Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
1.
Cells ; 10(7)2021 07 06.
Article in English | MEDLINE | ID: mdl-34359881

ABSTRACT

Proper functioning of the body depends on hormonal homeostasis. White adipose tissue is now known as an endocrine organ due to the secretion of multiple molecules called adipokines. These proteins exert direct effects on whole body functions, including lipid metabolism, angiogenesis, inflammation, and reproduction, whereas changes in their level are linked with pathological events, such as infertility, diabetes, and increased food intake. Vaspin-visceral adipose tissue-derived serine protease inhibitor, or SERPINA12 according to serpin nomenclature, is an adipokine discovered in 2005 that is connected to the development of insulin resistance, obesity, and inflammation. A significantly higher amount of vaspin was observed in obese patients. The objective of this review was to summarize the latest findings about vaspin expression and action in endocrine tissues, such as the hypothalamus, pituitary gland, adipose tissue, thyroid, ovary, placenta, and testis, as well as discuss the link between vaspin and pathologies connected with hormonal imbalance.


Subject(s)
Diabetes Mellitus/genetics , Endocrine Cells/metabolism , Hypothalamo-Hypophyseal System/metabolism , Infertility/genetics , Obesity/genetics , Serpins/genetics , Adipose Tissue/cytology , Adipose Tissue/metabolism , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Endocrine Cells/cytology , Female , Gene Expression Regulation , Gonads/cytology , Gonads/metabolism , Humans , Hypothalamo-Hypophyseal System/cytology , Infertility/metabolism , Infertility/pathology , Insulin Resistance , Lipid Metabolism/genetics , Male , Neovascularization, Physiologic/genetics , Obesity/metabolism , Obesity/pathology , Reproduction/genetics , Serpins/metabolism , Thyroid Gland/cytology , Thyroid Gland/metabolism
2.
In Vitro Cell Dev Biol Anim ; 57(6): 587-597, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34212340

ABSTRACT

Conventional methods for obtaining pancreatic ß cells are based on simulating the embryonic development phase of endocrine cells via hierarchical differentiation of pluripotent stem cells (PSCs). Accordingly, we attempted to modify the protocols for obtaining insulin-secreting cells (ISCs) by sequential differentiation of a human embryonic stem cell (hESC), using the HS181 cell line. Furthermore, we hypothesize that actual pancreatic endocrine cells may arise from trans-differentiation of mature ductal cells after the embryonic developmental stage and throughout the rest of life. According to the hypothesis, ductal cells are trans-differentiated into endocrine and exocrine cells, undergoing a partial epithelial to mesenchymal transition (EMT). To address this issue, we developed two new protocols based on hESC differentiation to obtain ductal cells and then induce EMT in cells to obtain hormone-secreting islet-like cells (HSCs). The ductal (pre-EMT exocrine) cells were then induced to undergo partial EMT by treating with Wnt3a and activin A, in hypoxia. The cell derived from the latter method significantly expressed the main endocrine cell-specific markers and also ß cells, in particular. These experiments not only support our hypothetical model but also offer a promising approach to develop new methods to compensate ß cell depletion in patients with type 1 diabetes mellitus (T1DM). Although this protocol of generating islet-like cells from ductal cells has a potential to treat T1DM, this strategy may be exploited to optimize the function of these cells in an animal model and future clinical applications.


Subject(s)
Cell Transdifferentiation/genetics , Diabetes Mellitus, Type 1/therapy , Human Embryonic Stem Cells/cytology , Pluripotent Stem Cells/cytology , Wnt3A Protein/genetics , Cell Culture Techniques , Cell Differentiation/genetics , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Endocrine Cells/cytology , Epithelial-Mesenchymal Transition/genetics , Human Embryonic Stem Cells/transplantation , Humans , Insulin/genetics , Insulin/metabolism , Insulin Secretion/genetics , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/transplantation , Pancreas/growth & development , Pancreas/pathology , Pluripotent Stem Cells/transplantation
3.
FASEB J ; 35(5): e21400, 2021 05.
Article in English | MEDLINE | ID: mdl-33793981

ABSTRACT

Anterior pituitary endocrine cells that release hormones such as growth hormone and prolactin are excitable and fire action potentials. In these cells, several studies previously showed that extracellular sodium (Na+ ) removal resulted in a negative shift of the resting membrane potential (RMP) and a subsequent inhibition of the spontaneous firing of action potentials, suggesting the contribution of a Na+ background conductance. Here, we show that the Na+ leak channel NALCN conducts a Ca2+ - Gd3+ -sensitive and TTX-resistant Na+ background conductance in the GH3 cell line, a cell model of pituitary endocrine cells. NALCN knockdown hyperpolarized the RMP, altered GH3 cell electrical properties and inhibited prolactin secretion. Conversely, the overexpression of NALCN depolarized the RMP, also reshaping the electrical properties of GH3 cells. Overall, our results indicate that NALCN is functional in GH3 cells and involved in endocrine cell excitability as well as in hormone secretion. Indeed, the GH3 cell line suitably models native pituitary cells that display a similar Na+ background conductance and appears as a proper cellular model to study the role of NALCN in cellular excitability.


Subject(s)
Action Potentials , Endocrine Cells/physiology , Ion Channels/metabolism , Membrane Potentials , Membrane Proteins/metabolism , Pituitary Gland/physiology , Sodium/metabolism , Animals , Endocrine Cells/cytology , Pituitary Gland/cytology , Rats
4.
J Neuroendocrinol ; 32(10): e12903, 2020 10.
Article in English | MEDLINE | ID: mdl-32959418

ABSTRACT

The anterior and intermediate lobes of the pituitary are composed of endocrine cells, as well as vasculature and supporting cells, such as folliculostellate cells. Folliculostellate cells form a network with several postulated roles in the pituitary, including production of paracrine signalling molecules and cytokines, coordination of endocrine cell hormone release, phagocytosis, and structural support. Folliculostellate cells in rats are characterised by expression of S100B protein, and in humans by glial fibrillary acid protein. However, there is evidence for another network of supporting cells in the anterior pituitary that has properties of mural cells, such as vascular smooth muscle cells and pericytes. The present study aims to characterise the distribution of cells that express the mural cell marker platelet derived growth factor receptor beta (PDGFRß) in the mouse pituitary and establish whether these cells are folliculostellate. By immunohistochemical localisation, we determine that approximately 80% of PDGFRß+ cells in the mouse pituitary have a non-perivascular location and 20% are pericytes. Investigation of gene expression in a magnetic cell sorted population of PDGFRß+ cells shows that, despite a mostly non-perivascular location, this population is enriched for mural cell markers but not enriched for rat or human folliculostellate cell markers. This is confirmed by immunohistochemistry. The present study concludes that a mural cell network is present throughout the anterior pituitary of the mouse and that this population does not express well-characterised human or rat folliculostellate cell markers.


Subject(s)
Cell Communication/physiology , Pituitary Gland/cytology , Animals , Biomarkers/metabolism , Endocrine Cells/cytology , Endocrine Cells/physiology , Endothelial Cells/cytology , Endothelial Cells/physiology , Mice , Mice, Inbred C57BL , Pericytes/cytology , Pericytes/physiology , Pituitary Gland/metabolism , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , SOXB1 Transcription Factors/metabolism
5.
Mol Cell Endocrinol ; 518: 110877, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32569857

ABSTRACT

The reproductive neuroendocrine axis, or hypothalamo-pituitary-gonadal (HPG) axis, is a paragon of complex biological system involving numerous cell types, spread over several anatomical levels communicating through entangled endocrine feedback loops. The HPG axis exhibits remarkable dynamic behaviors on multiple time and space scales, which are an inexhaustible source of studies for mathematical and computational biology. In this review, we will describe a variety of modeling approaches of the HPG axis from a cellular endocrinology viewpoint. We will in particular investigate the questions raised by some of the most striking features of the HPG axis: (i) the pulsatile secretion of hypothalamic and pituitary hormones, and its counterpart, the cell signaling induced by frequency-encoded hormonal signals, and (ii) the dual, gametogenic and glandular function of the gonads, which relies on the tight control of the somatic cell populations ensuring the proper maturation and timely release of the germ cells.


Subject(s)
Endocrine Cells/physiology , Gonads/cytology , Hypothalamo-Hypophyseal System/cytology , Models, Theoretical , Pituitary-Adrenal System/cytology , Animals , Endocrine Cells/cytology , Endocrinology/methods , Female , Gonads/physiology , Humans , Hypothalamo-Hypophyseal System/physiology , Male , Pituitary-Adrenal System/physiology , Reproduction/physiology , Signal Transduction/physiology
6.
Nat Commun ; 11(1): 2082, 2020 04 29.
Article in English | MEDLINE | ID: mdl-32350257

ABSTRACT

Developmental progression depends on temporally defined changes in gene expression mediated by transient exposure of lineage intermediates to signals in the progenitor niche. To determine whether cell-intrinsic epigenetic mechanisms contribute to signal-induced transcriptional responses, here we manipulate the signalling environment and activity of the histone demethylase LSD1 during differentiation of hESC-gut tube intermediates into pancreatic endocrine cells. We identify a transient requirement for LSD1 in endocrine cell differentiation spanning a short time-window early in pancreas development, a phenotype we reproduced in mice. Examination of enhancer and transcriptome landscapes revealed that LSD1 silences transiently active retinoic acid (RA)-induced enhancers and their target genes. Furthermore, prolonged RA exposure phenocopies LSD1 inhibition, suggesting that LSD1 regulates endocrine cell differentiation by limiting the duration of RA signalling. Our findings identify LSD1-mediated enhancer silencing as a cell-intrinsic epigenetic feedback mechanism by which the duration of the transcriptional response to a developmental signal is limited.


Subject(s)
Endocrine Cells/cytology , Endocrine Cells/metabolism , Enhancer Elements, Genetic/genetics , Gene Silencing , Histone Demethylases/metabolism , Islets of Langerhans/cytology , Signal Transduction , Tretinoin/metabolism , Adult , Animals , Base Sequence , Cell Differentiation/drug effects , Endocrine Cells/drug effects , Female , Gene Expression Regulation, Developmental/drug effects , Gene Silencing/drug effects , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/drug effects , Human Embryonic Stem Cells/metabolism , Humans , Islets of Langerhans/embryology , Male , Mice , Signal Transduction/drug effects , Transcription Factors/metabolism , Tretinoin/pharmacology , Young Adult
7.
Bull Exp Biol Med ; 168(6): 797-801, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32328948

ABSTRACT

We studied the expression of transcriptional factors regulating postnatal morphogenesis of the adrenal zona fasciculata in rats after developmental exposure to endocrine disruptor DDT. It was found that tissue reparation after trophic disorders and cell death triggered by prenatal and postnatal exposure to DDT was accompanied by an increase in the number of Oct4- and Shh-expressing cells forming a pool located outside the regeneration zones and involved in the maintenance of tissue homeostasis in the zona fasciculata. DDT exposure also disrupted the expression of antiproliferative factor Hhex. The data showed that proliferation of fasciculata cells after termination of adrenal cortex growth was downregulated by inhibition of the expression of Oct4 and Shh and suppression of canonical Wnt signaling, i.e. due to a decrease in the reserve cell pool essential for physiological regeneration, which can reduce the reactive potential of the zona fasciculata.


Subject(s)
DDT/pharmacology , Endocrine Cells/drug effects , Endocrine Disruptors/pharmacology , Prenatal Exposure Delayed Effects/genetics , Transcription, Genetic/drug effects , Zona Fasciculata/drug effects , Animals , Animals, Newborn , Cell Proliferation/drug effects , Endocrine Cells/cytology , Endocrine Cells/metabolism , Female , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Male , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/pathology , Rats , Rats, Wistar , Wnt Signaling Pathway , Zona Fasciculata/cytology , Zona Fasciculata/growth & development , Zona Fasciculata/metabolism
8.
Article in English | MEDLINE | ID: mdl-32296396

ABSTRACT

Insulin and glucagon are hormones secreted by pancreatic ß and α cells, respectively, which together regulate glucose homeostasis. Dysregulation of insulin or glucagon can result in loss of blood glucose control, characterized by hyperglycemia or hypoglycemia. To better understand the endocrine physiology of cetaceans, we cloned and characterized the insulin and glucagon genes from pygmy sperm whale (Kogia breviceps). We obtained the complete coding sequences of the preproinsulin and preproglucagon genes, which encodes the preproinsulin protein of 110 amino acid (aa) residues and encodes the preproglucagon protein of 179 aa residues, respectively. Sequence comparison and phylogenetic analyses demonstrate that protein structures were similar to other mammalian orthologs. Immunohistochemistry and immunofluorescence staining using insulin, glucagon, and somatostatin antibodies allowed analysis of pygmy sperm whale islet distribution, architecture, and composition. Our results showed the pygmy sperm whale islet was irregularly shaped and randomly distributed throughout the pancreas. The architecture of α, ß, and δ cells of the pygmy sperm whale was similar to that of artiodactyls species. This is the first report about insulin and glucagon genes in cetaceans, which provides new information about the structural conservation of the insulin and glucagon genes. Furthermore, offers novel information on the properties of endocrine cells in cetacean for further studies.


Subject(s)
Endocrine Cells/metabolism , Glucagon/genetics , Insulin/genetics , Whales , Animals , Cloning, Molecular , Endocrine Cells/cytology , Female , Glucagon/metabolism , Glucagon-Secreting Cells/cytology , Glucagon-Secreting Cells/metabolism , Glucagon-Secreting Cells/physiology , Insulin/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/physiology , Male , Mice , Mice, Inbred C57BL , Pancreas/cytology , Pancreas/metabolism , Phylogeny , Protein Precursors/genetics , Protein Precursors/metabolism , Sequence Analysis, DNA/veterinary , Whales/genetics , Whales/metabolism
9.
PLoS Comput Biol ; 16(4): e1007769, 2020 04.
Article in English | MEDLINE | ID: mdl-32251433

ABSTRACT

Endocrine cells in the pituitary gland typically display either spiking or bursting electrical activity, which is related to the level of hormone secretion. Recent work, which combines mathematical modelling with dynamic clamp experiments, suggests the difference is due to the presence or absence of a few large-conductance potassium channels. Since endocrine cells only contain a handful of these channels, it is likely that stochastic effects play an important role in the pattern of electrical activity. Here, for the first time, we explicitly determine the effect of such noise by studying a mathematical model that includes the realistic noisy opening and closing of ion channels. This allows us to investigate how noise affects the electrical activity, examine the origin of spiking and bursting, and determine which channel types are responsible for the greatest noise. Further, for the first time, we address the role of cell size in endocrine cell electrical activity, finding that larger cells typically display more bursting, while the smallest cells almost always only exhibit spiking behaviour.


Subject(s)
Action Potentials/physiology , Endocrine Cells , Ion Channels/physiology , Models, Neurological , Neurons , Animals , Computational Biology , Endocrine Cells/cytology , Endocrine Cells/physiology , Neurons/cytology , Neurons/physiology , Patch-Clamp Techniques , Pituitary Gland/cytology
10.
J Morphol ; 281(1): 55-67, 2020 01.
Article in English | MEDLINE | ID: mdl-31782555

ABSTRACT

Goblet cells (GCs) and endocrine cells (ECs) play an important role in intestine physiology, and few studies currently exist for Amazonian fishes. This study aimed to quantify the distribution of GCs and ECs producing cholecystokinin-8 and neuropeptide Y, assessed by mucin histochemistry and peptides immunohistochemistry, in the intestine of two Amazonian species with different feeding habits Tambaqui (Colossosoma macropomum) and hybrid catfish (Pseudoplatystoma reticulatum × Leiarius marmoratus), an omnivore and carnivore, respectively. A systematic literature review correlating feeding habit and GC and EC distribution was also included to contribute to the comparative study. The results of this study provided novel information about the gut cells of Tambaqui and hybrid catfish. Both, GCs and ECs can be found sweeping the entire intestine of Tambaqui and hybrid catfish although the cells can be more concentrated in certain segments. The GCs and ECs in Tambaqui were more uniformly distributed in the midgut segments (T1, T2, and T3). Unlike, in hybrid catfish GCs were more concentrated in the hindgut (C4) and ECs mainly in the two midgut segments (C1 and C2) of hybrid catfish. Based on the comparison between Tambaqui, hybrid catfish, and other fishes in the literature review, we suggest that cell distribution can be partially explained by feeding habits, carnivorous vs. omnivorous.


Subject(s)
Catfishes/genetics , Endocrine Cells/cytology , Fresh Water , Hybridization, Genetic , Intestines/cytology , Animals , Cell Count , Cholecystokinin/metabolism , Mucins/metabolism , Neuropeptide Y/metabolism , Peptide Fragments/metabolism , Systematic Reviews as Topic
11.
Development ; 146(12)2019 06 17.
Article in English | MEDLINE | ID: mdl-31160421

ABSTRACT

Deciphering mechanisms of endocrine cell induction, specification and lineage allocation in vivo will provide valuable insights into how the islets of Langerhans are generated. Currently, it is ill defined how endocrine progenitors segregate into different endocrine subtypes during development. Here, we generated a novel neurogenin 3 (Ngn3)-Venus fusion (NVF) reporter mouse line, that closely mirrors the transient endogenous Ngn3 protein expression. To define an in vivo roadmap of endocrinogenesis, we performed single cell RNA sequencing of 36,351 pancreatic epithelial and NVF+ cells during secondary transition. This allowed Ngn3low endocrine progenitors, Ngn3high endocrine precursors, Fev+ endocrine lineage and hormone+ endocrine subtypes to be distinguished and time-resolved, and molecular programs during the step-wise lineage restriction steps to be delineated. Strikingly, we identified 58 novel signature genes that show the same transient expression dynamics as Ngn3 in the 7260 profiled Ngn3-expressing cells. The differential expression of these genes in endocrine precursors associated with their cell-fate allocation towards distinct endocrine cell types. Thus, the generation of an accurately regulated NVF reporter allowed us to temporally resolve endocrine lineage development to provide a fine-grained single cell molecular profile of endocrinogenesis in vivo.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Nerve Tissue Proteins/genetics , Pancreas/embryology , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Animals , Cell Differentiation/genetics , Cell Lineage , Endocrine Cells/cytology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genes, Reporter , Insulin-Secreting Cells/cytology , Mice , Regeneration , Signal Transduction , Stem Cells/cytology , Wnt Proteins/metabolism
12.
PLoS One ; 14(5): e0216254, 2019.
Article in English | MEDLINE | ID: mdl-31075154

ABSTRACT

BACKGROUND: Pancreatic islet xenotransplantation is a potential treatment for diabetes mellitus, and porcine pancreas may provide a readily available source of islets. Islets in juvenile pigs are smaller than those in young adult pigs, but the insulin content is very similar. In addition, as juvenile pigs are more easily reared in uncontaminated conditions, many researchers have conducted studies using pancreatic islets from juvenile pigs. We aimed to analyze the distributions of endocrine cell clusters by comprehensively evaluating juvenile porcine pancreatic development and to propose an appropriate age at which islets could be isolated from the juvenile porcine pancreas. METHODS: Splenic (SL) and duodenal lobe (DL) samples were collected from the pancreases of pigs aged 0-180 days (n = 3/day after birth). The chronological changes in endocrine cell clustering were analyzed in relation to morphological changes, cell characterization, numbers, islet areas, and gene expression. RESULTS: In juvenile pigs aged 0-21 days, the pancreas contained numerous endocrine cells, and compact islets appeared from 21 days of age. Well-defined small islets were seen at 28 days of age, and the clusters were denser in the SL than in the DL. At 35 days of age, the islets were morphologically similar to those observed at 180 days of age, and the greater number of islets was similar to that seen at 90 days of age. The differences in the islets' cytoarchitecture between the lobes were negligible. The expression of ß-cell-related genes was higher in the juvenile pancreas than in the adult pancreas, and the expression of neurogenin-3 decreased dramatically over time. CONCLUSIONS: These findings may have implications for attempts to refine the most appropriate age for islet isolation from porcine donors. Focusing on porcine pancreatic islets isolated at around 35 days after birth may offer benefits regarding their xenotransplantation potential.


Subject(s)
Age Factors , Endocrine Cells/cytology , Islets of Langerhans Transplantation/methods , Islets of Langerhans/growth & development , Transplantation, Heterologous/methods , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cluster Analysis , Diabetes Mellitus/therapy , Humans , Islets of Langerhans/cytology , Nerve Tissue Proteins/metabolism , Swine
13.
Diabetes ; 68(6): 1230-1239, 2019 06.
Article in English | MEDLINE | ID: mdl-30936150

ABSTRACT

Pancreatic ß-cells play a pivotal role in maintaining normoglycemia. Recent studies have revealed that the ß-cell is not a homogeneous cell population but, rather, is heterogeneous in a number of properties such as electrical activity, gene expression, and cell surface markers. Identification of specific ß-cell subpopulations altered in diabetic conditions would open a new avenue to develop targeted therapeutic interventions. As intense studies of ß-cell heterogeneity are anticipated in the next decade, it is important that heterogeneity of the islet be recognized. Many studies in the past were undertaken with a small sample of islets, which might overlook important individual variance. In this study, by systematic analyses of the human islet in two and three dimensions, we demonstrate islet heterogeneity in size, number, architecture, cellular composition, and capillary density. There is no stereotypic human islet, and thus, a sufficient number of islets should be examined to ensure study reproducibility.


Subject(s)
Glucagon-Secreting Cells/cytology , Insulin-Secreting Cells/cytology , Islets of Langerhans/cytology , Somatostatin-Secreting Cells/cytology , Adolescent , Adult , Aged , Animals , Endocrine Cells/cytology , Endocrine Cells/metabolism , Female , Glucagon-Secreting Cells/metabolism , Humans , Imaging, Three-Dimensional , Immunohistochemistry , Insulin-Secreting Cells/metabolism , Islets of Langerhans/blood supply , Islets of Langerhans/metabolism , Male , Mice , Microscopy, Confocal , Middle Aged , Reproducibility of Results , Somatostatin-Secreting Cells/metabolism , Young Adult
14.
Nat Cell Biol ; 21(2): 263-274, 2019 02.
Article in English | MEDLINE | ID: mdl-30710150

ABSTRACT

Despite advances in the differentiation of insulin-producing cells from human embryonic stem cells, the generation of mature functional ß cells in vitro has remained elusive. To accomplish this goal, we have developed cell culture conditions to closely mimic events occurring during pancreatic islet organogenesis and ß cell maturation. In particular, we have focused on recapitulating endocrine cell clustering by isolating and reaggregating immature ß-like cells to form islet-sized enriched ß-clusters (eBCs). eBCs display physiological properties analogous to primary human ß cells, including robust dynamic insulin secretion, increased calcium signalling in response to secretagogues, and improved mitochondrial energization. Notably, endocrine cell clustering induces metabolic maturation by driving mitochondrial oxidative respiration, a process central to stimulus-secretion coupling in mature ß cells. eBCs display glucose-stimulated insulin secretion as early as three days after transplantation in mice. In summary, replicating aspects of endocrine cell clustering permits the generation of stem-cell-derived ß cells that resemble their endogenous counterparts.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Endocrine Cells/cytology , Fibroblasts/cytology , Human Embryonic Stem Cells/cytology , Insulin-Secreting Cells/cytology , Animals , Cells, Cultured , Embryonic Stem Cells/physiology , Endocrine Cells/physiology , Fibroblasts/physiology , Glucose/pharmacology , Human Embryonic Stem Cells/physiology , Humans , Insulin Secretion/drug effects , Insulin-Secreting Cells/physiology , Islets of Langerhans/cytology , Mice , Mitochondria/metabolism
15.
Sci Rep ; 9(1): 640, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30679498

ABSTRACT

Large numbers of hormone-releasing cells, approximately 109 endocrine cells, are required to treat type I diabetes patients by cell transplantation. The SOX9-positive pancreatic epithelium proliferates extensively during the early stages of pancreatic development. SOX9-positive pancreatic epithelium is thought to be an expandable cell source of ß cells for transplantation therapy. In this study, we attempted to expand pancreatic progenitors (PPs: PDX1+/SOX9+) derived from four human iPSC lines in three-dimensional (3D) culture using a chemically defined medium and examined the potential of the derived PPs to differentiate into ß-like cells. PPs from four human iPSC lines were maintained and effectively proliferated in a chemically defined medium containing epidermal growth factor and R-spondin-1, CHIR99021, fibroblast growth factor-7, and SB431542. PPs derived from one iPSC line can be expanded by more than 104-fold in chemically defined medium containing two of the fives, epidermal growth factor and R-spondin-1. The expanded PPs were also stable following cryopreservation. After freezing and thawing, the PPs proliferated without a decrease in the rate. PPs obtained after 50 days of culture successfully differentiated into insulin-positive ß-like cells, glucagon-positive α-like cells, and somatostatin-positive δ-like cells. The differentiation efficiency of expanded PPs was similar to that of PPs without expansion culture.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Pancreas/cytology , Benzamides/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Dioxoles/pharmacology , Endocrine Cells/cytology , Endocrine Cells/drug effects , Epidermal Growth Factor/pharmacology , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Pyridines/pharmacology , Pyrimidines/pharmacology
16.
Traffic ; 20(2): 137-151, 2019 02.
Article in English | MEDLINE | ID: mdl-30426623

ABSTRACT

The male seminal fluid contains factors that affect female post-mating behavior and physiology. In Drosophila, most of these factors are secreted by the two epithelial cell types that make up the male accessory gland: the main and secondary cells. Although secondary cells represent only ~4% of the cells of the accessory gland, their contribution to the male seminal fluid is essential for sustaining the female post-mating response. To better understand the function of the secondary cells, we investigated their molecular organization, particularly with respect to the intracellular membrane transport machinery. We determined that large vacuole-like structures found in the secondary cells are trafficking hubs labeled by Rab6, 7, 11 and 19. Furthermore, these organelles require Rab6 for their formation and many are essential in the process of creating the long-term postmating behavior of females. In order to better serve the intracellular membrane and protein trafficking communities, we have created a searchable, online, open-access imaging resource to display our complete findings regarding Rab localization in the accessory gland.


Subject(s)
Drosophila Proteins/metabolism , Endocrine Cells/cytology , Fertility , rab GTP-Binding Proteins/metabolism , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Endocrine Cells/metabolism , Genitalia, Male/cytology , Genitalia, Male/metabolism , Male , Protein Transport , Vacuoles/metabolism , Vacuoles/ultrastructure , rab GTP-Binding Proteins/genetics
17.
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
18.
Sci Rep ; 8(1): 10876, 2018 Jul 18.
Article in English | MEDLINE | ID: mdl-30022126

ABSTRACT

Pancreatic islets of Langerhans display characteristic spatial architecture of their endocrine cell types. This architecture is critical for cell-cell communication and coordinated hormone secretion. Islet architecture is disrupted in type-2 diabetes. Moreover, the generation of architecturally correct islets in vitro remains a challenge in regenerative approaches to type-1 diabetes. Although the characteristic islet architecture is well documented, the mechanisms controlling its formation remain obscure. Here, we report that correct endocrine cell type sorting and the formation of mature islet architecture require the expression of Roundabout (Robo) receptors in ß cells. Mice with whole-body deletion of Robo1 and conditional deletion of Robo2 either in all endocrine cells or selectively in ß cells show complete loss of endocrine cell type sorting, highlighting the importance of ß cells as the primary organizer of islet architecture. Conditional deletion of Robo in mature ß cells subsequent to islet formation results in a similar phenotype. Finally, we provide evidence to suggest that the loss of islet architecture in Robo KO mice is not due to ß cell transdifferentiation, cell death or loss of ß cell differentiation or maturation.


Subject(s)
Cell Differentiation , Endocrine Cells/cytology , Insulin-Secreting Cells/cytology , Islets of Langerhans/physiopathology , Nerve Tissue Proteins/physiology , Receptors, Immunologic/physiology , Animals , Cell Communication , Cell Movement , Endocrine Cells/metabolism , Female , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Male , Mice , Mice, Knockout , Roundabout Proteins
19.
Elife ; 72018 06 19.
Article in English | MEDLINE | ID: mdl-29916364

ABSTRACT

Pancreatic islets are innervated by autonomic and sensory nerves that influence their function. Analyzing the innervation process should provide insight into the nerve-endocrine interactions and their roles in development and disease. Here, using in vivo time-lapse imaging and genetic analyses in zebrafish, we determined the events leading to islet innervation. Comparable neural density in the absence of vasculature indicates that it is dispensable for early pancreatic innervation. Neural crest cells are in close contact with endocrine cells early in development. We find these cells give rise to neurons that extend axons toward the islet as they surprisingly migrate away. Specific ablation of these neurons partly prevents other neurons from migrating away from the islet resulting in diminished innervation. Thus, our studies establish the zebrafish as a model to interrogate mechanisms of organ innervation, and reveal a novel mode of innervation whereby neurons establish connections with their targets before migrating away.


Subject(s)
Endocrine Cells/physiology , Islets of Langerhans/innervation , Nerve Net/physiology , Neural Crest/physiology , Parasympathetic Nervous System/physiology , Synaptic Transmission/physiology , Animals , Animals, Genetically Modified , Biomarkers/metabolism , Cell Communication , Cell Differentiation , Cell Movement , Embryo, Nonmammalian , Endocrine Cells/cytology , Gene Expression , Insulin/genetics , Insulin/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/physiology , Nerve Net/cytology , Neural Crest/cytology , Parasympathetic Nervous System/cytology , Somatostatin/genetics , Somatostatin/metabolism , Tubulin/genetics , Tubulin/metabolism , Zebrafish
20.
PLoS One ; 13(4): e0196029, 2018.
Article in English | MEDLINE | ID: mdl-29684040

ABSTRACT

Pituitary endocrine cells are supplied by Sox2-expressing stem/progenitor cells in the anterior lobe of the adult pituitary. In relation to their microenvironment ("niche"), SOX2-positive cells exist in two types of niches; the marginal cell layer-niche and the parenchymal-niche. Recently, we isolated dense stem/progenitor cell clusters from the parenchymal-niche as parenchymal stem/progenitor cell (PS)-clusters. We classified these PS-clusters into three subtypes based on differences in S100ß-expression (S100ß-positive, -negative, and -mixed type), and reported that S100ß-positive PS-clusters exhibited the capacity for differentiation into endocrine cells under 3-dimensional cultivation system. In the present study, we further characterized S100ß-positive PS-clusters using an in vitro 2-dimensional cultivation system. The results demonstrated that S100ß-positive PS-clusters in the 2-dimensional cultivation system proliferated more actively than S100ß-negative clusters. Moreover, in 2-dimensional cultivation conditions, S100ß-positive PS-clusters showed differentiation capacity into non-endocrine cells (Myogenin-, αSMA-, NG2-, or SOX17-positive cells) but not into endocrine cells, whereas S100ß-negative PS-clusters did not. Collectively, PS-clusters were heterogeneous, exhibiting different proliferation and differentiation properties based on the difference in S100ß-expression. Specifically, a part of SOX2-positive cells in the parenchymal-niche had capacities for differentiation into non-endocrine cells, and S100ß-positive PS-clusters may be in more progressive stages toward differentiation than S100ß-negative clusters.


Subject(s)
Adult Stem Cells/cytology , Cell Culture Techniques/methods , Pituitary Gland, Anterior/cytology , S100 Calcium Binding Protein beta Subunit/metabolism , Adult Stem Cells/metabolism , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Plasticity , Cell Proliferation , Cells, Cultured , Endocrine Cells/cytology , Endocrine Cells/metabolism , Pituitary Gland, Anterior/metabolism , Rats , S100 Calcium Binding Protein beta Subunit/genetics , SOXB1 Transcription Factors/metabolism , Stem Cell Niche
SELECTION OF CITATIONS
SEARCH DETAIL