Ductal Cells

14,000,000 Leading Edge Experts on the ideXlab platform

Scan Science and Technology

Contact Leading Edge Experts & Companies

Scan Science and Technology

Contact Leading Edge Experts & Companies

The Experts below are selected from a list of 28869 Experts worldwide ranked by ideXlab platform

Riccardo Perfetti - One of the best experts on this subject based on the ideXlab platform.

  • glucagon like peptide 1 induces differentiation of islet duodenal homeobox 1 positive pancreatic Ductal Cells into insulin secreting Cells
    Diabetes, 2001
    Co-Authors: Christopher V E Wright, Riccardo Perfetti
    Abstract:

    Glucagon-like peptide-1 (GLP-1) is an incretin hormone capable of restoring normal glucose tolerance in aging glucose-intolerant Wistar rats. Whether the antidiabetic properties of GLP-1 are exclusively due to its insulin secretory activity remains to be determined. A GLP-1–dependent differentiation of pancreatic precursor Cells into mature β-Cells has recently been proposed. The aim of this study was to investigate whether pancreatic Ductal epithelial Cells could be differentiated into insulin-secreting Cells by exposing them to GLP-1. Rat (ARIP) and human (PANC-1) cell lines, both derived from the pancreatic Ductal epithelium, were used to test this hypothesis. A major difference distinguishes these two cell lines: whereas ARIP Cells spontaneously express the β-cell differentiation factor islet duodenal homeobox-1 (IDX-1), PANC-1 Cells are characteristically IDX-1 negative. GLP-1 induced the differentiation of ARIP Cells into insulin-synthesizing Cells, although it did not affect the phenotype of PANC-1 Cells, as determined by fluorescence-activated cell sorting (FACS) analysis. Differentiation of ARIP Cells by exposure to human GLP-1 occurs in a time- and dose-dependent manner, and this is associated with an increase in IDX-1 and insulin mRNA levels. Secretion of insulin was also induced in a parallel manner, and it was regulated by the concentration of glucose in the culture medium. Interestingly, PANC-1 Cells, when stably transfected with human IDX-1, gained responsiveness to GLP-1 and were able to differentiate into β-Cells, as determined by FACS analysis, insulin gene expression, intracellular insulin content, and insulin accumulation in the culture medium. Finally, we demonstrated that the receptor for GLP-1 is constitutively expressed by ARIP and PANC-1 Cells and that the mRNA level for this transcript was increased by cellular transfection with human IDX-1. In summary, our study provides evidence that GLP-1 is a differentiation factor for pancreatic Ductal Cells and that its effect requires the expression of IDX-1.

  • glucagon like peptide 1 induces differentiation of islet duodenal homeobox 1 positive pancreatic Ductal Cells into insulin secreting Cells
    Diabetes, 2001
    Co-Authors: Hongxiang Hui, Christopher V E Wright, Riccardo Perfetti
    Abstract:

    Glucagon-like peptide-1 (GLP-1) is an incretin hormone capable of restoring normal glucose tolerance in aging glucose-intolerant Wistar rats. Whether the antidiabetic properties of GLP-1 are exclusively due to its insulin secretory activity remains to be determined. A GLP-1-dependent differentiation of pancreatic precursor Cells into mature beta-Cells has recently been proposed. The aim of this study was to investigate whether pancreatic Ductal epithelial Cells could be differentiated into insulin-secreting Cells by exposing them to GLP-1. Rat (ARIP) and human (PANC-1) cell lines, both derived from the pancreatic Ductal epithelium, were used to test this hypothesis. A major difference distinguishes these two cell lines: whereas ARIP Cells spontaneously express the beta-cell differentiation factor islet duodenal homeobox-1 (IDX-1), PANC-1 Cells are characteristically IDX-1 negative. GLP-1 induced the differentiation of ARIP Cells into insulin-synthesizing Cells, although it did not affect the phenotype of PANC-1 Cells, as determined by fluorescence-activated cell sorting (FACS) analysis. Differentiation of ARIP Cells by exposure to human GLP-1 occurs in a time- and dose-dependent manner, and this is associated with an increase in IDX-1 and insulin mRNA levels. Secretion of insulin was also induced in a parallel manner, and it was regulated by the concentration of glucose in the culture medium. Interestingly, PANC-1 Cells, when stably transfected with human IDX-1, gained responsiveness to GLP-1 and were able to differentiate into beta-Cells, as determined by FACS analysis, insulin gene expression, intracellular insulin content, and insulin accumulation in the culture medium. Finally, we demonstrated that the receptor for GLP-1 is constitutively expressed by ARIP and PANC-1 Cells and that the mRNA level for this transcript was increased by cellular transfection with human IDX-1. In summary, our study provides evidence that GLP-1 is a differentiation factor for pancreatic Ductal Cells and that its effect requires the expression of IDX-1.

Anil K. Rustgi - One of the best experts on this subject based on the ideXlab platform.

  • Culturing primary mouse pancreatic Ductal Cells.
    Cold Spring Harbor protocols, 2015
    Co-Authors: Maximilian Reichert, Andrew D. Rhim, Anil K. Rustgi
    Abstract:

    The most common subtype of pancreatic cancer is pancreatic Ductal adenocarcinoma (PDAC). PDAC resembles Ductal Cells morphologically. To study pancreatic Ductal cell (PDC) and pancreatic intraepithelial neoplasia (PanIN)/PDAC biology, it is essential to have reliable in vitro culture conditions. Here we describe a methodology to isolate, culture, and passage PDCs and duct-like Cells from the mouse pancreas. It can be used to isolate Cells from genetically engineered mouse models (GEMMs), providing a valuable tool to study disease models in vitro to complement in vivo findings. The culture conditions allow epithelial Cells to outgrow fibroblast and other "contaminating" cell types within a few passages. However, the resulting cultures, although mostly epithelial, are not completely devoid of fibroblasts. Regardless, this protocol provides guidelines for a robust in vitro culture system to isolate, maintain, and expand primary pancreatic Ductal epithelial Cells. It can be applied to virtually all GEMMs of pancreatic disease and other diseases and cancers that arise from Ductal structures. Because most carcinomas resemble Ductal structures, this protocol has utility in the study of other cancers in addition to PDAC, such as breast and prostate cancers.

  • Isolation, culture and genetic manipulation of mouse pancreatic Ductal Cells
    Nature Protocols, 2013
    Co-Authors: Maximilian Reichert, Shigetsugu Takano, Steffen Heeg, Basil Bakir, Gregory P Botta, Anil K. Rustgi
    Abstract:

    The most common subtype of pancreatic cancer is pancreatic Ductal adenocarcinoma (PDAC). PDAC resembles duct Cells morphologically and, to some extent, at a molecular level. Recently, genetic-lineage labeling has become popular in the field of tumor biology in order to study cell-fate decisions or to trace cancer Cells in the mouse. However, certain biological questions require a nongenetic labeling approach to purify a distinct cell population in the pancreas. Here we describe a protocol for isolating mouse pancreatic Ductal epithelial Cells and ductlike Cells directly in vivo using Ductal-specific Dolichos biflorus agglutinin (DBA) lectin labeling followed by magnetic bead separation. Isolated Cells can be cultured (in two or three dimensions), manipulated by lentiviral transduction to modulate gene expression and directly used for molecular studies. This approach is fast (∼4 h), affordable, results in Cells with high viability, can be performed on the bench and is applicable to virtually all genetic and nongenetic disease models of the pancreas.

  • Pancreatic Ductal Cells in development, regeneration, and neoplasia
    The Journal of clinical investigation, 2011
    Co-Authors: Maximilian Reichert, Anil K. Rustgi
    Abstract:

    The pancreas is a complex organ comprised of three critical cell lineages: islet (endocrine), acinar, and Ductal. This review will focus upon recent insights and advances in the biology of pancreatic Ductal Cells. In particular, emphasis will be placed upon the regulation of Ductal Cells by specific transcriptional factors during development as well as the underpinnings of acinar-Ductal metaplasia as an important adaptive response during injury and regeneration. We also address the potential contributions of Ductal Cells to neoplastic transformation, specifically in pancreatic Ductal adenocarcinoma.

  • successful growth and characterization of mouse pancreatic Ductal Cells functional properties of the ki rasg12v oncogene
    Gastroenterology, 2004
    Co-Authors: Franz S Schreiber, Therese B Deramaudt, Thomas Brunner, Michael I Boretti, Keith J Gooch, Doris A Stoffers, Eric J Bernhard, Anil K. Rustgi
    Abstract:

    Abstract Background & Aims: The Ki-RAS oncogene is altered in pancreatic Ductal neoplasms. Pancreatic Ductal Cells (PDCs) were purified from cytokeratin 19 (K19)- Ki-RAS G12V transgenic mice and control littermates to identify properties of Ki-Ras activation in a cell-type-specific context. Because Ki-RAS mutation has prognostic significance in patients treated with radiation, we studied the influence of Ki-RAS status on radiation survival. Methods: Pancreatic Ductal fragments from mice with Ki-RAS G12V mutation or wild-type (WT)- Ki-RAS were cultured. Growth curves, electron microscopy, flow cytometry, and analysis of signaling and cell-cycle proteins were established. Farnesyltransferase inhibitor (FTI) treatment with R115777 before and after irradiation was used to determine the effect of Ki-Ras farnesylation on cell survival. Results: PDCs from WT and K19-Ki-RAS G12V mice had features of Ductal Cells with formation of 3-dimensional structures on collagen without differences in morphology, growth, and cell-cycle distribution. This may result from up-regulation of p16INK4 and p27 Kip1 and lack of hyperstimulation of the mitogen-activated protein kinase pathway in Ki-RAS G12V PDCs. No differences in radiation survival between Ki-RAS G12V PDCs and WT PDCs were observed. However, Ki-RAS G12V PDCs expressing mutant p53 V143A had enhanced survival compared with WT PDCs transduced with p53 V143A . R115777 treatment sensitized Ki-RAS G12V PDCs and Ki-RAS G12V /p53 V143A PDCs, but not WT PDCs. Conclusions: Novel characterization of murine WT PDCs and Ki-RAS G12V PDCs is described. Induction of cell-cycle regulators and lack of mitogen-activated protein kinase hyperstimulation likely are responsible for constraining activated Ki-RAS G12V -mediated proliferation. Because its activation was required for sensitization by an FTI, R115777 may be useful against pancreatic tumors expressing oncogenic Ki-Ras.

  • the mutant k ras oncogene causes pancreatic periDuctal lymphocytic infiltration and gastric mucous neck cell hyperplasia in transgenic mice
    Cancer Research, 2003
    Co-Authors: Felix H Brembeck, Franz S Schreiber, Therese B Deramaudt, Doris A Stoffers, Linden E Craig, Ben Rhoades, Gary P Swain, Paul J Grippo, Debra G Silberg, Anil K. Rustgi
    Abstract:

    A frequent genetic alteration found in premalignant stages of pancreatic adenocarcinoma is K-ras oncogene point mutation. The mechanistic basis for the inability of K-ras mutation to transform pancreatic Ductal Cells is unclear, although cooperating events with p16 inactivation, p53 mutation, and SMAD 4 mutation are recognized to be necessary. We have generated a novel mouse model in which the cytokeratin 19 promoter, specifically active in pancreatic Ductal Cells but not other cell types of the pancreas, is fused to mutant K-ras. This is of direct relevance to human pancreatic cancer because premalignant lesions are found specifically in Ductal Cells. There is dramatic periDuctal lymphocytic infiltration in the pancreata of transgenic mice, predominantly CD4+ T lymphocytes, which may act as an adaptive immune response to activated ras-mediated signaling. In addition, gene array analysis reveals an induction of N-cadherin in transgenic mice pancreatic Ductal Cells, the significance of which relates to promotion of cell adhesion and deterrence of cell migration. Apart from these important biological considerations, there is parallel activity of the cytokeratin 19 promoter in the stem cell region of the gastric epithelium, namely in mucous neck Cells. Activated K-ras in this context causes mucous neck cell hyperplasia, a precursor to gastric adenocarcinoma. There is concomitant parietal cell decrease, which is a key step toward gastric adenocarcinoma. Taken together, we have defined how mutant K-ras signaling modulates important molecular events in the initiating events of pancreatic and gastric carcinogenesis.

Francois Pattou - One of the best experts on this subject based on the ideXlab platform.

  • coxsackievirus b4 infection of human primary pancreatic Ductal cell cultures results in impairment of differentiation into insulin producing Cells
    Viruses, 2019
    Co-Authors: Antoine Bertin, Delphine Lobert, Arthur Dechaumes, Julie Kerrconte, Francois Pattou, Famara Sane, Valery Gmyr, Didier Hober
    Abstract:

    Coxsackievirus-B4 (CV-B4) E2 can persist in the pancreatic Ductal-like Cells (Panc-1 cell line), which results in an impaired differentiation of these Cells into islet-like cell aggregates (ICA). In this study, primary pancreatic Ductal Cells obtained as a by-product of islet isolation from the pancreas of seven brain-dead adults were inoculated with CV-B4 E2, followed-up for 29 days, and the impact was investigated. Viral titers in culture supernatants were analyzed throughout the culture. Intracellular viral RNA was detected by RT-PCR. Levels of Ductal cell marker CK19 mRNA and of insulin mRNA were evaluated by qRT-PCR. The concentration of c-peptide in supernatants was determined by ELISA. Ductal Cells exposed to trypsin and serum-free medium formed ICA and resulted in an increased insulin secretion. Ductal Cells from five brain-dead donors were severely damaged by CV-B4 E2, whereas the virus persisted in cultures of Cells obtained from the other two. The ICAs whose formation was induced on day 14 post-inoculation were scarce and appeared tiny in infected cultures. Also, insulin mRNA expression and c-peptide levels were strongly reduced compared to the controls. In conclusion, CV-B4 E2 lysed human primary pancreatic Ductal Cells or persisted in these Cells, which resulted in the impairment of differentiation into insulin-producing Cells.

  • coxsackievirus b4 can infect human pancreas Ductal Cells and persist in Ductal like cell cultures which results in inhibition of pdx1 expression and disturbed formation of islet like cell aggregates
    Cellular and Molecular Life Sciences, 2013
    Co-Authors: Famara Sane, Julie Kerrconte, Francois Pattou, Valery Gmyr, Delphine Caloone, Ilka Engelmann, Sandrine Belaich, Rachel Desailloud, Didier Hober
    Abstract:

    The role of enteroviruses, especially Coxsackievirus B (CVB), in type 1 diabetes is suspected, but the mechanisms of the virus-induced or aggravated pathogenesis of the disease are unknown. The hypothesis of an enterovirus-induced disturbance of pancreatic β-Cells regeneration has been investigated in the human system. The infection of human pancreas Ductal Cells and pancreatic duct cell line, PANC-1, with CVB4E2 has been studied. Primary Ductal Cells and PANC-1 Cells were infectable with CVB4E2 and a RT-PCR assay without extraction displayed that a larger proportion of Cells harbored viral RNA than predicted by the detection of the viral capsid protein VP1 by indirect immunofluorescence. The detection of intracellular positive- and negative-strands of enterovirus genomes in cellular extracts by RT-PCR and the presence of infectious particles in supernatant fluids during the 37 weeks of monitoring demonstrated that CVB4E2 could persist in the pancreatic duct cell line. A persistent infection of these Cells resulted in an impaired expression of Pdx1, a transcription factor required for the formation of endocrine pancreas, and a disturbed formation of islet-like cell aggregates of which the viability was decreased. These data support the hypothesis of an impact of enteroviruses onto pancreatic Ductal Cells which are involved in the renewal of pancreatic β-Cells.

  • rapid purification of human Ductal Cells from human pancreatic fractions with surface antibody ca19 9
    Biochemical and Biophysical Research Communications, 2004
    Co-Authors: Valery Gmyr, Francois Pattou, Sandrine Belaich, Ghaffar Muharram, Bruno Lukowiak, Brigitte Vandewalle, Julie Kerrconte
    Abstract:

    Abstract Generating human insulin-secreting Cells for cell therapy of diabetes represents a highly competitive world challenge. Human Ductal Cells can give rise to islets in vivo and in vitro. The goal of this study was to devise a rapid sorting method to highly purify human Ductal Cells from pancreatic tissue using a pan-Ductal membrane antibody carbohydrate antigen 19-9 (CA19-9). Human pancreatic sections confirmed antibody specificity. The human exocrine fraction (30% Ductal Cells) was sorted with magnetic bead technology or by FACS. Immunocytochemistry post-sorting determined Ductal cell content. The manual magnetic bead technique resulted in 74% ± 2 ( n =4) CA19 positive Cells. Whereas the automated AutoMACS technique ( n =5) yielded 92.6% ± 0.5 CA19-9 positive Cells with only a minor beta cell contamination (0.2% ± 0.03); cell yield post-sorting was 12.9% ± 2.5 (1.69 ± 0.41 × 10 6  Cells) with 51.7% + 6.5 ( n =5) viability post-sorting. The FACS ( n =6) resulted in 97.1% ± 0.82 CA19-9 positive Cells, a cell yield of 25.5% ± 5.6 (5.03 ± 1.0 × 10 6 ), with 72.1% ± 6.1 viability post-sorting.

  • adult human cytokeratin 19 positive Cells reexpress insulin promoter factor 1 in vitro further evidence for pluripotent pancreatic stem Cells in humans
    Diabetes, 2000
    Co-Authors: Valery Gmyr, Julie Kerrconte, Sandrine Belaich, Brigitte Vandewalle, Emmanuelle Leteurtre, M C Vantyghem, M Lecomtehoucke, C Proye, J Lefebvre, Francois Pattou
    Abstract:

    Human pancreatic Cells with a typical Ductal phenotype and potential to proliferate can be obtained in vitro, but the differentiation capacity of these putative human pancreatic stem Cells remains to be documented. We investigated the protein and mRNA expression of insulin promoter factor 1 (IPF-1) (or pancreas/duodenal homeobox 1), a transcription factor critical for pancreatic development and endocrine cell neogenesis, in human pancreatic Ductal Cells derived from cultured exocrine tissue. In vitro, exocrine Cells rapidly adhered (within 12 h) and were de-/transdifferentiated to Ductal Cells after 3 days with a dramatic loss of amylase protein (n = 4, 92 +/- 3.3%, P < 0.05 vs. day 1) and a simultaneous increase of Ductal cytokeratin 19 protein (n = 4, 3.4-fold on day 3 and 7-fold on day 9, P < 0.05 vs. day 1). IPF-1 protein and mRNA levels were low to undetectable in exocrine preparations before culture. After 2 days of culture, a 3.2-fold increase in IPF-1 protein was observed, corresponding to the characteristic 46-kDa protein in Western blots. Reverse transcriptase-polymerase chain reaction confirmed a 10.5-fold increase in IPF-1 mRNA levels after 3 days of culture (n = 5, P < 0.001 vs. day 1). Double immunocytochemistry showed direct evidence that IPF-1 appeared during culture in these exocrine-derived Ductal Cells (cytokeratin 7-positive) and was not merely in contaminating endocrine Cells (chromogranin A-positive). In conclusion, we describe herein the first converging evidence on both the molecular and protein level that human Cells with a typical Ductal phenotype derived ex vivo from pancreatic exocrine tissue (obtained from healthy donors) can reexpress IPF-1 in culture, suggesting their pancreatic precursor/stem cell potential.

Christopher V E Wright - One of the best experts on this subject based on the ideXlab platform.

  • glucagon like peptide 1 induces differentiation of islet duodenal homeobox 1 positive pancreatic Ductal Cells into insulin secreting Cells
    Diabetes, 2001
    Co-Authors: Christopher V E Wright, Riccardo Perfetti
    Abstract:

    Glucagon-like peptide-1 (GLP-1) is an incretin hormone capable of restoring normal glucose tolerance in aging glucose-intolerant Wistar rats. Whether the antidiabetic properties of GLP-1 are exclusively due to its insulin secretory activity remains to be determined. A GLP-1–dependent differentiation of pancreatic precursor Cells into mature β-Cells has recently been proposed. The aim of this study was to investigate whether pancreatic Ductal epithelial Cells could be differentiated into insulin-secreting Cells by exposing them to GLP-1. Rat (ARIP) and human (PANC-1) cell lines, both derived from the pancreatic Ductal epithelium, were used to test this hypothesis. A major difference distinguishes these two cell lines: whereas ARIP Cells spontaneously express the β-cell differentiation factor islet duodenal homeobox-1 (IDX-1), PANC-1 Cells are characteristically IDX-1 negative. GLP-1 induced the differentiation of ARIP Cells into insulin-synthesizing Cells, although it did not affect the phenotype of PANC-1 Cells, as determined by fluorescence-activated cell sorting (FACS) analysis. Differentiation of ARIP Cells by exposure to human GLP-1 occurs in a time- and dose-dependent manner, and this is associated with an increase in IDX-1 and insulin mRNA levels. Secretion of insulin was also induced in a parallel manner, and it was regulated by the concentration of glucose in the culture medium. Interestingly, PANC-1 Cells, when stably transfected with human IDX-1, gained responsiveness to GLP-1 and were able to differentiate into β-Cells, as determined by FACS analysis, insulin gene expression, intracellular insulin content, and insulin accumulation in the culture medium. Finally, we demonstrated that the receptor for GLP-1 is constitutively expressed by ARIP and PANC-1 Cells and that the mRNA level for this transcript was increased by cellular transfection with human IDX-1. In summary, our study provides evidence that GLP-1 is a differentiation factor for pancreatic Ductal Cells and that its effect requires the expression of IDX-1.

  • glucagon like peptide 1 induces differentiation of islet duodenal homeobox 1 positive pancreatic Ductal Cells into insulin secreting Cells
    Diabetes, 2001
    Co-Authors: Hongxiang Hui, Christopher V E Wright, Riccardo Perfetti
    Abstract:

    Glucagon-like peptide-1 (GLP-1) is an incretin hormone capable of restoring normal glucose tolerance in aging glucose-intolerant Wistar rats. Whether the antidiabetic properties of GLP-1 are exclusively due to its insulin secretory activity remains to be determined. A GLP-1-dependent differentiation of pancreatic precursor Cells into mature beta-Cells has recently been proposed. The aim of this study was to investigate whether pancreatic Ductal epithelial Cells could be differentiated into insulin-secreting Cells by exposing them to GLP-1. Rat (ARIP) and human (PANC-1) cell lines, both derived from the pancreatic Ductal epithelium, were used to test this hypothesis. A major difference distinguishes these two cell lines: whereas ARIP Cells spontaneously express the beta-cell differentiation factor islet duodenal homeobox-1 (IDX-1), PANC-1 Cells are characteristically IDX-1 negative. GLP-1 induced the differentiation of ARIP Cells into insulin-synthesizing Cells, although it did not affect the phenotype of PANC-1 Cells, as determined by fluorescence-activated cell sorting (FACS) analysis. Differentiation of ARIP Cells by exposure to human GLP-1 occurs in a time- and dose-dependent manner, and this is associated with an increase in IDX-1 and insulin mRNA levels. Secretion of insulin was also induced in a parallel manner, and it was regulated by the concentration of glucose in the culture medium. Interestingly, PANC-1 Cells, when stably transfected with human IDX-1, gained responsiveness to GLP-1 and were able to differentiate into beta-Cells, as determined by FACS analysis, insulin gene expression, intracellular insulin content, and insulin accumulation in the culture medium. Finally, we demonstrated that the receptor for GLP-1 is constitutively expressed by ARIP and PANC-1 Cells and that the mRNA level for this transcript was increased by cellular transfection with human IDX-1. In summary, our study provides evidence that GLP-1 is a differentiation factor for pancreatic Ductal Cells and that its effect requires the expression of IDX-1.

Piero Marchetti - One of the best experts on this subject based on the ideXlab platform.

  • sars cov 2 receptor angiotensin i converting enzyme type 2 ace2 is expressed in human pancreatic β Cells and in the human pancreas microvasculature
    Frontiers in Endocrinology, 2020
    Co-Authors: Daniela Fignani, Giada Licata, Noemi Brusco, Laura Nigi, Giuseppina Emanuela Grieco, Lorella Marselli, Lut Overbergh, Conny Gysemans, Maikel Luis Colli, Piero Marchetti
    Abstract:

    Increasing evidence demonstrated that the expression of Angiotensin I-Converting Enzyme type 2 (ACE2) is a necessary step for SARS-CoV-2 infection permissiveness. In light of the recent data highlighting an association between COVID-19 and diabetes, a detailed analysis aimed at evaluating ACE2 expression pattern distribution in human pancreas is still lacking. Here, we took advantage of INNODIA network EUnPOD biobank collection to thoroughly analyze ACE2, both at mRNA and protein level, in multiple human pancreatic tissues and using several methodologies. Using multiple reagents and antibodies, we showed that ACE2 is expressed in human pancreatic islets, where it is preferentially expressed in subsets of insulin producing β-Cells. ACE2 is also highly expressed in pancreas microvasculature pericytes and moderately expressed in rare scattered Ductal Cells. By using different ACE2 antibodies we showed that a recently described short-ACE2 isoform is also prevalently expressed in human β-Cells. Finally, using RT-qPCR, RNA-seq and High-Content imaging screening analysis, we demonstrated that pro-inflammatory cytokines, but not palmitate, increase ACE2 expression in the β-cell line EndoC-βH1 and in primary human pancreatic islets. Taken together, our data indicate a potential link between SARS-CoV-2 and diabetes through putative infection of pancreatic microvasculature and/or Ductal Cells and/or through direct β-cell virus tropism.

  • sars cov 2 receptor angiotensin i converting enzyme type 2 ace2 is expressed in human pancreatic β Cells and in the human pancreas microvasculature
    bioRxiv, 2020
    Co-Authors: Daniela Fignani, Giada Licata, Noemi Brusco, Laura Nigi, Giuseppina Emanuela Grieco, Lorella Marselli, Lut Overbergh, Conny Gysemans, Maikel Luis Colli, Piero Marchetti
    Abstract:

    Abstract Increasing evidence demonstrated that the expression of Angiotensin I-Converting Enzyme type 2 (ACE2), is a necessary step for SARS-CoV-2 infection permissiveness. In the light of the recent data highlighting an association between COVID-19 and diabetes, a detailed analysis aimed at evaluating ACE2 expression pattern distribution in human pancreas is still lacking. Here, we took advantage of INNODIA network EUnPOD biobank collection to thoroughly analyse ACE2, both at mRNA and protein level, in multiple human pancreatic tissues and using several methodologies. Using multiple reagents and antibodies, we showed that ACE2 is expressed in human pancreatic islets, where it is preferentially expressed in subsets of insulin producing β-Cells. ACE2 is also is highly expressed in pancreas microvasculature pericytes and moderately expressed in rare scattered Ductal Cells. By using different ACE2 antibodies we showed that a recently described short-ACE2 isoform is also prevalently expressed in human β-Cells. Finally, using RT-qPCR, RNA-seq and High-Content imaging screening analysis, we demonstrated that pro-inflammatory cytokines, but not palmitate, increases ACE2 expression in the β-cell line EndoC-βH1 and in primary human pancreatic islets. Taken together, our data indicate a potential link between SARS-CoV-2 and diabetes through putative infection of pancreatic microvasculature and/or Ductal Cells and/or through direct β-cell virus tropism.