Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.790
Filter
1.
Mol Med Rep ; 30(1)2024 07.
Article in English | MEDLINE | ID: mdl-38695254

ABSTRACT

As a pluripotent cell, activated pancreatic stellate cells (PSCs) can differentiate into various pancreatic parenchymal cells and participate in the secretion of extracellular matrix and the repair of pancreatic damage. Additionally, PSCs characteristics allow them to contribute to pancreatic inflammation and carcinogenesis. Moreover, a detailed study of the pathogenesis of activated PSCs in pancreatic disease can offer promise for the development of innovative therapeutic strategies and improved patient prognoses. Therefore, the present study review aimed to examine the involvement of activated PSCs in pancreatic diseases and elucidate the underlying mechanisms to provide a viable therapeutic strategy for the management of pancreas­related diseases.


Subject(s)
Pancreas , Pancreatic Diseases , Pancreatic Stellate Cells , Humans , Pancreatic Stellate Cells/metabolism , Pancreatic Stellate Cells/pathology , Pancreas/metabolism , Pancreas/pathology , Pancreas/cytology , Pancreatic Diseases/pathology , Pancreatic Diseases/metabolism , Animals , Extracellular Matrix/metabolism , Cell Differentiation , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism
2.
Nat Commun ; 15(1): 3744, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702321

ABSTRACT

Cellular composition and anatomical organization influence normal and aberrant organ functions. Emerging spatial single-cell proteomic assays such as Image Mass Cytometry (IMC) and Co-Detection by Indexing (CODEX) have facilitated the study of cellular composition and organization by enabling high-throughput measurement of cells and their localization directly in intact tissues. However, annotation of cell types and quantification of their relative localization in tissues remain challenging. To address these unmet needs for atlas-scale datasets like Human Pancreas Analysis Program (HPAP), we develop AnnoSpat (Annotator and Spatial Pattern Finder) that uses neural network and point process algorithms to automatically identify cell types and quantify cell-cell proximity relationships. Our study of data from IMC and CODEX shows the higher performance of AnnoSpat in rapid and accurate annotation of cell types compared to alternative approaches. Moreover, the application of AnnoSpat to type 1 diabetic, non-diabetic autoantibody-positive, and non-diabetic organ donor cohorts recapitulates known islet pathobiology and shows differential dynamics of pancreatic polypeptide (PP) cell abundance and CD8+ T cells infiltration in islets during type 1 diabetes progression.


Subject(s)
Algorithms , Diabetes Mellitus, Type 1 , Pancreas , Proteomics , Humans , Proteomics/methods , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/metabolism , Pancreas/cytology , Pancreas/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/cytology , Single-Cell Analysis/methods , Neural Networks, Computer , CD8-Positive T-Lymphocytes/metabolism , Image Cytometry/methods
3.
Cell Transplant ; 33: 9636897241248942, 2024.
Article in English | MEDLINE | ID: mdl-38712762

ABSTRACT

Recently, we and others generated induced tissue-specific stem/progenitor (iTS/iTP) cells. The advantages of iTS/iTP cells compared with induced pluripotent stem (iPS) cells are (1) easier generation, (2) efficient differentiation, and (3) no teratomas formation. In this study, we generated mouse induced pancreatic stem cells (iTS-P cells) by the plasmid vector expressing Yes-associated protein 1 (YAP). The iTS-P YAP9 cells expressed Foxa2 (endoderm marker) and Pdx1 (pancreatic marker) while the expressions of Oct3/4 and Nanog (marker of embryonic stem [ES] cells) in iTS-P YAP9 cells was significantly lower compared with those in ES cells. The iTS-P YAP9 cells efficiently differentiated into insulin-expressing cells compared with ES cells. The ability to generate autologous iTS cells may be applied to diverse applications of regenerative medicine.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells , YAP-Signaling Proteins , Animals , Mice , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Hepatocyte Nuclear Factor 3-beta/metabolism , Hepatocyte Nuclear Factor 3-beta/genetics , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism , Octamer Transcription Factor-3/genetics , Pancreas/cytology , Pancreas/metabolism , Phosphoproteins/metabolism , Phosphoproteins/genetics , Trans-Activators/metabolism , Trans-Activators/genetics
4.
Nature ; 629(8012): 679-687, 2024 May.
Article in English | MEDLINE | ID: mdl-38693266

ABSTRACT

Pancreatic intraepithelial neoplasias (PanINs) are the most common precursors of pancreatic cancer, but their small size and inaccessibility in humans make them challenging to study1. Critically, the number, dimensions and connectivity of human PanINs remain largely unknown, precluding important insights into early cancer development. Here, we provide a microanatomical survey of human PanINs by analysing 46 large samples of grossly normal human pancreas with a machine-learning pipeline for quantitative 3D histological reconstruction at single-cell resolution. To elucidate genetic relationships between and within PanINs, we developed a workflow in which 3D modelling guides multi-region microdissection and targeted and whole-exome sequencing. From these samples, we calculated a mean burden of 13 PanINs per cm3 and extrapolated that the normal intact adult pancreas harbours hundreds of PanINs, almost all with oncogenic KRAS hotspot mutations. We found that most PanINs originate as independent clones with distinct somatic mutation profiles. Some spatially continuous PanINs were found to contain multiple KRAS mutations; computational and in situ analyses demonstrated that different KRAS mutations localize to distinct cell subpopulations within these neoplasms, indicating their polyclonal origins. The extensive multifocality and genetic heterogeneity of PanINs raises important questions about mechanisms that drive precancer initiation and confer differential progression risk in the human pancreas. This detailed 3D genomic mapping of molecular alterations in human PanINs provides an empirical foundation for early detection and rational interception of pancreatic cancer.


Subject(s)
Genetic Heterogeneity , Genomics , Imaging, Three-Dimensional , Pancreatic Neoplasms , Precancerous Conditions , Single-Cell Analysis , Adult , Female , Humans , Male , Clone Cells/metabolism , Clone Cells/pathology , Exome Sequencing , Machine Learning , Mutation , Pancreas/anatomy & histology , Pancreas/cytology , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Workflow , Disease Progression , Early Detection of Cancer , Oncogenes/genetics
5.
BMC Genomics ; 25(1): 427, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38689254

ABSTRACT

BACKGROUND: Current approaches to profile the single-cell transcriptomics of human pancreatic endocrine cells almost exclusively rely on freshly isolated islets. However, human islets are limited in availability. Furthermore, the extensive processing steps during islet isolation and subsequent single cell dissolution might alter gene expressions. In this work, we report the development of a single-nucleus RNA sequencing (snRNA-seq) approach with targeted islet cell enrichment for endocrine-population focused transcriptomic profiling using frozen archival pancreatic tissues without islet isolation. RESULTS: We cross-compared five nuclei isolation protocols and selected the citric acid method as the best strategy to isolate nuclei with high RNA integrity and low cytoplasmic contamination from frozen archival human pancreata. We innovated fluorescence-activated nuclei sorting based on the positive signal of NKX2-2 antibody to enrich nuclei of the endocrine population from the entire nuclei pool of the pancreas. Our sample preparation procedure generated high-quality single-nucleus gene-expression libraries while preserving the endocrine population diversity. In comparison with single-cell RNA sequencing (scRNA-seq) library generated with live cells from freshly isolated human islets, the snRNA-seq library displayed comparable endocrine cellular composition and cell type signature gene expression. However, between these two types of libraries, differential enrichments of transcripts belonging to different functional classes could be observed. CONCLUSIONS: Our work fills a technological gap and helps to unleash frozen archival pancreatic tissues for molecular profiling targeting the endocrine population. This study opens doors to retrospective mappings of endocrine cell dynamics in pancreatic tissues of complex histopathology. We expect that our protocol is applicable to enrich nuclei for transcriptomics studies from various populations in different types of frozen archival tissues.


Subject(s)
Cell Nucleus , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Islets of Langerhans , Nuclear Proteins , Sequence Analysis, RNA , Single-Cell Analysis , Transcription Factors , Humans , Islets of Langerhans/metabolism , Islets of Langerhans/cytology , Single-Cell Analysis/methods , Sequence Analysis, RNA/methods , Cell Nucleus/genetics , Cell Nucleus/metabolism , Gene Expression Profiling/methods , Pancreas/metabolism , Pancreas/cytology , Transcriptome
6.
Diabetologia ; 67(6): 1066-1078, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38630142

ABSTRACT

AIMS/HYPOTHESIS: Rodent pancreas development has been described in great detail. On the other hand, there are still gaps in our understanding of the developmental trajectories of pancreatic cells during human ontogenesis. Here, our aim was to map the spatial and chronological dynamics of human pancreatic cell differentiation and proliferation by using 3D imaging of cleared human embryonic and fetal pancreases. METHODS: We combined tissue clearing with light-sheet fluorescence imaging in human embryonic and fetal pancreases during the first trimester of pregnancy. In addition, we validated an explant culture system enabling in vitro proliferation of pancreatic progenitors to determine the mitogenic effect of candidate molecules. RESULTS: We detected the first insulin-positive cells as early as five post-conceptional weeks, two weeks earlier than previously observed. We observed few insulin-positive clusters at five post-conceptional weeks (mean ± SD 9.25±5.65) with a sharp increase to 11 post-conceptional weeks (4307±152.34). We identified a central niche as the location of onset of the earliest insulin cell production and detected extra-pancreatic loci within the adjacent developing gut. Conversely, proliferating pancreatic progenitors were located in the periphery of the epithelium, suggesting the existence of two separated pancreatic niches for differentiation and proliferation. Additionally, we observed that the proliferation ratio of progenitors ranged between 20% and 30%, while for insulin-positive cells it was 1%. We next unveiled a mitogenic effect of the platelet-derived growth factor AA isoform (PDGFAA) in progenitors acting through the pancreatic mesenchyme by increasing threefold the number of proliferating progenitors. CONCLUSIONS/INTERPRETATION: This work presents a first 3D atlas of the human developing pancreas, charting both endocrine and proliferating cells across early development.


Subject(s)
Cell Differentiation , Cell Proliferation , Imaging, Three-Dimensional , Pancreas , Humans , Pancreas/embryology , Pancreas/cytology , Pancreas/metabolism , Cell Differentiation/physiology , Female , Stem Cells/cytology , Stem Cells/metabolism , Pregnancy , Insulin/metabolism
7.
Adv Sci (Weinh) ; 10(25): e2207691, 2023 09.
Article in English | MEDLINE | ID: mdl-37409821

ABSTRACT

Mitochondrial function impairment due to abnormal opening of the mitochondrial permeability transition pore (MPTP) is considered the central event in acute pancreatitis; however, therapeutic choices for this condition remain controversial. Mesenchymal stem cells (MSCs) are a family member of stem cells with immunomodulatory and anti-inflammatory capabilities that can mitigate damage in experimental pancreatitis. Here, it is shown that MSCs deliver hypoxia-treated functional mitochondria to damaged pancreatic acinar cells (PACs) via extracellular vesicles (EVs), which reverse the metabolic function of PACs, maintain ATP supply, and exhibit an excellent injury-inhibiting effect. Mechanistically, hypoxia inhibits superoxide accumulation in the mitochondria of MSCs and upregulates the membrane potential, which is internalized into PACs via EVs, thus, remodeling the metabolic state. In addition, cargocytes constructed via stem cell denucleation as mitochondrial vectors are shown to exert similar therapeutic effects to MSCs. These findings reveal an important mechanism underlying the role of mitochondria in MSC therapy and offer the possibility of applying mitochondrial therapy to patients with severe acute pancreatitis.


Subject(s)
Acinar Cells , Mesenchymal Stem Cells , Mitochondria , Pancreas , Pancreatitis , Acinar Cells/cytology , Acinar Cells/metabolism , Acute Disease , Adenosine Triphosphate/metabolism , Bile Acids and Salts/metabolism , Cell Hypoxia , Cellular Reprogramming , Extracellular Vesicles/metabolism , Membrane Potential, Mitochondrial , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mitochondria/metabolism , Mitochondrial Permeability Transition Pore/metabolism , Pancreas/cytology , Pancreas/metabolism , Pancreas/pathology , Pancreatitis/metabolism , Pancreatitis/pathology , Pancreatitis/therapy , Paracrine Communication , Superoxides/metabolism , Umbilical Cord/cytology , Humans
8.
Sci Rep ; 13(1): 11954, 2023 07 24.
Article in English | MEDLINE | ID: mdl-37488151

ABSTRACT

To explore the stability and repeatability of diffusion-weighted imaging (DWI) of normal pancreas with different field of views (FOV) on 5.0 T magnetic resonance imaging (MRI) system. Twenty healthy subjects underwent two sessions of large FOV (lFOV) and reduced FOV (rFOV) DWI sequence scanning. Two radiologists measured the apparent diffusion coefficient (ADC) values and the signal-to-noise ratio (SNR) of the pancreatic head, body, and tail on DWI images, simultaneously, using a 5-point scale, evaluate the artifacts and image quality. One radiologist re-measured the ADC on DWI images again after a 4-week interval. The test-retest repeatability of two scan sessions were also evaluated. Intra-observer and inter-observer at lFOV and rFOV, the ADC values were not significantly different (P > 0.05), intraclass correlation coefficients (ICCs) and coefficient of variations were excellence (ICCs 0.85-0.99, CVs < 8.0%). The ADC values were lower with rFOV than lFOV DWI for the head, body, tail, and overall pancreas. The consistency of the two scan sessions were high. The high stability and repeatability of pancreas DWI has been confirmed at 5.0 T. Scan durations are reduced while resolution and image quality are improved with rFOV DWI, which is more preferable than lFOV for routine pancreas imaging.


Subject(s)
Pancreas , Humans , Male , Female , Adolescent , Young Adult , Adult , Middle Aged , Aged , Pancreas/cytology , Magnetic Resonance Imaging , Artifacts , Reproducibility of Results
9.
Cancer Cytopathol ; 131(4): 234-244, 2023 04.
Article in English | MEDLINE | ID: mdl-36574153

ABSTRACT

BACKGROUND: Autoimmune pancreatitis (AIP) is a known mimicker of pancreatic ductal adenocarcinoma both clinically and radiologically. In this study, the authors present their institutional experience in diagnosing AIP on cytology and correlate results with the histologic findings. METHODS: A 14-year computerized search for patients who had histologically confirmed AIP with concurrent or prior cytology was performed. Clinical data, cytology findings, and surgical pathology results were reviewed for analysis. RESULTS: Eighteen patients were identified. The patients showed a male predominance, with a mean age of 59 years. Jaundice, weight loss, and abdominal pain were the most common clinical presentation. Five of 12 patients who were tested for serum immunoglobulin G4 had elevated levels. Cytologic findings of 16 cases that were available for review showed markedly inflamed fibrous stroma (54%) and cytologic atypia (50%). The final cytologic diagnoses were suspicious for adenocarcinoma (n = 1), atypical (n = 8), and benign/negative (n = 9). The corresponding surgical pathology diagnoses were classified as type 1 (n = 10), type 2 (n = 6), and AIP, not otherwise specified (n = 2). All type 2 AIP cases had at least atypical cytologic diagnoses, with one called suspicious for adenocarcinoma and another called adenocarcinoma at the time of rapid on-site evaluation. In contrast, eight of 10 type 1 AIP cases were negative/benign, and two of 10 were atypical. In these two atypical cases, the possibility of AIP was raised because of the presence of inflamed stroma. CONCLUSION: AIP is a pitfall in cytology because moderate-to-marked atypia can be present, especially in type 2 AIP. Because atypia can be severe, the presence of cellular fibrous stroma with lymphocytic stromal infiltrates and the integration of serum immunoglobulin G4 levels could be helpful in avoiding diagnostic overcall in AIP.


Subject(s)
Autoimmune Pancreatitis , Pancreas , Humans , Autoimmune Pancreatitis/complications , Autoimmune Pancreatitis/diagnosis , Autoimmune Pancreatitis/pathology , Retrospective Studies , Male , Female , Middle Aged , Pancreas/cytology , Adenocarcinoma/complications , Adenocarcinoma/diagnosis
10.
Pancreas ; 51(4): 330-337, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35695761

ABSTRACT

OBJECTIVES: Embryonic stem cells (ESCs)-derived pancreatic precursor cells have great potential for pancreas repair. Expression of pancreatic duodenal homeobox 1 (Pdx1) in definitive endoderm (DE) cells is the premise that DE cells differentiate into pancreatic cells. To achieve the required number of Pdx1-expressing DE cells for cell transplantation therapy, a valid model must be established. Using this model, researchers investigated how Pdx1 regulates ESC differentiation into pancreatic cells. METHODS: Tet-On inducible lentiviral vector encoding Pdx1 or mock vector was transduced into mouse ESC (ES-E14TG2a). The mouse ESCs were divided into 3 groups: control (ESC), mock vector (Pdx1 - -ESC), and vector encoding Pdx1 (Pdx1 + -ESC). All groups were separately cocultured with the DE cells sorted by immune beads containing CXCR-4 + (C-X-C chemokine receptor type-4) antibody. Doxycycline induced the expression of Pdx1 on the Pdx1 + -ESC cells. The markers of cell differentiation and Notch pathway were examined. RESULTS: Significantly increased expression levels of Ptf1a, CK19, and amylase on day (d) 3 and d7, Neuro-D1 on d10 and d14, Pax6 and insulin on d14, as well as Notch1, Notch2, Hes1, and Hes5 on d3 and thereafter declined on d14 were observed in Pdx1 + -ESC group. CONCLUSIONS: Pdx1 + -ESC could differentiate into pancreatic-like cells with involvement of the Notch pathway.


Subject(s)
Endoderm , Homeodomain Proteins , Mouse Embryonic Stem Cells , Pancreas , Trans-Activators , Animals , Cell Differentiation , Endoderm/cytology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Pancreas/cytology , Receptors, Notch/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
11.
Dev Cell ; 57(12): 1496-1511.e6, 2022 06 20.
Article in English | MEDLINE | ID: mdl-35675813

ABSTRACT

Diabetic patients show elevated plasma IL18 concentrations. IL18 has two receptors: the IL18 receptor (IL18r) and the Na-Cl co-transporter (NCC). Here, we report that IL18 is expressed on islet α cells, NCC on ß cells, and IL18r on acinar cells in human and mouse pancreases. The deficiency of these receptors reduces islet size, ß cell proliferation, and insulin secretion but increases ß cell apoptosis and exocrine macrophage accumulation after diet-induced glucose intolerance or streptozotocin-induced hyperglycemia. Together with the glucagon-like peptide-1 (GLP1), IL18 uses the NCC and GLP1 receptors on ß cells to trigger ß cell development and insulin secretion. IL18 also uses the IL18r on acinar cells to block hyperglycemic pancreas macrophage expansion. The ß cell-selective depletion of the NCC or acinar-cell-selective IL18r depletion reduces glucose tolerance and insulin sensitivity with impaired ß cell proliferation, enhanced ß cell apoptosis and macrophage expansion, and inflammation in mouse hyperglycemic pancreas. IL18 uses NCC, GLP1r, and IL18r to maintain islet ß cell function and homeostasis.


Subject(s)
Insulin-Secreting Cells , Interleukin-18 , Pancreas , Animals , Glucagon-Like Peptide 1/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Interleukin-18/metabolism , Mice , Pancreas/cytology , Pancreas/metabolism
12.
Elife ; 112022 01 21.
Article in English | MEDLINE | ID: mdl-35060900

ABSTRACT

Restoring damaged ß-cells in diabetic patients by harnessing the plasticity of other pancreatic cells raises the questions of the efficiency of the process and of the functionality of the new Insulin-expressing cells. To overcome the weak regenerative capacity of mammals, we used regeneration-prone zebrafish to study ß-cells arising following destruction. We show that most new insulin cells differ from the original ß-cells as they coexpress Somatostatin and Insulin. These bihormonal cells are abundant, functional and able to normalize glycemia. Their formation in response to ß-cell destruction is fast, efficient, and age-independent. Bihormonal cells are transcriptionally close to a subset of δ-cells that we identified in control islets and that are characterized by the expression of somatostatin 1.1 (sst1.1) and by genes essential for glucose-induced Insulin secretion in ß-cells such as pdx1, slc2a2 and gck. We observed in vivo the conversion of monohormonal sst1.1-expressing cells to sst1.1+ ins + bihormonal cells following ß-cell destruction. Our findings support the conclusion that sst1.1 δ-cells possess a pro-ß identity enabling them to contribute to the neogenesis of Insulin-producing cells during regeneration. This work unveils that abundant and functional bihormonal cells benefit to diabetes recovery in zebrafish.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Somatostatin-Secreting Cells/metabolism , Animals , Female , Male , Pancreas/cytology , Somatostatin/metabolism , Zebrafish
13.
Gastroenterology ; 162(2): 604-620.e20, 2022 02.
Article in English | MEDLINE | ID: mdl-34695382

ABSTRACT

BACKGROUND & AIMS: Acinar to ductal metaplasia (ADM) occurs in the pancreas in response to tissue injury and is a potential precursor for adenocarcinoma. The goal of these studies was to define the populations arising from ADM, the associated transcriptional changes, and markers of disease progression. METHODS: Acinar cells were lineage-traced with enhanced yellow fluorescent protein (EYFP) to follow their fate post-injury. Transcripts of more than 13,000 EYFP+ cells were determined using single-cell RNA sequencing (scRNA-seq). Developmental trajectories were generated. Data were compared with gastric metaplasia, KrasG12D-induced neoplasia, and human pancreatitis. Results were confirmed by immunostaining and electron microscopy. KrasG12D was expressed in injury-induced ADM using several inducible Cre drivers. Surgical specimens of chronic pancreatitis from 15 patients were evaluated by immunostaining. RESULTS: scRNA-seq of ADM revealed emergence of a mucin/ductal population resembling gastric pyloric metaplasia. Lineage trajectories suggest that some pyloric metaplasia cells can generate tuft and enteroendocrine cells (EECs). Comparison with KrasG12D-induced ADM identifies populations associated with disease progression. Activation of KrasG12D expression in HNF1B+ or POU2F3+ ADM populations leads to neoplastic transformation and formation of MUC5AC+ gastric-pit-like cells. Human pancreatitis samples also harbor pyloric metaplasia with a similar transcriptional phenotype. CONCLUSIONS: Under conditions of chronic injury, acinar cells undergo a pyloric-type metaplasia to mucinous progenitor-like populations, which seed disparate tuft cell and EEC lineages. ADM-derived EEC subtypes are diverse. KrasG12D expression is sufficient to drive neoplasia when targeted to injury-induced ADM populations and offers an alternative origin for tumorigenesis. This program is conserved in human pancreatitis, providing insight into early events in pancreas diseases.


Subject(s)
Acinar Cells/metabolism , Carcinoma, Pancreatic Ductal/genetics , Metaplasia/genetics , Pancreatic Ducts/metabolism , Pancreatic Neoplasms/genetics , Acinar Cells/cytology , Cell Plasticity/genetics , Enteroendocrine Cells/cytology , Enteroendocrine Cells/metabolism , Gene Expression Profiling , Humans , Metaplasia/metabolism , Mucin 5AC/genetics , Pancreas/cytology , Pancreas/metabolism , Pancreatic Ducts/cytology , Pancreatitis/genetics , Pancreatitis/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Single-Cell Analysis
14.
J Nutr Biochem ; 99: 108870, 2022 01.
Article in English | MEDLINE | ID: mdl-34563663

ABSTRACT

Emerging evidence has deemed vitamin D as a potential candidate for the intervention of type 2 diabetes (T2D). Herein, we explored the underlying mechanisms of T2D prevention by vitamin D, concentrating on pancreatic iron deposition reported recently. Zucker diabetic fatty (ZDF) rats were treated by vitamin D, with age-matched Zucker lean rats as control. As expected, vitamin D treatment for ZDF rats normalized islet morphology and ß-cell function. Moreover, vitamin D alleviated iron accumulation and apoptosis in pancreatic cells of ZDF rats, accompanied by lowered divalent metal transporter 1 (DMT1) expression. Consistently, similar results were observed in high glucose-stimulated INS-1 cells treated with or without vitamin D. Nuclear factor-κB (NF-κB), a transcription factor involving DMT1 regulation, was activated in pancreases of ZDF rats and INS-1 cells exposed to high glucose, but inactivated by vitamin D or BAY 11-7082, a NF-κB inhibitor. Futhermore, IL-1ß functioning as NF-κB activator abolished the suppression of NF-κB activation, DMT1 induction and the attenuation of apoptosis as a consequence of vitamin D incubation. Our study showed that iron overload in pancreas may contribute to T2D pathogenesis and uncovered a potentially protective role for vitamin D on iron deposition of diabetic pancreas through NF-κB- DMT1 signaling.


Subject(s)
Cation Transport Proteins/metabolism , Diabetes Mellitus, Type 2/drug therapy , Iron/metabolism , NF-kappa B/metabolism , Pancreas/metabolism , Vitamin D/administration & dosage , Animals , Apoptosis , Cation Transport Proteins/genetics , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Humans , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Male , NF-kappa B/genetics , Pancreas/cytology , Pancreas/drug effects , Rats , Rats, Zucker , Signal Transduction/drug effects
15.
STAR Protoc ; 2(4): 100989, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34927097

ABSTRACT

Preparation of single-cell suspension from primary tumor tissue can provide a valuable resource for functional, genetic, proteomic, and tumor microenvironment studies. Here, we describe an effective protocol for mouse pancreatic tumor dissociation with further processing of tumor suspension for single-cell RNA sequencing analysis of cellular populations. We further provide an outline of the bioinformatics processing of the data and clustering of heterogeneous cellular populations comprising pancreatic tumors using Common Workflow Language (CWL) pipelines within user-friendly Scientific Data Analysis Platform (https://SciDAP.com). For complete details on the use and execution of this protocol, please refer to Gabitova-Cornell et al. (2020).


Subject(s)
Computational Biology/methods , Pancreatic Neoplasms , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Animals , Female , Male , Mice , Pancreas/cytology , Pancreas/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Software
16.
PLoS One ; 16(12): e0260902, 2021.
Article in English | MEDLINE | ID: mdl-34879096

ABSTRACT

In the last few years, there has been a considerable increase in the use of organoids, which is a new three-dimensional culture technology applied in scientific research. The main reasons for their extensive use are their plasticity and multiple applications, including in regenerative medicine and the screening of new drugs. The aim of this study was to better understand these structures by focusing on the choice of the best housekeeping gene (HKG) to perform accurate molecular analysis on such a heterogeneous system. This feature should not be underestimated because the inappropriate use of a HKG can lead to misleading data and incorrect results, especially when the subject of the study is innovative and not totally explored like organoids. We focused our attention on the newly described human pancreatic organoids (hPOs) and compared 12 well-known HKGs (ACTB, B2M, EF1α, GAPDH, GUSB, HPRT, PPIA, RNA18S, RPL13A TBP, UBC and YWHAZ). Four different statistical algorithms (NormFinder, geNorm, BestKeeper and ΔCt) were applied to estimate the expression stability of each HKG, and RefFinder was used to identify the most suitable genes for RT-qPCR data normalization. Our results showed that the intragroup and intergroup comparisons could influence the best choice of the HKG, making clear that the identification of a stable reference gene for accurate and reproducible RT-qPCR data normalization remains a critical issue. In summary, this is the first report on HKGs in human organoids, and this work provides a strong basis to pave the way for further gene analysis in hPOs.


Subject(s)
Genes, Essential , Organoids/metabolism , Pancreas/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction/methods , Real-Time Polymerase Chain Reaction/standards , Algorithms , Humans , Organoids/cytology , Pancreas/cytology , RNA, Messenger/analysis , Reference Standards
17.
Sci China Life Sci ; 64(12): 1998-2029, 2021 12.
Article in English | MEDLINE | ID: mdl-34865207

ABSTRACT

Stem cells are undifferentiated cells capable of self-renewal and differentiation, giving rise to specialized functional cells. Stem cells are of pivotal importance for organ and tissue development, homeostasis, and injury and disease repair. Tissue-specific stem cells are a rare population residing in specific tissues and present powerful potential for regeneration when required. They are usually named based on the resident tissue, such as hematopoietic stem cells and germline stem cells. This review discusses the recent advances in stem cells of various tissues, including neural stem cells, muscle stem cells, liver progenitors, pancreatic islet stem/progenitor cells, intestinal stem cells, and prostate stem cells, and the future perspectives for tissue stem cell research.


Subject(s)
Stem Cells , Animals , Brain/cytology , Forecasting , Humans , Intestines/cytology , Liver/cytology , Liver/physiology , Male , Muscles/cytology , Pancreas/cytology , Prostate/cytology , Regeneration/physiology , Rodentia , Stem Cell Research , Stem Cells/physiology
18.
STAR Protoc ; 2(4): 100935, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34841274

ABSTRACT

Pancreatic cancer is characterized by bi-directional interactions between pancreatic cancer cells and stromal cells including neural cells. The absence of neural cells in pancreatic organoids limits the investigation of cell- cell interaction and tumor innervation. This protocol describes how to generate innervated wild type (WT) and Kras+/LSLG12D Trp53fl/f lp48+/Cre (KPC) murine pancreatic organoids. To specifically investigate neurogenesis, organoids are co-cultured with iPSCs-derived neural crest cells, while co-culture with dorsal root ganglia explants is used for comparing organoids with mature neurons. For complete details on the use and execution of this protocol, please refer to Huch et al. (2013), Boj et al. (2015), and Demir et al. (2014).


Subject(s)
Coculture Techniques/methods , Models, Biological , Organoids , Pancreas/cytology , Pancreatic Neoplasms/pathology , Animals , Cells, Cultured , Mice , Organoids/cytology , Organoids/pathology , Stromal Cells/cytology , Tumor Cells, Cultured/cytology
19.
Pancreas ; 50(7): 942-951, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34643609

ABSTRACT

ABSTRACT: Type 1 diabetes is an autoimmune disease, and its incidence is usually estimated in the range of 5% to 10%. Currently, the administration of exogenous insulin is the standard of care therapy. However, this therapy is not effective in some patients who may develop some chronic complications. Islet transplantation into the liver is another therapy with promising outcomes; however, the long-term efficacy of this therapeutic option is limited to a small number of patients. Because native extracellular matrix (ECM) components provide a suitable microenvironment for islet functions, engineering a 3-dimensional construct that recapitulates the native pancreatic environment could address these obstacles. Many attempts have been conducted to mimic an in vivo microenvironment to increase the survival of islets or islet-like clusters. With the advent of decellularization technology, it is possible to use a native ECM in organ engineering. Pancreatic decellularized bioscaffold provides proper cell-cell and cell-ECM interactions and retains growth factors that are critical in the determination of cell fate within a native organ. This review summarizes the current knowledge of decellularized matrix technology and addresses its possible limitations before use in the clinic.


Subject(s)
Extracellular Matrix/metabolism , Pancreas/metabolism , Tissue Engineering/methods , Tissue Scaffolds , Tumor Microenvironment , Animals , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Islets of Langerhans Transplantation/methods , Pancreas/cytology
20.
Cell Mol Life Sci ; 78(23): 7107-7132, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34613423

ABSTRACT

Over the past few years, extensive efforts have been made to generate in-vitro pancreatic micro-tissue, for disease modeling or cell replacement approaches in pancreatic related diseases such as diabetes mellitus. To obtain these goals, a closer look at the diverse cells participating in pancreatic development is necessary. Five major non-epithelial pancreatic (pN-Epi) cell populations namely, pancreatic endothelium, mesothelium, neural crests, pericytes, and stellate cells exist in pancreas throughout its development, and they are hypothesized to be endogenous inducers of the development. In this review, we discuss different pN-Epi cells migrating to and existing within the pancreas and their diverse effects on pancreatic epithelium during organ development mediated via associated signaling pathways, soluble factors or mechanical cell-cell interactions. In-vivo and in-vitro experiments, with a focus on N-Epi cells' impact on pancreas endocrine development, have also been considered. Pluripotent stem cell technology and multicellular three-dimensional organoids as new approaches to generate pancreatic micro-tissues have also been discussed. Main challenges for reaching a detailed understanding of the role of pN-Epi cells in pancreas development in utilizing for in-vitro recapitulation have been summarized. Finally, various novel and innovative large-scale bioengineering approaches which may help to recapitulate cell-cell interactions and are crucial for generation of large-scale in-vitro multicellular pancreatic micro-tissues, are discussed.


Subject(s)
Cell Communication/physiology , Cell- and Tissue-Based Therapy/methods , Diabetes Mellitus/therapy , Pancreas/growth & development , Tissue Engineering/methods , Cell Differentiation/physiology , Endothelial Cells/metabolism , Endothelium/cytology , Endothelium/metabolism , Humans , Organogenesis/physiology , Organoids/cytology , Pancreas/cytology , Pancreatic Diseases/therapy , Pluripotent Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...