Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Gastroenterology ; 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38908487

ABSTRACT

BACKGROUND AND AIMS: Pancreatic ducts form an intricate network of tubules that secrete bicarbonate and drive acinar secretions into the duodenum. This network is formed by centroacinar cells, terminal, intercalated, intracalated ducts, and the main pancreatic duct. Ductal heterogeneity at the single-cell level has been poorly characterized; therefore, our understanding of the role of ductal cells in pancreas regeneration and exocrine pathogenesis has been hampered by the limited knowledge and unexplained diversity within the ductal network. METHODS: We used scRNA-seq to comprehensively characterize mouse ductal heterogeneity at single-cell resolution of the entire ductal epithelium from centroacinar cells to the main duct. Moreover, we used organoid cultures, injury models and pancreatic tumor samples to interrogate the role of novel ductal populations in pancreas regeneration and exocrine pathogenesis. RESULTS: We have identified the coexistence of 15 ductal populations within the healthy pancreas and characterized their organoid formation capacity and endocrine differentiation potential. Cluster isolation and subsequent culturing let us identify ductal cell populations with high organoid formation capacity and endocrine and exocrine differentiation potential in vitro, including Wnt-responsive-population, ciliated-population and FLRT3+ cells. Moreover, we have characterized the location of these novel ductal populations in healthy pancreas, chronic pancreatitis, and tumor samples. The expression of WNT-responsive, IFN-responsive and EMT-population markers increases in chronic pancreatitis and tumor samples. CONCLUSIONS: In light of our discovery of previously unidentified ductal populations, we unmask potential roles of specific ductal populations in pancreas regeneration and exocrine pathogenesis. Thus, novel lineage tracing models are needed to investigate ductal specific populations in vivo.

2.
Nat Commun ; 15(1): 3744, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702321

ABSTRACT

Cellular composition and anatomical organization influence normal and aberrant organ functions. Emerging spatial single-cell proteomic assays such as Image Mass Cytometry (IMC) and Co-Detection by Indexing (CODEX) have facilitated the study of cellular composition and organization by enabling high-throughput measurement of cells and their localization directly in intact tissues. However, annotation of cell types and quantification of their relative localization in tissues remain challenging. To address these unmet needs for atlas-scale datasets like Human Pancreas Analysis Program (HPAP), we develop AnnoSpat (Annotator and Spatial Pattern Finder) that uses neural network and point process algorithms to automatically identify cell types and quantify cell-cell proximity relationships. Our study of data from IMC and CODEX shows the higher performance of AnnoSpat in rapid and accurate annotation of cell types compared to alternative approaches. Moreover, the application of AnnoSpat to type 1 diabetic, non-diabetic autoantibody-positive, and non-diabetic organ donor cohorts recapitulates known islet pathobiology and shows differential dynamics of pancreatic polypeptide (PP) cell abundance and CD8+ T cells infiltration in islets during type 1 diabetes progression.


Subject(s)
Algorithms , Diabetes Mellitus, Type 1 , Pancreas , Proteomics , Humans , Proteomics/methods , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/metabolism , Pancreas/cytology , Pancreas/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/cytology , Single-Cell Analysis/methods , Neural Networks, Computer , CD8-Positive T-Lymphocytes/metabolism , Image Cytometry/methods
3.
BMC Genomics ; 25(1): 427, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38689254

ABSTRACT

BACKGROUND: Current approaches to profile the single-cell transcriptomics of human pancreatic endocrine cells almost exclusively rely on freshly isolated islets. However, human islets are limited in availability. Furthermore, the extensive processing steps during islet isolation and subsequent single cell dissolution might alter gene expressions. In this work, we report the development of a single-nucleus RNA sequencing (snRNA-seq) approach with targeted islet cell enrichment for endocrine-population focused transcriptomic profiling using frozen archival pancreatic tissues without islet isolation. RESULTS: We cross-compared five nuclei isolation protocols and selected the citric acid method as the best strategy to isolate nuclei with high RNA integrity and low cytoplasmic contamination from frozen archival human pancreata. We innovated fluorescence-activated nuclei sorting based on the positive signal of NKX2-2 antibody to enrich nuclei of the endocrine population from the entire nuclei pool of the pancreas. Our sample preparation procedure generated high-quality single-nucleus gene-expression libraries while preserving the endocrine population diversity. In comparison with single-cell RNA sequencing (scRNA-seq) library generated with live cells from freshly isolated human islets, the snRNA-seq library displayed comparable endocrine cellular composition and cell type signature gene expression. However, between these two types of libraries, differential enrichments of transcripts belonging to different functional classes could be observed. CONCLUSIONS: Our work fills a technological gap and helps to unleash frozen archival pancreatic tissues for molecular profiling targeting the endocrine population. This study opens doors to retrospective mappings of endocrine cell dynamics in pancreatic tissues of complex histopathology. We expect that our protocol is applicable to enrich nuclei for transcriptomics studies from various populations in different types of frozen archival tissues.


Subject(s)
Cell Nucleus , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Islets of Langerhans , Nuclear Proteins , Sequence Analysis, RNA , Single-Cell Analysis , Transcription Factors , Humans , Islets of Langerhans/metabolism , Islets of Langerhans/cytology , Single-Cell Analysis/methods , Sequence Analysis, RNA/methods , Cell Nucleus/genetics , Cell Nucleus/metabolism , Gene Expression Profiling/methods , Pancreas/metabolism , Pancreas/cytology , Transcriptome
4.
bioRxiv ; 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38463969

ABSTRACT

Background and aims: Pancreatic ducts form an intricate network of tubules that secrete bicarbonate and drive acinar secretions into the duodenum. This network is formed by centroacinar cells, terminal, intercalated, intracalated ducts, and the main pancreatic duct. Ductal heterogeneity at the single-cell level has been poorly characterized; therefore, our understanding of the role of ductal cells in pancreas regeneration and exocrine pathogenesis has been hampered by the limited knowledge and unexplained diversity within the ductal network. Methods: We used scRNA-seq to comprehensively characterize mouse ductal heterogeneity at single-cell resolution of the entire ductal epithelium from centroacinar cells to the main duct. Moreover, we used organoid cultures, injury models and pancreatic tumor samples to interrogate the role of novel ductal populations in pancreas regeneration and exocrine pathogenesis. Results: We have identified the coexistence of 15 ductal populations within the healthy pancreas and characterized their organoid formation capacity and endocrine differentiation potential. Cluster isolation and subsequent culturing let us identify ductal cell populations with high organoid formation capacity and endocrine and exocrine differentiation potential in vitro , including Wnt-responsive-population, ciliated-population and FLRT3 + cells. Moreover, we have characterized the location of these novel ductal populations in healthy pancreas, chronic pancreatitis, and tumor samples, highlighting a putative role of WNT-responsive, IFN-responsive and EMT-populations in pancreatic exocrine pathogenesis as their expression increases in chronic pancreatitis and PanIN lesions. Conclusions: In light of our discovery of previously unidentified ductal populations, we unmask the potential roles of specific ductal populations in pancreas regeneration and exocrine pathogenesis.

5.
bioRxiv ; 2023 Jan 18.
Article in English | MEDLINE | ID: mdl-36712052

ABSTRACT

Cellular composition and anatomical organization influence normal and aberrant organ functions. Emerging spatial single-cell proteomic assays such as Image Mass Cytometry (IMC) and Co-Detection by Indexing (CODEX) have facilitated the study of cellular composition and organization by enabling high-throughput measurement of cells and their localization directly in intact tissues. However, annotation of cell types and quantification of their relative localization in tissues remain challenging. To address these unmet needs, we developed AnnoSpat (Annotator and Spatial Pattern Finder) that uses neural network and point process algorithms to automatically identify cell types and quantify cell-cell proximity relationships. Our study of data from IMC and CODEX show the superior performance of AnnoSpat in rapid and accurate annotation of cell types compared to alternative approaches. Moreover, the application of AnnoSpat to type 1 diabetic, non-diabetic autoantibody-positive, and non-diabetic organ donor cohorts recapitulated known islet pathobiology and showed differential dynamics of pancreatic polypeptide (PP) cell abundance and CD8+ T cells infiltration in islets during type 1 diabetes progression.

6.
Endocrinology ; 163(11)2022 10 11.
Article in English | MEDLINE | ID: mdl-36130190

ABSTRACT

Epidemiology studies demonstrate that women are at a significantly lower risk of developing type 2 diabetes (T2D) compared to men. However, the molecular basis of this risk difference is not well understood. In this study, we examined the sex differences in the genetic programs of pancreatic endocrine cells. We combined pancreas perifusion data and single-cell genomic data from our laboratory and from publicly available data sets to investigate multiple axes of the sex differences in the human pancreas at the single-cell type and single-cell level. We systematically compared female and male islet secretion function, gene expression program, and regulatory principles of pancreatic endocrine cells. The perifusion data indicate that female endocrine cells have a higher secretion capacity than male endocrine cells. Single-cell RNA-sequencing analysis suggests that endocrine cells in male controls have molecular signatures that resemble T2D. In addition, we identified genomic elements associated with genome-wide association study T2D loci to have differential accessibility between female and male delta cells. These genomic elements may play a sex-specific causal role in the pathogenesis of T2D. We provide molecular mechanisms that explain the differential risk of T2D between women and men. Knowledge gained from our study will accelerate the development of diagnostics and therapeutics in sex-aware precision medicine for diabetes.


Subject(s)
Diabetes Mellitus, Type 2 , Islets of Langerhans , Diabetes Mellitus, Type 2/metabolism , Female , Genome-Wide Association Study , Humans , Islets of Langerhans/metabolism , Male , Pancreas/metabolism , RNA/metabolism , Sex Characteristics
7.
Anal Chim Acta ; 1212: 339942, 2022 Jun 15.
Article in English | MEDLINE | ID: mdl-35623790

ABSTRACT

Insulin secreted from islets of Langerhans is the main hormone to reduce blood glucose. Examination of insulin secretion patterns at the single islet level reveals functional differences in the timings and patterns of release. This heterogeneous response highlights the importance of developing systems to measure dynamic release from small numbers of islets in parallel. Toward this, we describe fluorescence anisotropy imaging immunoassays as a relatively simple method for increased throughput of islet secretion measurements. In this system, vacuum pressure from a syringe pump pulled perfusate from 12 islet chambers and reagents into 12 parallel mixing channels for a competitive immunoassay. Light from a Xe arc lamp was filtered and polarized prior to focusing on the microfluidic device at the region where the 12 mixing channels converged. Emission was collected and passed through vertical and horizontal emission polarizers housed in an automated filter wheel before being imaged with a sCMOS camera for the determination of anisotropy. This microfluidic system was tested by monitoring insulin release from groups of murine and human islets. Heterogeneity was observed in the islet traces; however, the presence of islets affected the resistance of the islet chambers, hampering insulin quantification. Nonetheless, this microfluidic system is a step towards increasing the throughput of hormone release measurements from islets of Langerhans.


Subject(s)
Insulin , Optical Imaging , Animals , Anisotropy , Fluorescence Polarization , Humans , Immunoassay , Mice
8.
Nat Metab ; 4(2): 284-299, 2022 02.
Article in English | MEDLINE | ID: mdl-35228745

ABSTRACT

Type 1 diabetes (T1D) is an autoimmune disease in which immune cells destroy insulin-producing beta cells. The aetiology of this complex disease is dependent on the interplay of multiple heterogeneous cell types in the pancreatic environment. Here, we provide a single-cell atlas of pancreatic islets of 24 T1D, autoantibody-positive and nondiabetic organ donors across multiple quantitative modalities including ~80,000 cells using single-cell transcriptomics, ~7,000,000 cells using cytometry by time of flight and ~1,000,000 cells using in situ imaging mass cytometry. We develop an advanced integrative analytical strategy to assess pancreatic islets and identify canonical cell types. We show that a subset of exocrine ductal cells acquires a signature of tolerogenic dendritic cells in an apparent attempt at immune suppression in T1D donors. Our multimodal analyses delineate cell types and processes that may contribute to T1D immunopathogenesis and provide an integrative procedure for exploration and discovery of human pancreatic function.


Subject(s)
Diabetes Mellitus, Type 1 , Insulin-Secreting Cells , Islets of Langerhans , Humans , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Pancreas/metabolism , Pancreatic Hormones/metabolism
9.
Front Endocrinol (Lausanne) ; 12: 736286, 2021.
Article in English | MEDLINE | ID: mdl-34566896

ABSTRACT

NEUROGENIN3+ (NEUROG3+) cells are considered to be pancreatic endocrine progenitors. Our current knowledge on the molecular program of NEUROG3+ cells in humans is largely extrapolated from studies in mice. We hypothesized that single-cell RNA-seq enables in-depth exploration of the rare NEUROG3+ cells directly in humans. We aligned four large single-cell RNA-seq datasets from postnatal human pancreas. Our integrated analysis revealed 10 NEUROG3+ epithelial cells from a total of 11,174 pancreatic cells. Noticeably, human NEUROG3+ cells clustered with mature pancreatic cells and epsilon cells displayed the highest frequency of NEUROG3 positivity. We confirmed the co-expression of NEUROG3 with endocrine markers and the high percentage of NEUROG3+ cells among epsilon cells at the protein level based on immunostaining on pancreatic tissue sections. We further identified unique genetic signatures of the NEUROG3+ cells. Regulatory network inference revealed novel transcription factors including Prospero homeobox protein 1 (PROX1) may act jointly with NEUROG3. As NEUROG3 plays a central role in endocrine differentiation, knowledge gained from our study will accelerate the development of beta cell regeneration therapies to treat diabetes.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Endocrine Cells/metabolism , Nerve Tissue Proteins/genetics , Pancreas/metabolism , Stem Cells/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Humans , Nerve Tissue Proteins/metabolism
10.
JCI Insight ; 6(7)2021 04 08.
Article in English | MEDLINE | ID: mdl-33621209

ABSTRACT

Studies of human hepatitis B virus (HBV) immune pathogenesis are hampered by limited access to liver tissues and technologies for detailed analyses. Here, utilizing imaging mass cytometry (IMC) to simultaneously detect 30 immune, viral, and structural markers in liver biopsies from patients with hepatitis B e antigen+ (HBeAg+) chronic hepatitis B, we provide potentially novel comprehensive visualization, quantitation, and phenotypic characterizations of hepatic adaptive and innate immune subsets that correlated with hepatocellular injury, histological fibrosis, and age. We further show marked correlations between adaptive and innate immune cell frequencies and phenotype, highlighting complex immune interactions within the hepatic microenvironment with relevance to HBV pathogenesis.


Subject(s)
Hepatitis B, Chronic/pathology , Image Cytometry/methods , Liver/immunology , Liver/virology , Adolescent , Adult , Age Factors , Biopsy , Child , Female , Hepatitis B e Antigens/metabolism , Hepatitis B, Chronic/immunology , Humans , Image Processing, Computer-Assisted , Immunity, Innate , Leukocyte Common Antigens/metabolism , Liver/pathology , Male , Middle Aged , Young Adult
11.
Mol Metab ; 42: 101057, 2020 12.
Article in English | MEDLINE | ID: mdl-32739450

ABSTRACT

OBJECTIVE: Dedifferentiation of pancreatic ß-cells may reduce islet function in type 2 diabetes (T2D). However, the prevalence, plasticity and functional consequences of this cellular state remain unknown. METHODS: We employed single-cell RNAseq to detail the maturation program of α- and ß-cells during human ontogeny. We also compared islets from non-diabetic and T2D individuals. RESULTS: Both α- and ß-cells mature in part by repressing non-endocrine genes; however, α-cells retain hallmarks of an immature state, while ß-cells attain a full ß-cell specific gene expression program. In islets from T2D donors, both α- and ß-cells have a less mature expression profile, de-repressing the juvenile genetic program and exocrine genes and increasing expression of exocytosis, inflammation and stress response signalling pathways. These changes are consistent with the increased proportion of ß-cells displaying suboptimal function observed in T2D islets. CONCLUSIONS: These findings provide new insights into the molecular program underlying islet cell maturation during human ontogeny and the loss of transcriptomic maturity that occurs in islets of type 2 diabetics.


Subject(s)
Cell Dedifferentiation/genetics , Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/metabolism , Cell Dedifferentiation/physiology , Computational Biology/methods , Diabetes Mellitus, Type 2/physiopathology , Exocytosis/physiology , Gene Expression/genetics , Gene Expression Profiling/methods , Gene Expression Regulation/genetics , Glucagon-Secreting Cells/metabolism , Glucagon-Secreting Cells/physiology , Humans , Inflammation/metabolism , Insulin-Secreting Cells/physiology , Islets of Langerhans/metabolism , Pancreas/metabolism , Primary Cell Culture , Signal Transduction/physiology , Single-Cell Analysis/methods , Transcriptome/genetics
12.
Proc Natl Acad Sci U S A ; 117(29): 17177-17186, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32631996

ABSTRACT

Hepatocyte nuclear factor 4α (HNF4α) is a master regulator of liver function and a tumor suppressor in hepatocellular carcinoma (HCC). In this study, we explore the reciprocal negative regulation of HNF4α and cyclin D1, a key cell cycle protein in the liver. Transcriptomic analysis of cultured hepatocyte and HCC cells found that cyclin D1 knockdown induced the expression of a large network of HNF4α-regulated genes. Chromatin immunoprecipitation-sequencing (ChIP-seq) demonstrated that cyclin D1 inhibits the binding of HNF4α to thousands of targets in the liver, thereby diminishing the expression of associated genes that regulate diverse metabolic activities. Conversely, acute HNF4α deletion in the liver induces cyclin D1 and hepatocyte cell cycle progression; concurrent cyclin D1 ablation blocked this proliferation, suggesting that HNF4α maintains proliferative quiescence in the liver, at least, in part, via repression of cyclin D1. Acute cyclin D1 deletion in the regenerating liver markedly inhibited hepatocyte proliferation after partial hepatectomy, confirming its pivotal role in cell cycle progression in this in vivo model, and enhanced the expression of HNF4α target proteins. Hepatocyte cyclin D1 gene ablation caused markedly increased postprandial liver glycogen levels (in a HNF4α-dependent fashion), indicating that the cyclin D1-HNF4α axis regulates glucose metabolism in response to feeding. In AML12 hepatocytes, cyclin D1 depletion led to increased glucose uptake, which was negated if HNF4α was depleted simultaneously, and markedly elevated glycogen synthesis. To summarize, mutual repression by cyclin D1 and HNF4α coordinately controls the cell cycle machinery and metabolism in the liver.


Subject(s)
Cell Cycle/physiology , Cyclin D1/genetics , Cyclin D1/metabolism , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Liver/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Female , Gene Knockdown Techniques , Hepatocytes/metabolism , Hepatocytes/pathology , Liver Regeneration/genetics , Liver Regeneration/physiology , Male , Mice, Inbred BALB C , Mice, Knockout
13.
Cell Mol Gastroenterol Hepatol ; 9(1): 121-143, 2020.
Article in English | MEDLINE | ID: mdl-31629814

ABSTRACT

BACKGROUND & AIMS: The adult liver is the main detoxification organ and routinely is exposed to environmental insults but retains the ability to restore its mass and function upon tissue damage. However, extensive injury can lead to liver failure, and chronic injury causes fibrosis, cirrhosis, and hepatocellular carcinoma. Currently, the transcriptional regulation of organ repair in the adult liver is incompletely understood. METHODS: We isolated nuclei from quiescent as well as repopulating hepatocytes in a mouse model of hereditary tyrosinemia, which recapitulates the injury and repopulation seen in toxic liver injury in human beings. We then performed the assay for transposase accessible chromatin with high-throughput sequencing specifically in repopulating hepatocytes to identify differentially accessible chromatin regions and nucleosome positioning. In addition, we used motif analysis to predict differential transcription factor occupancy and validated the in silico results with chromatin immunoprecipitation followed by sequencing for hepatocyte nuclear factor 4α (HNF4α) and CCCTC-binding factor (CTCF). RESULTS: Chromatin accessibility in repopulating hepatocytes was increased in the regulatory regions of genes promoting proliferation and decreased in the regulatory regions of genes involved in metabolism. The epigenetic changes at promoters and liver enhancers correspond with the regulation of gene expression, with enhancers of many liver function genes showing a less accessible state during the regenerative process. Moreover, increased CTCF occupancy at promoters and decreased HNF4α binding at enhancers implicate these factors as key drivers of the transcriptomic changes in replicating hepatocytes that enable liver repopulation. CONCLUSIONS: Our analysis of hepatocyte-specific epigenomic changes during liver repopulation identified CTCF and HNF4α as key regulators of hepatocyte proliferation and regulation of metabolic programs. Thus, liver repopulation in the setting of toxic injury makes use of both general transcription factors (CTCF) for promoter activation, and reduced binding by a hepatocyte-enriched factor (HNF4α) to temporarily limit enhancer activity. All sequencing data in this study were deposited to the Gene Expression Omnibus database and can be downloaded with accession number GSE109466.


Subject(s)
CCCTC-Binding Factor/metabolism , Chromatin/metabolism , Hepatocyte Nuclear Factor 4/metabolism , Liver Regeneration/genetics , Tyrosinemias/pathology , Animals , CCCTC-Binding Factor/genetics , Cell Nucleus/metabolism , Cell Proliferation , Disease Models, Animal , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic , Gene Expression Profiling , Hepatocyte Nuclear Factor 4/genetics , Hepatocytes/cytology , Hepatocytes/physiology , High-Throughput Nucleotide Sequencing , Humans , Hydrolases/genetics , Liver/cytology , Liver/pathology , Mice , Mice, Knockout , Promoter Regions, Genetic/genetics , Tyrosinemias/genetics
14.
Gastroenterology ; 158(4): 1044-1057.e17, 2020 03.
Article in English | MEDLINE | ID: mdl-31759059

ABSTRACT

BACKGROUND & AIMS: Liver regeneration is impaired in mice with hepatocyte-specific deficiencies in microRNA (miRNA) processing, but it is not clear which miRNAs regulate this process. We developed a high-throughput screen to identify miRNAs that regulate hepatocyte repopulation after toxic liver injury using fumarylacetoacetate hydrolase-deficient mice. METHODS: We constructed plasmid pools encoding more than 30,000 tough decoy miRNA inhibitors (hairpin nucleic acids designed to specifically inhibit interactions between miRNAs and their targets) to target hepatocyte miRNAs in a pairwise manner. The plasmid libraries were delivered to hepatocytes in fumarylacetoacetate hydrolase-deficient mice at the time of liver injury via hydrodynamic tail-vein injection. Integrated transgene-containing transposons were quantified after liver repopulation via high-throughput sequencing. Changes in polysome-bound transcripts after miRNA inhibition were determined using translating ribosome affinity purification followed by high-throughput sequencing. RESULTS: Analyses of tough decoy abundance in hepatocyte genomic DNA and input plasmid pools identified several thousand miRNA inhibitors that were significantly depleted or increased after repopulation. We classified a subset of miRNA binding sites as those that have strong effects on liver repopulation, implicating the targeted hepatocyte miRNAs as regulators of this process. We then generated a high-content map of pairwise interactions between 171 miRNA-binding sites and identified synergistic and redundant effects. CONCLUSIONS: We developed a screen to identify miRNAs that regulate liver repopulation after injury in live mice.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Liver Regeneration/genetics , Liver/injuries , MicroRNAs/analysis , Animals , Chromosome Mapping , Hepatocytes/physiology , Hydrolases/deficiency , Liver/physiopathology , Mice , MicroRNAs/antagonists & inhibitors , Plasmids , RNA-Binding Proteins/analysis
15.
Cell Metab ; 29(3): 769-783.e4, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30713110

ABSTRACT

The interaction between the immune system and endocrine cells in the pancreas is crucial for the initiation and progression of type 1 diabetes (T1D). Imaging mass cytometry (IMC) enables multiplexed assessment of the abundance and localization of more than 30 proteins on the same tissue section at 1-µm resolution. Herein, we have developed a panel of 33 antibodies that allows for the quantification of key cell types including pancreatic exocrine cells, islet cells, immune cells, and stromal components. We employed this panel to analyze 12 pancreata obtained from donors with clinically diagnosed T1D and 6 pancreata from non-diabetic controls. In the pancreata from donors with T1D, we simultaneously visualized significant alterations in islet architecture, endocrine cell composition, and immune cell presentation. Indeed, we demonstrate the utility of IMC to investigate complex events on the cellular level that will provide new insights on the pathophysiology of T1D.


Subject(s)
Diabetes Mellitus, Type 1 , Image Cytometry/methods , Islets of Langerhans , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Endocrine Cells/immunology , Endocrine Cells/metabolism , Humans , Immune System/immunology , Immune System/metabolism , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Islets of Langerhans/ultrastructure
16.
J Clin Invest ; 129(1): 209-214, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30352048

ABSTRACT

The loss of insulin-secreting ß cells is characteristic among type I and type II diabetes. Stimulating proliferation to expand sources of ß cells for transplantation remains a challenge because adult ß cells do not proliferate readily. The cell cycle inhibitor p57 has been shown to control cell division in human ß cells. Expression of p57 is regulated by the DNA methylation status of the imprinting control region 2 (ICR2), which is commonly hypomethylated in Beckwith-Wiedemann syndrome patients who exhibit massive ß cell proliferation. We hypothesized that targeted demethylation of the ICR2 using a transcription activator-like effector protein fused to the catalytic domain of TET1 (ICR2-TET1) would repress p57 expression and promote cell proliferation. We report here that overexpression of ICR2-TET1 in human fibroblasts reduces p57 expression levels and increases proliferation. Furthermore, human islets overexpressing ICR2-TET1 exhibit repression of p57 with concomitant upregulation of Ki-67 while maintaining glucose-sensing functionality. When transplanted into diabetic, immunodeficient mice, the epigenetically edited islets show increased ß cell replication compared with control islets. These findings demonstrate that epigenetic editing is a promising tool for inducing ß cell proliferation, which may one day alleviate the scarcity of transplantable ß cells for the treatment of diabetes.


Subject(s)
Beckwith-Wiedemann Syndrome/metabolism , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , DNA Demethylation , Genetic Loci , Insulin-Secreting Cells/metabolism , Up-Regulation , Beckwith-Wiedemann Syndrome/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Insulin-Secreting Cells/pathology , Ki-67 Antigen/biosynthesis
17.
Cell Metab ; 29(3): 539-544, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30581120

ABSTRACT

In the past 3 years, we have seen a flurry of publications on single-cell RNA sequencing (RNA-seq) analyses of pancreatic islets from mouse and human. This technology holds the promise to refine cell-type signatures and discover cellular heterogeneity among the canonical endocrine cell types such as the glucagon-producing α and insulin-producing ß cells, going as far as suggesting new subtypes. In addition, single-cell RNA-seq has the ability to characterize rare endocrine cell types that are not captured by prior bulk analysis. With transcriptomics data from individual endocrine cells, cellular states can be profiled both along developmental processes and during the emergence of metabolic diseases. However, the promises of this new technology have not yet been met in full. While the methodology for the first time enabled the transcriptional definition of rare endocrine cell types such as ghrelin-producing ɛ cells, some of the conclusions regarding cell-type-specific gene expression changes in type 2 diabetes might need to be revisited once larger sample sizes become available. Data generation and analysis are continuously improving single-cell RNA-seq approaches and are helping us to understand the (mal)adaptations of the islet cells during development, metabolic challenge, and disease.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucagon-Secreting Cells/metabolism , Insulin-Secreting Cells/metabolism , RNA, Messenger/metabolism , RNA-Seq/methods , Single-Cell Analysis/methods , Animals , Gene Expression Profiling/methods , Glucagon-Secreting Cells/cytology , Glucagon-Secreting Cells/pathology , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/pathology , Mice
18.
Diabetes ; 67(9): 1807-1815, 2018 09.
Article in English | MEDLINE | ID: mdl-30084829

ABSTRACT

Type 2 diabetes mellitus (T2DM) is characterized by the inability of the insulin-producing ß-cells to overcome insulin resistance. We previously identified an imprinted region on chromosome 14, the DLK1-MEG3 locus, as being downregulated in islets from humans with T2DM. In this study, using targeted epigenetic modifiers, we prove that increased methylation at the promoter of Meg3 in mouse ßTC6 ß-cells results in decreased transcription of the maternal transcripts associated with this locus. As a result, the sensitivity of ß-cells to cytokine-mediated oxidative stress was increased. Additionally, we demonstrate that an evolutionarily conserved intronic region at the MEG3 locus can function as an enhancer in ßTC6 ß-cells. Using circular chromosome conformation capture followed by high-throughput sequencing, we demonstrate that the promoter of MEG3 physically interacts with this novel enhancer and other putative regulatory elements in this imprinted region in human islets. Remarkably, this enhancer is bound in an allele-specific manner by the transcription factors FOXA2, PDX1, and NKX2.2. Overall, these data suggest that the intronic MEG3 enhancer plays an important role in the regulation of allele-specific expression at the imprinted DLK1-MEG3 locus in human ß-cells, which in turn impacts the sensitivity of ß-cells to cytokine-mediated oxidative stress.


Subject(s)
DNA Methylation , Diabetes Mellitus, Type 2/metabolism , Gene Expression Regulation , Intercellular Signaling Peptides and Proteins/metabolism , Islets of Langerhans/metabolism , Membrane Proteins/metabolism , Promoter Regions, Genetic , RNA, Long Noncoding/metabolism , Animals , Calcium-Binding Proteins , Cell Line , Cytokines/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/chemistry , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Diabetes Mellitus, Type 2/pathology , Enhancer Elements, Genetic , Epigenesis, Genetic , Genetic Loci , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Islets of Langerhans/pathology , Locus Control Region , Membrane Proteins/genetics , Mice , Mutation , Nuclear Proteins , Oxidative Stress/drug effects , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tissue Banks , Transcription Factors/genetics , Transcription Factors/metabolism
19.
Nature ; 560(7718): E29, 2018 08.
Article in English | MEDLINE | ID: mdl-29977061

ABSTRACT

Change history: In this Letter, the surname of author Efi E. Massasa was misspelled 'Massassa'. This error has been corrected online.

20.
Nature ; 557(7704): 242-246, 2018 05.
Article in English | MEDLINE | ID: mdl-29720649

ABSTRACT

Tissues that undergo rapid cellular turnover, such as the mammalian haematopoietic system or the intestinal epithelium, are dependent on stem and progenitor cells that proliferate to provide differentiated cells to maintain organismal health. Stem and progenitor cells, in turn, are thought to rely on signals and growth factors provided by local niche cells to support their function and self-renewal. Several cell types have been hypothesized to provide the signals required for the proliferation and differentiation of the intestinal stem cells in intestinal crypts1-6. Here we identify subepithelial telocytes as an important source of Wnt proteins, without which intestinal stem cells cannot proliferate and support epithelial renewal. Telocytes are large but rare mesenchymal cells that are marked by expression of FOXL1 and form a subepithelial plexus that extends from the stomach to the colon. While supporting the entire epithelium, FOXL1+ telocytes compartmentalize the production of Wnt ligands and inhibitors to enable localized pathway activation. Conditional genetic ablation of porcupine (Porcn), which is required for functional maturation of all Wnt proteins, in mouse FOXL1+ telocytes causes rapid cessation of Wnt signalling to intestinal crypts, followed by loss of proliferation of stem and transit amplifying cells and impaired epithelial renewal. Thus, FOXL1+ telocytes are an important source of niche signals to intestinal stem cells.


Subject(s)
Cell Self Renewal , Intestinal Mucosa/cytology , Telocytes/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , Acyltransferases/deficiency , Acyltransferases/genetics , Acyltransferases/metabolism , Animals , Cell Proliferation , Forkhead Transcription Factors/metabolism , Ligands , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...