1
|
Wu N, Sun Q, Yang L, Sun H, Zhou Z, Hu Q, Li C, Wang D, Zhang L, Hu Y, Cong X. HDAC3 and Snail2 complex promotes melanoma metastasis by epigenetic repression of IGFBP3. Int J Biol Macromol 2025; 300:140310. [PMID: 39864688 DOI: 10.1016/j.ijbiomac.2025.140310] [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: 09/11/2024] [Revised: 01/09/2025] [Accepted: 01/23/2025] [Indexed: 01/28/2025]
Abstract
The treatment of metastatic melanoma has long posed a complex challenge within clinical practice. Previous studies have found that EMT transcription factors are essential in the development of various cancers through their induction of EMT. Here, we demonstrate that Snail2 expression is dramatically increased in melanoma and is associated with an adverse prognosis. Elevated Snail2 in melanoma cells enhanced migratory and invasive capabilities in vitro and in vivo. Furthermore, RNA-Seq analysis revealed a significant reduction of IGFBP3 expression in melanoma cells overexpressing Snail2. IGFBP3 might mitigate the Snail2's ability to promote melanoma metastasis via the PI3K-AKT pathway. Moreover, Snail2 and HDAC3 collaborate to suppress IGFBP3 transcription through H3K4 deacetylation and H4K5 delactylation. Additionally, the combination of HDAC3 and p-GSK-3β inhibitors significantly improved the treatment outcomes for lung metastasis in melanoma in vivo. The results of our study indicate that Snail2, HDAC3, and IGFBP3 play significant roles in melanoma progression and represent promising therapeutic targets.
Collapse
Affiliation(s)
- Nan Wu
- Department of Biobank, China-Japan Union Hospital of Jilin University, Changchun 130033, China; Phase I Clinical Trial Research Laboratory, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Qian Sun
- Department of Biobank, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Liehao Yang
- Department of Biobank, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Hongyan Sun
- Department of Biobank, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Zilong Zhou
- Department of Biobank, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Qianying Hu
- Department of Biobank, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Chunyi Li
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun 130033, China
| | - Dongxu Wang
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun 130033, China
| | - Ling Zhang
- Key Laboratory of Pathobiology, Ministry of Education, and Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Yue Hu
- Department of Biobank, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| | - Xianling Cong
- Department of Biobank, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| |
Collapse
|
2
|
Wan J, Xu Y, Qi T, Xue X, Li Y, Huang M, Guo Y, Guo Q, Lu Y, Huang Y. AG73-GelMA/AlgMA hydrogels provide a stable microenvironment for the generation of pancreatic progenitor organoids. J Nanobiotechnology 2025; 23:149. [PMID: 40016740 PMCID: PMC11866579 DOI: 10.1186/s12951-025-03266-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 02/21/2025] [Indexed: 03/01/2025] Open
Abstract
Patient specific induced pluripotent stem cells (iPSCs) derived β cells represent an effective means for disease modeling and autologous diabetes cell replacement therapy. In this study, an AG73-5%gelatin methacryloyl (GelMA) /2% alginate methacrylate (AlgMA) hydrogel was employed to generate pancreatic progenitor (PP) organoids and improve stem cell-derived β (SC-β) cell differentiation protocol. The laminin-derived homolog AG73, which mimics certain cell‒matrix interactions, facilitates AKT signaling pathway activation to promote PDX1+/NKX6.1+ PP organoid formation and effectively modulates subsequent epithelial-mesenchymal transition (EMT) in the endocrine lineage. The 5%GelMA/2%AlgMA hydrogel mimics the physiological stiffness of the pancreas, providing the optimal mechanical stress and spatial structure for PP organoid differentiation. The Syndecan-4 (SDC4)-ITGAV complex plays a pivotal role in the early stages of pancreatic development by facilitating the formation of SOX9+/PDX1+ bipotent PPs. Our findings demonstrate that AG73-GelMA/AlgMA hydrogel-derived SC-β cells exhibit enhanced insulin secretion and accelerated hyperglycemia reversal in vivo. This study presents a cost-effective, stable, and efficient alternative for the comprehensive 3D culture of SC-β cells in vitro by mitigating the uncertainties associated with conventional culture methods.
Collapse
Affiliation(s)
- Jian Wan
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Yang Xu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Tianmu Qi
- Medical School of Nantong University, Nantong, China
| | - Xiaoxia Xue
- Department of Nephrology, Rugao Hospital of Traditional Chinese Medicine, Nantong, China
| | - Yuxi Li
- Medical School of Nantong University, Nantong, China
| | - Minjie Huang
- Medical School of Nantong University, Nantong, China
| | - Yuchen Guo
- Medical School of Nantong University, Nantong, China
| | - Qingsong Guo
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China.
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China.
| | - Yuhua Lu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China.
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China.
| | - Yan Huang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China.
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China.
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co- Innovation Center of Neuroregeneration, Nantong University, Nantong, China.
| |
Collapse
|
3
|
Haq N, Toczyska KW, Wilson ME, Jacobs M, Zhao M, Lei Y, Shen Z, Pearson JA, Persaud SJ, Pullen TJ, Bewick GA. Reformed islets: a long-term primary cell platform for exploring mouse and human islet biology. Cell Death Discov 2024; 10:480. [PMID: 39580467 PMCID: PMC11585622 DOI: 10.1038/s41420-024-02234-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 10/19/2024] [Accepted: 10/30/2024] [Indexed: 11/25/2024] Open
Abstract
Pancreatic islets are 3D micro-organs that maintain β-cell functionality through cell-cell and cell-matrix communication. While primary islets, the gold standard for in vitro models, have a short culture life of approximately 1-2 weeks, we developed a novel protocol that employs reformed islets following dispersion coupled with a fine-tuned culture environment. Reformed islets exhibit physiological characteristics similar to primary islets, enabling high-resolution imaging and repeated functional assessment. Unlike other in vitro platforms, reformed islets retain an immune population, allowing the study of interactions between β cells and resident and infiltrating immune cells. Analyses showed that reformed islets have a similar composition and cytoarchitecture to primary islets, including macrophages and T cells, and can secrete insulin in response to glucose. Reformed islets exhibited partial dedifferentiation compared to native islets but were otherwise transcriptionally similar. The reformed islets offer a useful platform for studying diabetes pathology and can recapitulate both T1DM and T2DM disease milieus, providing an advantage over other models, such as mouse and human β-cell lines, which lack the input of non-β-endocrine cells and immune cell crosstalk.
Collapse
Affiliation(s)
- N Haq
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, Diabetes Endocrinology and Obesity Clinical Academic Partnership, King's College London and King's Health Partners, Guy's Campus, London, UK
| | - K W Toczyska
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, Diabetes Endocrinology and Obesity Clinical Academic Partnership, King's College London and King's Health Partners, Guy's Campus, London, UK
| | - M E Wilson
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, Diabetes Endocrinology and Obesity Clinical Academic Partnership, King's College London and King's Health Partners, Guy's Campus, London, UK
| | - M Jacobs
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, Diabetes Endocrinology and Obesity Clinical Academic Partnership, King's College London and King's Health Partners, Guy's Campus, London, UK
| | - Min Zhao
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, Diabetes Endocrinology and Obesity Clinical Academic Partnership, King's College London and King's Health Partners, Guy's Campus, London, UK
| | - Y Lei
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, Diabetes Endocrinology and Obesity Clinical Academic Partnership, King's College London and King's Health Partners, Guy's Campus, London, UK
| | - Z Shen
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, Diabetes Endocrinology and Obesity Clinical Academic Partnership, King's College London and King's Health Partners, Guy's Campus, London, UK
| | - J A Pearson
- Diabetes Research Group, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - S J Persaud
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, Diabetes Endocrinology and Obesity Clinical Academic Partnership, King's College London and King's Health Partners, Guy's Campus, London, UK
| | - T J Pullen
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, Diabetes Endocrinology and Obesity Clinical Academic Partnership, King's College London and King's Health Partners, Guy's Campus, London, UK
| | - G A Bewick
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, Diabetes Endocrinology and Obesity Clinical Academic Partnership, King's College London and King's Health Partners, Guy's Campus, London, UK.
| |
Collapse
|
4
|
Zhao J, Liang S, Cen HH, Li Y, Baker RK, Ruprai B, Gao G, Zhang C, Ren H, Tang C, Chen L, Liu Y, Lynn FC, Johnson JD, Kieffer TJ. PDX1+ cell budding morphogenesis in a stem cell-derived islet spheroid system. Nat Commun 2024; 15:5894. [PMID: 39003281 PMCID: PMC11246529 DOI: 10.1038/s41467-024-50109-2] [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: 01/06/2024] [Accepted: 07/01/2024] [Indexed: 07/15/2024] Open
Abstract
Remarkable advances in protocol development have been achieved to manufacture insulin-secreting islets from human pluripotent stem cells (hPSCs). Distinct from current approaches, we devised a tunable strategy to generate islet spheroids enriched for major islet cell types by incorporating PDX1+ cell budding morphogenesis into staged differentiation. In this process that appears to mimic normal islet morphogenesis, the differentiating islet spheroids organize with endocrine cells that are intermingled or arranged in a core-mantle architecture, accompanied with functional heterogeneity. Through in vitro modelling of human pancreas development, we illustrate the importance of PDX1 and the requirement for EphB3/4 signaling in eliciting cell budding morphogenesis. Using this new approach, we model Mitchell-Riley syndrome with RFX6 knockout hPSCs illustrating unexpected morphogenesis defects in the differentiation towards islet cells. The tunable differentiation system and stem cell-derived islet models described in this work may facilitate addressing fundamental questions in islet biology and probing human pancreas diseases.
Collapse
Affiliation(s)
- Jia Zhao
- Life Sciences Institute, Departments of Cellular & Physiological Sciences and Surgery, University of British Columbia, Vancouver, BC, Canada.
| | - Shenghui Liang
- Life Sciences Institute, Departments of Cellular & Physiological Sciences and Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Haoning Howard Cen
- Life Sciences Institute, Departments of Cellular & Physiological Sciences and Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Yanjun Li
- Institute of Molecular Medicine, School of Future Technology, National Biomedical Imaging Center, Peking University, Beijing, China
| | - Robert K Baker
- Life Sciences Institute, Departments of Cellular & Physiological Sciences and Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Balwinder Ruprai
- Life Sciences Institute, Departments of Cellular & Physiological Sciences and Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Guang Gao
- Imaging Core Facility, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Chloe Zhang
- Life Sciences Institute, Departments of Cellular & Physiological Sciences and Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Huixia Ren
- Institute of Molecular Medicine, School of Future Technology, National Biomedical Imaging Center, Peking University, Beijing, China
- Center for Quantitative Biology, Peking University, Beijing, China
| | - Chao Tang
- Center for Quantitative Biology, Peking University, Beijing, China
| | - Liangyi Chen
- Institute of Molecular Medicine, School of Future Technology, National Biomedical Imaging Center, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Yanmei Liu
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, South China Normal University, 510631, Guangzhou, China
- Institute for Brain Research and Rehabilitation, and Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, 510631, Guangzhou, China
| | - Francis C Lynn
- BC Children's Hospital Research Institute, Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - James D Johnson
- Life Sciences Institute, Departments of Cellular & Physiological Sciences and Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Timothy J Kieffer
- Life Sciences Institute, Departments of Cellular & Physiological Sciences and Surgery, University of British Columbia, Vancouver, BC, Canada.
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
5
|
Edri S, Rosenthal V, Ginsburg O, Newman Frisch A, Pierreux CE, Sharon N, Levenberg S. 3D model of mouse embryonic pancreas and endocrine compartment using stem cell-derived mesoderm and pancreatic progenitors. iScience 2024; 27:109959. [PMID: 38832019 PMCID: PMC11144751 DOI: 10.1016/j.isci.2024.109959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/21/2024] [Accepted: 05/08/2024] [Indexed: 06/05/2024] Open
Abstract
The developing mouse pancreas is surrounded by mesoderm compartments providing signals that induce pancreas formation. Most pancreatic organoid protocols lack this mesoderm niche and only partially capture the pancreatic cell repertoire. This work aims to generate pancreatic aggregates by differentiating mouse embryonic stem cells (mESCs) into mesoderm progenitors (MPs) and pancreas progenitors (PPs), without using Matrigel. First, mESCs were differentiated into epiblast stem cells (EpiSCs) to enhance the PP differentiation rate. Next, PPs and MPs aggregated together giving rise to various pancreatic cell types, including endocrine, acinar, and ductal cells, and to endothelial cells. Single-cell RNA sequencing analysis revealed a larger endocrine population within the PP + MP aggregates, as compared to PPs alone or PPs in Matrigel aggregates. The PP + MP aggregate gene expression signatures and its endocrine population percentage closely resembled those of the endocrine population found in the mouse embryonic pancreas, which holds promise for studying pancreas development.
Collapse
Affiliation(s)
- Shlomit Edri
- Faculty of Biomedical Engineering, Technion – Israel Institute of Technology, Haifa 3200003, Israel
| | - Vardit Rosenthal
- Faculty of Biomedical Engineering, Technion – Israel Institute of Technology, Haifa 3200003, Israel
| | - Or Ginsburg
- Faculty of Biomedical Engineering, Technion – Israel Institute of Technology, Haifa 3200003, Israel
| | - Abigail Newman Frisch
- Faculty of Biomedical Engineering, Technion – Israel Institute of Technology, Haifa 3200003, Israel
| | | | - Nadav Sharon
- Faculty of Biology, Technion – Israel Institute of Technology, Haifa 3200003, Israel
| | - Shulamit Levenberg
- Faculty of Biomedical Engineering, Technion – Israel Institute of Technology, Haifa 3200003, Israel
| |
Collapse
|
6
|
Xu Y, Mao S, Fan H, Wan J, Wang L, Zhang M, Zhu S, Yuan J, Lu Y, Wang Z, Yu B, Jiang Z, Huang Y. LINC MIR503HG Controls SC-β Cell Differentiation and Insulin Production by Targeting CDH1 and HES1. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305631. [PMID: 38243869 PMCID: PMC10987150 DOI: 10.1002/advs.202305631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 01/03/2024] [Indexed: 01/22/2024]
Abstract
Stem cell-derived pancreatic progenitors (SC-PPs), as an unlimited source of SC-derived β (SC-β) cells, offers a robust tool for diabetes treatment in stem cell-based transplantation, disease modeling, and drug screening. Whereas, PDX1+/NKX6.1+ PPs enhances the subsequent endocrine lineage specification and gives rise to glucose-responsive SC-β cells in vivo and in vitro. To identify the regulators that promote induction efficiency and cellular function maturation, single-cell RNA-sequencing is performed to decipher the transcriptional landscape during PPs differentiation. The comprehensive evaluation of functionality demonstrated that manipulating LINC MIR503HG using CRISPR in PP cell fate decision can improve insulin synthesis and secretion in mature SC-β cells, without effects on liver lineage specification. Importantly, transplantation of MIR503HG-/- SC-β cells in recipients significantly restored blood glucose homeostasis, accompanied by serum C-peptide release and an increase in body weight. Mechanistically, by releasing CtBP1 occupying the CDH1 and HES1 promoters, the decrease in MIR503HG expression levels provided an excellent extracellular niche and appropriate Notch signaling activation for PPs following differentiation. Furthermore, this exhibited higher crucial transcription factors and mature epithelial markers in CDH1High expressed clusters. Altogether, these findings highlighted MIR503HG as an essential and exclusive PP cell fate specification regulator with promising therapeutic potential for patients with diabetes.
Collapse
Affiliation(s)
- Yang Xu
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
- Center of Gallbladder DiseaseShanghai East HospitalInstitute of Gallstone DiseaseSchool of MedicineTongji UniversityShanghai200092China
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
| | - Susu Mao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of EducationNMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology ProductsCo‐innovation Center of NeuroregenerationNantong UniversityNantong226001China
| | - Haowen Fan
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
| | - Jian Wan
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
| | - Lin Wang
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
- Department of Graduate SchoolDalian Medical UniversityDalianLiaoning116000China
| | - Mingyu Zhang
- Department of Nuclear MedicineBeijing Friendship HospitalAffiliated to Capital Medical UniversityBeijing100050China
| | - Shajun Zhu
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
| | - Jin Yuan
- Department of Endocrinology and MetabolismAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
| | - Yuhua Lu
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
| | - Zhiwei Wang
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
| | - Bin Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of EducationNMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology ProductsCo‐innovation Center of NeuroregenerationNantong UniversityNantong226001China
| | - Zhaoyan Jiang
- Center of Gallbladder DiseaseShanghai East HospitalInstitute of Gallstone DiseaseSchool of MedicineTongji UniversityShanghai200092China
| | - Yan Huang
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of EducationNMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology ProductsCo‐innovation Center of NeuroregenerationNantong UniversityNantong226001China
| |
Collapse
|
7
|
Yang RM, Song SY, Wu FY, Yang RF, Shen YT, Tu PH, Wang Z, Zhang JX, Cheng F, Gao GQ, Liang J, Guo MM, Yang L, Zhou Y, Zhao SX, Zhan M, Song HD. Myeloid cells interact with a subset of thyrocytes to promote their migration and follicle formation through NF-κB. Nat Commun 2023; 14:8082. [PMID: 38057310 PMCID: PMC10700497 DOI: 10.1038/s41467-023-43895-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 11/23/2023] [Indexed: 12/08/2023] Open
Abstract
The pathogenesis of thyroid dysgenesis (TD) is not well understood. Here, using a combination of single-cell RNA and spatial transcriptome sequencing, we identify a subgroup of NF-κB-activated thyrocytes located at the center of thyroid tissues in postnatal mice, which maintained a partially mesenchymal phenotype. These cells actively protruded out of the thyroid primordium and generated new follicles in zebrafish embryos through continuous tracing. Suppressing NF-κB signaling affected thyrocyte migration and follicle formation, leading to a TD-like phenotype in both mice and zebrafish. Interestingly, during thyroid folliculogenesis, myeloid cells played a crucial role in promoting thyrocyte migration by maintaining close contact and secreting TNF-α. We found that cebpa mutant zebrafish, in which all myeloid cells were depleted, exhibited thyrocyte migration defects. Taken together, our results suggest that myeloid-derived TNF-α-induced NF-κB activation plays a critical role in promoting the migration of vertebrate thyrocytes for follicle generation.
Collapse
Affiliation(s)
- Rui-Meng Yang
- Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shi-Yang Song
- Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng-Yao Wu
- Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui-Feng Yang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan-Ting Shen
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping-Hui Tu
- Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Wang
- Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun-Xiu Zhang
- Department of Endocrinology, Maternal and Child Health Institute of Bozhou, Bozhou, China
| | - Feng Cheng
- Department of Laboratory Medicine, Fujian Children's Hospital, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Guan-Qi Gao
- Department of Endocrinology, The Linyi People's Hospital, Linyi, Shandong Province, China
| | - Jun Liang
- Department of Endocrinology, The Central Hospital of Xuzhou Affiliated to Xuzhou Medical College, Xuzhou, China
| | - Miao-Miao Guo
- Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liu Yang
- Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Zhou
- Stem Cell Program, Boston Children's Hospital and Harvard Stem Cell Institute, Boston, MA, USA
- Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Boston, MA, USA
| | - Shuang-Xia Zhao
- Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Ming Zhan
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Huai-Dong Song
- Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
8
|
Vacondio D, Nogueira Pinto H, Coenen L, Mulder IA, Fontijn R, van Het Hof B, Fung WK, Jongejan A, Kooij G, Zelcer N, Rozemuller AJ, de Vries HE, de Wit NM. Liver X receptor alpha ensures blood-brain barrier function by suppressing SNAI2. Cell Death Dis 2023; 14:781. [PMID: 38016947 PMCID: PMC10684660 DOI: 10.1038/s41419-023-06316-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 11/09/2023] [Accepted: 11/15/2023] [Indexed: 11/30/2023]
Abstract
In Alzheimer's disease (AD) more than 50% of the patients are affected by capillary cerebral amyloid-angiopathy (capCAA), which is characterized by localized hypoxia, neuro-inflammation and loss of blood-brain barrier (BBB) function. Moreover, AD patients with or without capCAA display increased vessel number, indicating a reactivation of the angiogenic program. The molecular mechanism(s) responsible for BBB dysfunction and angiogenesis in capCAA is still unclear, preventing a full understanding of disease pathophysiology. The Liver X receptor (LXR) family, consisting of LXRα and LXRβ, was reported to inhibit angiogenesis and particularly LXRα was shown to secure BBB stability, suggesting a major role in vascular function. In this study, we unravel the regulatory mechanism exerted by LXRα to preserve BBB integrity in human brain endothelial cells (BECs) and investigate its role during pathological conditions. We report that LXRα ensures BECs identity via constitutive inhibition of the transcription factor SNAI2. Accordingly, deletion of brain endothelial LXRα is associated with impaired DLL4-NOTCH signalling, a critical signalling pathway involved in vessel sprouting. A similar response was observed when BECs were exposed to hypoxia, with concomitant LXRα decrease and SNAI2 increase. In support of our cell-based observations, we report a general increase in vascular SNAI2 in the occipital cortex of AD patients with and without capCAA. Importantly, SNAI2 strongly associated with vascular amyloid-beta deposition and angiopoietin-like 4, a marker for hypoxia. In hypoxic capCAA vessels, the expression of LXRα may decrease leading to an increased expression of SNAI2, and consequently BECs de-differentiation and sprouting. Our findings indicate that LXRα is essential for BECs identity, thereby securing BBB stability and preventing aberrant angiogenesis. These results uncover a novel molecular pathway essential for BBB identity and vascular homeostasis providing new insights on the vascular pathology affecting AD patients.
Collapse
Affiliation(s)
- D Vacondio
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, De Boelelaan 1108, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - H Nogueira Pinto
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, De Boelelaan 1108, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - L Coenen
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, De Boelelaan 1108, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
- Biomedical Primate Research Centre, Department of Neurobiology and Aging, Rijswijk, the Netherlands
| | - I A Mulder
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, De Boelelaan 1108, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
- Amsterdam UMC location University of Amsterdam, Department of Biomedical Engineering and Physics, Meibergdreef 9, Amsterdam, the Netherlands
| | - R Fontijn
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, De Boelelaan 1108, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - B van Het Hof
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, De Boelelaan 1108, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - W K Fung
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, De Boelelaan 1108, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - A Jongejan
- Amsterdam UMC location University of Amsterdam, Epidemiology and Data Science, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Public Health, Methodology, Amsterdam, The Netherlands
- Amsterdam Infection and Immunity, Inflammatory Diseases, Amsterdam, The Netherlands
| | - G Kooij
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, De Boelelaan 1108, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - N Zelcer
- Amsterdam UMC location University of Amsterdam Department of Medical Biochemistry, Meibergdreef 9, Amsterdam, the Netherlands
- Amsterdam UMC location University of Amsterdam, Cardiovascular Sciences and Gastroenterology and Metabolism, Meibergdreef 9, Amsterdam, the Netherlands
| | - A J Rozemuller
- Amsterdam Neuroscience, Amsterdam, the Netherlands
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Pathology, De Boelelaan 1117, Amsterdam, the Netherlands
| | - H E de Vries
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, De Boelelaan 1108, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - N M de Wit
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, De Boelelaan 1108, Amsterdam, the Netherlands.
- Amsterdam Neuroscience, Amsterdam, the Netherlands.
| |
Collapse
|
9
|
Ma Z, Zhang X, Zhong W, Yi H, Chen X, Zhao Y, Ma Y, Song E, Xu T. Deciphering early human pancreas development at the single-cell level. Nat Commun 2023; 14:5354. [PMID: 37660175 PMCID: PMC10475098 DOI: 10.1038/s41467-023-40893-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 08/15/2023] [Indexed: 09/04/2023] Open
Abstract
Understanding pancreas development can provide clues for better treatments of pancreatic diseases. However, the molecular heterogeneity and developmental trajectory of the early human pancreas are poorly explored. Here, we performed large-scale single-cell RNA sequencing and single-cell assay for transposase accessible chromatin sequencing of human embryonic pancreas tissue obtained from first-trimester embryos. We unraveled the molecular heterogeneity, developmental trajectories and regulatory networks of the major cell types. The results reveal that dorsal pancreatic multipotent cells in humans exhibit different gene expression patterns than ventral multipotent cells. Pancreato-biliary progenitors that generate ventral multipotent cells in humans were identified. Notch and MAPK signals from mesenchymal cells regulate the differentiation of multipotent cells into trunk and duct cells. Notably, we identified endocrine progenitor subclusters with different differentiation potentials. Although the developmental trajectories are largely conserved between humans and mice, some distinct gene expression patterns have also been identified. Overall, we provide a comprehensive landscape of early human pancreas development to understand its lineage transitions and molecular complexity.
Collapse
Affiliation(s)
- Zhuo Ma
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaofei Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, 570102, China
| | - Wen Zhong
- Science for Life Laboratory, Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, 581 83, Sweden
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Hongyan Yi
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, 570102, China
| | - Xiaowei Chen
- Center for High Throughput Sequencing, Core Facility for Protein Research, Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yinsuo Zhao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yanlin Ma
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, 570102, China.
| | - Eli Song
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Tao Xu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
- Guangzhou Laboratory, Guangzhou, 510005, China.
- Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, China.
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250062, China.
| |
Collapse
|
10
|
Francis M, Bhaskar S, Komanduri S, Sheshadri P, Prasanna J, Kumar A. Deubiquitinase USP1 influences the dedifferentiation of mouse pancreatic β-cells. iScience 2023; 26:106771. [PMID: 37250303 PMCID: PMC10214732 DOI: 10.1016/j.isci.2023.106771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 03/08/2023] [Accepted: 04/24/2023] [Indexed: 05/31/2023] Open
Abstract
Loss of insulin-secreting β-cells in diabetes may be either due to apoptosis or dedifferentiation of β-cell mass. The ubiquitin-proteasome system comprising E3 ligase and deubiquitinases (DUBs) controls several aspects of β-cell functions. In this study, screening for key DUBs identified USP1 to be specifically involved in dedifferentiation process. Inhibition of USP1 either by genetic intervention or small-molecule inhibitor ML323 restored epithelial phenotype of β-cells, but not with inhibition of other DUBs. In absence of dedifferentiation cues, overexpression of USP1 was sufficient to induce dedifferentiation in β-cells; mechanistic insight showed USP1 to mediate its effect via modulating the expression of inhibitor of differentiation (ID) 2. In an in vivo streptozotocin (STZ)-induced dedifferentiation mouse model system, administering ML323 alleviated hyperglycemic state. Overall, this study identifies USP1 to be involved in dedifferentiation of β-cells and its inhibition may have a therapeutic application of reducing β-cell loss during diabetes.
Collapse
Affiliation(s)
- Meenal Francis
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India
| | - Smitha Bhaskar
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India
| | - Saarwani Komanduri
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India
| | - Preethi Sheshadri
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India
| | - Jyothi Prasanna
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India
| | - Anujith Kumar
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
11
|
Bele S, Wokasch AS, Gannon M. Epigenetic modulation of cell fate during pancreas development. TRENDS IN DEVELOPMENTAL BIOLOGY 2023; 16:1-27. [PMID: 38873037 PMCID: PMC11173269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Epigenetic modifications to DNA and its associated proteins affect cell plasticity and cell fate restrictions throughout embryonic development. Development of the vertebrate pancreas is characterized by initial is an over-lapping expression of a set of transcriptional regulators in a defined region of the posterior foregut endoderm that collectively promote pancreas progenitor specification and proliferation. As development progresses, these transcription factors segregate into distinct pancreatic lineages, with some being maintained in specific subsets of terminally differentiated pancreas cell types throughout adulthood. Here we describe the progressive stages and cell fate restrictions that occur during pancreas development and the relevant known epigenetic regulatory events that drive the dynamic expression patterns of transcription factors that regulate pancreas development. In addition, we highlight how changes in epigenetic marks can affect susceptibility to pancreas diseases (such as diabetes), adult pancreas cell plasticity, and the ability to derive replacement insulin-producing β cells for the treatment of diabetes.
Collapse
Affiliation(s)
- Shilpak Bele
- Department of Medicine, Vanderbilt University Medical Center, 2213 Garland Avenue, Nashville, TN, 37232, USA
| | - Anthony S. Wokasch
- Department of Cell and Developmental Biology, Vanderbilt University, 2213 Garland Avenue, Nashville, TN, 37232, USA
| | - Maureen Gannon
- Department of Medicine, Vanderbilt University Medical Center, 2213 Garland Avenue, Nashville, TN, 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University, 2213 Garland Avenue, Nashville, TN, 37232, USA
- Department of Veterans Affairs Tennessee Valley Authority, Research Division, 1310 24 Avenue South, Nashville, TN, 37212, USA
- Department of Molecular Physiology and Biophysics, 2213 Garland Avenue, Nashville, TN, 37232, USA
| |
Collapse
|
12
|
Bakhti M, Bastidas-Ponce A, Tritschler S, Czarnecki O, Tarquis-Medina M, Nedvedova E, Jaki J, Willmann SJ, Scheibner K, Cota P, Salinno C, Boldt K, Horn N, Ueffing M, Burtscher I, Theis FJ, Coskun Ü, Lickert H. Synaptotagmin-13 orchestrates pancreatic endocrine cell egression and islet morphogenesis. Nat Commun 2022; 13:4540. [PMID: 35927244 PMCID: PMC9352765 DOI: 10.1038/s41467-022-31862-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/05/2022] [Indexed: 12/12/2022] Open
Abstract
During pancreas development endocrine cells leave the ductal epithelium to form the islets of Langerhans, but the morphogenetic mechanisms are incompletely understood. Here, we identify the Ca2+-independent atypical Synaptotagmin-13 (Syt13) as a key regulator of endocrine cell egression and islet formation. We detect specific upregulation of the Syt13 gene and encoded protein in endocrine precursors and the respective lineage during islet formation. The Syt13 protein is localized to the apical membrane of endocrine precursors and to the front domain of egressing endocrine cells, marking a previously unidentified apical-basal to front-rear repolarization during endocrine precursor cell egression. Knockout of Syt13 impairs endocrine cell egression and skews the α-to-β-cell ratio. Mechanistically, Syt13 is a vesicle trafficking protein, transported via the microtubule cytoskeleton, and interacts with phosphatidylinositol phospholipids for polarized localization. By internalizing a subset of plasma membrane proteins at the front domain, including α6β4 integrins, Syt13 modulates cell-matrix adhesion and allows efficient endocrine cell egression. Altogether, these findings uncover an unexpected role for Syt13 as a morphogenetic driver of endocrinogenesis and islet formation.
Collapse
Affiliation(s)
- Mostafa Bakhti
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Aimée Bastidas-Ponce
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Sophie Tritschler
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
- Technical University of Munich, School of Life Sciences Weihenstephan, Freising, Germany
| | - Oliver Czarnecki
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Technische Universität München, School of Medicine, München, Germany
| | - Marta Tarquis-Medina
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Technische Universität München, School of Medicine, München, Germany
| | - Eva Nedvedova
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum Munich at the University Clinic Carl Gustav Carus, TU Dresden, Dresden, Germany
- SOTIO a.s, Jankovcova 1518/2, Prague, Czech Republic
| | - Jessica Jaki
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Stefanie J Willmann
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
| | - Katharina Scheibner
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Perla Cota
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Technische Universität München, School of Medicine, München, Germany
| | - Ciro Salinno
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Technische Universität München, School of Medicine, München, Germany
| | - Karsten Boldt
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Nicola Horn
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Marius Ueffing
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Ingo Burtscher
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
- Technical University of Munich, Department of Mathematics, Garching b, Munich, Germany
| | - Ünal Coskun
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum Munich at the University Clinic Carl Gustav Carus, TU Dresden, Dresden, Germany
- Center of Membrane Biochemistry and Lipid Research, Carl Gustav Carus School of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Technische Universität München, School of Medicine, München, Germany.
| |
Collapse
|
13
|
van Roey R, Brabletz T, Stemmler MP, Armstark I. Deregulation of Transcription Factor Networks Driving Cell Plasticity and Metastasis in Pancreatic Cancer. Front Cell Dev Biol 2021; 9:753456. [PMID: 34888306 PMCID: PMC8650502 DOI: 10.3389/fcell.2021.753456] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/27/2021] [Indexed: 12/15/2022] Open
Abstract
Pancreatic cancer is a very aggressive disease with 5-year survival rates of less than 10%. The constantly increasing incidence and stagnant patient outcomes despite changes in treatment regimens emphasize the requirement of a better understanding of the disease mechanisms. Challenges in treating pancreatic cancer include diagnosis at already progressed disease states due to the lack of early detection methods, rapid acquisition of therapy resistance, and high metastatic competence. Pancreatic ductal adenocarcinoma, the most prevalent type of pancreatic cancer, frequently shows dominant-active mutations in KRAS and TP53 as well as inactivation of genes involved in differentiation and cell-cycle regulation (e.g. SMAD4 and CDKN2A). Besides somatic mutations, deregulated transcription factor activities strongly contribute to disease progression. Specifically, transcriptional regulatory networks essential for proper lineage specification and differentiation during pancreas development are reactivated or become deregulated in the context of cancer and exacerbate progression towards an aggressive phenotype. This review summarizes the recent literature on transcription factor networks and epigenetic gene regulation that play a crucial role during tumorigenesis.
Collapse
Affiliation(s)
- Ruthger van Roey
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Marc P Stemmler
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Isabell Armstark
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
14
|
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
|
15
|
Schmidtlein PM, Volz C, Hackel A, Thürling I, Castven D, Braun R, Wellner UF, Konukiewitz B, Riemekasten G, Lehnert H, Marquardt JU, Ungefroren H. Activation of a Ductal-to-Endocrine Transdifferentiation Transcriptional Program in the Pancreatic Cancer Cell Line PANC-1 Is Controlled by RAC1 and RAC1b through Antagonistic Regulation of Stemness Factors. Cancers (Basel) 2021; 13:cancers13215541. [PMID: 34771704 PMCID: PMC8583136 DOI: 10.3390/cancers13215541] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/29/2021] [Accepted: 11/02/2021] [Indexed: 01/29/2023] Open
Abstract
Simple Summary For patients with metastatic pancreatic ductal adenocarcinoma (PDAC) there is currently no cure; hence, novel effective therapies are desperately needed. Among PDAC patients, the tumor cell phenotypes are heterogeneous as a result of epithelial–mesenchymal transition, a process that endows them with the ability to metastasize, resist therapy, and generate cancer stem cells. The heightened plasticity of quasimesenchymal and potentially metastatic tumor cells may, however, also be exploited for their transdifferentiation into benign, highly differentiated or post-mitotic cells. Since PDAC patients often have a need for replacement of insulin-producing cells, conversion of tumor cells with a ductal/exocrine origin to endocrine β cell-like cells is an attractive therapeutic option. Successful transdifferentiation into insulin-producing cells has been reported for the quasimesenchymal cell line PANC-1; however, the mechanistic basis of this transformation process is unknown. Here, we show that the small GTPases, RAC1 and RAC1b control this process by antagonistic regulation of stemness genes. Abstract Epithelial–mesenchymal transition (EMT) is a driving force for tumor growth, metastatic spread, therapy resistance, and the generation of cancer stem cells (CSCs). However, the regained stem cell character may also be exploited for therapeutic conversion of aggressive tumor cells to benign, highly differentiated cells. The PDAC-derived quasimesenchymal-type cell lines PANC-1 and MIA PaCa-2 have been successfully transdifferentiated to endocrine precursors or insulin-producing cells; however, the underlying mechanism of this increased plasticity remains elusive. Given its crucial role in normal pancreatic endocrine development and tumor progression, both of which involve EMT, we analyzed here the role of the small GTPase RAC1. Ectopic expression in PANC-1 cells of dominant negative or constitutively active mutants of RAC1 activation blocked or enhanced, respectively, the cytokine-induced activation of a ductal-to-endocrine transdifferentiation transcriptional program (deTDtP) as revealed by induction of the NEUROG3, INS, SLC2A2, and MAFA genes. Conversely, ectopic expression of RAC1b, a RAC1 splice isoform and functional antagonist of RAC1-driven EMT, decreased the deTDtP, while genetic knockout of RAC1b dramatically increased it. We further show that inhibition of RAC1 activation attenuated pluripotency marker expression and self-renewal ability, while depletion of RAC1b dramatically enhanced stemness features and clonogenic potential. Finally, rescue experiments involving pharmacological or RNA interference-mediated inhibition of RAC1 or RAC1b, respectively, confirmed that both RAC1 isoforms control the deTDtP in an opposite manner. We conclude that RAC1 and RAC1b antagonistically control growth factor-induced activation of an endocrine transcriptional program and the generation of CSCs in quasimesenchymal PDAC cells. Our results have clinical implications for PDAC patients, who in addition to eradication of tumor cells have a need for replacement of insulin-producing cells.
Collapse
Affiliation(s)
- Paula Marie Schmidtlein
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, D-23538 Lübeck, Germany; (P.M.S.); (C.V.); (I.T.); (D.C.); (J.-U.M.)
| | - Clara Volz
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, D-23538 Lübeck, Germany; (P.M.S.); (C.V.); (I.T.); (D.C.); (J.-U.M.)
| | - Alexander Hackel
- Department of Rheumatology and Clinical Immunology, University Hospital Schleswig-Holstein, Campus Lübeck, D-23538 Lübeck, Germany; (A.H.); (G.R.)
| | - Isabel Thürling
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, D-23538 Lübeck, Germany; (P.M.S.); (C.V.); (I.T.); (D.C.); (J.-U.M.)
| | - Darko Castven
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, D-23538 Lübeck, Germany; (P.M.S.); (C.V.); (I.T.); (D.C.); (J.-U.M.)
| | - Rüdiger Braun
- Clinic for Surgery, University Hospital Schleswig-Holstein, Campus Lübeck, D-23538 Lübeck, Germany; (R.B.); (U.F.W.)
| | - Ulrich Friedrich Wellner
- Clinic for Surgery, University Hospital Schleswig-Holstein, Campus Lübeck, D-23538 Lübeck, Germany; (R.B.); (U.F.W.)
| | - Björn Konukiewitz
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Kiel, D-24105 Kiel, Germany;
| | - Gabriela Riemekasten
- Department of Rheumatology and Clinical Immunology, University Hospital Schleswig-Holstein, Campus Lübeck, D-23538 Lübeck, Germany; (A.H.); (G.R.)
| | | | - Jens-Uwe Marquardt
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, D-23538 Lübeck, Germany; (P.M.S.); (C.V.); (I.T.); (D.C.); (J.-U.M.)
| | - Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, D-23538 Lübeck, Germany; (P.M.S.); (C.V.); (I.T.); (D.C.); (J.-U.M.)
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Kiel, D-24105 Kiel, Germany;
- Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, D-23538 Lübeck, Germany
- Correspondence:
| |
Collapse
|
16
|
Title AC, Silva PN, Godbersen S, Hasenöhrl L, Stoffel M. The miR-200-Zeb1 axis regulates key aspects of β-cell function and survival in vivo. Mol Metab 2021; 53:101267. [PMID: 34116231 PMCID: PMC8258987 DOI: 10.1016/j.molmet.2021.101267] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/24/2021] [Accepted: 06/03/2021] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE The miR-200-Zeb1 axis regulates the epithelial-to-mesenchymal transition (EMT), differentiation, and resistance to apoptosis. A better understanding of these processes in diabetes is highly relevant, as β-cell dedifferentiation and apoptosis contribute to the loss of functional β-cell mass and diabetes progression. Furthermore, EMT promotes the loss of β-cell identity in the in vitro expansion of human islets. Though the miR-200 family has previously been identified as a regulator of β-cell apoptosis in vivo, studies focusing on Zeb1 are lacking. The aim of this study was thus to investigate the role of Zeb1 in β-cell function and survival in vivo. METHODS miR-200 and Zeb1 are involved in a double-negative feedback loop. We characterized a mouse model in which miR-200 binding sites in the Zeb1 3'UTR are mutated (Zeb1200), leading to a physiologically relevant upregulation of Zeb1 mRNA expression. The role of Zeb1 was investigated in this model via metabolic tests and analysis of isolated islets. Further insights into the distinct contributions of the miR-200 and Zeb1 branches of the feedback loop were obtained by crossing the Zeb1200 allele into a background of miR-141-200c overexpression. RESULTS Mild Zeb1 derepression in vivo led to broad transcriptional changes in islets affecting β-cell identity, EMT, insulin secretion, cell-cell junctions, the unfolded protein response (UPR), and the response to ER stress. The aggregation and insulin secretion of dissociated islets of mice homozygous for the Zeb1200 mutation (Zeb1200M) were impaired, and Zeb1200M islets were resistant to thapsigargin-induced ER stress ex vivo. Zeb1200M mice had increased circulating proinsulin levels but no overt metabolic phenotype, reflecting the strong compensatory ability of islets to maintain glucose homeostasis. CONCLUSIONS This study signifies the importance of the miR-200-Zeb1 axis in regulating key aspects of β-cell function and survival. A better understanding of this axis is highly relevant in developing therapeutic strategies for inducing β-cell redifferentiation and maintaining β-cell identity in in vitro islet expansion.
Collapse
Affiliation(s)
- Alexandra C Title
- Institute of Molecular Health Sciences (IMHS), ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland; Competence Center Personalized Medicine, ETH Zürich, Voltastrasse 24, 8044, Zürich, Switzerland
| | - Pamuditha N Silva
- Institute of Molecular Health Sciences (IMHS), ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland
| | - Svenja Godbersen
- Institute of Molecular Health Sciences (IMHS), ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland
| | - Lynn Hasenöhrl
- Institute of Molecular Health Sciences (IMHS), ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland
| | - Markus Stoffel
- Institute of Molecular Health Sciences (IMHS), ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland; Competence Center Personalized Medicine, ETH Zürich, Voltastrasse 24, 8044, Zürich, Switzerland; Medical Faculty, University of Zürich, 8091, Zürich, Switzerland.
| |
Collapse
|
17
|
A Comparative Endocrine Trans-Differentiation Approach to Pancreatic Ductal Adenocarcinoma Cells with Different EMT Phenotypes Identifies Quasi-Mesenchymal Tumor Cells as Those with Highest Plasticity. Cancers (Basel) 2021; 13:cancers13184663. [PMID: 34572891 PMCID: PMC8466512 DOI: 10.3390/cancers13184663] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/01/2021] [Accepted: 09/14/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancer types with the quasi-mesenchymal (QM) subtype of PDAC having the worst prognosis. De-differentiation of the ductal tumor cells to a mesenchymal phenotype occurs as a result of epithelial–mesenchymal transition (EMT), a process associated with the acquisition of stem cell traits. While QM tumor cells are highly metastatic and drug-resistant, their increased plasticity opens a window of opportunity for trans-differentiation into non-malignant pancreatic cells. In this study we compared established PDAC-derived cell lines of either epithelial (E) or QM phenotype for their potential to be differentiated to pancreatic endocrine cells. We found that QM cells responded more strongly than E cells with transcriptional activation of a pancreatic progenitor or pancreatic β cell-specific program. Our results bear strong implications for a novel type of targeted therapy, namely EMT-based trans-differentiation of highly metastatic PDAC cells in vivo to non-malignant endocrine cells. Abstract Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive and therapy-resistant cancer types which is largely due to tumor heterogeneity, cancer cell de-differentiation, and early metastatic spread. The major molecular subtypes of PDAC are designated classical/epithelial (E) and quasi-mesenchymal (QM) subtypes, with the latter having the worst prognosis. Epithelial–mesenchymal transition (EMT) and the reverse process, mesenchymal-epithelial transition (MET), are involved in regulating invasion/metastasis and stem cell generation in cancer cells but also early pancreatic endocrine differentiation or de-differentiation of adult pancreatic islet cells in vitro, suggesting that pancreatic ductal exocrine and endocrine cells share common EMT programs. Using a panel of PDAC-derived cell lines classified by epithelial/mesenchymal expression as either E or QM, we compared their trans-differentiation (TD) potential to endocrine progenitor or β cell-like cells since studies with human pancreatic cancer cells for possible future TD therapy in PDAC patients are not available so far. We observed that QM cell lines responded strongly to TD culture using as inducers 5′-aza-2′-deoxycytidine or growth factors/cytokines, while their E counterparts were refractory or showed only a weak response. Moreover, the gain of plasticity was associated with a decrease in proliferative and migratory activities and was directly related to epigenetic changes acquired during selection of a metastatic phenotype as revealed by TD experiments using the paired isogenic COLO 357-L3.6pl model. Our data indicate that a QM phenotype in PDAC coincides with increased plasticity and heightened trans-differentiation potential to activate a pancreatic β cell-specific transcriptional program. We strongly assume that this specific biological feature has potential to be exploited clinically in TD-based therapy to convert metastatic PDAC cells into less malignant or even benign cells.
Collapse
|
18
|
Zhang A, Aslam H, Sharma N, Warmflash A, Fakhouri WD. Conservation of Epithelial-to-Mesenchymal Transition Process in Neural Crest Cells and Metastatic Cancer. Cells Tissues Organs 2021; 210:151-172. [PMID: 34218225 DOI: 10.1159/000516466] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 04/12/2021] [Indexed: 11/19/2022] Open
Abstract
Epithelial to mesenchymal transition (EMT) is a highly conserved cellular process in several species, from worms to humans. EMT plays a fundamental role in early embryogenesis, wound healing, and cancer metastasis. For neural crest cell (NCC) development, EMT typically results in forming a migratory and potent cell population that generates a wide variety of cell and tissue, including cartilage, bone, connective tissue, endocrine cells, neurons, and glia amongst many others. The degree of conservation between the signaling pathways that regulate EMT during development and metastatic cancer (MC) has not been fully established, despite ample studies. This systematic review and meta-analysis dissects the major signaling pathways involved in EMT of NCC development and MC to unravel the similarities and differences. While the FGF, TGFβ/BMP, SHH, and NOTCH pathways have been rigorously investigated in both systems, the EGF, IGF, HIPPO, Factor Receptor Superfamily, and their intracellular signaling cascades need to be the focus of future NCC studies. In general, meta-analyses of the associated signaling pathways show a significant number of overlapping genes (particularly ligands, transcription regulators, and targeted cadherins) involved in each signaling pathway of both systems without stratification by body segments and cancer type. Lack of stratification makes it difficult to meaningfully evaluate the intracellular downstream effectors of each signaling pathway. Finally, pediatric neuroblastoma and melanoma are NCC-derived malignancies, which emphasize the importance of uncovering the EMT events that convert NCC into treatment-resistant malignant cells.
Collapse
Affiliation(s)
- April Zhang
- Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Hira Aslam
- Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Neha Sharma
- Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Aryeh Warmflash
- Department of Biosciences, Rice University, Houston, Texas, USA
| | - Walid D Fakhouri
- Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, Texas, USA.,Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center, Houston, Texas, USA
| |
Collapse
|
19
|
Pancreas morphogenesis and homeostasis depends on tightly regulated Zeb1 levels in epithelial cells. Cell Death Discov 2021; 7:138. [PMID: 34112759 PMCID: PMC8192546 DOI: 10.1038/s41420-021-00522-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/26/2021] [Accepted: 05/13/2021] [Indexed: 02/08/2023] Open
Abstract
The pancreas is comprised of exocrine and endocrine compartments releasing digestive enzymes into the duodenum and regulating blood glucose levels by insulin and glucagon release. Tissue homeostasis is depending on transcription factor networks, involving Ptf1α, Ngn3, Nkx6.1, and Sox9, which are already activated during organogenesis. However, proper organ function is challenged by diets of high sugar and fat content, increasing the risk of type 2 diabetes and other disorders. A detailed understanding of processes that are important for homeostasis and are impaired during type 2 diabetes is lacking. Here, we show that Zeb1—a transcription factor known for its pivotal role in epithelial-mesenchymal transition, cell plasticity, and metastasis in cancer—is expressed at low levels in epithelial cells of the pancreas and is crucial for organogenesis and pancreas function. Loss of Zeb1 in these cells result in an increase of islet mass, impaired glucose tolerance, and sensitizes to develop liver and pancreas steatosis during diabetes and obesity. Interestingly, moderate overexpression of Zeb1 results in severe pancreas agenesis and lethality after birth, due to islet insufficiency and lack of acinar structures. We show that Zeb1 induction interferes with proper differentiation, cell survival, and proliferation during pancreas formation, due to deregulated expression of endocrine-specific transcription factors. In summary, our analysis suggests a novel role of Zeb1 for homeostasis in epithelial cells that is indispensable for pancreas morphogenesis and proper organ function involving a tight regulation of Zeb1 expression.
Collapse
|
20
|
Liu H, Li R, Liao HK, Min Z, Wang C, Yu Y, Shi L, Dan J, Hayek A, Martinez Martinez L, Nuñez Delicado E, Izpisua Belmonte JC. Chemical combinations potentiate human pluripotent stem cell-derived 3D pancreatic progenitor clusters toward functional β cells. Nat Commun 2021; 12:3330. [PMID: 34099664 PMCID: PMC8184986 DOI: 10.1038/s41467-021-23525-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 05/02/2021] [Indexed: 11/09/2022] Open
Abstract
Human pluripotent stem cell (hPSC)-derived pancreatic β cells are an attractive cell source for treating diabetes. However, current derivation methods remain inefficient, heterogeneous, and cell line dependent. To address these issues, we first devised a strategy to efficiently cluster hPSC-derived pancreatic progenitors into 3D structures. Through a systematic study, we discovered 10 chemicals that not only retain the pancreatic progenitors in 3D clusters but also enhance their potentiality towards NKX6.1+/INS+ β cells. We further systematically screened signaling pathway modulators in the three steps from pancreatic progenitors toward β cells. The implementation of all these strategies and chemical combinations resulted in generating β cells from different sources of hPSCs with high efficiency. The derived β cells are functional and can reverse hyperglycemia in mice within two weeks. Our protocol provides a robust platform for studying human β cells and developing hPSC-derived β cells for cell replacement therapy.
Collapse
Affiliation(s)
- Haisong Liu
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, California, USA
| | - Ronghui Li
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, California, USA
| | - Hsin-Kai Liao
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, California, USA
| | - Zheying Min
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Chao Wang
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, California, USA
| | - Yang Yu
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, California, USA.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Lei Shi
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, California, USA
| | - Jiameng Dan
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, California, USA
| | - Alberto Hayek
- Department of Pediatrics, UCSD-Medical School, La Jolla, California, USA
| | | | | | | |
Collapse
|
21
|
Wu Y, Aegerter P, Nipper M, Ramjit L, Liu J, Wang P. Hippo Signaling Pathway in Pancreas Development. Front Cell Dev Biol 2021; 9:663906. [PMID: 34079799 PMCID: PMC8165189 DOI: 10.3389/fcell.2021.663906] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/12/2021] [Indexed: 12/17/2022] Open
Abstract
The Hippo signaling pathway is a vital regulator of pancreatic development and homeostasis, directing cell fate decisions, morphogenesis, and adult pancreatic cellular plasticity. Through loss-of-function research, Hippo signaling has been found to play key roles in maintaining the proper balance between progenitor cell renewal, proliferation, and differentiation in pancreatic organogenesis. Other studies suggest that overactivation of YAP, a downstream effector of the pathway, promotes ductal cell development and suppresses endocrine cell fate specification via repression of Ngn3. After birth, disruptions in Hippo signaling have been found to lead to de-differentiation of acinar cells and pancreatitis-like phenotype. Further, Hippo signaling directs pancreatic morphogenesis by ensuring proper cell polarization and branching. Despite these findings, the mechanisms through which Hippo governs cell differentiation and pancreatic architecture are yet to be fully understood. Here, we review recent studies of Hippo functions in pancreatic development, including its crosstalk with NOTCH, WNT/β-catenin, and PI3K/Akt/mTOR signaling pathways.
Collapse
Affiliation(s)
- Yifan Wu
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, TX, United States.,Department of Obstetrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Pauline Aegerter
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, TX, United States
| | - Michael Nipper
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, TX, United States
| | - Logan Ramjit
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, TX, United States
| | - Jun Liu
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, TX, United States
| | - Pei Wang
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, TX, United States
| |
Collapse
|
22
|
Abstract
Over 50 years after its discovery in early chick embryos, the concept of epithelial-mesenchymal transition (EMT) is now widely applied to morphogenetic studies in both physiological and pathological contexts. Indeed, the EMT field has witnessed exponential growth in recent years, driven primarily by a rapid expansion of cancer-oriented EMT research. This has led to EMT-based therapeutic interventions that bear the prospect of fighting cancer, and has given developmental biologists new impetus to investigate EMT phenomena more closely and to find suitable models to address emerging EMT-related questions. Here, and in the accompanying poster, I provide a brief summary of the current status of EMT research and give an overview of EMT models that have been used in developmental studies. I also highlight dynamic epithelialization and de-epithelialization events that are involved in many developmental processes and that should be considered to provide a broader perspective of EMT. Finally, I put forward a set of criteria to separate morphogenetic phenomena that are EMT-related from those that are not.
Collapse
Affiliation(s)
- Guojun Sheng
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan
| |
Collapse
|
23
|
Zhang X, Ma Z, Song E, Xu T. Islet organoid as a promising model for diabetes. Protein Cell 2021; 13:239-257. [PMID: 33751396 PMCID: PMC7943334 DOI: 10.1007/s13238-021-00831-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 01/22/2021] [Indexed: 02/06/2023] Open
Abstract
Studies on diabetes have long been hampered by a lack of authentic disease models that, ideally, should be unlimited and able to recapitulate the abnormalities involved in the development, structure, and function of human pancreatic islets under pathological conditions. Stem cell-based islet organoids faithfully recapitulate islet development in vitro and provide large amounts of three-dimensional functional islet biomimetic materials with a morphological structure and cellular composition similar to those of native islets. Thus, islet organoids hold great promise for modeling islet development and function, deciphering the mechanisms underlying the onset of diabetes, providing an in vitro human organ model for infection of viruses such as SARS-CoV-2, and contributing to drug screening and autologous islet transplantation. However, the currently established islet organoids are generally immature compared with native islets, and further efforts should be made to improve the heterogeneity and functionality of islet organoids, making it an authentic and informative disease model for diabetes. Here, we review the advances and challenges in the generation of islet organoids, focusing on human pluripotent stem cell-derived islet organoids, and the potential applications of islet organoids as disease models and regenerative therapies for diabetes.
Collapse
Affiliation(s)
- Xiaofei Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Zhuo Ma
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Eli Song
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China. .,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Tao Xu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China. .,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China. .,Guangzhou Regenerative Medicine and Health Guangdong Laboratory (Bioland Laboratory), Guangzhou, 510005, China.
| |
Collapse
|
24
|
Wang D, Wang J, Bai L, Pan H, Feng H, Clevers H, Zeng YA. Long-Term Expansion of Pancreatic Islet Organoids from Resident Procr + Progenitors. Cell 2020; 180:1198-1211.e19. [PMID: 32200801 DOI: 10.1016/j.cell.2020.02.048] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 12/03/2019] [Accepted: 02/21/2020] [Indexed: 12/31/2022]
Abstract
It has generally proven challenging to produce functional β cells in vitro. Here, we describe a previously unidentified protein C receptor positive (Procr+) cell population in adult mouse pancreas through single-cell RNA sequencing (scRNA-seq). The cells reside in islets, do not express differentiation markers, and feature epithelial-to-mesenchymal transition characteristics. By genetic lineage tracing, Procr+ islet cells undergo clonal expansion and generate all four endocrine cell types during adult homeostasis. Sorted Procr+ cells, representing ∼1% of islet cells, can robustly form islet-like organoids when cultured at clonal density. Exponential expansion can be maintained over long periods by serial passaging, while differentiation can be induced at any time point in culture. β cells dominate in differentiated islet organoids, while α, δ, and PP cells occur at lower frequencies. The organoids are glucose-responsive and insulin-secreting. Upon transplantation in diabetic mice, these organoids reverse disease. These findings demonstrate that the adult mouse pancreatic islet contains a population of Procr+ endocrine progenitors.
Collapse
Affiliation(s)
- Daisong Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Jingqiang Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Lanyue Bai
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Hong Pan
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Hua Feng
- Omics Core, Bio-Med Big Data Center, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hans Clevers
- Hubrecht Institute and Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre Utrecht, Utrecht, the Netherlands; Utrecht University and Princess Maxima Center, Utrecht, the Netherlands
| | - Yi Arial Zeng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
25
|
The histopathological and molecular features of breast carcinoma with tumour budding-a systematic review and meta-analysis. Breast Cancer Res Treat 2020; 183:503-514. [PMID: 32710280 DOI: 10.1007/s10549-020-05810-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 07/15/2020] [Indexed: 01/04/2023]
Abstract
PURPOSE Tumour budding (TB) is an adverse histological feature in many epithelial cancers. It is thought to represent epithelial-mesenchymal transition, a key step in the metastatic process. The significance of TB in breast carcinoma (BC) remains unclear. The aim of this study is to investigate the relationship between TB and other histological and molecular features of BC. METHODS A systematic search was performed to identify studies that compared features of BC based on the presence or absence of high-grade TB. Dichotomous variables were pooled as odds ratios (OR) using the Der Simonian-Laird method. Quality assessment of the included studies was performed using the Newcastle-Ottawa scale (NOS). RESULTS Seven studies with a total of 1040 patients (high-grade TB n = 519, 49.9%; low-grade/absent TB n = 521, 50.1%) were included. A moderate to high risk of bias was noted. The median NOS was 7 (range 6-8). High-grade TB was significantly associated with lymph node metastasis (OR 2.32, 95% c.i. 1.77 to 3.03, P < 0.001) and lymphovascular invasion (OR 3.08, 95% c.i. 2.13 to 4.47, P < 0.001). With regard to molecular subtypes, there was an increased likelihood of high-grade TB in oestrogen (OR 1.66, 95% c.i. 1.21 to 2.29, P = 0.002) and progesterone receptor-positive (OR 1.48, 95% c.i. 1.09 to 2.02, P = 0.01) tumours. In contrast, triple-negative breast cancer had a reduced incidence of high-grade TB (OR 0.46, 95% c.i. 0.30 to 0.72, P = 0.0006). CONCLUSION High-grade TB is enriched in hormone receptor-positive BC and is associated with known adverse prognostic variables. TB may offer new insights into the metastatic process of BC.
Collapse
|
26
|
Dybala MP, Kuznetsov A, Motobu M, Hendren-Santiago BK, Philipson LH, Chervonsky AV, Hara M. Integrated Pancreatic Blood Flow: Bidirectional Microcirculation Between Endocrine and Exocrine Pancreas. Diabetes 2020; 69:1439-1450. [PMID: 32198213 PMCID: PMC7306124 DOI: 10.2337/db19-1034] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 03/09/2020] [Indexed: 02/06/2023]
Abstract
The pancreatic islet is a highly vascularized endocrine micro-organ. The unique architecture of rodent islets, a so-called core-mantle arrangement seen in two-dimensional images, led researchers to seek functional implications for islet hormone secretion. Three models of islet blood flow were previously proposed, all based on the assumption that islet microcirculation occurs in an enclosed structure. Recent electrophysiological and molecular biological studies using isolated islets also presumed unidirectional flow. Using intravital analysis of the islet microcirculation in mice, we found that islet capillaries were continuously integrated to those in the exocrine pancreas, which made the islet circulation rather open, not self-contained. Similarly in human islets, the capillary structure was integrated with pancreatic microvasculature in its entirety. Thus, islet microcirculation has no relation to islet cytoarchitecture, which explains its well-known variability throughout species. Furthermore, tracking fluorescent-labeled red blood cells at the endocrine-exocrine interface revealed bidirectional blood flow, with similar variability in blood flow speed in both the intra- and extra-islet vasculature. To date, the endocrine and exocrine pancreas have been studied separately by different fields of investigators. We propose that the open circulation model physically links both endocrine and exocrine parts of the pancreas as a single organ through the integrated vascular network.
Collapse
Affiliation(s)
| | | | - Maki Motobu
- Department of Pathology, The University of Chicago, Chicago, IL
| | | | - Louis H Philipson
- Department of Medicine, The University of Chicago, Chicago, IL
- Department of Pediatrics, The University of Chicago, Chicago, IL
| | | | - Manami Hara
- Department of Medicine, The University of Chicago, Chicago, IL
| |
Collapse
|
27
|
Chela H, Romana BS, Madabattula M, Albarrak AA, Yousef MH, Samiullah S, Tahan V. Stem cell therapy: a potential for the perils of pancreatitis. THE TURKISH JOURNAL OF GASTROENTEROLOGY : THE OFFICIAL JOURNAL OF TURKISH SOCIETY OF GASTROENTEROLOGY 2020; 31:415-424. [PMID: 32721912 PMCID: PMC7433995 DOI: 10.5152/tjg.2020.19143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 09/01/2019] [Indexed: 12/17/2022]
Abstract
Acute and chronic pancreatitis carry a significant disease burden and there is no definite treatment that exists for either. They are associated with local and systemic inflammation and lead to numerous complications. Stem cell therapy has been explored for other disease processes and is a topic of research that has gained momentum with regards to implications for acute and chronic pancreatitis. They not only carry the potential to aid in regeneration but also prevent pancreatic injury as well as injury of other organs and hence the resultant complications. Stem cells appear to have immunomodulatory properties and clinical potential as evidenced by numerous studies in animal models. This review article discusses the types of stem cells commonly used and the properties that show promise in the field of pancreatitis.
Collapse
Affiliation(s)
- Harleen Chela
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri, USA
| | - Bhupinder S Romana
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri, USA
| | - Markandeya Madabattula
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri, USA
| | - Abdulmajeed A Albarrak
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri, USA
| | - Mohamad H Yousef
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri, USA
| | - Sami Samiullah
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri, USA
| | - Veysel Tahan
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
28
|
Ledard N, Liboz A, Blondeau B, Babiak M, Moulin C, Vallin B, Guillas I, Mateo V, Jumeau C, Blirando K, Meilhac O, Limon I, Glorian M. Slug, a Cancer-Related Transcription Factor, is Involved in Vascular Smooth Muscle Cell Transdifferentiation Induced by Platelet-Derived Growth Factor-BB During Atherosclerosis. J Am Heart Assoc 2020; 9:e014276. [PMID: 31959031 PMCID: PMC7033846 DOI: 10.1161/jaha.119.014276] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background Heart attacks and stroke often result from occlusive thrombi following the rupture of vulnerable atherosclerotic plaques. Vascular smooth muscle cells (VSMCs) play a pivotal role in plaque vulnerability because of their switch towards a proinflammatory/macrophage-like phenotype when in the context of atherosclerosis. The prometastatic transcription factor Slug/Snail2 is a critical regulator of cell phenotypic transition. Here, we aimed to investigate the role of Slug in the transdifferentiation process of VSMCs occurring during atherogenesis. Methods and Results In rat and human primary aortic smooth muscle cells, Slug protein expression is strongly and rapidly increased by platelet-derived growth factor-BB (PDGF-BB). PDGF-BB increases Slug protein without affecting mRNA levels indicating that this growth factor stabilizes Slug protein. Immunocytochemistry and subcellular fractionation experiments reveal that PDGF-BB triggers a rapid accumulation of Slug in VSMC nuclei. Using pharmacological tools, we show that the PDGF-BB-dependent mechanism of Slug stabilization in VSMCs involves the extracellular signal-regulated kinase 1/2 pathway. Immunohistochemistry experiments on type V and type VI atherosclerotic lesions of human carotids show smooth muscle-specific myosin heavy chain-/Slug-positive cells surrounding the prothrombotic lipid core. In VSMCs, Slug siRNAs inhibit prostaglandin E2 secretion and prevent the inhibition of cholesterol efflux gene expression mediated by PDGF-BB, known to be involved in plaque vulnerability and/or thrombogenicity. Conclusions Our results highlight, for the first time, a role of Slug in aortic smooth muscle cell transdifferentiation and enable us to consider Slug as an actor playing a role in the atherosclerotic plaque progression towards a life-threatening phenotype. This also argues for common features between acute cardiovascular events and cancer.
Collapse
Affiliation(s)
- Nahéma Ledard
- Institut de Biologie Paris-Seine (IBPS) Biological Adaptation and Ageing UMR 8256 Sorbonne Université Paris France
| | - Alexandrine Liboz
- INSERM Saint-Antoine Research Center Sorbonne Université Paris France
| | - Bertrand Blondeau
- INSERM Saint-Antoine Research Center Sorbonne Université Paris France
| | - Mégane Babiak
- Institut de Biologie Paris-Seine (IBPS) Biological Adaptation and Ageing UMR 8256 Sorbonne Université Paris France
| | - Célia Moulin
- Institut de Biologie Paris-Seine (IBPS) Biological Adaptation and Ageing UMR 8256 Sorbonne Université Paris France
| | - Benjamin Vallin
- Institut de Biologie Paris-Seine (IBPS) Biological Adaptation and Ageing UMR 8256 Sorbonne Université Paris France
| | - Isabelle Guillas
- National Institute for Health and Medical Research (INSERM) Faculté de Médecine Pitié Salpétrière UMR-S 1166 ICAN Sorbonne Université Paris France
| | - Véronique Mateo
- CIMI-Paris INSERM U1135 Faculté de Médecine Sorbonne-Université Site Pitié-Salpêtrière Sorbonne Université Paris France
| | | | - Karl Blirando
- Institut de Biologie Paris-Seine (IBPS) Biological Adaptation and Ageing UMR 8256 Sorbonne Université Paris France
| | - Olivier Meilhac
- Université de La Réunion Diabète, Athérothrombose, Thérapies, Réunion, Océan Indien (UMR DéTROI U1188) - -CYROI- Sainte Clotilde La Réunion
| | - Isabelle Limon
- Institut de Biologie Paris-Seine (IBPS) Biological Adaptation and Ageing UMR 8256 Sorbonne Université Paris France
| | - Martine Glorian
- Institut de Biologie Paris-Seine (IBPS) Biological Adaptation and Ageing UMR 8256 Sorbonne Université Paris France
| |
Collapse
|
29
|
Rieger L, O’Connor R. Controlled Signaling-Insulin-Like Growth Factor Receptor Endocytosis and Presence at Intracellular Compartments. Front Endocrinol (Lausanne) 2020; 11:620013. [PMID: 33584548 PMCID: PMC7878670 DOI: 10.3389/fendo.2020.620013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/02/2020] [Indexed: 12/16/2022] Open
Abstract
Ligand-induced activation of the IGF-1 receptor triggers plasma-membrane-derived signal transduction but also triggers receptor endocytosis, which was previously thought to limit signaling. However, it is becoming ever more clear that IGF-1R endocytosis and trafficking to specific subcellular locations can define specific signaling responses that are important for key biological processes in normal cells and cancer cells. In different cell types, specific cell adhesion receptors and associated proteins can regulate IGF-1R endocytosis and trafficking. Once internalized, the IGF-1R may be recycled, degraded or translocated to the intracellular membrane compartments of the Golgi apparatus or the nucleus. The IGF-1R is present in the Golgi apparatus of migratory cancer cells where its signaling contributes to aggressive cancer behaviors including cell migration. The IGF-1R is also found in the nucleus of certain cancer cells where it can regulate gene expression. Nuclear IGF-1R is associated with poor clinical outcomes. IGF-1R signaling has also been shown to support mitochondrial biogenesis and function, and IGF-1R inhibition causes mitochondrial dysfunction. How IGF-1R intracellular trafficking and compartmentalized signaling is controlled is still unknown. This is an important area for further study, particularly in cancer.
Collapse
|
30
|
EMT and Stemness-Key Players in Pancreatic Cancer Stem Cells. Cancers (Basel) 2019; 11:cancers11081136. [PMID: 31398893 PMCID: PMC6721598 DOI: 10.3390/cancers11081136] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 08/01/2019] [Accepted: 08/06/2019] [Indexed: 12/15/2022] Open
Abstract
Metastasis and tumor progression are the major cause of death in patients suffering from pancreatic ductal adenocarcinoma. Tumor growth and especially dissemination are typically associated with activation of an epithelial-to-mesenchymal transition (EMT) program. This phenotypic transition from an epithelial to a mesenchymal state promotes migration and survival both during development and in cancer progression. When re-activated in pathological contexts such as cancer, this type of developmental process confers additional stemness properties to specific subsets of cells. Cancer stem cells (CSCs) are a subpopulation of cancer cells with stem-like features that are responsible for the propagation of the tumor as well as therapy resistance and cancer relapse, but also for circulating tumor cell release and metastasis. In support of this concept, EMT transcription factors generate cells with stem cell properties and mediate chemoresistance. However, their role in pancreatic ductal adenocarcinoma metastasis remains controversial. As such, a better characterization of CSC populations will be crucial in future development of therapies targeting these cells. In this review, we will discuss the latest updates on the mechanisms common to pancreas development and CSC-mediated tumor progression.
Collapse
|
31
|
Braitsch CM, Azizoglu DB, Htike Y, Barlow HR, Schnell U, Chaney CP, Carroll TJ, Stanger BZ, Cleaver O. LATS1/2 suppress NFκB and aberrant EMT initiation to permit pancreatic progenitor differentiation. PLoS Biol 2019; 17:e3000382. [PMID: 31323030 PMCID: PMC6668837 DOI: 10.1371/journal.pbio.3000382] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 07/31/2019] [Accepted: 07/02/2019] [Indexed: 12/25/2022] Open
Abstract
The Hippo pathway directs cell differentiation during organogenesis, in part by restricting proliferation. How Hippo signaling maintains a proliferation-differentiation balance in developing tissues via distinct molecular targets is only beginning to be understood. Our study makes the unexpected finding that Hippo suppresses nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) signaling in pancreatic progenitors to permit cell differentiation and epithelial morphogenesis. We find that pancreas-specific deletion of the large tumor suppressor kinases 1 and 2 (Lats1/2PanKO) from mouse progenitor epithelia results in failure to differentiate key pancreatic lineages: acinar, ductal, and endocrine. We carried out an unbiased transcriptome analysis to query differentiation defects in Lats1/2PanKO. This analysis revealed increased expression of NFκB activators, including the pantetheinase vanin1 (Vnn1). Using in vivo and ex vivo studies, we show that VNN1 activates a detrimental cascade of processes in Lats1/2PanKO epithelium, including (1) NFκB activation and (2) aberrant initiation of epithelial-mesenchymal transition (EMT), which together disrupt normal differentiation. We show that exogenous stimulation of VNN1 or NFκB can trigger this cascade in wild-type (WT) pancreatic progenitors. These findings reveal an unexpected requirement for active suppression of NFκB by LATS1/2 during pancreas development, which restrains a cell-autonomous deleterious transcriptional program and thereby allows epithelial differentiation.
Collapse
Affiliation(s)
- Caitlin M. Braitsch
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - D. Berfin Azizoglu
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Yadanar Htike
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Haley R. Barlow
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Ulrike Schnell
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Christopher P. Chaney
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Thomas J. Carroll
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Ben Z. Stanger
- Department of Medicine and Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ondine Cleaver
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| |
Collapse
|
32
|
Yu XX, Qiu WL, Yang L, Zhang Y, He MY, Li LC, Xu CR. Defining multistep cell fate decision pathways during pancreatic development at single-cell resolution. EMBO J 2019; 38:e100164. [PMID: 30737258 PMCID: PMC6463266 DOI: 10.15252/embj.2018100164] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 12/27/2018] [Accepted: 01/07/2019] [Indexed: 12/13/2022] Open
Abstract
The generation of terminally differentiated cell lineages during organogenesis requires multiple, coordinated cell fate choice steps. However, this process has not been clearly delineated, especially in complex solid organs such as the pancreas. Here, we performed single-cell RNA-sequencing in pancreatic cells sorted from multiple genetically modified reporter mouse strains at embryonic stages E9.5-E17.5. We deciphered the developmental trajectories and regulatory strategies of the exocrine and endocrine pancreatic lineages as well as intermediate progenitor populations along the developmental pathways. Notably, we discovered previously undefined programs representing the earliest events in islet α- and β-cell lineage allocation as well as the developmental pathway of the "first wave" of α-cell generation. Furthermore, we demonstrated that repressing ERK pathway activity is essential for inducing both α- and β-lineage differentiation. This study provides key insights into the regulatory mechanisms underlying cell fate choice and stepwise cell fate commitment and can be used as a resource to guide the induction of functional islet lineage cells from stem cells in vitro.
Collapse
Affiliation(s)
- Xin-Xin Yu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Wei-Lin Qiu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
- PKU-Tsinghua-NIBS Graduate Program, Peking University, Beijing, China
| | - Liu Yang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Yu Zhang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Mao-Yang He
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
- PKU-Tsinghua-NIBS Graduate Program, Peking University, Beijing, China
| | - Lin-Chen Li
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Cheng-Ran Xu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| |
Collapse
|
33
|
Nyeng P, Heilmann S, Löf-Öhlin ZM, Pettersson NF, Hermann FM, Reynolds AB, Semb H. p120ctn-Mediated Organ Patterning Precedes and Determines Pancreatic Progenitor Fate. Dev Cell 2019; 49:31-47.e9. [PMID: 30853440 DOI: 10.1016/j.devcel.2019.02.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 12/13/2018] [Accepted: 02/04/2019] [Indexed: 11/28/2022]
Abstract
The mechanism of how organ shape emerges and specifies cell fate is not understood. Pancreatic duct and endocrine lineages arise in a spatially distinct domain from the acinar lineage. Whether these lineages are pre-determined or settle once these niches have been established remains unknown. Here, we reconcile these two apparently opposing models, demonstrating that pancreatic progenitors re-localize to establish the niche that will determine their ultimate fate. We identify a p120ctn-regulated mechanism for coordination of organ architecture and cellular fate mediated by differential E-cadherin based cell sorting. Reduced p120ctn expression is necessary and sufficient to re-localize a subset of progenitors to the peripheral tip domain, where they acquire an acinar fate. The same mechanism is used re-iteratively during endocrine specification, where it balances the choice between the alpha and beta cell fates. In conclusion, organ patterning is regulated by p120ctn-mediated cellular positioning, which precedes and determines pancreatic progenitor fate.
Collapse
Affiliation(s)
- Pia Nyeng
- Novo Nordisk Foundation Center for Stem Cell Biology (Danstem), University of Copenhagen, 2200 Copenhagen N, Denmark.
| | - Silja Heilmann
- Novo Nordisk Foundation Center for Stem Cell Biology (Danstem), University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Zarah M Löf-Öhlin
- Novo Nordisk Foundation Center for Stem Cell Biology (Danstem), University of Copenhagen, 2200 Copenhagen N, Denmark
| | | | - Florian Malte Hermann
- Novo Nordisk Foundation Center for Stem Cell Biology (Danstem), University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Albert B Reynolds
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Henrik Semb
- Novo Nordisk Foundation Center for Stem Cell Biology (Danstem), University of Copenhagen, 2200 Copenhagen N, Denmark; Institute of Translational Stem Cell Research, Helmholtz Zentrum München, Neuherberg, Germany.
| |
Collapse
|
34
|
Abstract
Diabetes mellitus is a multifactorial disease affecting increasing numbers of patients worldwide. Progression to insulin-dependent diabetes mellitus is characterized by the loss or dysfunction of pancreatic β-cells, but the pathomechanisms underlying β-cell failure in type 1 diabetes mellitus and type 2 diabetes mellitus are still poorly defined. Regeneration of β-cell mass from residual islet cells or replacement by β-like cells derived from stem cells holds great promise to stop or reverse disease progression. However, the development of new treatment options is hampered by our limited understanding of human pancreas organogenesis due to the restricted access to primary tissues. Therefore, the challenge is to translate results obtained from preclinical model systems to humans, which requires comparative modelling of β-cell biology in health and disease. Here, we discuss diverse modelling systems across different species that provide spatial and temporal resolution of cellular and molecular mechanisms to understand the evolutionary conserved genotype-phenotype relationship and translate them to humans. In addition, we summarize the latest knowledge on organoids, stem cell differentiation platforms, primary micro-islets and pseudo-islets, bioengineering and microfluidic systems for studying human pancreas development and homeostasis ex vivo. These new modelling systems and platforms have opened novel avenues for exploring the developmental trajectory, physiology, biology and pathology of the human pancreas.
Collapse
Affiliation(s)
- Mostafa Bakhti
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany.
- Institute of Stem Cell Research, Helmholtz Zentrum München, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Anika Böttcher
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany.
- Institute of Stem Cell Research, Helmholtz Zentrum München, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany.
- Institute of Stem Cell Research, Helmholtz Zentrum München, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Technical University of Munich, Medical Faculty, Munich, Germany.
| |
Collapse
|
35
|
Sharon N, Chawla R, Mueller J, Vanderhooft J, Whitehorn LJ, Rosenthal B, Gürtler M, Estanboulieh RR, Shvartsman D, Gifford DK, Trapnell C, Melton D. A Peninsular Structure Coordinates Asynchronous Differentiation with Morphogenesis to Generate Pancreatic Islets. Cell 2019; 176:790-804.e13. [PMID: 30661759 PMCID: PMC6705176 DOI: 10.1016/j.cell.2018.12.003] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 08/20/2018] [Accepted: 12/03/2018] [Indexed: 12/15/2022]
Abstract
The pancreatic islets of Langerhans regulate glucose homeostasis. The loss of insulin-producing β cells within islets results in diabetes, and islet transplantation from cadaveric donors can cure the disease. In vitro production of whole islets, not just β cells, will benefit from a better understanding of endocrine differentiation and islet morphogenesis. We used single-cell mRNA sequencing to obtain a detailed description of pancreatic islet development. Contrary to the prevailing dogma, we find islet morphology and endocrine differentiation to be directly related. As endocrine progenitors differentiate, they migrate in cohesion and form bud-like islet precursors, or "peninsulas" (literally "almost islands"). α cells, the first to develop, constitute the peninsular outer layer, and β cells form later, beneath them. This spatiotemporal collinearity leads to the typical core-mantle architecture of the mature, spherical islet. Finally, we induce peninsula-like structures in differentiating human embryonic stem cells, laying the ground for the generation of entire islets in vitro.
Collapse
Affiliation(s)
- Nadav Sharon
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Raghav Chawla
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA; Division of Hematology/Oncology, Seattle Children's Hospital, Seattle, WA 98105, USA; Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Jonas Mueller
- Computer Science and Artificial Intelligence Laboratory, MIT, Cambridge, MA 02412, USA
| | - Jordan Vanderhooft
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | | | - Benjamin Rosenthal
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Mads Gürtler
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | | | - Dmitry Shvartsman
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - David K Gifford
- Computer Science and Artificial Intelligence Laboratory, MIT, Cambridge, MA 02412, USA
| | - Cole Trapnell
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA; Molecular & Cellular Biology Program, University of Washington, Seattle, WA 98195, USA.
| | - Doug Melton
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
36
|
Kowalska M, Rupik W. Development of endocrine pancreatic islets in embryos of the grass snake Natrix natrix
(Lepidosauria, Serpentes). J Morphol 2018; 280:103-118. [DOI: 10.1002/jmor.20921] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 09/04/2018] [Accepted: 10/29/2018] [Indexed: 12/29/2022]
Affiliation(s)
- Magdalena Kowalska
- Department of Animal Histology and Embryology; University of Silesia in Katowice; Poland
| | - Weronika Rupik
- Department of Animal Histology and Embryology; University of Silesia in Katowice; Poland
| |
Collapse
|
37
|
Mesodermal induction of pancreatic fate commitment. Semin Cell Dev Biol 2018; 92:77-88. [PMID: 30142440 DOI: 10.1016/j.semcdb.2018.08.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 07/29/2018] [Accepted: 08/20/2018] [Indexed: 12/27/2022]
Abstract
The pancreas is a compound gland comprised of both exocrine acinar and duct cells as well as endocrine islet cells. Most notable amongst the latter are the insulin-synthesizing β-cells, loss or dysfunction of which manifests in diabetes mellitus. All exocrine and endocrine cells derive from multipotent pancreatic progenitor cells arising from the primitive gut epithelium via inductive interactions with adjacent mesodermal tissues. Research in the last two decades has revealed the identity of many of these extrinsic cues and they include signaling molecules used in many other developmental contexts such as retinoic acid, fibroblast growth factors, and members of the TGF-β superfamily. As important as these inductive cues is the absence of other signaling molecules such as hedgehog family members. Much has been learned about the interactions of extrinsic factors with fate regulators intrinsic to the pancreatic endoderm. This new knowledge has had tremendous impact on the development of directed differentiation protocols for converting pluripotent stem cells to β-cells in vitro.
Collapse
|
38
|
Functional Role of Non-Coding RNAs during Epithelial-To-Mesenchymal Transition. Noncoding RNA 2018; 4:ncrna4020014. [PMID: 29843425 PMCID: PMC6027143 DOI: 10.3390/ncrna4020014] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/22/2018] [Accepted: 05/23/2018] [Indexed: 01/17/2023] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a key biological process involved in a multitude of developmental and pathological events. It is characterized by the progressive loss of cell-to-cell contacts and actin cytoskeletal rearrangements, leading to filopodia formation and the progressive up-regulation of a mesenchymal gene expression pattern enabling cell migration. Epithelial-to-mesenchymal transition is already observed in early embryonic stages such as gastrulation, when the epiblast undergoes an EMT process and therefore leads to the formation of the third embryonic layer, the mesoderm. Epithelial-to-mesenchymal transition is pivotal in multiple embryonic processes, such as for example during cardiovascular system development, as valve primordia are formed and the cardiac jelly is progressively invaded by endocardium-derived mesenchyme or as the external cardiac cell layer is established, i.e., the epicardium and cells detached migrate into the embryonic myocardial to form the cardiac fibrous skeleton and the coronary vasculature. Strikingly, the most important biological event in which EMT is pivotal is cancer development and metastasis. Over the last years, understanding of the transcriptional regulatory networks involved in EMT has greatly advanced. Several transcriptional factors such as Snail, Slug, Twist, Zeb1 and Zeb2 have been reported to play fundamental roles in EMT, leading in most cases to transcriptional repression of cell⁻cell interacting proteins such as ZO-1 and cadherins and activation of cytoskeletal markers such as vimentin. In recent years, a fundamental role for non-coding RNAs, particularly microRNAs and more recently long non-coding RNAs, has been identified in normal tissue development and homeostasis as well as in several oncogenic processes. In this study, we will provide a state-of-the-art review of the functional roles of non-coding RNAs, particularly microRNAs, in epithelial-to-mesenchymal transition in both developmental and pathological EMT.
Collapse
|
39
|
Krivova YS, Proshchina AE, Barabanov VM, Barinova IV, Saveliev SV. Immunohistochemical detection of vimentin in pancreatic islet β- and α-cells of macrosomic infants of diabetic and nondiabetic mothers. Early Hum Dev 2018; 117:44-49. [PMID: 29275072 DOI: 10.1016/j.earlhumdev.2017.12.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 12/08/2017] [Accepted: 12/10/2017] [Indexed: 11/18/2022]
Abstract
BACKGROUND Expression of the intermediate filament protein vimentin has been recently observed in the pancreatic islet β- and α-cells of humans with type 2 diabetes mellitus. It was suggested that the presence of vimentin in endocrine cells may indicate islet tissue renewal, or potentially represent the dedifferentiation of endocrine cells, which could contribute to the onset of type 2 diabetes or islet cell dysfunction. AIM To analyze the expression of vimentin in pancreatic β- and α-cells of macrosomic infants of diabetic and nondiabetic mothers. SUBJECTS Pancreatic samples of five macrosomic infants (gestational age 34-40weeks) from three diabetic and two nondiabetic mothers were compared to six control infants (32-40weeks, weight appropriate for gestational age) from normoglycemic mothers. METHODS Pancreatic autopsy samples were examined by double immunofluorescent labeling with antibodies against vimentin and either insulin or glucagon. Alterations in the endocrine pancreas were measured using morphometric methods, then data were statistically analyzed. RESULTS In the pancreatic islets of macrosomic infants from diabetic and nondiabetic mothers, we observed vimentin-positive cells, some of which simultaneously contained insulin or glucagon. We also quantitatively showed that the presence of such cells was associated with hypertrophy and hyperplasia of the islets, and with an increase in β- and α-cell density. CONCLUSIONS We speculate that the appearance of vimentin-positive islet cells may reflect induction of differentiation in response to the increased insulin demand, and vimentin may serve as an early marker of endocrine pancreas disorders.
Collapse
Affiliation(s)
- Yuliya S Krivova
- Laboratory of Nervous System Development, Research Institute of Human Morphology, 3 Tsurupy St., Moscow 117418, Russia.
| | - Alexandra E Proshchina
- Laboratory of Nervous System Development, Research Institute of Human Morphology, 3 Tsurupy St., Moscow 117418, Russia
| | - Valeriy M Barabanov
- Laboratory of Nervous System Development, Research Institute of Human Morphology, 3 Tsurupy St., Moscow 117418, Russia
| | - Irina V Barinova
- Moscow Regional Research Institute of Obstetrics and Gynecology, Ministry of Health of the Moscow Region, 22a Pokrovka St., Moscow 101000, Russia
| | - Sergey V Saveliev
- Laboratory of Nervous System Development, Research Institute of Human Morphology, 3 Tsurupy St., Moscow 117418, Russia
| |
Collapse
|
40
|
Warsito D, Lin Y, Gnirck AC, Sehat B, Larsson O. Nuclearly translocated insulin-like growth factor 1 receptor phosphorylates histone H3 at tyrosine 41 and induces SNAI2 expression via Brg1 chromatin remodeling protein. Oncotarget 2018; 7:42288-42302. [PMID: 27275536 PMCID: PMC5173135 DOI: 10.18632/oncotarget.9785] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 04/26/2016] [Indexed: 01/14/2023] Open
Abstract
The insulin-like growth factor-1 receptor (IGF-1R) is a receptor tyrosine kinase that has crucial roles in cell proliferation and protection from apoptosis. It is therefore not surprising that IGF-1R is often found overexpressed in many types of tumors. This has made IGF-1R a prominent target molecule for pharmacological companies to develop new anti-cancer agents. However, several clinical trials during the last 5 years using IGF-1R specific antibodies have shown disappointing results. We have previously shown that upon IGF-1 stimulation, the receptor becomes SUMOylated and translocates into the nucleus of cancer cells to act as a transcription co-factor. Soon after our original study, several others have reported nuclear IGF-1R (nIGF-1R) as well, and some of them have demonstrated a prognostic value of nIGF-1R expression in cancer. In the current study we demonstrate that nIGF-1R binds to and phosphorylates histone H3 at tyrosine 41 (H3Y41) in HeLa cells. Furthermore, our results suggest that phosphorylation of H3Y41 by nIGF-1R, stabilizes the binding of Brg1 chromatin remodeling protein to Histone H3. Our findings suggest that phosphorylated nIGF-1R, rather than total nIGF-1R, plays a superior role in these contexts. We identified SNAI2 oncogene as a target gene for nIGF-1R and its expression was decreased upon mutation of H3Y41 or by Brg1 knockdown. Furthermore, we demonstrate that both IGF-1R and Brg1 binds to the SNAI2 promoter. As SNAI2 protein is implicated in e.g. cancer invasion and metastasis, the nIGF-1R-mediated effects shown in this study may influence such important tumor phenotypic actions.
Collapse
Affiliation(s)
- Dudi Warsito
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | - Yingbo Lin
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | - Ann-Christin Gnirck
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | - Bita Sehat
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | - Olle Larsson
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| |
Collapse
|
41
|
Petersen MB, Gonçalves CA, Kim YH, Grapin-Botton A. Recapitulating and Deciphering Human Pancreas Development From Human Pluripotent Stem Cells in a Dish. Curr Top Dev Biol 2018; 129:143-190. [DOI: 10.1016/bs.ctdb.2018.02.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
42
|
Bastidas-Ponce A, Scheibner K, Lickert H, Bakhti M. Cellular and molecular mechanisms coordinating pancreas development. Development 2017; 144:2873-2888. [PMID: 28811309 DOI: 10.1242/dev.140756] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The pancreas is an endoderm-derived glandular organ that participates in the regulation of systemic glucose metabolism and food digestion through the function of its endocrine and exocrine compartments, respectively. While intensive research has explored the signaling pathways and transcriptional programs that govern pancreas development, much remains to be discovered regarding the cellular processes that orchestrate pancreas morphogenesis. Here, we discuss the developmental mechanisms and principles that are known to underlie pancreas development, from induction and lineage formation to morphogenesis and organogenesis. Elucidating such principles will help to identify novel candidate disease genes and unravel the pathogenesis of pancreas-related diseases, such as diabetes, pancreatitis and cancer.
Collapse
Affiliation(s)
- Aimée Bastidas-Ponce
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany.,Technical University of Munich, Medical Faculty, 81675 Munich, Germany
| | - Katharina Scheibner
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany.,Technical University of Munich, Medical Faculty, 81675 Munich, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany.,Technical University of Munich, Medical Faculty, 81675 Munich, Germany
| | - Mostafa Bakhti
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany .,Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany
| |
Collapse
|
43
|
Scoville DW, Kang HS, Jetten AM. GLIS1-3: emerging roles in reprogramming, stem and progenitor cell differentiation and maintenance. Stem Cell Investig 2017; 4:80. [PMID: 29057252 DOI: 10.21037/sci.2017.09.01] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 08/27/2017] [Indexed: 12/12/2022]
Abstract
Recent studies have provided evidence for a regulatory role of GLI-similar (GLIS) transcription factors in reprogramming, maintenance and differentiation of several stem and progenitor cell populations. GLIS1, in conjunction with several other reprogramming factors, was shown to markedly increase the efficiency of generating induced pluripotent stem cells (iPSC) from somatic cells. GLIS2 has been reported to contribute to the maintenance of the pluripotent state in hPSCs. In addition, GLIS2 has a function in regulating self-renewal of hematopoietic progenitors and megakaryocytic differentiation. GLIS3 plays a critical role during the development of several tissues. GLIS3 is able to promote reprogramming of human fibroblasts into retinal pigmented epithelial (RPE) cells. Moreover, GLIS3 is essential for spermatogonial stem cell renewal and spermatogonial progenitor cell differentiation. During pancreas development, GLIS3 protein is first detectable in bipotent pancreatic progenitors and pro-endocrine progenitors and plays a critical role in the generation of pancreatic beta cells. Here, we review the current status of the roles of GLIS proteins in the maintenance and differentiation of these different stem and progenitor cells.
Collapse
Affiliation(s)
- David W Scoville
- Cell Biology Section, Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Hong Soon Kang
- Cell Biology Section, Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Anton M Jetten
- Cell Biology Section, Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| |
Collapse
|
44
|
Pauerstein PT, Tellez K, Willmarth KB, Park KM, Hsueh B, Efsun Arda H, Gu X, Aghajanian H, Deisseroth K, Epstein JA, Kim SK. A radial axis defined by semaphorin-to-neuropilin signaling controls pancreatic islet morphogenesis. Development 2017; 144:3744-3754. [PMID: 28893946 DOI: 10.1242/dev.148684] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 09/04/2017] [Indexed: 12/24/2022]
Abstract
The islets of Langerhans are endocrine organs characteristically dispersed throughout the pancreas. During development, endocrine progenitors delaminate, migrate radially and cluster to form islets. Despite the distinctive distribution of islets, spatially localized signals that control islet morphogenesis have not been discovered. Here, we identify a radial signaling axis that instructs developing islet cells to disperse throughout the pancreas. A screen of pancreatic extracellular signals identified factors that stimulated islet cell development. These included semaphorin 3a, a guidance cue in neural development without known functions in the pancreas. In the fetal pancreas, peripheral mesenchymal cells expressed Sema3a, while central nascent islet cells produced the semaphorin receptor neuropilin 2 (Nrp2). Nrp2 mutant islet cells developed in proper numbers, but had defects in migration and were unresponsive to purified Sema3a. Mutant Nrp2 islets aggregated centrally and failed to disperse radially. Thus, Sema3a-Nrp2 signaling along an unrecognized pancreatic developmental axis constitutes a chemoattractant system essential for generating the hallmark morphogenetic properties of pancreatic islets. Unexpectedly, Sema3a- and Nrp2-mediated control of islet morphogenesis is strikingly homologous to mechanisms that regulate radial neuronal migration and cortical lamination in the developing mammalian brain.
Collapse
Affiliation(s)
- Philip T Pauerstein
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Krissie Tellez
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kirk B Willmarth
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Keon Min Park
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Brian Hsueh
- Departments of Bioengineering and of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - H Efsun Arda
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xueying Gu
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Haig Aghajanian
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Karl Deisseroth
- Departments of Bioengineering and of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.,Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jonathan A Epstein
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Seung K Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
45
|
Modeling coexistence of oscillation and Delta/Notch-mediated lateral inhibition in pancreas development and neurogenesis. J Theor Biol 2017; 430:32-44. [PMID: 28652000 DOI: 10.1016/j.jtbi.2017.06.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 06/02/2017] [Accepted: 06/07/2017] [Indexed: 11/23/2022]
Abstract
During pancreas development, Neurog3 positive endocrine progenitors are specified by Delta/Notch (D/N) mediated lateral inhibition in the growing ducts. During neurogenesis, genes that determine the transition from the proneural state to neuronal or glial lineages are oscillating before their expression is sustained. Although the basic gene regulatory network is very similar, cycling gene expression in pancreatic development was not investigated yet, and previous simulations of lateral inhibition in pancreas development excluded by design the possibility of oscillations. To explore this possibility, we developed a dynamic model of a growing duct that results in an oscillatory phase before the determination of endocrine progenitors by lateral inhibition. The basic network (D/N + Hes1 + Neurog3) shows scattered, stable Neurog3 expression after displaying transient expression. Furthermore, we included the Hes1 negative feedback as previously discussed in neurogenesis and show the consequences for Neurog3 expression in pancreatic duct development. Interestingly, a weakened HES1 action on the Hes1 promoter allows the coexistence of stable patterning and oscillations. In conclusion, cycling gene expression and lateral inhibition are not mutually exclusive. In this way, we argue for a unified mode of D/N mediated lateral inhibition in neurogenic and pancreatic progenitor specification.
Collapse
|
46
|
Sinagoga KL, Stone WJ, Schiesser JV, Schweitzer JI, Sampson L, Zheng Y, Wells JM. Distinct roles for the mTOR pathway in postnatal morphogenesis, maturation and function of pancreatic islets. Development 2017; 144:2402-2414. [PMID: 28576773 DOI: 10.1242/dev.146316] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 05/26/2017] [Indexed: 02/03/2023]
Abstract
While much is known about the molecular pathways that regulate embryonic development and adult homeostasis of the endocrine pancreas, little is known about what regulates early postnatal development and maturation of islets. Given that birth marks the first exposure to enteral nutrition, we investigated how nutrient-regulated signaling pathways influence postnatal islet development in mice. We performed loss-of-function studies of mechanistic target of rapamycin (mTOR), a highly conserved kinase within a nutrient-sensing pathway known to regulate cellular growth, morphogenesis and metabolism. Deletion of Mtor in pancreatic endocrine cells had no significant effect on their embryonic development. However, within the first 2 weeks after birth, mTOR-deficient islets became dysmorphic, β-cell maturation and function were impaired, and animals lost islet mass. Moreover, we discovered that these distinct functions of mTOR are mediated by separate downstream branches of the pathway, in that mTORC1 (with adaptor protein Raptor) is the main complex mediating the maturation and function of islets, whereas mTORC2 (with adaptor protein Rictor) impacts islet mass and architecture. Taken together, these findings suggest that nutrient sensing may be an essential trigger for postnatal β-cell maturation and islet development.
Collapse
Affiliation(s)
- Katie L Sinagoga
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229-3039, USA
| | - William J Stone
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229-3039, USA
| | - Jacqueline V Schiesser
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229-3039, USA
| | - Jamie I Schweitzer
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229-3039, USA
| | - Leesa Sampson
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229-3039, USA
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229-3039, USA
| | - James M Wells
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229-3039, USA .,Division of Endocrinology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229-3039, USA
| |
Collapse
|
47
|
Azzarelli R, Hurley C, Sznurkowska MK, Rulands S, Hardwick L, Gamper I, Ali F, McCracken L, Hindley C, McDuff F, Nestorowa S, Kemp R, Jones K, Göttgens B, Huch M, Evan G, Simons BD, Winton D, Philpott A. Multi-site Neurogenin3 Phosphorylation Controls Pancreatic Endocrine Differentiation. Dev Cell 2017; 41:274-286.e5. [PMID: 28457793 PMCID: PMC5425251 DOI: 10.1016/j.devcel.2017.04.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 03/10/2017] [Accepted: 04/04/2017] [Indexed: 12/02/2022]
Abstract
The proneural transcription factor Neurogenin3 (Ngn3) plays a critical role in pancreatic endocrine cell differentiation, although regulation of Ngn3 protein is largely unexplored. Here we demonstrate that Ngn3 protein undergoes cyclin-dependent kinase (Cdk)-mediated phosphorylation on multiple serine-proline sites. Replacing wild-type protein with a phosphomutant form of Ngn3 increases α cell generation, the earliest endocrine cell type to be formed in the developing pancreas. Moreover, un(der)phosphorylated Ngn3 maintains insulin expression in adult β cells in the presence of elevated c-Myc and enhances endocrine specification during ductal reprogramming. Mechanistically, preventing multi-site phosphorylation enhances both Ngn3 stability and DNA binding, promoting the increased expression of target genes that drive differentiation. Therefore, multi-site phosphorylation of Ngn3 controls its ability to promote pancreatic endocrine differentiation and to maintain β cell function in the presence of pro-proliferation cues and could be manipulated to promote and maintain endocrine differentiation in vitro and in vivo.
Collapse
Affiliation(s)
- Roberta Azzarelli
- Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, UK; Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Christopher Hurley
- Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, UK; Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Magdalena K Sznurkowska
- Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, UK; Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Steffen Rulands
- Max Planck Institute for the Physics of Complex Systems, Nöthnitzer Straße 38, 01187 Dresden, Germany; Center for Systems Biology Dresden, Pfotenhauer Straße 108, 01307 Dresden, Germany
| | - Laura Hardwick
- Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, UK; Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Ivonne Gamper
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, UK
| | - Fahad Ali
- Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, UK; Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Laura McCracken
- Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, UK; Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Christopher Hindley
- The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK; Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge CB3 0HE, UK
| | - Fiona McDuff
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, UK
| | - Sonia Nestorowa
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK; Department of Haematology, Cambridge Institute for Medical Research, Hills Road, Cambridge CB2 0XY, UK
| | - Richard Kemp
- Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Kenneth Jones
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Berthold Göttgens
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK; Department of Haematology, Cambridge Institute for Medical Research, Hills Road, Cambridge CB2 0XY, UK
| | - Meritxell Huch
- The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK
| | - Gerard Evan
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, UK
| | - Benjamin D Simons
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK; The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK; Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge CB3 0HE, UK
| | - Douglas Winton
- Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Anna Philpott
- Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, UK; Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK.
| |
Collapse
|
48
|
Larsen HL, Grapin-Botton A. The molecular and morphogenetic basis of pancreas organogenesis. Semin Cell Dev Biol 2017; 66:51-68. [PMID: 28089869 DOI: 10.1016/j.semcdb.2017.01.005] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 01/06/2017] [Accepted: 01/09/2017] [Indexed: 01/08/2023]
Abstract
The pancreas is an essential endoderm-derived organ that ensures nutrient metabolism via its endocrine and exocrine functions. Here we review the essential processes governing the embryonic and early postnatal development of the pancreas discussing both the mechanisms and molecules controlling progenitor specification, expansion and differentiation. We elaborate on how these processes are orchestrated in space and coordinated with morphogenesis. We draw mainly from experiments conducted in the mouse model but also from investigations in other model organisms, complementing a recent comprehensive review of human pancreas development (Jennings et al., 2015) [1]. The understanding of pancreas development in model organisms provides a framework to interpret how human mutations lead to neonatal diabetes and may contribute to other forms of diabetes and to guide the production of desired pancreatic cell types from pluripotent stem cells for therapeutic purposes.
Collapse
Affiliation(s)
- Hjalte List Larsen
- DanStem, University of Copenhagen, 3 B Blegdamsvej, DK-2200 Copenhagen N, Denmark
| | - Anne Grapin-Botton
- DanStem, University of Copenhagen, 3 B Blegdamsvej, DK-2200 Copenhagen N, Denmark.
| |
Collapse
|
49
|
Abstract
The zebrafish pancreas shares its basic organization and cell types with the mammalian pancreas. In addition, the developmental pathways that lead to the establishment of the pancreatic islets of Langherhans are generally conserved from fish to mammals. Zebrafish provides a powerful tool to probe the mechanisms controlling establishment of the pancreatic endocrine cell types from early embryonic progenitor cells, as well as the regeneration of endocrine cells after damage. This knowledge is, in turn, applicable to refining protocols to generate renewable sources of human pancreatic islet cells that are critical for regulation of blood sugar levels. Here, we review how previous and ongoing studies in zebrafish and beyond are influencing the understanding of molecular mechanisms underlying various forms of diabetes and efforts to develop cell-based approaches to cure this increasingly widespread disease.
Collapse
|
50
|
Efrat S. Mechanisms of adult human β-cell in vitro dedifferentiation and redifferentiation. Diabetes Obes Metab 2016; 18 Suppl 1:97-101. [PMID: 27615137 DOI: 10.1111/dom.12724] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 04/20/2016] [Indexed: 12/13/2022]
Abstract
Recent studies in animal models and human pathological specimens suggest the involvement of β-cell dedifferentiation in β-cell dysfunction associated with type 2 diabetes. Dedifferentiated β-cells may be exploited for endogenous renewal of the β-cell mass. However, studying human β-cell dedifferentiation in diabetes presents major difficulties. We have analysed mechanisms involved in human β-cell dedifferentiation in vitro, under conditions that allow cell proliferation. Although there are important differences between the two cellular environments, β-cell dedifferentiation in the two conditions is likely to share a number of common pathways. Insights from the in vitro studies may lead to development of approaches for redifferentiation of endogenous dedifferentiated β-cells.
Collapse
Affiliation(s)
- S Efrat
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv, Israel.
| |
Collapse
|