18 results on '"Wagner, Bridget K."'
Search Results
2. The Power of Sophisticated Phenotypic Screening and Modern Mechanism-of-Action Methods.
- Author
-
Wagner, Bridget K. and Schreiber, Stuart L.
- Subjects
- *
PHENOTYPES , *BIOCHEMICAL mechanism of action , *SMALL molecules , *CELL culture , *CHEMICAL biology , *DRUG use testing - Abstract
The enthusiasm for phenotypic screening as an approach for small-molecule discovery has increased dramatically over the last several years. The recent increase in phenotype-based discoveries is in part due to advancements in phenotypic readouts in improved disease models that recapitulate clinically relevant biology in cell culture. Of course, a major historical barrier to using phenotypic assays in chemical biology has been the challenge in determining the mechanism of action (MoA) for compounds of interest. With the combination of medically inspired phenotypic screening and the development of modern MoA methods, we can now start implementing this approach in chemical probe and drug discovery. In this Perspective, we highlight recent advances in phenotypic readouts and MoA determination by discussing several case studies in which both activities were required for understanding the chemical biology involved and, in some cases, advancing toward clinical development. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
3. Phenotypic approaches to small-molecule discovery in chemical biology.
- Author
-
Wagner, Bridget K. and Dey, Mishtu
- Subjects
- *
CHEMICAL biology , *PHENOTYPES - Published
- 2021
- Full Text
- View/download PDF
4. When Small Molecules Are Like Real Estate: It's All about Location, Location, Location.
- Author
-
DeRan, Michael and Wagner, Bridget K.
- Subjects
- *
SMALL molecules , *APOPTOSIS - Abstract
In this issue of Cell Chemical Biology , Park et al. (2018) demonstrate that targeting apoptazole, an Hsp70 inhibitor, to mitochondria induces apoptosis by a distinct mechanism of action different from unmodified apoptazole, which accumulates in the lysosome. These results highlight the power of subcellular localization in small-molecule selectivity. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
5. Nuisance compounds in cellular assays.
- Author
-
Dahlin, Jayme L., Auld, Douglas S., Rothenaigner, Ina, Haney, Steve, Sexton, Jonathan Z., Nissink, J. Willem M., Walsh, Jarrod, Lee, Jonathan A., Strelow, John M., Willard, Francis S., Ferrins, Lori, Baell, Jonathan B., Walters, Michael A., Hua, Bruce K., Hadian, Kamyar, and Wagner, Bridget K.
- Subjects
- *
NUISANCES , *CHEMICAL biology , *REFUSE containers , *CHEMICAL potential , *PHENOTYPES - Abstract
Compounds that exhibit assay interference or undesirable mechanisms of bioactivity ("nuisance compounds") are routinely encountered in cellular assays, including phenotypic and high-content screening assays. Much is known regarding compound-dependent assay interferences in cell-free assays. However, despite the essential role of cellular assays in chemical biology and drug discovery, there is considerably less known about nuisance compounds in more complex cell-based assays. In our view, a major obstacle to realizing the full potential of chemical biology will not just be difficult-to-drug targets or even the sheer number of targets, but rather nuisance compounds, due to their ability to waste significant resources and erode scientific trust. In this review, we summarize our collective academic, government, and industry experiences regarding cellular nuisance compounds. We describe assay design strategies to mitigate the impact of nuisance compounds and suggest best practices to efficiently address these compounds in complex biological settings. [Display omitted] Nuisance compounds can waste significant resources by producing promising bioactivities that are attributable to undesirable mechanisms of action. Addressing nuisance compounds is particularly challenging in cellular assays. Dahlin et al. summarize academic, government, and industry experiences with assay design and hit triage to specifically address cellular nuisance compounds. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
6. A Small-Molecule Inducer of PDX1 Expression Identified by High-Throughput Screening.
- Author
-
Yuan, Yuan, Hartland, Kate, Boskovic, Zarko, Wang, Yikai, Walpita, Deepika, Lysy, Philippe?A., Zhong, Cheng, Young, Damian?W., Kim, Young-kwon, Tolliday, Nicola?J., Sokal, Etienne?M., Schreiber, Stuart?L., and Wagner, Bridget?K.
- Subjects
- *
DUODENAL diseases , *HOMEOBOX genes , *GENE expression , *GENETIC testing , *GENETIC transcription , *CELL physiology , *LABORATORY mice - Abstract
Summary: Pancreatic and duodenal homeobox 1 (PDX1), a member of the homeodomain-containing transcription factor family, is a key transcription factor important for both pancreas development and mature β cell function. The ectopic overexpression of Pdx1, Neurog3, and MafA in mice reprograms acinar cells to insulin-producing cells. We developed a quantitative PCR-based gene expression assay to screen more than 60,000 compounds for expression of each of these genes in the human PANC-1 ductal carcinoma cell line. We identified BRD7552, which upregulated PDX1 expression in both primary human islets and ductal cells, and induced epigenetic changes in the PDX1 promoter consistent with transcriptional activation. Prolonged compound treatment induced both insulin mRNA and protein and also enhanced insulin expression induced by the three-gene combination. These results provide a proof of principle for identifying small molecules that induce expression of transcription factors to control cellular reprogramming. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
7. Inhibition of Histone Deacetylase 3 Protects Beta Cells from Cytokine-Induced Apoptosis
- Author
-
Chou, Danny Hung-Chieh, Holson, Edward B., Wagner, Florence F., Tang, Alicia J., Maglathlin, Rebecca L., Lewis, Timothy A., Schreiber, Stuart L., and Wagner, Bridget K.
- Subjects
- *
HISTONE deacetylase , *PANCREATIC beta cells , *CYTOKINES , *APOPTOSIS , *GLUCOSE , *HYDROXAMIC acids - Abstract
Summary: Cytokine-induced beta-cell apoptosis is important to the etiology of type-1 diabetes. Although previous reports have shown that general inhibitors of histone deacetylase (HDAC) activity, such as suberoylanilide hydroxamic acid and trichostatin A, can partially prevent beta-cell death, they do not fully restore beta-cell function. To understand HDAC isoform selectivity in beta cells, we measured the cellular effects of 11 structurally diverse HDAC inhibitors on cytokine-induced apoptosis in the rat INS-1E cell line. All 11 compounds restored ATP levels and reduced nitrite secretion. However, caspase-3 activity was reduced only by MS-275 and CI-994, both of which target HDAC1, 2, and 3. Importantly, both MS-275 and genetic knockdown of Hdac3 alone were sufficient to restore glucose-stimulated insulin secretion in the presence of cytokines. These results suggest that HDAC3-selective inhibitors may be effective in preventing cytokine-induced beta-cell apoptosis. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
8. 6-Phosphogluconate Dehydrogenase Links Cytosolic Carbohydrate Metabolism to Protein Secretion via Modulation of Glutathione Levels.
- Author
-
Li, Haoxin, Ericsson, Maria, Rabasha, Bokang, Budnik, Bogdan, Chan, Sze Ham, Freinkman, Elizaveta, Lewis, Caroline A., Doench, John G., Wagner, Bridget K., Garraway, Levi A., and Schreiber, Stuart L.
- Subjects
- *
CARBOHYDRATE metabolism , *PROTEIN metabolism , *SECRETION , *SUPPRESSOR mutation , *CYTOSKELETAL proteins , *GLUTATHIONE - Abstract
The proteinaceous extracellular matrix (ECM) is vital for the survival, proliferation, migration, and differentiation of many types of cancer. However, little is known regarding metabolic pathways required for ECM secretion. By using an unbiased computational approach, we searched for enzymes whose suppression may lead to disruptions in protein secretion. Here, we show that 6-phosphogluconate dehydrogenase (PGD), a cytosolic enzyme involved in carbohydrate metabolism, is required for ER structural integrity and protein secretion. Chemical inhibition or genetic suppression of PGD activity led to cell stress accompanied by significantly expanded ER volume and was rescued by compensating endogenous glutathione supplies. Our results also suggest that this characteristic ER-dilation phenotype may be a general marker indicating increased ECM protein congestion inside cells and decreased secretion. Thus, PGD serves as a link between cytosolic carbohydrate metabolism and protein secretion. • Compounds targeting distinct protein secretion steps might cause similar phenotypes • PGD is a link between cytosolic carbohydrate metabolism and protein secretion • PGD suppression leads to ER dilation by modulating GSH levels • ER dilation is a marker of increased ECM protein congestion and decreased secretion Li et al. discovered 6-phosphogluconate dehydrogenase (PGD) as a link between cytosolic carbohydrate metabolism and protein secretion. By showing that PGD suppression causes a characteristic ER-dilation phenotype that is associated with increased ECM protein inside cells as well as decreased secretion, the authors propose a "congestion-to-dilation" model. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
9. A Small-Molecule Inducer of PDX1 Expression Identified by High-Throughput Screening.
- Author
-
Yuan, Yuan, Hartland, Kate, Boskovic, Zarko, Wang, Yikai, Walpita, Deepika, Lysy, Philippe?A., Zhong, Cheng, Young, Damian?W., Kim, Young-kwon, Tolliday, Nicola?J., Sokal, Etienne?M., Schreiber, Stuart?L., and Wagner, Bridget?K.
- Published
- 2014
- Full Text
- View/download PDF
10. Excess pancreatic elastase alters acinar-β cell communication by impairing the mechano-signaling and the PAR2 pathways.
- Author
-
Basile G, Vetere A, Hu J, Ijaduola O, Zhang Y, Liu KC, Eltony AM, De Jesus DF, Fukuda K, Doherty G, Leech CA, Chepurny OG, Holz GG, Yun SH, Andersson O, Choudhary A, Wagner BK, and Kulkarni RN
- Subjects
- Humans, Mice, Animals, Acinar Cells metabolism, Pancreatic Elastase metabolism, Insulin metabolism, Cell Communication, Diabetes Mellitus, Type 2 metabolism, Insulin-Secreting Cells metabolism
- Abstract
Type 1 (T1D) or type 2 diabetes (T2D) are caused by a deficit of functional insulin-producing β cells. Thus, the identification of β cell trophic agents could allow the development of therapeutic strategies to counteract diabetes. The discovery of SerpinB1, an elastase inhibitor that promotes human β cell growth, prompted us to hypothesize that pancreatic elastase (PE) regulates β cell viability. Here, we report that PE is up-regulated in acinar cells and in islets from T2D patients, and negatively impacts β cell viability. Using high-throughput screening assays, we identified telaprevir as a potent PE inhibitor that can increase human and rodent β cell viability in vitro and in vivo and improve glucose tolerance in insulin-resistant mice. Phospho-antibody microarrays and single-cell RNA sequencing analysis identified PAR2 and mechano-signaling pathways as potential mediators of PE. Taken together, our work highlights PE as a potential regulator of acinar-β cell crosstalk that acts to limit β cell viability, leading to T2D., Competing Interests: Declaration of interests R.N.K. is a member of the Scientific Advisory Board and served as a consultant for Novo Nordisk, Biomea, and Inversago Pharma., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
11. FALCON systematically interrogates free fatty acid biology and identifies a novel mediator of lipotoxicity.
- Author
-
Wieder N, Fried JC, Kim C, Sidhom EH, Brown MR, Marshall JL, Arevalo C, Dvela-Levitt M, Kost-Alimova M, Sieber J, Gabriel KR, Pacheco J, Clish C, Abbasi HS, Singh S, Rutter JC, Therrien M, Yoon H, Lai ZW, Baublis A, Subramanian R, Devkota R, Small J, Sreekanth V, Han M, Lim D, Carpenter AE, Flannick J, Finucane H, Haigis MC, Claussnitzer M, Sheu E, Stevens B, Wagner BK, Choudhary A, Shaw JL, Pablo JL, and Greka A
- Subjects
- Humans, Fatty Acids, Signal Transduction, Biology, Fatty Acids, Nonesterified metabolism, Diabetes Mellitus, Type 2
- Abstract
Cellular exposure to free fatty acids (FFAs) is implicated in the pathogenesis of obesity-associated diseases. However, there are no scalable approaches to comprehensively assess the diverse FFAs circulating in human plasma. Furthermore, assessing how FFA-mediated processes interact with genetic risk for disease remains elusive. Here, we report the design and implementation of fatty acid library for comprehensive ontologies (FALCON), an unbiased, scalable, and multimodal interrogation of 61 structurally diverse FFAs. We identified a subset of lipotoxic monounsaturated fatty acids associated with decreased membrane fluidity. Furthermore, we prioritized genes that reflect the combined effects of harmful FFA exposure and genetic risk for type 2 diabetes (T2D). We found that c-MAF-inducing protein (CMIP) protects cells from FFA exposure by modulating Akt signaling. In sum, FALCON empowers the study of fundamental FFA biology and offers an integrative approach to identify much needed targets for diverse diseases associated with disordered FFA metabolism., Competing Interests: Declaration of interests N.W., J.C.F., and A.G. are co-inventors of a patent on the composition, method, and use for FFA screening, application no: 52199-550P01US. A.G. serves as a founding advisor to a new company launched by Atlas Ventures, an agreement reviewed and managed by Brigham and Women’s Hospital, Mass General Brigham, and the Broad Institute of MIT and Harvard in accordance with their conflict of interest policies., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
12. SMNDC1 links chromatin remodeling and splicing to regulate pancreatic hormone expression.
- Author
-
Casteels T, Bajew S, Reiniš J, Enders L, Schuster M, Fontaine F, Müller AC, Wagner BK, Bock C, and Kubicek S
- Subjects
- Animals, Humans, Insulin metabolism, Insulin Secretion, Mice, Transcription Factors metabolism, Chromatin Assembly and Disassembly, Glucagon-Secreting Cells metabolism, Insulin-Secreting Cells metabolism, Islets of Langerhans metabolism, RNA Splicing genetics, RNA Splicing Factors metabolism, SMN Complex Proteins metabolism
- Abstract
Insulin expression is primarily restricted to the pancreatic β cells, which are physically or functionally depleted in diabetes. Identifying targetable pathways repressing insulin in non-β cells, particularly in the developmentally related glucagon-secreting α cells, is an important aim of regenerative medicine. Here, we perform an RNA interference screen in a murine α cell line to identify silencers of insulin expression. We discover that knockdown of the splicing factor Smndc1 triggers a global repression of α cell gene-expression programs in favor of increased β cell markers. Mechanistically, Smndc1 knockdown upregulates the β cell transcription factor Pdx1 by modulating the activities of the BAF and Atrx chromatin remodeling complexes. SMNDC1's repressive role is conserved in human pancreatic islets, its loss triggering enhanced insulin secretion and PDX1 expression. Our study identifies Smndc1 as a key factor connecting splicing and chromatin remodeling to the control of insulin expression in human and mouse islet cells., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
13. An expanded universe of cancer targets.
- Author
-
Hahn WC, Bader JS, Braun TP, Califano A, Clemons PA, Druker BJ, Ewald AJ, Fu H, Jagu S, Kemp CJ, Kim W, Kuo CJ, McManus M, B Mills G, Mo X, Sahni N, Schreiber SL, Talamas JA, Tamayo P, Tyner JW, Wagner BK, Weiss WA, and Gerhard DS
- Subjects
- Animals, Clinical Trials as Topic, Disease Models, Animal, Genomics, Humans, Neoplasms genetics, Neoplasms pathology, Tumor Escape drug effects, Tumor Microenvironment drug effects, Drug Delivery Systems, Neoplasms drug therapy
- Abstract
The characterization of cancer genomes has provided insight into somatically altered genes across tumors, transformed our understanding of cancer biology, and enabled tailoring of therapeutic strategies. However, the function of most cancer alleles remains mysterious, and many cancer features transcend their genomes. Consequently, tumor genomic characterization does not influence therapy for most patients. Approaches to understand the function and circuitry of cancer genes provide complementary approaches to elucidate both oncogene and non-oncogene dependencies. Emerging work indicates that the diversity of therapeutic targets engendered by non-oncogene dependencies is much larger than the list of recurrently mutated genes. Here we describe a framework for this expanded list of cancer targets, providing novel opportunities for clinical translation., Competing Interests: Declaration of interests G.B.M. is a science advisory board (SAB) member/consultant with AstraZeneca, Chrysallis Biotechnology, GSK, ImmunoMET, Ionis, Lilly, PDX Pharmaceuticals, Signalchem Lifesciences, Symphogen, Tarveda, Turbine, and Zentalis Pharmaceuticals; has stock/options/financial engagement with Catena Pharmaceuticals, ImmunoMet, SignalChem, and Tarveda; has licensed technology: HRD assay to Myriad Genetics and DSP patents with Nanostring; has sponsored research: Nanostring Center of Excellence and Ionis (Provision of tool compounds); and clinical trials support (funding or in kind) with AstraZeneca, Genentech, GSK, Lilly. S.L.S. is a shareholder and serves on the board of directors of Jnana Therapeutics; is a shareholder of Forma Therapeutics and Decibel Therapeutics; is a shareholder and advises Kojin Therapeutics, Kisbee Therapeutics, and Eikonizo Therapeutics; serves on the SABs of Eisai Co., Ono Pharma Foundation, Exo Therapeutics, and F-Prime Capital Partners; serves on the board of advisers of the Genomics Institute of the Novartis Research Foundation; and is a Novartis Faculty Scholar. W.C.H. is a consultant for ThermoFisher, Solasta, MPM Capital, iTeos, RAPPTA Therapeutics, Jubilant Therapeutics, and Paraxel and is a scientific founder and serves on the SAB for KSQ Therapeutics. J.S.B. is a founder and director of Neochromosome and holds an ownership equity interest in the company. A.C. is the founder and an equity holder in DarwinHealth, a company that has licensed algorithms for the analysis of regulatory networks and master regulator proteins from Columbia University. Columbia University is also an equity holder in DarwinHealth. C.J. Kemp is a founder and an equity holder in SEngine Precision Medicine, a company that harnesses 3D organoid technology and AI for more effective treatment options and accelerated drug development. W.K. and P.T. receive research support from Pfizer Oncology. W.A.W. is co-founder of StemSynergy Therapeutics. B.J.D. is an SAB member of Aileron Therapeutics, Therapy Architects (ALLCRON), Cepheid, Vivid Biosciences, Celgene, RUNX1 Research Program, Novartis, Gilead Sciences (inactive), Monojul (inactive); is an SAB member and holds stock in Aptose Biosciences, Blueprint Medicines, EnLiven Therapeutics, Iterion Therapeutics, Third Coast Therapeutics, GRAIL (SAB inactive); is a scientific founder of MolecularMD (inactive, acquired by ICON); is a member of the board of directors holds stock in Amgen; is a member of the board of directors of Burroughs Wellcome Fund and CureOne; is a member of the Joint Steering Committee of Beat AML LLS; is a founder of VB Therapeutics; has a sponsored research agreement for EnLiven Therapeutics; has clinical trial funding from Novartis, Bristol-Myers Squibb, Pfizer; and collects royalties from patent 6958335 (Novartis) and OHSU and Merck and one CytoImage exclusive license., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
14. A High-Throughput Platform to Identify Small-Molecule Inhibitors of CRISPR-Cas9.
- Author
-
Maji B, Gangopadhyay SA, Lee M, Shi M, Wu P, Heler R, Mok B, Lim D, Siriwardena SU, Paul B, Dančík V, Vetere A, Mesleh MF, Marraffini LA, Liu DR, Clemons PA, Wagner BK, and Choudhary A
- Subjects
- CRISPR-Associated Protein 9 metabolism, Clustered Regularly Interspaced Short Palindromic Repeats physiology, DNA metabolism, Endonucleases metabolism, Gene Editing methods, Genome, Small Molecule Libraries, Streptococcus pyogenes genetics, Substrate Specificity, CRISPR-Associated Protein 9 antagonists & inhibitors, CRISPR-Cas Systems physiology, High-Throughput Screening Assays methods
- Abstract
The precise control of CRISPR-Cas9 activity is required for a number of genome engineering technologies. Here, we report a generalizable platform that provided the first synthetic small-molecule inhibitors of Streptococcus pyogenes Cas9 (SpCas9) that weigh <500 Da and are cell permeable, reversible, and stable under physiological conditions. We developed a suite of high-throughput assays for SpCas9 functions, including a primary screening assay for SpCas9 binding to the protospacer adjacent motif, and used these assays to screen a structurally diverse collection of natural-product-like small molecules to ultimately identify compounds that disrupt the SpCas9-DNA interaction. Using these synthetic anti-CRISPR small molecules, we demonstrated dose and temporal control of SpCas9 and catalytically impaired SpCas9 technologies, including transcription activation, and identified a pharmacophore for SpCas9 inhibition using structure-activity relationships. These studies establish a platform for rapidly identifying synthetic, miniature, cell-permeable, and reversible inhibitors against both SpCas9 and next-generation CRISPR-associated nucleases., (Published by Elsevier Inc.)
- Published
- 2019
- Full Text
- View/download PDF
15. Type 2 Diabetes Variants Disrupt Function of SLC16A11 through Two Distinct Mechanisms.
- Author
-
Rusu V, Hoch E, Mercader JM, Tenen DE, Gymrek M, Hartigan CR, DeRan M, von Grotthuss M, Fontanillas P, Spooner A, Guzman G, Deik AA, Pierce KA, Dennis C, Clish CB, Carr SA, Wagner BK, Schenone M, Ng MCY, Chen BH, Centeno-Cruz F, Zerrweck C, Orozco L, Altshuler DM, Schreiber SL, Florez JC, Jacobs SBR, and Lander ES
- Subjects
- Basigin metabolism, Cell Membrane metabolism, Chromosomes, Human, Pair 17 metabolism, Gene Knockdown Techniques, Haplotypes, Hepatocytes metabolism, Heterozygote, Histone Code, Humans, Liver metabolism, Models, Molecular, Monocarboxylic Acid Transporters chemistry, Diabetes Mellitus, Type 2 metabolism, Monocarboxylic Acid Transporters genetics, Monocarboxylic Acid Transporters metabolism
- Abstract
Type 2 diabetes (T2D) affects Latinos at twice the rate seen in populations of European descent. We recently identified a risk haplotype spanning SLC16A11 that explains ∼20% of the increased T2D prevalence in Mexico. Here, through genetic fine-mapping, we define a set of tightly linked variants likely to contain the causal allele(s). We show that variants on the T2D-associated haplotype have two distinct effects: (1) decreasing SLC16A11 expression in liver and (2) disrupting a key interaction with basigin, thereby reducing cell-surface localization. Both independent mechanisms reduce SLC16A11 function and suggest SLC16A11 is the causal gene at this locus. To gain insight into how SLC16A11 disruption impacts T2D risk, we demonstrate that SLC16A11 is a proton-coupled monocarboxylate transporter and that genetic perturbation of SLC16A11 induces changes in fatty acid and lipid metabolism that are associated with increased T2D risk. Our findings suggest that increasing SLC16A11 function could be therapeutically beneficial for T2D. VIDEO ABSTRACT., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
16. A Single-Cell Transcriptomic Map of the Human and Mouse Pancreas Reveals Inter- and Intra-cell Population Structure.
- Author
-
Baron M, Veres A, Wolock SL, Faust AL, Gaujoux R, Vetere A, Ryu JH, Wagner BK, Shen-Orr SS, Klein AM, Melton DA, and Yanai I
- Subjects
- Animals, Cell Differentiation, Gene Expression Profiling, Gene Expression Regulation, Developmental, Humans, Islets of Langerhans, Mice, Pancreas, Pancreas, Exocrine, Single-Cell Analysis, Transcription Factors, Transcriptome
- Abstract
Although the function of the mammalian pancreas hinges on complex interactions of distinct cell types, gene expression profiles have primarily been described with bulk mixtures. Here we implemented a droplet-based, single-cell RNA-seq method to determine the transcriptomes of over 12,000 individual pancreatic cells from four human donors and two mouse strains. Cells could be divided into 15 clusters that matched previously characterized cell types: all endocrine cell types, including rare epsilon-cells; exocrine cell types; vascular cells; Schwann cells; quiescent and activated stellate cells; and four types of immune cells. We detected subpopulations of ductal cells with distinct expression profiles and validated their existence with immuno-histochemistry stains. Moreover, among human beta- cells, we detected heterogeneity in the regulation of genes relating to functional maturation and levels of ER stress. Finally, we deconvolved bulk gene expression samples using the single-cell data to detect disease-associated differential expression. Our dataset provides a resource for the discovery of novel cell type-specific transcription factors, signaling receptors, and medically relevant genes., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
17. Preserved DNA Damage Checkpoint Pathway Protects against Complications in Long-Standing Type 1 Diabetes.
- Author
-
Bhatt S, Gupta MK, Khamaisi M, Martinez R, Gritsenko MA, Wagner BK, Guye P, Busskamp V, Shirakawa J, Wu G, Liew CW, Clauss TR, Valdez I, El Ouaamari A, Dirice E, Takatani T, Keenan HA, Smith RD, Church G, Weiss R, Wagers AJ, Qian WJ, King GL, and Kulkarni RN
- Subjects
- Aged, Diabetes Complications metabolism, Diabetes Complications pathology, Diabetes Complications prevention & control, Diabetes Mellitus, Type 1 pathology, Female, Humans, Induced Pluripotent Stem Cells metabolism, Induced Pluripotent Stem Cells pathology, Male, Middle Aged, Neurons metabolism, Neurons pathology, Cell Cycle Checkpoints, DNA Damage, Diabetes Mellitus, Type 1 metabolism, Gene Expression Regulation, MicroRNAs biosynthesis, Models, Biological
- Abstract
The mechanisms underlying the development of complications in type 1 diabetes (T1D) are poorly understood. Disease modeling of induced pluripotent stem cells (iPSCs) from patients with longstanding T1D (disease duration ≥ 50 years) with severe (Medalist +C) or absent to mild complications (Medalist -C) revealed impaired growth, reprogramming, and differentiation in Medalist +C. Genomics and proteomics analyses suggested differential regulation of DNA damage checkpoint proteins favoring protection from cellular apoptosis in Medalist -C. In silico analyses showed altered expression patterns of DNA damage checkpoint factors among the Medalist groups to be targets of miR200, whose expression was significantly elevated in Medalist +C serum. Notably, neurons differentiated from Medalist +C iPSCs exhibited enhanced susceptibility to genotoxic stress that worsened upon miR200 overexpression. Furthermore, knockdown of miR200 in Medalist +C fibroblasts and iPSCs rescued checkpoint protein expression and reduced DNA damage. We propose miR200-regulated DNA damage checkpoint pathway as a potential therapeutic target for treating complications of diabetes., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
18. High-throughput luminescent reporter of insulin secretion for discovering regulators of pancreatic Beta-cell function.
- Author
-
Burns SM, Vetere A, Walpita D, Dančík V, Khodier C, Perez J, Clemons PA, Wagner BK, and Altshuler D
- Subjects
- Cells, Cultured, Cytokines pharmacology, Enzyme-Linked Immunosorbent Assay, Genes, Reporter, Glucose pharmacology, High-Throughput Screening Assays, Humans, Insulin genetics, Insulin Secretion, Insulin-Secreting Cells cytology, Insulin-Secreting Cells drug effects, Luciferases genetics, Luciferases metabolism, Recombinant Fusion Proteins biosynthesis, Recombinant Fusion Proteins genetics, Thapsigargin toxicity, Fatty Acids pharmacology, Insulin metabolism, Insulin-Secreting Cells metabolism
- Abstract
Defects in insulin secretion play a central role in the pathogenesis of type 2 diabetes, yet the mechanisms driving beta-cell dysfunction remain poorly understood, and therapies to preserve glucose-dependent insulin release are inadequate. We report a luminescent insulin secretion assay that enables large-scale investigations of beta-cell function, created by inserting Gaussia luciferase into the C-peptide portion of proinsulin. Beta-cell lines expressing this construct cosecrete luciferase and insulin in close correlation, under both standard conditions or when stressed by cytokines, fatty acids, or ER toxins. We adapted the reporter for high-throughput assays and performed a 1,600-compound pilot screen, which identified several classes of drugs inhibiting secretion, as well as glucose-potentiated secretagogues that were confirmed to have activity in primary human islets. Requiring 40-fold less time and expense than the traditional ELISA, this assay may accelerate the identification of pathways governing insulin secretion and compounds that safely augment beta-cell function in diabetes., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.