Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
Add more filters

Publication year range
1.
Stem Cells ; 41(6): 643-657, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37085274

ABSTRACT

During early embryogenesis, the transcription factor SOX17 contributes to hepato-pancreato-biliary system formation and vascular-hematopoietic emergence. To better understand Sox17 function in the developing endoderm and endothelium, we developed a dual-color temporal lineage-tracing strategy in mice combined with single-cell RNA sequencing to analyze 6934 cells from Sox17-expressing lineages at embryonic days 9.0-9.5. Our analyses showed 19 distinct cellular clusters combined from all 3 germ layers. Differential gene expression, trajectory and RNA-velocity analyses of endothelial cells revealed a heterogenous population of uncommitted and specialized endothelial subtypes, including 2 hemogenic populations that arise from different origins. Similarly, analyses of posterior foregut endoderm revealed subsets of hepatic, pancreatic, and biliary progenitors with overlapping developmental potency. Calculated gene-regulatory networks predict gene regulons that are dominated by cell type-specific transcription factors unique to each lineage. Vastly different Sox17 regulons found in endoderm versus endothelial cells support the differential interactions of SOX17 with other regulatory factors thereby enabling lineage-specific regulatory actions.


Subject(s)
Embryonic Development , Endothelial Cells , Gene Expression Regulation, Developmental , Gene Regulatory Networks , SOXF Transcription Factors , Animals , Mice , Cell Differentiation , Cell Lineage/genetics , Endoderm/metabolism , Endothelial Cells/metabolism , HMGB Proteins/genetics , HMGB Proteins/metabolism , Sequence Analysis, RNA , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Transcription Factors/metabolism , Embryonic Development/genetics
2.
Cell ; 139(1): 161-74, 2009 Oct 02.
Article in English | MEDLINE | ID: mdl-19804761

ABSTRACT

GABAergic interneurons are key elements in neural coding, but the mechanisms that assemble inhibitory circuits remain unclear. In the spinal cord, the transfer of sensory signals to motor neurons is filtered by GABAergic interneurons that act presynaptically to inhibit sensory transmitter release and postsynaptically to inhibit motor neuron excitability. We show here that the connectivity and synaptic differentiation of GABAergic interneurons that mediate presynaptic inhibition is directed by their sensory targets. In the absence of sensory terminals these GABAergic neurons shun other available targets, fail to undergo presynaptic differentiation, and withdraw axons from the ventral spinal cord. A sensory-specific source of brain derived neurotrophic factor induces synaptic expression of the GABA synthetic enzyme GAD65--a defining biochemical feature of this set of interneurons. The organization of a GABAergic circuit that mediates presynaptic inhibition in the mammalian CNS is therefore controlled by a stringent program of sensory recognition and signaling.


Subject(s)
Interneurons/physiology , Spinal Cord/physiology , gamma-Aminobutyric Acid/physiology , Animals , Brain-Derived Neurotrophic Factor/metabolism , Glutamate Decarboxylase , Mice , Motor Neurons/physiology , Presynaptic Terminals , Proprioception , Sensory Receptor Cells/physiology , Spinal Cord/cytology
3.
Genes Dev ; 30(16): 1852-65, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27585590

ABSTRACT

The current model for endocrine cell specification in the pancreas invokes high-level production of the transcription factor Neurogenin 3 (Neurog3) in Sox9(+) bipotent epithelial cells as the trigger for endocrine commitment, cell cycle exit, and rapid delamination toward proto-islet clusters. This model posits a transient Neurog3 expression state and short epithelial residence period. We show, however, that a Neurog3(TA.LO) cell population, defined as Neurog3 transcriptionally active and Sox9(+) and often containing nonimmunodetectable Neurog3 protein, has a relatively high mitotic index and prolonged epithelial residency. We propose that this endocrine-biased mitotic progenitor state is functionally separated from a pro-ductal pool and endows them with long-term capacity to make endocrine fate-directed progeny. A novel BAC transgenic Neurog3 reporter detected two types of mitotic behavior in Sox9(+) Neurog3(TA.LO) progenitors, associated with progenitor pool maintenance or derivation of endocrine-committed Neurog3(HI) cells, respectively. Moreover, limiting Neurog3 expression dramatically increased the proportional representation of Sox9(+) Neurog3(TA.LO) progenitors, with a doubling of its mitotic index relative to normal Neurog3 expression, suggesting that low Neurog3 expression is a defining feature of this cycling endocrine-biased state. We propose that Sox9(+) Neurog3(TA.LO) endocrine-biased progenitors feed production of Neurog3(HI) endocrine-committed cells during pancreas organogenesis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Endocrine Cells/cytology , Gene Expression Regulation, Developmental , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Stem Cells/cytology , Animals , Cell Differentiation , Cell Proliferation/genetics , Mice , Mitosis , Pancreas/cytology
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(20): 2203-16, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26494792

ABSTRACT

In the mammalian pancreas, endocrine cells undergo lineage allocation upon emergence from a bipotent duct/endocrine progenitor pool, which resides in the "trunk epithelium." Major questions remain regarding how niche environments are organized within this epithelium to coordinate endocrine differentiation with programs of epithelial growth, maturation, and morphogenesis. We used EdU pulse-chase and tissue-reconstruction approaches to analyze how endocrine progenitors and their differentiating progeny are assembled within the trunk as it undergoes remodeling from an irregular plexus of tubules to form the eventual mature, branched ductal arbor. The bulk of endocrine progenitors is maintained in an epithelial "plexus state," which is a transient intermediate during epithelial maturation within which endocrine cell differentiation is continually robust and surprisingly long-lived. Within the plexus, local feedback effects derived from the differentiating and delaminating endocrine cells nonautonomously regulate the flux of endocrine cell birth as well as proliferative growth of the bipotent cell population using Notch-dependent and Notch-independent influences, respectively. These feedback effects in turn maintain the plexus state to ensure prolonged allocation of endocrine cells late into gestation. These findings begin to define a niche-like environment guiding the genesis of the endocrine pancreas and advance current models for how differentiation is coordinated with the growth and morphogenesis of the developing pancreatic epithelium.


Subject(s)
Cell Differentiation/genetics , Endocrine Cells/cytology , Epithelial Cells/cytology , Feedback, Physiological , Pancreas/cytology , Pancreas/embryology , Stem Cells/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle/genetics , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Organogenesis/genetics , Receptors, Notch/genetics , Receptors, Notch/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism
6.
Development ; 145(18)2018 09 21.
Article in English | MEDLINE | ID: mdl-30126902

ABSTRACT

During mouse pancreas organogenesis, endocrine cells are born from progenitors residing in an epithelial plexus niche. After a period in a lineage-primed Neurog3LO state, progenitors become endocrine committed via upregulation of Neurog3 We find that the Neurog3LO to Neurog3HI transition is associated with distinct stages of an epithelial egression process: narrowing the apical surface of the cell, basalward cell movement and eventual cell-rear detachment from the apical lumen surface to allow clustering as nascent islets under the basement membrane. Apical narrowing, basalward movement and Neurog3 transcriptional upregulation still occur without Neurog3 protein, suggesting that morphogenetic cues deployed within the plexus initiate endocrine commitment upstream or independently of Neurog3. Neurog3 is required for cell-rear detachment and complete endocrine-cell birth. The ROCK-nmMyoII pathway coordinates epithelial-cell morphogenesis and the progression through Neurog3-expressing states. NmMyoII is necessary for apical narrowing, basalward cell displacement and Neurog3 upregulation, but all three are limited by ROCK activity. We propose that ROCK-nmMyoII activity, Neurog3 gene-dose and Notch signaling integrate endocrine fate allocation with epithelial plexus growth and morphogenesis, representing a feedback control circuit that coordinates morphogenesis with lineage diversification in the endocrine-birth niche.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Endocrine Cells/cytology , Gene Dosage/genetics , Nerve Tissue Proteins/genetics , Organogenesis/genetics , Pancreas/embryology , Receptors, Notch/genetics , rho-Associated Kinases/genetics , Animals , Cell Differentiation/genetics , Cell Movement , Mice , Mice, Transgenic , Pancreas/cytology , Stem Cells/cytology , Transcriptional Activation/genetics
7.
Development ; 144(2): 248-257, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27993987

ABSTRACT

The transcription factor Pdx1 is required for multiple aspects of pancreatic organogenesis. It remains unclear to what extent Pdx1 expression and function depend upon trans-activation through 5' conserved cis-regulatory regions and, in particular, whether the mammal-specific Area II (-2139 to -1958 bp) affects minor or major aspects of organogenesis. We show that Area II is a primary effector of endocrine-selective transcription in epithelial multipotent cells, nascent endocrine progenitors, and differentiating and mature ß cells in vivo Pdx1ΔAREAII/- mice exhibit a massive reduction in endocrine progenitor cells and progeny hormone-producing cells, indicating that Area II activity is fundamental to mounting an effective endocrine lineage-specification program within the multipotent cell population. Creating an Area II-deleted state within already specified Neurog3-expressing endocrine progenitor cells increased the proportion of glucagon+ α relative to insulin+ ß cells, associated with the transcriptional and epigenetic derepression of the α-cell-determining Arx gene in endocrine progenitors. There were also glucagon and insulin co-expressing cells, and ß cells that were incapable of maturation. Creating the Pdx1ΔAREAII state after cells entered an insulin-expressing stage led to immature and dysfunctional islet ß cells carrying abnormal chromatin marking in vital ß-cell-associated genes. Therefore, trans-regulatory integration through Area II mediates a surprisingly extensive range of progenitor and ß-cell-specific Pdx1 functions.


Subject(s)
Cell Differentiation/genetics , Enhancer Elements, Genetic , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/physiology , Islets of Langerhans/embryology , Trans-Activators/metabolism , Animals , Binding Sites/genetics , Embryo, Mammalian , Gene Expression Regulation, Developmental , Insulin-Secreting Cells/cytology , Islets of Langerhans/growth & development , Islets of Langerhans/metabolism , Mammals/embryology , Mammals/genetics , Mice , Mice, Transgenic , Organogenesis/genetics , Species Specificity
8.
Genes Dev ; 25(16): 1680-5, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21852533

ABSTRACT

Using single transcription factors to reprogram cells could produce important insights into the epigenetic mechanisms that direct normal differentiation, or counter inappropriate plasticity, or even provide new ways of manipulating normal ontogeny in vitro to control lineage diversification and differentiation. We enforced Pdx1 expression from the Neurogenin-3-expressing endocrine commitment point onward and found during the embryonic period a minor increased ß-cell allocation with accompanying reduced α-cell numbers. More surprisingly, almost all remaining Pdx1-containing glucagon/Arx-producing cells underwent a fairly rapid conversion at postnatal stages, through glucagon-insulin double positivity, to a state indistinguishable from normal ß cells, resulting in complete α-cell absence. This α-to-ß conversion was not caused by activating Pdx1 in the later glucagon-expressing state. Our findings reveal that Pdx1 can work single-handedly as a potent context-dependent autonomous reprogramming agent, and suggest a postnatal differentiation evaluation stage involved in normal endocrine maturation.


Subject(s)
Glucagon-Secreting Cells/metabolism , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/metabolism , Trans-Activators/metabolism , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Glucagon/genetics , Glucagon/metabolism , Glucagon-Secreting Cells/cytology , Homeodomain Proteins/genetics , Immunohistochemistry , Insulin/genetics , Insulin/metabolism , Insulin-Secreting Cells/cytology , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Pancreas/embryology , Pancreas/growth & development , Pancreas/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/genetics
9.
Development ; 142(21): 3637-48, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26534984

ABSTRACT

Homozygous Mnx1 mutation causes permanent neonatal diabetes in humans, but via unknown mechanisms. Our systematic and longitudinal analysis of Mnx1 function during murine pancreas organogenesis and into the adult uncovered novel stage-specific roles for Mnx1 in endocrine lineage allocation and ß-cell fate maintenance. Inactivation in the endocrine-progenitor stage shows that Mnx1 promotes ß-cell while suppressing δ-cell differentiation programs, and is crucial for postnatal ß-cell fate maintenance. Inactivating Mnx1 in embryonic ß-cells (Mnx1(Δbeta)) caused ß-to-δ-like cell transdifferentiation, which was delayed until postnatal stages. In the latter context, ß-cells escaping Mnx1 inactivation unexpectedly upregulated Mnx1 expression and underwent an age-independent persistent proliferation. Escaper ß-cells restored, but then eventually surpassed, the normal pancreatic ß-cell mass, leading to islet hyperplasia in aged mice. In vitro analysis of islets isolated from Mnx1(Δbeta) mice showed higher insulin secretory activity and greater insulin mRNA content than in wild-type islets. Mnx1(Δbeta) mice also showed a much faster return to euglycemia after ß-cell ablation, suggesting that the new ß-cells derived from the escaper population are functional. Our findings identify Mnx1 as an important factor in ß-cell differentiation and proliferation, with the potential for targeting to increase the number of endogenous ß-cells for diabetes therapy.


Subject(s)
Diabetes Mellitus/pathology , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/metabolism , Transcription Factors/metabolism , Animals , Cell Transdifferentiation , Cellular Senescence , Eye Proteins/metabolism , Homeodomain Proteins/genetics , Humans , Hyperplasia/metabolism , Insulin-Secreting Cells/cytology , Mice , PAX6 Transcription Factor , Paired Box Transcription Factors/metabolism , Repressor Proteins/metabolism , Somatostatin-Secreting Cells/metabolism , Transcription Factors/genetics
10.
Genesis ; 55(9)2017 09.
Article in English | MEDLINE | ID: mdl-28772022

ABSTRACT

During pancreas organogenesis, Neurog3HI endocrine-committing cells are generated from a population of Sox9+ mitotic progenitors with only a low level of Neurog3 transcriptional activity (Neurog3TA.LO ). Low-level Neurog3 protein, in Neurog3TA.LO cells, is required to maintain their mitotic endocrine-lineage-primed status. Herein, we describe a Neurog3-driven FUCCI cell-cycle reporter (Neurog3P2A.FUCCI ) derived from a Neurog3 BAC transgenic reporter that functions as a loxed cassette acceptor (LCA). In cycling Sox9+ Neurog3TA.LO progenitors, the majority of cells in S-G2 -M phases have undetectable levels of Neurog3 with increased expression of endocrine progenitor markers, while those in G1 have low Neurog3 levels with increased expression of endocrine differentiation markers. These findings support a model in which variations in Neurog3 protein levels are coordinated with cell-cycle phase progression in Neurog3TA.LO progenitors with entrance into G1 triggering a concerted effort, beyond increasing Neurog3 levels, to maintain an endocrine-lineage-primed state by initiating expression of the downstream endocrine differentiation program prior to endocrine-commitment.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Cycle , Embryonic Stem Cells/metabolism , Islets of Langerhans/metabolism , Nerve Tissue Proteins/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cells, Cultured , Embryonic Stem Cells/cytology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Islets of Langerhans/cytology , Mice , Nerve Tissue Proteins/metabolism
11.
Development ; 141(22): 4385-94, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25371369

ABSTRACT

The timing and gene regulatory logic of organ-fate commitment from within the posterior foregut of the mammalian endoderm is largely unexplored. Transient misexpression of a presumed pancreatic-commitment transcription factor, Ptf1a, in embryonic mouse endoderm (Ptf1a(EDD)) dramatically expanded the pancreatic gene regulatory network within the foregut. Ptf1a(EDD) temporarily suppressed Sox2 broadly over the anterior endoderm. Pancreas-proximal organ territories underwent full tissue conversion. Early-stage Ptf1a(EDD) rapidly expanded the endogenous endodermal Pdx1-positive domain and recruited other pancreas-fate-instructive genes, thereby spatially enlarging the potential for pancreatic multipotency. Early Ptf1a(EDD) converted essentially the entire glandular stomach, rostral duodenum and extrahepatic biliary system to pancreas, with formation of many endocrine cell clusters of the type found in normal islets of Langerhans. Sliding the Ptf1a(EDD) expression window through embryogenesis revealed differential temporal competencies for stomach-pancreas respecification. The response to later-stage Ptf1a(EDD) changed radically towards unipotent, acinar-restricted conversion. We provide strong evidence, beyond previous Ptf1a inactivation or misexpression experiments in frog embryos, for spatiotemporally context-dependent activity of Ptf1a as a potent gain-of-function trigger of pro-pancreatic commitment.


Subject(s)
Endoderm/embryology , Gastrointestinal Tract/embryology , Gene Expression Regulation, Developmental/physiology , Organogenesis/physiology , Pancreas/embryology , Transcription Factors/metabolism , Animals , Endoderm/metabolism , Fluorescent Antibody Technique , Gastrointestinal Tract/metabolism , Gene Expression Regulation, Developmental/genetics , Gene Regulatory Networks/genetics , Histological Techniques , Mice , Microscopy, Confocal , Organogenesis/genetics , SOXB1 Transcription Factors/metabolism
12.
Development ; 141(7): 1480-91, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24574008

ABSTRACT

Neurovascular alignment is a common anatomical feature of organs, but the mechanisms leading to this arrangement are incompletely understood. Here, we show that vascular endothelial growth factor (VEGF) signaling profoundly affects both vascularization and innervation of the pancreatic islet. In mature islets, nerves are closely associated with capillaries, but the islet vascularization process during embryonic organogenesis significantly precedes islet innervation. Although a simple neuronal meshwork interconnects the developing islet clusters as they begin to form at E14.5, the substantial ingrowth of nerve fibers into islets occurs postnatally, when islet vascularization is already complete. Using genetic mouse models, we demonstrate that VEGF regulates islet innervation indirectly through its effects on intra-islet endothelial cells. Our data indicate that formation of a VEGF-directed, intra-islet vascular plexus is required for development of islet innervation, and that VEGF-induced islet hypervascularization leads to increased nerve fiber ingrowth. Transcriptome analysis of hypervascularized islets revealed an increased expression of extracellular matrix components and axon guidance molecules, with these transcripts being enriched in the islet-derived endothelial cell population. We propose a mechanism for coordinated neurovascular development within pancreatic islets, in which endocrine cell-derived VEGF directs the patterning of intra-islet capillaries during embryogenesis, forming a scaffold for the postnatal ingrowth of essential autonomic nerve fibers.


Subject(s)
Blood Vessels/physiology , Cell Communication/genetics , Islets of Langerhans/blood supply , Islets of Langerhans/innervation , Neovascularization, Physiologic/physiology , Vascular Endothelial Growth Factor A/physiology , Animals , Blood Vessels/embryology , Cells, Cultured , Embryo, Mammalian , Endothelium, Vascular/embryology , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiology , Female , Islets of Langerhans/embryology , Mice , Mice, Transgenic , Vascular Endothelial Growth Factor A/genetics
13.
Development ; 140(4): 751-64, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23325761

ABSTRACT

Pancreatic multipotent progenitor cells (MPCs) produce acinar, endocrine and duct cells during organogenesis, but their existence and location in the mature organ remain contentious. We used inducible lineage-tracing from the MPC-instructive gene Ptf1a to define systematically in mice the switch of Ptf1a(+) MPCs to unipotent proacinar competence during the secondary transition, their rapid decline during organogenesis, and absence from the mature organ. Between E11.5 and E15.5, we describe tip epithelium heterogeneity, suggesting that putative Ptf1a(+)Sox9(+)Hnf1ß(+) MPCs are intermingled with Ptf1a(HI)Sox9(LO) proacinar progenitors. In the adult, pancreatic duct ligation (PDL) caused facultative reactivation of multipotency factors (Sox9 and Hnf1ß) in Ptf1a(+) acini, which undergo rapid reprogramming to duct cells and longer-term reprogramming to endocrine cells, including insulin(+) ß-cells that are mature by the criteria of producing Pdx1(HI), Nkx6.1(+) and MafA(+). These Ptf1a lineage-derived endocrine/ß-cells are likely formed via Ck19(+)/Hnf1ß(+)/Sox9(+) ductal and Ngn3(+) endocrine progenitor intermediates. Acinar to endocrine/ß-cell transdifferentiation was enhanced by combining PDL with pharmacological elimination of pre-existing ß-cells. Thus, we show that acinar cells, without exogenously introduced factors, can regain aspects of embryonic multipotentiality under injury, and convert into mature ß-cells.


Subject(s)
Cell Differentiation/physiology , Multipotent Stem Cells/physiology , Organogenesis/physiology , Pancreas/embryology , Recovery of Function/physiology , Signal Transduction/physiology , Transcription Factors/metabolism , Acinar Cells/cytology , Animals , Body Weights and Measures , Gene Knock-In Techniques , Mice , Microscopy, Confocal , Multipotent Stem Cells/metabolism , Pancreas/physiology , Tamoxifen , Time Factors
14.
Development ; 140(13): 2669-79, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23720049

ABSTRACT

Although the liver and ventral pancreas are thought to arise from a common multipotent progenitor pool, it is unclear whether these progenitors of the hepatopancreas system are specified by a common genetic mechanism. Efforts to determine the role of Hnf1b and Wnt signaling in this crucial process have been confounded by a combination of factors, including a narrow time frame for hepatopancreas specification, functional redundancy among Wnt ligands, and pleiotropic defects caused by either severe loss of Wnt signaling or Hnf1b function. Using a novel hypomorphic hnf1ba zebrafish mutant that exhibits pancreas hypoplasia, as observed in HNF1B monogenic diabetes, we show that hnf1ba plays essential roles in regulating ß-cell number and pancreas specification, distinct from its function in regulating pancreas size and liver specification, respectively. By combining Hnf1ba partial loss of function with conditional loss of Wnt signaling, we uncover a crucial developmental window when these pathways synergize to specify the entire ventrally derived hepatopancreas progenitor population. Furthermore, our in vivo genetic studies demonstrate that hnf1ba generates a permissive domain for Wnt signaling activity in the foregut endoderm. Collectively, our findings provide a new model for HNF1B function, yield insight into pancreas and ß-cell development, and suggest a new mechanism for hepatopancreatic specification.


Subject(s)
Hepatocyte Nuclear Factor 1-beta/metabolism , Hepatopancreas/cytology , Hepatopancreas/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Wnt Proteins/metabolism , Zebrafish Proteins/metabolism , Animals , Animals, Genetically Modified , Cell Differentiation/genetics , Cell Differentiation/physiology , Hepatocyte Nuclear Factor 1-beta/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Wnt Proteins/genetics , Zebrafish , Zebrafish Proteins/genetics
15.
Proc Natl Acad Sci U S A ; 110(31): 12691-6, 2013 Jul 30.
Article in English | MEDLINE | ID: mdl-23852729

ABSTRACT

Developmental biology is challenged to reveal the function of numerous candidate genes implicated by recent genome-scale studies as regulators of organ development and diseases. Recapitulating organogenesis from purified progenitor cells that can be genetically manipulated would provide powerful opportunities to dissect such gene functions. Here we describe systems for reconstructing pancreas development, including islet ß-cell and α-cell differentiation, from single fetal progenitor cells. A strict requirement for native genetic regulators of in vivo pancreas development, such as Ngn3, Arx, and Pax4, revealed the authenticity of differentiation programs in vitro. Efficient genetic screens permitted by this system revealed that Prdm16 is required for pancreatic islet development in vivo. Discovering the function of genes regulating pancreas development with our system should enrich strategies for regenerating islets for treating diabetes mellitus.


Subject(s)
Cell Differentiation , Glucagon-Secreting Cells/metabolism , Insulin-Secreting Cells/metabolism , Stem Cells/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/genetics , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Diabetes Mellitus/metabolism , Diabetes Mellitus/therapy , Female , Glucagon-Secreting Cells/cytology , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Insulin-Secreting Cells/cytology , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Paired Box Transcription Factors/biosynthesis , Paired Box Transcription Factors/genetics , Stem Cells/cytology , Transcription Factors/biosynthesis , Transcription Factors/genetics
16.
Dev Biol ; 393(1): 109-23, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24972089

ABSTRACT

Asymmetric development of the vertebrate embryo has fascinated embryologists for over a century. Much has been learned since the asymmetric Nodal signaling cascade in the left lateral plate mesoderm was detected, and began to be unraveled over the past decade or two. When and how symmetry is initially broken, however, has remained a matter of debate. Two essentially mutually exclusive models prevail. Cilia-driven leftward flow of extracellular fluids occurs in mammalian, fish and amphibian embryos. A great deal of experimental evidence indicates that this flow is indeed required for symmetry breaking. An alternative model has argued, however, that flow simply acts as an amplification step for early asymmetric cues generated by ion flux during the first cleavage divisions. In this review we critically evaluate the experimental basis of both models. Although a number of open questions persist, the available evidence is best compatible with flow-based symmetry breakage as the archetypical mode of symmetry breakage.


Subject(s)
Body Patterning , Vertebrates/embryology , Animals , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/embryology , Embryo, Nonmammalian/anatomy & histology , Embryo, Nonmammalian/embryology , Fishes/embryology , Gene Expression Regulation, Developmental , H(+)-K(+)-Exchanging ATPase/genetics , H(+)-K(+)-Exchanging ATPase/metabolism , Left-Right Determination Factors/metabolism , Mammals/embryology , Mesoderm/metabolism , Nodal Protein/metabolism , Organizers, Embryonic/physiology , Serotonin/metabolism , Signal Transduction/genetics , Vertebrates/anatomy & histology , Xenopus/embryology
17.
Gastroenterology ; 146(1): 233-44.e5, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23999170

ABSTRACT

BACKGROUND & AIMS: Metaplasias often have characteristics of developmentally related tissues. Pancreatic metaplastic ducts are usually associated with pancreatitis and pancreatic ductal adenocarcinoma. The tuft cell is a chemosensory cell that responds to signals in the extracellular environment via effector molecules. Commonly found in the biliary tract, tuft cells are absent from normal murine pancreas. Using the aberrant appearance of tuft cells as an indicator, we tested if pancreatic metaplasia represents transdifferentiation to a biliary phenotype and what effect this has on pancreatic tumorigenesis. METHODS: We analyzed pancreatic tissue and tumors that developed in mice that express an activated form of Kras (Kras(LSL-G12D/+);Ptf1a(Cre/+) mice). Normal bile duct, pancreatic duct, and tumor-associated metaplasias from the mice were analyzed for tuft cell and biliary progenitor markers, including SOX17, a transcription factor that regulates biliary development. We also analyzed pancreatic tissues from mice expressing transgenic SOX17 alone (ROSA(tTa/+);Ptf1(CreERTM/+);tetO-SOX17) or along with activated Kras (ROSAtT(a/+);Ptf1a(CreERTM/+);tetO-SOX17;Kras(LSL-G12D;+)). RESULTS: Tuft cells were frequently found in areas of pancreatic metaplasia, decreased throughout tumor progression, and absent from invasive tumors. Analysis of the pancreatobiliary ductal systems of mice revealed tuft cells in the biliary tract but not the normal pancreatic duct. Analysis for biliary markers revealed expression of SOX17 in pancreatic metaplasia and tumors. Pancreas-specific overexpression of SOX17 led to ductal metaplasia along with inflammation and collagen deposition. Mice that overexpressed SOX17 along with Kras(G12D) had a greater degree of transformed tissue compared with mice expressing only Kras(G12D). Immunofluorescence analysis of human pancreatic tissue arrays revealed the presence of tuft cells in metaplasia and early-stage tumors, along with SOX17 expression, consistent with a biliary phenotype. CONCLUSIONS: Expression of Kras(G12D) and SOX17 in mice induces development of metaplasias with a biliary phenotype containing tuft cells. Tuft cells express a number of tumorigenic factors that can alter the microenvironment. Expression of SOX17 induces pancreatitis and promotes Kras(G12D)-induced tumorigenesis in mice.


Subject(s)
Bile Ducts/cytology , Carcinoma, Pancreatic Ductal/pathology , Cell Transformation, Neoplastic/pathology , HMGB Proteins/metabolism , Pancreatic Ducts/pathology , Pancreatic Neoplasms/pathology , Precancerous Conditions/pathology , SOXF Transcription Factors/metabolism , Animals , Bile Ducts/metabolism , Carcinoma, Pancreatic Ductal/complications , Carcinoma, Pancreatic Ductal/metabolism , Cell Transformation, Neoplastic/metabolism , Humans , Metaplasia/complications , Metaplasia/metabolism , Metaplasia/pathology , Mice , Mice, Transgenic , Pancreatic Ducts/cytology , Pancreatic Ducts/metabolism , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/metabolism , Pancreatitis/metabolism , Precancerous Conditions/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction
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.
Semin Cell Dev Biol ; 23(6): 673-84, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22728667

ABSTRACT

Pancreatic endocrine cells are produced from a dynamic epithelium in a process that, as in any developing organ, is driven by interacting programs of spatiotemporally regulated intercellular signals and autonomous gene regulatory networks. These algorithms work to push progenitors and their transitional intermediates through a series of railroad-station-like switching decisions to regulate flux along specific differentiation tracks. Extensive research on pancreas organogenesis over the last 20 years, greatly spurred by the potential to restore functional ß-cell mass in diabetic patients by transplantation therapy, is advancing our knowledge of how endocrine lineage bias is established and allocation is promoted. The field is working towards the goal of generating a detailed blueprint of how heterogeneous cell populations interact and respond to each other, and other influences such as the extracellular matrix, to move into progressively refined and mature cell states. Here, we highlight how signaling codes and transcriptional networks might determine endocrine lineage within a complex and dynamic architecture, based largely on studies in the mouse. The process begins with the designation of multipotent progenitor cells (MPC) to pancreatic buds that subsequently move through a newly proposed period involving epithelial plexus formation-remodeling, and ends with formation of clustered endocrine islets connected to the vascular and peripheral nervous systems. Developing this knowledge base, and increasing the emphasis on direct comparisons between mouse and human, will yield a more complete and focused picture of pancreas development, and thereby inform ß-cell-directed differentiation from human embryonic stem or induced pluripotent stem cells (hESC, iPSC). Additionally, a deeper understanding may provide surprising therapeutic angles by defining conditions that allow the controllable reprogramming of endodermal or pancreatic cell populations.


Subject(s)
Islets of Langerhans/cytology , Islets of Langerhans/embryology , Multipotent Stem Cells/metabolism , Organogenesis , Animals , Gene Regulatory Networks , Humans , Islets of Langerhans/metabolism , Pancreas/cytology , Pancreas/embryology , Pancreas/metabolism
20.
Cell Mol Gastroenterol Hepatol ; 18(2): 101347, 2024.
Article in English | MEDLINE | ID: mdl-38670488

ABSTRACT

BACKGROUND & AIM: Telocytes, a recently identified type of subepithelial interstitial cell, have garnered attention for their potential roles in tissue homeostasis and repair. However, their contribution to gastric metaplasia remains unexplored. This study elucidates the role of telocytes in the development of metaplasia within the gastric environment. METHODS: To investigate the presence and behavior of telocytes during metaplastic transitions, we used drug-induced acute injury models (using DMP-777 or L635) and a genetically engineered mouse model (Mist1-Kras). Lineage tracing via the Foxl1-CreERT2;R26R-tdTomato mouse model was used to track telocyte migratory dynamics. Immunofluorescence staining was used to identify telocyte markers and evaluate their correlation with metaplasia-related changes. RESULTS: We confirmed the existence of FOXL1+/PDGFRα+ double-positive telocytes in the stomach's isthmus region. As metaplasia developed, we observed a marked increase in the telocyte population. The distribution of telocytes expanded beyond the isthmus to encompass the entire gland and closely reflected the expansion of the proliferative cell zone. Rather than a general response to mucosal damage, the shift in telocyte distribution was associated with the establishment of a metaplastic cell niche at the gland base. Furthermore, lineage-tracing experiments highlighted the active recruitment of telocytes to the emerging metaplastic cell niche, and we observed expression of Wnt5a, Bmp4, and Bmp7 in PDGFRα+ telocytes. CONCLUSIONS: These results suggest that telocytes contribute to the evolution of a gastric metaplasia niche. The dynamic behavior of these stromal cells, their responsiveness to metaplastic changes, and potential association with Wnt5a, Bmp4, and Bmp7 signaling emphasize the significance of telocytes in tissue adaptation and repair.


Subject(s)
Bone Morphogenetic Protein 4 , Gastric Mucosa , Metaplasia , Receptor, Platelet-Derived Growth Factor alpha , Telocytes , Wnt-5a Protein , Animals , Metaplasia/pathology , Mice , Telocytes/metabolism , Telocytes/pathology , Wnt-5a Protein/metabolism , Gastric Mucosa/pathology , Gastric Mucosa/metabolism , Bone Morphogenetic Protein 4/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Stomach/pathology , Bone Morphogenetic Protein 7/metabolism , Cell Movement , Mice, Transgenic , Disease Models, Animal , Forkhead Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL