Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Am J Physiol Gastrointest Liver Physiol ; 325(5): G458-G470, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37698169

ABSTRACT

Notch signaling regulates gastrointestinal stem cell proliferation and differentiation yet Notch-regulated transcriptional effectors of gastric epithelial cell differentiation are poorly understood. Here we tested the role of the bHLH transcription factor Achaete-Scute homolog 1 (ASCL1) in gastric epithelial cell differentiation, and its regulation by Notch. Newborn Ascl1 null mice showed a loss of expression of markers of neurogenin-3-dependent enteroendocrine cells, with normal expression of enterochromaffin-like cells, mucous cells, chief cells, and parietal cells. In adult mice, Ascl1 gene expression was observed in the stomach, but not the intestine, with higher expression in antral than corpus epithelium. Lineage tracing in Ascl1-CreERT2; Rosa26-LSL-tdTomato mice revealed single, scattered ASCL1+ cells in the gastric epithelium, demonstrating expression in antral gastrin- and serotonin-producing endocrine cells. ASCL1-expressing endocrine cells persisted for several weeks posttamoxifen labeling with a half-life of approximately 2 months. Lineage tracing in Gastrin-CreERT2 mice demonstrated a similar lifespan for gastrin-producing cells, confirming that gastric endocrine cells are long-lived. Finally, treatment of Ascl1-CreERT2; Rosa26-LSL-tdTomato mice with the pan-Notch inhibitor dibenzazepine increased the number of lineage-labeled cells in the gastric antrum, suggesting that Notch signaling normally inhibits Ascl1 expression. Notch regulation of Ascl1 was also demonstrated in a genetic mouse model of Notch activation, as well as Notch-manipulated antral organoid cultures, thus suggesting that ASCL1 is a key downstream Notch pathway effector promoting endocrine cell differentiation in the gastric epithelium.NEW & NOTEWORTHY Although Notch signaling is known to regulate cellular differentiation in the stomach, downstream effectors are poorly described. Here we demonstrate that the bHLH transcription factor ASCL1 is expressed in endocrine cells in the stomach and is required for formation of neurogenin-3-dependent enteroendocrine cells but not enterochromaffin-like cells. We also demonstrate that Ascl1 expression is inhibited by Notch signaling, suggesting that ASCL1 is a Notch-regulated transcriptional effector directing enteroendocrine cell fate in the mouse stomach.


Subject(s)
Gastrins , Stomach , Animals , Mice , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Enteroendocrine Cells/metabolism , Mice, Knockout
2.
Cell Rep Methods ; 3(7): 100512, 2023 07 24.
Article in English | MEDLINE | ID: mdl-37533652

ABSTRACT

Time-specific modulation of gene expression during differentiation by transcription factors promotes cell diversity. However, estimating their dynamic regulatory activity at the single-cell level and in a high-throughput manner remains challenging. We present FateCompass, an integrative approach that utilizes single-cell transcriptomics data to identify lineage-specific transcription factors throughout differentiation. By combining a probabilistic framework with RNA velocities or differentiation potential, we estimate transition probabilities, while a linear model of gene regulation is employed to compute transcription factor activities. Considering dynamic changes and correlations of expression and activities, FateCompass identifies lineage-specific regulators. Our validation using in silico data and application to pancreatic endocrine cell differentiation datasets highlight both known and potentially novel lineage-specific regulators. Notably, we uncovered undescribed transcription factors of an enterochromaffin-like population during in vitro differentiation toward ß-like cells. FateCompass provides a valuable framework for hypothesis generation, advancing our understanding of the gene regulatory networks driving cell-fate decisions.


Subject(s)
Gene Expression Regulation , Transcription Factors , Transcription Factors/genetics , Cell Differentiation/genetics , Gene Regulatory Networks , Gene Expression Profiling
3.
Dev Biol ; 499: 59-74, 2023 07.
Article in English | MEDLINE | ID: mdl-37172642

ABSTRACT

The molecular links between tissue-level morphogenesis and the differentiation of cell lineages in the pancreas remain elusive despite a decade of studies. We previously showed that in pancreas both processes depend on proper lumenogenesis. The Rab GTPase Rab11 is essential for epithelial lumen formation in vitro, however few studies have addressed its functions in vivo and none have tested its requirement in pancreas. Here, we show that Rab11 is critical for proper pancreas development. Co-deletion of the Rab11 isoforms Rab11A and Rab11B in the developing pancreatic epithelium (Rab11pancDKO) results in ∼50% neonatal lethality and surviving adult Rab11pancDKO mice exhibit defective endocrine function. Loss of both Rab11A and Rab11B in the embryonic pancreas results in morphogenetic defects of the epithelium, including defective lumen formation and lumen interconnection. In contrast to wildtype cells, Rab11pancDKO cells initiate the formation of multiple ectopic lumens, resulting in a failure to coordinate a single apical membrane initiation site (AMIS) between groups of cells. This results in an inability to form ducts with continuous lumens. Here, we show that these defects are due to failures in vesicle trafficking, as apical and junctional components remain trapped within Rab11pancDKO cells. Together, these observations suggest that Rab11 directly regulates epithelial lumen formation and morphogenesis. Our report links intracellular trafficking to organ morphogenesis in vivo and presents a novel framework for decoding pancreatic development.


Subject(s)
Pancreas , rab GTP-Binding Proteins , Mice , Animals , Epithelium/metabolism , Cell Membrane/metabolism , Protein Isoforms/metabolism , Pancreas/metabolism , Morphogenesis , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
5.
Front Cell Dev Biol ; 10: 915206, 2022.
Article in English | MEDLINE | ID: mdl-35874834

ABSTRACT

Pancreatic islet ß cells regulate glucose homeostasis via glucose-stimulated insulin secretion (GSIS). Cytoskeletal polymers microtubules (MTs) serve as tracks for the transport and positioning of secretory insulin granules. MT network in ß cells has unique morphology with several distinct features, which support granule biogenesis (via Golgi-derived MT array), net non-directional transport (via interlocked MT mesh), and control availability of granules at secretion sites (via submembrane MT bundle). The submembrane MT array, which is parallel to the plasma membrane and serves to withdraw excessive granules from the secretion hot spots, is destabilized and fragmented downstream of high glucose stimulation, allowing for regulated secretion. The origin of such an unusual MT network, the features that define its functionality, and metabolic pathways that regulate it are still to a large extent elusive and are a matter of active investigation and debate. Besides the MT network itself, it is important to consider the interplay of molecular motors that drive and fine-tune insulin granule transport. Importantly, activity of kinesin-1, which is the major MT-dependent motor in ß cells, transports insulin granules, and has a capacity to remodel MT network, is also regulated by glucose. We discuss yet unknown potential avenues toward understanding how MT network and motor proteins provide control for secretion in coordination with other GSIS-regulating mechanisms.

6.
Biol Pharm Bull ; 45(8): 1185-1190, 2022.
Article in English | MEDLINE | ID: mdl-35908900

ABSTRACT

Bitter taste receptors (TAS2Rs) are expressed by oral cavity cells in mammals and classically function as sensors for bitter compounds. There are 25 functional isoforms of human TAS2Rs, with individual bitter ligands. Each human TAS2R isoform is distributed in several tissues, such as the airway epithelia and gastrointestinal tract, and plays an important role in physiological functions. However, quantification of each isoform is difficult because of highly homologous sequences between some TAS2R isoforms. Therefore, differentiating the isoforms by their expression levels is suitable for clarifying the tissue-specific effects of bitter compounds. In this study, we developed a real-time quantitative PCR (qPCR) method to determine the expression of each TAS2R isoform. Using plasmid standards harboring each isoform, we confirmed that the current assay can quantify the gene expression of each isoform, with negligible interference from other isoforms. In addition, our methods can successfully discriminate between the mRNA expression of each isoform in human cell lines and tissues. Therefore, this qPCR method can successfully quantify the mRNA level of each TAS2R isoform. This method will contribute to a better understanding of the molecular mechanisms underlying the TAS2R ligand-activated signal transduction.


Subject(s)
Protein Isoforms , Receptors, G-Protein-Coupled , Taste , Animals , Humans , Ligands , Protein Isoforms/genetics , RNA, Messenger , Real-Time Polymerase Chain Reaction , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Transcription, Genetic
7.
Diagn Pathol ; 17(1): 37, 2022 Apr 14.
Article in English | MEDLINE | ID: mdl-35422044

ABSTRACT

The occurrence of endocrine cell micronests in ovarian tumors is rarely reported. To our knowledge, there are only three prior cases reported to date: one occurring in an ovarian mucinous cystadenoma, one in an ovarian mucinous cystadenofibroma, and another in an ovarian mucinous carcinoma with a predominant borderline component. This is a 27-year-old woman that presented with a one-month history of abdominal pain and fullness. Imaging studies revealed a large multiloculated cystic and solid mass measuring 23 cm occupying the majority of the pelvis and abdomen concerning for a primary ovarian malignancy. The patient underwent a right salpingo-oophorectomy with appendectomy. Histologic sections from the ovary showed a multiloculated, cystic and focally solid mass lined by gastrointestinal-type mucinous epithelium with variable degrees of proliferation accounting for greater than 10% of the tumor. In addition to the mucinous epithelial component, there were several foci of bland, monotonous epithelioid cells arranged in solid nests with focal tubular/acinar formation within the fibrous septa and mucinous epithelium. Immunohistochemical studies showed that these cells were positive for cytokeratin, EMA, and synaptophysin, while negative for inhibin. The Ki-67 proliferation index was low (<1%). The presence of endocrine cell nests associated with an ovarian mucinous neoplasm is a rare phenomenon. Whether this represents preservation of endocrine cells in the context of epithelial degeneration or an independent neoplastic component is unclear. Progression related to this endocrine cell proliferation is unlikely and the recognition of this phenomenon holds more diagnostic value than prognostic significance, as it could be confused with microinvasion or sex cord stromal elements.


Subject(s)
Cystadenoma, Mucinous , Endocrine Cells , Neoplasms, Cystic, Mucinous, and Serous , Ovarian Neoplasms , Adult , Cystadenoma, Mucinous/diagnosis , Cystadenoma, Mucinous/pathology , Endocrine Cells/pathology , Female , Humans , Neoplasms, Cystic, Mucinous, and Serous/diagnosis , Neoplasms, Cystic, Mucinous, and Serous/surgery , Ovarian Neoplasms/pathology
8.
Case Rep Gastroenterol ; 16(1): 37-43, 2022.
Article in English | MEDLINE | ID: mdl-35350675

ABSTRACT

Case Report: A 65-year-old man without any symptoms received colonoscopy for cancer screening and underwent cold snare polypectomy (CSP) for a 3-mm rectal lesion at a local clinic. A histopathological examination revealed neuroendocrine tumor (NET) G1 with a positive margin. The patient was referred to our hospital for further treatment. Then, the post-CSP scar was removed by endoscopic submucosal dissection (ESD), with a sufficient endoscopically normal margin. Histopathology showed 4 NETs and endocrine cell micronests (ECMs) distant from the post-CSP scar, with a positive lateral margin. We considered that the possibility of other NETs was high. Additional surgery was performed. After a histopathological examination, 11 NETs and ECMs were found in the whole rectum, without lymph node metastasis. The patient had no recurrence at 24 months after surgery. In the past 10 years, we have experienced 4 cases (including the present case) of multiple rectal NETs among 56 cases of rectal NETs of ≤10 mm (7.1%). None of our 4 cases showed any recurrence (follow-up period: 12-32 months). Conclusions: We herein report a case involving a patient with 15 rectal NETs and ECMs. We reviewed our experience with multiple rectal NETs, and the rate of multiple rectal NETs was 7.1%. Endoscopists should consider that multiple lesions may be present in cases of rectal NET and be aware that some cannot be detected endoscopically.

9.
Acta Physiol (Oxf) ; 234(2): e13773, 2022 02.
Article in English | MEDLINE | ID: mdl-34985199

ABSTRACT

AIMS: The mammalian gut is the largest endocrine organ. Dozens of hormones secreted by enteroendocrine cells regulate a variety of physiological functions of the gut but also of the pancreas and brain. Here, we examined the role of the helix-loop-helix transcription factor ID2 during the differentiation of intestinal stem cells along the enteroendocrine lineage. METHODS: To assess the functions of ID2 in the adult mouse small intestine, we used single-cell RNA sequencing, genetically modified mice, and organoid assays. RESULTS: We found that in the adult intestinal epithelium Id2 is predominantly expressed in enterochromaffin and peptidergic enteroendocrine cells. Consistently, the loss of Id2 leads to the reduction of Chromogranin A-positive enteroendocrine cells. In contrast, the numbers of tuft cells are increased in Id2 mutant small intestine. Moreover, ablation of Id2 elevates the numbers of Serotonin+ enterochromaffin cells and Ghrelin+ X-cells in the posterior part of the small intestine. Finally, ID2 acts downstream of BMP signalling during the differentiation of Glucagon-like peptide-1+ L-cells and Cholecystokinin+ I-cells towards Neurotensin+ PYY+ N-cells. CONCLUSION: ID2 plays an important role in cell fate decisions in the adult small intestine. First, ID2 is essential for establishing a differentiation gradient for enterochromaffin and X-cells along the anterior-posterior axis of the gut. Next, ID2 is necessary for the differentiation of N-cells thus ensuring a differentiation gradient along the crypt-villi axis. Finally, ID2 suppresses the commitment of secretory intestinal epithelial progenitors towards tuft cell lineage and thus controls host immune response to commensal and parasitic microbiota.


Subject(s)
Cell Differentiation , Enteroendocrine Cells , Inhibitor of Differentiation Protein 2/genetics , Transcription Factors , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Intestinal Mucosa , Intestine, Small/cytology , Mammals , Mice , Transcription Factors/genetics
10.
Int J Mol Sci ; 22(16)2021 Aug 09.
Article in English | MEDLINE | ID: mdl-34445257

ABSTRACT

The production of pancreatic ß cells is the most challenging step for curing diabetes using next-generation treatments. Adult pancreatic endocrine cells are thought to be maintained by the self-duplication of differentiated cells, and pancreatic endocrine neogenesis can only be observed when the tissue is severely damaged. Experimentally, this can be performed using a method named partial duct ligation (PDL). As the success rate of PDL surgery is low because of difficulties in identifying the pancreatic duct, we previously proposed a method for fluorescently labeling the duct in live animals. Using this method, we performed PDL on neurogenin3 (Ngn3)-GFP transgenic mice to determine the origin of endocrine precursor cells and evaluate their potential to differentiate into multiple cell types. Ngn3-activated cells, which were marked with GFP, appeared after PDL operation. Because some GFP-positive cells were aligned proximally to the duct, we hypothesized that Ngn3-positive cells arise from the pancreatic duct. Therefore, we next developed an in vitro pancreatic duct culture system using Ngn3-GFP mice and examined whether Ngn3-positive cells emerge from this duct. We observed GFP expressions in ductal organoid cultures. GFP expressions were correlated with Ngn3 expressions and endocrine cell lineage markers. Interestingly, tuft cell markers were also correlated with GFP expressions. Our results demonstrate that in adult mice, Ngn3-positive endocrine precursor cells arise from the pancreatic ducts both in vivo and in vitro experiments indicating that the pancreatic duct could be a potential donor for therapeutic use.


Subject(s)
Antigens, Differentiation/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Insulin-Secreting Cells/metabolism , Nerve Tissue Proteins/metabolism , Pancreatic Ducts/metabolism , Stem Cells/metabolism , Animals , Antigens, Differentiation/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Insulin-Secreting Cells/cytology , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Organoids/cytology , Organoids/metabolism , Pancreatic Ducts/cytology , Stem Cells/cytology
11.
Front Physiol ; 12: 562817, 2021.
Article in English | MEDLINE | ID: mdl-34267669

ABSTRACT

It is well-established that anterior pituitary contains multiple endocrine cell populations, and each of them can secrete one/two hormone(s) to regulate vital physiological processes of vertebrates. However, the gene expression profiles of each pituitary cell population remains poorly characterized in most vertebrate groups. Here we analyzed the transcriptome of each cell population in adult chicken anterior pituitaries using single-cell RNA sequencing technology. The results showed that: (1) four out of five known endocrine cell clusters have been identified and designated as the lactotrophs, thyrotrophs, corticotrophs, and gonadotrophs, respectively. Somatotrophs were not analyzed in the current study. Each cell cluster can express at least one known endocrine hormone, and novel marker genes (e.g., CD24 and HSPB1 in lactotrophs, NPBWR2 and NDRG1 in corticotrophs; DIO2 and SOUL in thyrotrophs, C5H11ORF96 and HPGDS in gonadotrophs) are identified. Interestingly, gonadotrophs were shown to abundantly express five peptide hormones: FSH, LH, GRP, CART and RLN3; (2) four non-endocrine/secretory cell types, including endothelial cells (expressing IGFBP7 and CFD) and folliculo-stellate cells (FS-cells, expressing S100A6 and S100A10), were identified in chicken anterior pituitaries. Among them, FS-cells can express many growth factors, peptides (e.g., WNT5A, HBEGF, Activins, VEGFC, NPY, and BMP4), and progenitor/stem cell-associated genes (e.g., Notch signaling components, CDH1), implying that the FS-cell cluster may act as a paracrine/autocrine signaling center and enrich pituitary progenitor/stem cells; (3) sexually dimorphic expression of many genes were identified in most cell clusters, including gonadotrophs and lactotrophs. Taken together, our data provides a bird's-eye view on the diverse aspects of anterior pituitaries, including cell composition, heterogeneity, cell-to-cell communication, and gene expression profiles, which facilitates our comprehensive understanding of vertebrate pituitary biology.

12.
Diabetologia ; 64(10): 2279-2291, 2021 10.
Article in English | MEDLINE | ID: mdl-34274990

ABSTRACT

AIMS/HYPOTHESIS: Normal cellular prion protein (PrPC) is a conserved mammalian glycoprotein found on the outer plasma membrane leaflet through a glycophosphatidylinositol anchor. Although PrPC is expressed by a wide range of tissues throughout the body, the complete repertoire of its functions has not been fully determined. The misfolded pathogenic isoform PrPSc (the scrapie form of PrP) is a causative agent of neurodegenerative prion diseases. The aim of this study is to evaluate PrPC localisation, expression and trafficking in pancreases from organ donors with and without type 1 diabetes and to infer PrPC function through studies on interacting protein partners. METHODS: In order to evaluate localisation and trafficking of PrPC in the human pancreas, 12 non-diabetic, 12 type 1 diabetic and 12 autoantibody-positive organ donor tissue samples were analysed using immunofluorescence analysis. Furthermore, total RNA was isolated from 29 non-diabetic, 29 type 1 diabetic and 24 autoantibody-positive donors to estimate PrPC expression in the human pancreas. Additionally, we performed PrPC-specific immunoblot analysis on total pancreatic protein from non-diabetic and type 1 diabetic organ donors to test whether changes in PrPC mRNA levels leads to a concomitant increase in PrPC protein levels in human pancreases. RESULTS: In non-diabetic and type 1 diabetic pancreases (the latter displaying both insulin-positive [INS(+)] and -negative [INS(-)] islets), we found PrPC in islets co-registering with beta cells in all INS(+) islets and, strikingly, unexpected activation of PrPC in alpha cells within diabetic INS(-) islets. We found PrPC localised to the plasma membrane and endoplasmic reticulum (ER) but not the Golgi, defining two cellular pools and an unconventional protein trafficking mechanism bypassing the Golgi. We demonstrate PrPC co-registration with established protein partners, neural cell adhesion molecule 1 (NCAM1) and stress-inducible phosphoprotein 1 (STI1; encoded by STIP1) on the plasma membrane and ER, respectively, linking PrPC function with cyto-protection, signalling, differentiation and morphogenesis. We demonstrate that both PRNP (encoding PrPC) and STIP1 gene expression are dramatically altered in type 1 diabetic and autoantibody-positive pancreases. CONCLUSIONS/INTERPRETATION: As the first study to address PrPC expression in non-diabetic and type 1 diabetic human pancreas, we provide new insights for PrPC in the pathogenesis of type 1 diabetes. We evaluated the cell-type specific expression of PrPC in the human pancreas and discovered possible connections with potential interacting proteins that we speculate might address mechanisms relevant to the role of PrPC in the human pancreas.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Pancreas/metabolism , PrPC Proteins/metabolism , Adolescent , Adult , Autoantibodies/blood , CD56 Antigen/metabolism , Cell Membrane/metabolism , Child , Endoplasmic Reticulum/metabolism , Female , Gene Expression Regulation/physiology , Heat-Shock Proteins/metabolism , Humans , Immunohistochemistry , Insulin Antibodies/immunology , Male , PrPC Proteins/genetics , Prion Proteins/genetics , Prion Proteins/metabolism , Protein Transport , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Tissue Donors , Young Adult
13.
J Histotechnol ; 44(1): 2-11, 2021 03.
Article in English | MEDLINE | ID: mdl-32909928

ABSTRACT

The research on hatching ecology of the Chinese softshell turtle Trionyx sinensis has essential guiding roles to clarify the physiological and ecological mechanism of reptile evolution. The aim of this study is to describe the histological changes, differentiation, and maturation of some functional cells during the genesis and development of the liver and pancreas of the Chinese softshell turtle T. sinensis. Softshell turtle eggs were incubated under artificial conditions and hatched within 41-45 days. Hematoxylin and eosin-stained embryonic pancreas and liver were examined at various time points from 2 to 31 days and compared with that of other reptiles, amphibians, fishes, and birds in the literature. Immunohistochemical assay for glucagon and insulin was performed on paraformaldehyde-fixed embryos to identify functional cells in the pancreas. Pancreatic endocrine cells of T. sinensis have secretory ability at day 26 of embryonic development, and the dispersed pancreatic endocrine cells may be the result of the incomplete pancreatic development.


Subject(s)
Liver/embryology , Pancreas/embryology , Turtles , Animals , China , Embryonic Development , Pancreatic Hormones
14.
Vet World ; 13(9): 1737-1742, 2020 Sep.
Article in English | MEDLINE | ID: mdl-33132583

ABSTRACT

AIM: The aim of the study was to identify the distribution pattern and frequency of endocrine cell types in the digestive tract of Varanus salvator. MATERIALS AND METHODS: The presence of endocrine cells (glucagon, somatostatin, and serotonin) in the digestive tract (esophagus, stomach, and intestine) was detected using the avidin-biotin complex (ABC) method. RESULTS: Three types of endocrine cells immunoreactive to antisera glucagon, serotonin, and somatostatin were found in the caudal portion of the small and large intestines but were not observed in the esophagus, stomach, and caput and medial sections of the small intestine. Endocrine cells distributed in the digestive tract of V. salvator vary in color intensity, from weak to sharp, in response to the primer antibody. CONCLUSION: Endocrine cells in the digestive tract that is immunoreactive to glucagon, somatostatin, and serotonin are those found in the caudal portion of the small and large intestines. They are varied in distribution pattern, frequency, and color intensity.

15.
Intern Med ; 59(5): 619-623, 2020 Mar 01.
Article in English | MEDLINE | ID: mdl-31666465

ABSTRACT

Although a few reports of neuroendocrine tumor (NET) in the stomach or appendix with surrounding micronests have been published, cases of rectal NET are rare. We herein report a unique case of a patient with single rectal NET treated endoscopically. A pathological examination revealed multiple endocrine cell micronests (ECMs) in the submucosal layer around the main NET lesion. Neither lymph node metastasis nor distant metastasis in computed tomography was observed six years after the treatment. Because case reports of multiple ECM are very rare, the significance of malignancy is unclear. It therefore appears to be necessary to accumulate similar cases.


Subject(s)
Neuroendocrine Tumors/pathology , Rectal Neoplasms/pathology , Endocrine Cells/pathology , Humans , Male , Middle Aged , Neuroendocrine Tumors/surgery , Rectal Neoplasms/surgery , Tomography, X-Ray Computed
16.
Insect Biochem Mol Biol ; 116: 103257, 2020 01.
Article in English | MEDLINE | ID: mdl-31678581

ABSTRACT

Peptides modulate physiological/behavioral control systems in all animals. In arthropods, midgut epithelial endocrine cells are one of the largest sources of these signaling agents. At present, little is known about the identity of the peptides that form arthropod midgut enteroendocrine peptidomes. While many techniques can be used for peptide structural identification, in silico transcriptome mining is one that has been used extensively for arthropod neuropeptidome prediction; this strategy has yet to be used for large-scale arthropod enteroendocrine peptide discovery. Here, a tissue-specific transcriptome was used to assess putative enteroendocrine peptide complement in the honey bee, Apis mellifera, midgut. Searches for transcripts encoding members of 42 peptide families were conducted, with evidence of expression for 15 groups found in the assembly: adipokinetic hormone, allatostatin A, allatostatin C, bursicon, CCHamide, CNMamide, diuretic hormone 31, diuretic hormone 44, insulin-like peptide, myosuppressin, neuropeptide F, pigment dispersing hormone, pyrokinin, short neuropeptide F, and tachykinin-related peptide. The proteins deduced from the midgut transcripts are identical in sequence, or nearly so, to those of Apis pre/preprohormones deposited previously into NCBI, providing increased confidence in the accuracy of the reported data. Seventy-five peptides were predicted from the deduced precursor proteins, 26 being members of known peptide families. Comparisons to previously published mass spectrometric data support the existence of many of the predicted Apis peptides. This study is the first prediction of an arthropod midgut peptidome using transcriptomics, and provides a powerful new resource for investigating enteroendocrine peptide signaling within/from the Apis midgut, a species of significant ecological/economic importance.


Subject(s)
Bees/genetics , Insect Proteins/genetics , Peptides/genetics , Transcriptome , Amino Acid Sequence , Animals , Bees/metabolism , Gastrointestinal Tract , Insect Proteins/chemistry , Insect Proteins/metabolism , Multigene Family , Peptides/chemistry , Peptides/metabolism , Sequence Alignment
17.
Cell Tissue Res ; 377(3): 505-525, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31359140

ABSTRACT

The insect midgut epithelium represents an interface between the internal and the external environment and it is the almost unique epithelial tissue by which these arthropods acquire nutrients. This epithelium is indeed able to produce digestive enzymes and to support vectorial transport of small organic nutrients, ions, and water. Moreover, it plays a key role in the defense against pathogenic microorganisms and in shaping gut microbiota. Another important midgut function is the ability to produce signaling molecules that regulate its own physiology and the activity of other organs. The two main mature cell types present in the midgut of all insects, i.e., columnar and endocrine cells, are responsible for these functions. In addition, stem cells, located at the base of the midgut epithelium, ensure the growth and renewal of the midgut during development and after injury. In insects belonging to specific orders, midgut physiology is deeply conditioned by the presence of unique cell types, i.e., goblet and copper cells, which confer peculiar features to this organ. This review reports current knowledge on the cells that form the insect midgut epithelium, focusing attention on their morphological and functional features. Notwithstanding the apparent structural simplicity of this organ, the properties of the cells make the midgut a key player in insect development and homeostasis.


Subject(s)
Digestive System/ultrastructure , Endoderm/ultrastructure , Insecta/anatomy & histology , Animals
18.
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
19.
Int J Mol Sci ; 20(8)2019 Apr 16.
Article in English | MEDLINE | ID: mdl-31014006

ABSTRACT

Pancreatic endocrine cells expressing the ghrelin gene and producing the ghrelin hormone were first identified in 2002. These cells, named ε cells, were recognized as the fifth type of endocrine cells. Differentiation of ε cells is induced by various transcription factors, including Nk2 homeobox 2, paired box proteins Pax-4 and Pax6, and the aristaless-related homeobox. Ghrelin is generally considered to be a "hunger hormone" that stimulates the appetite and is produced mainly by the stomach. Although the population of ε cells is small in adults, they play important roles in regulating other endocrine cells, especially ß cells, by releasing ghrelin. However, the roles of ghrelin in ß cells are complex. Ghrelin contributes to increased blood glucose levels by suppressing insulin release from ß cells and is also involved in the growth and proliferation of ß cells and the prevention of ß cell apoptosis. Despite increasing evidence and clarification of the mechanisms of ε cells over the last 20 years, many questions remain to be answered. In this review, we present the current evidence for the participation of ε cells in differentiation and clarify their characteristics by focusing on the roles of ghrelin.


Subject(s)
Islets of Langerhans/metabolism , Animals , Ghrelin/genetics , Ghrelin/metabolism , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Insulin/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/growth & development , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
20.
Paediatr Respir Rev ; 30: 65-71, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30552058

ABSTRACT

Interstitial lung diseases in children (chILD) are rare and diverse. The current classifications include a group of early onset chILD specific to infancy, namely neuro-endocrine cell hyperplasia of infancy (NEHI), pulmonary interstitial glycogenosis (PIG) and the alveolar capillary-congenital acinar dysplasia (ACD-CAD) spectrum, as well as alveolar growth disorders. NEHI and PIG cells are seen in the normal developing foetal lung. We hypothesise that these conditions are in fact overlapping manifestations of pulmonary dysmaturity, respectively of airway, mesenchymal and vascular elements, rather than discrete clinical conditions in their own right. Clinically, these present as respiratory distress in early life. Mild cases rightly never undergo lung biopsy, and for these the clinical description 'persistent tachypnoea of infancy' has been proposed. In terms of pathology, we reviewed current literature, which showed that NEHI cells decline with age, and are not specific to NEHI, which we confirmed by unpublished re-analysis of a second dataset. Furthermore, specific genetic disorders which affect pulmonary maturation lead to a histological picture indistinguishable from NEHI. PIG and ACD-CAD are also associated with pulmonary growth disorders, and manifestations of PIG and NEHI may be present in the same child. We conclude that, contrary to current classifications, NEHI, PIG, and ACD-CAD should be considered as overlapping manifestations of pulmonary dysmaturation, frequently associated with disorders of alveolar growth, rather than as separate conditions. Identification of one of these patterns should be the start, not the end of the diagnostic journey, and underlying in particular genetic causes should be sought.


Subject(s)
Infant, Premature, Diseases/physiopathology , Lung Diseases, Interstitial/physiopathology , Child , Child, Preschool , Fetal Organ Maturity , Humans , Hyperplasia , Infant , Infant, Newborn , Infant, Premature, Diseases/diagnostic imaging , Infant, Premature, Diseases/pathology , Lung , Lung Diseases, Interstitial/diagnostic imaging , Lung Diseases, Interstitial/pathology , Neuroendocrine Cells/pathology , Persistent Fetal Circulation Syndrome/diagnostic imaging , Persistent Fetal Circulation Syndrome/pathology , Persistent Fetal Circulation Syndrome/physiopathology , Pulmonary Alveoli/abnormalities , Pulmonary Alveoli/diagnostic imaging , Pulmonary Alveoli/pathology , Pulmonary Alveoli/physiopathology , Tomography, X-Ray Computed
SELECTION OF CITATIONS
SEARCH DETAIL
...