammation, and metaplasia. Overall, we investigated the lifelong adaptation of gastric growth. Wistar rats were divided into suckling-control (S) and EW groups, and gastric samples were collected at 18, 30, and 60 days for morphology, RNA, and protein isolation. Inflammation and metaplasia were not identified, but we observed that EW promptly increased Ki-67-proliferative index (PI) and mucosa thickness (18 days). From 18 to 30 days, PI increased in S rats, whereas it was stable in EW animals, and such developmental change in S made its PI higher than in EW. At 60 days, the PI decreased in S, making the indices similar between groups. Spatially, during development, proliferative cells spread along the gland, whereas, in adults, they concentrate at the isthmus-neck area. EW pushed dividing cells to this compartment (18 days), increased PI at the gland base (60 days), but it did not interfere in expression of cell cycle molecules. At 18 days, EW reduced Tgfβ2, Tgfβ3, and Tgfbr2 and TβRII and p27 levels, which might regulate the proliferative increase at this age. We demonstrated that gastric cell proliferation is immediately upregulated by EW, corroborating previous results, but for the first time, we showed that such increased PI is stable during growth and aging. We suggest that suckling and early weaning might use TGFβs and p27 to trigger different proliferative profiles during life course.
Collapse
Affiliation(s)
- Kethleen Mesquita da Silva
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Isadora Campos Rattes
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Gizela Maria Agostini Pereira
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Patrícia Gama
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
21
|
Wölffling S, Daddi AA, Imai-Matsushima A, Fritsche K, Goosmann C, Traulsen J, Lisle R, Schmid M, Reines-Benassar MDM, Pfannkuch L, Brinkmann V, Bornschein J, Malfertheiner P, Ordemann J, Link A, Meyer TF, Boccellato F. EGF and BMPs Govern Differentiation and Patterning in Human Gastric Glands. Gastroenterology 2021; 161:623-636.e16. [PMID: 33957136 DOI: 10.1053/j.gastro.2021.04.062] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/22/2021] [Accepted: 04/24/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS The homeostasis of the gastrointestinal epithelium relies on cell regeneration and differentiation into distinct lineages organized inside glands and crypts. Regeneration depends on Wnt/β-catenin pathway activation, but to understand homeostasis and its dysregulation in disease, we need to identify the signaling microenvironment governing cell differentiation. By using gastric glands as a model, we have identified the signals inducing differentiation of surface mucus-, zymogen-, and gastric acid-producing cells. METHODS We generated mucosoid cultures from the human stomach and exposed them to different growth factors to obtain cells with features of differentiated foveolar, chief, and parietal cells. We localized the source of the growth factors in the tissue of origin. RESULTS We show that epidermal growth factor is the major fate determinant distinguishing the surface and inner part of human gastric glands. In combination with bone morphogenetic factor/Noggin signals, epidermal growth factor controls the differentiation of foveolar cells vs parietal or chief cells. We also show that epidermal growth factor is likely to underlie alteration of the gastric mucosa in the precancerous condition atrophic gastritis. CONCLUSIONS Use of our recently established mucosoid cultures in combination with analysis of the tissue of origin provided a robust strategy to understand differentiation and patterning of human tissue and allowed us to draw a new, detailed map of the signaling microenvironment in the human gastric glands.
Collapse
Affiliation(s)
- Sarah Wölffling
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Alice Anna Daddi
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, United Kingdom
| | - Aki Imai-Matsushima
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany; Preemptive Medicine and Lifestyle-Related Diseases Research Center, Kyoto University Hospital, Kyoto, Japan
| | - Kristin Fritsche
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Christian Goosmann
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Jan Traulsen
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, United Kingdom
| | - Richard Lisle
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, United Kingdom
| | - Monika Schmid
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | | | - Lennart Pfannkuch
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Volker Brinkmann
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Jan Bornschein
- Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford University Hospitals, Oxford, United Kingdom; Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Peter Malfertheiner
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Jürgen Ordemann
- Department of Bariatric and Metabolic Surgery, Helios Klinikum, Berlin, Germany; Center for Bariatric and Metabolic Surgery, Vivantes Klinikum Spandau, Berlin, Germany
| | - Alexander Link
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Thomas F Meyer
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany; Laboratory of Infection Oncology, Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel and University Hospital Schleswig-Holstein, Kiel, Germany.
| | - Francesco Boccellato
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany; Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, United Kingdom.
| |
Collapse
|
22
|
Hayakawa Y, Nakagawa H, Rustgi AK, Que J, Wang TC. Stem cells and origins of cancer in the upper gastrointestinal tract. Cell Stem Cell 2021; 28:1343-1361. [PMID: 34129814 DOI: 10.1016/j.stem.2021.05.012] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The esophagus and stomach, joined by a unique transitional zone, contain actively dividing epithelial stem cells required for organ homeostasis. Upon prolonged inflammation, epithelial cells in both organs can undergo a cell fate switch leading to intestinal metaplasia, predisposing to malignancy. Here we discuss the biology of gastroesophageal stem cells and their role as cells of origin in cancer. We summarize the interactions between the stromal niche and gastroesophageal stem cells in metaplasia and early expansion of mutated stem-cell-derived clones during carcinogenesis. Finally, we review new approaches under development to better study gastroesophageal stem cells and advance the field.
Collapse
Affiliation(s)
- Yoku Hayakawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyoku, Tokyo 113-8655, Japan
| | - Hiroshi Nakagawa
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, College of Physicians and Surgeons, 1130 St. Nicholas Avenue, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, 1130 St. Nicholas Avenue, New York, NY 10032, USA
| | - Anil K Rustgi
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, College of Physicians and Surgeons, 1130 St. Nicholas Avenue, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, 1130 St. Nicholas Avenue, New York, NY 10032, USA
| | - Jianwen Que
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, College of Physicians and Surgeons, 1130 St. Nicholas Avenue, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, 1130 St. Nicholas Avenue, New York, NY 10032, USA; Columbia Center for Human Development, Department of Medicine, Columbia University, College of Physicians and Surgeons, 1130 St. Nicholas Avenue, New York, NY 10032, USA.
| | - Timothy C Wang
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, College of Physicians and Surgeons, 1130 St. Nicholas Avenue, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, 1130 St. Nicholas Avenue, New York, NY 10032, USA.
| |
Collapse
|
23
|
Traulsen J, Zagami C, Daddi AA, Boccellato F. Molecular modelling of the gastric barrier response, from infection to carcinogenesis. Best Pract Res Clin Gastroenterol 2021; 50-51:101737. [PMID: 33975688 DOI: 10.1016/j.bpg.2021.101737] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/15/2021] [Accepted: 02/18/2021] [Indexed: 02/07/2023]
Abstract
The lining of the stomach is a tight monolayer of epithelial cells performing functions in digestion and a protective barrier against gastric acid, toxic metabolites and infectious agents, including Helicobacter pylori. The response of the epithelial barrier to infections underlies gastric pathologies, including gastric cancer. H. pylori has the unique capacity to colonise the gastric mucosa while evading the immune system. The colonised mucosa initiates an inflammatory response to fight the infection and a strong regenerative program to avoid barrier failure and ulceration. This response changes the morphology and cell composition of the gastric epithelium and in parallel it might contribute to the accumulation of somatic mutations leading to cellular transformation. Genetically modified mice, cell lines and human-derived organoids are the main biological models to study the gastric epithelial barrier. With these models it is possible to dissect the stepwise process of tissue adaptation to infection that places the epithelium at risk of malignant transformation.
Collapse
Affiliation(s)
- Jan Traulsen
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, United Kingdom.
| | - Claudia Zagami
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, United Kingdom.
| | - Alice Anna Daddi
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, United Kingdom.
| | - Francesco Boccellato
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, United Kingdom.
| |
Collapse
|
24
|
Dysregulated Immune Responses by ASK1 Deficiency Alter Epithelial Progenitor Cell Fate and Accelerate Metaplasia Development during H. pylori Infection. Microorganisms 2020; 8:microorganisms8121995. [PMID: 33542169 PMCID: PMC7765114 DOI: 10.3390/microorganisms8121995] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/11/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023] Open
Abstract
The mechanism of H. pylori-induced atrophy and metaplasia has not been fully understood. Here, we demonstrate the novel role of Apoptosis signal-regulating kinase 1 (ASK1) and downstream MAPKs as a regulator of host immune responses and epithelial maintenance against H. pylori infection. ASK1 gene deficiency resulted in enhanced inflammation with numerous inflammatory cells including Gr-1+CD11b+ myeloid-derived suppressor cells (MDSCs) recruited into the infected stomach. Increase of IL-1β release from apoptotic macrophages and enhancement of TH1-polarized immune responses caused STAT1 and NF-κB activation in epithelial cells in ASK1 knockout mice. Dysregulated immune and epithelial activation in ASK1 knockout mice led to dramatic expansion of gastric progenitor cells and massive metaplasia development. Bone marrow transplantation experiments revealed that ASK1 in inflammatory cells is critical for inducing immune disorder and metaplastic changes in epithelium, while ASK1 in epithelial cells regulates cell proliferation in stem/progenitor zone without changes in inflammation and differentiation. These results suggest that H. pylori-induced immune cells may regulate epithelial homeostasis and cell fate as an inflammatory niche via ASK1 signaling.
Collapse
|
25
|
Kanetaka K, Eguchi S. Regenerative medicine for the upper gastrointestinal tract. Regen Ther 2020; 15:129-137. [PMID: 33426211 PMCID: PMC7770370 DOI: 10.1016/j.reth.2020.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/21/2020] [Accepted: 07/01/2020] [Indexed: 12/12/2022] Open
Abstract
The main surgical strategy for gastrointestinal tract malignancy is en bloc resection, which consists of not only resection of the involved organs but also simultaneous resection of the surrounding or adjacent mesenteries that contain lymph vessels and nodes. After resection of the diseased organs, the defect of the gastrointestinal conduit is replaced with organs located downstream, such as the stomach and jejunum. However, esophageal and gastric reconstruction using these natural substitutes is associated with a diminished quality of life due to the loss of the reserve function, damage to the antireflux barrier, and dumping syndrome. Thus, replacement of the deficit after resection with the patient's own regenerated tissue to compensate for the lost function and tissue using regenerative medicine will be an ideal treatment. Many researchers have been trying to construct artificial organs through tissue engineering techniques; however, none have yet succeeded in growing a whole organ because of the complicated functions these organs perform, such as the processing and absorption of nutrients. While exciting results have been reported with regard to tissue engineering techniques concerning the upper gastrointestinal tract, such as the esophagus and stomach, most of these achievements have been observed in animal models, and few successful approaches in the clinical setting have been reported for the replacement of mucosal defects. We review the recent progress in regenerative medicine in relation to the upper gastrointestinal tract, such as the esophagus and stomach. We also focus on the functional capacity of regenerated tissue and its role as a culture system to recapitulate the mechanisms underlying infectious disease. With the emergence of technology such as the fabrication of decellularized constructs, organoids and cell sheet medicine, collaboration between gastrointestinal surgery and regenerative medicine is expected to help establish novel therapeutic modalities in the future.
Collapse
Affiliation(s)
- Kengo Kanetaka
- Tissue Engineering and Regenerative Therapeutics in Gastrointestinal Surgery, Nagasaki University Graduate School of Biomedical Sciences, Japan
| | - Susumu Eguchi
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Japan
| |
Collapse
|
26
|
Xiao S, Zhou L. Gastric Stem Cells: Physiological and Pathological Perspectives. Front Cell Dev Biol 2020; 8:571536. [PMID: 33043003 PMCID: PMC7527738 DOI: 10.3389/fcell.2020.571536] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/20/2020] [Indexed: 12/24/2022] Open
Abstract
Gastric epithelium operates in a hazardous environment that curtails the lifespan of the constituent cells, imposing a requirement for continuous epithelial renewal. Stem cells that reside in the stomach are thus essential for regulating physiological tissue renewal and injury repair because of their self-renewal, high proliferation capacity and multiple differentiation potentials. Recent investigations using lineage tracing models have identified diverse populations of gastric stem cells and even fully differentiated cells that can regain stem cell capacity, so enriching our understanding on the identity and plasticity of gastric stem cells. These cell populations include the Villin promotor, Lgr5+, CCKR2+, Axin2+ and AQP5+ stem cells in the antrum, TFF2 mRNA, Mist1+ cells and Troy+ mature chief cells in the corpus, as well as Sox2, eR1, Lrig1, Bmi1-marked cell in both the antrum and the corpus. Establishment of gastric organoids derived from primary gastric tissues and pluripotent stem cells or embryonic stem cells characterizes niche factors required by the gastric stem cell populations, and further provides new insights into stomach development, host-Helicobacter pylori interactions and malignant transformation. Furthermore, focus on the gastric stem cells and their niches uncovers the initiation of stomach precancerous lesions and origin of gastric cancer, providing options for cancer prevention and intervention. In summary, with the development of stem cell research, gastric stem cells give us more opportunities to prevent and treat stomach diseases.
Collapse
Affiliation(s)
- Shiyu Xiao
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Peking University Third Hospital, Beijing, China
| | - Liya Zhou
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Peking University Third Hospital, Beijing, China
| |
Collapse
|
27
|
Sayols S, Klassek J, Werner C, Möckel S, Ritz S, Mendez-Lago M, Soshnikova N. Signalling codes for the maintenance and lineage commitment of embryonic gastric epithelial progenitors. Development 2020; 147:dev.188839. [PMID: 32878924 DOI: 10.1242/dev.188839] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 08/11/2020] [Indexed: 12/16/2022]
Abstract
The identity of embryonic gastric epithelial progenitors is unknown. We used single-cell RNA-sequencing, genetic lineage tracing and organoid assays to assess whether Axin2- and Lgr5-expressing cells are gastric progenitors in the developing mouse stomach. We show that Axin2 + cells represent a transient population of embryonic epithelial cells in the forestomach. Lgr5 + cells generate both glandular corpus and squamous forestomach organoids ex vivo Only Lgr5 + progenitors give rise to zymogenic cells in culture. Modulating the activity of the WNT, BMP and Notch pathways in vivo and ex vivo, we found that WNTs are essential for the maintenance of Lgr5 + epithelial cells. Notch prevents differentiation of the embryonic epithelial cells along all secretory lineages and hence ensures their maintenance. Whereas WNTs promote differentiation of the embryonic progenitors along the zymogenic cell lineage, BMPs enhance their differentiation along the parietal lineage. In contrast, WNTs and BMPs are required to suppress differentiation of embryonic gastric epithelium along the pit cell lineage. Thus, coordinated action of the WNT, BMP and Notch pathways controls cell fate determination in the embryonic gastric epithelium.
Collapse
Affiliation(s)
- Sergi Sayols
- Institute of Molecular Biology gGmbH, Mainz 55128, Germany
| | - Jakub Klassek
- Institute of Molecular Biology gGmbH, Mainz 55128, Germany
| | - Clara Werner
- Institute of Molecular Biology gGmbH, Mainz 55128, Germany
| | | | - Sandra Ritz
- Institute of Molecular Biology gGmbH, Mainz 55128, Germany
| | | | - Natalia Soshnikova
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University, Mainz 55131, Germany
| |
Collapse
|
28
|
Sheng W, Malagola E, Nienhüser H, Zhang Z, Kim W, Zamechek L, Sepulveda A, Hata M, Hayakawa Y, Zhao CM, Chen D, Wang TC. Hypergastrinemia Expands Gastric ECL Cells Through CCK2R + Progenitor Cells via ERK Activation. Cell Mol Gastroenterol Hepatol 2020; 10:434-449.e1. [PMID: 32330731 PMCID: PMC7371950 DOI: 10.1016/j.jcmgh.2020.04.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 04/11/2020] [Accepted: 04/13/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Enterochromaffin-like (ECL) cells in the stomach express gastrin/cholecystokinin 2 receptor CCK2R and are known to expand under hypergastrinemia, but whether this results from expansion of existing ECL cells or increased production from progenitors has not been clarified. METHODS We used mice with green fluorescent protein fluorescent reporter expression in ECL cells (histidine decarboxylase [Hdc]-green fluorescent protein), as well as Cck2r- and Hdc-driven Tamoxifen inducible recombinase Cre (Cck2r-CreERT2, Hdc-CreERT2) mice combined with Rosa26Sor-tdTomato (R26-tdTomato) mice, and studied their expression and cell fate in the gastric corpus by using models of hypergastrinemia (gastrin infusion, omeprazole treatment). RESULTS Hdc-GFP marked the majority of ECL cells, located in the lower third of the gastric glands. Hypergastrinemia led to expansion of ECL cells that was not restricted to the gland base, and promoted cellular proliferation (Ki67) in the gastric isthmus but not in basal ECL cells. Cck2r-CreERT2 mice marked most ECL cells, as well as scattered cell types located higher up in the glands, whose number was increased during hypergastrinemia. Cck2r-CreERT2+ isthmus progenitors, but not Hdc+ mature ECL cells, were the source of ECL cell hyperplasia during hypergastrinemia and could grow as 3-dimensional spheroids in vitro. Moreover, gastrin treatment in vitro promoted sphere formation from sorted Cck2r+Hdc- cells, and increased chromogranin A and phosphorylated- extracellular signal-regulated kinase expression in CCK2R-derived organoids. Gastrin activates extracellular signal-regulated kinase pathways in vivo and in vitro, and treatment with the Mitogen-activated protein kinase kinase 1 inhibitor U0126 blocked hypergastrinemia-mediated changes, including CCK2R-derived ECL cell hyperplasia in vivo as well as sphere formation and chromogranin A expression in vitro. CONCLUSIONS We show here that hypergastrinemia induces ECL cell hyperplasia that is derived primarily from CCK2R+ progenitors in the corpus. Gastrin-dependent function of CCK2R+ progenitors is regulated by the extracellular signal-regulated kinase pathway.
Collapse
Affiliation(s)
- Weiwei Sheng
- Division of Digestive and Liver Diseases, Department of Medicine,Department of Gastrointestinal Surgery, the First Hospital, China Medical University, Shenyang, China
| | - Ermanno Malagola
- Division of Digestive and Liver Diseases, Department of Medicine
| | - Henrik Nienhüser
- Division of Digestive and Liver Diseases, Department of Medicine
| | - Zhengyu Zhang
- Division of Digestive and Liver Diseases, Department of Medicine
| | - Woosook Kim
- Division of Digestive and Liver Diseases, Department of Medicine
| | - Leah Zamechek
- Division of Digestive and Liver Diseases, Department of Medicine
| | - Antonia Sepulveda
- Department of Pathology, Columbia University, College of Physicians and Surgeons, New York, New York
| | - Masahiro Hata
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoku Hayakawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Chun-Mei Zhao
- Department of Clinical and Molecular Medicine (Institutt for klinisk og molekylær medisin), Norwegian University of Science and Technology (Norges teknisk-naturvitenskaplige universitet), Trondheim, Norway
| | - Duan Chen
- Department of Clinical and Molecular Medicine (Institutt for klinisk og molekylær medisin), Norwegian University of Science and Technology (Norges teknisk-naturvitenskaplige universitet), Trondheim, Norway
| | - Timothy C. Wang
- Division of Digestive and Liver Diseases, Department of Medicine,Correspondence Address correspondence to: Timothy C. Wang, MD, Division of Digestive and Liver Disease, Department of Medicine, Columbia University Medical Center, New York, New York; fax: (212) 851-4590.
| |
Collapse
|
29
|
Wizenty J, Tacke F, Sigal M. Responses of gastric epithelial stem cells and their niche to Helicobacter pylori infection. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:568. [PMID: 32775369 PMCID: PMC7347775 DOI: 10.21037/atm.2020.02.178] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Helicobacter pylori (H. pylori) are gram-negative bacteria that are able to colonize and persist in the stomach. Gastric cancer is tightly linked to chronic infection with this bacterium. Research over the last decades has illuminated the molecular interactions between H. pylori and host cells. It is now well established that H. pylori have multiple sophisticated means to adhere to epithelial cells and to manipulate their behavior. This interaction with the epithelium can lead to altered cell signaling, DNA damage and aberrant epithelial immunity. H. pylori are known to colonize the mucus layer of the stomach and surface epithelial cells. In addition, it has recently become clear that they can also penetrate the glands and directly interact with specialized epithelial cells deep in the glands. Understanding the biogeography of infection is important because gastric epithelial glands are composed of various types of short-lived differentiated cells that are constantly regenerated by a limited pool of long-lived stem cells located in base of gastric glands. Recent advances in gastric stem cell research not only led to identification of stem cell populations using specific markers but has also uncovered specific regulatory pathways and principles that govern gastric stem cell behavior and regeneration. Particularly, the stem cell state is largely dependent on signals from the niche cells that surround the stem cell compartment. The subpopulation of H. pylori that colonizes in the stem cell compartment triggers specific inflammatory responses and drives epithelial pathology. Colonization of gastric glands induces responses of the stem cell niche, simultaneously enhancing the cell turnover kinetics and driving the formation of antimicrobial cells in the gland base. These data reveal the high plasticity of the epithelium and its ability to adapt to the environment, which is necessary to regenerate and counterbalance infection, but simultaneously lays the grounds for development of gastric pathology and carcinogenesis.
Collapse
Affiliation(s)
- Jonas Wizenty
- Department of Hepatology & Gastroenterology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Sigal
- Department of Hepatology & Gastroenterology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| |
Collapse
|
30
|
Wang R, Zeng X, Liu B, Yi R, Zhou X, Mu J, Zhao X. Prophylactic effect of Lactobacillus plantarum KSFY06 on HCl/ethanol-induced gastric injury in mice. Food Funct 2020; 11:2679-2692. [PMID: 32162630 DOI: 10.1039/c9fo02474c] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
The present study was conducted to determine the prophylactic effect of Lactobacillus plantarum KSFY06 (LP-KSFY06) on HCl/ethanol-induced gastric injury in Kunming mice. The experimental mice were allocated into six groups: the normal group, HCl/ethanol treated group, HCl/ethanol + ranitidine treated group, HCl/ethanol + Lactobacillus delbrueckii subsp. Bulgaricus (LB) treated group, HCl/ethanol + low concentration of Lactobacillus plantans KSFY06 (LP-KSFY06-L) treated group, and HCl/ethanol + high concentration of Lactobacillus plantans KSFY06 (LP-KSFY06-H) treated group. The changes in daily body weight and food intake of the mice in the HCl/ethanol + LP-KSFY06-H treated group were the closest to those of the HCl/ethanol + ranitidine treated and normal groups. LP-KSFY06 significantly inhibited the formation of gastric mucosal lesions, reduced the area of gastric lesions, inhibited gastric-juice secretion, and increased pH compared with the HCl/ethanol treated group. After the treatment, the serum interleukin-6 (IL)-6, IL-12, tumor necrosis factor-α (TNF-α), and interferon-γ levels and the gastric-tissue IL-6 and IL-12 levels in the LP-KSFY06 (including LP-KSFY06-L and LP-KSFY06-H) group decreased compared with those in the HCl/ethanol treated group. The level of serum and gastric tissue malondialdehyde was lower and the nitric oxide, total superoxide dismutase, and glutathione activities in the LP-KSFY06 treated mice were higher than those in the HCl/ethanol treated mice. Quantitative polymerase chain reaction analysis and western blot analysis showed that LP-KSFY06 increased the mRNA and protein expression of the epidermal growth factor, epidermal growth factor receptor, vascular endothelial growth factor, inhibitor kappaB-α, neuronal nitric oxide synthase, and endothelial NOS and reduced the mRNA and protein expression of nuclear factor kappaB, inducible NOS, cyclooxygenase-2, TNF-α, and IL-1β in gastric tissues compared with the HCl/ethanol treated mice. These experimental results showed that a high concentration (1.0 × 109 CFU per kg B.W.) of LP-KSFY06 had a stronger effect on preventing gastric injury than a low concentration (1.0 × 108 CFU per kg B.W.) of LP-KSFY06. These results suggest that LP-KSFY06 has a potential probiotic effect in preventing gastric injury.
Collapse
Affiliation(s)
- Ranran Wang
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China. and Chongqing Engineering Research Center of Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China and Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China and College of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, P.R. China
| | - Xiaofei Zeng
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, P.R. China
| | - Bihui Liu
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China. and Chongqing Engineering Research Center of Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China and Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China and College of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, P.R. China
| | - Ruokun Yi
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China. and Chongqing Engineering Research Center of Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China and Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China and College of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, P.R. China
| | - Xianrong Zhou
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China. and Chongqing Engineering Research Center of Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China and Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China
| | - Jianfei Mu
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China. and Chongqing Engineering Research Center of Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China and Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China
| | - Xin Zhao
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China. and Chongqing Engineering Research Center of Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China and Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China and College of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, P.R. China
| |
Collapse
|
31
|
Han S, Fink J, Jörg DJ, Lee E, Yum MK, Chatzeli L, Merker SR, Josserand M, Trendafilova T, Andersson-Rolf A, Dabrowska C, Kim H, Naumann R, Lee JH, Sasaki N, Mort RL, Basak O, Clevers H, Stange DE, Philpott A, Kim JK, Simons BD, Koo BK. Defining the Identity and Dynamics of Adult Gastric Isthmus Stem Cells. Cell Stem Cell 2019; 25:342-356.e7. [PMID: 31422913 PMCID: PMC6739486 DOI: 10.1016/j.stem.2019.07.008] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 04/11/2019] [Accepted: 07/18/2019] [Indexed: 12/13/2022]
Abstract
The gastric corpus epithelium is the thickest part of the gastrointestinal tract and is rapidly turned over. Several markers have been proposed for gastric corpus stem cells in both isthmus and base regions. However, the identity of isthmus stem cells (IsthSCs) and the interaction between distinct stem cell populations is still under debate. Here, based on unbiased genetic labeling and biophysical modeling, we show that corpus glands are compartmentalized into two independent zones, with slow-cycling stem cells maintaining the base and actively cycling stem cells maintaining the pit-isthmus-neck region through a process of "punctuated" neutral drift dynamics. Independent lineage tracing based on Stmn1 and Ki67 expression confirmed that rapidly cycling IsthSCs maintain the pit-isthmus-neck region. Finally, single-cell RNA sequencing (RNA-seq) analysis is used to define the molecular identity and lineage relationship of a single, cycling, IsthSC population. These observations define the identity and functional behavior of IsthSCs.
Collapse
Affiliation(s)
- Seungmin Han
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, UK; The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK
| | - Juergen Fink
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, UK; Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | - David J Jörg
- The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK; Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, University of Cambridge, Wilberforce Road, Cambridge CB3 0WA, UK
| | - Eunmin Lee
- Department of New Biology, DGIST, Daegu 42988, Republic of Korea
| | - Min Kyu Yum
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, UK; The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK
| | - Lemonia Chatzeli
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, UK; The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK
| | - Sebastian R Merker
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Manon Josserand
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, UK
| | - Teodora Trendafilova
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, UK
| | - Amanda Andersson-Rolf
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, UK; Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | - Catherine Dabrowska
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, UK
| | - Hyunki Kim
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, UK
| | - Ronald Naumann
- MPI of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
| | - Ji-Hyun Lee
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Nobuo Sasaki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Richard Lester Mort
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Furness Building, Lancaster University, Bailrigg, Lancaster LA1 4YG, UK
| | - Onur Basak
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, 3584 Utrecht, the Netherlands
| | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, 3584 Utrecht, the Netherlands
| | - Daniel E Stange
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Anna Philpott
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, UK; Department of Oncology, Hutchison/MRC Research Centre, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XZ, UK
| | - Jong Kyoung Kim
- Department of New Biology, DGIST, Daegu 42988, Republic of Korea.
| | - Benjamin D Simons
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, UK; The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK; Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, University of Cambridge, Wilberforce Road, Cambridge CB3 0WA, UK.
| | - Bon-Kyoung Koo
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, UK; Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK; Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria.
| |
Collapse
|