Catalogue Search | MBRL
Search Results Heading
Explore the vast range of titles available.
MBRLSearchResults
-
DisciplineDiscipline
-
Is Peer ReviewedIs Peer Reviewed
-
Item TypeItem Type
-
SubjectSubject
-
YearFrom:-To:
-
More FiltersMore FiltersSourceLanguage
Done
Filters
Reset
7,002
result(s) for
"Pancreas - growth "
Sort by:
The Map3k12 (Dlk)/JNK3 signaling pathway is required for pancreatic beta-cell proliferation during postnatal development
2021
Unveiling the key pathways underlying postnatal beta-cell proliferation can be instrumental to decipher the mechanisms of beta-cell mass plasticity to increased physiological demand of insulin during weight gain and pregnancy. Using transcriptome and global Serine Threonine Kinase activity (STK) analyses of islets from newborn (10 days old) and adult rats, we found that highly proliferative neonatal rat islet cells display a substantially elevated activity of the mitogen activated protein 3 kinase 12, also called dual leucine zipper-bearing kinase (Dlk). As a key upstream component of the c-Jun amino terminal kinase (Jnk) pathway, Dlk overexpression was associated with increased Jnk3 activity and was mainly localized in the beta-cell cytoplasm. We provide the evidence that Dlk associates with and activates Jnk3, and that this cascade stimulates the expression of Ccnd1 and Ccnd2, two essential cyclins controlling postnatal beta-cell replication. Silencing of Dlk or of Jnk3 in neonatal islet cells dramatically hampered primary beta-cell replication and the expression of the two cyclins. Moreover, the expression of Dlk,Jnk3,Ccnd1 and Ccnd2 was induced in high replicative islet beta cells from ob/ob mice during weight gain, and from pregnant female rats. In human islets from non-diabetic obese individuals, DLK expression was also cytoplasmic and the rise of the mRNA level was associated with an increase of JNK3,CCND1andCCND2 mRNA levels, when compared to islets from lean and obese patients with diabetes. In conclusion, we find that activation of Jnk3 signalling by Dlk could be a key mechanism for adapting islet beta-cell mass during postnatal development and weight gain.
Journal Article
Pancreas regeneration
2018
The pancreas is made from two distinct components: the exocrine pancreas, a reservoir of digestive enzymes, and the endocrine islets, the source of the vital metabolic hormone insulin. Human islets possess limited regenerative ability; loss of islet β-cells in diseases such as type 1 diabetes requires therapeutic intervention. The leading strategy for restoration of β-cell mass is through the generation and transplantation of new β-cells derived from human pluripotent stem cells. Other approaches include stimulating endogenous β-cell proliferation, reprogramming non-β-cells to β-like cells, and harvesting islets from genetically engineered animals. Together these approaches form a rich pipeline of therapeutic development for pancreatic regeneration.
Journal Article
Simultaneous lineage tracing and cell-type identification using CRISPR–Cas9-induced genetic scars
2018
LINNAEUS reconstructs developmental lineages using RNA sequencing data and lineage markers from the same single cells.
A key goal of developmental biology is to understand how a single cell is transformed into a full-grown organism comprising many different cell types. Single-cell RNA-sequencing (scRNA-seq) is commonly used to identify cell types in a tissue or organ
1
. However, organizing the resulting taxonomy of cell types into lineage trees to understand the developmental origin of cells remains challenging. Here we present LINNAEUS (lineage tracing by nuclease-activated editing of ubiquitous sequences)—a strategy for simultaneous lineage tracing and transcriptome profiling in thousands of single cells. By combining scRNA-seq with computational analysis of lineage barcodes, generated by genome editing of transgenic reporter genes, we reconstruct developmental lineage trees in zebrafish larvae, and in heart, liver, pancreas, and telencephalon of adult fish. LINNAEUS provides a systematic approach for tracing the origin of novel cell types, or known cell types under different conditions.
Journal Article
Human pluripotent stem cell-derived acinar/ductal organoids generate human pancreas upon orthotopic transplantation and allow disease modelling
by
Lechel, André
,
Costa, Ivan G
,
Mayerle, Julia
in
Acinar Cells - cytology
,
Animals
,
Cystic fibrosis
2017
ObjectiveThe generation of acinar and ductal cells from human pluripotent stem cells (PSCs) is a poorly studied process, although various diseases arise from this compartment.DesignWe designed a straightforward approach to direct human PSCs towards pancreatic organoids resembling acinar and ductal progeny.ResultsExtensive phenotyping of the organoids not only shows the appropriate marker profile but also ultrastructural, global gene expression and functional hallmarks of the human pancreas in the dish. Upon orthotopic transplantation into immunodeficient mice, these organoids form normal pancreatic ducts and acinar tissue resembling fetal human pancreas without evidence of tumour formation or transformation. Finally, we implemented this unique phenotyping tool as a model to study the pancreatic facets of cystic fibrosis (CF). For the first time, we provide evidence that in vitro, but also in our xenograft transplantation assay, pancreatic commitment occurs generally unhindered in CF. Importantly, cystic fibrosis transmembrane conductance regulator (CFTR) activation in mutated pancreatic organoids not only mirrors the CF phenotype in functional assays but also at a global expression level. We also conducted a scalable proof-of-concept screen in CF pancreatic organoids using a set of CFTR correctors and activators, and established an mRNA-mediated gene therapy approach in CF organoids.ConclusionsTaken together, our platform provides novel opportunities to model pancreatic disease and development, screen for disease-rescuing agents and to test therapeutic procedures.
Journal Article
Spatiotemporal profiling of adhesion G protein-coupled receptors in developing mouse and human pancreas reveals a role for GPR56 in islet development
2025
Introduction
G protein-coupled receptors (GPCRs) are cell-surface proteins that are targeted therapeutically for a range of disorders, including diabetes. Adhesion GPCRs (aGPCRs) are the second largest class of the GPCR superfamily and some members of this family have been implicated in appropriate organ development. However, the role of aGPCRs in endocrine pancreas specification is not yet known.
Methods
Here, we systematically characterised expression of mRNAs encoding aGPCRs and their ligands in developing mouse and human pancreas using our own and publicly available single-cell RNA sequencing and spatial transcriptomics data, and we conducted qPCR analysis of aGPCR expression in human pancreas at different gestational stages. We then investigated the role of GPR56 (ADGRG1), the most abundant aGPCR in pancreatic endocrine progenitors, in islet development using
Gpr56
null mice and their wildtype littermates.
Results
We demonstrated that aGPCRs are dynamically expressed during mouse and human pancreas development, with specific aGPCR mRNAs expressed in distinct endocrine, endothelial, mesenchymal, acinar, ductal, and immune cell clusters. aGPCR ligand mRNAs were mostly expressed by non-endocrine cells, and the most highly expressed receptor-ligand interacting mRNA pairs were those encoding GPR56 and COL3A1. Deletion of
Gpr56
in neonatal mice was associated with an altered α-/β-/δ-cell ratio and reduced β-cell proliferation.
Conclusion
Our data show that aGPCRs are expressed at key stages of human and mouse pancreas endocrine lineage decisions, and analysis of pancreases from
Gpr56
knockout mice implicate this aGPCR in the development of a full complement of β-cells.
Journal Article
Pancreatic Ppy-expressing γ-cells display mixed phenotypic traits and the adaptive plasticity to engage insulin production
2021
The cellular identity of pancreatic polypeptide (Ppy)-expressing γ-cells, one of the rarest pancreatic islet cell-type, remains elusive. Within islets, glucagon and somatostatin, released respectively from α- and δ-cells, modulate the secretion of insulin by β-cells. Dysregulation of insulin production raises blood glucose levels, leading to diabetes onset. Here, we present the genetic signature of human and mouse γ-cells. Using different approaches, we identified a set of genes and pathways defining their functional identity. We found that the γ-cell population is heterogeneous, with subsets of cells producing another hormone in addition to Ppy. These bihormonal cells share identity markers typical of the other islet cell-types. In mice,
Ppy
gene inactivation or conditional γ-cell ablation did not alter glycemia nor body weight. Interestingly, upon β-cell injury induction, γ-cells exhibited gene expression changes and some of them engaged insulin production, like α- and δ-cells. In conclusion, we provide a comprehensive characterization of γ-cells and highlight their plasticity and therapeutic potential.
The cellular identity and function of the pancreatic polypeptide (Ppy)-producing γ-cells are incompletely understood. Here the authors show that these cells are heterogeneous and display adaptive plasticity to engage in insulin production following β-cell injury, but loss of the Ppy gene or γ-cells in mice does not affect weight or glycemia under basal conditions.
Journal Article
β-Cell Replication Is the Primary Mechanism Subserving the Postnatal Expansion of β-Cell Mass in Humans
2008
β-Cell Replication Is the Primary Mechanism Subserving the Postnatal Expansion of β-Cell Mass in Humans
Juris J. Meier 1 ,
Alexandra E. Butler 1 ,
Yoshifumi Saisho 1 ,
Travis Monchamp 2 ,
Ryan Galasso 1 ,
Anil Bhushan 1 ,
Robert A. Rizza 3 and
Peter C. Butler 1
1 Larry Hillblom Islet Research Center, UCLA David Geffen School of Medicine, Los Angeles, California
2 Division of Endocrinology, Diabetes and Hypertension, UCLA David Geffen School of Medicine, Los Angeles, California
3 Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, Minnesota
Corresponding author: Peter C. Butler, Larry Hillblom Islet Research Center, UCLA David Geffen School of Medicine, 24-130
Warren Hall, 900 Veteran Ave., Los Angeles, CA 90095-7073. E-mail: pbutler{at}mednet.ucla.edu
Abstract
OBJECTIVE— Little is known about the capacity, mechanisms, or timing of growth in β-cell mass in humans. We sought to establish if the
predominant expansion of β-cell mass in humans occurs in early childhood and if, as in rodents, this coincides with relatively
abundant β-cell replication. We also sought to establish if there is a secondary growth in β-cell mass coincident with the
accelerated somatic growth in adolescence.
RESEARCH DESIGN AND METHODS— To address these questions, pancreas volume was determined from abdominal computer tomographies in 135 children aged 4 weeks
to 20 years, and morphometric analyses were performed in human pancreatic tissue obtained at autopsy from 46 children aged
2 weeks to 21 years.
RESULTS— We report that 1 ) β-cell mass expands by severalfold from birth to adulthood, 2 ) islets grow in size rather than in number during this transition, 3 ) the relative rate of β-cell growth is highest in infancy and gradually declines thereafter to adulthood with no secondary
accelerated growth phase during adolescence, 4 ) β-cell mass (and presumably growth) is highly variable between individuals, and 5 ) a high rate of β-cell replication is coincident with the major postnatal expansion of β-cell mass.
CONCLUSIONS— These data imply that regulation of β-cell replication during infancy plays a major role in β-cell mass in adult humans.
CT, computed tomography
Footnotes
Published ahead of print at http://diabetes.diabetesjournals.org on 11 March 2008. DOI: 10.2337/db07-1369.
The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore
be hereby marked “advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.
Accepted February 26, 2008.
Received September 26, 2007.
DIABETES
Journal Article
Pancreatic Islet Production of Vascular Endothelial Growth Factor-A Is Essential for Islet Vascularization, Revascularization, and Function
by
Jin Chen
,
Maureen Gannon
,
Peter O. Wiebe
in
Angiogenesis
,
Animals
,
Biological and medical sciences
2006
Pancreatic Islet Production of Vascular Endothelial Growth Factor-A Is Essential for Islet Vascularization, Revascularization,
and Function
Marcela Brissova 1 ,
Alena Shostak 1 ,
Masakazu Shiota 2 ,
Peter O. Wiebe 2 ,
Greg Poffenberger 1 ,
Jeannelle Kantz 2 ,
Zhongyi Chen 1 ,
Chad Carr 1 ,
W. Gray Jerome 3 4 ,
Jin Chen 4 5 ,
H. Scott Baldwin 6 ,
Wendell Nicholson 1 ,
David M. Bader 7 ,
Thomas Jetton 8 ,
Maureen Gannon 1 2 and
Alvin C. Powers 1 2 9
1 Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville,
Tennessee
2 Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee
3 Department of Pathology, Vanderbilt University Medical Center, Nashville, Tennessee
4 Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee
5 Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee
6 Division of Pediatric Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
7 Stahlman Laboratory, Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville,
Tennessee
8 Division of Endocrinology and Metabolism, Department of Medicine, University of Vermont College of Medicine, Burlington, Vermont
9 Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee
Address correspondence and reprint requests to Alvin C. Powers, Division of Diabetes, Endocrinology, and Metabolism, 715 PRB,
Vanderbilt University, Nashville, TN 37232. E-mail: al.powers{at}vanderbilt.edu
Abstract
To investigate molecular mechanisms controlling islet vascularization and revascularization after transplantation, we examined
pancreatic expression of three families of angiogenic factors and their receptors in differentiating endocrine cells and adult
islets. Using intravital lectin labeling, we demonstrated that development of islet microvasculature and establishment of
islet blood flow occur concomitantly with islet morphogenesis. Our genetic data indicate that vascular endothelial growth
factor (VEGF)-A is a major regulator of islet vascularization and revascularization of transplanted islets. In spite of normal
pancreatic insulin content and β-cell mass, mice with β-cell–reduced VEGF-A expression had impaired glucose-stimulated insulin
secretion. By vascular or diffusion delivery of β-cell secretagogues to islets, we showed that reduced insulin output is not
a result of β-cell dysfunction but rather caused by vascular alterations in islets. Taken together, our data indicate that
the microvasculature plays an integral role in islet function. Factors modulating VEGF-A expression may influence islet vascularity
and, consequently, the amount of insulin delivered into the systemic circulation.
Ang, angiopoietin
VEGF, vascular endothelial growth factor
Footnotes
Additional information for this article can be found in an online appendix at http://diabetes.diabetesjournals.org .
The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore
be hereby marked “advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.
Accepted July 19, 2006.
Received May 18, 2006.
DIABETES
Journal Article
PPDPF is not a key regulator of human pancreas development
by
Simon, Eric
,
Hauff, Natalie
,
Kleger, Alexander
in
Animals
,
Biology and Life Sciences
,
Cell differentiation
2025
Given their capability to differentiate into each cell type of the human body, human pluripotent stem cells (hPSCs) provide a unique platform for developmental studies. In the current study, we employed this cell system to understand the role of pancreatic progenitor differentiation and proliferation factor (PPDPF), a protein that has been little explored so far. While the zebrafish orthologue exdpf is essential for exocrine pancreas specification, its importance for mammalian and human development has not been studied yet. We implemented a four times CRISPR/Cas9 nicking approach to knockout PPDPF in human embryonic stem cells (hESCs) and differentiated PPDPF KO/KO and PPDPF WT/WT cells towards the pancreatic lineage. In contrast to data obtained from zebrafish, a very modest effect of the knockout was observed in the development of pancreatic progenitors in vitro , not affecting lineage specification upon orthotopic transplantation in vivo . The modest effect is in line with the finding that genetic variants near PPDPF are associated with random glucose levels in humans, but not with type 2 diabetes risk, supporting that dysregulation of this gene may only result in minor alterations of glycaemic balance in humans. In addition, PPDPF is less organ- and cell type specifically expressed in higher vertebrates and its so far reported functions appear highly context-dependent.
Journal Article
The transcription factors Tfeb and Tfe3 are required for survival and embryonic development of pancreas and liver in zebrafish
by
Chen, Vicky
,
Burgess, Shawn M.
,
La Spina, Martina
in
Animals
,
Apoptosis
,
Apoptosis - genetics
2025
The transcription factors TFEB and TFE3 modulate expression of lysosomal, autophagic, and metabolic genes to restore energy and cellular homeostasis in response to a variety of stress conditions. Since their role during vertebrate development is less characterized, we used CRISPR/Cas9 to deplete tfeb , tfe3a , and tfe3b in zebrafish. The simultaneous lack of these genes compromised embryo survival during early development, with an almost complete lethality of the larvae by 8–10 dpf. The knockout animals showed apoptosis in brain and retina and alterations in pancreas, liver, and gut. Exocrine pancreas presented the most severe defects, with accumulation of abnormal zymogen granules leading to acinar atrophy in embryos and pancreatitis-like phenotypes in adults; likely due to a block of the autophagy machinery implicated in removal of damaged granules. Knockout animals displayed increased susceptibility to oxidative and heat-shock stress. Our work reveals an essential role of Tfeb and Tfe3 in maintaining cellular and tissue homeostasis during development.
Journal Article