1
|
Lynch EB, Kapur N, Goretsky T, Bradford EM, Vekaria H, Bhogoju S, Hassan SA, Pauw E, Avdiushko MG, Lee G, Gao T, Sullivan PG, Barrett TA. Phosphatidylinositol 3-Kinase Signaling Enhances Intestinal Crypt Epithelial Cell Recovery after Radiation. THE AMERICAN JOURNAL OF PATHOLOGY 2025:S0002-9440(25)00151-8. [PMID: 40316215 DOI: 10.1016/j.ajpath.2025.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 03/24/2025] [Accepted: 04/10/2025] [Indexed: 05/04/2025]
Abstract
Intestinal stem cell (ISC) signaling maintains the balance of self-renewal and differentiation. The role of phosphatidylinositol 3-kinase (PI3K) signaling in ISC responses to radiation was interrogated using Villin-Cre pik3r1lox/lox (p85ΔIEC) mice and p85α-deficient human enteroids (shp85α). Lethal whole-body irradiation in mice was performed to monitor PI3K-mediated survival responses. Rectal biopsies from patients with radiation proctitis were examined by immunohistochemistry for the PI3K/Akt- and Wnt-target survivin. The intestinal epithelial cells (IECs) from p85ΔIEC mice showed increased protein levels of phosphorylated phosphatase and tensin homolog, phosphorylated AktSer473, survivin, cyclin D1, and ρ-β-cateninSer552, as well as increased mRNA for ISC/progenitor cell. In situ hybridization showed that enhanced PI3K signaling reduced Lgr5+ cells but expansion of Axin2+ cells. The shp85α enteroids showed increased mRNA expression of Wnt targets and transcription factor ASCL2, needed for dedifferentiation-mediated restoration of ablated ISCs. The p85α-deficient enteroids showed reduced HES1 mRNA and increases in secretory (ATOH1/MATH1) signaling determinants GFI1 and SPDEF, indicative of reduced NOTCH signaling. Seahorse analyses and phosphorylated p38 staining in IECΔp85 mice indicated that enhanced PI3K signaling led to increased IEC mitochondrial respiration and reactive oxygen species generation. Expression of survivin correlated with the radiation injury in patients. The current data indicate that PI3K signaling increases mitochondrial reactive oxygen species generation and ISC activation that improves IEC recovery from radiation-induced injury. The results suggest that increasing PI3K signaling and induced mitochondrial respiration may improve mucosal healing from radiation injury in patients.
Collapse
Affiliation(s)
- Evan B Lynch
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, Kentucky; Division of Plastic Surgery, Department of Surgery, University of Kentucky, Lexington, Kentucky; Division of Digestive Diseases and Nutrition, Department of Medicine, University of Kentucky, Lexington, Kentucky
| | - Neeraj Kapur
- Division of Digestive Diseases and Nutrition, Department of Medicine, University of Kentucky, Lexington, Kentucky; Lexington VA Healthcare System, Lexington, Kentucky
| | - Tatiana Goretsky
- Division of Digestive Diseases and Nutrition, Department of Medicine, University of Kentucky, Lexington, Kentucky; Lexington VA Healthcare System, Lexington, Kentucky
| | - Emily M Bradford
- Division of Digestive Diseases and Nutrition, Department of Medicine, University of Kentucky, Lexington, Kentucky; Lexington VA Healthcare System, Lexington, Kentucky
| | - Hemendra Vekaria
- Lexington VA Healthcare System, Lexington, Kentucky; Department of Neuroscience, University of Kentucky, Lexington, Kentucky
| | - Sarayu Bhogoju
- Division of Digestive Diseases and Nutrition, Department of Medicine, University of Kentucky, Lexington, Kentucky
| | - Syed A Hassan
- Division of Digestive Diseases and Nutrition, Department of Medicine, University of Kentucky, Lexington, Kentucky; Lexington VA Healthcare System, Lexington, Kentucky
| | - Emily Pauw
- College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Margarita G Avdiushko
- Division of Digestive Diseases and Nutrition, Department of Medicine, University of Kentucky, Lexington, Kentucky; Lexington VA Healthcare System, Lexington, Kentucky
| | - Goo Lee
- The University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, Alabama
| | - Tianyan Gao
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky
| | - Patrick G Sullivan
- Lexington VA Healthcare System, Lexington, Kentucky; Department of Neuroscience, University of Kentucky, Lexington, Kentucky
| | - Terrence A Barrett
- Division of Digestive Diseases and Nutrition, Department of Medicine, University of Kentucky, Lexington, Kentucky; Lexington VA Healthcare System, Lexington, Kentucky.
| |
Collapse
|
2
|
Smith NR, Giske NR, Sengupta SK, Conley P, Swain JR, Nair A, Fowler KL, Klocke C, Yoo YJ, Anderson AN, Sanati N, Torkenczy K, Adey AC, Fischer JM, Wu G, Wong MH. Dual states of murine Bmi1-expressing intestinal stem cells drive epithelial development utilizing non-canonical Wnt signaling. Dev Cell 2025:S1534-5807(25)00177-7. [PMID: 40262610 DOI: 10.1016/j.devcel.2025.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 11/07/2024] [Accepted: 03/28/2025] [Indexed: 04/24/2025]
Abstract
Intestinal epithelial development and homeostasis critically rely upon balanced stem cell proliferation, involving slow-cycling/label-retaining and active-cycling/canonical Wnt-dependent intestinal stem cell (ISC) subtypes. ISC regulation during development remains poorly understood but has important implications for establishing key mechanisms governing tissue maintenance. Herein, we identify Bmi1+ cells as functional stem cells present in early murine intestinal development, prior to Lgr5-expressing ISCs. Lineage tracing and single-cell RNA sequencing identify that Bmi1+ ISCs can trace to Lgr5+ ISCs and other differentiated lineages. Initially highly proliferative, Bmi1+ ISCs transition to slow-cycling states as Lgr5+ ISCs emerge. Non-canonical Wnt signaling regulates the proliferative Bmi1+ cell state. These findings highlight the dynamic interplay between stem cell populations and the opposing Wnt pathways that govern proliferation-ultimately having implications for tissue development, homeostasis, regeneration, and tumorigenesis. Understanding these fundamental developmental mechanisms is critical for understanding adult intestinal maintenance.
Collapse
Affiliation(s)
- Nicholas R Smith
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97201, USA
| | - Nicole R Giske
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97201, USA
| | - Sidharth K Sengupta
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97201, USA
| | - Patrick Conley
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR 97239, USA
| | - John R Swain
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97201, USA
| | - Ashvin Nair
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97201, USA
| | - Kathryn L Fowler
- Department of Surgery, Oregon Health & Science University, Portland, OR 97239, USA
| | - Christopher Klocke
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Yeon Jung Yoo
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97201, USA
| | - Ashley N Anderson
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97201, USA
| | - Nasim Sanati
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Kristof Torkenczy
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA
| | - Andrew C Adey
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA; The Knight Cancer Institute, Oregon Health & Science University, Oregon Health & Science University, Portland, OR 97201, USA
| | - Jared M Fischer
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA; Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR 97201 USA; The Knight Cancer Institute, Oregon Health & Science University, Oregon Health & Science University, Portland, OR 97201, USA
| | - Guanming Wu
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR 97239, USA; The Knight Cancer Institute, Oregon Health & Science University, Oregon Health & Science University, Portland, OR 97201, USA
| | - Melissa H Wong
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97201, USA; The Knight Cancer Institute, Oregon Health & Science University, Oregon Health & Science University, Portland, OR 97201, USA.
| |
Collapse
|
3
|
Marefati M, Fernandez-Vallone V, Leprovots M, Vasile G, Libert F, Lefort A, Dinsart G, Weber A, Jetzer J, Garcia MI, Vassart G. A Lgr5-independent developmental lineage is involved in mouse intestinal regeneration. Development 2025; 152:dev204654. [PMID: 40013494 PMCID: PMC12045596 DOI: 10.1242/dev.204654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 02/06/2025] [Indexed: 02/28/2025]
Abstract
Collagenase and dispase treatment of intestinal tissue from adult mice generates cells growing in matrigel as stably replatable cystic spheroids, in addition to differentiated organoids. Contrary to classical EDTA-derived organoids, these spheroids display poor intestinal differentiation and grow independently of Rspondin, noggin and EGF. Their transcriptome strikingly resembles that of fetal intestinal spheroids, with downregulation of crypt base columnar cell (CBC) markers (Lgr5, Ascl2, Smoc2 and Olfm4). In addition, they display upregulation of inflammatory and mesenchymal genetic programs, together with robust expression of YAP target genes. Lineage tracing, cell-sorting and single cell RNA sequencing experiments demonstrate that adult spheroid-generating cells belong to a hitherto undescribed developmental lineage, independent of Lgr5-positive CBCs, and are involved in regeneration of the epithelium following CBC ablation.
Collapse
Affiliation(s)
- Maryam Marefati
- Institute of Interdisciplinary Research in Molecular Human Biology (IRIBHM, https://iribhm.org/), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Valeria Fernandez-Vallone
- Institute of Interdisciplinary Research in Molecular Human Biology (IRIBHM, https://iribhm.org/), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Morgane Leprovots
- Institute of Interdisciplinary Research in Molecular Human Biology (IRIBHM, https://iribhm.org/), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Gabriella Vasile
- Institute of Interdisciplinary Research in Molecular Human Biology (IRIBHM, https://iribhm.org/), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Frédérick Libert
- Institute of Interdisciplinary Research in Molecular Human Biology (IRIBHM, https://iribhm.org/), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Anne Lefort
- Institute of Interdisciplinary Research in Molecular Human Biology (IRIBHM, https://iribhm.org/), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Gilles Dinsart
- Institute of Interdisciplinary Research in Molecular Human Biology (IRIBHM, https://iribhm.org/), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Achim Weber
- Institute of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, CH-8091 Zurich, Switzerland
| | - Jasna Jetzer
- Institute of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, CH-8091 Zurich, Switzerland
- Institute of Molecular Cancer Research, University of Zurich, CH-8091 Zurich, Switzerland
| | - Marie-Isabelle Garcia
- Institute of Interdisciplinary Research in Molecular Human Biology (IRIBHM, https://iribhm.org/), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Gilbert Vassart
- Institute of Interdisciplinary Research in Molecular Human Biology (IRIBHM, https://iribhm.org/), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| |
Collapse
|
4
|
Shay JES, Yilmaz ÖH. Dietary and metabolic effects on intestinal stem cells in health and disease. Nat Rev Gastroenterol Hepatol 2025; 22:23-38. [PMID: 39358589 PMCID: PMC12105169 DOI: 10.1038/s41575-024-00980-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/05/2024] [Indexed: 10/04/2024]
Abstract
Diet and nutritional metabolites exhibit wide-ranging effects on health and disease partly by altering tissue composition and function. With rapidly rising rates of obesity, there is particular interest in how obesogenic diets influence tissue homeostasis and risk of tumorigenesis; epidemiologically, these diets have a positive correlation with various cancers, including colorectal cancer. The gastrointestinal tract is a highly specialized, continuously renewing tissue with a fundamental role in nutrient uptake and is, in turn, influenced by diet composition and host metabolic state. Intestinal stem cells are found at the base of the intestinal crypt and can generate all mature lineages that comprise the intestinal epithelium and are uniquely influenced by host diet, metabolic by-products and energy dynamics. Similarly, tumour growth and metabolism can also be shaped by nutrient availability and host diet. In this Review, we discuss how different diets and metabolic changes influence intestinal stem cells in homeostatic and pathological conditions, as well as tumorigenesis. We also discuss how dietary changes and composition affect the intestinal epithelium and its surrounding microenvironment.
Collapse
Affiliation(s)
- Jessica E S Shay
- Department of Biology, The David H. Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA, USA
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ömer H Yilmaz
- Department of Biology, The David H. Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA.
| |
Collapse
|
5
|
Tu J, Wang B, Wang X, Huo K, Hu W, Zhang R, Li J, Zhu S, Liang Q, Han S. Current status and new directions for hepatocellular carcinoma diagnosis. LIVER RESEARCH 2024; 8:218-236. [PMID: 39958920 PMCID: PMC11771281 DOI: 10.1016/j.livres.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/17/2024] [Accepted: 12/01/2024] [Indexed: 02/18/2025]
Abstract
Liver cancer ranks as the sixth most common cancer globally, with hepatocellular carcinoma (HCC) accounting for approximately 75%-85% of cases. Most patients present with moderately advanced disease, while those with advanced HCC face limited and ineffective treatment options. Despite diagnostic efforts, no ideal tumor marker exists to date, highlighting the urgent clinical need for improved early detection of HCC. A key research objective is the development of assays that target specific pathways involved in HCC progression. This review explores the pathological origin and development of HCC, providing insights into the mechanistic rationale, clinical statistics, and the advantages and limitations of commonly used diagnostic tumor markers. Additionally, it discusses the potential of emerging biomarkers for early diagnosis and offers a brief overview of relevant assay methodologies. This review aims to summarize existing markers and investigate new ones, providing a basis for subsequent research.
Collapse
Affiliation(s)
- Jinqi Tu
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang, China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, China
| | - Bo Wang
- Animal Experimental Center, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xiaoming Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, China
| | - Kugeng Huo
- Cyagen Biosciences (Guangzhou) Inc., Guangzhou, Guangdong, China
| | - Wanting Hu
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Beijing Key Lab of Microanalytical Methods & Instrumentation, Center for Synthetic and Systems Biology, Department of Chemistry, Tsinghua University, Beijing, China
| | - Rongli Zhang
- Department of Medicine, Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Jinyao Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang, China
| | - Shijie Zhu
- Department of Oncology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qionglin Liang
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Beijing Key Lab of Microanalytical Methods & Instrumentation, Center for Synthetic and Systems Biology, Department of Chemistry, Tsinghua University, Beijing, China
| | - Shuxin Han
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang, China
| |
Collapse
|
6
|
Verma S, Moreno IY, Gesteira TF, Coulson-Thomas VJ. Toxicity of nuclear-localized GFP in reporter mice. Sci Rep 2024; 14:24642. [PMID: 39428407 PMCID: PMC11491490 DOI: 10.1038/s41598-024-75741-2] [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: 04/10/2024] [Accepted: 10/08/2024] [Indexed: 10/22/2024] Open
Abstract
Various techniques using fluorescent reporter probes have been developed, such as GFP transgenic mouse lines that are used to detect spatial-temporal expression levels of genes. Although GFP expression is largely considered non-toxic, recent reports have indicated that under certain conditions GFP can display cellular toxicity. We hereby report the nuclear toxicity of H2B-GFP using a K14 specific Tet-on reporter mouse system. Using this system, GFP accumulates in the nucleus of all K14 expressing cells, such as the ocular surface epithelia and ocular adnexa. Expression of high levels of nuclear GFP during embryonic stages led to an eye open-at-birth (EOB) phenotype and abnormal ocular adnexa development and during adult and aging stages showed notable toxicity to ocular tissues. Other tissues, such as skin, also presented multiple defects associated with H2B-GFP expression. This toxicity was found to be concentration dependent, with homozygous mice presenting extremely high toxicity, while heterozygous mice presented limited toxicity. Upon induction, the accumulation of H2B-GFP in the nucleus of homozygous mice led to apoptosis within 2 weeks. This study therefore shows that although the use of nuclear GFP reporter mice is a valuable tool, at high levels, nuclear GFP can be toxic, leading to cell death and affecting tissue function.
Collapse
Affiliation(s)
- Sudhir Verma
- College of Optometry, University of Houston, 4401 Martin Luther King Boulevard, Houston, TX, 77204-2020, USA
- Department of Zoology, Deen Dayal Upadhyaya College, University of Delhi, Delhi, 110078, India
| | - Isabel Y Moreno
- College of Optometry, University of Houston, 4401 Martin Luther King Boulevard, Houston, TX, 77204-2020, USA
| | - Tarsis F Gesteira
- College of Optometry, University of Houston, 4401 Martin Luther King Boulevard, Houston, TX, 77204-2020, USA
| | - Vivien J Coulson-Thomas
- College of Optometry, University of Houston, 4401 Martin Luther King Boulevard, Houston, TX, 77204-2020, USA.
| |
Collapse
|
7
|
Oh SJ, Seo Y, Kim HS. Navigating the Landscape of Intestinal Regeneration: A Spotlight on Quiescence Regulation and Fetal Reprogramming. Int J Stem Cells 2024; 17:213-223. [PMID: 38267367 PMCID: PMC11361849 DOI: 10.15283/ijsc23176] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/20/2023] [Accepted: 01/02/2024] [Indexed: 01/26/2024] Open
Abstract
Tissue-specific adult stem cells are pivotal in maintaining tissue homeostasis, especially in the rapidly renewing intestinal epithelium. At the heart of this process are leucine-rich repeat-containing G protein-coupled receptor 5-expressing crypt base columnar cells (CBCs) that differentiate into various intestinal epithelial cells. However, while these CBCs are vital for tissue turnover, they are vulnerable to cytotoxic agents. Recent advances indicate that alternative stem cell sources drive the epithelial regeneration post-injury. Techniques like lineage tracing and single-cell RNA sequencing, combined with in vitro organoid systems, highlight the remarkable cellular adaptability of the intestinal epithelium during repair. These regenerative responses are mediated by the reactivation of conserved stem cells, predominantly quiescent stem cells and revival stem cells. With focus on these cells, this review unpacks underlying mechanisms governing intestinal regeneration and explores their potential clinical applications.
Collapse
Affiliation(s)
- Su-Jeong Oh
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, Korea
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan, Korea
- Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan, Korea
| | - Yoojin Seo
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, Korea
| | - Hyung-Sik Kim
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, Korea
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan, Korea
- Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan, Korea
| |
Collapse
|
8
|
Arif W, Narendran S, Kannan NB, Ramasamy K, Veerappan M, Chidambaranathan GP. Age-related reduction in the functional properties of adult stem cells located in the peripheral region of human retinal pigment epithelium. Indian J Ophthalmol 2024; 72:S688-S695. [PMID: 38623706 PMCID: PMC11338411 DOI: 10.4103/ijo.ijo_2491_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/06/2024] [Accepted: 01/15/2024] [Indexed: 04/17/2024] Open
Abstract
PURPOSE Adult stem cells (SCs) with self-renewal and multilineage potential have been reported upon culturing human retinal pigment epithelial (RPE) cells. The current study aimed to identify the location of SCs in human RPE and to elucidate the age-related changes. METHODS Peripheral, equatorial, and central RPE cells from donors of three age groups were analyzed for their sphere-forming, clonal, and label-retaining cell properties. Furthermore, native human RPE flatmounts were immunostained for SC and proliferating cell markers. RESULTS Cells with higher sphere-forming and clonal ability were identified only in young donors (<30 years) and were restricted to the periphery. Upon culturing, cells from peripheral and equatorial regions had the label-retaining cell (LRC) property. With aging, the LRCs were restricted to the periphery and were reduced. In young donors, Ki67 + proliferating cells were not observed in native RPE. However, such cells were observed in the peripheral RPE of older donors correlating with the need for regeneration. The native RPE cells were negative for SC marker expression. CONCLUSION The above findings highlighted the presence of SCs with the ability to proliferate in the peripheral RPE and a reduction in these functional properties of SCs with aging.
Collapse
Affiliation(s)
- Waseema Arif
- Department of Immunology and Stem Cell Biology, Aravind Medical Research Foundation, Madurai, Tamil Nadu, India
- Department of Biotechnology, Aravind Medical Research Foundation -Affiliated to Alagappa University, Karaikudi, Tamil Nadu, India
- Unit of One Health, ICMR- Vector Control Research Centre, Puducherry, India
| | | | - Naresh Babu Kannan
- Retina and Vitreous Services, Aravind Eye Hospital and Post Graduate Institute of Ophthalmology, Madurai, Tamil Nadu, India
| | - Kim Ramasamy
- Retina and Vitreous Services, Aravind Eye Hospital and Post Graduate Institute of Ophthalmology, Madurai, Tamil Nadu, India
| | - Muthukkaruppan Veerappan
- Department of Immunology and Stem Cell Biology, Aravind Medical Research Foundation, Madurai, Tamil Nadu, India
| | - Gowri Priya Chidambaranathan
- Department of Immunology and Stem Cell Biology, Aravind Medical Research Foundation, Madurai, Tamil Nadu, India
- Department of Biotechnology, Aravind Medical Research Foundation -Affiliated to Alagappa University, Karaikudi, Tamil Nadu, India
| |
Collapse
|
9
|
Yang F, Duan Y, Li Y, Zhu D, Wang Z, Luo Z, Zhang Y, Zhang G, He X, Kang X. S100A6 Regulates nucleus pulposus cell apoptosis via Wnt/β-catenin signaling pathway: an in vitro and in vivo study. Mol Med 2024; 30:87. [PMID: 38877413 PMCID: PMC11179208 DOI: 10.1186/s10020-024-00853-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 06/03/2024] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND Intervertebral disc degeneration (IDD) is a common musculoskeletal degenerative disease, which often leads to low back pain and even disability, resulting in loss of labor ability and decreased quality of life. Although many progresses have been made in the current research, the underlying mechanism of IDD remains unclear. The apoptosis of nucleus pulposus (NP) cells (NPCs) is an important pathological mechanism in intervertebral disc degeneration (IDD). This study evaluated the relationship between S100A6 and NPCs and its underlying mechanism. METHODS Mass spectrometry, bioinformatics, and quantitative real-time polymerase chain reaction (qRT-PCR) analyses were used to screen and verify hub genes for IDD in human IVD specimens with different degeneration degrees. Western blotting, immunohistochemistry (IHC), and/or immunofluorescence (IF) were used to detect the expression level of S100A6 in human NP tissues and NPCs. The apoptotic phenotype of NPCs and Wnt/β-catenin signaling pathway were evaluated using flow cytometry, western blotting, and IF. S100A6 was overexpressed or knocked down in NPCs to determine its impact on apoptosis and Wnt/β-catenin signaling pathway activity. Moreover, we used the XAV-939 to inhibit and SKL2001 to activate the Wnt/β-catenin signaling pathway. The therapeutic effect of S100A6 inhibition on IDD was also evaluated. RESULTS S100A6 expression increased in IDD. In vitro, increased S100A6 expression promoted apoptosis in interleukin (IL)-1β-induced NPCs. In contrast, the inhibition of S100A6 expression partially alleviated the progression of annulus fibrosus (AF) puncture-induced IDD in rats. Mechanistic studies revealed that S100A6 regulates NPC apoptosis via Wnt/β-catenin signaling pathway. CONCLUSIONS This study showed that S100A6 expression increased during IDD and promoted NPCs apoptosis by regulating the Wnt/β-catenin signaling pathway, suggesting that S100A6 is a promising new therapeutic target for IDD.
Collapse
Affiliation(s)
- Fengguang Yang
- Department of Orthopedics, The Second Hospital of Lanzhou University, 82 Cuiying Men, Lanzhou, Gansu Province, 730030, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Yanni Duan
- Department of Orthopedics, The Second Hospital of Lanzhou University, 82 Cuiying Men, Lanzhou, Gansu Province, 730030, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Yanhu Li
- Department of Orthopedics, The Second Hospital of Lanzhou University, 82 Cuiying Men, Lanzhou, Gansu Province, 730030, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Daxue Zhu
- Department of Orthopedics, The Second Hospital of Lanzhou University, 82 Cuiying Men, Lanzhou, Gansu Province, 730030, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Zhaoheng Wang
- Department of Orthopedics, The Second Hospital of Lanzhou University, 82 Cuiying Men, Lanzhou, Gansu Province, 730030, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Zhangbin Luo
- Department of Orthopedics, The Second Hospital of Lanzhou University, 82 Cuiying Men, Lanzhou, Gansu Province, 730030, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Yizhi Zhang
- Department of Orthopedics, The Second Hospital of Lanzhou University, 82 Cuiying Men, Lanzhou, Gansu Province, 730030, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Guangzhi Zhang
- Department of Orthopedics, The Second Hospital of Lanzhou University, 82 Cuiying Men, Lanzhou, Gansu Province, 730030, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Xuegang He
- Department of Orthopedics, The Second Hospital of Lanzhou University, 82 Cuiying Men, Lanzhou, Gansu Province, 730030, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Xuewen Kang
- Department of Orthopedics, The Second Hospital of Lanzhou University, 82 Cuiying Men, Lanzhou, Gansu Province, 730030, China.
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China.
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, 730030, China.
| |
Collapse
|
10
|
Capdevila C, Miller J, Cheng L, Kornberg A, George JJ, Lee H, Botella T, Moon CS, Murray JW, Lam S, Calderon RI, Malagola E, Whelan G, Lin CS, Han A, Wang TC, Sims PA, Yan KS. Time-resolved fate mapping identifies the intestinal upper crypt zone as an origin of Lgr5+ crypt base columnar cells. Cell 2024; 187:3039-3055.e14. [PMID: 38848677 PMCID: PMC11770878 DOI: 10.1016/j.cell.2024.05.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 01/16/2024] [Accepted: 05/01/2024] [Indexed: 06/09/2024]
Abstract
In the prevailing model, Lgr5+ cells are the only intestinal stem cells (ISCs) that sustain homeostatic epithelial regeneration by upward migration of progeny through elusive upper crypt transit-amplifying (TA) intermediates. Here, we identify a proliferative upper crypt population marked by Fgfbp1, in the location of putative TA cells, that is transcriptionally distinct from Lgr5+ cells. Using a kinetic reporter for time-resolved fate mapping and Fgfbp1-CreERT2 lineage tracing, we establish that Fgfbp1+ cells are multi-potent and give rise to Lgr5+ cells, consistent with their ISC function. Fgfbp1+ cells also sustain epithelial regeneration following Lgr5+ cell depletion. We demonstrate that FGFBP1, produced by the upper crypt cells, is an essential factor for crypt proliferation and epithelial homeostasis. Our findings support a model in which tissue regeneration originates from upper crypt Fgfbp1+ cells that generate progeny propagating bi-directionally along the crypt-villus axis and serve as a source of Lgr5+ cells in the crypt base.
Collapse
Affiliation(s)
- Claudia Capdevila
- Department of Medicine, Division of Digestive & Liver Diseases, Columbia University Irving Medical Center, New York, NY, USA; Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA; Digestive & Liver Diseases Research Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Jonathan Miller
- Department of Medicine, Division of Digestive & Liver Diseases, Columbia University Irving Medical Center, New York, NY, USA; Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA; Digestive & Liver Diseases Research Center, Columbia University Irving Medical Center, New York, NY, USA; Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Liang Cheng
- Department of Medicine, Division of Digestive & Liver Diseases, Columbia University Irving Medical Center, New York, NY, USA; Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA; Digestive & Liver Diseases Research Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Adam Kornberg
- Department of Medicine, Division of Digestive & Liver Diseases, Columbia University Irving Medical Center, New York, NY, USA; Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA; Digestive & Liver Diseases Research Center, Columbia University Irving Medical Center, New York, NY, USA; Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Joel J George
- Department of Medicine, Division of Digestive & Liver Diseases, Columbia University Irving Medical Center, New York, NY, USA; Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA; Digestive & Liver Diseases Research Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Hyeonjeong Lee
- Department of Medicine, Division of Digestive & Liver Diseases, Columbia University Irving Medical Center, New York, NY, USA; Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA; Digestive & Liver Diseases Research Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Theo Botella
- Department of Medicine, Division of Digestive & Liver Diseases, Columbia University Irving Medical Center, New York, NY, USA; Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA; Digestive & Liver Diseases Research Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Christine S Moon
- Department of Medicine, Division of Digestive & Liver Diseases, Columbia University Irving Medical Center, New York, NY, USA; Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA; Digestive & Liver Diseases Research Center, Columbia University Irving Medical Center, New York, NY, USA
| | - John W Murray
- Department of Medicine, Division of Digestive & Liver Diseases, Columbia University Irving Medical Center, New York, NY, USA
| | - Stephanie Lam
- Department of Medicine, Division of Digestive & Liver Diseases, Columbia University Irving Medical Center, New York, NY, USA; Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA; Digestive & Liver Diseases Research Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Ruben I Calderon
- Department of Medicine, Division of Digestive & Liver Diseases, Columbia University Irving Medical Center, New York, NY, USA; Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA; Digestive & Liver Diseases Research Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Ermanno Malagola
- Department of Medicine, Division of Digestive & Liver Diseases, Columbia University Irving Medical Center, New York, NY, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA; Digestive & Liver Diseases Research Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Gary Whelan
- Department of Medicine, Division of Digestive & Liver Diseases, Columbia University Irving Medical Center, New York, NY, USA; Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA; Digestive & Liver Diseases Research Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Chyuan-Sheng Lin
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA; Department of Pathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Arnold Han
- Department of Medicine, Division of Digestive & Liver Diseases, Columbia University Irving Medical Center, New York, NY, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA; Digestive & Liver Diseases Research Center, Columbia University Irving Medical Center, New York, NY, USA; Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Timothy C Wang
- Department of Medicine, Division of Digestive & Liver Diseases, Columbia University Irving Medical Center, New York, NY, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA; Digestive & Liver Diseases Research Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Peter A Sims
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA; Digestive & Liver Diseases Research Center, Columbia University Irving Medical Center, New York, NY, USA; Departments of Biochemistry & Molecular Biophysics and of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Kelley S Yan
- Department of Medicine, Division of Digestive & Liver Diseases, Columbia University Irving Medical Center, New York, NY, USA; Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA; Digestive & Liver Diseases Research Center, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
11
|
Sun M, Tan Z, Lin K, Li X, Zhu J, Zhan L, Zheng H. Advanced Progression for the Heterogeneity and Homeostasis of Intestinal Stem Cells. Stem Cell Rev Rep 2023; 19:2109-2119. [PMID: 37351833 DOI: 10.1007/s12015-023-10578-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2023] [Indexed: 06/24/2023]
Abstract
Current understanding of the leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5) in intestinal stem cells (ISCs) is well established, however, the implications of ISC heterogeneity and homeostasis are poorly understood. Prior studies have provided important evidence for the association between heterogeneity of ISC pools with pathogenesis and therapeutic response of malignant disease. Leveraging the advantages of organoids and single cell RNA sequencing (scRNA-seq), glandular development has been simulated and cell heterogeneity has been clarified. Based on this research, several potential ISCs were identified, such as LGR5 + p27 + quiescent ISCs, LGR5 + Mex3a + slowly proliferating stem cells, and CLU + reverse stem cells. We also illustrated major factors responsible for ISC homeostasis including metabolism-related (LKB1, TGR5, HMGCS2), inflammation-related (IFB-b, IFN2, TNF), and Wnt signaling-related (CREPT, Mex3a, MTG16) factors. ISCs play complex roles in intestinal tumorigenesis, chemoresistance and occasional relapse of colon cancer, which bear discussion. In this review, we focus on novel technical challenges in ISCs fate drawing upon recent research with the goals of clarifying our understanding of complex ISCs, elucidating the integrated intestinal crypt niche, and creating new opportunities for therapeutic development.
Collapse
Affiliation(s)
- Minqiong Sun
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui, China
| | - Zhenya Tan
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui, China
| | - Keqiong Lin
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui, China
| | - Xiaofei Li
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui, China
| | - Jicheng Zhu
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui, China
| | - Li Zhan
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui, China
| | - Hong Zheng
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
12
|
Wang Y, Kang X, Kang X, Yang F. S100A6: molecular function and biomarker role. Biomark Res 2023; 11:78. [PMID: 37670392 PMCID: PMC10481514 DOI: 10.1186/s40364-023-00515-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/03/2023] [Indexed: 09/07/2023] Open
Abstract
S100A6 (also called calcyclin) is a Ca2+-binding protein that belongs to the S100 protein family. S100A6 has many functions related to the cytoskeleton, cell stress, proliferation, and differentiation. S100A6 also has many interacting proteins that are distributed in the cytoplasm, nucleus, cell membrane, and outside the cell. Almost all these proteins interact with S100A6 in a Ca2+-dependent manner, and some also have specific motifs responsible for binding to S100A6. The expression of S100A6 is regulated by several transcription factors (such as c-Myc, P53, NF-κB, USF, Nrf2, etc.). The expression level depends on the specific cell type and the transcription factors activated in specific physical and chemical environments, and is also related to histone acetylation, DNA methylation, and other epigenetic modifications. The differential expression of S100A6 in various diseases, and at different stages of those diseases, makes it a good biomarker for differential diagnosis and prognosis evaluation, as well as a potential therapeutic target. In this review, we mainly focus on the S100A6 ligand and its transcriptional regulation, molecular function (cytoskeleton, cell stress, cell differentiation), and role as a biomarker in human disease and stem cells.
Collapse
Affiliation(s)
- Yidian Wang
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi, China
| | - Xuewen Kang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Xin Kang
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi, China.
| | - Fengguang Yang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China.
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China.
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China.
- The Orthopedics Department of the Second Hospital of Lanzhou University, 82 Cuiying Men, Lanzhou, Gansu Province, 730000, PR China.
| |
Collapse
|
13
|
Creff J, Nowosad A, Prel A, Pizzoccaro A, Aguirrebengoa M, Duquesnes N, Callot C, Jungas T, Dozier C, Besson A. p57 Kip2 acts as a transcriptional corepressor to regulate intestinal stem cell fate and proliferation. Cell Rep 2023; 42:112659. [PMID: 37327110 DOI: 10.1016/j.celrep.2023.112659] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/01/2022] [Accepted: 06/01/2023] [Indexed: 06/18/2023] Open
Abstract
p57Kip2 is a cyclin/CDK inhibitor and a negative regulator of cell proliferation. Here, we report that p57 regulates intestinal stem cell (ISC) fate and proliferation in a CDK-independent manner during intestinal development. In the absence of p57, intestinal crypts exhibit an increased proliferation and an amplification of transit-amplifying cells and of Hopx+ ISCs, which are no longer quiescent, while Lgr5+ ISCs are unaffected. RNA sequencing (RNA-seq) analyses of Hopx+ ISCs show major gene expression changes in the absence of p57. We found that p57 binds to and inhibits the activity of Ascl2, a transcription factor critical for ISC specification and maintenance, by participating in the recruitment of a corepressor complex to Ascl2 target gene promoters. Thus, our data suggest that, during intestinal development, p57 plays a key role in maintaining Hopx+ ISC quiescence and repressing the ISC phenotype outside of the crypt bottom by inhibiting the transcription factor Ascl2 in a CDK-independent manner.
Collapse
Affiliation(s)
- Justine Creff
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), University of Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Ada Nowosad
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), University of Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Anne Prel
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), University of Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Anne Pizzoccaro
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), University of Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Marion Aguirrebengoa
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), University of Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Nicolas Duquesnes
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), University of Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Caroline Callot
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), University of Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Thomas Jungas
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), University of Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Christine Dozier
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), University of Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Arnaud Besson
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), University of Toulouse, CNRS, UPS, 31062 Toulouse, France.
| |
Collapse
|
14
|
Dong J, Wu X, Zhou X, Gao Y, Wang C, Wang W, He W, Li J, Deng W, Liao J, Wu X, Lu Y, Chen AK, Wen L, Fu W, Tang F. Spatially resolved expression landscape and gene-regulatory network of human gastric corpus epithelium. Protein Cell 2023; 14:433-447. [PMID: 37402315 PMCID: PMC10319429 DOI: 10.1093/procel/pwac059] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/30/2022] [Indexed: 07/20/2023] Open
Abstract
Molecular knowledge of human gastric corpus epithelium remains incomplete. Here, by integrated analyses using single-cell RNA sequencing (scRNA-seq), spatial transcriptomics, and single-cell assay for transposase accessible chromatin sequencing (scATAC-seq) techniques, we uncovered the spatially resolved expression landscape and gene-regulatory network of human gastric corpus epithelium. Specifically, we identified a stem/progenitor cell population in the isthmus of human gastric corpus, where EGF and WNT signaling pathways were activated. Meanwhile, LGR4, but not LGR5, was responsible for the activation of WNT signaling pathway. Importantly, FABP5 and NME1 were identified and validated as crucial for both normal gastric stem/progenitor cells and gastric cancer cells. Finally, we explored the epigenetic regulation of critical genes for gastric corpus epithelium at chromatin state level, and identified several important cell-type-specific transcription factors. In summary, our work provides novel insights to systematically understand the cellular diversity and homeostasis of human gastric corpus epithelium in vivo.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Weiya He
- GMU-GIBH Joint School of Life Sciences, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou 510799, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510320, China
| | - Jingyun Li
- Biomedical Pioneering Innovation Center, Department of General Surgery, College of Life Sciences, Third Hospital, Peking University, Beijing 100871, China
| | - Wenjun Deng
- GMU-GIBH Joint School of Life Sciences, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou 510799, China
| | - Jiayu Liao
- GMU-GIBH Joint School of Life Sciences, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou 510799, China
| | - Xiaotian Wu
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Yongqu Lu
- Biomedical Pioneering Innovation Center, Department of General Surgery, College of Life Sciences, Third Hospital, Peking University, Beijing 100871, China
| | - Antony K Chen
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Lu Wen
- Biomedical Pioneering Innovation Center, Department of General Surgery, College of Life Sciences, Third Hospital, Peking University, Beijing 100871, China
- Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing 100871, China
| | | | | |
Collapse
|
15
|
Karo-Atar D, Gregorieff A, King IL. Dangerous liaisons: how helminths manipulate the intestinal epithelium. Trends Parasitol 2023; 39:414-422. [PMID: 37076358 DOI: 10.1016/j.pt.2023.03.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/10/2023] [Accepted: 03/16/2023] [Indexed: 04/21/2023]
Abstract
Intestinal helminths remain highly pervasive throughout the animal kingdom by modulating multiple aspects of the host immune response. The intestinal epithelium functions as a physical barrier as well as a sentinel innate immune tissue with the ability to sense and respond to infectious agents. Although helminths form intimate interactions with the epithelium, comprehensive knowledge about host-helminth interactions at this dynamic interface is lacking. In addition, little is known about the ability of helminths to directly shape the fate of this barrier tissue. Here, we review the diverse pathways by which helminths regulate the epithelium and highlight the emerging field of direct helminth regulation of intestinal stem cell (ISC) fate and function.
Collapse
Affiliation(s)
- Danielle Karo-Atar
- Department of Microbiology and Immunology, Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; McGill Interdisciplinary Initiative in Infection and Immunity, Montreal, Quebec, Canada; McGill Regenerative Medicine Network, Montreal, Quebec, Canada.
| | - Alex Gregorieff
- McGill Regenerative Medicine Network, Montreal, Quebec, Canada; Department of Pathology, McGill University and Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Irah L King
- Department of Microbiology and Immunology, Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; McGill Interdisciplinary Initiative in Infection and Immunity, Montreal, Quebec, Canada; McGill Regenerative Medicine Network, Montreal, Quebec, Canada; McGill Centre for Microbiome Research, Montreal, Quebec, Canada.
| |
Collapse
|
16
|
Banjac I, Maimets M, Jensen KB. Maintenance of high-turnover tissues during and beyond homeostasis. Cell Stem Cell 2023; 30:348-361. [PMID: 37028402 DOI: 10.1016/j.stem.2023.03.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/23/2023] [Accepted: 03/15/2023] [Indexed: 04/09/2023]
Abstract
Tissues with a high turnover rate produce millions of cells daily and have abundant regenerative capacity. At the core of their maintenance are populations of stem cells that balance self-renewal and differentiation to produce the adequate numbers of specialized cells required for carrying out essential tissue functions. Here, we compare and contrast the intricate mechanisms and elements of homeostasis and injury-driven regeneration in the epidermis, hematopoietic system, and intestinal epithelium-the fastest renewing tissues in mammals. We highlight the functional relevance of the main mechanisms and identify open questions in the field of tissue maintenance.
Collapse
Affiliation(s)
- Isidora Banjac
- The Novo Nordisk Foundation Center for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Martti Maimets
- The Novo Nordisk Foundation Center for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark.
| | - Kim B Jensen
- The Novo Nordisk Foundation Center for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark.
| |
Collapse
|
17
|
Huang XT, Li T, Li T, Xing S, Tian JZ, Ding YF, Cai SL, Yang YS, Wood C, Yang JS, Yang WJ. Embryogenic stem cell-derived intestinal crypt fission directs de novo crypt genesis. Cell Rep 2022; 41:111796. [PMID: 36516755 DOI: 10.1016/j.celrep.2022.111796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 09/30/2022] [Accepted: 11/16/2022] [Indexed: 12/15/2022] Open
Abstract
Intestinal epithelial replenishment is fueled by continuously dividing intestinal stem cells (ISCs) resident at the crypt niche. However, the cell type(s) enabling replenishment upon damage and subsequent loss of whole crypts remain largely unclear. Using Set domain-containing protein 4 (Setd4), we identify a small population with reserve stem cell characteristics in the mouse intestine. Upon irradiation-induced injury, Setd4-expressing (Setd4+) cells survive radiation exposure and then activate to produce Sca-1-expressing cell types to restore the epithelial wall and regenerate crypts de novo via crypt fission. Setd4+ cells are confirmed to originate from the early fetal period, subsequently contributing to the development of embryonic gut and the establishment of postnatal crypts. Setd4+ cells are therefore represented as both originators and key regenerators of the intestine.
Collapse
Affiliation(s)
- Xue-Ting Huang
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ting Li
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tong Li
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Sheng Xing
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jin-Ze Tian
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yan-Fu Ding
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Sun-Li Cai
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yao-Shun Yang
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Christopher Wood
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jin-Shu Yang
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wei-Jun Yang
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou 310058, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266000, China.
| |
Collapse
|
18
|
Johnson NM, Parham LR, Na J, Monaghan KE, Kolev HM, Klochkova A, Kim MS, Danan CH, Cramer Z, Simon LA, Naughton KE, Adams‐Tzivelekidis S, Tian Y, Williams PA, Leu NA, Sidoli S, Whelan KA, Li N, Lengner CJ, Hamilton KE. Autophagic state prospectively identifies facultative stem cells in the intestinal epithelium. EMBO Rep 2022; 23:e55209. [PMID: 36120829 PMCID: PMC9638868 DOI: 10.15252/embr.202255209] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/24/2022] [Accepted: 09/05/2022] [Indexed: 01/25/2023] Open
Abstract
The intestinal epithelium exhibits a rapid and efficient regenerative response to injury. Emerging evidence supports a model where plasticity of differentiated cells, particularly those in the secretory lineages, contributes to epithelial regeneration upon ablation of injury-sensitive stem cells. However, such facultative stem cell activity is rare within secretory populations. Here, we ask whether specific functional properties predict facultative stem cell activity. We utilize in vivo labeling combined with ex vivo organoid formation assays to evaluate how cell age and autophagic state contribute to facultative stem cell activity within secretory lineages. Strikingly, we find that cell age (time elapsed since cell cycle exit) does not correlate with secretory cell plasticity. Instead, high autophagic vesicle content predicts plasticity and resistance to DNA damaging injury independently of cell lineage. Our findings indicate that autophagic status prior to injury serves as a lineage-agnostic marker for the prospective identification of facultative stem cells.
Collapse
Affiliation(s)
- Nicolette M Johnson
- Medical Scientist Training Program, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Biomedical Sciences, School of Veterinary MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Louis R Parham
- Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Division of Gastroenterology, Hepatology, and NutritionChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Jeeyoon Na
- Department of Biomedical Sciences, School of Veterinary MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Keara E Monaghan
- Department of Biomedical Sciences, School of Veterinary MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Hannah M Kolev
- Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Alena Klochkova
- Fels Institute for Cancer Research & Molecular BiologyLewis Katz School of Medicine at Temple UniversityPhiladelphiaPennsylvaniaUSA
| | - Melissa S Kim
- Department of Biomedical Sciences, School of Veterinary MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Charles H Danan
- Medical Scientist Training Program, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Division of Gastroenterology, Hepatology, and NutritionChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Zvi Cramer
- Department of Biomedical Sciences, School of Veterinary MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Lauren A Simon
- Division of Gastroenterology, Hepatology, and NutritionChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Kaitlyn E Naughton
- Division of Gastroenterology, Hepatology, and NutritionChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Stephanie Adams‐Tzivelekidis
- Department of Biomedical Sciences, School of Veterinary MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Yuhua Tian
- Department of Biomedical Sciences, School of Veterinary MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Patrick A Williams
- Division of Gastroenterology, Hepatology, and NutritionChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - N Adrian Leu
- Department of Biomedical Sciences, School of Veterinary MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Simone Sidoli
- Department of BiochemistryAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Kelly A Whelan
- Fels Institute for Cancer Research & Molecular BiologyLewis Katz School of Medicine at Temple UniversityPhiladelphiaPennsylvaniaUSA
- Department of Pathology & Laboratory MedicineLewis Katz School of Medicine at Temple UniversityPhiladelphiaPennsylvaniaUSA
| | - Ning Li
- Department of Biomedical Sciences, School of Veterinary MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Institute for Regenerative MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Christopher J Lengner
- Department of Biomedical Sciences, School of Veterinary MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Institute for Regenerative MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Cell & Developmental Biology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Kathryn E Hamilton
- Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Division of Gastroenterology, Hepatology, and NutritionChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
- Institute for Regenerative MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
19
|
Ishikawa K, Sugimoto S, Oda M, Fujii M, Takahashi S, Ohta Y, Takano A, Ishimaru K, Matano M, Yoshida K, Hanyu H, Toshimitsu K, Sawada K, Shimokawa M, Saito M, Kawasaki K, Ishii R, Taniguchi K, Imamura T, Kanai T, Sato T. Identification of Quiescent LGR5 + Stem Cells in the Human Colon. Gastroenterology 2022; 163:1391-1406.e24. [PMID: 35963362 DOI: 10.1053/j.gastro.2022.07.081] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 07/16/2022] [Accepted: 07/19/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS In the mouse intestinal epithelium, Lgr5+ stem cells are vulnerable to injury, owing to their predominantly cycling nature, and their progenies de-differentiate to replenish the stem cell pool. However, how human colonic stem cells behave in homeostasis and during regeneration remains unknown. METHODS Transcriptional heterogeneity among colonic epithelial cells was analyzed by means of single-cell RNA sequencing analysis of human and mouse colonic epithelial cells. To trace the fate of human colonic stem or differentiated cells, we generated LGR5-tdTomato, LGR5-iCasase9-tdTomato, LGR5-split-Cre, and KRT20-ERCreER knock-in human colon organoids via genome engineering. p27+ dormant cells were further visualized with the p27-mVenus reporter. To analyze the dynamics of human colonic stem cells in vivo, we orthotopically xenotransplanted fluorescence-labeled human colon organoids into immune-deficient mice. The cell cycle dynamics in xenograft cells were evaluated using 5-ethynyl-2'-deoxyuridine pulse-chase analysis. The clonogenic capacity of slow-cycling human stem cells or differentiated cells was analyzed in the context of homeostasis, LGR5 ablation, and 5-fluorouracil-induced mucosal injury. RESULTS Single-cell RNA sequencing analysis illuminated the presence of nondividing LGR5+ stem cells in the human colon. Visualization and lineage tracing of slow-cycling LGR5+p27+ cells and orthotopic xenotransplantation validated their homeostatic lineage-forming capability in vivo, which was augmented by 5-FU-induced mucosal damage. Transforming growth factor-β signaling regulated the quiescent state of LGR5+ cells. Despite the plasticity of differentiated KRT20+ cells, they did not display clonal growth after 5-FU-induced injury, suggesting that occupation of the niche environment by LGR5+p27+ cells prevented neighboring differentiated cells from de-differentiating. CONCLUSIONS Our results highlight the quiescent nature of human LGR5+ colonic stem cells and their contribution to post-injury regeneration.
Collapse
Affiliation(s)
- Keiko Ishikawa
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan; Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Shinya Sugimoto
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan; Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Mayumi Oda
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
| | - Masayuki Fujii
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
| | - Sirirat Takahashi
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
| | - Yuki Ohta
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
| | - Ai Takano
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
| | - Kazuhiro Ishimaru
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
| | - Mami Matano
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
| | - Kosuke Yoshida
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan; Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Hikaru Hanyu
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
| | - Kohta Toshimitsu
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
| | - Kazuaki Sawada
- Center for Integrated Medical Research, School of Medicine, Keio University, Tokyo, Japan
| | - Mariko Shimokawa
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
| | - Megumu Saito
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan; Fujii Memorial Research Institute, Otsuka Pharmaceutical Company, Limited, Shiga, Japan
| | - Kenta Kawasaki
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan; Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Ryota Ishii
- Department of Biostatistics, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Koji Taniguchi
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan; Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Takeshi Imamura
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Takanori Kanai
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Toshiro Sato
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
20
|
Leśniak W, Filipek A. S100A6 as a Constituent and Potential Marker of Adult and Cancer Stem Cells. Stem Cell Rev Rep 2022; 18:2699-2708. [PMID: 35796891 DOI: 10.1007/s12015-022-10403-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2022] [Indexed: 10/17/2022]
Abstract
Adult or tissue stem cells are present in various tissues of the organism where they reside in a specific environment called the niche. Owing to their ability to generate a progeny that can proliferate and differentiate into specialized cell types, adult stem cells constitute a source of new cells necessary for tissue maintenance and/or regeneration. Under normal conditions they divide with a frequency matching the pace of tissue renewal but, following tissue damage, they can migrate to the site of injury and expand/divide intensively to facilitate tissue repair. For this reason much hope is being placed on the use of adult stem cells in regenerative therapies, including tissue engineering. Identification and characterization of tissue stem cells has been a laborious process due to their scarcity and lack of universal markers. Nonetheless, recent studies, employing various types of transcriptomic analyses, revealed some common trends in gene expression pattern among stem cells derived from different tissues, suggesting the importance of certain genes/proteins for the unique properties of these cells. S100A6, a small calcium binding protein, has been recognized as an important factor influencing cell proliferation and differentiation. Accumulating results show that S100A6 is a constituent of adult stem cells and, in some cases, may even be considered as their marker. Thus, in this review we summarize literature data concerning the presence of S100A6 in adult and cancer stem cells and speculate on its potential role and usefulness as a marker of these cells.
Collapse
Affiliation(s)
- Wiesława Leśniak
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02- 093, Warsaw, Poland.
| | - Anna Filipek
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02- 093, Warsaw, Poland
| |
Collapse
|
21
|
Meyer AR, Brown ME, McGrath PS, Dempsey PJ. Injury-Induced Cellular Plasticity Drives Intestinal Regeneration. Cell Mol Gastroenterol Hepatol 2021; 13:843-856. [PMID: 34915204 PMCID: PMC8803615 DOI: 10.1016/j.jcmgh.2021.12.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 12/14/2022]
Abstract
The epithelial lining of the intestine, particularly the stem cell compartment, is affected by harsh conditions in the luminal environment and also is susceptible to genotoxic agents such as radiation and chemotherapy. Therefore, the ability for intestinal epithelial cells to revert to a stem cell state is an important physiological damage response to regenerate the intestinal epithelium at sites of mucosal injury. Many signaling networks involved in maintaining the stem cell niche are activated as part of the damage response to promote cellular plasticity and regeneration. The relative contribution of each cell type and signaling pathway is a critical area of ongoing research, likely dependent on the nature of injury as well as the regional specification within the intestine. Here, we review the current understanding of the multicellular cooperation to restore the intestinal epithelium after damage.
Collapse
Affiliation(s)
| | | | | | - Peter J. Dempsey
- Correspondence Address correspondence to: Peter J. Dempsey, PhD, Section of Developmental Biology, Department of Pediatrics, University of Colorado School of Medicine, 1775 Aurora Court, Barbara Davis Center, M20–3306, Aurora, Colorado 80045. fax: (303) 724-6538.
| |
Collapse
|
22
|
Fu X, He Q, Tao Y, Wang M, Wang W, Wang Y, Yu QC, Zhang F, Zhang X, Chen YG, Gao D, Hu P, Hui L, Wang X, Zeng YA. Recent advances in tissue stem cells. SCIENCE CHINA. LIFE SCIENCES 2021; 64:1998-2029. [PMID: 34865207 DOI: 10.1007/s11427-021-2007-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 06/08/2021] [Indexed: 12/13/2022]
Abstract
Stem cells are undifferentiated cells capable of self-renewal and differentiation, giving rise to specialized functional cells. Stem cells are of pivotal importance for organ and tissue development, homeostasis, and injury and disease repair. Tissue-specific stem cells are a rare population residing in specific tissues and present powerful potential for regeneration when required. They are usually named based on the resident tissue, such as hematopoietic stem cells and germline stem cells. This review discusses the recent advances in stem cells of various tissues, including neural stem cells, muscle stem cells, liver progenitors, pancreatic islet stem/progenitor cells, intestinal stem cells, and prostate stem cells, and the future perspectives for tissue stem cell research.
Collapse
Affiliation(s)
- Xin Fu
- Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200233, China
| | - Qiang He
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yu Tao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Mengdi Wang
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology, Bioland Laboratory (Guangzhou), Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wei Wang
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology, Bioland Laboratory (Guangzhou), Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yalong Wang
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Qing Cissy Yu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Fang Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xiaoyu Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- Max-Planck Center for Tissue Stem Cell Research and Regenerative Medicine, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510530, China.
| | - Dong Gao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Ping Hu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200233, China.
- Max-Planck Center for Tissue Stem Cell Research and Regenerative Medicine, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510530, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Bio-Research Innovation Center, Shanghai Institute of Biochemistry and Cell Biology, Suzhou, 215121, China.
| | - Lijian Hui
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Bio-Research Innovation Center, Shanghai Institute of Biochemistry and Cell Biology, Suzhou, 215121, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, 310024, China.
| | - Xiaoqun Wang
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology, Bioland Laboratory (Guangzhou), Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Advanced Innovation Center for Human Brain Protection, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China.
| | - Yi Arial Zeng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- Bio-Research Innovation Center, Shanghai Institute of Biochemistry and Cell Biology, Suzhou, 215121, China.
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, 310024, China.
| |
Collapse
|
23
|
Wang S, Li S, Li Y, Jiang Q, Li X, Wang Y, Han JD, Liu Y, Chen YG. Non-muscle myosin heavy chain 9 maintains intestinal homeostasis by preventing epithelium necroptosis and colitis adenoma formation. Stem Cell Reports 2021; 16:1290-1301. [PMID: 33891868 PMCID: PMC8185465 DOI: 10.1016/j.stemcr.2021.03.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/12/2022] Open
Abstract
Non-muscle myosin IIA plays an important role in cell adhesion, cell migration, and tissue architecture. We previously showed that low activity of the heavy chain of non-muscle myosin II Myh9 is beneficial to LGR5+ intestinal stem cell maintenance. However, the function of Myh9 in adult mouse intestinal epithelium is largely unclear. In this study, we used the inducible Villin-creERT2 knockout approach to delete Myh9 in adult mouse intestinal epithelium and observed that homozygous deletion of Myh9 causes colitis-like morphologic changes in intestine, leads to a high sensitivity to dextran sulfate sodium and promotes colitis-related adenoma formation in the colon. Myh9 deletion disturbs cell junctions and impairs intestinal lumen barrier integrity, promoting the necroptosis of epithelial cells. Consistently, these changes can be partially rescued by Ripk3 knockout. Our results indicate that Myh9 is required for the maintenance of intestinal epithelium integrity and the prevention of cell necroptosis.
Collapse
Affiliation(s)
- Shan Wang
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Siqi Li
- Max-Planck Center for Tissue Stem Cell Research and Regenerative Medicine, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Yehua Li
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Quanlong Jiang
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology, Peking University, Beijing 100871, P.R. China
| | - Xintong Li
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yalong Wang
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jing-Dong Han
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology, Peking University, Beijing 100871, P.R. China
| | - Yuan Liu
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China.
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China; Max-Planck Center for Tissue Stem Cell Research and Regenerative Medicine, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.
| |
Collapse
|
24
|
Cycling Stem Cells Are Radioresistant and Regenerate the Intestine. Cell Rep 2021; 32:107952. [PMID: 32726617 PMCID: PMC7789978 DOI: 10.1016/j.celrep.2020.107952] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 01/09/2020] [Accepted: 07/02/2020] [Indexed: 01/17/2023] Open
Abstract
A certain number of epithelial cells in intestinal crypts are DNA damage resistant and contribute to regeneration. However, the cellular mechanism underlying intestinal regeneration remains unclear. Using lineage tracing, we show that cells marked by an Msi1 reporter (Msi1+) are right above Lgr5high cells in intestinal crypts and exhibit DNA damage resistance. Single-cell RNA sequencing reveals that the Msi1+ cells are heterogeneous with the majority being intestinal stem cells (ISCs). The DNA damage-resistant subpopulation of Msi1+ cells is characterized by low-to-negative Lgr5 expression and is more rapidly cycling than Lgr5high radiosensitive crypt base columnar stem cells (CBCs). This enables an efficient repopulation of the intestinal epithelium at early stage when Lgr5high cells are not emerging. Furthermore, relative to CBCs, Msi1+ cells preferentially produce Paneth cells during homeostasis and upon radiation repair. Together, we demonstrate that the DNA damage-resistant Msi1+ cells are cycling ISCs that maintain and regenerate the intestinal epithelium. Quiescent reserve stem cells in the intestine are thought to activate following irradiation to restore the depleted Lgr5high CBCs. Now, Sheng et al. demonstrate that cycling Msi1+ cells represent DNA damage-resistant ISCs that support efficient repopulation of the intestinal epithelium at the early stage of post-radiation repair, ahead of Lgr5high CBCs.
Collapse
|
25
|
Sphyris N, Hodder MC, Sansom OJ. Subversion of Niche-Signalling Pathways in Colorectal Cancer: What Makes and Breaks the Intestinal Stem Cell. Cancers (Basel) 2021; 13:1000. [PMID: 33673710 PMCID: PMC7957493 DOI: 10.3390/cancers13051000] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/15/2021] [Accepted: 02/17/2021] [Indexed: 12/12/2022] Open
Abstract
The intestinal epithelium fulfils pleiotropic functions in nutrient uptake, waste elimination, and immune surveillance while also forming a barrier against luminal toxins and gut-resident microbiota. Incessantly barraged by extraneous stresses, the intestine must continuously replenish its epithelial lining and regenerate the full gamut of specialized cell types that underpin its functions. Homeostatic remodelling is orchestrated by the intestinal stem cell (ISC) niche: a convergence of epithelial- and stromal-derived cues, which maintains ISCs in a multipotent state. Following demise of homeostatic ISCs post injury, plasticity is pervasive among multiple populations of reserve stem-like cells, lineage-committed progenitors, and/or fully differentiated cell types, all of which can contribute to regeneration and repair. Failure to restore the epithelial barrier risks seepage of toxic luminal contents, resulting in inflammation and likely predisposing to tumour formation. Here, we explore how homeostatic niche-signalling pathways are subverted in tumorigenesis, enabling ISCs to gain autonomy from niche restraints ("ISC emancipation") and transform into cancer stem cells capable of driving tumour initiation, progression, and therapy resistance. We further consider the implications of the pervasive plasticity of the intestinal epithelium for the trajectory of colorectal cancer, the emergence of distinct molecular subtypes, the propensity to metastasize, and the development of effective therapeutic strategies.
Collapse
Affiliation(s)
- Nathalie Sphyris
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK; (N.S.); (M.C.H.)
| | - Michael C. Hodder
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK; (N.S.); (M.C.H.)
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| | - Owen J. Sansom
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK; (N.S.); (M.C.H.)
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| |
Collapse
|
26
|
Non-canonical Wnt/PCP signalling regulates intestinal stem cell lineage priming towards enteroendocrine and Paneth cell fates. Nat Cell Biol 2021; 23:23-31. [PMID: 33398177 DOI: 10.1038/s41556-020-00617-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 11/27/2020] [Indexed: 02/07/2023]
Abstract
A detailed understanding of intestinal stem cell (ISC) self-renewal and differentiation is required to treat chronic intestinal diseases. However, the different models of ISC lineage hierarchy1-6 and segregation7-12 are subject to debate. Here, we have discovered non-canonical Wnt/planar cell polarity (PCP)-activated ISCs that are primed towards the enteroendocrine or Paneth cell lineage. Strikingly, integration of time-resolved lineage labelling with single-cell gene expression analysis revealed that both lineages are directly recruited from ISCs via unipotent transition states, challenging the existence of formerly predicted bi- or multipotent secretory progenitors7-12. Transitory cells that mature into Paneth cells are quiescent and express both stem cell and secretory lineage genes, indicating that these cells are the previously described Lgr5+ label-retaining cells7. Finally, Wnt/PCP-activated Lgr5+ ISCs are molecularly indistinguishable from Wnt/β-catenin-activated Lgr5+ ISCs, suggesting that lineage priming and cell-cycle exit is triggered at the post-transcriptional level by polarity cues and a switch from canonical to non-canonical Wnt/PCP signalling. Taken together, we redefine the mechanisms underlying ISC lineage hierarchy and identify the Wnt/PCP pathway as a new niche signal preceding lateral inhibition in ISC lineage priming and segregation.
Collapse
|
27
|
Li X, Tong M, Wang L, Qin Y, Yu H, Yu Y. Age-Dependent Activation and Neuronal Differentiation of Lgr5+ Basal Cells in Injured Olfactory Epithelium via Notch Signaling Pathway. Front Aging Neurosci 2020; 12:602688. [PMID: 33390928 PMCID: PMC7773941 DOI: 10.3389/fnagi.2020.602688] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/16/2020] [Indexed: 01/15/2023] Open
Abstract
Aging is an important factor affecting function of smell, leading to the degeneration of mature olfactory sensory neurons and inducing the occurrence of smell loss. The mammalian olfactory epithelium (OE) can regenerate when subjected to chemical assaults. However, this capacity is not limitless. Inactivation of globose basal cells and failure to generate sensory neurons are the main obstacles to prevent the OE regeneration. Here, we found the significant attenuation in mature sensory neuronal generation and apparent transcriptional alternation in the OE from aged mice compared with young ones. The recruitment of leucine-rich repeat-containing G-protein coupled receptor 5 (Lgr5)-positive cells in injured OE was weakened in aged mice, and more Lgr5+ cells remained quiescence in aged OE postinjury. Lineage-traced progenies from Lgr5+ cells were significantly fewer in the OE with aging. Moreover, Notch activation enhanced the neuronal regeneration in aged OE, making the regenerative capacity of aged OE comparable with that of young animals after injury. The growth and morphology of three-dimensional (3D)-cultured organoids from the OE of young and aged mice varied and was modulated by small molecules regulating the Notch signaling pathway. Thus, we concluded that activation of Lgr5+ cells in injured OE was age dependent and Notch activation could enhance the capacity of neuronal generation from Lgr5+ cells in aged OE after injury.
Collapse
Affiliation(s)
- Xuewen Li
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Meimei Tong
- Ear, Nose and Throat Department, Yuecheng People's Hospital, Shaoxing, China
| | - Li Wang
- Department of Otolaryngology, Eye, Ear, Nose and Throat Hospital, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Fudan University, Shanghai, China
| | - Yumei Qin
- School of Food Science and Bioengineering, Zhejiang Gongshang University, Hangzhou, China
| | - Hongmeng Yu
- Department of Otolaryngology, Eye, Ear, Nose and Throat Hospital, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Fudan University, Shanghai, China.,Research Units of New Technologies of Endoscopic Surgery in Skull Base Tumor, Chinese Academy of Medical Sciences, Beijing, China
| | - Yiqun Yu
- School of Life Sciences, Shanghai University, Shanghai, China.,Department of Otolaryngology, Eye, Ear, Nose and Throat Hospital, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Fudan University, Shanghai, China
| |
Collapse
|
28
|
Gan L, Liu Y, Cui DX, Pan Y, Wan M. New insight into dental epithelial stem cells: Identification, regulation, and function in tooth homeostasis and repair. World J Stem Cells 2020; 12:1327-1340. [PMID: 33312401 PMCID: PMC7705464 DOI: 10.4252/wjsc.v12.i11.1327] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/21/2020] [Accepted: 09/15/2020] [Indexed: 02/06/2023] Open
Abstract
Tooth enamel, a highly mineralized tissue covering the outermost area of teeth, is always damaged by dental caries or trauma. Tooth enamel rarely repairs or renews itself, due to the loss of ameloblasts and dental epithelial stem cells (DESCs) once the tooth erupts. Unlike human teeth, mouse incisors grow continuously due to the presence of DESCs that generate enamel-producing ameloblasts and other supporting dental epithelial lineages. The ready accessibility of mouse DESCs and wide availability of related transgenic mouse lines make mouse incisors an excellent model to examine the identity and heterogeneity of dental epithelial stem/progenitor cells; explore the regulatory mechanisms underlying enamel formation; and help answer the open question regarding the therapeutic development of enamel engineering. In the present review, we update the current understanding about the identification of DESCs in mouse incisors and summarize the regulatory mechanisms of enamel formation driven by DESCs. The roles of DESCs during homeostasis and repair are also discussed, which should improve our knowledge regarding enamel tissue engineering.
Collapse
Affiliation(s)
- Lu Gan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Ying Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Di-Xin Cui
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yue Pan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Mian Wan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
29
|
Escudero-Hernández C. Epithelial cell dysfunction in coeliac disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 358:133-164. [PMID: 33707053 DOI: 10.1016/bs.ircmb.2020.09.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The intestinal epithelium limits host-luminal interactions and maintains gut homeostasis. Breakdown of the epithelial barrier and villous atrophy are hallmarks of coeliac disease. Besides the well characterized immune-mediated epithelial damage induced in coeliac mucosa, constitutional changes and early gluten direct effects disturb intestinal epithelial cells. The subsequent modifications in key epithelial signaling pathways leads to outnumbered immature epithelial cells that, in turn, facilitate epithelial dysfunction, promote crypt hyperplasia, and increase intestinal permeability. Consequently, underlying immune cells have a greater access to gluten, which boosts the proinflammatory immune response against gluten and positively feedback the epithelial damage loop. Gluten-free diet is an indispensable treatment for coeliac disease patients, but additional therapies are under development, including those that reinforce intestinal epithelial healing. In this chapter, we provide an overview of intestinal epithelial cell disturbances that develop during gluten intake in coeliac disease mucosa.
Collapse
|
30
|
Sei Y, Feng J, Zhao X, Wank SA. Role of an active reserve stem cell subset of enteroendocrine cells in intestinal stem cell dynamics and the genesis of small intestinal neuroendocrine tumors. Am J Physiol Gastrointest Liver Physiol 2020; 319:G494-G501. [PMID: 32845170 PMCID: PMC7654644 DOI: 10.1152/ajpgi.00278.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Small intestinal neuroendocrine tumors (SI-NET) are serotonin-secreting well-differentiated neuroendocrine tumors of putative enterochromaffin (EC) cell origin. Recent studies recognize a subset of EC cells that is label-retaining at the +4 position in the crypt and functions as a reserve intestinal stem cell. Importantly, this +4 reserve EC cell subset not only contributes to regeneration of the intestinal epithelium during injury and inflammation but also to basal crypt homeostasis at a constant rate. The latter function suggests that the +4 EC cell subset serves as an active reserve stem cell via a constant rate of dedifferentiation. Characterization of early tumor formation of SI-NET, observed as crypt-based EC cell clusters in many cases of familial SI-NETs, suggests that the +4 active reserve EC cell subset is the cell of origin. This newly discovered active reserve stem cell property of EC cells can account for unique biological mechanisms and processes associated with the genesis and development of SI-NETs. The recognition of this property of the +4 active reserve EC cell subset may provide novel opportunities to explore NETs in the gastrointestinal tract and other organs.
Collapse
Affiliation(s)
- Yoshitatsu Sei
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Jianying Feng
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Xilin Zhao
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Stephen A. Wank
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
31
|
Liu Y, Chen YG. Intestinal epithelial plasticity and regeneration via cell dedifferentiation. CELL REGENERATION 2020; 9:14. [PMID: 32869114 PMCID: PMC7459029 DOI: 10.1186/s13619-020-00053-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/01/2020] [Indexed: 12/21/2022]
Abstract
The intestinal epithelium possesses a great capacity of self-renewal under normal homeostatic conditions and of regeneration upon damages. The renewal and regenerative processes are driven by intestinal stem cells (ISCs), which reside at the base of crypts and are marked by Lgr5. As Lgr5+ ISCs undergo fast cycling and are vulnerable to damages, there must be other types of cells that can replenish the lost Lgr5+ ISCs and then regenerate the damage epithelium. In addition to Lgr5+ ISCs, quiescent ISCs at the + 4 position in the crypt have been proposed to convert to Lgr5+ ISCs during regeneration. However, this “reserve stem cell” model still remains controversial. Different from the traditional view of a hierarchical organization of the intestinal epithelium, recent works support the dynamic “dedifferentiation” model, in which various cell types within the epithelium can de-differentiate to revert to the stem cell state and then regenerate the epithelium upon tissue injury. Here, we provide an overview of the cell identity and features of two distinct models and discuss the possible mechanisms underlying the intestinal epithelial plasticity.
Collapse
Affiliation(s)
- Yuan Liu
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
32
|
Chen F, Zhang Y, Hu S, Shi X, Wang Z, Deng Z, Lin L, Zhang J, Pan Y, Bai Y, Liu F, Zhang H, Shao C. TIGAR/AP-1 axis accelerates the division of Lgr5 - reserve intestinal stem cells to reestablish intestinal architecture after lethal radiation. Cell Death Dis 2020; 11:501. [PMID: 32632140 PMCID: PMC7338449 DOI: 10.1038/s41419-020-2715-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 06/18/2020] [Accepted: 06/22/2020] [Indexed: 11/30/2022]
Abstract
During radiologic or nuclear accidents, high-dose ionizing radiation (IR) can cause gastrointestinal syndrome (GIS), a deadly disorder that urgently needs effective therapy. Unfortunately, current treatments based on natural products and antioxidants have shown very limited effects in alleviating deadly GIS. Reserve intestinal stem cells (ISCs) and secretory progenitor cells are both reported to replenish damaged cells and contribute to crypt regeneration. However, the suppressed β-catenin/c-MYC axis within these slow-cycling cells leads to limited regenerative response to restore intestinal integrity during fatal accidental injury. Current study demonstrates that post-IR overexpression of TIGAR, a critical downstream target of c-MYC in mouse intestine, mounts a hyperplastic response in Bmi1-creERT+ reserve ISCs, and thus rescues mice from lethal IR exposure. Critically, by eliminating damaging reactive oxygen species (ROS) yet retaining the proliferative ROS signals, TIGAR-overexpression enhances the activity of activator protein 1, which is indispensable for initiating reserve-ISC division after lethal radiation. In addition, it is identified that TIGAR-induction exclusively gears the Lgr5− subpopulation of reserve ISCs to regenerate crypts, and intestinal TIGAR-overexpression displays equivalent intestinal reconstruction to reserve-ISC-restricted TIGAR-induction. Our findings imply that precise administrations toward Lgr5− reserve ISCs are promising strategies for unpredictable lethal injury, and TIGAR can be employed as a therapeutic target for unexpected radiation-induced GIS.
Collapse
Affiliation(s)
- Fei Chen
- Institute of Radiation Medicine, Fudan University, Shanghai, 200032, China.,State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou, 215123, China
| | - Yushuo Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou, 215123, China
| | - Songling Hu
- Institute of Radiation Medicine, Fudan University, Shanghai, 200032, China
| | - Xiaolin Shi
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou, 215123, China
| | - Zhongmin Wang
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Department of Interventional Radiology, The Third Affiliated Hospital of the Medical College of Shihezi University, Xinjiang, 832008, China
| | - Zicheng Deng
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou, 215123, China
| | - Longxin Lin
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou, 215123, China
| | - Jianghong Zhang
- Institute of Radiation Medicine, Fudan University, Shanghai, 200032, China
| | - Yan Pan
- Institute of Radiation Medicine, Fudan University, Shanghai, 200032, China
| | - Yang Bai
- Institute of Radiation Medicine, Fudan University, Shanghai, 200032, China
| | - Fenju Liu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou, 215123, China
| | - Haowen Zhang
- Institute of Radiation Medicine, Fudan University, Shanghai, 200032, China. .,State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou, 215123, China.
| | - Chunlin Shao
- Institute of Radiation Medicine, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
33
|
Blutt SE, Klein OD, Donowitz M, Shroyer N, Guha C, Estes MK. Use of organoids to study regenerative responses to intestinal damage. Am J Physiol Gastrointest Liver Physiol 2019; 317:G845-G852. [PMID: 31589468 PMCID: PMC7132322 DOI: 10.1152/ajpgi.00346.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Intestinal organoid cultures provide an in vitro model system for studying pathways and mechanisms involved in epithelial damage and repair. Derived from either embryonic or induced pluripotent stem cells or adult intestinal stem cells or tissues, these self-organizing, multicellular structures contain polarized mature cells that recapitulate both the physiology and heterogeneity of the intestinal epithelium. These cultures provide a cutting-edge technology for defining regenerative pathways that are induced following radiation or chemical damage, which directly target the cycling intestinal stem cell, or damage resulting from viral, bacterial, or parasitic infection of the epithelium. Novel signaling pathways or biological mechanisms identified from organoid studies that mediate regeneration of the epithelium following damage are likely to be important targets of preventive or therapeutic modalities to mitigate intestinal injury. The evolution of these cultures to include more components of the intestinal wall and the ability to genetically modify them are key components for defining the mechanisms that modulate epithelial regeneration.
Collapse
Affiliation(s)
- Sarah E. Blutt
- 1Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | - Ophir D. Klein
- 2Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, California,3Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, California
| | - Mark Donowitz
- 4Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland,5Department of Medicine, Gastroenterology and Hepatology Division, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Noah Shroyer
- 6Department of Medicine, Divisions of Gastroenterology and Hepatology and Infectious Diseases, Baylor College of Medicine, Houston, Texas
| | - Chandan Guha
- 7Department of Radiation Oncology, Albert Einstein, Bronx, New York
| | - Mary K. Estes
- 1Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas,6Department of Medicine, Divisions of Gastroenterology and Hepatology and Infectious Diseases, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
34
|
Kim CK, Saxena M, Maharjan K, Song JJ, Shroyer KR, Bialkowska AB, Shivdasani RA, Yang VW. Krüppel-like Factor 5 Regulates Stemness, Lineage Specification, and Regeneration of Intestinal Epithelial Stem Cells. Cell Mol Gastroenterol Hepatol 2019; 9:587-609. [PMID: 31778829 PMCID: PMC7078555 DOI: 10.1016/j.jcmgh.2019.11.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 11/15/2019] [Accepted: 11/18/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Self-renewal and multipotent differentiation are cardinal properties of intestinal stem cells (ISCs), mediated in part by WNT and NOTCH signaling. Although these pathways are well characterized, the molecular mechanisms that control the 'stemness' of ISCs are still not well defined. Here, we investigated the role of Krüppel-like factor 5 (KLF5) in regulating ISC functions. METHODS We performed studies in adult Lgr5EGFP-IRES-creERT2;Rosa26LSLtdTomato (Lgr5Ctrl) and Lgr5EGFP-IRES-creERT2;Klf5fl/fl;Rosa26LSLtdTomato (Lgr5ΔKlf5) mice. Mice were injected with tamoxifen to activate Cre recombinase, which deletes Klf5 from the intestinal epithelium in Lgr5ΔKlf5 but not Lgr5Crtl mice. In experiments involving irradiation, mice were subjected to 12 Gy total body irradiation (TBI). Tissues were collected for immunofluorescence (IF) analysis and next generation sequencing. Oganoids were derived from fluoresecence activated cell sorted- (FACS-) single cells from tamoxifen-treated Lgr5ΔKlf5 or Lgr5Crtl mice and examined by immunofluorescence stain. RESULTS Lgr5+ ISCs lacking KLF5 proliferate faster than control ISCs but fail to self-renew, resulting in a depleted ISC compartment. Transcriptome analysis revealed that Klf5-null Lgr5+ cells lose ISC identity and prematurely differentiate. Following irradiation injury, which depletes Lgr5+ ISCs, reserve Klf5-null progenitor cells fail to dedifferentiate and regenerate the epithelium. Absence of KLF5 inactivates numerous selected enhancer elements and direct transcriptional targets including canonical WNT- and NOTCH-responsive genes. Analysis of human intestinal tissues showed increased levels of KLF5 in the regenerating epithelium as compared to those of healthy controls. CONCLUSION We conclude that ISC self-renewal, lineage specification, and precursor dedifferentiation require KLF5, by its ability to regulate epigenetic and transcriptional activities of ISC-specific gene sets. These findings have the potential for modulating ISC functions by targeting KLF5 in the intestinal epithelium.
Collapse
Affiliation(s)
- Chang-Kyung Kim
- Department of Medicine, Stony Brook University Renaissance School of Medicine, Stony Brook, New York
| | - Madhurima Saxena
- Department of Medical Oncology and Center for Functional Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Kasmika Maharjan
- Department of Medicine, Stony Brook University Renaissance School of Medicine, Stony Brook, New York
| | - Jane J. Song
- Department of Medicine, Stony Brook University Renaissance School of Medicine, Stony Brook, New York
| | - Kenneth R. Shroyer
- Department of Pathology, Stony Brook University Renaissance School of Medicine, Stony Brook, New York
| | - Agnieszka B. Bialkowska
- Department of Medicine, Stony Brook University Renaissance School of Medicine, Stony Brook, New York
| | - Ramesh A. Shivdasani
- Department of Medical Oncology and Center for Functional Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Vincent W. Yang
- Department of Medicine, Stony Brook University Renaissance School of Medicine, Stony Brook, New York,Department of Physiology and Biophysics, Stony Brook University Renaissance School of Medicine, Stony Brook, New York,Correspondence Address correspondence to: Vincent W. Yang, MD, PhD, Department of Medicine, Stony Brook University School of Medicine, HSC T-16, Room 020, Stony Brook, New York, 11794. fax: (631) 444-3144.
| |
Collapse
|
35
|
Wang Y, Chiang IL, Ohara TE, Fujii S, Cheng J, Muegge BD, Ver Heul A, Han ND, Lu Q, Xiong S, Chen F, Lai CW, Janova H, Wu R, Whitehurst CE, VanDussen KL, Liu TC, Gordon JI, Sibley LD, Stappenbeck TS. Long-Term Culture Captures Injury-Repair Cycles of Colonic Stem Cells. Cell 2019; 179:1144-1159.e15. [PMID: 31708126 PMCID: PMC6904908 DOI: 10.1016/j.cell.2019.10.015] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 07/15/2019] [Accepted: 10/16/2019] [Indexed: 02/07/2023]
Abstract
The colonic epithelium can undergo multiple rounds of damage and repair, often in response to excessive inflammation. The responsive stem cell that mediates this process is unclear, in part because of a lack of in vitro models that recapitulate key epithelial changes that occur in vivo during damage and repair. Here, we identify a Hopx+ colitis-associated regenerative stem cell (CARSC) population that functionally contributes to mucosal repair in mouse models of colitis. Hopx+ CARSCs, enriched for fetal-like markers, transiently arose from hypertrophic crypts known to facilitate regeneration. Importantly, we established a long-term, self-organizing two-dimensional (2D) epithelial monolayer system to model the regenerative properties and responses of Hopx+ CARSCs. This system can reenact the "homeostasis-injury-regeneration" cycles of epithelial alterations that occur in vivo. Using this system, we found that hypoxia and endoplasmic reticulum stress, insults commonly present in inflammatory bowel diseases, mediated the cyclic switch of cellular status in this process.
Collapse
Affiliation(s)
- Yi Wang
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - I-Ling Chiang
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Takahiro E Ohara
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Satoru Fujii
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Jiye Cheng
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Brian D Muegge
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Aaron Ver Heul
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Nathan D Han
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Qiuhe Lu
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Shanshan Xiong
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Feidi Chen
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Chin-Wen Lai
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Hana Janova
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Renee Wu
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Charles E Whitehurst
- Boehringer Ingelheim Pharmaceuticals, Immunology and Respiratory Disease Research, Ridgefield, CT 06877, USA
| | - Kelli L VanDussen
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Ta-Chiang Liu
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Jeffrey I Gordon
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - L David Sibley
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Thaddeus S Stappenbeck
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| |
Collapse
|
36
|
Zwick RK, Ohlstein B, Klein OD. Intestinal renewal across the animal kingdom: comparing stem cell activity in mouse and Drosophila. Am J Physiol Gastrointest Liver Physiol 2019; 316:G313-G322. [PMID: 30543448 PMCID: PMC6415738 DOI: 10.1152/ajpgi.00353.2018] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The gastrointestinal (GI) tract renews frequently to sustain nutrient digestion and absorption in the face of consistent tissue stress. In many species, proliferative intestinal stem cells (ISCs) are responsible for the repair of the damage arising from chemical and mechanical aspects of food breakdown and exposure to pathogens. As the cellular source of all mature cell types of the intestinal epithelium throughout adulthood, ISCs hold tremendous therapeutic potential for understanding and treating GI disease in humans. This review focuses on recent advances in our understanding of ISC identity, behavior, and regulation during homeostasis and injury-induced repair, as revealed by two major animal models used to study regeneration of the small intestine: Drosophila melanogaster and Mus musculus. We emphasize recent findings from Drosophila that are likely to translate to the mammalian GI system, as well as challenging topics in mouse ISC biology that may be ideally suited for investigation in flies. For context, we begin by reviewing major physiological similarities and distinctions between the Drosophila midgut and mouse small intestine.
Collapse
Affiliation(s)
- Rachel K. Zwick
- 1Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, California
| | - Benjamin Ohlstein
- 2Department of Genetics and Development, Columbia University Medical Center, New York, New York
| | - Ophir D. Klein
- 1Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, California,3Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, California
| |
Collapse
|
37
|
Abstract
The intestinal epithelium withstands continuous mechanical, chemical and biological insults despite its single-layered, simple epithelial structure. The crypt-villus tissue architecture in combination with rapid cell turnover enables the intestine to act both as a barrier and as the primary site of nutrient uptake. Constant tissue replenishment is fuelled by continuously dividing stem cells that reside at the bottom of crypts. These cells are nurtured and protected by specialized epithelial and mesenchymal cells, and together constitute the intestinal stem cell niche. Intestinal stem cells and early progenitor cells compete for limited niche space and, therefore, the ability to retain or regain stemness. Those cells unable to do so differentiate to one of six different mature cell types and move upwards towards the villus, where they are shed into the intestinal lumen after 3-5 days. In this Review, we discuss the signals, cell types and mechanisms that control homeostasis and regeneration in the intestinal epithelium. We investigate how the niche protects and instructs intestinal stem cells, which processes drive differentiation of mature cells and how imbalance in key signalling pathways can cause human disease.
Collapse
|
38
|
Abstract
The intestinal epithelium is one the fastest renewing tissues in mammals and is endowed with extensive adaptability. The more traditional view of a hierarchical organization of the gut has recently given way to a more dynamic model in which various cell types within the intestinal epithelium can de-differentiate and function as an alternative source of stem cells upon tissue damage and stress conditions such as inflammation and tumorigenesis. Here, we will review the mechanistic principles and key players involved in intestinal plasticity and discuss potential therapeutic implications of cellular plasticity in regenerative medicine and cancer.
Collapse
|
39
|
Jones JC, Brindley CD, Elder NH, Myers MG, Rajala MW, Dekaney CM, McNamee EN, Frey MR, Shroyer NF, Dempsey PJ. Cellular Plasticity of Defa4 Cre-Expressing Paneth Cells in Response to Notch Activation and Intestinal Injury. Cell Mol Gastroenterol Hepatol 2018; 7:533-554. [PMID: 30827941 PMCID: PMC6402430 DOI: 10.1016/j.jcmgh.2018.11.004] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 11/13/2018] [Accepted: 11/16/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Loss of leucine-rich repeat-containing G-protein-coupled receptor 5-positive crypt base columnar cells provides permissive conditions for different facultative stem cell populations to dedifferentiate and repopulate the stem cell compartment. In this study, we used a defensin α4-Cre recombinase (Defa4Cre) line to define the potential of Paneth cells to dedifferentiate and contribute to intestinal stem cell (ISC) maintenance during normal homeostasis and after intestinal injury. METHODS Small intestine and enteroids from Defa4Cre;Rosa26 tandem dimer Tomato (tdTomato), a red fluoresent protein, (or Rosa26 Enhanced Yellow Fluorescent Protein (EYFP)) reporter, Notch gain-of-function (Defa4Cre;Rosa26 Notch Intracellular Domain (NICD)-ires-nuclear Green Fluorescent Protein (nGFP) and Defa4Cre;Rosa26reverse tetracycline transactivator-ires Enhanced Green Fluorescent Protein (EGFP);TetONICD), A Disintegrin and Metalloproteinase domain-containing protein 10 (ADAM10) loss-of-function (Defa4Cre;ADAM10flox/flox), and Adenomatous polyposis coli (APC) inactivation (Defa4Cre;APCflox/flox) mice were analyzed. Doxorubicin treatment was used as an acute intestinal injury model. Lineage tracing, proliferation, and differentiation were assessed in vitro and in vivo. RESULTS Defa4Cre-expressing cells are fated to become mature Paneth cells and do not contribute to ISC maintenance during normal homeostasis in vivo. However, spontaneous lineage tracing was observed in enteroids, and fluorescent-activated cell sorter-sorted Defa4Cre-marked cells showed clonogenic enteroid growth. Notch activation in Defa4Cre-expressing cells caused dedifferentiation to multipotent ISCs in vivo and was required for adenoma formation. ADAM10 deletion had no significant effect on crypt homeostasis. However, after acute doxorubicin-induced injury, Defa4Cre-expressing cells contributed to regeneration in an ADAM10-Notch-dependent manner. CONCLUSIONS Our studies have shown that Defa4Cre-expressing Paneth cells possess cellular plasticity, can dedifferentiate into multipotent stem cells upon Notch activation, and can contribute to intestinal regeneration in an acute injury model.
Collapse
Affiliation(s)
- Jennifer C. Jones
- Cell Biology, Stem Cells and Development Graduate Program, University of Colorado Medical School, Aurora, Colorado
| | - Constance D. Brindley
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Colorado Medical School, Aurora, Colorado
| | - Nicholas H. Elder
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Colorado Medical School, Aurora, Colorado
| | - Martin G. Myers
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Michael W. Rajala
- Division of Gastroenterology, Department of Digestive Disease and Transplantation, Einstein Health Network, Philadelphia, Pennsylvania
| | - Christopher M. Dekaney
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Eoin N. McNamee
- Mucosal Immunology Program, University of Colorado Medical School, Aurora, Colorado
| | - Mark R. Frey
- Saban Research Institute, Children's Hospital Los Angeles, Department of Pediatrics, Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Noah F. Shroyer
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Peter J. Dempsey
- Cell Biology, Stem Cells and Development Graduate Program, University of Colorado Medical School, Aurora, Colorado,Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Colorado Medical School, Aurora, Colorado,Correspondence Address correspondence to: Peter J. Dempsey, PhD, Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Colorado Medical School, 12700 East 19th Avenue, Building RC2 Room 6113, Aurora, Colorado 80045. fax: (303) 724-6538.
| |
Collapse
|
40
|
Bohin N, Carlson EA, Samuelson LC. Genome Toxicity and Impaired Stem Cell Function after Conditional Activation of CreER T2 in the Intestine. Stem Cell Reports 2018; 11:1337-1346. [PMID: 30449703 PMCID: PMC6294112 DOI: 10.1016/j.stemcr.2018.10.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 10/15/2018] [Accepted: 10/17/2018] [Indexed: 12/11/2022] Open
Abstract
With the tamoxifen-inducible CreERT2 system, genetic recombination can be temporally controlled in a cell-type-specific manner in intact animals, permitting dissection of the molecular underpinnings of mammalian physiology. Here we present a significant drawback to CreERT2 technology for analysis of intestinal stem cells. Using the intestine-specific Villin-CreERT2 mouse strain, we observed delayed intestinal regeneration post irradiation. Villin-CreERT2 activation was associated with DNA damage and cryptic loxP site cleavage. Analysis of stem cell-specific CreERT2 strains showed that the genome toxicity impairs function of crypt base columnar stem cells, resulting in loss of organoid initiating activity. Importantly, the stem cell impairment is short-lived, with return to normal by 7 days post tamoxifen treatment. Our findings demonstrate that mouse genetic experiments that utilize CreERT2 should consider the confounding effects of enhanced stem cell sensitivity to genome toxicity resulting from CreERT2 activation. Intestinal stem cell (ISC) toxicity induced in mice by CreERT2 activation Impaired organoid formation after activation of ISC-specific CreERT2 strains Genotoxicity and impaired crypt regeneration in Villin-CreERT2 mice Impaired ISC function and genotoxicity repaired by 7 days after activation
Collapse
Affiliation(s)
- Natacha Bohin
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA; Cellular & Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Elizabeth A Carlson
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Linda C Samuelson
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA; Cellular & Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
41
|
Bankaitis ED, Ha A, Kuo CJ, Magness ST. Reserve Stem Cells in Intestinal Homeostasis and Injury. Gastroenterology 2018; 155:1348-1361. [PMID: 30118745 PMCID: PMC7493459 DOI: 10.1053/j.gastro.2018.08.016] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/17/2018] [Accepted: 08/01/2018] [Indexed: 02/07/2023]
Abstract
Renewal of the intestinal epithelium occurs approximately every week and requires a careful balance between cell proliferation and differentiation to maintain proper lineage ratios and support absorptive, secretory, and barrier functions. We review models used to study the mechanisms by which intestinal stem cells (ISCs) fuel the rapid turnover of the epithelium during homeostasis and might support epithelial regeneration after injury. In anatomically defined zones of the crypt stem cell niche, phenotypically distinct active and reserve ISC populations are believed to support homeostatic epithelial renewal and injury-induced regeneration, respectively. However, other cell types previously thought to be committed to differentiated states might also have ISC activity and participate in regeneration. Efforts are underway to reconcile the proposed relatively strict hierarchical relationships between reserve and active ISC pools and their differentiated progeny; findings from models provide evidence for phenotypic plasticity that is common among many if not all crypt-resident intestinal epithelial cells. We discuss the challenges to consensus on ISC nomenclature, technical considerations, and limitations inherent to methodologies used to define reserve ISCs, and the need for standardized metrics to quantify and compare the relative contributions of different epithelial cell types to homeostatic turnover and post-injury regeneration. Increasing our understanding of the high-resolution genetic and epigenetic mechanisms that regulate reserve ISC function and cell plasticity will help refine these models and could affect approaches to promote tissue regeneration after intestinal injury.
Collapse
Affiliation(s)
- Eric D. Bankaitis
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC,Center for Gastrointestinal Biology & Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Andrew Ha
- Department of Medicine, Hematology Division, and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305,Department of Biology, Stanford University, Stanford, CA 94305
| | - Calvin J. Kuo
- Department of Medicine, Hematology Division, and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305,Co-Corresponding Authors: Calvin J. Kuo: , Scott T. Magness: , Calvin J. Kuo: Stanford University School of Medicine, Lokey Stem Cell Research Building G2034A, 265 Campus Drive, Stanford, CA 94305; Scott T. Magness, University of North Carolina at Chapel Hill, 111 Mason Farm Rd. CB# 7032, MBRB Rm 4337, Chapel Hill, NC, 27599
| | - Scott T. Magness
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC,Joint Departments of Biomedical Engineering, University of North Carolina at Chapel Hill/North Carolina State University, Chapel Hill, NC,Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC,Center for Gastrointestinal Biology & Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC,Co-Corresponding Authors: Calvin J. Kuo: , Scott T. Magness: , Calvin J. Kuo: Stanford University School of Medicine, Lokey Stem Cell Research Building G2034A, 265 Campus Drive, Stanford, CA 94305; Scott T. Magness, University of North Carolina at Chapel Hill, 111 Mason Farm Rd. CB# 7032, MBRB Rm 4337, Chapel Hill, NC, 27599
| |
Collapse
|
42
|
Demystifying the Differences Between Tumor-Initiating Cells and Cancer Stem Cells in Colon Cancer. CURRENT COLORECTAL CANCER REPORTS 2018. [DOI: 10.1007/s11888-018-0421-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
43
|
Sei Y, Feng J, Samsel L, White A, Zhao X, Yun S, Citrin D, McCoy JP, Sundaresan S, Hayes MM, Merchant JL, Leiter A, Wank SA. Mature enteroendocrine cells contribute to basal and pathological stem cell dynamics in the small intestine. Am J Physiol Gastrointest Liver Physiol 2018; 315:G495-G510. [PMID: 29848020 PMCID: PMC6230697 DOI: 10.1152/ajpgi.00036.2018] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Lgr5-expressing intestinal stem cells (ISCs) maintain continuous and rapid generation of the intestinal epithelium. Here, we present evidence that dedifferentiation of committed enteroendocrine cells (EECs) contributes to maintenance of the epithelium under both basal conditions and in response to injury. Lineage-tracing studies identified a subset of EECs that reside at +4 position for more than 2 wk, most of which were BrdU-label-retaining cells. Under basal conditions, cells derived from these EECs grow from the bottom of the crypt to generate intestinal epithelium according to neutral drift kinetics that is consistent with dedifferentiation of mature EECs to ISCs. The lineage tracing of EECs demonstrated reserve stem cell properties in response to radiation-induced injury with the generation of reparative EEC-derived epithelial patches. Finally, the enterochromaffin (EC) cell was the predominant EEC type participating in these stem cell dynamics. These results provide novel insights into the +4 reserve ISC hypothesis, stem cell dynamics of the intestinal epithelium, and in the development of EC-derived small intestinal tumors. NEW & NOTEWORTHY The current manuscript demonstrating that a subset of mature enteroendocrine cells (EECs), predominantly enterochromaffin cells, dedifferentiates to fully functional intestinal stem cells (ISCs) is novel, timely, and important. These cells dedifferentiate to ISCs not only in response to injury but also under basal homeostatic conditions. These novel findings provide a mechanism in which a specified cell can dedifferentiate and contribute to normal tissue plasticity as well as the development of EEC-derived intestinal tumors under pathologic conditions.
Collapse
Affiliation(s)
- Yoshitatsu Sei
- 1Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Jianying Feng
- 1Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Leigh Samsel
- 2Flow Cytometry Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Ayla White
- 3Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Xilin Zhao
- 1Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Sajung Yun
- 1Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Deborah Citrin
- 3Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - J. Philip McCoy
- 2Flow Cytometry Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Sinju Sundaresan
- 4Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Michael M. Hayes
- 4Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Juanita L. Merchant
- 5Department of Molecular and Integrative Physiology, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Andrew Leiter
- 6Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Stephen A. Wank
- 1Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
44
|
Agudo J, Park ES, Rose SA, Alibo E, Sweeney R, Dhainaut M, Kobayashi KS, Sachidanandam R, Baccarini A, Merad M, Brown BD. Quiescent Tissue Stem Cells Evade Immune Surveillance. Immunity 2018; 48:271-285.e5. [PMID: 29466757 DOI: 10.1016/j.immuni.2018.02.001] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 12/05/2017] [Accepted: 01/31/2018] [Indexed: 12/17/2022]
Abstract
Stem cells are critical for the maintenance of many tissues, but whether their integrity is maintained in the face of immunity is unclear. Here we found that cycling epithelial stem cells, including Lgr5+ intestinal stem cells, as well as ovary and mammary stem cells, were eliminated by activated T cells, but quiescent stem cells in the hair follicle and muscle were resistant to T cell killing. Immune evasion was an intrinsic property of the quiescent stem cells resulting from systemic downregulation of the antigen presentation machinery, including MHC class I and TAP proteins, and is mediated by the transactivator NLRC5. This process was reversed upon stem cell entry into the cell cycle. These studies identify a link between stem cell quiescence, antigen presentation, and immune evasion. As cancer-initiating cells can derive from stem cells, these findings may help explain how the earliest cancer cells evade immune surveillance.
Collapse
Affiliation(s)
- Judith Agudo
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Eun Sook Park
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Samuel A Rose
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Eziwoma Alibo
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Robert Sweeney
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Maxime Dhainaut
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Koichi S Kobayashi
- Hokkaido University Faculty of Medicine, Sapporo, Hokkaido 060-8638, Japan
| | - Ravi Sachidanandam
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Alessia Baccarini
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Miriam Merad
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Brian D Brown
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
45
|
Burclaff J, Mills JC. Plasticity of differentiated cells in wound repair and tumorigenesis, part II: skin and intestine. Dis Model Mech 2018; 11:11/9/dmm035071. [PMID: 30171151 PMCID: PMC6177008 DOI: 10.1242/dmm.035071] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Recent studies have identified and begun to characterize the roles of regenerative cellular plasticity in many organs. In Part I of our two-part Review, we discussed how cells reprogram following injury to the stomach and pancreas. We introduced the concept of a conserved cellular program, much like those governing division and death, which may allow mature cells to become regenerative. This program, paligenosis, is likely necessary to help organs repair the numerous injuries they face over the lifetime of an organism; however, we also postulated that rounds of paligenosis and redifferentiation may allow long-lived cells to accumulate and store oncogenic mutations, and could thereby contribute to tumorigenesis. We have termed the model wherein differentiated cells can store mutations and then unmask them upon cell cycle re-entry the ‘cyclical hit’ model of tumorigenesis. In the present Review (Part II), we discuss these concepts, and cell plasticity as a whole, in the skin and intestine. Although differentiation and repair are arguably more thoroughly studied in skin and intestine than in stomach and pancreas, it is less clear how mature skin and intestinal cells contribute to tumorigenesis. Moreover, we conclude our Review by discussing plasticity in all four organs, and look for conserved mechanisms and concepts that might help advance our knowledge of tumor formation and advance the development of therapies for treating or preventing cancers that might be shared across multiple organs. Summary: This final installment of a two-part Review discusses how cycles of dedifferentiation and redifferentiation can promote tumorigenesis in the skin and intestine, showing how plasticity in these continuously renewing tissues might contribute to tumorigenesis.
Collapse
Affiliation(s)
- Joseph Burclaff
- Division of Gastroenterology, Departments of Medicine, Pathology & Immunology, and Developmental Biology, Washington University, St Louis, MO 63110, USA
| | - Jason C Mills
- Division of Gastroenterology, Departments of Medicine, Pathology & Immunology, and Developmental Biology, Washington University, St Louis, MO 63110, USA
| |
Collapse
|
46
|
Yu S, Tong K, Zhao Y, Balasubramanian I, Yap GS, Ferraris RP, Bonder EM, Verzi MP, Gao N. Paneth Cell Multipotency Induced by Notch Activation following Injury. Cell Stem Cell 2018; 23:46-59.e5. [PMID: 29887318 PMCID: PMC6035085 DOI: 10.1016/j.stem.2018.05.002] [Citation(s) in RCA: 207] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 01/08/2018] [Accepted: 05/04/2018] [Indexed: 02/08/2023]
Abstract
Paneth cells are post-mitotic intestinal epithelial cells supporting the stem cell niche and mucosal immunity. Paneth cell pathologies are observed in various gastrointestinal diseases, but their plasticity and response to genomic and environmental challenges remain unclear. Using a knockin allele engineered at the mouse Lyz1 locus, we performed detailed Paneth cell-lineage tracing. Irradiation induced a subset of Paneth cells to proliferate and differentiate into villus epithelial cells. RNA sequencing (RNA-seq) revealed that Paneth cells sorted from irradiated mice acquired a stem cell-like transcriptome; when cultured in vitro, these individual Paneth cells formed organoids. Irradiation activated Notch signaling, and forced expression of Notch intracellular domain (NICD) in Paneth cells, but not Wnt/β-catenin pathway activation, induced their dedifferentiation. This study documents Paneth cell plasticity, particularly their ability to participate in epithelial replenishment following stem cell loss, adding to a growing body of knowledge detailing the molecular pathways controlling injury-induced regeneration.
Collapse
Affiliation(s)
- Shiyan Yu
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Kevin Tong
- Department of Genetics, Rutgers University, Piscataway, NJ 08854, USA
| | - Yanlin Zhao
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07101, USA
| | | | - George S Yap
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07101, USA
| | - Ronaldo P Ferraris
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ 07101, USA
| | - Edward M Bonder
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Michael P Verzi
- Department of Genetics, Rutgers University, Piscataway, NJ 08854, USA; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
| | - Nan Gao
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA.
| |
Collapse
|
47
|
Mouse Intestinal Krt15+ Crypt Cells Are Radio-Resistant and Tumor Initiating. Stem Cell Reports 2018; 10:1947-1958. [PMID: 29805107 PMCID: PMC5993649 DOI: 10.1016/j.stemcr.2018.04.022] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 04/23/2018] [Accepted: 04/24/2018] [Indexed: 12/17/2022] Open
Abstract
Two principal stem cell pools orchestrate the rapid cell turnover in the intestinal epithelium. Rapidly cycling Lgr5+ stem cells are intercalated between the Paneth cells at the crypt base (CBCs) and injury-resistant reserve stem cells reside above the crypt base. The intermediate filament Keratin 15 (Krt15) marks either stem cells or long-lived progenitor cells that contribute to tissue repair in the hair follicle or the esophageal epithelium. Herein, we demonstrate that Krt15 labels long-lived and multipotent cells in the small intestinal crypt by lineage tracing. Krt15+ crypt cells display self-renewal potential in vivo and in 3D organoid cultures. Krt15+ crypt cells are resistant to high-dose radiation and contribute to epithelial regeneration following injury. Notably, loss of the tumor suppressor Apc in Krt15+ cells leads to adenoma and adenocarcinoma formation. These results indicate that Krt15 marks long-lived, multipotent, and injury-resistant crypt cells that may function as a cell of origin in intestinal cancer. Krt15 marks multipotent and self-renewing crypt cells in the mouse small intestine Krt15+ crypt cells are radio-resistant and contribute to regeneration following injury Apc loss in Krt15+ cells leads to intestinal adenoma and adenocarcinoma formation Krt15+ cells may function as a cell of origin in intestinal cancer
Collapse
|
48
|
Diverse mechanisms for endogenous regeneration and repair in mammalian organs. Nature 2018; 557:322-328. [PMID: 29769669 DOI: 10.1038/s41586-018-0073-7] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 03/07/2018] [Indexed: 12/11/2022]
Abstract
Mammalian organs comprise an extraordinary diversity of cell and tissue types. Regenerative organs, such as the skin and gastrointestinal tract, use resident stem cells to maintain tissue function. Organs with a lower cellular turnover, such as the liver and lungs, mostly rely on proliferation of committed progenitor cells. In many organs, injury reveals the plasticity of both resident stem cells and differentiated cells. The ability of resident cells to maintain and repair organs diminishes with age, whereas, paradoxically, the risk of cancer increases. New therapeutic approaches aim to harness cell plasticity for tissue repair and regeneration while avoiding the risk of malignant transformation of cells.
Collapse
|
49
|
Sáenz JB, Mills JC. Acid and the basis for cellular plasticity and reprogramming in gastric repair and cancer. Nat Rev Gastroenterol Hepatol 2018; 15:257-273. [PMID: 29463907 PMCID: PMC6016373 DOI: 10.1038/nrgastro.2018.5] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Subjected to countless daily injuries, the stomach still functions as a remarkably efficient digestive organ and microbial filter. In this Review, we follow the lead of the earliest gastroenterologists who were fascinated by the antiseptic and digestive powers of gastric secretions. We propose that it is easiest to understand how the stomach responds to injury by stressing the central role of the most important gastric secretion, acid. The stomach follows two basic patterns of adaptation. The superficial response is a pattern whereby the surface epithelial cells migrate and rapidly proliferate to repair erosions induced by acid or other irritants. The stomach can also adapt through a glandular response when the source of acid is lost or compromised (that is, the process of oxyntic atrophy). We primarily review the mechanisms governing the glandular response, which is characterized by a metaplastic change in cellular differentiation known as spasmolytic polypeptide-expressing metaplasia (SPEM). We propose that the stomach, like other organs, exhibits marked cellular plasticity: the glandular response involves reprogramming mature cells to serve as auxiliary stem cells that replace lost cells. Unfortunately, such plasticity might mean that the gastric epithelium undergoes cycles of differentiation and de-differentiation that increase the risk of accumulating cancer-predisposing mutations.
Collapse
Affiliation(s)
- José B. Sáenz
- Division of Gastroenterology, Department of Internal Medicine, Washington University School of Medicine
| | - Jason C. Mills
- Division of Gastroenterology, Department of Internal Medicine, Washington University School of Medicine
- Department of Developmental Biology, Washington University School of Medicine
- Department of Pathology and Immunology, Washington University School of Medicine
| |
Collapse
|
50
|
Testa U, Pelosi E, Castelli G. Colorectal cancer: genetic abnormalities, tumor progression, tumor heterogeneity, clonal evolution and tumor-initiating cells. Med Sci (Basel) 2018; 6:E31. [PMID: 29652830 PMCID: PMC6024750 DOI: 10.3390/medsci6020031] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 03/24/2018] [Accepted: 04/03/2018] [Indexed: 02/08/2023] Open
Abstract
Colon cancer is the third most common cancer worldwide. Most colorectal cancer occurrences are sporadic, not related to genetic predisposition or family history; however, 20-30% of patients with colorectal cancer have a family history of colorectal cancer and 5% of these tumors arise in the setting of a Mendelian inheritance syndrome. In many patients, the development of a colorectal cancer is preceded by a benign neoplastic lesion: either an adenomatous polyp or a serrated polyp. Studies carried out in the last years have characterized the main molecular alterations occurring in colorectal cancers, showing that the tumor of each patient displays from two to eight driver mutations. The ensemble of molecular studies, including gene expression studies, has led to two proposed classifications of colorectal cancers, with the identification of four/five non-overlapping groups. The homeostasis of the rapidly renewing intestinal epithelium is ensured by few stem cells present at the level of the base of intestinal crypts. Various experimental evidence suggests that colorectal cancers may derive from the malignant transformation of intestinal stem cells or of intestinal cells that acquire stem cell properties following malignant transformation. Colon cancer stem cells seem to be involved in tumor chemoresistance, radioresistance and relapse.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy.
| | - Elvira Pelosi
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy.
| | - Germana Castelli
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy.
| |
Collapse
|