17,246 results on '"insulin-secreting cells"'
Search Results
2. Dysfunctional β-cell longevity in diabetes relies on energy conservation and positive epistasis.
- Author
-
Raval, Kavit, Jamshidi, Neema, Seyran, Berfin, Salwinski, Lukasz, Pillai, Raju, Yang, Lixin, Ma, Feiyang, Pellegrini, Matteo, Shin, Juliana, Yang, Xia, and Tudzarova, Slavica
- Subjects
Insulin-Secreting Cells ,Animals ,Diabetes Mellitus ,Type 2 ,Humans ,Mice ,Phosphofructokinase-2 ,Energy Metabolism ,Epistasis ,Genetic ,Male ,Transcriptome ,Apoptosis ,Mice ,Inbred C57BL - Abstract
Long-lived PFKFB3-expressing β-cells are dysfunctional partly because of prevailing glycolysis that compromises metabolic coupling of insulin secretion. Their accumulation in type 2 diabetes (T2D) appears to be related to the loss of apoptotic competency of cell fitness competition that maintains islet function by favoring constant selection of healthy winner cells. To investigate how PFKFB3 can disguise the competitive traits of dysfunctional loser β-cells, we analyzed the overlap between human β-cells with bona fide loser signature across diabetes pathologies using the HPAP scRNA-seq and spatial transcriptomics of PFKFB3-positive β-cells from nPOD T2D pancreata. The overlapping transcriptional profile of loser β-cells was represented by down-regulated ribosomal biosynthesis and genes encoding for mitochondrial respiration. PFKFB3-positive loser β-cells had the reduced expression of HLA class I and II genes. Gene-gene interaction analysis revealed that PFKFB3 rs1983890 can interact with the anti-apoptotic gene MAIP1 implicating positive epistasis as a mechanism for prolonged survival of loser β-cells in T2D. Inhibition of PFKFB3 resulted in the clearance of dysfunctional loser β-cells leading to restored glucose tolerance in the mouse model of T2D.
- Published
- 2024
3. Calorie restriction increases insulin sensitivity to promote beta cell homeostasis and longevity in mice
- Author
-
dos Santos, Cristiane, Cambraia, Amanda, Shrestha, Shristi, Cutler, Melanie, Cottam, Matthew, Perkins, Guy, Lev-Ram, Varda, Roy, Birbickram, Acree, Christopher, Kim, Keun-Young, Deerinck, Thomas, Dean, Danielle, Cartailler, Jean Philippe, MacDonald, Patrick E, Hetzer, Martin, Ellisman, Mark, and Arrojo e Drigo, Rafael
- Subjects
Medical Biochemistry and Metabolomics ,Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Diabetes ,Genetics ,Aging ,Nutrition ,1.1 Normal biological development and functioning ,Metabolic and endocrine ,Animals ,Caloric Restriction ,Insulin-Secreting Cells ,Longevity ,Homeostasis ,Insulin Resistance ,Mice ,Male ,Diet ,High-Fat ,Mice ,Inbred C57BL ,Mitochondria ,Cell Proliferation ,Mitophagy ,Insulin ,Gene Regulatory Networks - Abstract
Caloric restriction (CR) can extend the organism life- and health-span by improving glucose homeostasis. How CR affects the structure-function of pancreatic beta cells remains unknown. We used single nucleus transcriptomics to show that CR increases the expression of genes for beta cell identity, protein processing, and organelle homeostasis. Gene regulatory network analysis reveal that CR activates transcription factors important for beta cell identity and homeostasis, while imaging metabolomics demonstrates that beta cells upon CR are more energetically competent. In fact, high-resolution microscopy show that CR reduces beta cell mitophagy to increase mitochondria mass and the potential for ATP generation. However, CR beta cells have impaired adaptive proliferation in response to high fat diet feeding. Finally, we show that long-term CR delays the onset of beta cell aging hallmarks and promotes cell longevity by reducing beta cell turnover. Therefore, CR could be a feasible approach to preserve compromised beta cell structure-function during aging and diabetes.
- Published
- 2024
4. The fate of intracellular S1P regulates lipid droplet turnover and lipotoxicity in pancreatic beta-cells.
- Author
-
Tang, Yadi, Majewska, Mariola, Leß, Britta, Mehmeti, Ilir, Wollnitzke, Philipp, Semleit, Nina, Levkau, Bodo, Saba, Julie, van Echten-Deckert, Gerhild, and Gurgul-Convey, Ewa
- Subjects
beta-cells ,ceramide ,diabetes ,free fatty acids ,insulin-secreting cells ,lipid droplets ,mitochondria ,sphingosine-1 phosphate ,Insulin-Secreting Cells ,Lysophospholipids ,Sphingosine ,Rats ,Animals ,Lipid Droplets ,Fatty Acids ,Nonesterified ,Aldehyde-Lyases ,Lipid Metabolism ,Humans ,Cell Line ,Oxidative Stress ,Intracellular Space - Abstract
Lipotoxicity has been considered the main cause of pancreatic beta-cell failure during type 2 diabetes development. Lipid droplets (LD) are believed to regulate the beta-cell sensitivity to free fatty acids (FFA), but the underlying molecular mechanisms are largely unclear. Accumulating evidence points, however, to an important role of intracellular sphingosine-1-phosphate (S1P) metabolism in lipotoxicity-mediated disturbances of beta-cell function. In the present study, we compared the effects of an increased irreversible S1P degradation (S1P-lyase, SPL overexpression) with those associated with an enhanced S1P recycling (overexpression of S1P phosphatase 1, SGPP1) on LD formation and lipotoxicity in rat INS1E beta-cells. Interestingly, although both approaches led to a reduced S1P concentration, they had opposite effects on the susceptibility to FFA. Overexpression of SGPP1 prevented FFA-mediated caspase-3 activation by a mechanism involving an enhanced lipid storage capacity and prevention of oxidative stress. In contrast, SPL overexpression limited LD biogenesis, content, and size, while accelerating lipophagy. This was associated with FFA-induced hydrogen peroxide formation, mitochondrial fragmentation, and dysfunction, as well as ER stress. These changes coincided with the upregulation of proapoptotic ceramides but were independent of lipid peroxidation rate. Also in human EndoC-βH1 beta-cells, suppression of SPL with simultaneous overexpression of SGPP1 led to a similar and even more pronounced LD phenotype as that in INS1E-SGPP1 cells. Thus, intracellular S1P turnover significantly regulates LD content and size and influences beta-cell sensitivity to FFA.
- Published
- 2024
5. Network representation of multicellular activity in pancreatic islets: Technical considerations for functional connectivity analysis
- Author
-
Šterk, Marko, Zhang, Yaowen, Pohorec, Viljem, Leitgeb, Eva Paradiž, Dolenšek, Jurij, Benninger, Richard KP, Stožer, Andraž, Kravets, Vira, and Gosak, Marko
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Diabetes ,1.1 Normal biological development and functioning ,Underpinning research ,Metabolic and endocrine ,Islets of Langerhans ,Animals ,Computational Biology ,Mice ,Insulin ,Humans ,Insulin-Secreting Cells ,Insulin Secretion ,Models ,Biological ,Calcium ,Calcium Signaling ,Mathematical Sciences ,Information and Computing Sciences ,Bioinformatics - Abstract
Within the islets of Langerhans, beta cells orchestrate synchronized insulin secretion, a pivotal aspect of metabolic homeostasis. Despite the inherent heterogeneity and multimodal activity of individual cells, intercellular coupling acts as a homogenizing force, enabling coordinated responses through the propagation of intercellular waves. Disruptions in this coordination are implicated in irregular insulin secretion, a hallmark of diabetes. Recently, innovative approaches, such as integrating multicellular calcium imaging with network analysis, have emerged for a quantitative assessment of the cellular activity in islets. However, different groups use distinct experimental preparations, microscopic techniques, apply different methods to process the measured signals and use various methods to derive functional connectivity patterns. This makes comparisons between findings and their integration into a bigger picture difficult and has led to disputes in functional connectivity interpretations. To address these issues, we present here a systematic analysis of how different approaches influence the network representation of islet activity. Our findings show that the choice of methods used to construct networks is not crucial, although care is needed when combining data from different islets. Conversely, the conclusions drawn from network analysis can be heavily affected by the pre-processing of the time series, the type of the oscillatory component in the signals, and by the experimental preparation. Our tutorial-like investigation aims to resolve interpretational issues, reconcile conflicting views, advance functional implications, and encourage researchers to adopt connectivity analysis. As we conclude, we outline challenges for future research, emphasizing the broader applicability of our conclusions to other tissues exhibiting complex multicellular dynamics.
- Published
- 2024
6. In situ structure of actin remodeling during glucose-stimulated insulin secretion using cryo-electron tomography.
- Author
-
Li, Weimin, Li, Angdi, Yu, Bing, Zhang, Xiaoxiao, Liu, Xiaoyan, White, Kate, Stevens, Raymond, Baumeister, Wolfgang, Jasnin, Marion, Sun, Liping, and Sali, Andrej
- Subjects
Insulin Secretion ,Actins ,Glucose ,Electron Microscope Tomography ,Insulin ,Insulin-Secreting Cells ,Actin Cytoskeleton - Abstract
Actin mediates insulin secretion in pancreatic β-cells through remodeling. Hampered by limited resolution, previous studies have offered an ambiguous depiction as depolymerization and repolymerization. We report the in situ structure of actin remodeling in INS-1E β-cells during glucose-stimulated insulin secretion at nanoscale resolution. After remodeling, the actin filament network at the cell periphery exhibits three marked differences: 12% of actin filaments reorient quasi-orthogonally to the ventral membrane; the filament network mainly remains as cell-stabilizing bundles but partially reconfigures into a less compact arrangement; actin filaments anchored to the ventral membrane reorganize from a netlike to a blooming architecture. Furthermore, the density of actin filaments and microtubules around insulin secretory granules decreases, while actin filaments and microtubules become more densely packed. The actin filament network after remodeling potentially precedes the transport and release of insulin secretory granules. These findings advance our understanding of actin remodeling and its role in glucose-stimulated insulin secretion.
- Published
- 2024
7. Differential lipid signaling from CD4+ and CD8+ T cells contributes to type 1 diabetes development
- Author
-
White, Tayleur D, Almutairi, Abdulaziz, Gai-Tusing, Ying, Stephenson, Daniel J, Stephenson, Benjamin D, Chalfant, Charles E, Lei, Xiaoyong, Lu, Brian, Hammock, Bruce D, DiLorenzo, Teresa P, and Ramanadham, Sasanka
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Immunology ,Autoimmune Disease ,Diabetes ,Pediatric ,Metabolic and endocrine ,Animals ,Diabetes Mellitus ,Type 1 ,CD8-Positive T-Lymphocytes ,Mice ,Inbred NOD ,Mice ,CD4-Positive T-Lymphocytes ,Signal Transduction ,Insulin-Secreting Cells ,Group VI Phospholipases A2 ,Lipid Metabolism ,Mice ,SCID ,Female ,T-lymphocytes ,lipid signaling ,type 1 diabetes ,adoptive transfer ,islet microscopy ,flow cytometry ,lipidomics ,Medical Microbiology ,Biochemistry and cell biology ,Genetics - Abstract
IntroductionWe reported that Ca2+-independent phospholipase A2β (iPLA2β)-derived lipids (iDLs) contribute to type 1 diabetes (T1D) onset. As CD4+ and CD8+ T cells are critical in promoting β-cell death, we tested the hypothesis that iDL signaling from these cells participates in T1D development.MethodsCD4+ and CD8+ T cells from wild-type non-obese diabetic (NOD) and NOD.iPLA2β+/- (NOD.HET) mice were administered in different combinations to immunodeficient NOD.scid.ResultsIn mice receiving only NOD T cells, T1D onset was rapid (5 weeks), incidence 100% by 20 weeks, and islets absent. In contrast, onset was delayed 1 week and incidence reduced 40%-50% in mice receiving combinations that included NOD.HET T cells. Consistently, islets from these non-diabetic mice were devoid of infiltrate and contained insulin-positive β-cells. Reduced iPLA2β led to decreased production of proinflammatory lipids from CD4+ T cells including prostaglandins and dihydroxyeicosatrienoic acids (DHETs), products of soluble epoxide hydrolase (sEH), and inhibition of their signaling decreased (by 82%) IFNγ+CD4+ cells abundance. However, only DHETs production was reduced from CD8+ T cells and was accompanied by decreases in sEH and granzyme B.DiscussionThese findings suggest that differential select iDL signaling in CD4+ and CD8+ T cells contributes to T1D development, and that therapeutics targeting such signaling might be considered to counter T1D.
- Published
- 2024
8. Subcellular Feature-Based Classification of α and β Cells Using Soft X-ray Tomography
- Author
-
Deshmukh, Aneesh, Chang, Kevin, Cuala, Janielle, Vanslembrouck, Bieke, Georgia, Senta, Loconte, Valentina, and White, Kate L
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Diabetes ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning ,Metabolic and endocrine ,Insulin-Secreting Cells ,Glucagon-Secreting Cells ,Animals ,Tomography ,X-Ray ,Mice ,Humans ,Insulin ,soft X-ray tomography ,cryogenic fluorescence microscopy ,pancreatic islets ,alpha cells ,beta cells ,3D cell mapping ,machine learning ,Uniform Manifold Approximation and Projection ,α cells ,β cells ,Biological sciences ,Biomedical and clinical sciences - Abstract
The dysfunction of α and β cells in pancreatic islets can lead to diabetes. Many questions remain on the subcellular organization of islet cells during the progression of disease. Existing three-dimensional cellular mapping approaches face challenges such as time-intensive sample sectioning and subjective cellular identification. To address these challenges, we have developed a subcellular feature-based classification approach, which allows us to identify α and β cells and quantify their subcellular structural characteristics using soft X-ray tomography (SXT). We observed significant differences in whole-cell morphological and organelle statistics between the two cell types. Additionally, we characterize subtle biophysical differences between individual insulin and glucagon vesicles by analyzing vesicle size and molecular density distributions, which were not previously possible using other methods. These sub-vesicular parameters enable us to predict cell types systematically using supervised machine learning. We also visualize distinct vesicle and cell subtypes using Uniform Manifold Approximation and Projection (UMAP) embeddings, which provides us with an innovative approach to explore structural heterogeneity in islet cells. This methodology presents an innovative approach for tracking biologically meaningful heterogeneity in cells that can be applied to any cellular system.
- Published
- 2024
9. 췌도부전당뇨병에 대한 입장 성명서.
- Author
-
Kim, Ji Yoon, Jin, Sang-Man, Kim, Gyuri, Kim, Soo Kyoung, Kim, Won Jun, Moon, Sun Joon, Yoo, Jee Hee, Lee, Da Young, Lee, Seung-Eun, Jun, Ji Eun, and Kim, Jae Hyeon
- Abstract
Diabetes mellitus is a heterogeneous disease that encompasses a wide range of conditions, from mild cases to severe conditions where survival depends on insulin therapy. The Korean Diabetes Association Task Force Team for Diabetes with β-Cell Failure has established the term to classify severe refractory disease with β-cell failure. Individuals with β-cell failure are at high risk of diabetes-related complications. We propose that diabetes with β-cell failure can be diagnosed when individuals treated with multiple daily insulin injections or insulin pumps meet at least one of the following criteria: fasting C-peptide ≤ 0.6 ng/mL, non-fasting C-peptide ≤ 1.8 ng/mL, 24-hour urine C-peptide < 30 μg/day, or spot urine C-peptide/creatinine ratio ≤ 0.6 nmol/mmol. Among cases of diabetes with β-cell failure, β-cell failure with absolute insulin deficiency can be diagnosed when at least one of the following criteria is met: fasting C-peptide < 0.24 ng/mL, non-fasting C-peptide < 0.6 ng/mL, or spot urine C-peptide/creatinine ratio < 0.2 nmol/mmol. Multiple daily insulin injections with long-acting insulin analogs and rapid-acting insulin analogs or insulin pumps are required for treatment of diabetes with β-cell failure. Continuous glucose monitoring and an automated insulin delivery system, sensor-augmented pump, or smart insulin pen, along with structured education, are necessary. We call for improvements in the relevant systems to ensure that such treatments can be provided. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. Species-specific roles for the MAFA and MAFB transcription factors in regulating islet β cell identity
- Author
-
Cha, Jeeyeon, Tong, Xin, Walker, Emily M, Dahan, Tehila, Cochrane, Veronica A, Ashe, Sudipta, Russell, Ronan, Osipovich, Anna B, Mawla, Alex M, Guo, Min, Liu, Jin-hua, Loyd, Zachary A, Huising, Mark O, Magnuson, Mark A, Hebrok, Matthias, Dor, Yuval, and Stein, Roland
- Subjects
Biomedical and Clinical Sciences ,Health Sciences ,Genetics ,Diabetes ,2.1 Biological and endogenous factors ,Metabolic and endocrine ,Adult ,Humans ,Animals ,Mice ,MafB Transcription Factor ,Diabetes Mellitus ,Type 2 ,Islets of Langerhans ,Insulin-Secreting Cells ,Insulin ,Beta cells ,Cell Biology ,Endocrinology ,Islet cells ,Biomedical and clinical sciences ,Health sciences - Abstract
Type 2 diabetes (T2D) is associated with compromised identity of insulin-producing pancreatic islet β cells, characterized by inappropriate production of other islet cell-enriched hormones. Here, we examined how hormone misexpression was influenced by the MAFA and MAFB transcription factors, closely related proteins that maintain islet cell function. Mice specifically lacking MafA in β cells demonstrated broad, population-wide changes in hormone gene expression with an overall gene signature closely resembling islet gastrin+ (Gast+) cells generated under conditions of chronic hyperglycemia and obesity. A human β cell line deficient in MAFB, but not one lacking MAFA, also produced a GAST+ gene expression pattern. In addition, GAST was detected in human T2D β cells with low levels of MAFB. Moreover, evidence is provided that human MAFB can directly repress GAST gene transcription. These results support a potentially novel, species-specific role for MafA and MAFB in maintaining adult mouse and human β cell identity, respectively. Here, we discuss the possibility that induction of Gast/GAST and other non-β cell hormones, by reduction in the levels of these transcription factors, represents a dysfunctional β cell signature.
- Published
- 2023
11. Is 14-3-3 the Combination to Unlock New Pathways to Improve Metabolic Homeostasis and β-Cell Function?
- Author
-
Rial, Sabri, Shishani, Rahaf, Cummings, Bethany, and Lim, Gareth
- Subjects
Humans ,14-3-3 Proteins ,Insulin-Secreting Cells ,Diabetes Mellitus ,Homeostasis ,Glucose ,Insulin - Abstract
UNLABELLED: Since their discovery nearly five decades ago, molecular scaffolds belonging to the 14-3-3 protein family have been recognized as pleiotropic regulators of diverse cellular and physiological functions. With their ability to bind to proteins harboring specific serine and threonine phosphorylation motifs, 14-3-3 proteins can interact with and influence the function of docking proteins, enzymes, transcription factors, and transporters that have essential roles in metabolism and glucose homeostasis. Here, we will discuss the regulatory functions of 14-3-3 proteins that will be of great interest to the fields of metabolism, pancreatic β-cell biology, and diabetes. We first describe how 14-3-3 proteins play a central role in glucose and lipid homeostasis by modulating key pathways of glucose uptake, glycolysis, oxidative phosphorylation, and adipogenesis. This is followed by a discussion of the contributions of 14-3-3 proteins to calcium-dependent exocytosis and how this relates to insulin secretion from β-cells. As 14-3-3 proteins are major modulators of apoptosis and cell cycle progression, we will explore if 14-3-3 proteins represent a viable target for promoting β-cell regeneration and discuss the feasibility of targeting 14-3-3 proteins to treat metabolic diseases such as diabetes. ARTICLE HIGHLIGHTS: 14-3-3 proteins are ubiquitously expressed scaffolds with multiple roles in glucose homeostasis and metabolism. 14-3-3ζ regulates adipogenesis via distinct mechanisms and is required for postnatal adiposity and adipocyte function. 14-3-3ζ controls glucose-stimulated insulin secretion from pancreatic β-cells by regulating mitochondrial function and ATP synthesis as well as facilitating cross talk between β-cells and α-cells.
- Published
- 2023
12. Loss of ZNF148 enhances insulin secretion in human pancreatic β cells
- Author
-
de Klerk, Eleonora, Xiao, Yini, Emfinger, Christopher H, Keller, Mark P, Berrios, David I, Loconte, Valentina, Ekman, Axel A, White, Kate L, Cardone, Rebecca L, Kibbey, Richard G, Attie, Alan D, and Hebrok, Matthias
- Subjects
Biomedical and Clinical Sciences ,Health Sciences ,Genetics ,Stem Cell Research ,Diabetes ,1.1 Normal biological development and functioning ,Generic health relevance ,Metabolic and endocrine ,Humans ,Insulin-Secreting Cells ,Insulin Secretion ,Glucose ,Insulin ,Exocytosis ,DNA-Binding Proteins ,Transcription Factors ,Embryonic stem cells ,Islet cells ,Metabolism ,Stem cells ,Biomedical and clinical sciences ,Health sciences - Abstract
Insulin secretion from pancreatic β cells is essential to the maintenance of glucose homeostasis. Defects in this process result in diabetes. Identifying genetic regulators that impair insulin secretion is crucial for the identification of novel therapeutic targets. Here, we show that reduction of ZNF148 in human islets, and its deletion in stem cell-derived β cells (SC-β cells), enhances insulin secretion. Transcriptomics of ZNF148-deficient SC-β cells identifies increased expression of annexin and S100 genes whose proteins form tetrameric complexes involved in regulation of insulin vesicle trafficking and exocytosis. ZNF148 in SC-β cells prevents translocation of annexin A2 from the nucleus to its functional place at the cell membrane via direct repression of S100A16 expression. These findings point to ZNF148 as a regulator of annexin-S100 complexes in human β cells and suggest that suppression of ZNF148 may provide a novel therapeutic strategy to enhance insulin secretion.
- Published
- 2023
13. Integrating genetics with single-cell multiomic measurements across disease states identifies mechanisms of beta cell dysfunction in type 2 diabetes
- Author
-
Wang, Gaowei, Chiou, Joshua, Zeng, Chun, Miller, Michael, Matta, Ileana, Han, Jee Yun, Kadakia, Nikita, Okino, Mei-Lin, Beebe, Elisha, Mallick, Medhavi, Camunas-Soler, Joan, dos Santos, Theodore, Dai, Xiao-Qing, Ellis, Cara, Hang, Yan, Kim, Seung K, MacDonald, Patrick E, Kandeel, Fouad R, Preissl, Sebastian, Gaulton, Kyle J, and Sander, Maike
- Subjects
Biochemistry and Cell Biology ,Genetics ,Biological Sciences ,Obesity ,Machine Learning and Artificial Intelligence ,Human Genome ,Diabetes ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning ,Metabolic and endocrine ,Humans ,Diabetes Mellitus ,Type 2 ,Multiomics ,Insulin-Secreting Cells ,Gene Expression Regulation ,Chromatin ,Medical and Health Sciences ,Developmental Biology ,Agricultural biotechnology ,Bioinformatics and computational biology - Abstract
Dysfunctional pancreatic islet beta cells are a hallmark of type 2 diabetes (T2D), but a comprehensive understanding of the underlying mechanisms, including gene dysregulation, is lacking. Here we integrate information from measurements of chromatin accessibility, gene expression and function in single beta cells with genetic association data to nominate disease-causal gene regulatory changes in T2D. Using machine learning on chromatin accessibility data from 34 nondiabetic, pre-T2D and T2D donors, we identify two transcriptionally and functionally distinct beta cell subtypes that undergo an abundance shift during T2D progression. Subtype-defining accessible chromatin is enriched for T2D risk variants, suggesting a causal contribution of subtype identity to T2D. Both beta cell subtypes exhibit activation of a stress-response transcriptional program and functional impairment in T2D, which is probably induced by the T2D-associated metabolic environment. Our findings demonstrate the power of multimodal single-cell measurements combined with machine learning for characterizing mechanisms of complex diseases.
- Published
- 2023
14. Understanding cell fate acquisition in stem-cell-derived pancreatic islets using single-cell multiome-inferred regulomes
- Author
-
Zhu, Han, Wang, Gaowei, Nguyen-Ngoc, Kim-Vy, Kim, Dongsu, Miller, Michael, Goss, Georgina, Kovsky, Jenna, Harrington, Austin R, Saunders, Diane C, Hopkirk, Alexander L, Melton, Rebecca, Powers, Alvin C, Preissl, Sebastian, Spagnoli, Francesca M, Gaulton, Kyle J, and Sander, Maike
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Autoimmune Disease ,Stem Cell Research ,Stem Cell Research - Nonembryonic - Human ,Stem Cell Research - Induced Pluripotent Stem Cell ,Diabetes ,Stem Cell Research - Embryonic - Human ,Genetics ,Regenerative Medicine ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Metabolic and endocrine ,Adult ,Humans ,Islets of Langerhans ,Pancreas ,Cell Differentiation ,Insulin-Secreting Cells ,Pluripotent Stem Cells ,ATAC-seq ,CDX2 ,RNA-seq ,development ,fetal pancreas ,gene regulatory network ,human pluripotent stem cells ,islets ,pancreas ,serotonin ,signals ,single-cell genomics ,transcription factors ,β cell ,Medical and Health Sciences ,Developmental Biology ,Biochemistry and cell biology - Abstract
Pancreatic islet cells derived from human pluripotent stem cells hold great promise for modeling and treating diabetes. Differences between stem-cell-derived and primary islets remain, but molecular insights to inform improvements are limited. Here, we acquire single-cell transcriptomes and accessible chromatin profiles during in vitro islet differentiation and pancreas from childhood and adult donors for comparison. We delineate major cell types, define their regulomes, and describe spatiotemporal gene regulatory relationships between transcription factors. CDX2 emerged as a regulator of enterochromaffin-like cells, which we show resemble a transient, previously unrecognized, serotonin-producing pre-β cell population in fetal pancreas, arguing against a proposed non-pancreatic origin. Furthermore, we observe insufficient activation of signal-dependent transcriptional programs during in vitro β cell maturation and identify sex hormones as drivers of β cell proliferation in childhood. Altogether, our analysis provides a comprehensive understanding of cell fate acquisition in stem-cell-derived islets and a framework for manipulating cell identities and maturity.
- Published
- 2023
15. Effects of Saccharomyces cerevisiae on Pancreatic Alpha and Beta Cells and Metabolic Profile in Broilers
- Author
-
Peña B., Silvana J., Salazar J., Johan S., Pardo, Jhon F., Roa, Maria L., Corredor-Matus, José R., and Ochoa-Amaya, Julieta E.
- Published
- 2024
- Full Text
- View/download PDF
16. Nutrient regulation of the islet epigenome controls adaptive insulin secretion
- Author
-
Wortham, Matthew, Liu, Fenfen, Harrington, Austin R, Fleischman, Johanna Y, Wallace, Martina, Mulas, Francesca, Mallick, Medhavi, Vinckier, Nicholas K, Cross, Benjamin R, Chiou, Joshua, Patel, Nisha A, Sui, Yinghui, McGrail, Carolyn, Jun, Yesl, Wang, Gaowei, Jhala, Ulupi S, Schüle, Roland, Shirihai, Orian S, Huising, Mark O, Gaulton, Kyle J, Metallo, Christian M, and Sander, Maike
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Diabetes ,Autoimmune Disease ,Genetics ,Nutrition ,1.1 Normal biological development and functioning ,Generic health relevance ,Metabolic and endocrine ,Mice ,Humans ,Animals ,Insulin Secretion ,Diabetes Mellitus ,Type 2 ,Histones ,Epigenome ,Islets of Langerhans ,Insulin ,Insulin-Secreting Cells ,Glucose ,Endocrinology ,Epigenetics ,Islet cells ,Metabolism ,Medical and Health Sciences ,Immunology ,Biological sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
Adaptation of the islet β cell insulin-secretory response to changing insulin demand is critical for blood glucose homeostasis, yet the mechanisms underlying this adaptation are unknown. Here, we have shown that nutrient-stimulated histone acetylation plays a key role in adapting insulin secretion through regulation of genes involved in β cell nutrient sensing and metabolism. Nutrient regulation of the epigenome occurred at sites occupied by the chromatin-modifying enzyme lysine-specific demethylase 1 (Lsd1) in islets. β Cell-specific deletion of Lsd1 led to insulin hypersecretion, aberrant expression of nutrient-response genes, and histone hyperacetylation. Islets from mice adapted to chronically increased insulin demand exhibited shared epigenetic and transcriptional changes. Moreover, we found that genetic variants associated with type 2 diabetes were enriched at LSD1-bound sites in human islets, suggesting that interpretation of nutrient signals is genetically determined and clinically relevant. Overall, these studies revealed that adaptive insulin secretion involves Lsd1-mediated coupling of nutrient state to regulation of the islet epigenome.
- Published
- 2023
17. A beta cell subset with enhanced insulin secretion and glucose metabolism is reduced in type 2 diabetes.
- Author
-
Rubio-Navarro, Alfonso, Gómez-Banoy, Nicolás, Stoll, Lisa, Dündar, Friederike, Mawla, Alex, Ma, Lunkun, Cortada, Eric, Zumbo, Paul, Li, Ang, Reiterer, Moritz, Montoya-Oviedo, Nathalia, Homan, Edwin, Imai, Norihiro, Gilani, Ankit, Liu, Chengyang, Naji, Ali, Yang, Boris, Chong, Angie, Cohen, David, Chen, Shuibing, Cao, Jingli, Pitt, Geoffrey, Betel, Doron, Lo, James, and Huising, Mark
- Subjects
Humans ,Mice ,Animals ,Diabetes Mellitus ,Type 2 ,Insulin Secretion ,Insulin ,Diabetes Mellitus ,Experimental ,Insulin-Secreting Cells ,Glucose - Abstract
The pancreatic islets are composed of discrete hormone-producing cells that orchestrate systemic glucose homeostasis. Here we identify subsets of beta cells using a single-cell transcriptomic approach. One subset of beta cells marked by high CD63 expression is enriched for the expression of mitochondrial metabolism genes and exhibits higher mitochondrial respiration compared with CD63lo beta cells. Human and murine pseudo-islets derived from CD63hi beta cells demonstrate enhanced glucose-stimulated insulin secretion compared with pseudo-islets from CD63lo beta cells. We show that CD63hi beta cells are diminished in mouse models of and in humans with type 2 diabetes. Finally, transplantation of pseudo-islets generated from CD63hi but not CD63lo beta cells into diabetic mice restores glucose homeostasis. These findings suggest that loss of a specific subset of beta cells may lead to diabetes. Strategies to reconstitute or maintain CD63hi beta cells may represent a potential anti-diabetic therapy.
- Published
- 2023
18. Protector Role of Cx30.2 in Pancreatic β-Cell against Glucotoxicity-Induced Apoptosis.
- Author
-
Ortega-Cuellar, Daniel, González-Sánchez, Ignacio, Piñón-Zárate, Gabriela, Cerbón, Marco A., De la Rosa, Víctor, Franco-Juárez, Yuliana, Castell-Rodríguez, Andrés, Islas, León D., and Coronel-Cruz, Cristina
- Subjects
- *
MEMBRANE proteins , *PANCREATIC beta cells , *TYPE 2 diabetes , *GENE expression , *ANNEXINS - Abstract
Simple Summary: Glucotoxicity is a condition that leads to β-cell death, driving a decrease in insulin secretion and leading to hyperglycemia and, eventually, diabetes. Here, we investigate whether connexin Cx30.2 could protect against glucotoxicity-induced apoptosis in β cells. We found that RIN-m5F β cells exposed to high glucose exhibited increased Cx30.2 protein expression. Interestingly, decreased expression of Cx30.2 by specific siRNAs resulted in augmented β-cell death, suggesting that Cx30.2 has an important role in maintaining cell survival during glucotoxicity. Overall, Cx30.2 could be an attractive pharmacological target to avoid high glucose-induced β-cell apoptosis. Glucotoxicity may exert its deleterious effects on pancreatic β-cell function via a myriad of mechanisms, leading to impaired insulin secretion and, eventually, type 2 diabetes. β-cell communication requires gap junction channels to be present among these cells. Gap junctions are constituted by transmembrane proteins of the connexins (Cxs) family. Two Cx genes have been identified in β cells, Cx36 and Cx30.2. We have found evidence that the glucose concentration on its own is sufficient to regulate Cx30.2 gene expression in mouse islets. In this work, we examine the involvement of the Cx30.2 protein in the survival of β cells (RIN-m5F). Methods: RIN-m5F cells were cultured in 5 mM D-glucose (normal) or 30 mM D-glucose (high glucose) for 24 h. Cx30.2 siRNAs was used to downregulate Cx30.2 expression. Apoptosis was measured by means of TUNEL, an annexin V staining method, and the cleaved form of the caspase-3 protein was determined using Western blot. Results: High glucose did not induce apoptosis in RIN-m5F β cells after 24 h; interestingly, high glucose increased the Cx30.2 total protein levels. Moreover, this work found that the downregulation of Cx30.2 expression in high glucose promoted apoptosis in RIN-m5F cells. Conclusion: The data suggest that the upregulation of Cx30.2 protects β cells from hyperglycemia-induced apoptosis. Furthermore, Cx30.2 may be a promising avenue of therapeutic investigation for the treatment of glucose metabolic disorders. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
19. The fate of intracellular S1P regulates lipid droplet turnover and lipotoxicity in pancreatic beta-cells
- Author
-
Yadi Tang, Mariola Majewska, Britta Leß, Ilir Mehmeti, Philipp Wollnitzke, Nina Semleit, Bodo Levkau, Julie D. Saba, Gerhild van Echten-Deckert, and Ewa Gurgul-Convey
- Subjects
diabetes ,beta-cells ,lipid droplets ,free fatty acids ,insulin-secreting cells ,sphingosine-1 phosphate ,Biochemistry ,QD415-436 - Abstract
Lipotoxicity has been considered the main cause of pancreatic beta-cell failure during type 2 diabetes development. Lipid droplets (LD) are believed to regulate the beta-cell sensitivity to free fatty acids (FFA), but the underlying molecular mechanisms are largely unclear. Accumulating evidence points, however, to an important role of intracellular sphingosine-1-phosphate (S1P) metabolism in lipotoxicity-mediated disturbances of beta-cell function. In the present study, we compared the effects of an increased irreversible S1P degradation (S1P-lyase, SPL overexpression) with those associated with an enhanced S1P recycling (overexpression of S1P phosphatase 1, SGPP1) on LD formation and lipotoxicity in rat INS1E beta-cells. Interestingly, although both approaches led to a reduced S1P concentration, they had opposite effects on the susceptibility to FFA. Overexpression of SGPP1 prevented FFA-mediated caspase-3 activation by a mechanism involving an enhanced lipid storage capacity and prevention of oxidative stress. In contrast, SPL overexpression limited LD biogenesis, content, and size, while accelerating lipophagy. This was associated with FFA-induced hydrogen peroxide formation, mitochondrial fragmentation, and dysfunction, as well as ER stress. These changes coincided with the upregulation of proapoptotic ceramides but were independent of lipid peroxidation rate. Also in human EndoC-βH1 beta-cells, suppression of SPL with simultaneous overexpression of SGPP1 led to a similar and even more pronounced LD phenotype as that in INS1E-SGPP1 cells. Thus, intracellular S1P turnover significantly regulates LD content and size and influences beta-cell sensitivity to FFA.
- Published
- 2024
- Full Text
- View/download PDF
20. Pharmacological conversion of gut epithelial cells into insulin-producing cells lowers glycemia in diabetic animals
- Author
-
Du, Wen, Wang, Junqiang, Kuo, Taiyi, Wang, Liheng, McKimpson, Wendy M, Son, Jinsook, Watanabe, Hitoshi, Kitamoto, Takumi, Lee, Yun-Kyoung, Creusot, Remi J, Ratner, Lloyd E, McCune, Kasi, Chen, Ya-Wen, Grubbs, Brendan H, Thornton, Matthew E, Fan, Jason, Sultana, Nishat, Diaz, Bryan S, Balasubramanian, Iyshwarya, Gao, Nan, Belvedere, Sandro, and Accili, Domenico
- Subjects
Diabetes ,Autoimmune Disease ,Digestive Diseases ,Aetiology ,2.1 Biological and endogenous factors ,Metabolic and endocrine ,Humans ,Mice ,Animals ,Forkhead Box Protein O1 ,Forkhead Transcription Factors ,Mice ,Inbred NOD ,Insulin-Secreting Cells ,Insulin ,Diabetes Mellitus ,Beta cells ,Endocrinology ,Medical and Health Sciences ,Immunology - Abstract
As a highly regenerative organ, the intestine is a promising source for cellular reprogramming for replacing lost pancreatic β cells in diabetes. Gut enterochromaffin cells can be converted to insulin-producing cells by forkhead box O1 (FoxO1) ablation, but their numbers are limited. In this study, we report that insulin-immunoreactive cells with Paneth/goblet cell features are present in human fetal intestine. Accordingly, lineage-tracing experiments show that, upon genetic or pharmacologic FoxO1 ablation, the Paneth/goblet lineage can also undergo conversion to the insulin lineage. We designed a screening platform in gut organoids to accurately quantitate β-like cell reprogramming and fine-tune a combination treatment to increase the efficiency of the conversion process in mice and human adult intestinal organoids. We identified a triple blockade of FOXO1, Notch, and TGF-β that, when tested in insulin-deficient streptozotocin (STZ) or NOD diabetic animals, resulted in near normalization of glucose levels, associated with the generation of intestinal insulin-producing cells. The findings illustrate a therapeutic approach for replacing insulin treatment in diabetes.
- Published
- 2022
21. Stem cell-based multi-tissue platforms to model human autoimmune diabetes
- Author
-
Leavens, Karla F, Alvarez-Dominguez, Juan R, Vo, Linda T, Russ, Holger A, and Parent, Audrey V
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Stem Cell Research - Embryonic - Human ,Regenerative Medicine ,Stem Cell Research ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Pediatric ,Autoimmune Disease ,Stem Cell Research - Induced Pluripotent Stem Cell ,Diabetes ,Metabolic and endocrine ,Humans ,Diabetes Mellitus ,Type 1 ,Pluripotent Stem Cells ,Insulin-Secreting Cells ,Induced Pluripotent Stem Cells ,Cell Differentiation ,Type 1 diabetes ,Autoimmunity ,Disease modeling ,Pluripotent stem cells ,Direct differentiation ,T cells ,Thymus ,Pancreatic &beta ,cells ,Genome engineering ,Pancreatic β cells ,Physiology ,Biochemistry and cell biology - Abstract
BackgroundType 1 diabetes (T1D) is an autoimmune disease in which pancreatic insulin-producing β cells are specifically destroyed by the immune system. Understanding the initiation and progression of human T1D has been hampered by the lack of appropriate models that can reproduce the complexity and heterogeneity of the disease. The development of platforms combining multiple human pluripotent stem cell (hPSC) derived tissues to model distinct aspects of T1D has the potential to provide critical novel insights into the etiology and pathogenesis of the human disease.Scope of reviewIn this review, we summarize the state of hPSC differentiation approaches to generate cell types and tissues relevant to T1D, with a particular focus on pancreatic islet cells, T cells, and thymic epithelium. We present current applications as well as limitations of using these hPSC-derived cells for disease modeling and discuss efforts to optimize platforms combining multiple cell types to model human T1D. Finally, we outline remaining challenges and emphasize future improvements needed to accelerate progress in this emerging field of research.Major conclusionsRecent advances in reprogramming approaches to create patient-specific induced pluripotent stem cell lines (iPSCs), genome engineering technologies to efficiently modify DNA of hPSCs, and protocols to direct their differentiation into mature cell types have empowered the use of stem cell derivatives to accurately model human disease. While challenges remain before complex interactions occurring in human T1D can be modeled with these derivatives, experiments combining hPSC-derived β cells and immune cells are already providing exciting insight into how these cells interact in the context of T1D, supporting the viability of this approach.
- Published
- 2022
22. Docosahexanoic Acid Attenuates Palmitate-Induced Apoptosis by Autophagy Upregulation via GPR120/mTOR Axis in Insulin-Secreting Cells
- Author
-
Seok-Woo Hong, Jinmi Lee, Sun Joon Moon, Hyemi Kwon, Se Eun Park, Eun-Jung Rhee, and Won-Young Lee
- Subjects
docosahexaenoic acids ,autophagy ,insulin-secreting cells ,g-protein-coupled receptor ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
Background Polyunsaturated fatty acids (PUFAs) reportedly have protective effects on pancreatic β-cells; however, the underlying mechanisms are unknown. Methods To investigate the cellular mechanism of PUFA-induced cell protection, mouse insulinoma 6 (MIN6) cells were cultured with palmitic acid (PA) and/or docosahexaenoic acid (DHA), and alterations in cellular signaling and apoptosis were examined. Results DHA treatment remarkably repressed caspase-3 cleavage and terminal deoxynucleotidyl transferase-mediated UTP nick end labeling (TUNEL)-positive red dot signals in PA-treated MIN6 cells, with upregulation of autophagy, an increase in microtubule-associated protein 1-light chain 3 (LC3)-II, autophagy-related 5 (Atg5), and decreased p62. Upstream factors involved in autophagy regulation (Beclin-1, unc51 like autophagy activating kinase 1 [ULK1], phosphorylated mammalian target of rapamycin [mTOR], and protein kinase B) were also altered by DHA treatment. DHA specifically induced phosphorylation on S2448 in mTOR; however, phosphorylation on S2481 decreased. The role of G protein-coupled receptor 120 (GPR120) in the effect of DHA was demonstrated using a GPR120 agonist and antagonist. Additional treatment with AH7614, a GPR120 antagonist, significantly attenuated DHA-induced autophagy and protection. Taken together, DHA-induced autophagy activation with protection against PA-induced apoptosis mediated by the GPR120/mTOR axis. Conclusion These findings indicate that DHA has therapeutic effects on PA-induced pancreatic β-cells, and that the cellular mechanism of β-cell protection by DHA may be a new research target with potential pharmacotherapeutic implications in β-cell protection.
- Published
- 2024
- Full Text
- View/download PDF
23. UDP-glucose, cereblon-dependent proinsulin degrader.
- Author
-
Cho, Jaeyong, Miyagawa, Atsushi, Yamaguchi, Kazuki, Abe, Wakana, Tsugawa, Yoji, Yamamura, Hatsuo, Imai, Takeshi, and Tsugawa, Yusuke
- Subjects
Animals ,Diabetes Mellitus ,Type 2 ,Glucose ,Insulin ,Insulin-Secreting Cells ,Mice ,Proinsulin ,Uridine ,Uridine Diphosphate Glucose - Abstract
Insulin secretion is regulated in multiple steps, and one of the main steps is in the endoplasmic reticulum (ER). Here, we show that UDP-glucose induces proinsulin ubiquitination by cereblon, and uridine binds and competes for proinsulin degradation and behaves as sustainable insulin secretagogue. Using insulin mutagenesis of neonatal diabetes variant-C43G and maturity-onset diabetes of the young 10 (MODY10) variant-R46Q, UDP-glucose:glycoprotein glucosyltransferase 1 (UGGT1) protects cereblon-dependent proinsulin ubiquitination in the ER. Cereblon is a ligand-inducible E3 ubiquitin ligase, and we found that UDP-glucose is the first identified endogenous proinsulin protein degrader. Uridine-containing compounds, such as uridine, UMP, UTP, and UDP-galactose, inhibit cereblon-dependent proinsulin degradation and stimulate insulin secretion from 3 to 24 h after administration in β-cell lines as well as mice. This late and long-term insulin secretion stimulation is designated a day sustainable insulin secretion stimulation. Uridine-containing compounds are designated as proinsulin degradation regulators.
- Published
- 2022
24. Insulin-degrading enzyme ablation in mouse pancreatic alpha cells triggers cell proliferation, hyperplasia and glucagon secretion dysregulation
- Author
-
Merino, Beatriz, Casanueva-Álvarez, Elena, Quesada, Iván, González-Casimiro, Carlos M, Fernández-Díaz, Cristina M, Postigo-Casado, Tamara, Leissring, Malcolm A, Kaestner, Klaus H, Perdomo, Germán, and Cózar-Castellano, Irene
- Subjects
Public Health ,Biomedical and Clinical Sciences ,Clinical Sciences ,Health Sciences ,Diabetes ,Nutrition ,1.1 Normal biological development and functioning ,Aetiology ,2.1 Biological and endogenous factors ,Underpinning research ,Metabolic and endocrine ,Animals ,Cell Proliferation ,Diabetes Mellitus ,Type 2 ,Glucagon ,Glucagon-Secreting Cells ,Hyperplasia ,Insulin ,Insulin-Secreting Cells ,Insulysin ,Mice ,alpha-Synuclein ,Alpha cells ,Cytoskeleton ,Hyperglucagonaemia ,Insulin-degrading enzyme ,Primary cilia ,Proliferation ,Type 2 diabetes ,Paediatrics and Reproductive Medicine ,Public Health and Health Services ,Endocrinology & Metabolism ,Clinical sciences ,Public health - Abstract
Type 2 diabetes is characterised by hyperglucagonaemia and perturbed function of pancreatic glucagon-secreting alpha cells but the molecular mechanisms contributing to these phenotypes are poorly understood. Insulin-degrading enzyme (IDE) is present within all islet cells, mostly in alpha cells, in both mice and humans. Furthermore, IDE can degrade glucagon as well as insulin, suggesting that IDE may play an important role in alpha cell function in vivo. We have generated and characterised a novel mouse model with alpha cell-specific deletion of Ide, the A-IDE-KO mouse line. Glucose metabolism and glucagon secretion in vivo was characterised; isolated islets were tested for glucagon and insulin secretion; alpha cell mass, alpha cell proliferation and α-synuclein levels were determined in pancreas sections by immunostaining. Targeted deletion of Ide exclusively in alpha cells triggers hyperglucagonaemia and alpha cell hyperplasia, resulting in elevated constitutive glucagon secretion. The hyperglucagonaemia is attributable in part to dysregulation of glucagon secretion, specifically an impaired ability of IDE-deficient alpha cells to suppress glucagon release in the presence of high glucose or insulin. IDE deficiency also leads to α-synuclein aggregation in alpha cells, which may contribute to impaired glucagon secretion via cytoskeletal dysfunction. We showed further that IDE deficiency triggers impairments in cilia formation, inducing alpha cell hyperplasia and possibly also contributing to dysregulated glucagon secretion and hyperglucagonaemia. We propose that loss of IDE function in alpha cells contributes to hyperglucagonaemia in type 2 diabetes.
- Published
- 2022
25. Covid-19 y diabetes mellitus tipo 2: implicaciones en las células beta pancreáticas.
- Author
-
Grisel Sánchez-Cervantes, Ivonne, González-Sánchez, Ignacio, Elena López-Martínez, Irma, Liliana AguirreBenítez, Elsa, and Coronel-Cruz, Cristina
- Abstract
The prevalence of chronic metabolic diseases in Mexico is high, being type 2 Diabetes mellitus (T2DM) as the most common disease. Several studies have shown that, compared with healthy individuals, patients with T2DM suffer a higher severity and mortality of Coronavirus disease 2019 (COVID-19). Therefore, it is important to the knowledge of the bidirectional relationship between these diseases. T2DM can increase SARS-CoV-2 virus pathogenicity in part due to metabolic disturbance. As a result, COVID-19 susceptibility and severity rise in diabetic individuals, which makes them a high-risk population. On the other hand, the infection caused by SARS-CoV-2 can lead individuals to hyperglycemia or new-onset diabetes. In order to understand the relationship between COVID-19 and T2DM, this review aims to emphasize the tropism of the SARS-CoV-2 virus to pancreatic beta-cells, as well as the physiologic effects of these. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
26. Oxidative and ER stress by elevated insulin biosynthesis and palmitic acid in insulin-producing cells.
- Author
-
Vidrio-Huerta, Brenda, Plötz, Thomas, and Lortz, Stephan
- Subjects
- *
PALMITIC acid , *INSULIN , *BIOSYNTHESIS , *TYPE 2 diabetes , *OXIDATIVE stress , *INSULIN receptors , *FLUORESCENT proteins , *PROTEIN folding - Abstract
The early phase of type 2 diabetes mellitus (T2DM) is characterised by insulin resistance, which can initially be compensated by elevated insulin secretion. However, as postulated by the workload hypothesis, over time harming insulin requirements contribute to β-cell dysfunction and death. The mechanisms behind this transition are complex and not fully understood but involve factors such as endoplasmic reticulum (ER) stress raised by gluco/lipotoxicity. To investigate the effect of excessive insulin folding on ER luminal H2O2 generation, ER stress and viability, insulin was expressed glucose-independently by a doxycycline-regulated Tet-On system in insulin-producing RINm5F cells. Additionally, the effect of palmitic acid (PA) as a subsidiary T2DM-associated factor was examined in this model system. Elevated insulin expression increased ER luminal H2O2 concentration quantified by the fluorescent sensor protein TriPer and reduced viability, but did not activate apoptosis. However, when combined with PA, insulin expression resulted in a significant increase in ER stress and apoptosis. Expression of ER-localised catalase verified the specificity of the applied H2O2 detection method without attenuating ER stress, caspase activation or viability loss. These findings suggest that hyperinsulinism alone can cause increased ER luminal H2O2 generation, mild ER stress and reduced viability, while hyperinsulinism in combination with PA accelerates these processes and triggers apoptosis. The inability of ER catalase to counteract these effects suggests that further damaging factors besides H2O2 are involved in cell dysfunction. Finally, reducing the high insulin demand in the initial phase of T2DM may be crucial in preventing further β-cell damage caused by gluco/lipotoxicity. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
27. β-Cell Succinate Dehydrogenase Deficiency Triggers Metabolic Dysfunction and Insulinopenic Diabetes
- Author
-
Lee, Sooyeon, Xu, Haixia, Van Vleck, Aidan, Mawla, Alex M, Li, Albert Mao, Ye, Jiangbin, Huising, Mark O, and Annes, Justin P
- Subjects
Biomedical and Clinical Sciences ,Diabetes ,Rare Diseases ,Nutrition ,2.1 Biological and endogenous factors ,Metabolic and endocrine ,Animals ,Diabetes Mellitus ,Type 2 ,Electron Transport Complex II ,Glucose ,Insulin-Secreting Cells ,Metabolism ,Inborn Errors ,Mice ,Mitochondrial Diseases ,Succinate Dehydrogenase ,TOR Serine-Threonine Kinases ,Medical and Health Sciences ,Endocrinology & Metabolism ,Biomedical and clinical sciences - Abstract
Mitochondrial dysfunction plays a central role in type 2 diabetes (T2D); however, the pathogenic mechanisms in pancreatic β-cells are incompletely elucidated. Succinate dehydrogenase (SDH) is a key mitochondrial enzyme with dual functions in the tricarboxylic acid cycle and electron transport chain. Using samples from human with diabetes and a mouse model of β-cell-specific SDH ablation (SDHBβKO), we define SDH deficiency as a driver of mitochondrial dysfunction in β-cell failure and insulinopenic diabetes. β-Cell SDH deficiency impairs glucose-induced respiratory oxidative phosphorylation and mitochondrial membrane potential collapse, thereby compromising glucose-stimulated ATP production, insulin secretion, and β-cell growth. Mechanistically, metabolomic and transcriptomic studies reveal that the loss of SDH causes excess succinate accumulation, which inappropriately activates mammalian target of rapamycin (mTOR) complex 1-regulated metabolic anabolism, including increased SREBP-regulated lipid synthesis. These alterations, which mirror diabetes-associated human β-cell dysfunction, are partially reversed by acute mTOR inhibition with rapamycin. We propose SDH deficiency as a contributing mechanism to the progressive β-cell failure of diabetes and identify mTOR complex 1 inhibition as a potential mitigation strategy.
- Published
- 2022
28. Pancreatic β-Cell Development and Regeneration.
- Author
-
Kerper, Natanya, Ashe, Sudipta, and Hebrok, Matthias
- Subjects
Cell Differentiation ,Diabetes Mellitus ,Humans ,Insulin ,Insulin-Secreting Cells - Abstract
The pancreatic β-cells are essential for regulating glucose homeostasis through the coordinated release of the insulin hormone. Dysfunction of the highly specialized β-cells results in diabetes mellitus, a growing global health epidemic. In this review, we describe the development and function of β-cells the emerging concept of heterogeneity within insulin-producing cells, and the potential of other cell types to assume β-cell functionality via transdifferentiation. We also discuss emerging routes to design cells with minimal β-cell properties and human stem cell differentiation efforts that carry the promise to restore normoglycemia in patients suffering from diabetes.
- Published
- 2022
29. Urocortin3: Local inducer of somatostatin release and bellwether of beta cell maturity
- Author
-
Flisher, Marcus F, Shin, Donghan, and Huising, Mark O
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Clinical Sciences ,Biological Sciences ,Autoimmune Disease ,Diabetes ,1.1 Normal biological development and functioning ,Metabolic and endocrine ,Generic health relevance ,Animals ,Corticotropin-Releasing Hormone ,Humans ,Insulin ,Insulin Secretion ,Insulin-Secreting Cells ,Islets of Langerhans ,Mice ,Somatostatin ,Urocortins ,Pancreatic islet ,UCN3 ,CRH ,Glucagon ,SST ,Beta cell maturity ,Medicinal and Biomolecular Chemistry ,Physiology ,Pharmacology and Pharmaceutical Sciences ,Endocrinology & Metabolism ,Biochemistry and cell biology ,Clinical sciences ,Medicinal and biomolecular chemistry - Abstract
Urocortin 3 (UCN3) is a peptide hormone expressed in pancreatic islets of Langerhans of both human alpha and human beta cells and solely in murine beta cells. UCN3 signaling acts locally within the islet to activate its cognate receptor, corticotropin releasing hormone receptor 2 (CRHR2), which is expressed by delta cells, to potentiate somatostatin (SST) negative feedback to reduce islet cell hormone output. The functional importance of UCN3 signaling in the islet is to modulate the amount of SST tone allowing for finely tuned regulation of insulin and glucagon secretion. UCN3 signaling is a hallmark of functional beta cell maturation, increasing the beta cell glucose threshold for insulin secretion. In doing so, UCN3 plays a relevant functional role in accurately maintaining blood glucose homeostasis. Additionally, UCN3 acts as an indicator of beta cell maturation and health, as UCN3 is not expressed in immature beta cells and is downregulated in dedifferentiated and dysfunctional beta cell states. Here, we review the mechanistic underpinnings of UCN3 signaling, its net effect on islet cell hormone output, as well as its value as a marker for beta cell maturation and functional status.
- Published
- 2022
30. Development of a scalable method to isolate subsets of stem cell-derived pancreatic islet cells
- Author
-
Parent, Audrey V, Ashe, Sudipta, Nair, Gopika G, Li, Mei-Lan, Chavez, Jessica, Liu, Jennifer S, Zhong, Yongping, Streeter, Philip R, and Hebrok, Matthias
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Stem Cell Research ,Biotechnology ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Stem Cell Research - Nonembryonic - Human ,Regenerative Medicine ,Stem Cell Research - Embryonic - Human ,Stem Cell Research - Nonembryonic - Non-Human ,Transplantation ,Diabetes ,Stem Cell Research - Induced Pluripotent Stem Cell ,Autoimmune Disease ,5.2 Cellular and gene therapies ,Metabolic and endocrine ,Cell Differentiation ,Glucose ,Humans ,Insulin ,Insulin Secretion ,Insulin-Secreting Cells ,Islets of Langerhans ,Pluripotent Stem Cells ,cell therapy ,diabetes ,directed differentiation ,pancreatic beta cells ,regenerative medicine ,Clinical Sciences ,Biochemistry and cell biology - Abstract
Cell replacement therapy using β cells derived from stem cells is a promising alternative to conventional diabetes treatment options. Although current differentiation methods produce glucose-responsive β cells, they can also yield populations of undesired endocrine progenitors and other proliferating cell types that might interfere with long-term islet function and safety of transplanted cells. Here, we describe the generation of an array of monoclonal antibodies against cell surface markers that selectively label stem cell-derived islet cells. A high-throughput screen identified promising candidates, including three clones that mark a high proportion of endocrine cells in differentiated cultures. A scalable magnetic sorting method was developed to enrich for human pluripotent stem cell (hPSC)-derived islet cells using these three antibodies, leading to the formation of islet-like clusters with improved glucose-stimulated insulin secretion and reduced growth upon transplantation. This strategy should facilitate large-scale production of functional islet clusters from stem cells for disease modeling and cell replacement therapy.
- Published
- 2022
31. Modeling human T1D-associated autoimmune processes.
- Author
-
Khosravi-Maharlooei, Mohsen, Madley, Rachel, Borsotti, Chiara, Ferreira, Leonardo MR, Sharp, Robert C, Brehm, Michael A, Greiner, Dale L, Parent, Audrey V, Anderson, Mark S, Sykes, Megan, and Creusot, Remi J
- Subjects
Immune System ,Animals ,Humans ,Mice ,Diabetes Mellitus ,Type 1 ,Insulin-Secreting Cells ,Autoimmunity ,Beta cell destruction ,Disease modeling ,Humanized mice ,In vitro models ,Type 1 diabetes ,Clinical Research ,Autoimmune Disease ,Pediatric ,Prevention ,Diabetes ,Genetics ,Aetiology ,2.1 Biological and endogenous factors ,Inflammatory and immune system ,Metabolic and endocrine ,In vitro models ,Biochemistry and Cell Biology ,Physiology - Abstract
BackgroundType 1 diabetes (T1D) is an autoimmune disease characterized by impaired immune tolerance to β-cell antigens and progressive destruction of insulin-producing β-cells. Animal models have provided valuable insights for understanding the etiology and pathogenesis of this disease, but they fall short of reflecting the extensive heterogeneity of the disease in humans, which is contributed by various combinations of risk gene alleles and unique environmental factors. Collectively, these factors have been used to define subgroups of patients, termed endotypes, with distinct predominating disease characteristics.Scope of reviewHere, we review the gaps filled by these models in understanding the intricate involvement and regulation of the immune system in human T1D pathogenesis. We describe the various models developed so far and the scientific questions that have been addressed using them. Finally, we discuss the limitations of these models, primarily ascribed to hosting a human immune system (HIS) in a xenogeneic recipient, and what remains to be done to improve their physiological relevance.Major conclusionsTo understand the role of genetic and environmental factors or evaluate immune-modifying therapies in humans, it is critical to develop and apply models in which human cells can be manipulated and their functions studied under conditions that recapitulate as closely as possible the physiological conditions of the human body. While microphysiological systems and living tissue slices provide some of these conditions, HIS mice enable more extensive analyses using in vivo systems.
- Published
- 2022
32. Deletion of ABCB10 in beta-cells protects from high-fat diet induced insulin resistance
- Author
-
Shum, Michael, Segawa, Mayuko, Gharakhanian, Raffi, Viñuela, Ana, Wortham, Matthew, Baghdasarian, Siyouneh, Wolf, Dane M, Sereda, Samuel B, Nocito, Laura, Stiles, Linsey, Zhou, Zhiqiang, Gutierrez, Vincent, Sander, Maike, Shirihai, Orian S, and Liesa, Marc
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Obesity ,Nutrition ,Genetics ,Diabetes ,2.1 Biological and endogenous factors ,Metabolic and endocrine ,ATP-Binding Cassette Transporters ,Animals ,Blood Glucose ,Diabetes Mellitus ,Type 2 ,Diet ,High-Fat ,Female ,Glucose ,Glucose Tolerance Test ,Insulin ,Insulin Resistance ,Insulin Secretion ,Insulin-Secreting Cells ,Islets of Langerhans ,Male ,Mice ,Mice ,Inbred C57BL ,Mice ,Knockout ,Mitochondria ,Insulin resistance ,Beta-cell ,ABCB10 ,Physiology ,Biochemistry and cell biology - Abstract
ObjectiveThe contribution of beta-cell dysfunction to type 2 diabetes (T2D) is not restricted to insulinopenia in the late stages of the disease. Elevated fasting insulinemia in normoglycemic humans is a major factor predicting the onset of insulin resistance and T2D, demonstrating an early alteration of beta-cell function in T2D. Moreover, an early and chronic increase in fasting insulinemia contributes to insulin resistance in high-fat diet (HFD)-fed mice. However, whether there are genetic factors that promote beta-cell-initiated insulin resistance remains undefined. Human variants of the mitochondrial transporter ABCB10, which regulates redox by increasing bilirubin synthesis, have been associated with an elevated risk of T2D. The effects of T2D ABCB10 variants on ABCB10 expression and the actions of ABCB10 in beta-cells are unknown.MethodsThe expression of beta-cell ABCB10 was analyzed in published transcriptome datasets from human beta-cells carrying the T2D-risk ABCB10 variant. Insulin sensitivity, beta-cell proliferation, and secretory function were measured in beta-cell-specific ABCB10 KO mice (Ins1Cre-Abcb10flox/flox). The short-term role of beta-cell ABCB10 activity on glucose-stimulated insulin secretion (GSIS) was determined in isolated islets.ResultsCarrying the T2Drisk allele G of ABCB10 rs348330 variant was associated with increased ABCB10 expression in human beta-cells. Constitutive deletion of Abcb10 in beta-cells protected mice from hyperinsulinemia and insulin resistance by limiting HFD-induced beta-cell expansion. An early limitation in GSIS and H2O2-mediated signaling caused by elevated ABCB10 activity can initiate an over-compensatory expansion of beta-cell mass in response to HFD. Accordingly, increasing ABCB10 expression was sufficient to limit GSIS capacity. In health, ABCB10 protein was decreased during islet maturation, with maturation restricting beta-cell proliferation and elevating GSIS. Finally, ex-vivo and short-term deletion of ABCB10 in islets isolated from HFD-fed mice increased H2O2 and GSIS, which was reversed by bilirubin treatments.ConclusionsBeta-cell ABCB10 is required for HFD to induce insulin resistance in mice by amplifying beta-cell mass expansion to maladaptive levels that cause fasting hyperinsulinemia.
- Published
- 2022
33. IAPP-induced beta cell stress recapitulates the islet transcriptome in type 2 diabetes.
- Author
-
Blencowe, Montgomery, Furterer, Allison, Wang, Qing, Gao, Fuying, Rosenberger, Madeline, Pei, Lina, Nomoto, Hiroshi, Mawla, Alex M, Huising, Mark O, Coppola, Giovanni, Yang, Xia, Butler, Peter C, and Gurlo, Tatyana
- Subjects
Islets of Langerhans ,Animals ,Mice ,Transgenic ,Mice ,Diabetes Mellitus ,Type 2 ,Amyloid ,Insulin-Secreting Cells ,Islet Amyloid Polypeptide ,Transcriptome ,Beta cell ,Cell cycle ,Dedifferentiation ,Inflammation ,Islet amyloid polypeptide ,Prediabetes ,Protein misfolding ,Type 2 diabetes ,Unfolded protein response ,Genetics ,Diabetes ,Biotechnology ,2.1 Biological and endogenous factors ,Aetiology ,Metabolic and endocrine ,Clinical Sciences ,Paediatrics and Reproductive Medicine ,Public Health and Health Services ,Endocrinology & Metabolism - Abstract
Aims/hypothesisType 2 diabetes is characterised by islet amyloid and toxic oligomers of islet amyloid polypeptide (IAPP). We posed the questions, (1) does IAPP toxicity induce an islet response comparable to that in humans with type 2 diabetes, and if so, (2) what are the key transcriptional drivers of this response?MethodsThe islet transcriptome was evaluated in five groups of mice: beta cell specific transgenic for (1) human IAPP, (2) rodent IAPP, (3) human calpastatin, (4) human calpastatin and human IAPP, and (5) wild-type mice. RNA sequencing data was analysed by differential expression analysis and gene co-expression network analysis to establish the islet response to adaptation to an increased beta cell workload of soluble rodent IAPP, the islet response to increased expression of oligomeric human IAPP, and the extent to which the latter was rescued by suppression of calpain hyperactivation by calpastatin. Rank-rank hypergeometric overlap analysis was used to compare the transcriptome of islets from human or rodent IAPP transgenic mice vs humans with prediabetes or type 2 diabetes.ResultsThe islet transcriptomes in humans with prediabetes and type 2 diabetes are remarkably similar. Beta cell overexpression of soluble rodent or oligomer-prone human IAPP induced changes in islet transcriptome present in prediabetes and type 2 diabetes, including decreased expression of genes that confer beta cell identity. Increased expression of human IAPP, but not rodent IAPP, induced islet inflammation present in prediabetes and type 2 diabetes in humans. Key mediators of the injury responses in islets transgenic for human IAPP or those from individuals with type 2 diabetes include STAT3, NF-κB, ESR1 and CTNNB1 by transcription factor analysis and COL3A1, NID1 and ZNF800 by gene regulatory network analysis.Conclusions/interpretationBeta cell injury mediated by IAPP is a plausible mechanism to contribute to islet inflammation and dedifferentiation in type 2 diabetes. Inhibition of IAPP toxicity is a potential therapeutic target in type 2 diabetes.
- Published
- 2022
34. Small molecule SWELL1 complex induction improves glycemic control and nonalcoholic fatty liver disease in murine Type 2 diabetes
- Author
-
Gunasekar, Susheel K, Xie, Litao, Kumar, Ashutosh, Hong, Juan, Chheda, Pratik R, Kang, Chen, Kern, David M, My-Ta, Chau, Maurer, Joshua, Heebink, John, Gerber, Eva E, Grzesik, Wojciech J, Elliot-Hudson, Macaulay, Zhang, Yanhui, Key, Phillip, Kulkarni, Chaitanya A, Beals, Joseph W, Smith, Gordon I, Samuel, Isaac, Smith, Jessica K, Nau, Peter, Imai, Yumi, Sheldon, Ryan D, Taylor, Eric B, Lerner, Daniel J, Norris, Andrew W, Klein, Samuel, Brohawn, Stephen G, Kerns, Robert, and Sah, Rajan
- Subjects
Autoimmune Disease ,Digestive Diseases ,Diabetes ,Chronic Liver Disease and Cirrhosis ,Liver Disease ,Nutrition ,Development of treatments and therapeutic interventions ,5.1 Pharmaceuticals ,2.1 Biological and endogenous factors ,Aetiology ,Metabolic and endocrine ,Adipose Tissue ,Animals ,Cryoelectron Microscopy ,Diabetes Mellitus ,Experimental ,Diabetes Mellitus ,Type 2 ,Glucose ,Glycemic Control ,Insulin ,Insulin Resistance ,Insulin Secretion ,Insulin-Secreting Cells ,Liver ,Male ,Membrane Proteins ,Mice ,Mice ,Inbred C57BL ,Molecular Docking Simulation ,Non-alcoholic Fatty Liver Disease ,Signal Transduction ,Transcriptome - Abstract
Type 2 diabetes is associated with insulin resistance, impaired pancreatic β-cell insulin secretion, and nonalcoholic fatty liver disease. Tissue-specific SWELL1 ablation impairs insulin signaling in adipose, skeletal muscle, and endothelium, and impairs β-cell insulin secretion and glycemic control. Here, we show that ICl,SWELL and SWELL1 protein are reduced in adipose and β-cells in murine and human diabetes. Combining cryo-electron microscopy, molecular docking, medicinal chemistry, and functional studies, we define a structure activity relationship to rationally-design active derivatives of a SWELL1 channel inhibitor (DCPIB/SN-401), that bind the SWELL1 hexameric complex, restore SWELL1 protein, plasma membrane trafficking, signaling, glycemic control and islet insulin secretion via SWELL1-dependent mechanisms. In vivo, SN-401 restores glycemic control, reduces hepatic steatosis/injury, improves insulin-sensitivity and insulin secretion in murine diabetes. These findings demonstrate that SWELL1 channel modulators improve SWELL1-dependent systemic metabolism in Type 2 diabetes, representing a first-in-class therapeutic approach for diabetes and nonalcoholic fatty liver disease.
- Published
- 2022
35. Accumulation of microbial DNAs promotes to islet inflammation and β cell abnormalities in obesity in mice
- Author
-
Gao, Hong, Luo, Zhenlong, Ji, Yudong, Tang, Kechun, Jin, Zhongmou, Ly, Crystal, Sears, Dorothy D, Mahata, Sushil, and Ying, Wei
- Subjects
Microbiology ,Biological Sciences ,Biomedical and Clinical Sciences ,Nutrition ,Obesity ,Prevention ,2.1 Biological and endogenous factors ,Metabolic and endocrine ,Oral and gastrointestinal ,Animals ,DNA ,Bacterial ,Diet ,High-Fat ,Extracellular Vesicles ,Gastrointestinal Microbiome ,Humans ,Inflammation ,Insulin Secretion ,Insulin-Secreting Cells ,Islets of Langerhans ,Macrophages ,Membrane Proteins ,Mice ,Inbred C57BL ,Mice ,Knockout ,Mice ,Transgenic ,Nucleotidyltransferases ,Receptors ,Complement ,Signal Transduction ,Mice - Abstract
Various microbial products leaked from gut lumen exacerbate tissue inflammation and metabolic disorders in obesity. Vsig4+ macrophages are key players preventing infiltration of bacteria and their products into host tissues. However, roles of islet Vsig4+ macrophages in the communication between microbiota and β cells in pathogenesis of obesity-associated islet abnormalities are unknown. Here, we find that bacterial DNAs are enriched in β cells of individuals with obesity. Intestinal microbial DNA-containing extracellular vesicles (mEVs) readily pass through obese gut barrier and deliver microbial DNAs into β cells, resulting in elevated inflammation and impaired insulin secretion by triggering cGAS/STING activation. Vsig4+ macrophages prevent mEV infiltration into β cells through a C3-dependent opsonization, whereas loss of Vsig4 leads to microbial DNA enrichment in β cells after mEV treatment. Removal of microbial DNAs blunts mEV effects. Loss of Vsig4+ macrophages leads to microbial DNA accumulation in β cells and subsequently obesity-associated islet abnormalities.
- Published
- 2022
36. What is a β cell? – Chapter I in the Human Islet Research Network (HIRN) review series
- Author
-
Kaestner, Klaus H, Campbell–Thompson, Martha, Dor, Yuval, Gill, Ronald G, Glaser, Benjamin, Kim, Seung K, Sander, Maike, Stabler, Cherie, Stewart, Andrew F, and Powers, Alvin C
- Subjects
Autoimmune Disease ,Diabetes ,Metabolic and endocrine ,Diabetes Mellitus ,Type 1 ,Diabetes Mellitus ,Type 2 ,Humans ,Insulin ,Insulin-Secreting Cells ,Beta cell ,Islet ,Islet transplantation ,Biochemistry and Cell Biology ,Physiology - Abstract
BackgroundThe pancreatic β cell, as the sole source of the vital hormone insulin, has been under intensive study for more than a century. Given the potential of newly created insulin-producing cells as a treatment or even cure of type 1 diabetes (T1D) and possibly in severe cases of type 2 diabetes (T2D), multiple academic and commercial laboratories are working to derive surrogate glucose-responsive, insulin-producing cells.Scope of reviewThe recent development of advanced phenotyping technologies, including molecular, epigenomic, histological, or functional, have greatly improved our understanding of the critical properties of human β cells. Using this information, here we summarize the salient features of normal, fully functional adult human β cells, and propose minimal criteria for what should rightfully be termed 'β cells' as opposed to insulin-producing but not fully-functional surrogates that we propose should be referred to as 'β-like' cells or insulin-producing cells.Major conclusionsClear criteria can be established to differentiate fully functional, mature β cells from 'β-like' surrogates. In addition, we outline important knowledge gaps that must be addressed to enable a greater understanding of the β cell.
- Published
- 2021
37. Antagonistic epistasis of Hnf4α and FoxO1 metabolic networks through enhancer interactions in β-cell function
- Author
-
Kuo, Taiyi, Du, Wen, Miyachi, Yasutaka, Dadi, Prasanna K, Jacobson, David A, Segrè, Daniel, and Accili, Domenico
- Subjects
Biochemistry and Cell Biology ,Genetics ,Biological Sciences ,Diabetes ,Human Genome ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning ,Animals ,Epistasis ,Genetic ,Forkhead Box Protein O1 ,Hepatocyte Nuclear Factor 4 ,Insulin-Secreting Cells ,Mice ,Mice ,Knockout ,Mutation ,Beta cell ,Hnf4a ,Foxo1 ,Enhancer ,Insulin ,Epistasis ,Physiology ,Biochemistry and cell biology - Abstract
ObjectiveGenetic and acquired abnormalities contribute to pancreatic β-cell failure in diabetes. Transcription factors Hnf4α (MODY1) and FoxO1 are respective examples of these two components and act through β-cell-specific enhancers. However, their relationship is unclear.MethodsIn this report, we show by genome-wide interrogation of chromatin modifications that ablation of FoxO1 in mature β-cells enriches active Hnf4α enhancers according to a HOMER analysis.ResultsTo model the functional significance of this predicted unusual enhancer utilization, we generated single and compound knockouts of FoxO1 and Hnf4α in β-cells. Single knockout of either gene impaired insulin secretion in mechanistically distinct fashions as indicated by their responses to sulfonylurea and calcium fluxes. Surprisingly, the defective β-cell secretory function of either single mutant in hyperglycemic clamps and isolated islets treated with various secretagogues was completely reversed in double mutants lacking FoxO1 and Hnf4α. Gene expression analyses revealed distinct epistatic modalities by which the two transcription factors regulate networks associated with reversal of β-cell dysfunction. An antagonistic network regulating glycolysis, including β-cell "disallowed" genes, and a synergistic network regulating protocadherins emerged as likely mediators of the functional restoration of insulin secretion.ConclusionsThe findings provide evidence of antagonistic epistasis as a model of gene/environment interactions in the pathogenesis of β-cell dysfunction.
- Published
- 2021
38. Restoring normal islet mass and function in type 1 diabetes through regenerative medicine and tissue engineering
- Author
-
Krentz, Nicole AJ, Shea, Lonnie D, Huising, Mark O, and Shaw, James AM
- Subjects
Medical Biotechnology ,Biomedical and Clinical Sciences ,Regenerative Medicine ,Diabetes ,Transplantation ,Bioengineering ,Stem Cell Research ,Autoimmune Disease ,5.2 Cellular and gene therapies ,Metabolic and endocrine ,Quality Education ,Animals ,Cell Count ,Cell Proliferation ,Diabetes Mellitus ,Type 1 ,History ,21st Century ,Humans ,Insulin-Secreting Cells ,Islets of Langerhans ,Islets of Langerhans Transplantation ,Regeneration ,Tissue Engineering ,Medical Biochemistry and Metabolomics ,Clinical Sciences ,Public Health and Health Services ,Clinical sciences ,Medical biochemistry and metabolomics - Abstract
Type 1 diabetes is characterised by autoimmune-mediated destruction of pancreatic β-cell mass. With the advent of insulin therapy a century ago, type 1 diabetes changed from a progressive, fatal disease to one that requires lifelong complex self-management. Replacing the lost β-cell mass through transplantation has proven successful, but limited donor supply and need for lifelong immunosuppression restricts widespread use. In this Review, we highlight incremental advances over the past 20 years and remaining challenges in regenerative medicine approaches to restoring β-cell mass and function in type 1 diabetes. We begin by summarising the role of endocrine islets in glucose homoeostasis and how this is altered in disease. We then discuss the potential regenerative capacity of the remaining islet cells and the utility of stem cell-derived β-like cells to restore β-cell function. We conclude with tissue engineering approaches that might improve the engraftment, function, and survival of β-cell replacement therapies.
- Published
- 2021
39. Glucagon Potentiates Insulin Secretion Via β-Cell GCGR at Physiological Concentrations of Glucose.
- Author
-
Zhang, Yulin, Han, Chengsheng, Zhu, Wenzhen, Yang, Guoyi, Peng, Xiaohong, Mehta, Sohum, Zhang, Jin, Chen, Liangyi, and Liu, Yanmei
- Subjects
GCGR ,GLP-1R ,cAMP ,glucagon ,glucose-stimulated insulin secretion ,α-cells ,β-cells ,Animals ,Glucagon ,Glucagon-Like Peptide 1 ,Glucagon-Like Peptide-1 Receptor ,Glucose ,Glucose Intolerance ,Insulin ,Insulin Secretion ,Insulin-Secreting Cells ,Male ,Mice ,Mice ,Inbred C57BL ,Mice ,Knockout ,Receptors ,Glucagon ,Signal Transduction - Abstract
Incretin-potentiated glucose-stimulated insulin secretion (GSIS) is critical to maintaining euglycemia, of which GLP-1 receptor (GLP-1R) on β-cells plays an indispensable role. Recently, α-cell-derived glucagon but not intestine-derived GLP-1 has been proposed as the critical hormone that potentiates GSIS via GLP-1R. However, the function of glucagon receptors (GCGR) on β-cells remains elusive. Here, using GCGR or GLP-1R antagonists, in combination with glucagon, to treat single β-cells, α-β cell clusters and isolated islets, we found that glucagon potentiates insulin secretion via β-cell GCGR at physiological but not high concentrations of glucose. Furthermore, we transfected primary mouse β-cells with RAB-ICUE (a genetically encoded cAMP fluorescence indicator) to monitor cAMP level after glucose stimulation and GCGR activation. Using specific inhibitors of different adenylyl cyclase (AC) family members, we revealed that high glucose concentration or GCGR activation independently evoked cAMP elevation via AC5 in β-cells, thus high glucose stimulation bypassed GCGR in promoting insulin secretion. Additionally, we generated β-cell-specific GCGR knockout mice which glucose intolerance was more severe when fed a high-fat diet (HFD). We further found that β-cell GCGR activation promoted GSIS more than GLP-1R in HFD, indicating the critical role of GCGR in maintaining glucose homeostasis during nutrient overload.
- Published
- 2021
40. SARS-CoV-2 infects human pancreatic β cells and elicits β cell impairment
- Author
-
Wu, Chien-Ting, Lidsky, Peter V, Xiao, Yinghong, Lee, Ivan T, Cheng, Ran, Nakayama, Tsuguhisa, Jiang, Sizun, Demeter, Janos, Bevacqua, Romina J, Chang, Charles A, Whitener, Robert L, Stalder, Anna K, Zhu, Bokai, Chen, Han, Goltsev, Yury, Tzankov, Alexandar, Nayak, Jayakar V, Nolan, Garry P, Matter, Matthias S, Andino, Raul, and Jackson, Peter K
- Subjects
Diabetes ,Pneumonia & Influenza ,Prevention ,Lung ,Autoimmune Disease ,Pneumonia ,Infectious Diseases ,Emerging Infectious Diseases ,Clinical Research ,Aetiology ,2.1 Biological and endogenous factors ,Metabolic and endocrine ,Good Health and Well Being ,A549 Cells ,Adult ,Aged ,Aged ,80 and over ,Angiotensin-Converting Enzyme 2 ,Antigens ,CD ,Apoptosis ,Apoptosis Regulatory Proteins ,COVID-19 ,Case-Control Studies ,Diabetes Mellitus ,Female ,Host-Pathogen Interactions ,Humans ,Insulin ,Insulin-Secreting Cells ,Male ,Middle Aged ,Neuropilin-1 ,Receptors ,Transferrin ,Receptors ,Virus ,SARS-CoV-2 ,Serine Endopeptidases ,Spike Glycoprotein ,Coronavirus ,Virus Internalization ,ACE2 ,SARS-CoV-2 spike protein ,apoptosis ,insulin ,neuropilin 1 ,pancreatic beta cell ,phosphoproteomics ,type 1 diabetes ,Biochemistry and Cell Biology ,Medical Biochemistry and Metabolomics ,Endocrinology & Metabolism - Abstract
Emerging evidence points toward an intricate relationship between the pandemic of coronavirus disease 2019 (COVID-19) and diabetes. While preexisting diabetes is associated with severe COVID-19, it is unclear whether COVID-19 severity is a cause or consequence of diabetes. To mechanistically link COVID-19 to diabetes, we tested whether insulin-producing pancreatic β cells can be infected by SARS-CoV-2 and cause β cell depletion. We found that the SARS-CoV-2 receptor, ACE2, and related entry factors (TMPRSS2, NRP1, and TRFC) are expressed in β cells, with selectively high expression of NRP1. We discovered that SARS-CoV-2 infects human pancreatic β cells in patients who succumbed to COVID-19 and selectively infects human islet β cells in vitro. We demonstrated that SARS-CoV-2 infection attenuates pancreatic insulin levels and secretion and induces β cell apoptosis, each rescued by NRP1 inhibition. Phosphoproteomic pathway analysis of infected islets indicates apoptotic β cell signaling, similar to that observed in type 1 diabetes (T1D). In summary, our study shows SARS-CoV-2 can directly induce β cell killing.
- Published
- 2021
41. Chop/Ddit3 depletion in β cells alleviates ER stress and corrects hepatic steatosis in mice
- Author
-
Yong, Jing, Parekh, Vishal S, Reilly, Shannon M, Nayak, Jonamani, Chen, Zhouji, Lebeaupin, Cynthia, Jang, Insook, Zhang, Jiangwei, Prakash, Thazha P, Sun, Hong, Murray, Sue, Guo, Shuling, Ayala, Julio E, Satin, Leslie S, Saltiel, Alan R, and Kaufman, Randal J
- Subjects
Medical Biotechnology ,Biomedical and Clinical Sciences ,Liver Disease ,Genetics ,Chronic Liver Disease and Cirrhosis ,Digestive Diseases ,Diabetes ,Nutrition ,2.1 Biological and endogenous factors ,Metabolic and endocrine ,Animals ,Diabetes Mellitus ,Type 2 ,Diet ,High-Fat ,Endoplasmic Reticulum Stress ,Fatty Liver ,Insulin ,Insulin Secretion ,Insulin-Secreting Cells ,Mice ,Mice ,Inbred C57BL ,Biological Sciences ,Medical and Health Sciences ,Medical biotechnology ,Biomedical engineering - Abstract
Type 2 diabetes (T2D) is a metabolic disorder characterized by hyperglycemia, hyperinsulinemia, and insulin resistance (IR). During the early phase of T2D, insulin synthesis and secretion by pancreatic β cells is enhanced, which can lead to proinsulin misfolding that aggravates endoplasmic reticulum (ER) protein homeostasis in β cells. Moreover, increased circulating insulin may contribute to fatty liver disease. Medical interventions aimed at alleviating ER stress in β cells while maintaining optimal insulin secretion are therefore an attractive therapeutic strategy for T2D. Previously, we demonstrated that germline Chop gene deletion preserved β cells in high-fat diet (HFD)-fed mice and in leptin receptor-deficient db/db mice. In the current study, we further investigated whether targeting Chop/Ddit3 specifically in murine β cells conferred therapeutic benefits. First, we showed that Chop deletion in β cells alleviated β cell ER stress and delayed glucose-stimulated insulin secretion (GSIS) in HFD-fed mice. Second, β cell-specific Chop deletion prevented liver steatosis and hepatomegaly in aged HFD-fed mice without affecting basal glucose homeostasis. Third, we provide mechanistic evidence that Chop depletion reduces ER Ca2+ buffering capacity and modulates glucose-induced islet Ca2+ oscillations, leading to transcriptional changes of ER chaperone profile ("ER remodeling"). Last, we demonstrated that a GLP1-conjugated Chop antisense oligonucleotide strategy recapitulated the reduction in liver triglycerides and pancreatic insulin content. In summary, our results demonstrate that Chop depletion in β cells provides a therapeutic strategy to alleviate dysregulated insulin secretion and consequent fatty liver disease in T2D.
- Published
- 2021
42. Transcriptional mechanisms of pancreatic β-cell maturation and functional adaptation
- Author
-
Wortham, Matthew and Sander, Maike
- Subjects
Reproductive Medicine ,Biomedical and Clinical Sciences ,Genetics ,Diabetes ,Stem Cell Research ,Stem Cell Research - Nonembryonic - Non-Human ,Nutrition ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Underpinning research ,Aetiology ,Metabolic and endocrine ,Cell Differentiation ,Insulin ,Insulin Secretion ,Insulin-Secreting Cells ,Stem Cells ,adaptation ,insulin secretion ,maturation ,metabolism ,transcription factors ,β-cell ,Clinical Sciences ,Paediatrics and Reproductive Medicine ,Endocrinology & Metabolism ,Reproductive medicine - Abstract
Pancreatic β-cells secrete insulin commensurate to circulating nutrient levels to maintain normoglycemia. The ability of β-cells to couple insulin secretion to nutrient stimuli is acquired during a postnatal maturation process. In mature β-cells the insulin secretory response adapts to changes in nutrient state. Both β-cell maturation and functional adaptation rely on the interplay between extracellular cues and cell type-specific transcriptional programs. Here we review emerging evidence that developmental and homeostatic regulation of β-cell function involves collaboration between lineage-determining and signal-dependent transcription factors (LDTFs and SDTFs, respectively). A deeper understanding of β-cell SDTFs and their cognate signals would delineate mechanisms of β-cell maturation and functional adaptation, which has direct implications for diabetes therapies and for generating mature β-cells from stem cells.
- Published
- 2021
43. Virgin β-Cells at the Neogenic Niche Proliferate Normally and Mature Slowly
- Author
-
Lee, Sharon, Zhang, Jing, Saravanakumar, Supraja, Flisher, Marcus F, Grimm, David R, van der Meulen, Talitha, and Huising, Mark O
- Subjects
Biomedical and Clinical Sciences ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Animals ,Cell Differentiation ,Cell Proliferation ,Female ,Flow Cytometry ,Fluorescent Antibody Technique ,Glucose Tolerance Test ,Humans ,Insulin-Secreting Cells ,Islets of Langerhans ,Male ,Mice ,Mice ,Inbred C57BL ,Medical and Health Sciences ,Endocrinology & Metabolism ,Biomedical and clinical sciences - Abstract
Proliferation of pancreatic β-cells has long been known to reach its peak in the neonatal stages and decline during adulthood. However, β-cell proliferation has been studied under the assumption that all β-cells constitute a single, homogenous population. It is unknown whether a subpopulation of β-cells retains the capacity to proliferate at a higher rate and thus contributes disproportionately to the maintenance of mature β-cell mass in adults. We therefore assessed the proliferative capacity and turnover potential of virgin β-cells, a novel population of immature β-cells found at the islet periphery. We demonstrate that virgin β-cells can proliferate but do so at rates similar to those of mature β-cells from the same islet under normal and challenged conditions. Virgin β-cell proliferation rates also conform to the age-dependent decline previously reported for β-cells at large. We further show that virgin β-cells represent a long-lived, stable subpopulation of β-cells with low turnover into mature β-cells under healthy conditions. Our observations indicate that virgin β-cells at the islet periphery can divide but do not contribute disproportionately to the maintenance of adult β-cell mass.
- Published
- 2021
44. Regulation of Cellular Senescence in Type 2 Diabetes Mellitus: From Mechanisms to Clinical Applications
- Author
-
Kanako Iwasaki, Cristian Abarca, and Cristina Aguayo-Mazzucato
- Subjects
aging ,diabetes mellitus ,type 2 ,insulin-secreting cells ,senotherapeutics ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
Cellular senescence is accelerated by hyperglycemia through multiple pathways. Therefore, senescence is an important cellular mechanism to consider in the pathophysiology of type 2 diabetes mellitus (T2DM) and an additional therapeutic target. The use of drugs that remove senescent cells has led to improvements in blood glucose levels and diabetic complications in animal studies. Although the removal of senescent cells is a promising approach for the treatment of T2DM, two main challenges limit its clinical application: the molecular basis of cellular senescence in each organ is yet to be understood, and the specific effect of removing senescent cells in each organ has to be determined. This review aims to discuss future applications of targeting senescence as a therapeutic option in T2DM and elucidate the characteristics of cellular senescence and senescence-associated secretory phenotype in the tissues important for regulating glucose levels: pancreas, liver, adipocytes, and skeletal muscle.
- Published
- 2023
- Full Text
- View/download PDF
45. Rediscovering Primary Cilia in Pancreatic Islets
- Author
-
Eun Young Lee and Jing W. Hughes
- Subjects
cilia ,ciliopathies ,insulin ,insulin-secreting cells ,islets of langerhans ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
Primary cilia are microtubule-based sensory and signaling organelles on the surfaces of most eukaryotic cells. Despite their early description by microscopy studies, islet cilia had not been examined in the functional context until recent decades. In pancreatic islets as in other tissues, primary cilia facilitate crucial developmental and signaling pathways in response to extracellular stimuli. Many human developmental and genetic disorders are associated with ciliary dysfunction, some manifesting as obesity and diabetes. Understanding the basis for metabolic diseases in human ciliopathies has been aided by close examination of cilia action in pancreatic islets at cellular and molecular levels. In this article, we review the evidence for ciliary expression on islet cells, known roles of cilia in pancreas development and islet hormone secretion, and summarize metabolic manifestations of human ciliopathy syndromes. We discuss emerging data on primary cilia regulation of islet cell signaling and the structural basis of cilia-mediated cell crosstalk, and offer our interpretation on the role of cilia in glucose homeostasis and human diseases.
- Published
- 2023
- Full Text
- View/download PDF
46. Human pluripotent stem cell-derived insulin-producing cells: A regenerative medicine perspective
- Author
-
Migliorini, Adriana, Nostro, Maria Cristina, and Sneddon, Julie B
- Subjects
Medical Biotechnology ,Biomedical and Clinical Sciences ,Transplantation ,Stem Cell Research - Embryonic - Human ,Stem Cell Research - Nonembryonic - Human ,Regenerative Medicine ,Stem Cell Research - Induced Pluripotent Stem Cell ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Stem Cell Research ,Biotechnology ,Autoimmune Disease ,Diabetes ,5.2 Cellular and gene therapies ,Development of treatments and therapeutic interventions ,Metabolic and endocrine ,Cell Differentiation ,Cell- and Tissue-Based Therapy ,Diabetes Mellitus ,Type 1 ,Diabetes Mellitus ,Type 2 ,Humans ,Insulin ,Insulin-Secreting Cells ,Pluripotent Stem Cells ,Transcription Factors ,Biochemistry and Cell Biology ,Medical Biochemistry and Metabolomics ,Endocrinology & Metabolism ,Biochemistry and cell biology ,Medical biochemistry and metabolomics - Abstract
Tremendous progress has been made over the last two decades in the field of pancreatic beta cell replacement therapy as a curative measure for diabetes. Transplantation studies have demonstrated therapeutic efficacy, and cGMP-grade cell products are currently being deployed for the first time in human clinical trials. In this perspective, we discuss current challenges surrounding the generation, delivery, and engraftment of stem cell-derived islet-like cells, along with strategies to induce durable tolerance to grafted cells, with an eye toward a functional cellular-based therapy enabling insulin independence for patients with diabetes.
- Published
- 2021
47. Single-cell chromatin accessibility identifies pancreatic islet cell type– and state-specific regulatory programs of diabetes risk
- Author
-
Chiou, Joshua, Zeng, Chun, Cheng, Zhang, Han, Jee Yun, Schlichting, Michael, Miller, Michael, Mendez, Robert, Huang, Serina, Wang, Jinzhao, Sui, Yinghui, Deogaygay, Allison, Okino, Mei-Lin, Qiu, Yunjiang, Sun, Ying, Kudtarkar, Parul, Fang, Rongxin, Preissl, Sebastian, Sander, Maike, Gorkin, David U, and Gaulton, Kyle J
- Subjects
Biochemistry and Cell Biology ,Genetics ,Biological Sciences ,Biotechnology ,Stem Cell Research - Embryonic - Non-Human ,Human Genome ,Stem Cell Research ,Diabetes ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Metabolic and endocrine ,Blood Glucose ,Cell Differentiation ,Chromatin ,Diabetes Mellitus ,Type 2 ,Epigenomics ,Fasting ,Gene Expression Profiling ,Genome-Wide Association Study ,Glucagon-Secreting Cells ,High-Throughput Nucleotide Sequencing ,Human Embryonic Stem Cells ,Humans ,Insulin-Secreting Cells ,KCNQ1 Potassium Channel ,Multigene Family ,Pancreatic Polypeptide-Secreting Cells ,Polymorphism ,Genetic ,Single-Cell Analysis ,Somatostatin-Secreting Cells ,Transcription Factors ,Medical and Health Sciences ,Developmental Biology ,Agricultural biotechnology ,Bioinformatics and computational biology - Abstract
Single-nucleus assay for transposase-accessible chromatin using sequencing (snATAC-seq) creates new opportunities to dissect cell type-specific mechanisms of complex diseases. Since pancreatic islets are central to type 2 diabetes (T2D), we profiled 15,298 islet cells by using combinatorial barcoding snATAC-seq and identified 12 clusters, including multiple alpha, beta and delta cell states. We cataloged 228,873 accessible chromatin sites and identified transcription factors underlying lineage- and state-specific regulation. We observed state-specific enrichment of fasting glucose and T2D genome-wide association studies for beta cells and enrichment for other endocrine cell types. At T2D signals localized to islet-accessible chromatin, we prioritized variants with predicted regulatory function and co-accessibility with target genes. A causal T2D variant rs231361 at the KCNQ1 locus had predicted effects on a beta cell enhancer co-accessible with INS and genome editing in embryonic stem cell-derived beta cells affected INS levels. Together our findings demonstrate the power of single-cell epigenomics for interpreting complex disease genetics.
- Published
- 2021
48. Free fatty acid receptor 4 inhibitory signaling in delta cells regulates islet hormone secretion in mice
- Author
-
Croze, Marine L, Flisher, Marcus F, Guillaume, Arthur, Tremblay, Caroline, Noguchi, Glyn M, Granziera, Sabrina, Vivot, Kevin, Castillo, Vincent C, Campbell, Scott A, Ghislain, Julien, Huising, Mark O, and Poitout, Vincent
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Diabetes ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,5.1 Pharmaceuticals ,Metabolic and endocrine ,Animals ,Fatty Acids ,Nonesterified ,Female ,Glucagon ,Glucagon-Secreting Cells ,Glucose ,Glucose Tolerance Test ,Homeostasis ,Insulin ,Insulin Secretion ,Insulin-Secreting Cells ,Islets of Langerhans ,Male ,Mice ,Mice ,Inbred C57BL ,Mice ,Knockout ,Receptors ,G-Protein-Coupled ,Signal Transduction ,Somatostatin ,Somatostatin-Secreting Cells ,FFAR4 ,GPR120 ,Islet of langerhans ,Physiology ,Biochemistry and cell biology - Abstract
ObjectiveMaintenance of glucose homeostasis requires the precise regulation of hormone secretion from the endocrine pancreas. Free fatty acid receptor 4 (FFAR4/GPR120) is a G protein-coupled receptor whose activation in islets of Langerhans promotes insulin and glucagon secretion and inhibits somatostatin secretion. However, the contribution of individual islet cell types (α, β, and δ cells) to the insulinotropic and glucagonotropic effects of GPR120 remains unclear. As gpr120 mRNA is enriched in somatostatin-secreting δ cells, we hypothesized that GPR120 activation stimulates insulin and glucagon secretion via inhibition of somatostatin release.MethodsGlucose tolerance tests were performed in mice after administration of selective GPR120 agonist Compound A. Insulin, glucagon, and somatostatin secretion were measured in static incubations of isolated mouse islets in response to endogenous (ω-3 polyunsaturated fatty acids) and/or pharmacological (Compound A and AZ-13581837) GPR120 agonists. The effect of Compound A on hormone secretion was tested further in islets isolated from mice with global or somatostatin cell-specific knock-out of gpr120. Gpr120 expression was assessed in pancreatic sections by RNA in situ hybridization. Cyclic AMP (cAMP) and calcium dynamics in response to pharmacological GPR120 agonists were measured specifically in α, β, and δ cells in intact islets using cAMPER and GCaMP6 reporter mice, respectively.ResultsAcute exposure to Compound A increased glucose tolerance, circulating insulin, and glucagon levels in vivo. Endogenous and/or pharmacological GPR120 agonists reduced somatostatin secretion in isolated islets and concomitantly demonstrated dose-dependent potentiation of glucose-stimulated insulin secretion and arginine-stimulated glucagon secretion. Gpr120 was enriched in δ cells. Pharmacological GPR120 agonists reduced cAMP and calcium levels in δ cells but increased these signals in α and β cells. Compound A-mediated inhibition of somatostatin secretion was insensitive to pertussis toxin. The effect of Compound A on hormone secretion was completely absent in islets from mice with either global or somatostatin cell-specific deletion of gpr120 and partially reduced upon blockade of somatostatin receptor signaling by cyclosomatostatin.ConclusionsInhibitory GPR120 signaling in δ cells contributes to both insulin and glucagon secretion in part by mitigating somatostatin release.
- Published
- 2021
49. GLP-1 receptor signaling increases PCSK1 and β cell features in human α cells.
- Author
-
Saikia, Mridusmita, Holter, Marlena, Donahue, Leanne, Lee, Isaac, Zheng, Qiaonan, Wise, Journey, Todero, Jenna, Phuong, Daryl, Garibay, Darline, Coch, Reilly, Sloop, Kyle, Garcia-Ocana, Adolfo, Danko, Charles, and Cummings, Bethany
- Subjects
Diabetes ,Endocrinology ,Islet cells ,Metabolism ,Animals ,Female ,Gene Knockdown Techniques ,Glucagon-Like Peptide-1 Receptor ,Glucagon-Secreting Cells ,Humans ,Hypoglycemic Agents ,In Vitro Techniques ,Insulin-Secreting Cells ,Liraglutide ,Male ,Mice ,Mice ,Inbred C57BL ,Mice ,Knockout ,Proprotein Convertase 1 ,RNA-Seq ,Signal Transduction - Abstract
Glucagon-like peptide-1 (GLP-1) is an incretin hormone that potentiates glucose-stimulated insulin secretion. GLP-1 is classically produced by gut L cells; however, under certain circumstances α cells can express the prohormone convertase required for proglucagon processing to GLP-1, prohormone convertase 1/3 (PC1/3), and can produce GLP-1. However, the mechanisms through which this occurs are poorly defined. Understanding the mechanisms by which α cell PC1/3 expression can be activated may reveal new targets for diabetes treatment. Here, we demonstrate that the GLP-1 receptor (GLP-1R) agonist, liraglutide, increased α cell GLP-1 expression in a β cell GLP-1R-dependent manner. We demonstrate that this effect of liraglutide was translationally relevant in human islets through application of a new scRNA-seq technology, DART-Seq. We found that the effect of liraglutide to increase α cell PC1/3 mRNA expression occurred in a subcluster of α cells and was associated with increased expression of other β cell-like genes, which we confirmed by IHC. Finally, we found that the effect of liraglutide to increase bihormonal insulin+ glucagon+ cells was mediated by the β cell GLP-1R in mice. Together, our data validate a high-sensitivity method for scRNA-seq in human islets and identify a potentially novel GLP-1-mediated pathway regulating human α cell function.
- Published
- 2021
50. The Role of Oxidative Stress in Pancreatic β Cell Dysfunction in Diabetes.
- Author
-
Eguchi, Natsuki, Vaziri, Nosratola D, Dafoe, Donald C, and Ichii, Hirohito
- Subjects
Animals ,Humans ,Diabetes Mellitus ,Signal Transduction ,Oxidative Stress ,Insulin-Secreting Cells ,anti-oxidants ,diabetes ,oxidative stress ,pancreatic β cells ,pancreatic β ,cells ,Digestive Diseases ,Diabetes ,Nutrition ,2.1 Biological and endogenous factors ,Metabolic and endocrine ,Generic health relevance ,Chemical Physics ,Other Chemical Sciences ,Genetics ,Other Biological Sciences - Abstract
Diabetes is a chronic metabolic disorder characterized by inappropriately elevated glucose levels as a result of impaired pancreatic β cell function and insulin resistance. Extensive studies have been conducted to elucidate the mechanism involved in the development of β cell failure and death under diabetic conditions such as hyperglycemia, hyperlipidemia, and inflammation. Of the plethora of proposed mechanisms, endoplasmic reticulum (ER) stress, mitochondrial dysfunction, and oxidative stress have been shown to play a central role in promoting β cell dysfunction. It has become more evident in recent years that these 3 factors are closely interrelated and importantly aggravate each other. Oxidative stress in particular is of great interest to β cell health and survival as it has been shown that β cells exhibit lower antioxidative capacity. Therefore, this review will focus on discussing factors that contribute to the development of oxidative stress in pancreatic β cells and explore the downstream effects of oxidative stress on β cell function and health. Furthermore, antioxidative capacity of β cells to counteract these effects will be discussed along with new approaches focused on preserving β cells under oxidative conditions.
- Published
- 2021
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.