Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 126
Filter
Add more filters

Publication year range
1.
Cell ; 163(6): 1457-67, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26627735

ABSTRACT

A variety of signals finely tune insulin secretion by pancreatic ß cells to prevent both hyper-and hypoglycemic states. Here, we show that post-translational regulation of the transcription factors ETV1, ETV4, and ETV5 by the ubiquitin ligase COP1 (also called RFWD2) in ß cells is critical for insulin secretion. Mice lacking COP1 in ß cells developed diabetes due to insulin granule docking defects that were fully rescued by genetic deletion of Etv1, Etv4, and Etv5. Genes regulated by ETV1, ETV4, or ETV5 in the absence of mouse COP1 were enriched in human diabetes-associated genes, suggesting that they also influence human ß-cell pathophysiology. In normal ß cells, ETV4 was stabilized upon membrane depolarization and limited insulin secretion under hyperglycemic conditions. Collectively, our data reveal that ETVs negatively regulate insulin secretion for the maintenance of normoglycemia.


Subject(s)
Insulin-Secreting Cells/metabolism , Insulin/metabolism , Nuclear Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , DNA-Binding Proteins/metabolism , Diabetes Mellitus/metabolism , Exocytosis , Gene Deletion , Glucose/metabolism , Humans , Hyperglycemia/metabolism , Insulin Secretion , Mice , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-ets/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/genetics
2.
Cell ; 150(6): 1103-4, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22980973

ABSTRACT

ß cell dysfunction with subsequent apoptosis is considered a significant contributor to the development of type 2 diabetes. Emerging data from Talchai et al. suggest ß cell dedifferentiation as an alternative mechanism of insulin insufficiency that might be more amenable to intervention in at least a subset of patients.

3.
Genes Dev ; 31(3): 228-240, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28270515

ABSTRACT

Following differentiation during fetal development, ß cells further adapt to their postnatal role through functional maturation. While adult islets are thought to contain functionally mature ß cells, recent analyses of transgenic rodent and human pancreata reveal a number of novel heterogeneity markers in mammalian ß cells. The marked heterogeneity long after maturation raises the prospect that diverse populations harbor distinct roles aside from glucose-stimulated insulin secretion. In this review, we outline our current understanding of the ß-cell maturation process, emphasize recent literature on novel heterogeneity markers, and offer perspectives on reconciling the findings from these two areas.


Subject(s)
Cell Differentiation , Glucose/metabolism , Insulin-Secreting Cells/cytology , Insulin/metabolism , Islets of Langerhans/cytology , Animals , Biomarkers , Humans , Insulin-Secreting Cells/metabolism
4.
Genes Dev ; 30(24): 2669-2683, 2016 12 15.
Article in English | MEDLINE | ID: mdl-28087712

ABSTRACT

Aberrant activation of embryonic signaling pathways is frequent in pancreatic ductal adenocarcinoma (PDA), making developmental regulators therapeutically attractive. Here we demonstrate diverse functions for pancreatic and duodenal homeobox 1 (PDX1), a transcription factor indispensable for pancreas development, in the progression from normal exocrine cells to metastatic PDA. We identify a critical role for PDX1 in maintaining acinar cell identity, thus resisting the formation of pancreatic intraepithelial neoplasia (PanIN)-derived PDA. Upon neoplastic transformation, the role of PDX1 changes from tumor-suppressive to oncogenic. Interestingly, subsets of malignant cells lose PDX1 expression while undergoing epithelial-to-mesenchymal transition (EMT), and PDX1 loss is associated with poor outcome. This stage-specific functionality arises from profound shifts in PDX1 chromatin occupancy from acinar cells to PDA. In summary, we report distinct roles of PDX1 at different stages of PDA, suggesting that therapeutic approaches against this potential target need to account for its changing functions at different stages of carcinogenesis. These findings provide insight into the complexity of PDA pathogenesis and advocate a rigorous investigation of therapeutically tractable targets at distinct phases of PDA development and progression.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Pancreatic Neoplasms/genetics , Trans-Activators/metabolism , Acinar Cells/pathology , Animals , Carcinoma, Pancreatic Ductal/physiopathology , Gene Deletion , Homeodomain Proteins/genetics , Humans , Mice , Pancreatic Neoplasms/physiopathology , Tissue Array Analysis , Trans-Activators/genetics , Tumor Cells, Cultured
5.
Genes Dev ; 29(6): 658-71, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25792600

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) develops predominantly through pancreatic intraepithelial neoplasia (PanIN) and intraductal papillary mucinous neoplasm (IPMN) precursor lesions. Pancreatic acinar cells are reprogrammed to a "ductal-like" state during PanIN-PDA formation. Here, we demonstrate a parallel mechanism operative in mature duct cells during which functional cells undergo "ductal retrogression" to form IPMN-PDA. We further identify critical antagonistic roles for Brahma-related gene 1 (Brg1), a catalytic subunit of the SWI/SNF complexes, during IPMN-PDA development. In mature duct cells, Brg1 inhibits the dedifferentiation that precedes neoplastic transformation, thus attenuating tumor initiation. In contrast, Brg1 promotes tumorigenesis in full-blown PDA by supporting a mesenchymal-like transcriptional landscape. We further show that JQ1, a drug that is currently being tested in clinical trials for hematological malignancies, impairs PDA tumorigenesis by both mimicking some and inhibiting other Brg1-mediated functions. In summary, our study demonstrates the context-dependent roles of Brg1 and points to potential therapeutic treatment options based on epigenetic regulation in PDA.


Subject(s)
Carcinoma, Pancreatic Ductal/physiopathology , Cell Transformation, Neoplastic/genetics , DNA Helicases/metabolism , Nuclear Proteins/metabolism , Pancreatic Neoplasms/physiopathology , Transcription Factors/metabolism , Animals , Azepines/pharmacology , Azepines/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Cell Transformation, Neoplastic/drug effects , DNA Helicases/genetics , Gene Expression Regulation, Neoplastic , Humans , Mice , Nuclear Proteins/genetics , Pancreatic Neoplasms/drug therapy , Proto-Oncogene Proteins p21(ras)/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Transcription Factors/genetics , Triazoles/pharmacology , Triazoles/therapeutic use , Tumor Cells, Cultured
6.
Biotechnol Bioeng ; 118(2): 979-991, 2021 02.
Article in English | MEDLINE | ID: mdl-33205831

ABSTRACT

Scalable processes are requisite for the robust biomanufacturing of human pluripotent stem cell (hPSC)-derived therapeutics. Toward this end, we demonstrate the xeno-free expansion and directed differentiation of human embryonic and induced pluripotent stem cells to definitive endoderm (DE) in a controlled stirred suspension bioreactor (SSB). Based on previous work on converting hPSCs to insulin-producing progeny, differentiation of two hPSC lines was optimized in planar cultures yielding up to 87% FOXA2+ /SOX17+ cells. Next, hPSCs were propagated in an SSB with controlled pH and dissolved oxygen. Cultures displayed a 10- to 12-fold increase in cell number over 5-6 days with the maintenance of pluripotency (>85% OCT4+ ) and viability (>85%). For differentiation, SSB cultures yielded up to 89% FOXA2+ /SOX17+ cells or ~ 8 DE cells per seeded hPSC. Specification to DE cell fate was consistently more efficient in the bioreactor compared to planar cultures. Hence, a tunable strategy is established that is suitable for the xeno-free manufacturing of DE cells from different hPSC lines in scalable SSBs. This study advances bioprocess development for producing a wide gamut of human DE cell-derived therapeutics.


Subject(s)
Bioreactors , Endoderm/metabolism , Human Embryonic Stem Cells/metabolism , Cell Line , Endoderm/cytology , Human Embryonic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology
7.
Genes Dev ; 27(23): 2563-75, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24298056

ABSTRACT

Precise functioning of the pancreatic ß cell is paramount to whole-body glucose homeostasis, and ß-cell dysfunction contributes significantly to diabetes mellitus. Using transgenic mouse models, we demonstrate that deletion of the von Hippel-Lindau (Vhlh) gene (encoding an E3 ubiquitin ligase implicated in, among other functions, oxygen sensing in pancreatic ß cells) is deleterious to canonical ß-cell gene expression. This triggers erroneous expression of factors normally active in progenitor cells, including effectors of the Notch, Wnt, and Hedgehog signaling cascades. Significantly, an up-regulation of the transcription factor Sox9, normally excluded from functional ß cells, occurs upon deletion of Vhlh. Sox9 plays important roles during pancreas development but does not have a described role in the adult ß cell. ß-Cell-specific ectopic expression of Sox9 results in diabetes mellitus from similar perturbations in ß-cell identity. These findings reveal that assaults on the ß cell that impact the differentiation state of the cell have clear implications toward our understanding of diabetes mellitus.


Subject(s)
Diabetes Mellitus/genetics , Diabetes Mellitus/physiopathology , Gene Expression Regulation/genetics , Insulin-Secreting Cells/cytology , SOX9 Transcription Factor/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Animals , Cell Hypoxia/physiology , Cell Line , Gene Deletion , Humans , Insulin-Secreting Cells/metabolism , Male , Mice , Von Hippel-Lindau Tumor Suppressor Protein/genetics
8.
Dev Biol ; 449(1): 14-20, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30771302

ABSTRACT

The embryonic origin of pericytes is heterogeneous, both between and within organs. While pericytes of coelomic organs were proposed to differentiate from the mesothelium, a single-layer squamous epithelium, the embryonic origin of pancreatic pericytes has yet to be reported. Here, we show that adult pancreatic pericytes originate from the embryonic pancreatic mesenchyme. Our analysis indicates that pericytes of the adult mouse pancreas originate from cells expressing the transcription factor Nkx3.2. In the embryonic pancreas, Nkx3.2-expressing cells constitute the multilayered mesenchyme, which surrounds the pancreatic epithelium and supports multiple events in its development. Thus, we traced the fate of the pancreatic mesenchyme. Our analysis reveals that pancreatic mesenchymal cells acquire various pericyte characteristics, including gene expression, typical morphology, and periendothelial location, during embryogenesis. Importantly, we show that the vast majority of pancreatic mesenchymal cells differentiate into pericytes already at embryonic day 13.5 and progressively acquires a more mature pericyte phenotype during later stages of pancreas organogenesis. Thus, our study indicates the embryonic pancreatic mesenchyme as the primary origin to adult pancreatic pericytes. As pericytes of other coelomic organs were suggested to differentiate from the mesothelium, our findings point to a distinct origin of these cells in the pancreas. Thus, our study proposes a complex ontogeny of pericytes of coelomic organs.


Subject(s)
Mesoderm/cytology , Mesoderm/embryology , Pancreas/cytology , Pancreas/embryology , Pericytes/cytology , Animals , Biomarkers/metabolism , Embryonic Development/genetics , Endothelial Cells/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Mice , Receptor, Platelet-Derived Growth Factor beta/metabolism , Transcription Factors/metabolism
9.
EMBO J ; 34(13): 1759-72, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-25908839

ABSTRACT

Directed differentiation of human pluripotent stem cells into functional insulin-producing beta-like cells holds great promise for cell replacement therapy for patients suffering from diabetes. This approach also offers the unique opportunity to study otherwise inaccessible aspects of human beta cell development and function in vitro. Here, we show that current pancreatic progenitor differentiation protocols promote precocious endocrine commitment, ultimately resulting in the generation of non-functional polyhormonal cells. Omission of commonly used BMP inhibitors during pancreatic specification prevents precocious endocrine formation while treatment with retinoic acid followed by combined EGF/KGF efficiently generates both PDX1(+) and subsequent PDX1(+)/NKX6.1(+) pancreatic progenitor populations, respectively. Precise temporal activation of endocrine differentiation in PDX1(+)/NKX6.1(+) progenitors produces glucose-responsive beta-like cells in vitro that exhibit key features of bona fide human beta cells, remain functional after short-term transplantation, and reduce blood glucose levels in diabetic mice. Thus, our simplified and scalable system accurately recapitulates key steps of human pancreas development and provides a fast and reproducible supply of functional human beta-like cells.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation , Embryonic Stem Cells/physiology , Insulin-Secreting Cells/physiology , Pancreas/cytology , Animals , Blood Glucose/metabolism , Cells, Cultured , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/therapy , Embryonic Stem Cells/cytology , Glucose/pharmacology , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/transplantation , Mice , Mice, SCID , Mice, Transgenic , Streptozocin
10.
EMBO J ; 34(4): 517-30, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25586376

ABSTRACT

In adaptation to oncogenic signals, pancreatic ductal adenocarcinoma (PDAC) cells undergo epithelial-mesenchymal transition (EMT), a process combining tumor cell dedifferentiation with acquisition of stemness features. However, the mechanisms linking oncogene-induced signaling pathways with EMT and stemness remain largely elusive. Here, we uncover the inflammation-induced transcription factor NFATc1 as a central regulator of pancreatic cancer cell plasticity. In particular, we show that NFATc1 drives EMT reprogramming and maintains pancreatic cancer cells in a stem cell-like state through Sox2-dependent transcription of EMT and stemness factors. Intriguingly, NFATc1-Sox2 complex-mediated PDAC dedifferentiation and progression is opposed by antithetical p53-miR200c signaling, and inactivation of the tumor suppressor pathway is essential for tumor dedifferentiation and dissemination both in genetically engineered mouse models (GEMM) and human PDAC. Based on these findings, we propose the existence of a hierarchical signaling network regulating PDAC cell plasticity and suggest that the molecular decision between epithelial cell preservation and conversion into a dedifferentiated cancer stem cell-like phenotype depends on opposing levels of p53 and NFATc1 signaling activities.


Subject(s)
MicroRNAs/metabolism , NFATC Transcription Factors/metabolism , Pancreatic Neoplasms/metabolism , SOXB1 Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/physiology , Humans , Mice , MicroRNAs/genetics , NFATC Transcription Factors/genetics , SOXB1 Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics
11.
EMBO J ; 33(19): 2135-6, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25154606

ABSTRACT

The in vivo assessment of epigenetic changes during mouse pancreatic beta­cell differentiation reveals surprising differences to directed, in vitro differentiation of human embryonic stem cells. New findings reported in this issue of The EMBO Journal further identify Ezh2 as a critical determinant of endocrine progenitor number and could instruct improved protocols for stem cell-based therapies.


Subject(s)
Endocrine Cells/cytology , Histones/metabolism , Islets of Langerhans/cytology , Jumonji Domain-Containing Histone Demethylases/genetics , Polycomb Repressive Complex 2/physiology , Animals , Enhancer of Zeste Homolog 2 Protein , Humans
12.
Development ; 142(23): 4010-25, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26483210

ABSTRACT

Mechanisms of initial cell fate decisions differ among species. To gain insights into lineage allocation in humans, we derived ten human embryonic stem cell lines (designated UCSFB1-10) from single blastomeres of four 8-cell embryos and one 12-cell embryo from a single couple. Compared with numerous conventional lines from blastocysts, they had unique gene expression and DNA methylation patterns that were, in part, indicative of trophoblast competence. At a transcriptional level, UCSFB lines from different embryos were often more closely related than those from the same embryo. As predicted by the transcriptomic data, immunolocalization of EOMES, T brachyury, GDF15 and active ß-catenin revealed differential expression among blastomeres of 8- to 10-cell human embryos. The UCSFB lines formed derivatives of the three germ layers and CDX2-positive progeny, from which we derived the first human trophoblast stem cell line. Our data suggest heterogeneity among early-stage blastomeres and that the UCSFB lines have unique properties, indicative of a more immature state than conventional lines.


Subject(s)
Blastomeres/cytology , Embryo Culture Techniques , Embryonic Stem Cells/cytology , Trophoblasts/cytology , Blastocyst/cytology , Cell Differentiation , Cell Line , Cell Lineage , DNA Methylation , Endoderm/metabolism , Fibroblasts/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Growth Differentiation Factor 15/metabolism , Humans , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Neural Stem Cells/cytology , Oligonucleotide Array Sequence Analysis , Transcription, Genetic , Transcriptome , beta Catenin/metabolism
13.
Diabetologia ; 60(1): 35-38, 2017 01.
Article in English | MEDLINE | ID: mdl-27785529

ABSTRACT

The asymptomatic phase of type 1 diabetes is recognised by the presence of beta cell autoantibodies in the absence of hyperglycaemia. We propose that an accurate description of this stage is provided by the name 'Autoimmune Beta Cell Disorder' (ABCD). Specifically, we suggest that this nomenclature and diagnosis will, in a proactive manner, shift the paradigm towards type 1 diabetes being first and foremost an immune-mediated disease and only later a metabolic disease, presaging more active therapeutic intervention in the asymptomatic stage of disease, before end-stage beta cell failure. Furthermore, we argue that accepting ABCD as a diagnosis will be critical in order to accelerate pharmaceutical, academic and public activities leading to clinical trials that could reverse beta cell autoimmunity and halt progression to symptomatic insulin-requiring type 1 diabetes. We recognize that there are both opportunities and challenges in the implementation of the ABCD concept but hope that the notion of 'asymptomatic autoimmune disease' as a disorder will be widely discussed and eventually accepted.


Subject(s)
Autoimmunity/physiology , Diabetes Mellitus, Type 1/complications , Insulin-Secreting Cells/pathology , Diabetes Mellitus, Type 1/immunology , Humans , Insulin-Secreting Cells/immunology
14.
Pancreatology ; 17(3): 350-353, 2017.
Article in English | MEDLINE | ID: mdl-28473229

ABSTRACT

OBJECTIVES: Pancreatic ductal adenocarcinoma (PDAC) is thought to derive from different precursor lesions including the recently identified atypical flat lesions (AFL). While all precursor lesions and PDAC share ductal characteristics, there is an ongoing debate about the cellular origin of the different PDAC precursor lesions. In particular, pancreatic acinar cells have previously been shown to display a remarkable plasticity being able to undergo ductal dedifferentiation in the context of oncogenic stimuli. METHODS: Histological analyses were performed in a murine PDAC model that specifically expresses oncogenic Kras in adult pancreatic acinar cells. Occurrence, characterization, and lineage tracing of AFLs were investigated. RESULTS: Upon expression of oncogenic Kras in adult pancreatic acinar cells, AFLs with typical morphology and expression profile arise. Lineage tracing confirmed that the AFLs were of acinar origin. CONCLUSIONS: Using a murine PDAC model, this study identifies pancreatic acinar cells as a cellular source for AFLs.


Subject(s)
Acinar Cells/pathology , Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/pathology , Animals , Carcinoma, Pancreatic Ductal/chemically induced , Carcinoma, Pancreatic Ductal/metabolism , Cell Differentiation , Cell Transformation, Neoplastic/pathology , Estrogen Antagonists , Immunohistochemistry , Mice , Pancreatic Neoplasms/chemically induced , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/biosynthesis , Proto-Oncogene Proteins p21(ras)/genetics , Tamoxifen
15.
Genes Dev ; 23(1): 24-36, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19136624

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is characterized by the deregulation of the hedgehog signaling pathway. The Sonic Hedgehog ligand (Shh), absent in the normal pancreas, is highly expressed in pancreatic tumors and is sufficient to induce neoplastic precursor lesions in mouse models. We investigated the mechanism of Shh signaling in PDAC carcinogenesis by genetically ablating the canonical bottleneck of hedgehog signaling, the transmembrane protein Smoothened (Smo), in the pancreatic epithelium of PDAC-susceptible mice. We report that multistage development of PDAC tumors is not affected by the deletion of Smo in the pancreas, demonstrating that autocrine Shh-Ptch-Smo signaling is not required in pancreatic ductal cells for PDAC progression. However, the expression of Gli target genes is maintained in Smo-negative ducts, implicating alternative means of regulating Gli transcription in the neoplastic ductal epithelium. In PDAC tumor cells, we find that Gli transcription is decoupled from upstream Shh-Ptch-Smo signaling and is regulated by TGF-beta and KRAS, and we show that Gli1 is required both for survival and for the KRAS-mediated transformed phenotype of cultured PDAC cancer cells.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic , Kruppel-Like Transcription Factors/metabolism , Pancreatic Ducts/metabolism , Pancreatic Neoplasms/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line , Cell Survival , Cells, Cultured , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Kruppel-Like Transcription Factors/genetics , Mice , Pancreatic Ducts/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Smoothened Receptor , Transforming Growth Factor beta/metabolism , Zinc Finger Protein GLI1
16.
Gastroenterology ; 148(5): 1024-1034.e9, 2015 May.
Article in English | MEDLINE | ID: mdl-25623042

ABSTRACT

BACKGROUND & AIMS: Oncogenic mutations in KRAS contribute to the development of pancreatic ductal adenocarcinoma, but are not sufficient to initiate carcinogenesis. Secondary events, such as inflammation-induced signaling via the epidermal growth factor receptor (EGFR) and expression of the SOX9 gene, are required for tumor formation. Herein we sought to identify the mechanisms that link EGFR signaling with activation of SOX9 during acinar-ductal metaplasia, a transdifferentiation process that precedes pancreatic carcinogenesis. METHODS: We analyzed pancreatic tissues from Kras(G12D);pdx1-Cre and Kras(G12D);NFATc1(Δ/Δ);pdx1-Cre mice after intraperitoneal administration of caerulein, vs cyclosporin A or dimethyl sulfoxide (controls). Induction of EGFR signaling and its effects on the expression of Nuclear factor of activated T cells c1 (NFATc1) or SOX9 were investigated by quantitative reverse-transcription polymerase chain reaction, immunoblot, and immunohistochemical analyses of mouse and human tissues and acinar cell explants. Interactions between NFATc1 and partner proteins and effects on DNA binding or chromatin modifications were studied using co-immunoprecipitation and chromatin immunoprecipitation assays in acinar cell explants and mouse tissue. RESULTS: EGFR activation induced expression of NFATc1 in metaplastic areas from patients with chronic pancreatitis and in pancreatic tissue from Kras(G12D) mice. EGFR signaling also promoted formation of a complex between NFATc1 and C-JUN in dedifferentiating mouse acinar cells, leading to activation of Sox9 transcription and induction of acinar-ductal metaplasia. Pharmacologic inhibition of NFATc1 or disruption of the Nfatc1 gene inhibited EGFR-mediated induction of Sox9 transcription and blocked acinar-ductal transdifferentiation and pancreatic cancer initiation in mice. CONCLUSIONS: EGFR signaling induces expression of NFATc1 and Sox9, leading to acinar cell transdifferentiation and initiation of pancreatic cancer. Strategies designed to disrupt this pathway might be developed to prevent pancreatic cancer initiation in high-risk patients with chronic pancreatitis.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Cell Transdifferentiation , ErbB Receptors/metabolism , NFATC Transcription Factors/metabolism , Pancreas, Exocrine/metabolism , Pancreatic Ducts/metabolism , Pancreatic Neoplasms/metabolism , Pancreatitis/metabolism , Precancerous Conditions/metabolism , SOX9 Transcription Factor/metabolism , Signal Transduction , Animals , Carcinoma, Pancreatic Ductal/chemically induced , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Ceruletide , Cyclosporine , Disease Models, Animal , ErbB Receptors/genetics , Gene Expression Regulation , Humans , Male , Metaplasia , Mice, Inbred C57BL , Mice, Knockout , Mutation , NFATC Transcription Factors/deficiency , NFATC Transcription Factors/genetics , Pancreas, Exocrine/pathology , Pancreatic Ducts/pathology , Pancreatic Neoplasms/chemically induced , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatitis/chemically induced , Pancreatitis/genetics , Pancreatitis/pathology , Precancerous Conditions/chemically induced , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Proto-Oncogene Proteins p21(ras)/genetics , SOX9 Transcription Factor/genetics , Tissue Culture Techniques , Transcriptional Activation
17.
J Autoimmun ; 71: 51-8, 2016 07.
Article in English | MEDLINE | ID: mdl-27017348

ABSTRACT

Type 1 diabetes (T1D) is an autoimmune disorder characterized by the destruction of insulin-producing pancreatic ß cells. Immune modulators have achieved some success in modifying the course of disease progression in T1D. However, there are parallel declines in C-peptide levels in treated and control groups after initial responses. In this review, we discuss mechanisms of ß cell death in T1D that involve necrosis and apoptosis. New technologies are being developed to enable visualization of insulitis and ß cell mass involving positron emission transmission that identifies ß cell ligands and magnetic resonance imaging that can identify vascular leakage. Molecular signatures that identify ß cell derived insulin DNA that is released from dying cells have been described and applied to clinical settings. We also consider changes in ß cells that occur during disease progression including the induction of DNA methyltransferases that may affect the function and differentiation of ß cells. Our findings from newer data suggest that the model of chronic long standing ß cell killing should be reconsidered. These studies indicate that the pathophysiology is accelerated in the peridiagnosis period and manifest by increased rates of ß cell killing and insulin secretory impairments over a shorter period than previously thought. Finally, we consider cellular explanations to account for the ongoing loss of insulin production despite continued immune therapy that may identify potential targets for treatment. The progressive decline in ß cell function raises the question as to whether ß cell failure that is independent of immune attack may be involved.


Subject(s)
Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/metabolism , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/metabolism , Animals , Autoimmunity , Biomarkers , Cell Death/genetics , Cell Death/immunology , Cell Survival/genetics , Cell Survival/immunology , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/therapy , Genetic Predisposition to Disease , Humans , Immunotherapy/adverse effects , Immunotherapy/methods , Insulin-Secreting Cells/pathology , Necrosis , Stress, Physiological
18.
Gut ; 63(4): 656-64, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23645620

ABSTRACT

OBJECTIVES: Emerging evidence from mouse models suggests that mutant Kras can drive the development of pancreatic ductal adenocarcinoma (PDA) precursors from acinar cells by enforcing ductal de-differentiation at the expense of acinar identity. Recently, human genome-wide association studies have identified NR5A2, a key regulator of acinar function, as a susceptibility locus for human PDA. We investigated the role of Nr5a2 in exocrine maintenance, regeneration and Kras driven neoplasia. DESIGN: To investigate the function of Nr5a2 in the pancreas, we generated mice with conditional pancreatic Nr5a2 deletion (PdxCre(late); Nr5a2(c/c)). Using this model, we evaluated acinar differentiation, regeneration after caerulein pancreatitis and Kras driven pancreatic neoplasia in the setting of Nr5a2 deletion. RESULTS: We show that Nr5a2 is not required for the development of the pancreatic acinar lineage but is important for maintenance of acinar identity. Nr5a2 deletion leads to destabilisation of the mature acinar differentiation state, acinar to ductal metaplasia and loss of regenerative capacity following acute caerulein pancreatitis. Loss of Nr5a2 also dramatically accelerates the development of oncogenic Kras driven acinar to ductal metaplasia and PDA precursor lesions. CONCLUSIONS: Nr5a2 is a key regulator of acinar plasticity. It is required for maintenance of acinar identity and re-establishing acinar fate during regeneration. Nr5a2 also constrains pancreatic neoplasia driven by oncogenic Kras, providing functional evidence supporting a potential role as a susceptibility gene for human PDA.


Subject(s)
Carcinoma, Acinar Cell/physiopathology , Carcinoma, Pancreatic Ductal/physiopathology , Pancreatic Neoplasms/physiopathology , Proto-Oncogene Proteins p21(ras)/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Cell Differentiation/physiology , Cell Line , Cell Transformation, Neoplastic , Ceruletide/pharmacology , Mice , Pancreatitis/chemically induced , Pancreatitis/physiopathology , Real-Time Polymerase Chain Reaction
19.
Hum Mol Genet ; 21(9): 2054-67, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22286172

ABSTRACT

Pluripotent stem cells are derived from culture of early embryos or the germline and can be induced by reprogramming of somatic cells. Barriers to reprogramming that stabilize the differentiated state and have tumor suppression functions are expected to exist. However, we have a limited understanding of what such barriers might be. To find novel barriers to reprogramming to pluripotency, we compared the transcriptional profiles of the mouse germline with pluripotent and somatic cells, in vivo and in vitro. There is a remarkable global expression of the transcriptional program for pluripotency in primordial germ cells (PGCs). We identify parallels between PGC reprogramming to pluripotency and human germ cell tumorigenesis, including the loss of LATS2, a tumor suppressor kinase of the Hippo pathway. We show that knockdown of LATS2 increases the efficiency of induction of pluripotency in human cells. LATS2 RNAi, unlike p53 RNAi, specifically enhances the generation of fully reprogrammed iPS cells without accelerating cell proliferation. We further show that LATS2 represses reprogramming in human cells by post-transcriptionally antagonizing TAZ but not YAP, two downstream effectors of the Hippo pathway. These results reveal transcriptional parallels between germ cell transformation and the generation of iPS cells and indicate that the Hippo pathway constitutes a barrier to cellular reprogramming.


Subject(s)
Cellular Reprogramming/genetics , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Acyltransferases , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression Profiling , Gene Knockdown Techniques , Genes, p53 , Germ Cells/cytology , Germ Cells/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mice , Mice, Transgenic , Models, Biological , Neoplasms, Germ Cell and Embryonal/etiology , Neoplasms, Germ Cell and Embryonal/genetics , Neoplasms, Germ Cell and Embryonal/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , RNA Interference , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors , YAP-Signaling Proteins
20.
Gastroenterology ; 144(6): 1170-9, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23622126

ABSTRACT

The endocrine and exocrine cells in the adult pancreas are not static, but can change their differentiation state in response to injury or stress. This concept of cells in flux means that there may be ways to generate certain types of cells (such as insulin-producing ß-cells) and prevent formation of others (such as transformed neoplastic cells). We review different aspects of cell identity in the pancreas, discussing how cells achieve their identity during embryonic development and maturation, and how this identity remains plastic, even in the adult pancreas.


Subject(s)
Cell Differentiation , Cell Lineage , Pancreas/metabolism , Pancreatic Diseases/metabolism , Regeneration , Signal Transduction , Stem Cells/metabolism , Adult Stem Cells/metabolism , Adult Stem Cells/pathology , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Proliferation , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/pathology , Gene Expression Regulation, Developmental , Humans , Pancreas/embryology , Pancreas/growth & development , Pancreas/pathology , Pancreas/physiopathology , Pancreatic Diseases/genetics , Pancreatic Diseases/pathology , Pancreatic Diseases/physiopathology , Pancreatic Diseases/therapy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/physiopathology , Signal Transduction/genetics , Stem Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL