1
|
Huang X, Zhao H, Chen H, Liu Z, Liu K, Lv Z, Liu X, Han X, Han M, Lu J, Zhou Q, Zhou B. Ductal or Ngn3 + cells do not contribute to adult pancreatic islet beta-cell neogenesis in homeostasis. EMBO J 2025:10.1038/s44318-025-00434-z. [PMID: 40205162 DOI: 10.1038/s44318-025-00434-z] [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: 04/17/2024] [Revised: 03/15/2025] [Accepted: 03/21/2025] [Indexed: 04/11/2025] Open
Abstract
The adult pancreatic ducts have long been proposed to contain rare progenitors, some of which expressing Ngn3, that generate new beta cells in endocrine-islet homeostasis. Due to their postulated rarity and the lack of definitive markers, the existence or absence of ductal endocrine progenitors remains unsettled despite many studies. Genetic lineage tracing of ductal cells or Ngn3+ cells with currently available CreER drivers has been complicated by off-target labeling of pre-existing beta cells. Here, using dual-recombinase-mediated intersectional genetic strategy and newly-derived Ngn3-2A-CreER and Hnf1b-2A-CreER knock-in drivers, we succeeded in specifically labeling Ngn3-positive cells and Hnf1b-positive ductal cells without marking pre-existing beta cells. These data revealed no evidence of de novo generation of insulin-producing beta cells from ductal cells or endogenous Ngn3-positive cells in the adult pancreas during homeostasis.
Collapse
Affiliation(s)
- Xiuzhen Huang
- New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Huan Zhao
- New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China.
| | - Hui Chen
- New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Zixin Liu
- New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Kuo Liu
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, 310024, Hangzhou, China
| | - Zan Lv
- New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Xiuxiu Liu
- New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Ximeng Han
- New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Maoying Han
- New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Jie Lu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 200080, Shanghai, China
| | - Qiao Zhou
- Division of Regenerative Medicine & Hartman Institute for Organ Regeneration, Department of Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Bin Zhou
- New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China.
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, 310024, Hangzhou, China.
- School of Life Science and Technology, ShanghaiTech University, 100 Haike Road, 201210, Shanghai, China.
| |
Collapse
|
2
|
Yang L, Yu XX, Wang X, Jin CT, Xu CR. The expression order determines the pioneer functions of NGN3 and NEUROD1 in pancreatic endocrine differentiation. SCIENCE ADVANCES 2025; 11:eadt4770. [PMID: 40138419 PMCID: PMC11939047 DOI: 10.1126/sciadv.adt4770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 02/20/2025] [Indexed: 03/29/2025]
Abstract
Pioneer transcription factors (TFs) initiate chromatin remodeling, which is crucial for gene regulation and cell differentiation. In this study, we investigated how the sequential expression of neurogenin 3 (NGN3) and NEUROD1 affects their pioneering functions during pancreatic endocrine differentiation. Using a genetically engineered mouse model, we mapped NGN3-binding sites, confirming the pivotal role of this molecule in regulating chromatin accessibility. The pioneering function of NGN3 involves dose tolerance, and low doses are sufficient. Although NEUROD1 generally acts as a conventional TF, it can assume a pioneering role in the absence of NGN3. The sequential expression of NeuroD1 and Ngn3 predominantly drives α cell generation, which may explain the inefficient β cell induction observed in vitro. Our findings demonstrate that pioneer activity is dynamically shaped by temporal TF expression and inter-TF interactions, providing insights into transcriptional regulation and its implications for disease mechanisms and therapeutic targeting and enhancing in vitro differentiation strategies.
Collapse
Affiliation(s)
- Liu Yang
- State Key Laboratory of Female Fertility Promotion, Department of Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing 100191, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Xin-Xin Yu
- State Key Laboratory of Female Fertility Promotion, Department of Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing 100191, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Xin Wang
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
- School of Life Sciences, Peking University, Beijing 100871, China
| | - Chen-Tao Jin
- State Key Laboratory of Female Fertility Promotion, Department of Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Cheng-Ran Xu
- State Key Laboratory of Female Fertility Promotion, Department of Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing 100191, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| |
Collapse
|
3
|
Arai T, Hayashi E, Maeda S, Matsubara T, Fujii H, Shinohara K, Sogabe A, Wainai S, Tanaka D, Ono Y, Ono Y, Yoshikai M, Sorimachi Y, Kok CYY, Shimoda M, Tanaka M, Kawada N, Goda N. Liver-derived Neuregulin1α stimulates compensatory pancreatic β cell hyperplasia in insulin resistance. Nat Commun 2025; 16:1950. [PMID: 40082404 PMCID: PMC11906622 DOI: 10.1038/s41467-025-57167-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 02/13/2025] [Indexed: 03/16/2025] Open
Abstract
Compensatory pancreatic islet hyperplasia is an adaptive response to increased systemic insulin demand, although factors meditating this response remain poorly understood. Here, we show that a liver-derived secreted protein, Neuregulin1α, promotes compensatory proliferation of pancreatic β cells in type 2 diabetes. Liver Neuregulin1α expression and serum Neuregulin1α levels increase in male mice fed an obesity-inducing diet. Male mice lacking either Neuregulin1 in liver or its receptor, ErbB3, in β cells deteriorate systemic glucose disposal due to impaired β cell expansion with reduced insulin secretion when fed the obesity-inducing diet. Mechanistically, Neuregulin1α activates ERBB2/3-ERK signaling to stimulate β cell proliferation without altering glucose-stimulated insulin secretion potential. In patients with metabolic dysfunction-associated steatotic liver disease (MASLD) and obesity but without type 2 diabetes serum Neuregulin1α levels increase, while in patient with MASLD and type 2 diabetes show markedly reduced levels of Neuregulin1α. These results suggest that Neuregulin1α serves as a hepatokine that can expand functional β cell mass in type 2 diabetes.
Collapse
Affiliation(s)
- Takatomo Arai
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Eriko Hayashi
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Sumie Maeda
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Tsutomu Matsubara
- Department of Anatomy and Regenerative Biology, Osaka Metropolitan University, Osaka, Japan
| | - Hideki Fujii
- Department of Hepatology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Koya Shinohara
- Department of Pancreatic Islet Cell Transplantation, National Center for Global Health and Medicine, Tokyo, Japan
| | - Arisu Sogabe
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Sadatomo Wainai
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Daishi Tanaka
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Yutaro Ono
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Yumika Ono
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Minami Yoshikai
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Yuriko Sorimachi
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Cindy Yuet-Yin Kok
- Neuroscience Research Australia, Sydney, NSW, Australia
- Discipline of Medicine, Randwick Clinical Campus, University of New South Wales, Sydney, NSW, Australia
| | - Masayuki Shimoda
- Department of Pancreatic Islet Cell Transplantation, National Center for Global Health and Medicine, Tokyo, Japan
| | - Minoru Tanaka
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Norifumi Kawada
- Department of Hepatology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Nobuhito Goda
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan.
| |
Collapse
|
4
|
Wong A, Alejandro EU. Post translational modification regulation of transcription factors governing pancreatic β-cell identity and functional mass. Front Endocrinol (Lausanne) 2025; 16:1562646. [PMID: 40134803 PMCID: PMC11932907 DOI: 10.3389/fendo.2025.1562646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 02/17/2025] [Indexed: 03/27/2025] Open
Abstract
Dysfunction of the insulin-secreting β-cells is a key hallmark of Type 2 diabetes (T2D). In the natural history of the progression of T2D, factors such as genetics, early life exposures, lifestyle, and obesity dictate an individual's susceptibility risk to disease. Obesity is associated with insulin resistance and increased demand for insulin to maintain glucose homeostasis. Studies in both mouse and human islets have implicated the β-cell's ability to compensate through proliferation and survival (increasing functional β-cell mass) as a tipping point toward the development of disease. A growing body of evidence suggests the reduction of β-cell mass in T2D is driven majorly by loss of β-cell identity, rather than by apoptosis alone. The development and maintenance of pancreatic β-cell identity, function, and adaptation to stress is governed, in part, by the spatiotemporal expression of transcription factors (TFs), whose activity is regulated by signal-dependent post-translational modifications (PTM). In this review, we examine the role of these TFs in the developing pancreas and in the mature β-cell. We discuss functional implications of post-translational modifications on these transcription factors' activities and how an understanding of the pathways they regulate can inform therapies to promoteβ-cell regeneration, proliferation, and survival in diabetes.
Collapse
Affiliation(s)
- Alicia Wong
- Department of Genetics, Cell Biology, and Development, University of Minnesota Twin Cities, Minneapolis, MN, United States
| | - Emilyn U. Alejandro
- Department of Integrative Biology and Physiology, University of Minnesota Twin Cities, Minneapolis, MN, United States
| |
Collapse
|
5
|
Kosheleva L, Koshelev D, Lagunas-Rangel FA, Levit S, Rabinovitch A, Schiöth HB. Disease-modifying pharmacological treatments of type 1 diabetes: Molecular mechanisms, target checkpoints, and possible combinatorial treatments. Pharmacol Rev 2025; 77:100044. [PMID: 40014914 PMCID: PMC11964952 DOI: 10.1016/j.pharmr.2025.100044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/10/2025] [Indexed: 03/01/2025] Open
Abstract
After a century of extensive scientific investigations, there is still no curative or disease-modifying treatment available that can provide long-lasting remission for patients diagnosed with type 1 diabetes (T1D). Although T1D has historically been regarded as a classic autoimmune disorder targeting and destroying pancreatic islet β-cells, significant research has recently demonstrated that β-cells themselves also play a substantial role in the disease's progression, which could explain some of the unfavorable clinical outcomes. We offer a thorough review of scientific and clinical insights pertaining to molecular mechanisms behind pathogenesis and the different therapeutic interventions in T1D covering over 20 possible pharmaceutical intervention treatments. The interventions are categorized as immune therapies, treatments targeting islet endocrine dysfunctions, medications with dual modes of action in immune and islet endocrine cells, and combination treatments with a broader spectrum of activity. We suggest that these collective findings can provide a valuable platform to discover new combinatorial synergies in search of the curative disease-modifying intervention for T1D. SIGNIFICANCE STATEMENT: This research delves into the underlying causes of T1D and identifies critical mechanisms governing β-cell function in both healthy and diseased states. Thus, we identify specific pathways that could be manipulated by existing or new pharmacological interventions. These interventions fall into several categories: (1) immunomodifying therapies individually targeting immune cell processes, (2) interventions targeting β-cells, (3) compounds that act simultaneously on both immune cell and β-cell pathways, and (4) combinations of compounds simultaneously targeting immune and β-cell pathways.
Collapse
Affiliation(s)
- Liudmila Kosheleva
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden
| | - Daniil Koshelev
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden
| | - Francisco Alejandro Lagunas-Rangel
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden; Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Shmuel Levit
- Diabetes and Metabolism Institute, Assuta Medical Centers, Tel Aviv, Israel
| | | | - Helgi B Schiöth
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden; Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia.
| |
Collapse
|
6
|
Ziojła NM, Socha M, Guerra MC, Kizewska D, Blaszczyk K, Urbaniak E, Henry S, Grabowska M, Niakan KK, Warmflash A, Borowiak M. ETVs dictate hPSC differentiation by tuning biophysical properties. Nat Commun 2025; 16:1999. [PMID: 40011454 PMCID: PMC11865489 DOI: 10.1038/s41467-025-56591-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 01/20/2025] [Indexed: 02/28/2025] Open
Abstract
Stem cells maintain a dynamic dialog with their niche, integrating biochemical and biophysical cues to modulate cellular behavior. Yet, the transcriptional networks that regulate cellular biophysical properties remain poorly defined. Here, we leverage human pluripotent stem cells (hPSCs) and two morphogenesis models - gastruloids and pancreatic differentiation - to establish ETV transcription factors as critical regulators of biophysical parameters and lineage commitment. Genetic ablation of ETV1 or ETV1/ETV4/ETV5 in hPSCs enhances cell-cell and cell-ECM adhesion, leading to aberrant multilineage differentiation including disrupted germ-layer organization, ectoderm loss, and extraembryonic cell overgrowth in gastruloids. Furthermore, ETV1 loss abolishes pancreatic progenitor formation. Single-cell RNA sequencing and follow-up assays reveal dysregulated mechanotransduction via the PI3K/AKT signaling. Our findings highlight the importance of transcriptional control over cell biophysical properties and suggest that manipulating these properties may improve in vitro cell and tissue engineering strategies.
Collapse
Affiliation(s)
- Natalia M Ziojła
- Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, Poznan, Poland
| | - Magdalena Socha
- Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, Poznan, Poland
| | | | - Dorota Kizewska
- Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, Poznan, Poland
| | - Katarzyna Blaszczyk
- Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, Poznan, Poland
| | - Edyta Urbaniak
- Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, Poznan, Poland
| | - Sara Henry
- Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, Poznan, Poland
| | - Malgorzata Grabowska
- Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, Poznan, Poland
| | - Kathy K Niakan
- The Loke Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Aryeh Warmflash
- Department of Biosciences, Rice University, Houston, TX, USA
| | - Malgorzata Borowiak
- Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, Poznan, Poland.
- McNair Medical Institute, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
7
|
Jacques K, Coles BLK, van der Kooy D. Pancreatic stem cells originate during the pancreatic progenitor developmental stage. Front Cell Dev Biol 2025; 13:1521411. [PMID: 40040790 PMCID: PMC11876382 DOI: 10.3389/fcell.2025.1521411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 01/22/2025] [Indexed: 03/06/2025] Open
Abstract
Previously isolated adult pancreatic precursors called pancreatic multipotent progenitors (which make both pancreatic endocrine and exocrine cell types) originate from the Pancreatic Duodenal Homeobox 1 (PDX1) pancreatic developmental lineage. The embryonic time point at which adult pancreatic multipotent progenitor cells emerge has not been established. We have employed the use of two models: a human embryonic stem cell (hESC) to beta-cell cytokine-induced differentiation protocol and a mouse lineage tracing model during early development to isolate clonal pancreatic spheres. The results show that insulin-positive clonal spheres can be isolated as early as the pancreatic endoderm stage as well as the pancreatic progenitor stage during the hESC to beta-cell lineage differentiation model and that they can be isolated only as early as the pancreatic progenitor stage during mouse embryogenesis. Further, pancreatic clonal sphere-forming cells isolated from the pancreatic progenitor stage in embryonic mice display multipotentiality, and those isolated at a later gestational age demonstrate self-renewal ability. These findings suggest that pancreatic precursors isolated from mouse embryonic time points have stem cell properties and that the pancreatic progenitor stage in hESC development may be the optimal time to capture and expand these stem cells and make large numbers of beta cells.
Collapse
Affiliation(s)
- Krystal Jacques
- Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Brenda L. K. Coles
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Derek van der Kooy
- Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
8
|
Li KR, Yu PL, Zheng QQ, Wang X, Fang X, Li LC, Xu CR. Spatiotemporal and genetic cell lineage tracing of endodermal organogenesis at single-cell resolution. Cell 2025; 188:796-813.e24. [PMID: 39824184 DOI: 10.1016/j.cell.2024.12.012] [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: 05/13/2024] [Revised: 09/30/2024] [Accepted: 12/09/2024] [Indexed: 01/20/2025]
Abstract
During early mammalian development, the endoderm germ layer forms the foundation of the respiratory and digestive systems through complex patterning. This intricate process, guided by a series of cell fate decisions, remains only partially understood. Our study introduces innovative genetic tracing codes for 14 distinct endodermal regions using novel mouse strains. By integrating high-throughput and high-precision single-cell RNA sequencing with sophisticated imaging, we detailed the spatiotemporal and genetic lineage differentiation of the endoderm at single-cell resolution. We discovered an unexpected multipotentiality within early endodermal regions, allowing differentiation into various organ primordia. This research illuminates the complex and underestimated phenomenon where endodermal organs develop from multiple origins, prompting a reevaluation of traditional differentiation models. Our findings advance understanding in developmental biology and have significant implications for regenerative medicine and the development of advanced organoid models, providing insights into the intricate mechanisms that guide organogenesis.
Collapse
Affiliation(s)
- Ke-Ran Li
- State Key Laboratory of Female Fertility Promotion, Department of Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Pei-Long Yu
- State Key Laboratory of Female Fertility Promotion, Department of Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Qi-Qi Zheng
- PKU-Tsinghua-NIBS Graduate Program, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Xin Wang
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China; School of Life Sciences, Peking University, Beijing 100871, China
| | - Xuan Fang
- Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Lin-Chen Li
- Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Cheng-Ran Xu
- State Key Laboratory of Female Fertility Promotion, Department of Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China.
| |
Collapse
|
9
|
Brooks EP, Casey MR, Wells KL, Liu TY, Van Orman M, Sussel L. NKX2.2 and KLF4 cooperate to regulate α-cell identity. Genes Dev 2025; 39:242-260. [PMID: 39797760 PMCID: PMC11789634 DOI: 10.1101/gad.352193.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/12/2024] [Indexed: 01/13/2025]
Abstract
Transcription factors (TFs) are indispensable for maintaining cell identity through regulating cell-specific gene expression. Distinct cell identities derived from a common progenitor are frequently perpetuated by shared TFs, yet the mechanisms that enable these TFs to regulate cell-specific targets are poorly characterized. We report that the TF NKX2.2 is critical for the identity of pancreatic islet α cells by directly activating α-cell genes and repressing alternate islet cell fate genes. When compared with the known role of NKX2.2 in islet β cells, we demonstrate that NKX2.2 regulates α-cell genes, facilitated in part by α-cell-specific DNA binding at gene promoters. Furthermore, we have identified the reprogramming factor KLF4 as having enriched expression in α cells, where it co-occupies NKX2.2-bound α-cell promoters, is necessary for NKX2.2 promoter occupancy in α cells, and coregulates many NKX2.2 α-cell transcriptional targets. Overexpression of Klf4 in β cells is sufficient to manipulate chromatin accessibility, increase binding of NKX2.2 at α-cell-specific promoter sites, and alter expression of NKX2.2-regulated cell-specific targets. This study identifies KLF4 as a novel α-cell factor that cooperates with NKX2.2 to regulate α-cell identity.
Collapse
Affiliation(s)
- Elliott P Brooks
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - McKenna R Casey
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - Kristen L Wells
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - Tsung-Yun Liu
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - Madeline Van Orman
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - Lori Sussel
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| |
Collapse
|
10
|
Zhao H, Zhou B. Lineage tracing of pancreatic cells for mechanistic and therapeutic insights. Trends Endocrinol Metab 2025:S1043-2760(24)00330-8. [PMID: 39828453 DOI: 10.1016/j.tem.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 01/22/2025]
Abstract
Recent advances in lineage-tracing technologies have significantly improved our understanding of pancreatic cell biology, particularly in elucidating the ontogeny and regenerative capacity of pancreatic cells. A deeper appreciation of the mechanisms underlying pancreatic cell identity and plasticity holds the potential to inform the development of new therapeutic modalities for conditions such as diabetes and pancreatitis. With this goal in mind, here we summarize advances, challenges, and future directions in tracing pancreatic cell origins and fates using lineage-tracing technologies. Given their essential role for blood glucose regulation, we pay particular attention on the insights gained from endocrine cells, especially β-cells.
Collapse
Affiliation(s)
- Huan Zhao
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Bin Zhou
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, China; Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
| |
Collapse
|
11
|
Bandral M, Sussel L, Lorberbaum DS. Retinoid signaling in pancreas development, islet function, and disease. Curr Top Dev Biol 2024; 161:297-318. [PMID: 39870436 DOI: 10.1016/bs.ctdb.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
All-trans retinoic acid (ATRA) signaling is essential in numerous different biological contexts. This review highlights the diverse roles of ATRA during development, function, and diseases of the pancreas. ATRA is essential to specify pancreatic progenitors from gut tube endoderm, endocrine and exocrine differentiation, and adult islet function. ATRA concentration must be carefully regulated during the derivation of islet-like cells from human pluripotent stem cells (hPSCs) to optimize the expression of key pancreatic transcription factors while mitigating adverse and unwanted cell-types in these cultures. The ATRA pathway is integral to the pancreas and here we will present selected studies from decades of research that has laid the essential groundwork for ongoing projects dedicated to unraveling the complexities of ATRA signaling in the pancreas.
Collapse
Affiliation(s)
- Manuj Bandral
- University of Michigan, Department of Pharmacology, Caswell Diabetes Institute, Ann Arbor, MI, United States
| | - Lori Sussel
- University of Colorado Denver Anschutz Medical Campus, Barbara Davis Center for Diabetes, Aurora, CO, United States
| | - David S Lorberbaum
- University of Michigan, Department of Pharmacology, Caswell Diabetes Institute, Ann Arbor, MI, United States.
| |
Collapse
|
12
|
Badder LM, Davies JA, Meniel VS, Marušková M, Salvador-Barbero B, Bayliss RJ, Phesse TJ, Hogan C, Parker AL. The αvβ6 integrin specific virotherapy, Ad5 NULL-A20.FCU1, selectively delivers potent "in-tumour" chemotherapy to pancreatic ductal adenocarcinoma. Br J Cancer 2024; 131:1694-1706. [PMID: 39369056 PMCID: PMC11555051 DOI: 10.1038/s41416-024-02869-3] [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: 05/06/2024] [Revised: 09/21/2024] [Accepted: 09/25/2024] [Indexed: 10/07/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) represent an unmet clinical need. Approximately 90% of PDACs express high levels of αvβ6 integrin. We have previously described Ad5NULL-A20, an adenovirus vector with ablated native means of cell entry and retargeted to αvβ6 integrin by incorporation of an A20 peptide. METHODS Here, we incorporate suicide genes FCY1 and FCU1 encoding for cytosine deaminase (CDase) or a combination of CDase and UPRTase, capable of catalysing a non-toxic prodrug, 5-FC into the chemotherapeutic 5-FU and downstream metabolites, into replication-deficient Ad5 and Ad5NULL-A20. RESULTS We show that Ad5NULL-A20 enables the transfer of suicide genes to αvβ6 integrin-positive PDAC cells which, in combination with 5-FC, results in cell death in vitro which is further mediated by a bystander effect in non-transduced cells. Intratumoural delivery of Ad5NULL-A20.FCU1 in combination with intraperitoneal delivery of 5-FC further results in tumour growth inhibition in a cell line xenograft in vivo. Using clinically-relevant 3D organoid models, we show selective transduction and therapeutic efficacy of FCU1 transgenes in combination with 5-FC. CONCLUSION Taken together these data provide the preclinical rationale for combined Ad5NULL-A20.FCU1 plus 5-FC as a promising targeted therapy to mediate "in-tumour chemotherapy" and merits further investigation for the treatment of PDAC patients.
Collapse
Affiliation(s)
- Luned M Badder
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK
| | - James A Davies
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK
| | - Valerie S Meniel
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff, CF24 4HQ, UK
| | - Mahulena Marušková
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK
| | - Beatriz Salvador-Barbero
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff, CF24 4HQ, UK
| | - Rebecca J Bayliss
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK
| | - Toby J Phesse
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff, CF24 4HQ, UK
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Catherine Hogan
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff, CF24 4HQ, UK
| | - Alan L Parker
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK.
- Systems Immunity University Research Institute, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK.
| |
Collapse
|
13
|
Zhang T, Wang N, Liao Z, Chen J, Meng H, Lin H, Xu T, Chen L, Zhu LQ, Liu H. A differentiation protocol for generating pancreatic delta cells from human pluripotent stem cells. Front Cell Dev Biol 2024; 12:1490040. [PMID: 39493348 PMCID: PMC11527672 DOI: 10.3389/fcell.2024.1490040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 10/07/2024] [Indexed: 11/05/2024] Open
Abstract
In this protocol, we detail a seven-stage differentiation methodology for generating pancreatic delta cells (SC-delta cells) from human pluripotent stem cells (hPSCs). In the first step, definitive endoderm is generated by activin A and CHIR99021, followed by induction of primitive gut tube and posterior foregut by treatment with FGF7, SANT1, LDN193189, PdBU, and retinoic acid (RA). The subsequent endocrine generation and directed SC-delta cell induction is achieved by a combined treatment of the FGF7 with FGF2 during stage 4 and 5, together with RA, XXI, ALK5 inhibitor II, SANT1, Betacellulin and LDN193189. The planar cultivation is converted to a suspended system after stage 5, allowing cells to aggregate into delta cell-containing spheroids. The differentiation takes approximately 4-5 weeks for delta cell generation and an additional 1-2 weeks for cell expansion and evaluation. We believe that this amenable and simplified protocol can provide a stable source of SC-delta cells from efficient differentiation, facilitating further investigation of the physiological role of delta cells as well as refinement of islet cell therapeutic strategies.
Collapse
Affiliation(s)
- Tongran Zhang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Testing and Diagnosis Technology Research, Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Nannan Wang
- Department of Testing and Diagnosis Technology Research, Guangzhou National Laboratory, Guangzhou, Guangdong, China
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhiying Liao
- Department of Testing and Diagnosis Technology Research, Guangzhou National Laboratory, Guangzhou, Guangdong, China
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jingyi Chen
- Department of Testing and Diagnosis Technology Research, Guangzhou National Laboratory, Guangzhou, Guangdong, China
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, China
| | - Hao Meng
- Department of Testing and Diagnosis Technology Research, Guangzhou National Laboratory, Guangzhou, Guangdong, China
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Haopeng Lin
- Department of Testing and Diagnosis Technology Research, Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Tao Xu
- Department of Testing and Diagnosis Technology Research, Guangzhou National Laboratory, Guangzhou, Guangdong, China
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lihua Chen
- Department of Testing and Diagnosis Technology Research, Guangzhou National Laboratory, Guangzhou, Guangdong, China
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ling-Qiang Zhu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Huisheng Liu
- Department of Testing and Diagnosis Technology Research, Guangzhou National Laboratory, Guangzhou, Guangdong, China
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong, China
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, China
| |
Collapse
|
14
|
Salazar Leon LE, Kim LH, Sillitoe RV. Cerebellar deep brain stimulation as a dual-function therapeutic for restoring movement and sleep in dystonic mice. Neurotherapeutics 2024; 21:e00467. [PMID: 39448336 PMCID: PMC11585869 DOI: 10.1016/j.neurot.2024.e00467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/26/2024] Open
Abstract
Dystonia arises with cerebellar dysfunction, which plays a key role in the emergence of multiple pathophysiological deficits that range from abnormal movements and postures to disrupted sleep. Current therapeutic interventions typically do not simultaneously address both the motor and non-motor symptoms of dystonia, underscoring the necessity for a multi-functional therapeutic strategy. Deep brain stimulation (DBS) is effectively used to reduce motor symptoms in dystonia, with existing parallel evidence arguing for its potential to correct sleep disturbances. However, the simultaneous efficacy of DBS for improving sleep and motor dysfunction, specifically by targeting the cerebellum, remains underexplored. Here, we test the effect of cerebellar DBS in two genetic mouse models with dystonia that exhibit sleep defects-Ptf1aCre;Vglut2fx/fx and Pdx1Cre;Vglut2fx/fx-which have overlapping cerebellar circuit miswiring defects but differing severity in motor phenotypes. By targeting DBS to the fiber tracts located between the cerebellar fastigial and the interposed nuclei (FN + INT-DBS), we modulated sleep dysfunction by enhancing sleep quality and timing. This DBS paradigm improved wakefulness and rapid eye movement sleep in both mutants. Additionally, the latency to reach REM sleep, a deficit observed in human dystonia patients, was reduced in both models. Cerebellar DBS also induced alterations in the electrocorticogram (ECoG) patterns that define sleep states. As expected, DBS reduced the severe dystonic twisting motor symptoms that are observed in the Ptf1aCre;Vglut2fx/fx mice. These findings highlight the potential for using cerebellar DBS to simultaneously improve sleep and reduce motor dysfunction in dystonia and uncover its potential as a dual-effect in vivo therapeutic strategy.
Collapse
Affiliation(s)
- Luis E Salazar Leon
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA; Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, 77030, USA
| | - Linda H Kim
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, 77030, USA
| | - Roy V Sillitoe
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA; Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, 77030, USA.
| |
Collapse
|
15
|
Tong X, Yagan M, Hu R, Nevills S, Doss TD, Stein RW, Balamurugan AN, Gu G. Metabolic Stress Levels Influence the Ability of Myelin Transcription Factors to Regulate β-Cell Identity and Survival. Diabetes 2024; 73:1662-1672. [PMID: 39058602 PMCID: PMC11417441 DOI: 10.2337/db23-0528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 07/14/2024] [Indexed: 07/28/2024]
Abstract
A hallmark of type 2 diabetes (T2D) is endocrine islet β-cell failure, which can occur via cell dysfunction, loss of identity, and/or death. How each is induced remains largely unknown. We used mouse β-cells deficient for myelin transcription factors (Myt TFs; including Myt1, -2, and -3) to address this question. We previously reported that inactivating all three Myt genes in pancreatic progenitor cells (MytPancΔ) caused β-cell failure and late-onset diabetes in mice. Their lower expression in human β-cells is correlated with β-cell dysfunction, and single nucleotide polymorphisms in MYT2 and MYT3 are associated with a higher risk of T2D. We now show that these Myt TF-deficient postnatal β-cells also dedifferentiate by reactivating several progenitor markers. Intriguingly, mosaic Myt TF inactivation in only a portion of islet β-cells did not result in overt diabetes, but this created a condition where Myt TF-deficient β-cells remained alive while activating several markers of Ppy-expressing islet cells. By transplanting MytPancΔ islets into the anterior eye chambers of immune-compromised mice, we directly show that glycemic and obesity-related conditions influence cell fate, with euglycemia inducing several Ppy+ cell markers and hyperglycemia and insulin resistance inducing additional cell death. These findings suggest that the observed β-cell defects in T2D depend not only on their inherent genetic/epigenetic defects but also on the metabolic load. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Xin Tong
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Mahircan Yagan
- Program in Developmental Biology, Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
- Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Ruiying Hu
- Program in Developmental Biology, Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
- Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Simone Nevills
- Program in Developmental Biology, Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
- Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Teri D. Doss
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Roland W. Stein
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Appakalai N. Balamurugan
- Center for Clinical and Translational Research, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH
| | - Guoqiang Gu
- Program in Developmental Biology, Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
- Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN
| |
Collapse
|
16
|
Rangel-Sosa MM, Mann F, Chauvet S. Pancreatic Schwann cell reprogramming supports cancer-associated neuronal remodeling. Glia 2024; 72:1840-1861. [PMID: 38961612 DOI: 10.1002/glia.24586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 07/05/2024]
Abstract
The peripheral nervous system is a key regulator of cancer progression. In pancreatic ductal adenocarcinoma (PDAC), the sympathetic branch of the autonomic nervous system inhibits cancer development. This inhibition is associated with extensive sympathetic nerve sprouting in early pancreatic cancer precursor lesions. However, the underlying mechanisms behind this process remain unclear. This study aimed to investigate the roles of pancreatic Schwann cells in the structural plasticity of sympathetic neurons. We examined the changes in the number and distribution of Schwann cells in a transgenic mouse model of PDAC and in a model of metaplastic pancreatic lesions induced by chronic inflammation. Schwann cells proliferated and expanded simultaneously with new sympathetic nerve sprouts in metaplastic/neoplastic pancreatic lesions. Sparse genetic labeling showed that individual Schwann cells in these lesions had a more elongated and branched structure than those under physiological conditions. Schwann cells overexpressed neurotrophic factors, including glial cell-derived neurotrophic factor (GDNF). Sympathetic neurons upregulated the GDNF receptors and exhibited enhanced neurite growth in response to GDNF in vitro. Selective genetic deletion of Gdnf in Schwann cells completely blocked sympathetic nerve sprouting in metaplastic pancreatic lesions in vivo. This study demonstrated that pancreatic Schwann cells underwent adaptive reprogramming during early cancer development, supporting a protective antitumor neuronal response. These finding could help to develop new strategies to modulate cancer associated neural plasticity.
Collapse
Affiliation(s)
| | - Fanny Mann
- Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | | |
Collapse
|
17
|
Al-Adsani AM, Al-Qattan KK, Barhoush SA, Abbood MS, Al-Bustan SA. Garlic Extract Promotes Pancreatic Islet Neogenesis Through α-to-β-Cell Transdifferentiation and Normalizes Glucose Homeostasis in Diabetic Rats. Mol Nutr Food Res 2024; 68:e2400362. [PMID: 39205537 DOI: 10.1002/mnfr.202400362] [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: 05/16/2024] [Revised: 07/01/2024] [Indexed: 09/04/2024]
Abstract
SCOPE Garlic extract (GE) has been shown to ameliorate hyperglycemia in diabetic rats (DRs) by increasing insulin production. However, the mechanism through which it exerts its effects remains unclear. Here, it investigates the molecular process and the origin of regenerating β-cell in rats with streptozotocin (STZ)-induced diabetes in response to GE. METHODS AND RESULTS In this study, quantitative RT-PCR (qRT-PCR), western blotting, and immunohistochemical analysis are carried out after pancreas isolation. These findings show that 1 week of GE treatment increases the expression of the endocrine progenitor cell markers Neurogenin3 (Neurog3), pancreatic and duodenal homeobox 1 (Pdx1), neurogenic differentiation factor 1 (Neurod1), paired box proteins (Pax)4, V-maf musculoaponeurotic fibrosarcoma oncogene homolog B (Mafb), and NK homeobox factors (Nkx)6-1 in STZ-induced DRs. Continuation with GE treatment for 8 weeks causes the expression of the mature β-cell markers insulin(Ins)2, urocortin3 (Ucn3), and glucose transporter 2 (Glut2) to peak. Comprehensive examination of the islet through immunohistochemical analysis reveals the presence of a heterogeneous cell population including INS+/GLUT2- and INS+/GLUT2+ β-cell subpopulations with few bihormonal INS+/GCG+ cells after 4 weeks. By week 8, islet architecture is reestablished, and glucose-stimulated insulin secretion was restored through the upregulation of Ucn3. CONCLUSION GE induces β-cell neogenesis in DRs and restores islet architecture. The newly formed mature β-like cells could have originated through the differentiation of endocrine progenitor cells as well as α- to β-cell transdifferentiation.
Collapse
Affiliation(s)
- Amani M Al-Adsani
- Department of Biological Sciences, Faculty of Science, Kuwait University, P.O. Box 5969, Safat, 13060, Kuwait
| | - Khaled K Al-Qattan
- Department of Biological Sciences, Faculty of Science, Kuwait University, P.O. Box 5969, Safat, 13060, Kuwait
| | - Sahar A Barhoush
- Department of Biological Sciences, Faculty of Science, Kuwait University, P.O. Box 5969, Safat, 13060, Kuwait
| | - Manal S Abbood
- Department of Biological Sciences, Faculty of Science, Kuwait University, P.O. Box 5969, Safat, 13060, Kuwait
| | - Suzanne A Al-Bustan
- Department of Biological Sciences, Faculty of Science, Kuwait University, P.O. Box 5969, Safat, 13060, Kuwait
| |
Collapse
|
18
|
van der Heijden ME, Brown AM, Kizek DJ, Sillitoe RV. Cerebellar nuclei cells produce distinct pathogenic spike signatures in mouse models of ataxia, dystonia, and tremor. eLife 2024; 12:RP91483. [PMID: 39072369 DOI: 10.7554/elife.91483] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024] Open
Abstract
The cerebellum contributes to a diverse array of motor conditions, including ataxia, dystonia, and tremor. The neural substrates that encode this diversity are unclear. Here, we tested whether the neural spike activity of cerebellar output neurons is distinct between movement disorders with different impairments, generalizable across movement disorders with similar impairments, and capable of causing distinct movement impairments. Using in vivo awake recordings as input data, we trained a supervised classifier model to differentiate the spike parameters between mouse models for ataxia, dystonia, and tremor. The classifier model correctly assigned mouse phenotypes based on single-neuron signatures. Spike signatures were shared across etiologically distinct but phenotypically similar disease models. Mimicking these pathophysiological spike signatures with optogenetics induced the predicted motor impairments in otherwise healthy mice. These data show that distinct spike signatures promote the behavioral presentation of cerebellar diseases.
Collapse
Affiliation(s)
- Meike E van der Heijden
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, United States
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
| | - Amanda M Brown
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, United States
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
| | - Dominic J Kizek
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
| | - Roy V Sillitoe
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, United States
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
- Department of Pediatrics, Baylor College of Medicine, Houston, United States
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, United States
| |
Collapse
|
19
|
Chen L, Wang N, Zhang T, Zhang F, Zhang W, Meng H, Chen J, Liao Z, Xu X, Ma Z, Xu T, Liu H. Directed differentiation of pancreatic δ cells from human pluripotent stem cells. Nat Commun 2024; 15:6344. [PMID: 39068220 PMCID: PMC11283558 DOI: 10.1038/s41467-024-50611-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 07/11/2024] [Indexed: 07/30/2024] Open
Abstract
Dysfunction of pancreatic δ cells contributes to the etiology of diabetes. Despite their important role, human δ cells are scarce, limiting physiological studies and drug discovery targeting δ cells. To date, no directed δ-cell differentiation method has been established. Here, we demonstrate that fibroblast growth factor (FGF) 7 promotes pancreatic endoderm/progenitor differentiation, whereas FGF2 biases cells towards the pancreatic δ-cell lineage via FGF receptor 1. We develop a differentiation method to generate δ cells from human stem cells by combining FGF2 with FGF7, which synergistically directs pancreatic lineage differentiation and modulates the expression of transcription factors and SST activators during endoderm/endocrine precursor induction. These δ cells display mature RNA profiles and fine secretory granules, secrete somatostatin in response to various stimuli, and suppress insulin secretion from in vitro co-cultured β cells and mouse β cells upon transplantation. The generation of human pancreatic δ cells from stem cells in vitro would provide an unprecedented cell source for drug discovery and cell transplantation studies in diabetes.
Collapse
Affiliation(s)
- Lihua Chen
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Nannan Wang
- Guangzhou National Laboratory, Guangzhou, Guangdong, China
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tongran Zhang
- Guangzhou National Laboratory, Guangzhou, Guangdong, China
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Feng Zhang
- Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Wei Zhang
- Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Hao Meng
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Jingyi Chen
- Guangzhou National Laboratory, Guangzhou, Guangdong, China
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, China
| | - Zhiying Liao
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Xiaopeng Xu
- Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Zhuo Ma
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Tao Xu
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong, China.
- Guangzhou National Laboratory, Guangzhou, Guangdong, China.
| | - Huisheng Liu
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong, China.
- Guangzhou National Laboratory, Guangzhou, Guangdong, China.
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, China.
| |
Collapse
|
20
|
Kane E, Mak TC, Latreille M. MicroRNA-7 regulates endocrine progenitor delamination and endocrine cell mass in developing pancreatic islets. iScience 2024; 27:110332. [PMID: 39055950 PMCID: PMC11269303 DOI: 10.1016/j.isci.2024.110332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/28/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
β-cell replenishment in patients with diabetes through cadaveric islet transplantation has been successful; however, it requires long-term immunosuppression and suitable islet donors are scarce. Stepwise in vitro differentiation of pluripotent stem cells into β-cells represents a viable alternative, but limitations in our current understanding of in vivo islet endocrine differentiation constrains its clinical use. Here, we show that microRNA-7 (miR-7) is highly expressed in embryonic pancreatic endocrine progenitors. Genetic deletion of the miR-7 gene family in endocrine progenitors leads to reduced islet endocrine cell mass, due to endocrine progenitors failing to delaminate from the epithelial plexus. This is associated with a reduction in neurogenin-3 levels and increased expression of Sry-box transcription factor 9. Further, we observe that a significant number of endocrine progenitors lacking miR-7 differentiate into ductal cells. Our study suggests that increasing miR-7 expression could improve efficiency of in vitro differentiation and augment stem cell-derived β-cell terminal maturity.
Collapse
Affiliation(s)
- Eva Kane
- MRC Laboratory of Medical Sciences, Du Cane Road, London W12 0NN, UK
| | - Tracy C.S. Mak
- MRC Laboratory of Medical Sciences, Du Cane Road, London W12 0NN, UK
| | - Mathieu Latreille
- MRC Laboratory of Medical Sciences, Du Cane Road, London W12 0NN, UK
| |
Collapse
|
21
|
Fuentes ME, Lu X, Flores NM, Hausmann S, Mazur PK. Combined deletion of MEN1, ATRX and PTEN triggers development of high-grade pancreatic neuroendocrine tumors in mice. Sci Rep 2024; 14:8510. [PMID: 38609433 PMCID: PMC11014914 DOI: 10.1038/s41598-024-58874-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
Pancreatic neuroendocrine tumors (PanNETs) are a heterogeneous group of tumors that exhibit an unpredictable and broad spectrum of clinical presentations and biological aggressiveness. Surgical resection is still the only curative therapeutic option for localized PanNET, but the majority of patients are diagnosed at an advanced and metastatic stage with limited therapeutic options. Key factors limiting the development of new therapeutics are the extensive heterogeneity of PanNETs and the lack of appropriate clinically relevant models. In that context, genomic sequencing of human PanNETs revealed recurrent mutations and structural alterations in several tumor suppressors. Here, we demonstrated that combined loss of MEN1, ATRX, and PTEN, tumor suppressors commonly mutated in human PanNETs, triggers the development of high-grade pancreatic neuroendocrine tumors in mice. Histopathological evaluation and gene expression analyses of the developed tumors confirm the presence of PanNET hallmarks and significant overlap in gene expression patterns found in human disease. Thus, we postulate that the presented novel genetically defined mouse model is the first clinically relevant immunocompetent high-grade PanNET mouse model.
Collapse
Affiliation(s)
- Mary Esmeralda Fuentes
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- The University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Xiaoyin Lu
- The University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Natasha M Flores
- The University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Simone Hausmann
- The University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Pawel K Mazur
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- The University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, TX, 77030, USA.
| |
Collapse
|
22
|
Liu S, Zhang Y, Luo Y, Liu J. Traditional and emerging strategies using hepatocytes for pancreatic regenerative medicine. J Diabetes 2024; 16:e13545. [PMID: 38599852 PMCID: PMC11006621 DOI: 10.1111/1753-0407.13545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 01/23/2024] [Accepted: 02/04/2024] [Indexed: 04/12/2024] Open
Abstract
Although pancreas and islet cell transplantation are the only ways to prevent the late complications of insulin-dependent diabetes, a shortage of donors is a major obstacle to tissue and organ transplantation. Stem cell therapy is an effective treatment for diabetes and other pancreatic-related diseases, which can be achieved by inducing their differentiation into insulin-secreting cells. The liver is considered an ideal source of pancreatic cells due to its similar developmental origin and strong regenerative ability as the pancreas. This article reviews the traditional and emerging strategies using hepatocytes for pancreatic regenerative medicine and evaluates their advantages and challenges. Gene reprogramming and chemical reprogramming technologies are traditional strategies with potential to improve the efficiency and specificity of cell reprogramming and promote the transformation of hepatocytes into islet cells. At the same time, organoid technology, as an emerging strategy, has received extensive attention. Biomaterials provide a three-dimensional culture microenvironment for cells, which helps improve cell survival and differentiation efficiency. In addition, clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 gene editing technology has brought new opportunities and challenges to the development of organoid technology.
Collapse
Affiliation(s)
- Shuang Liu
- Department of Metabolism and Endocrinology, the Second Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - YuYing Zhang
- Department of Metabolism and Endocrinology, the Second Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - YunFei Luo
- Department of Metabolism and Endocrinology, the Second Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - JianPing Liu
- Department of Metabolism and Endocrinology, the Second Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| |
Collapse
|
23
|
Ku B, Eisenbarth D, Baek S, Jeong TK, Kang JG, Hwang D, Noh MG, Choi C, Choi S, Seol T, Kim H, Kim YH, Woo SM, Kong SY, Lim DS. PRMT1 promotes pancreatic cancer development and resistance to chemotherapy. Cell Rep Med 2024; 5:101461. [PMID: 38460517 PMCID: PMC10983040 DOI: 10.1016/j.xcrm.2024.101461] [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: 02/28/2023] [Revised: 12/28/2023] [Accepted: 02/14/2024] [Indexed: 03/11/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal types of cancer, and novel treatment regimens are direly needed. Epigenetic regulation contributes to the development of various cancer types, but its role in the development of and potential as a therapeutic target for PDAC remains underexplored. Here, we show that PRMT1 is highly expressed in murine and human pancreatic cancer and is essential for cancer cell proliferation and tumorigenesis. Deletion of PRMT1 delays pancreatic cancer development in a KRAS-dependent mouse model, and multi-omics analyses reveal that PRMT1 depletion leads to global changes in chromatin accessibility and transcription, resulting in reduced glycolysis and a decrease in tumorigenic capacity. Pharmacological inhibition of PRMT1 in combination with gemcitabine has a synergistic effect on pancreatic tumor growth in vitro and in vivo. Collectively, our findings implicate PRMT1 as a key regulator of pancreatic cancer development and a promising target for combination therapy.
Collapse
Affiliation(s)
- Bomin Ku
- National Creative Research Center for Cell Plasticity, KAIST Stem Cell Center, Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - David Eisenbarth
- National Creative Research Center for Cell Plasticity, KAIST Stem Cell Center, Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea; Brown Center for Immunotherapy, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Seonguk Baek
- National Creative Research Center for Cell Plasticity, KAIST Stem Cell Center, Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Tae-Keun Jeong
- National Creative Research Center for Cell Plasticity, KAIST Stem Cell Center, Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Ju-Gyeong Kang
- National Creative Research Center for Cell Plasticity, KAIST Stem Cell Center, Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Daehee Hwang
- National Creative Research Center for Cell Plasticity, KAIST Stem Cell Center, Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Myung-Giun Noh
- Department of Pathology, Chonnam National University Medical School and Hwasun Hospital, Hwasun-gun, Jeonnam 58128, Republic of Korea
| | - Chan Choi
- Department of Pathology, Chonnam National University Medical School and Hwasun Hospital, Hwasun-gun, Jeonnam 58128, Republic of Korea
| | - Sungwoo Choi
- National Creative Research Center for Cell Plasticity, KAIST Stem Cell Center, Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Taejun Seol
- National Creative Research Center for Cell Plasticity, KAIST Stem Cell Center, Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Hail Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Yun-Hee Kim
- Research Institute, National Cancer Center, Goyang 10408, Republic of Korea
| | - Sang Myung Woo
- Research Institute, National Cancer Center, Goyang 10408, Republic of Korea
| | - Sun-Young Kong
- Targeted Therapy Branch, Division of Rare and Refractory Cancer, Research Institute, National Cancer Center, Goyang 10408, Republic of Korea
| | - Dae-Sik Lim
- National Creative Research Center for Cell Plasticity, KAIST Stem Cell Center, Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea.
| |
Collapse
|
24
|
Gu G, Brown M, Agan V, Nevills S, Hu R, Simmons A, Xu Y, Yang Y, Yagan M, Najam S, Dadi P, Sampson L, Magnuson M, Jacobson D, Lau K, Hodges E. Endocrine islet β-cell subtypes with differential function are derived from biochemically distinct embryonic endocrine islet progenitors that are regulated by maternal nutrients. RESEARCH SQUARE 2024:rs.3.rs-3946483. [PMID: 38496675 PMCID: PMC10942487 DOI: 10.21203/rs.3.rs-3946483/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Endocrine islet b cells comprise heterogenous cell subsets. Yet when/how these subsets are produced and how stable they are remain unknown. Addressing these questions is important for preventing/curing diabetes, because lower numbers of b cells with better secretory function is a high risk of this disease. Using combinatorial cell lineage tracing, scRNA-seq, and DNA methylation analysis, we show here that embryonic islet progenitors with distinct gene expression and DNA methylation produce b-cell subtypes of different function and viability in adult mice. The subtype with better function is enriched for genes involved in vesicular production/trafficking, stress response, and Ca2+-secretion coupling, which further correspond to differential DNA methylation in putative enhancers of these genes. Maternal overnutrition, a major diabetes risk factor, reduces the proportion of endocrine progenitors of the b-cell subtype with better-function via deregulating DNA methyl transferase 3a. Intriguingly, the gene signature that defines mouse b-cell subtypes can reliably divide human cells into two sub-populations while the proportion of b cells with better-function is reduced in diabetic donors. The implication of these results is that modulating DNA methylation in islet progenitors using maternal food supplements can be explored to improve b-cell function in the prevention and therapy of diabetes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yilin Yang
- Vanderbilty University School of Medicine
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Zheng C, Wang J, Wang J, Zhang Q, Liang T. Cell of Origin of Pancreatic cancer: Novel Findings and Current Understanding. Pancreas 2024; 53:e288-e297. [PMID: 38277420 PMCID: PMC11882172 DOI: 10.1097/mpa.0000000000002301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/08/2023] [Indexed: 01/28/2024]
Abstract
ABSTRACT Pancreatic ductal adenocarcinoma (PDAC) stands as one of the most lethal diseases globally, boasting a grim 5-year survival prognosis. The origin cell and the molecular signaling pathways that drive PDAC progression are not entirely understood. This review comprehensively outlines the categorization of PDAC and its precursor lesions, expounds on the creation and utility of genetically engineered mouse models used in PDAC research, compiles a roster of commonly used markers for pancreatic progenitors, duct cells, and acinar cells, and briefly addresses the mechanisms involved in the progression of PDAC. We acknowledge the value of precise markers and suitable tracing tools to discern the cell of origin, as it can facilitate the creation of more effective models for PDAC exploration. These conclusions shed light on our existing understanding of foundational genetically engineered mouse models and focus on the origin and development of PDAC.
Collapse
Affiliation(s)
- Chenlei Zheng
- From the Department of Hepatobiliary and Pancreatic Surgery
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine
| | - Jianing Wang
- From the Department of Hepatobiliary and Pancreatic Surgery
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine
| | - Junli Wang
- From the Department of Hepatobiliary and Pancreatic Surgery
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine
| | - Qi Zhang
- From the Department of Hepatobiliary and Pancreatic Surgery
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province
- Zhejiang University Cancer Center, Hangzhou, China
| | - Tingbo Liang
- From the Department of Hepatobiliary and Pancreatic Surgery
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province
- Zhejiang University Cancer Center, Hangzhou, China
| |
Collapse
|
26
|
Wang Y, Liu Z, Li S, Su X, Lai KP, Li R. Biochemical pancreatic β-cell lineage reprogramming: Various cell fate shifts. Curr Res Transl Med 2024; 72:103412. [PMID: 38246021 DOI: 10.1016/j.retram.2023.103412] [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: 10/22/2022] [Revised: 07/12/2023] [Accepted: 09/19/2023] [Indexed: 01/23/2024]
Abstract
The incidence of pancreatic diseases has been continuously rising in recent years. Thus, research on pancreatic regeneration is becoming more popular. Chronic hyperglycemia is detrimental to pancreatic β-cells, leading to impairment of insulin secretion which is the main hallmark of pancreatic diseases. Obtaining plenty of functional pancreatic β-cells is the most crucial aspect when studying pancreatic biology and treating diabetes. According to the International Diabetes Federation, diabetes has become a global epidemic, with about 3 million people suffering from diabetes worldwide. Hyperglycemia can lead to many dangerous diseases, including amputation, blindness, neuropathy, stroke, and cardiovascular and kidney diseases. Insulin is widely used in the treatment of diabetes; however, innovative approaches are needed in the academic and preclinical stages. A new approach aims at synthesizing patient-specific functional pancreatic β-cells. The present article focuses on how cells from different tissues can be transformed into pancreatic β-cells.
Collapse
Affiliation(s)
- Yuqin Wang
- Key Laboratory of Environmental Pollution and Integrative Omics, Education Department of Guangxi Zhuang Autonomous Region, Guilin Medical University, 1 Zhiyuan Road, Lingui District, Guilin 541199, China
| | - Zhuoqing Liu
- School of Pharmacy, Guilin Medical University, Guilin, China
| | - Shengren Li
- Lingui Clinical College of Guilin Medical University, Guilin, China
| | - Xuejuan Su
- Lingui Clinical College of Guilin Medical University, Guilin, China
| | - Keng Po Lai
- Key Laboratory of Environmental Pollution and Integrative Omics, Education Department of Guangxi Zhuang Autonomous Region, Guilin Medical University, 1 Zhiyuan Road, Lingui District, Guilin 541199, China
| | - Rong Li
- Key Laboratory of Environmental Pollution and Integrative Omics, Education Department of Guangxi Zhuang Autonomous Region, Guilin Medical University, 1 Zhiyuan Road, Lingui District, Guilin 541199, China.
| |
Collapse
|
27
|
Wortham M, Ramms B, Zeng C, Benthuysen JR, Sai S, Pollow DP, Liu F, Schlichting M, Harrington AR, Liu B, Prakash TP, Pirie EC, Zhu H, Baghdasarian S, Auwerx J, Shirihai OS, Sander M. Metabolic control of adaptive β-cell proliferation by the protein deacetylase SIRT2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.24.581864. [PMID: 38464227 PMCID: PMC10925077 DOI: 10.1101/2024.02.24.581864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Selective and controlled expansion of endogenous β-cells has been pursued as a potential therapy for diabetes. Ideally, such therapies would preserve feedback control of β-cell proliferation to avoid excessive β-cell expansion and an increased risk of hypoglycemia. Here, we identified a regulator of β-cell proliferation whose inactivation results in controlled β-cell expansion: the protein deacetylase Sirtuin 2 (SIRT2). Sirt2 deletion in β-cells of mice increased β-cell proliferation during hyperglycemia with little effect in homeostatic conditions, indicating preservation of feedback control of β-cell mass. SIRT2 restrains proliferation of human islet β-cells cultured in glucose concentrations above the glycemic set point, demonstrating conserved SIRT2 function. Analysis of acetylated proteins in islets treated with a SIRT2 inhibitor revealed that SIRT2 deacetylates enzymes involved in oxidative phosphorylation, dampening the adaptive increase in oxygen consumption during hyperglycemia. At the transcriptomic level, Sirt2 inactivation has context-dependent effects on β-cells, with Sirt2 controlling how β-cells interpret hyperglycemia as a stress. Finally, we provide proof-of-principle that systemic administration of a GLP1-coupled Sirt2-targeting antisense oligonucleotide achieves β-cell selective Sirt2 inactivation and stimulates β-cell proliferation under hyperglycemic conditions. Overall, these studies identify a therapeutic strategy for increasing β-cell mass in diabetes without circumventing feedback control of β-cell proliferation.
Collapse
Affiliation(s)
- Matthew Wortham
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
| | - Bastian Ramms
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
| | - Chun Zeng
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
| | - Jacqueline R Benthuysen
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
| | - Somesh Sai
- Institute of Chemistry and Biochemistry, Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Dennis P Pollow
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
| | - Fenfen Liu
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
| | - Michael Schlichting
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
| | - Austin R Harrington
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
| | - Bradley Liu
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
| | - Thazha P Prakash
- Department of Antisense Drug Discovery, Ionis Pharmaceuticals Inc., Carlsbad, CA, USA
| | - Elaine C Pirie
- Department of Antisense Drug Discovery, Ionis Pharmaceuticals Inc., Carlsbad, CA, USA
| | - Han Zhu
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
| | - Siyouneh Baghdasarian
- Departments of Medicine and Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Johan Auwerx
- Laboratory of Integrated Systems Physiology, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Orian S Shirihai
- Departments of Medicine and Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Maike Sander
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| |
Collapse
|
28
|
Sun J, Wang Y, Fu H, Kang F, Song J, Xu M, Ning G, Wang J, Wang W, Wang Q. Mettl3-Mediated m6A Methylation Controls Pancreatic Bipotent Progenitor Fate and Islet Formation. Diabetes 2024; 73:237-249. [PMID: 37963393 DOI: 10.2337/db23-0360] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 11/06/2023] [Indexed: 11/16/2023]
Abstract
The important role of m6A RNA modification in β-cell function has been established; however, how it regulates pancreatic development and endocrine differentiation remains unknown. Here, we generated transgenic mice lacking RNA methyltransferase-like 3 (Mettl3) specifically in Pdx1+ pancreatic progenitor cells and found the mice with the mutation developed hyperglycemia and hypoinsulinemia at age 2 weeks, along with an atrophic pancreas, reduced islet mass, and abnormal increase in ductal formation. At embryonic day 15.5, Mettl3 deletion had caused a significant loss of Ngn3+ endocrine progenitor cells, which was accompanied by increased Sox9+ ductal precursor cells. We identified histone deacetylase 1 (Hdac1) as the critical direct m6A target in bipotent progenitors, the degeneration of which caused abnormal activation of the Wnt/Notch signaling pathway and blocked endocrine differentiation. This transformation could be manipulated in embryonic pancreatic culture in vitro through regulation of the Mettl3-Hdac1-Wnt/Notch signaling axis. Our finding that Mettl3 determines endocrine lineage by modulating Hdac1 activity during the transition of bipotent progenitors might help in the development of targeted endocrine cell protocols for diabetes treatment. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Jiajun Sun
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the People's Republic of China, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanqiu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the People's Republic of China, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Fu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the People's Republic of China, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fuyun Kang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the People's Republic of China, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaxi Song
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the People's Republic of China, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the People's Republic of China, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the People's Republic of China, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Wang
- International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the People's Republic of China, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qidi Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the People's Republic of China, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
29
|
Puri S, Maachi H, Nair G, Russ HA, Chen R, Pulimeno P, Cutts Z, Ntranos V, Hebrok M. Sox9 regulates alternative splicing and pancreatic beta cell function. Nat Commun 2024; 15:588. [PMID: 38238288 PMCID: PMC10796970 DOI: 10.1038/s41467-023-44384-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 12/12/2023] [Indexed: 01/22/2024] Open
Abstract
Despite significant research, mechanisms underlying the failure of islet beta cells that result in type 2 diabetes (T2D) are still under investigation. Here, we report that Sox9, a transcriptional regulator of pancreas development, also functions in mature beta cells. Our results show that Sox9-depleted rodent beta cells have defective insulin secretion, and aging animals develop glucose intolerance, mimicking the progressive degeneration observed in T2D. Using genome editing in human stem cells, we show that beta cells lacking SOX9 have stunted first-phase insulin secretion. In human and rodent cells, loss of Sox9 disrupts alternative splicing and triggers accumulation of non-functional isoforms of genes with key roles in beta cell function. Sox9 depletion reduces expression of protein-coding splice variants of the serine-rich splicing factor arginine SRSF5, a major splicing enhancer that regulates alternative splicing. Our data highlight the role of SOX9 as a regulator of alternative splicing in mature beta cell function.
Collapse
Affiliation(s)
- Sapna Puri
- Diabetes Center, Department of Medicine, University of California, San Francisco, CA, USA
- Minutia Inc., Oakland, CA, USA
| | - Hasna Maachi
- Diabetes Center, Department of Medicine, University of California, San Francisco, CA, USA
- Center for Organoid Systems, Klinikum Rechts der Isar (MRI) and Technical University Munich, 85748, Garching, Germany
- Institute for Diabetes Organoid Technology, Helmholtz Munich, Helmholtz Diabetes Center, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Munich Institute of Biomedical Engineering (MIBE), Technical University Munich, Munich, Germany
- German Center for Diabetes Research (DZD), Ingolstaedter Landstrasse 1, 85764, Neuherberg, Germany
| | - Gopika Nair
- Diabetes Center, Department of Medicine, University of California, San Francisco, CA, USA
- Eli Lilly, Indianapolis, IN, USA
| | - Holger A Russ
- Diabetes Center, Department of Medicine, University of California, San Francisco, CA, USA
- Diabetes Institute, University of Florida, Gainesville, FL, USA
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Richard Chen
- Diabetes Center, Department of Medicine, University of California, San Francisco, CA, USA
| | - Pamela Pulimeno
- Diabetes Center, Department of Medicine, University of California, San Francisco, CA, USA
| | - Zachary Cutts
- Graduate Program in Bioinformatics, University of California, San Francisco, CA, USA
| | - Vasilis Ntranos
- Diabetes Center, Department of Medicine, University of California, San Francisco, CA, USA
| | - Matthias Hebrok
- Diabetes Center, Department of Medicine, University of California, San Francisco, CA, USA.
- Center for Organoid Systems, Klinikum Rechts der Isar (MRI) and Technical University Munich, 85748, Garching, Germany.
- Institute for Diabetes Organoid Technology, Helmholtz Munich, Helmholtz Diabetes Center, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.
- Munich Institute of Biomedical Engineering (MIBE), Technical University Munich, Munich, Germany.
- German Center for Diabetes Research (DZD), Ingolstaedter Landstrasse 1, 85764, Neuherberg, Germany.
| |
Collapse
|
30
|
Sue N, Thai LM, Saito A, Boyer CK, Fordham AM, Yan C, Davenport A, Tao J, Bensellam M, Cantley J, Shi YC, Stephens SB, Imaizumi K, Biden TJ. Independent activation of CREB3L2 by glucose fills a regulatory gap in mouse β-cells by co-ordinating insulin biosynthesis with secretory granule formation. Mol Metab 2024; 79:101845. [PMID: 38013154 PMCID: PMC10755490 DOI: 10.1016/j.molmet.2023.101845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 11/29/2023] Open
Abstract
OBJECTIVE Although individual steps have been characterized, there is little understanding of the overall process whereby glucose co-ordinates the biosynthesis of insulin with its export out of the endoplasmic reticulum (ER) and incorporation into insulin secretory granules (ISGs). Here we investigate a role for the transcription factor CREB3L2 in this context. METHODS MIN6 cells and mouse islets were analysed by immunoblotting after treatment with glucose, fatty acids, thapsigargin and various inhibitors. Knockdown of CREB3L2 was achieved using si or sh constructs by transfection, or viral delivery. In vivo metabolic phenotyping was conducted after deletion of CREB3L2 in β-cells of adult mice using Ins1-CreER+. Islets were isolated for RNAseq and assays of glucose-stimulated insulin secretion (GSIS). Trafficking was monitored in islet monolayers using a GFP-tagged proinsulin construct that allows for synchronised release from the ER. RESULTS With a Km ≈3.5 mM, glucose rapidly (T1/2 0.9 h) increased full length (FL) CREB3L2 followed by a slower rise (T1/2 2.5 h) in its transcriptionally-active cleavage product, P60 CREB3L2. Glucose stimulation repressed the ER stress marker, CHOP, and this was partially reverted by knockdown of CREB3L2. Activation of CREB3L2 by glucose was not due to ER stress, however, but a combination of O-GlcNAcylation, which impaired proteasomal degradation of FL-CREB3L2, and mTORC1 stimulation, which enhanced its conversion to P60. cAMP generation also activated CREB3L2, but independently of glucose. Deletion of CREB3L2 inhibited GSIS ex vivo and, following a high-fat diet (HFD), impaired glucose tolerance and insulin secretion in vivo. RNAseq revealed that CREB3L2 regulated genes controlling trafficking to-and-from the Golgi, as well as a broader cohort associated with β-cell compensation during a HFD. Although post-Golgi trafficking appeared intact, knockdown of CREB3L2 impaired the generation of both nascent ISGs and proinsulin condensates in the Golgi, implying a defect in ER export of proinsulin and/or its processing in the Golgi. CONCLUSION The stimulation of CREB3L2 by glucose defines a novel, rapid and direct mechanism for co-ordinating the synthesis, packaging and storage of insulin, thereby minimizing ER overload and optimizing β-cell function under conditions of high secretory demand. Upregulation of CREB3L2 also potentially contributes to the benefits of GLP1 agonism and might in itself constitute a novel means of treating β-cell failure.
Collapse
Affiliation(s)
- Nancy Sue
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW 2010, Australia
| | - Le May Thai
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW 2010, Australia
| | - Atsushi Saito
- Department of Biochemistry, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Cierra K Boyer
- Fraternal Order of Eagles Diabetes Research Center, Department of Internal Medicine, University of Iowa, Iowa City, IA 52246, USA
| | - Ashleigh M Fordham
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW 2010, Australia
| | - Chenxu Yan
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW 2010, Australia
| | - Aimee Davenport
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW 2010, Australia
| | - Jiang Tao
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW 2010, Australia
| | - Mohammed Bensellam
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW 2010, Australia
| | - James Cantley
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW 2010, Australia
| | - Yan-Chuan Shi
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, Faculty of Medicine, The University of New South Wales, Sydney, Australia
| | - Samuel B Stephens
- Fraternal Order of Eagles Diabetes Research Center, Department of Internal Medicine, University of Iowa, Iowa City, IA 52246, USA
| | - Kazunori Imaizumi
- Department of Biochemistry, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Trevor J Biden
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, Faculty of Medicine, The University of New South Wales, Sydney, Australia.
| |
Collapse
|
31
|
Himuro M, Wakabayashi Y, Taguchi T, Katahira T, Suzuki L, Iida H, Ogihara T, Nishida Y, Sasaki S, Lynn FC, Hiraoka Y, Oshima S, Okamoto R, Fujitani Y, Watada H, Miyatsuka T. Novel time-resolved reporter mouse reveals spatial and transcriptional heterogeneity during alpha cell differentiation. Diabetologia 2024; 67:156-169. [PMID: 37870650 DOI: 10.1007/s00125-023-06028-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 08/25/2023] [Indexed: 10/24/2023]
Abstract
AIMS/HYPOTHESIS Glucagon-expressing pancreatic alpha cells have attracted much attention for their plasticity to transdifferentiate into insulin-producing beta cells; however, it remains unclear precisely when, and from where, alpha cells emerge and what regulates alpha cell fate. We therefore explored the spatial and transcriptional heterogeneity of alpha cell differentiation using a novel time-resolved reporter system. METHODS We established the mouse model, 'Gcg-Timer', in which newly generated alpha cells can be distinguished from more-differentiated cells by their fluorescence. Fluorescence imaging and transcriptome analysis were performed with Gcg-Timer mice during the embryonic and postnatal stages. RESULTS Fluorescence imaging and flow cytometry demonstrated that green fluorescence-dominant cells were present in Gcg-Timer mice at the embryonic and neonatal stages but not after 1 week of age, suggesting that alpha cell neogenesis occurs during embryogenesis and early neonatal stages under physiological conditions. Transcriptome analysis of Gcg-Timer embryos revealed that the mRNAs related to angiogenesis were enriched in newly generated alpha cells. Histological analysis revealed that some alpha cells arise close to the pancreatic ducts, whereas the others arise away from the ducts and adjacent to the blood vessels. Notably, when the glucagon signal was suppressed by genetic ablation or by chemicals, such as neutralising glucagon antibody, green-dominant cells emerged again in adult mice. CONCLUSIONS/INTERPRETATION Novel time-resolved analysis with Gcg-Timer reporter mice uncovered spatiotemporal features of alpha cell neogenesis that will enhance our understanding of cellular identity and plasticity within the islets. DATA AVAILABILITY Raw and processed RNA sequencing data for this study has been deposited in the Gene Expression Omnibus under accession number GSE229090.
Collapse
Affiliation(s)
- Miwa Himuro
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuka Wakabayashi
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tomomi Taguchi
- Department of Endocrinology, Diabetes and Metabolism, Kitasato University School of Medicine, Sagamihara, Japan
| | - Takehiro Katahira
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Luka Suzuki
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hitoshi Iida
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takeshi Ogihara
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuya Nishida
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shugo Sasaki
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, BC, Canada
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Francis C Lynn
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Yuichi Hiraoka
- Laboratory of Genome Editing for Biomedical Research, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shigeru Oshima
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ryuichi Okamoto
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshio Fujitani
- Laboratory of Developmental Biology & Metabolism, Institute for Molecular & Cellular Regulation, Gunma University, Maebashi, Japan
| | - Hirotaka Watada
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan.
- Center for Identification of Diabetic Therapeutic Targets, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| | - Takeshi Miyatsuka
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan.
- Department of Endocrinology, Diabetes and Metabolism, Kitasato University School of Medicine, Sagamihara, Japan.
| |
Collapse
|
32
|
Cota P, Caliskan ÖS, Bastidas-Ponce A, Jing C, Jaki J, Saber L, Czarnecki O, Taskin D, Blöchinger AK, Kurth T, Sterr M, Burtscher I, Krahmer N, Lickert H, Bakhti M. Insulin regulates human pancreatic endocrine cell differentiation in vitro. Mol Metab 2024; 79:101853. [PMID: 38103636 PMCID: PMC10765254 DOI: 10.1016/j.molmet.2023.101853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/21/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023] Open
Abstract
OBJECTIVE The consequences of mutations in genes associated with monogenic forms of diabetes on human pancreas development cannot be studied in a time-resolved fashion in vivo. More specifically, if recessive mutations in the insulin gene influence human pancreatic endocrine lineage formation is still an unresolved question. METHODS To model the extremely reduced insulin levels in patients with recessive insulin gene mutations, we generated a novel knock-in H2B-Cherry reporter human induced pluripotent stem cell (iPSC) line expressing no insulin upon differentiation to stem cell-derived (SC-) β cells in vitro. Differentiation of iPSCs into the pancreatic and endocrine lineage, combined with immunostaining, Western blotting and proteomics analysis phenotypically characterized the insulin gene deficiency in SC-islets. Furthermore, we leveraged FACS analysis and confocal microscopy to explore the impact of insulin shortage on human endocrine cell induction, composition, differentiation and proliferation. RESULTS Interestingly, insulin-deficient SC-islets exhibited low insulin receptor (IR) signaling when stimulated with glucose but displayed increased IR sensitivity upon treatment with exogenous insulin. Furthermore, insulin shortage did not alter neurogenin-3 (NGN3)-mediated endocrine lineage induction. Nevertheless, lack of insulin skewed the SC-islet cell composition with an increased number in SC-β cell formation at the expense of SC-α cells. Finally, insulin deficiency reduced the rate of SC-β cell proliferation but had no impact on the expansion of SC-α cells. CONCLUSIONS Using iPSC disease modelling, we provide first evidence of insulin function in human pancreatic endocrine lineage formation. These findings help to better understand the phenotypic impact of recessive insulin gene mutations during pancreas development and shed light on insulin gene function beside its physiological role in blood glucose regulation.
Collapse
Affiliation(s)
- Perla Cota
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Özüm Sehnaz Caliskan
- German Center for Diabetes Research (DZD), Neuherberg, Germany; Institute of Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
| | - Aimée Bastidas-Ponce
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Changying Jing
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Munich medical research school (MMRS), Ludwig Maximilian University (LMU), Munich, Germany
| | - Jessica Jaki
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Lama Saber
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Oliver Czarnecki
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Damla Taskin
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany
| | - Anna Karolina Blöchinger
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Thomas Kurth
- Center for Molecular and Cellular Bioengineering (CMCB), Technology Platform Core Facility Electron Microscopy and Histology, Technische Universität Dresden, Dresden, Germany
| | - Michael Sterr
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Ingo Burtscher
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Natalie Krahmer
- German Center for Diabetes Research (DZD), Neuherberg, Germany; Institute of Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; School of Medicine, Technical University of Munich (TUM), Munich, Germany.
| | - Mostafa Bakhti
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany.
| |
Collapse
|
33
|
Bora J, Dey A, Lyngdoh AR, Dhasmana A, Ranjan A, Kishore S, Rustagi S, Tuli HS, Chauhan A, Rath P, Malik S. A critical review on therapeutic approaches of CRISPR-Cas9 in diabetes mellitus. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:3459-3481. [PMID: 37522916 DOI: 10.1007/s00210-023-02631-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 07/14/2023] [Indexed: 08/01/2023]
Abstract
Diabetes mellitus (D.M.) is a common metabolic disorder caused mainly by combining two primary factors, which are (1) defects in insulin production by the pancreatic β-cells and (2) responsiveness of insulin-sensitive tissues towards insulin. Despite the rapid advancement in medicine to suppress elevated blood glucose levels (hyperglycemia) and insulin resistance associated with this hazard, a demand has undoubtedly emerged to find more effective and curative dimensions in therapeutic approaches against D.M. The administration of diabetes treatment that emphasizes insulin production and sensitivity may result in unfavorable side effects, reduced adherence, and potential treatment ineffectiveness. Recent progressions in genome editing technologies, for instance, in zinc-finger nucleases, transcription activator-like effector nucleases, and clustered regularly interspaced short palindromic repeat (CRISPR-Cas)-associated nucleases, have greatly influenced the gene editing technology from concepts to clinical practices. Improvements in genome editing technologies have also opened up the possibility to target and modify specific genome sequences in a cell directly. CRISPR/Cas9 has proven effective in utilizing ex vivo gene editing in embryonic stem cells and stem cells derived from patients. This application has facilitated the exploration of pancreatic beta-cell development and function. Furthermore, CRISPR/Cas9 enables the creation of innovative animal models for diabetes and assesses the effectiveness of different therapeutic strategies in treating the condition. We, therefore, present a critical review of the therapeutic approaches of the genome editing tool CRISPR-Cas9 in treating D.M., discussing the challenges and limitations of implementing this technology.
Collapse
Affiliation(s)
- Jutishna Bora
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, 834001, India
| | - Ankita Dey
- Department of Biochemistry, North Eastern Hill University, Shillong, Meghalaya, 793022, India
| | - Antonia R Lyngdoh
- Department of Biochemistry, North Eastern Hill University, Shillong, Meghalaya, 793022, India
| | - Archna Dhasmana
- Himalayan School of Biosciences, Swami Rama Himalayan University, Jolly Grant, Dehradun, Uttarakhand, India
| | - Anuj Ranjan
- Academy of Biology and Biotechnology, Southern Federal University, Stachki 194/1, Rostov-On-Don, 344090, Russia
| | - Shristi Kishore
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, 834001, India
| | - Sarvesh Rustagi
- School of Applied and Life Sciences, Uttaranchal University, 22 Dehradun, Uttarakhand, India
| | - Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana-Ambala, 133207, India
| | - Abhishek Chauhan
- Amity Institute of Environmental Toxicology Safety and Management, Amity University, Sector 125, Noida, Uttar Pradesh, India
| | - Prangya Rath
- Amity Institute of Environmental Sciences, Amity University, Noida, Uttar Pradesh, 201303, India
| | - Sumira Malik
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, 834001, India.
- School of Applied and Life Sciences, Uttaranchal University, 22 Dehradun, Uttarakhand, India.
- Guru Nanak College of Pharmaceutical Sciences, Dehradun, Uttarakhand, India.
| |
Collapse
|
34
|
Hu C, Huang C, Hsu M, Chien H, Wu P, Chen Y, Jeng Y, Tang S, Chung M, Shen C, Chang M, Chang Y, Tien Y, Lee W. Oncogenic KRAS, Mucin 4, and Activin A-Mediated Fibroblast Activation Cooperate for PanIN Initiation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301240. [PMID: 37964407 PMCID: PMC10754145 DOI: 10.1002/advs.202301240] [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: 02/23/2023] [Revised: 08/22/2023] [Indexed: 11/16/2023]
Abstract
Over 90% of patients with pancreatic ductal adenocarcinoma (PDAC) have oncogenic KRAS mutations. Nevertheless, mutated KRAS alone is insufficient to initiate pancreatic intraepithelial neoplasia (PanIN), the precursor of PDAC. The identities of the other factors/events required to drive PanIN formation remain elusive. Here, optic-clear 3D histology is used to analyze entire pancreases of 2-week-old Pdx1-Cre; LSL-KrasG12D/+ (KC) mice to detect the earliest emergence of PanIN and observed that the occurrence is independent of physical location. Instead, it is found that the earliest PanINs overexpress Muc4 and associate with αSMA+ fibroblasts in both transgenic mice and human specimens. Mechanistically, KrasG12D/+ pancreatic cells upregulate Muc4 through genetic alterations to increase proliferation and fibroblast recruitments via Activin A secretion and consequently enhance cell transformation for PanIN formation. Inhibition of Activin A signaling using Follistatin (FST) diminishes early PanIN-associated fibroblast recruitment, effectively curtailing PanIN initiation and growth in KC mice. These findings emphasize the vital role of interactions between oncogenic KrasG12D/+ -driven genetic alterations and induced microenvironmental changes in PanIN initiation, suggesting potential avenues for early PDAC diagnostic and management approaches.
Collapse
Affiliation(s)
- Chun‐Mei Hu
- Genomics Research CenterAcademia SinicaTaipei11529Taiwan
| | - Chien‐Chang Huang
- Genomics Research CenterAcademia SinicaTaipei11529Taiwan
- Biomedical Translation Research CenterAcademia SinicaTaipei11529Taiwan
| | - Min‐Fen Hsu
- Genomics Research CenterAcademia SinicaTaipei11529Taiwan
| | - Hung‐Jen Chien
- Genomics Research CenterAcademia SinicaTaipei11529Taiwan
| | - Pei‐Jung Wu
- Genomics Research CenterAcademia SinicaTaipei11529Taiwan
| | - Yi‐Ing Chen
- Genomics Research CenterAcademia SinicaTaipei11529Taiwan
| | - Yung‐Ming Jeng
- Department of PathologyNational Taiwan University HospitalTaipei10041Taiwan
- Graduate Institute of Pathology, College of MedicineNational Taiwan UniversityTaipei10041Taiwan
| | - Shiue‐Cheng Tang
- Department of Medical ScienceNational Tsing Hua UniversityHsinchu30013Taiwan
| | - Mei‐Hsin Chung
- Department of PathologyNational Taiwan University Hospital−Hsinchu BranchHsinchu30331Taiwan
| | - Chia‐Ning Shen
- Genomics Research CenterAcademia SinicaTaipei11529Taiwan
- Biomedical Translation Research CenterAcademia SinicaTaipei11529Taiwan
| | - Ming‐Chu Chang
- Department of Internal MedicineNational Taiwan University HospitalTaipei10041Taiwan
| | - Yu‐Ting Chang
- Department of Internal MedicineNational Taiwan University HospitalTaipei10041Taiwan
| | - Yu‐Wen Tien
- Department of SurgeryNational Taiwan University HospitalTaipei10041Taiwan
| | - Wen‐Hwa Lee
- Genomics Research CenterAcademia SinicaTaipei11529Taiwan
- Drug Development CenterChina Medical UniversityTaichung40402Taiwan
- Department of Biological ChemistryUniversity of CaliforniaIrvineCA92697USA
| |
Collapse
|
35
|
Yang ZZ, Parchem RJ. The role of noncoding RNAs in pancreatic birth defects. Birth Defects Res 2023; 115:1785-1808. [PMID: 37066622 PMCID: PMC10579456 DOI: 10.1002/bdr2.2178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/19/2023] [Accepted: 04/03/2023] [Indexed: 04/18/2023]
Abstract
Congenital defects in the pancreas can cause severe health issues such as pancreatic cancer and diabetes which require lifelong treatment. Regenerating healthy pancreatic cells to replace malfunctioning cells has been considered a promising cure for pancreatic diseases including birth defects. However, such therapies are currently unavailable in the clinic. The developmental gene regulatory network underlying pancreatic development must be reactivated for in vivo regeneration and recapitulated in vitro for cell replacement therapy. Thus, understanding the mechanisms driving pancreatic development will pave the way for regenerative therapies. Pancreatic progenitor cells are the precursors of all pancreatic cells which use epigenetic changes to control gene expression during differentiation to generate all of the distinct pancreatic cell types. Epigenetic changes involving DNA methylation and histone modifications can be controlled by noncoding RNAs (ncRNAs). Indeed, increasing evidence suggests that ncRNAs are indispensable for proper organogenesis. Here, we summarize recent insight into the role of ncRNAs in the epigenetic regulation of pancreatic development. We further discuss how disruptions in ncRNA biogenesis and expression lead to developmental defects and diseases. This review summarizes in vivo data from animal models and in vitro studies using stem cell differentiation as a model for pancreatic development.
Collapse
Affiliation(s)
- Ziyue Zoey Yang
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, Texas, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Ronald J Parchem
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, Texas, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
36
|
Beydag-Tasöz BS, D'Costa JV, Hersemann L, Lee BH, Luppino F, Kim YH, Zechner C, Grapin-Botton A. Integrating single-cell imaging and RNA sequencing datasets links differentiation and morphogenetic dynamics of human pancreatic endocrine progenitors. Dev Cell 2023; 58:2292-2308.e6. [PMID: 37591246 DOI: 10.1016/j.devcel.2023.07.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 05/20/2023] [Accepted: 07/27/2023] [Indexed: 08/19/2023]
Abstract
Basic helix-loop-helix genes, particularly proneural genes, are well-described triggers of cell differentiation, yet information on their dynamics is limited, notably in human development. Here, we focus on Neurogenin 3 (NEUROG3), which is crucial for pancreatic endocrine lineage initiation. By monitoring both NEUROG3 gene expression and protein in single cells using a knockin dual reporter in 2D and 3D models of human pancreas development, we show an approximately 2-fold slower expression of human NEUROG3 than that of the mouse. We observe heterogeneous peak levels of NEUROG3 expression and reveal through long-term live imaging that both low and high NEUROG3 peak levels can trigger differentiation into hormone-expressing cells. Based on fluorescence intensity, we statistically integrate single-cell transcriptome with dynamic behaviors of live cells and propose a data-mapping methodology applicable to other contexts. Using this methodology, we identify a role for KLK12 in motility at the onset of NEUROG3 expression.
Collapse
Affiliation(s)
- Belin Selcen Beydag-Tasöz
- The Novo Nordisk Foundation Center for Stem Cell Biology, Copenhagen 2200, Denmark; Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Saxony 01307, Germany
| | - Joyson Verner D'Costa
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Saxony 01307, Germany
| | - Lena Hersemann
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Saxony 01307, Germany
| | - Byung Ho Lee
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Saxony 01307, Germany
| | - Federica Luppino
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Saxony 01307, Germany; Center for Systems Biology Dresden Dresden 01307, Germany
| | - Yung Hae Kim
- The Novo Nordisk Foundation Center for Stem Cell Biology, Copenhagen 2200, Denmark; Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Saxony 01307, Germany
| | - Christoph Zechner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Saxony 01307, Germany; Center for Systems Biology Dresden Dresden 01307, Germany; Cluster of Excellence Physics of Life, TU Dresden, Dresden 01062, Germany
| | - Anne Grapin-Botton
- The Novo Nordisk Foundation Center for Stem Cell Biology, Copenhagen 2200, Denmark; Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Saxony 01307, Germany; Center for Systems Biology Dresden Dresden 01307, Germany; Cluster of Excellence Physics of Life, TU Dresden, Dresden 01062, Germany; Paul Langerhans Institute Dresden of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany.
| |
Collapse
|
37
|
Zhang J, Katada K, Mosleh E, Yuhas A, Peng G, Golson ML. The leptin receptor has no role in delta-cell control of beta-cell function in the mouse. Front Endocrinol (Lausanne) 2023; 14:1257671. [PMID: 37850099 PMCID: PMC10577419 DOI: 10.3389/fendo.2023.1257671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 09/05/2023] [Indexed: 10/19/2023] Open
Abstract
Introduction Leptin inhibits insulin secretion from isolated islets from multiple species, but the cell type that mediates this process remains elusive. Several mouse models have been used to explore this question. Ablation of the leptin receptor (Lepr) throughout the pancreatic epithelium results in altered glucose homeostasis and ex vivo insulin secretion and Ca2+ dynamics. However, Lepr removal from neither alpha nor beta cells mimics this result. Moreover, scRNAseq data has revealed an enrichment of LEPR in human islet delta cells. Methods We confirmed LEPR upregulation in human delta cells by performing RNAseq on fixed, sorted beta and delta cells. We then used a mouse model to test whether delta cells mediate the diminished glucose-stimulated insulin secretion in response to leptin. Results Ablation of Lepr within mouse delta cells did not change glucose homeostasis or insulin secretion, whether mice were fed a chow or high-fat diet. We further show, using a publicly available scRNAseq dataset, that islet cells expressing Lepr lie within endothelial cell clusters. Conclusions In mice, leptin does not influence beta-cell function through delta cells.
Collapse
Affiliation(s)
- Jia Zhang
- Department of Genetics, University of Pennsylvania, Philadephia, PA, United States
| | - Kay Katada
- School of Medicine, University of Pennsylvania, Philadephia, PA, United States
| | - Elham Mosleh
- Department of Genetics, University of Pennsylvania, Philadephia, PA, United States
- School of Medicine, University of Pennsylvania, Philadephia, PA, United States
| | - Andrew Yuhas
- Department of Genetics, University of Pennsylvania, Philadephia, PA, United States
- School of Medicine, University of Pennsylvania, Philadephia, PA, United States
| | - Guihong Peng
- Department of Medicine, Divison of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University, Baltimore, MD, United States
| | - Maria L. Golson
- Department of Genetics, University of Pennsylvania, Philadephia, PA, United States
- School of Medicine, University of Pennsylvania, Philadephia, PA, United States
- Department of Medicine, Divison of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
38
|
Bohuslavova R, Fabriciova V, Smolik O, Lebrón-Mora L, Abaffy P, Benesova S, Zucha D, Valihrach L, Berkova Z, Saudek F, Pavlinkova G. NEUROD1 reinforces endocrine cell fate acquisition in pancreatic development. Nat Commun 2023; 14:5554. [PMID: 37689751 PMCID: PMC10492842 DOI: 10.1038/s41467-023-41306-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 08/30/2023] [Indexed: 09/11/2023] Open
Abstract
NEUROD1 is a transcription factor that helps maintain a mature phenotype of pancreatic β cells. Disruption of Neurod1 during pancreatic development causes severe neonatal diabetes; however, the exact role of NEUROD1 in the differentiation programs of endocrine cells is unknown. Here, we report a crucial role of the NEUROD1 regulatory network in endocrine lineage commitment and differentiation. Mechanistically, transcriptome and chromatin landscape analyses demonstrate that Neurod1 inactivation triggers a downregulation of endocrine differentiation transcription factors and upregulation of non-endocrine genes within the Neurod1-deficient endocrine cell population, disturbing endocrine identity acquisition. Neurod1 deficiency altered the H3K27me3 histone modification pattern in promoter regions of differentially expressed genes, which resulted in gene regulatory network changes in the differentiation pathway of endocrine cells, compromising endocrine cell potential, differentiation, and functional properties.
Collapse
Affiliation(s)
- Romana Bohuslavova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Valeria Fabriciova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Ondrej Smolik
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Laura Lebrón-Mora
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Pavel Abaffy
- Laboratory of Gene Expression, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Sarka Benesova
- Laboratory of Gene Expression, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Daniel Zucha
- Laboratory of Gene Expression, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Lukas Valihrach
- Laboratory of Gene Expression, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Zuzana Berkova
- Diabetes Centre, Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, 14021, Prague, Czechia
| | - Frantisek Saudek
- Diabetes Centre, Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, 14021, Prague, Czechia
| | - Gabriela Pavlinkova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology CAS, 25250, Vestec, Czechia.
| |
Collapse
|
39
|
Nakamura T, Nishikawa Y, Shiokawa M, Takeda H, Yokode M, Matsumoto S, Muramoto Y, Ota S, Yoshida H, Okada H, Kuwada T, Marui S, Matsumori T, Maruno T, Uza N, Kodama Y, Hatano E, Seno H. ELF3 suppresses gallbladder cancer development through downregulation of the EREG/EGFR/mTOR complex 1 signalling pathway. J Pathol 2023; 261:28-42. [PMID: 37345534 DOI: 10.1002/path.6144] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 05/01/2023] [Accepted: 05/16/2023] [Indexed: 06/23/2023]
Abstract
The prognosis of gallbladder cancer (GBC) remains poor, and a better understanding of GBC molecular mechanisms is important. Genome sequencing of human GBC has demonstrated that loss-of-function mutations of E74-like ETS transcription factor 3 (ELF3) are frequently observed, with ELF3 considered to be a tumour suppressor in GBC. To clarify the underlying molecular mechanisms by which ELF3 suppresses GBC development, we performed in vivo analysis using a combination of autochthonous and allograft mouse models. We first evaluated the clinical significance of ELF3 expression in human GBC tissues and found that low ELF3 expression was associated with advanced clinical stage and deep tumour invasion. For in vivo analysis, we generated Pdx1-Cre; KrasG12D ; Trp53R172H ; Elf3f/f (KPCE) mice and Pdx1-Cre; KrasG12D ; Trp53R172H ; Elf3wt/wt (KPC) mice as a control and analysed their gallbladders histologically. KPCE mice developed larger papillary lesions in the gallbladder than those developed by KPC mice. Organoids established from the gallbladders of KPCE and KPC mice were analysed in vitro. RNA sequencing showed upregulated expression of epiregulin (Ereg) in KPCE organoids, and western blotting revealed that EGFR/mechanical targets of rapamycin complex 1 (mTORC1) were upregulated in KPCE organoids. In addition, ChIP assays on Elf3-overexpressing KPCE organoids showed that ELF3 directly regulated Ereg. Ereg deletion in KPCE organoids (using CRISPR/Cas9) induced EGFR/mTORC1 downregulation, indicating that ELF3 controlled EGFR/mTORC1 activity through regulation of Ereg expression. We also generated allograft mouse models using KPCE and KPC organoids and found that KPCE organoid allograft tumours exhibited poorly differentiated structures with mTORC1 upregulation and mesenchymal phenotype, which were suppressed by Ereg deletion. Furthermore, EGFR/mTORC1 inhibition suppressed cell proliferation and epithelial-mesenchymal transition in KPCE organoids. Our results suggest that ELF3 suppresses GBC development via downregulation of EREG/EGFR/mTORC1 signalling. EGFR/mTORC1 inhibition is a potential therapeutic option for GBC with ELF3 mutation. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Takeharu Nakamura
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yoshihiro Nishikawa
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Gastroenterology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masahiro Shiokawa
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Haruhiko Takeda
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masataka Yokode
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shimpei Matsumoto
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuya Muramoto
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Sakiko Ota
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroyuki Yoshida
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hirokazu Okada
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takeshi Kuwada
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Saiko Marui
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tomoaki Matsumori
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takahisa Maruno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Norimitsu Uza
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuzo Kodama
- Department of Gastroenterology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Etsuro Hatano
- Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
40
|
Tixi W, Maldonado M, Chang YT, Chiu A, Yeung W, Parveen N, Nelson MS, Hart R, Wang S, Hsu WJ, Fueger P, Kopp JL, Huising MO, Dhawan S, Shih HP. Coordination between ECM and cell-cell adhesion regulates the development of islet aggregation, architecture, and functional maturation. eLife 2023; 12:e90006. [PMID: 37610090 PMCID: PMC10482429 DOI: 10.7554/elife.90006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/12/2023] [Indexed: 08/24/2023] Open
Abstract
Pancreatic islets are three-dimensional cell aggregates consisting of unique cellular composition, cell-to-cell contacts, and interactions with blood vessels. Cell aggregation is essential for islet endocrine function; however, it remains unclear how developing islets establish aggregation. By combining genetic animal models, imaging tools, and gene expression profiling, we demonstrate that islet aggregation is regulated by extracellular matrix signaling and cell-cell adhesion. Islet endocrine cell-specific inactivation of extracellular matrix receptor integrin β1 disrupted blood vessel interactions but promoted cell-cell adhesion and the formation of larger islets. In contrast, ablation of cell-cell adhesion molecule α-catenin promoted blood vessel interactions yet compromised islet clustering. Simultaneous removal of integrin β1 and α-catenin disrupts islet aggregation and the endocrine cell maturation process, demonstrating that establishment of islet aggregates is essential for functional maturation. Our study provides new insights into understanding the fundamental self-organizing mechanism for islet aggregation, architecture, and functional maturation.
Collapse
Affiliation(s)
- Wilma Tixi
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of HopeDuarteUnited States
| | - Maricela Maldonado
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of HopeDuarteUnited States
- Department of Biomedical Engineering, College of Engineering, California State University, Long BeachLong BeachUnited States
| | - Ya-Ting Chang
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of HopeDuarteUnited States
| | - Amy Chiu
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of HopeDuarteUnited States
| | - Wilson Yeung
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of HopeDuarteUnited States
| | - Nazia Parveen
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of HopeDuarteUnited States
| | - Michael S Nelson
- Light Microscopy Core, Beckman Research Institute, City of HopeDuarteUnited States
| | - Ryan Hart
- Department of Neurobiology, Physiology and Behavior, University of California, DavisDavisUnited States
| | - Shihao Wang
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British ColumbiaVancouverCanada
| | - Wu Jih Hsu
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British ColumbiaVancouverCanada
| | - Patrick Fueger
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of HopeDuarteUnited States
| | - Janel L Kopp
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British ColumbiaVancouverCanada
| | - Mark O Huising
- Department of Neurobiology, Physiology and Behavior, University of California, DavisDavisUnited States
- Department of Physiology and Membrane Biology, School of Medicine, University of California, DavisDavisUnited States
| | - Sangeeta Dhawan
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of HopeDuarteUnited States
| | - Hung Ping Shih
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of HopeDuarteUnited States
| |
Collapse
|
41
|
Sasaki S, Nian C, Xu EE, Pasula DJ, Winata H, Grover S, Luciani DS, Lynn FC. Type 2 diabetes susceptibility gene GRK5 regulates physiological pancreatic β-cell proliferation via phosphorylation of HDAC5. iScience 2023; 26:107311. [PMID: 37520700 PMCID: PMC10382860 DOI: 10.1016/j.isci.2023.107311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/24/2023] [Accepted: 07/04/2023] [Indexed: 08/01/2023] Open
Abstract
Restoring functional β cell mass is a potential therapy for those with diabetes. However, the pathways regulating β cell mass are not fully understood. Previously, we demonstrated that Sox4 is required for β cell proliferation during prediabetes. Here, we report that Sox4 regulates β cell mass through modulating expression of the type 2 diabetes (T2D) susceptibility gene GRK5. β cell-specific Grk5 knockout mice showed impaired glucose tolerance with reduced β cell mass, which was accompanied by upregulation of cell cycle inhibitor gene Cdkn1a. Furthermore, we found that Grk5 may drive β cell proliferation through a pathway that includes phosphorylation of HDAC5 and subsequent transcription of immediate-early genes (IEGs) such as Nr4a1, Fosb, Junb, Arc, Egr1, and Srf. Together, these studies suggest GRK5 is linked to T2D through regulation of β cell growth and that it may be a target to preserve β cells during the development of T2D.
Collapse
Affiliation(s)
- Shugo Sasaki
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Surgery, The University of British Columbia, Vancouver, BC, Canada
| | - Cuilan Nian
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Surgery, The University of British Columbia, Vancouver, BC, Canada
- Department of Cellular and Physiological Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Eric E. Xu
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Cellular and Physiological Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Daniel J. Pasula
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Surgery, The University of British Columbia, Vancouver, BC, Canada
| | - Helena Winata
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Surgery, The University of British Columbia, Vancouver, BC, Canada
- Department of Cellular and Physiological Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Sanya Grover
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Dan S. Luciani
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Surgery, The University of British Columbia, Vancouver, BC, Canada
| | - Francis C. Lynn
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Surgery, The University of British Columbia, Vancouver, BC, Canada
- Department of Cellular and Physiological Sciences, The University of British Columbia, Vancouver, BC, Canada
- School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
42
|
de la O S, Yao X, Chang S, Liu Z, Sneddon JB. Single-cell chromatin accessibility of developing murine pancreas identifies cell state-specific gene regulatory programs. Mol Metab 2023; 73:101735. [PMID: 37178817 PMCID: PMC10230264 DOI: 10.1016/j.molmet.2023.101735] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 04/20/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Numerous studies have characterized the existence of cell subtypes, along with their corresponding transcriptional profiles, within the developing mouse pancreas. The upstream mechanisms that initiate and maintain gene expression programs across cell states, however, remain largely unknown. Here, we generate single-nucleus ATAC-Sequencing data of developing murine pancreas and perform an integrated, multi-omic analysis of both chromatin accessibility and RNA expression to describe the chromatin landscape of the developing pancreas at both E14.5 and E17.5 at single-cell resolution. We identify candidate transcription factors regulating cell fate and construct gene regulatory networks of active transcription factor binding to regulatory regions of downstream target genes. This work serves as a valuable resource for the field of pancreatic biology in general and contributes to our understanding of lineage plasticity among endocrine cell types. In addition, these data identify which epigenetic states should be represented in the differentiation of stem cells to the pancreatic beta cell fate to best recapitulate in vitro the gene regulatory networks that are critical for progression along the beta cell lineage in vivo.
Collapse
Affiliation(s)
- Sean de la O
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, 94143, USA; Department of Anatomy, University of California, San Francisco, San Francisco, CA, 94143, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA, 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Xinkai Yao
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, 94143, USA; Department of Anatomy, University of California, San Francisco, San Francisco, CA, 94143, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA, 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Sean Chang
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, 94143, USA; Department of Anatomy, University of California, San Francisco, San Francisco, CA, 94143, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA, 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Zhe Liu
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, 94143, USA; Department of Anatomy, University of California, San Francisco, San Francisco, CA, 94143, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA, 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Julie B Sneddon
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, 94143, USA; Department of Anatomy, University of California, San Francisco, San Francisco, CA, 94143, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA, 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
43
|
Diane A, Mohammed LI, Al-Siddiqi HH. Islets in the body are never flat: transitioning from two-dimensional (2D) monolayer culture to three-dimensional (3D) spheroid for better efficiency in the generation of functional hPSC-derived pancreatic β cells in vitro. Cell Commun Signal 2023; 21:151. [PMID: 37349801 PMCID: PMC10286450 DOI: 10.1186/s12964-023-01171-8] [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: 03/01/2023] [Accepted: 05/20/2023] [Indexed: 06/24/2023] Open
Abstract
Diabetes mellitus (DM), currently affecting more than 537 million people worldwide is a chronic disease characterized by impaired glucose metabolism resulting from a defect in insulin secretion, action, or both due to the loss or dysfunction of pancreatic β cells. Since cadaveric islet transplantation using Edmonton protocol has served as an effective intervention to restore normoglycaemia in T1D patients for months, stem cell-derived β cells have been explored for cell replacement therapy for diabetes. Thus, great effort has been concentrated by scientists on developing in vitro differentiation protocols to realize the therapeutic potential of hPSC-derived β cells. However, most of the 2D traditional monolayer culture could mainly generate insulin-producing β cells with immature phenotype. In the body, pancreatic islets are 3D cell arrangements with complex cell-cell and cell-ECM interactions. Therefore, it is important to consider the spatial organization of the cell in the culture environment. More recently, 3D cell culture platforms have emerged as powerful tools with huge translational potential, particularly for stem cell research. 3D protocols provide a better model to recapitulate not only the in vivo morphology, but also the cell connectivity, polarity, and gene expression mimicking more physiologically the in vivo cell niche. Therefore, the 3D culture constitutes a more relevant model that may help to fill the gap between in vitro and in vivo models. Interestingly, most of the 2D planar methodologies that successfully generated functional hPSC-derived β cells have switched to a 3D arrangement of cells from pancreatic progenitor stage either as suspension clusters or as aggregates, suggesting the effect of 3D on β cell functionality. In this review we highlight the role of dimensionality (2D vs 3D) on the differentiation efficiency for generation of hPSC-derived insulin-producing β cells in vitro. Consequently, how transitioning from 2D monolayer culture to 3D spheroid would provide a better model for an efficient generation of fully functional hPSC-derived β cells mimicking in vivo islet niche for diabetes therapy or drug screening. Video Abstract.
Collapse
Affiliation(s)
- Abdoulaye Diane
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar.
| | - Layla Ibrahim Mohammed
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Heba H Al-Siddiqi
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| |
Collapse
|
44
|
Galindo-Vega A, Maldonado-Lagunas V, Mitre-Aguilar IB, Melendez-Zajgla J. Tumor Microenvironment Role in Pancreatic Cancer Stem Cells. Cells 2023; 12:1560. [PMID: 37371030 DOI: 10.3390/cells12121560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/18/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy with a majority of patients presenting with unresectable or metastatic disease, resulting in a poor 5-year survival rate. This, in turn, is due to a highly complex tumor microenvironment and the presence of cancer stem cells, both of which induce therapy resistance and tumor relapse. Therefore, understanding and targeting the tumor microenvironment and cancer stem cells may be key strategies for designing effective PDAC therapies. In the present review, we summarized recent advances in the role of tumor microenvironment in pancreatic neoplastic progression.
Collapse
Affiliation(s)
- Aaron Galindo-Vega
- Functional Genomics Laboratory, Instituto Nacional de Medicina Genómica, Mexico City 04710, Mexico
| | | | - Irma B Mitre-Aguilar
- Biochemistry Unit, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City 14080, Mexico
| | - Jorge Melendez-Zajgla
- Functional Genomics Laboratory, Instituto Nacional de Medicina Genómica, Mexico City 04710, Mexico
| |
Collapse
|
45
|
Vivot K, Meszaros G, Pangou E, Zhang Z, Qu M, Erbs E, Yeghiazaryan G, Quiñones M, Grandgirard E, Schneider A, Clauss-Creusot E, Charlet A, Faour M, Martin C, Berditchevski F, Sumara I, Luquet S, Kloppenburg P, Nogueiras R, Ricci R. CaMK1D signalling in AgRP neurons promotes ghrelin-mediated food intake. Nat Metab 2023; 5:1045-1058. [PMID: 37277610 DOI: 10.1038/s42255-023-00814-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/25/2023] [Indexed: 06/07/2023]
Abstract
Hypothalamic AgRP/NPY neurons are key players in the control of feeding behaviour. Ghrelin, a major orexigenic hormone, activates AgRP/NPY neurons to stimulate food intake and adiposity. However, cell-autonomous ghrelin-dependent signalling mechanisms in AgRP/NPY neurons remain poorly defined. Here we show that calcium/calmodulin-dependent protein kinase ID (CaMK1D), a genetic hot spot in type 2 diabetes, is activated upon ghrelin stimulation and acts in AgRP/NPY neurons to mediate ghrelin-dependent food intake. Global Camk1d-knockout male mice are resistant to ghrelin, gain less body weight and are protected against high-fat-diet-induced obesity. Deletion of Camk1d in AgRP/NPY, but not in POMC, neurons is sufficient to recapitulate above phenotypes. In response to ghrelin, lack of CaMK1D attenuates phosphorylation of CREB and CREB-dependent expression of the orexigenic neuropeptides AgRP/NPY in fibre projections to the paraventricular nucleus (PVN). Hence, CaMK1D links ghrelin action to transcriptional control of orexigenic neuropeptide availability in AgRP neurons.
Collapse
Affiliation(s)
- Karl Vivot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.
- Centre National de la Recherche Scientifique, Illkirch, France.
- Institut National de la Santé et de la Recherche Médicale, Illkirch, France.
- Université de Strasbourg, Strasbourg, France.
| | - Gergö Meszaros
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Evanthia Pangou
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Zhirong Zhang
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Mengdi Qu
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Eric Erbs
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Gagik Yeghiazaryan
- Biocenter, Institute for Zoology, and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, (CECAD), University of Cologne, Cologne, Germany
| | - Mar Quiñones
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Santiago de Compostela, Spain
| | - Erwan Grandgirard
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Anna Schneider
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Etienne Clauss-Creusot
- Université de Strasbourg, Strasbourg, France
- Centre National de la Recherche Scientifique, Institute of Cellular and Integrative Neurosciences, Strasbourg, France
| | - Alexandre Charlet
- Université de Strasbourg, Strasbourg, France
- Centre National de la Recherche Scientifique, Institute of Cellular and Integrative Neurosciences, Strasbourg, France
| | - Maya Faour
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Claire Martin
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Fedor Berditchevski
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Birmingham, UK
| | - Izabela Sumara
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Serge Luquet
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Peter Kloppenburg
- Biocenter, Institute for Zoology, and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, (CECAD), University of Cologne, Cologne, Germany
| | - Ruben Nogueiras
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Spain
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Romeo Ricci
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.
- Centre National de la Recherche Scientifique, Illkirch, France.
- Institut National de la Santé et de la Recherche Médicale, Illkirch, France.
- Université de Strasbourg, Strasbourg, France.
- Laboratoire de Biochimie et de Biologie Moléculaire, Nouvel Hôpital Civil, Strasbourg, France.
| |
Collapse
|
46
|
Augsornworawat P, Hogrebe NJ, Ishahak M, Schmidt MD, Marquez E, Maestas MM, Veronese-Paniagua DA, Gale SE, Miller JR, Velazco-Cruz L, Millman JR. Single-nucleus multi-omics of human stem cell-derived islets identifies deficiencies in lineage specification. Nat Cell Biol 2023; 25:904-916. [PMID: 37188763 PMCID: PMC10264244 DOI: 10.1038/s41556-023-01150-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 04/17/2023] [Indexed: 05/17/2023]
Abstract
Insulin-producing β cells created from human pluripotent stem cells have potential as a therapy for insulin-dependent diabetes, but human pluripotent stem cell-derived islets (SC-islets) still differ from their in vivo counterparts. To better understand the state of cell types within SC-islets and identify lineage specification deficiencies, we used single-nucleus multi-omic sequencing to analyse chromatin accessibility and transcriptional profiles of SC-islets and primary human islets. Here we provide an analysis that enabled the derivation of gene lists and activity for identifying each SC-islet cell type compared with primary islets. Within SC-islets, we found that the difference between β cells and awry enterochromaffin-like cells is a gradient of cell states rather than a stark difference in identity. Furthermore, transplantation of SC-islets in vivo improved cellular identities overtime, while long-term in vitro culture did not. Collectively, our results highlight the importance of chromatin and transcriptional landscapes during islet cell specification and maturation.
Collapse
Affiliation(s)
- Punn Augsornworawat
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Nathaniel J Hogrebe
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, MO, USA
| | - Matthew Ishahak
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, MO, USA
| | - Mason D Schmidt
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, MO, USA
| | - Erica Marquez
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, MO, USA
| | - Marlie M Maestas
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, MO, USA
| | - Daniel A Veronese-Paniagua
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, MO, USA
| | - Sarah E Gale
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, MO, USA
| | - Julia R Miller
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Leonardo Velazco-Cruz
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, MO, USA
| | - Jeffrey R Millman
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, MO, USA.
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
47
|
Su Q, Yuan F, Li X, Wang X, Yang K, Shao L, Li W. Wfs1 loss-of-function disrupts the composition of mouse pancreatic endocrine cells from birth and impairs Glut2 localization to cytomembrane in pancreatic β cells. Biochem Biophys Res Commun 2023; 658:80-87. [PMID: 37027908 DOI: 10.1016/j.bbrc.2023.03.074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023]
Abstract
Wfs1 is an endoplasmic reticulum (ER) membrane located protein highly expressed in pancreatic β cells and brain. Wfs1 deficiency causes adult pancreatic β cells dysfunction following β cells apoptosis. Previous studies mainly focus on the Wfs1 function in adult mouse pancreatic β cells. However, whether Wfs1 loss-of-function impairs mouse pancreatic β cell from its early development is unknown. In our study, Wfs1 deficiency disrupts the composition of mouse pancreatic endocrine cells from early postnatal day 0 (P0) to 8 weeks old, with decreased percentage of β cells and increased percentage of α and δ cells. Meanwhile, Wfs1 loss-of-function leads to reduced intracellular insulin content. Notably, Wfs1 deficiency impairs Glut2 localization and causes the accumulation of Glut2 in mouse pancreatic β cell cytoplasm. In Wfs1-deficient mice, glucose homeostasis is disturbed from early 3 weeks old to 8 weeks old. This work reveals that Wfs1 is significantly required for the composition of pancreatic endocrine cells and is essential for Glut2 localization in mouse pancreatic β cells.
Collapse
Affiliation(s)
- Qiang Su
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Fei Yuan
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xiaobo Li
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xuan Wang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Kaijiang Yang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Li Shao
- Department of VIP Clinic, Shanghai East Hospital, Tongji University School of Medicine, No. 1800 Yuntai Road, Pudong District, Shanghai, 200092, China.
| | - Weida Li
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China; Reg-Verse Therapeutics (Shanghai) Co. Ltd., Shanghai, 200120, China.
| |
Collapse
|
48
|
Ruiz-Otero N, Kuruvilla R. Role of Delta/Notch-like EGF-related receptor in blood glucose homeostasis. Front Endocrinol (Lausanne) 2023; 14:1161085. [PMID: 37223028 PMCID: PMC10200888 DOI: 10.3389/fendo.2023.1161085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/18/2023] [Indexed: 05/25/2023] Open
Abstract
Cell-cell interactions are necessary for optimal endocrine functions in the pancreas. β-cells, characterized by the expression and secretion of the hormone insulin, are a major constituent of functional micro-organs in the pancreas known as islets of Langerhans. Cell-cell contacts between β-cells are required to regulate insulin production and glucose-stimulated insulin secretion, which are key determinants of blood glucose homeostasis. Contact-dependent interactions between β-cells are mediated by gap junctions and cell adhesion molecules such as E-cadherin and N-CAM. Recent genome-wide studies have implicated Delta/Notch-like EGF-related receptor (Dner) as a potential susceptibility locus for Type 2 Diabetes in humans. DNER is a transmembrane protein and a proposed Notch ligand. DNER has been implicated in neuron-glia development and cell-cell interactions. Studies herein demonstrate that DNER is expressed in β-cells with an onset during early postnatal life and sustained throughout adulthood in mice. DNER loss in adult β-cells in mice (β-Dner cKO mice) disrupted islet architecture and decreased the expression of N-CAM and E-cadherin. β-Dner cKO mice also exhibited impaired glucose tolerance, defects in glucose- and KCl-induced insulin secretion, and decreased insulin sensitivity. Together, these studies suggest that DNER plays a crucial role in mediating islet cell-cell interactions and glucose homeostasis.
Collapse
Affiliation(s)
- Nelmari Ruiz-Otero
- Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Rejji Kuruvilla
- Department of Biology, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
49
|
Harithpriya K, Jayasuriya R, Adhikari T, Rai A, Ramkumar KM. Modulation of transcription factors by small molecules in β-cell development and differentiation. Eur J Pharmacol 2023; 946:175606. [PMID: 36809813 DOI: 10.1016/j.ejphar.2023.175606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/21/2023]
Abstract
Transcription factors regulate gene expression and play crucial roles in development and differentiation of pancreatic β-cell. The expression and/or activities of these transcription factors are reduced when β-cells are chronically exposed to hyperglycemia, which results in loss of β-cell function. Optimal expression of such transcription factors is required to maintain normal pancreatic development and β-cell function. Over many other methods of regenerating β-cells, using small molecules to activate transcription factors has gained insights, resulting in β-cells regeneration and survival. In this review, we discuss the broad spectrum of transcription factors regulating pancreatic β-cell development, differentiation and regulation of these factors in normal and pathological states. Also, we have presented set of potential pharmacological effects of natural and synthetic compounds on activities of transcription factor involved in pancreatic β-cell regeneration and survival. Exploring these compounds and their action on transcription factors responsible for pancreatic β-cell function and survival could be useful in providing new insights for development of small molecule modulators.
Collapse
Affiliation(s)
- Kannan Harithpriya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamil Nadu, India
| | - Ravichandran Jayasuriya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamil Nadu, India
| | - Trishla Adhikari
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamil Nadu, India
| | - Awantika Rai
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamil Nadu, India
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamil Nadu, India.
| |
Collapse
|
50
|
Jiang H, Jiang FX. Human pluripotent stem cell-derived β cells: Truly immature islet β cells for type 1 diabetes therapy? World J Stem Cells 2023; 15:182-195. [PMID: 37180999 PMCID: PMC10173812 DOI: 10.4252/wjsc.v15.i4.182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/30/2023] [Accepted: 03/20/2023] [Indexed: 04/26/2023] Open
Abstract
A century has passed since the Nobel Prize winning discovery of insulin, which still remains the mainstay treatment for type 1 diabetes mellitus (T1DM) to this day. True to the words of its discoverer Sir Frederick Banting, “insulin is not a cure for diabetes, it is a treatment”, millions of people with T1DM are dependent on daily insulin medications for life. Clinical donor islet transplantation has proven that T1DM is curable, however due to profound shortages of donor islets, it is not a mainstream treatment option for T1DM. Human pluripotent stem cell derived insulin-secreting cells, pervasively known as stem cell-derived β cells (SC-β cells), are a promising alternative source and have the potential to become a T1DM treatment through cell replacement therapy. Here we briefly review how islet β cells develop and mature in vivo and several types of reported SC-β cells produced using different ex vivo protocols in the last decade. Although some markers of maturation were expressed and glucose stimulated insulin secretion was shown, the SC-β cells have not been directly compared to their in vivo counterparts, generally have limited glucose response, and are not yet fully matured. Due to the presence of extra-pancreatic insulin-expressing cells, and ethical and technological issues, further clarification of the true nature of these SC-β cells is required.
Collapse
Affiliation(s)
- Helen Jiang
- Sir Charles Gairdner Hospital, University of Western Australia, Perth 6009, Australia
| | - Fang-Xu Jiang
- School of Biomedical Sciences, University of Western Australia, Perth 6009, Australia
- School of Health and Medical Sciences, Edith Cowan University, Perth 6027, Australia
| |
Collapse
|