Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Biomed Pharmacother ; 173: 116292, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38394848

ABSTRACT

Single-cell sequencing is a novel and rapidly advancing high-throughput technique that can be used to investigating genomics, transcriptomics, and epigenetics at a single-cell level. Currently, single-cell sequencing can not only be used to draw the pancreatic islet cells map and uncover the characteristics of cellular heterogeneity in type 2 diabetes, but can also be used to label and purify functional beta cells in pancreatic stem cells, improving stem cells and islet organoids therapies. In addition, this technology helps to analyze islet cell dedifferentiation and can be applied to the treatment of type 2 diabetes. In this review, we summarize the development and process of single-cell sequencing, describe the potential applications of single-cell sequencing in the field of type 2 diabetes, and discuss the prospects and limitations of single-cell sequencing to provide a new direction for exploring the pathogenesis of type 2 diabetes and finding therapeutic targets.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Islets of Langerhans , Humans , Diabetes Mellitus, Type 2/metabolism , Islets of Langerhans/metabolism , Pancreas/metabolism , Insulin-Secreting Cells/metabolism , Gene Expression Profiling
2.
Matrix Biol ; 121: 194-216, 2023 08.
Article in English | MEDLINE | ID: mdl-37402431

ABSTRACT

Fibrolamellar carcinomas (FLCs), lethal tumors occurring in children to young adults, have genetic signatures implicating derivation from biliary tree stem cell (BTSC) subpopulations, co-hepato/pancreatic stem cells, involved in hepatic and pancreatic regeneration. FLCs and BTSCs express pluripotency genes, endodermal transcription factors, and stem cell surface, cytoplasmic and proliferation biomarkers. The FLC-PDX model, FLC-TD-2010, is driven ex vivo to express pancreatic acinar traits, hypothesized responsible for this model's propensity for enzymatic degradation of cultures. A stable ex vivo model of FLC-TD-2010 was achieved using organoids in serum-free Kubota's Medium (KM) supplemented with 0.1% hyaluronans (KM/HA). Heparins (10 ng/ml) caused slow expansion of organoids with doubling times of ∼7-9 days. Spheroids, organoids depleted of mesenchymal cells, survived indefinitely in KM/HA in a state of growth arrest for more than 2 months. Expansion was restored with FLCs co-cultured with mesenchymal cell precursors in a ratio of 3:7, implicating paracrine signaling. Signals identified included FGFs, VEGFs, EGFs, Wnts, and others, produced by associated stellate and endothelial cell precursors. Fifty-three, unique heparan sulfate (HS) oligosaccharides were synthesized, assessed for formation of high affinity complexes with paracrine signals, and each complex screened for biological activity(ies) on organoids. Ten distinct HS-oligosaccharides, all 10-12 mers or larger, and in specific paracrine signal complexes elicited particular biological responses. Of note, complexes of paracrine signals and 3-O sulfated HS-oligosaccharides elicited slowed growth, and with Wnt3a, elicited growth arrest of organoids for months. If future efforts are used to prepare HS-oligosaccharides resistant to breakdown in vivo, then [paracrine signal-HS-oligosaccharide] complexes are potential therapeutic agents for clinical treatments of FLCs, an exciting prospect for a deadly disease.


Subject(s)
Carcinoma , Sulfates , Child , Humans , Paracrine Communication , Heparitin Sulfate/metabolism , Oligosaccharides/pharmacology , Oligosaccharides/metabolism
3.
Cancers (Basel) ; 13(13)2021 Jun 30.
Article in English | MEDLINE | ID: mdl-34209288

ABSTRACT

Cell generation and renewal are essential processes to develop, maintain, and regenerate tissues. New cells can be generated from immature cell types, such as stem-like cells, or originate from more differentiated pre-existing cells that self-renew or transdifferentiate. The adult pancreas is a dormant organ with limited regeneration capacity, which complicates studying these processes. As a result, there is still discussion about the existence of stem cells in the adult pancreas. Interestingly, in contrast to the classical stem cell concept, stem cell properties seem to be plastic, and, in circumstances of injury, differentiated cells can revert back to a more immature cellular state. Importantly, deregulation of the balance between cellular proliferation and differentiation can lead to disease initiation, in particular to cancer formation. Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease with a 5-year survival rate of only ~9%. Unfortunately, metastasis formation often occurs prior to diagnosis, and most tumors are resistant to current treatment strategies. It has been proposed that a specific subpopulation of cells, i.e., cancer stem cells (CSCs), are responsible for tumor expansion, metastasis formation, and therapy resistance. Understanding the underlying mechanisms of pancreatic stem cells during homeostasis and injury might lead to new insights to understand the role of CSCs in PDAC. Therefore, in this review, we present an overview of the current literature regarding the stem cell dynamics in the pancreas during health and disease. Furthermore, we highlight the influence of the tumor microenvironment on the growth behavior of PDAC.

4.
J Steroid Biochem Mol Biol ; 198: 105537, 2020 04.
Article in English | MEDLINE | ID: mdl-31785377

ABSTRACT

Porcine pancreatic stem cells (pPSCs) can be induced to insulin-secreting cells and therefore considered the most promising seeding cells for curing human diabetes in future. However, insufficient pPSCs number is one of the bottleneck problems before its clinical application. SerpinB1 is a serine protease inhibitor in neutrophils and can directly promote the proliferation of ß cells. Whether SerpinB1 is involved in pPSC proliferation and differentiation remains unknown. The effects of SerpinB1 on pPSCs proliferation were measured by Cell Counting Kit-8, 5-ethynyl-2'-deoxyuridine, qRT-PCR, western blot, and flow cytometry assays. We found that pPSCs did not efficiently reach the S phase when SerpinB1 expression was knocked down with short hairpin RNA (sh-SerpinB1), the expression of Cyclin D1, CDK-2, and PCNA also decreased. Meanwhile, cell viability and proliferation ability were both declined. Further analyses showed that the expression level of phosphorylated STAT3/STAT3was downregulated, along with an upregulation of p53 and p21. We used a two-step induction method to induce pPSCs to insulin-secreting cells and found that SerpinB1 expression in insulin-secreting cells was higher than in pPSCs. Meanwhile, the protein expression level of phosphorylated STAT3/STAT3 was increased while p53 and p21 was decreased in induced insulin-secreting cells in comparison with control cells. The insulin-secreting cells derived from the sh-SerpinB1 cells secreted less insulin and showed poor sensitivity to high glucose than control group. However, the insulin-secreting cells derived from the ov-SerpinB1 cells has a quite contrary tendency. In conclusion, this study demonstrates that SerpinB1 promotes the proliferation of pPSCs through the STAT3 signaling pathway, and SerpinB1 is a key factor for maintaining the viability of pPSCs during the transition to insulin-secreting cells.


Subject(s)
Adult Stem Cells/cytology , Adult Stem Cells/drug effects , Pancreas/cytology , Pancreas/drug effects , STAT3 Transcription Factor/metabolism , Serpins/physiology , Animals , Cell Culture Techniques , Cell Differentiation , Cell Line , Cell Proliferation , Cyclin D1/metabolism , Cyclin-Dependent Kinase 2/metabolism , Disease Progression , Inflammation , Insulin/metabolism , Insulin-Secreting Cells/cytology , Phosphorylation , Proliferating Cell Nuclear Antigen/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , Swine , Tumor Suppressor Protein p53/metabolism
5.
Int J Mol Sci ; 20(19)2019 Oct 03.
Article in English | MEDLINE | ID: mdl-31623299

ABSTRACT

Pancreatic organogenesis is a multistep process that requires the cooperation of several signaling pathways. In this context, the role of pancreatic mesenchyme is important to define the epithelium development; nevertheless, the precise space-temporal signaling activation still needs to be clarified. This study reports a dissection of the pancreatic embryogenesis, highlighting the molecular network surrounding the epithelium-mesenchyme interaction. To investigate this crosstalk, pancreatic epithelium and surrounding mesenchyme, at embryonic day 10.5, were collected through laser capture microdissection (LCM) and characterized based on their global gene expression. We performed a bioinformatic analysis to hypothesize crosstalk interactions, validating the most promising genes and verifying the precise localization of their expression in the compartments, by RNA in situ hybridization (ISH). Our analyses pointed out also the c-Met gene, a very well-known factor involved in stimulating motility, morphogenesis, and organ regeneration. We also highlighted the potential crosstalk between Versican (Vcan) and Syndecan4 (Sdc4) since these genes are involved in pancreatic tissue repair, strengthening the concept that the same signaling pathways required during pancreatic embryogenesis are also involved in tissue repair. This finding leads to novel strategies for obtaining functional pancreatic stem cells for cell replacement therapies.


Subject(s)
Gene Expression Regulation, Developmental , Mesoderm/embryology , Organogenesis , Pancreas/embryology , Pancreas/metabolism , Signal Transduction , Animals , Computational Biology/methods , Embryonic Development , Gene Expression Profiling , Mice
6.
In Vitro Cell Dev Biol Anim ; 55(6): 453-461, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31140102

ABSTRACT

Although bone marrow-derived mesenchymal stem cells (BMSCs) have been reported to be effective for the attenuation of diabetes, they have limitations. Whether BMSCs can be target-induced by pancreatic stem cells (PSCs) to have effectiveness for the restoration of diabetic islet injury was unknown. In this study, based on their successful isolation and cultivation, BMSCs were co-cultured with PSCs. The pancreatic stem cells markers, Nestin and Neurogenin3 in co-cultured BMSCs were detected to evaluate the target-induction effects. After the diabetic rats were intravenously injected with the target-induced BMSCs, general indicators and islet morphology were detected. The islet insulin generation, and serum insulin and C-peptide contents were measured. It was found that after co-culture, the mRNA expressions, protein contents and distributions of Nestin and Neurogenin3, were dramatically high in BMSCs, indicating that they were successfully target-induced to pancreatic stem-like cells. Furthermore, the target-induced BMSCs had beneficial effects on serum glycated albumin levels and glycogen contents as well as islet morphology of the diabetic rats. Besides elevation of islet insulin generation, the target-induced BMSCs had significant effect on serum insulin and C-peptide contents. In conclusion, BMSCs could be target-induced by PSCs to have effectiveness on the pancreatic restoration of diabetic rats.


Subject(s)
Diabetes Mellitus, Experimental/therapy , Islets of Langerhans/pathology , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/physiology , Pancreas/cytology , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Marrow Cells/cytology , C-Peptide/metabolism , Coculture Techniques , Diabetes Mellitus, Experimental/pathology , Glycation End Products, Advanced , Glycogen/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , Male , Mesenchymal Stem Cells/cytology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin/genetics , Nestin/metabolism , Rats, Sprague-Dawley , Serum Albumin/analysis , Serum Albumin/metabolism , Stem Cells/cytology , Glycated Serum Albumin
7.
Mater Sci Eng C Mater Biol Appl ; 97: 593-601, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30678946

ABSTRACT

Diabetes mellitus is an epidemic worldwide. Pancreatic stem cells can be induced to differentiate into insulin-secreting cells, this method is an effective way to solve the shortage of islet donor. Poly (lactic acid-co-glycolic acid (PLGA) copolymer is an excellent scaffold for tissue engineering as it presents good biocompatibility and film forming properties. In this study, we adopted biological methods, using fibroblast-coated PLGA diaphragm to form a biological membrane, and then pancreatic stem cells were cultured on the fibroblast-modified PLGA membrane and the two-step induction method was utilized to induce the differentiation of pancreatic stem cells into insulin-secreting cells. The proliferation and differentiation of pancreatic stem cells on the fibroblast-modified PLGA membrane as well as the expression of genes related to the differentiation of pancreatic stem cells were examined in both normal and induced cultures to explore the potential of fibroblast-modified PLGA membrane for the transplantation to treat diabetes mellitus. The results indicated that fibroblasts can effectively improve the cell compatibility and histocompatibility of the PLGA membrane and promote the proliferation and differentiation of pancreatic stem cells. After induction, real-time fluorescence quantitative PCR (FQRT-PCR) results showed there were more Notch receptors and its ligands expressed in the membranes of pancreatic stem cells than non-induced pancreatic stem cells or fibroblast. Semiconductor quantum dot coupled-anti-complex probe experiments revealed that induced pancreatic stem cells had higher expression levels of Notch 2 and Delta-like 1 than non-induced ones, which may regulate the expression of Neurogenin-3 (Ngn3) and Hairy/Enhancer of split-1 gene (Hes1) through Notch signaling interaction between fibroblasts and pancreatic stem cells as well as enhance the proliferation of pancreatic stem cells and their differentiation into insulin-secreting cells. Further, our study suggests that the fibroblast-modified PLGA membrane can be used as matrix material composed of pancreatic stem cells or other stem cells to construct artificial islet tissue for the treatment of diabetes mellitus.


Subject(s)
Cell Differentiation , Insulin-Secreting Cells/metabolism , Pancreas/cytology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Stem Cells/cytology , Animals , Cell Culture Techniques/methods , Cells, Cultured , Collagen Type IV/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Glucose/pharmacology , Humans , Insulin/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Proteoglycans/metabolism , Rats, Wistar , Receptors, Notch/genetics , Receptors, Notch/metabolism , Stem Cells/metabolism , Tissue Scaffolds/chemistry
8.
Artif Cells Nanomed Biotechnol ; 46(1): 62-74, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29037070

ABSTRACT

Although melatonin has been shown to exhibit a wide variety of biological functions, its effects on promotion of self-renewal in pancreatic stem cells remain unknown. In this study, we incubated murine pancreatic stem cells (PSCs) with various concentrations of melatonin (0.01, 0.1, 1, 10 or 100 µM) to screen for the optimum culture medium for increasing cell proliferation. We found that 10 µM melatonin can significantly increase proliferation and enhance expression of a stem cell marker, nestin, in PSCs via melatonin receptor 2 (MT2). Thus, we used 10 µM melatonin to study the melatonin-mediated molecular mechanisms of cell proliferation in PSCs. We applied extracellular signal-regulated kinase (ERK) pathway inhibitor SCH772984 and transforming growth factor beta (TGF-ß) pathway inhibitor SB431542, along with interfering RNAs siERK1, siERK2, siSmad2, siSmad3, siSmad4 and siNestin, to melatonin-treated PSCs to research the roles of these genes in self-renewal. The results revealed a novel molecular mechanism by which melatonin promotes self-renewal of PSCs: a chain reaction in the MT2/ERK/SMAD/nestin axis promoted the aforementioned self-renewal as well as inhibited differentiation. In addition, upregulation of nestin created a positive feedback loop in the regulation of the transforming growth factor beta 1 (TGF-ß1)/SMADs pathway by promoting expression of Smad4. Conversely, knockdown of nestin significantly suppressed the proliferative effect in melatonin-treated PSCs. These are all novel mechanisms through which the ERK pathway cooperatively crosstalks with the SMAD pathway to regulate nestin expression, thereby enhancing self-renewal in PSCs.


Subject(s)
Cell Self Renewal/drug effects , Melatonin/pharmacology , Pancreas/cytology , Stem Cells/cytology , Stem Cells/drug effects , Animals , Cell Proliferation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Mice , Nestin/metabolism , Receptor, Melatonin, MT2/metabolism , Signal Transduction/drug effects
9.
Curr Stem Cell Res Ther ; 13(1): 60-79, 2018.
Article in English | MEDLINE | ID: mdl-29110621

ABSTRACT

BACKGROUND: Some research studies provided evidence for the differentiation capacity of adult stem cells (ASCs) into germ cells (GCs). Since the generation of GCs from stem cells (SCs) has been proposed as a potential way for treatment of infertility, many research groups have begun their creative studies on generation of new GCs both in vitro and in vivo, and utilized different ASC types such as bone marrow mesenchymal stem cells (BM-MSCs), skin stem cells, pancreatic stem cells, and adipose tissue MSCs. Despite many interesting reports with promising results, an obvious problem in the research projects was the functionality of the produced GCs. OBJECTIVE: In this paper, we have reviewed the results of almost all previously published reports on derivation of male and female GCs from ASCs to provide a better insight into this field of research. RESULTS: The most evaluated papers have shown that ASCs from various tissues can differentiate into GCs but rarely were the produced GCs functional and could form fertile gametes neither in vitro, nor in vivo (after transplantation into the gonads). CONCLUSION: There are still so many unknown issues about gametogenesis. Perhaps making alterations in treatment methods and utilizing creative techniques like tissue engineering and gene targeting help to achieve a standard method of in vitro GC production from ASCs.


Subject(s)
Adult Stem Cells/cytology , Adult Stem Cells/physiology , Cell Differentiation , Germ Cells/cytology , Germ Cells/physiology , Tissue Engineering/methods , Adult , Humans
10.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-811733

ABSTRACT

@#To further evaluate the effect of miR-217 in the proliferation of mouse adult pancreatic stem cells, we firstly transfected adult pancreatic stem cells with miR-217 mimics and studied the effect of miR-217 on proliferation through Western blot and immunofluorescence. Results showed that during the proliferation of adult pancreatic stem cells, miR-217 inhibited the protein expression of Ki-67 and Cyclin D1, which are related to cell propagation. As well as that, to investigate the target genes of miR-217 and their conserved sites bound by the seed region of miR-217, we used bioinformatic algorithms to find a potential target of miR-217 and verified by dual-luciferase activity assay. Surprisingly, dual-luciferase activity assay revealed that miR-217 could decrease PMIR-REPORT-Sirt1-3′UTR luciferase activity and Sirt1 is a direct target of miR-217. Finally, we verified the function of Sirt1 in the proliferation of pancreatic stem cells. Overexpression of miR-217 in pancreatic stem cells inhibited the level of Sirt1 in protein level but not in mRNA level. Furthermore, activator of Sirt1 played positive effect on colony formation ability and cell proliferation and inhibitor of Sirt1 showed the opposite function. In conclusion, miR-217 inhibits the proliferation of mouse adult pancreatic stem cells through Sirt1 and decreased expression of miR-217 to contribute to the pancreatic stem cells development.

11.
J Tissue Eng ; 8: 2041731417738145, 2017.
Article in English | MEDLINE | ID: mdl-29118967

ABSTRACT

Despite progress in the field of decellularization and recellularization, the outcome for pancreas has not been adequate. This might be due to the challenging dual nature of pancreas with both endocrine and exocrine tissues. We aimed to develop a novel and efficient cold-perfusion method for decellularization of porcine pancreas and recellularize acellular scaffolds with human fetal pancreatic stem cells. Decellularization of whole porcine pancreas at 4°C with sodium deoxycholate, Triton X-100 and DNase efficiently removed cellular material, while preserving the extracellular matrix structure. Furthermore, recellularization of acellular pieces with human fetal pancreatic stem cells for 14 days showed attached and proliferating cells. Both endocrine (C-peptide and PDX1) and exocrine (glucagon and α-amylase) markers were expressed in recellularized tissues. Thus, cold-perfusion can successfully decellularize porcine pancreas, which when recellularized with human fetal pancreatic stem cells shows relevant endocrine and exocrine phenotypes. Decellularized pancreas is a promising biomaterial and might translate to clinical relevance for treatment of diabetes.

12.
Curr Stem Cell Res Ther ; 12(8): 611-623, 2017.
Article in English | MEDLINE | ID: mdl-28831915

ABSTRACT

BACKGROUND: Diabetes mellitus, the widely prevalent disease of pancreas, is a metabolic disorder caused by autoimmune destruction of ß cells or insulin insufficiency or insulin resistance. Replacement of damaged ß cells by cell therapy can mitigate the condition and re-establish normal metabolic control. This has opened up new horizons for research, such as stem cells, cellular reprogramming and ß cell regeneration. OBJECTIVE: The goal of the study was to summarize the available literature on the use of stem cells for the regeneration of pancreatic ß cells and treatment of diabetes mellitus. RESULTS AND CONCLUSION: Stem cells are exceptional having the potential to self renew and differentiate in many lineages. Stem cells hold tremendous potential to regenerate ß cells and treat diabetes mellitus but many milestones on the way are yet to be achieved. But researchers do believe that stem cells and regenerative medicines will be widely used in clinical practices and possibly new effective methodology would be designed for even cure, mitigate and reduce the social burden of diabetes mellitus.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Diabetes Mellitus/therapy , Stem Cell Transplantation/methods , Animals , Cellular Reprogramming , Humans , Insulin-Secreting Cells , Regenerative Medicine/methods
13.
J Biomater Appl ; 32(3): 362-372, 2017 09.
Article in English | MEDLINE | ID: mdl-28747082

ABSTRACT

Objective To improve the biocompatibility between polylactic- co-glycolic acid membrane and pancreatic stem cells, rat fibroblasts were used to modify the polylactic- co-glycolic acid membrane. Meanwhile, we constructed artificial islet tissue by compound culturing the pancreatic stem cells and the fibroblast-modified polylactic- co-glycolic acid membrane and explored the function of artificial islets in diabetic nude mice. Methods Pancreatic stem cells were cultured on the fibroblast-modified polylactic- co-glycolic acid membrane in dulbecco's modified eagle medium containing activin-A, ß-catenin, and exendin-4. The differentiated pancreatic stem cells combined with modified polylactic- co-glycolic acid membrane were implanted subcutaneously in diabetic nude mice. The function of artificial islet tissue was explored by detecting blood levels of glucose and insulin in diabetic nude mice. Moreover, the proliferation and differentiation of pancreatic stem cells on modified polylactic- co-glycolic acid membrane as well as the changes on the tissue structure of artificial islets were investigated by immunofluorescence and haematoxylin and eosin staining. Results The pancreatic stem cells differentiated into islet-like cells and secreted insulin when cultured on fibroblast-modified polylactic- co-glycolic acid membrane. Furthermore, when the artificial islet tissues were implanted into diabetic nude mice, the pancreatic stem cells combined with polylactic- co-glycolic acid membrane modified by fibroblasts proliferated, differentiated, and secreted insulin to reduce blood glucose levels in diabetic nude mice. Conclusion Pancreatic stem cells can be induced to differentiate into islet-like cells in vitro. In vivo, the artificial islet tissue can effectively regulate the blood glucose level in nude mice within a short period. However, as time increased, the structure of the artificial islets was destroyed due to the erosion of blood cells that resulted in the gradual loss of artificial islet function.


Subject(s)
Biocompatible Materials/chemistry , Diabetes Mellitus, Experimental/therapy , Islets of Langerhans Transplantation , Islets of Langerhans/cytology , Lactic Acid/chemistry , Polyglycolic Acid/chemistry , Stem Cells/cytology , Tissue Engineering/methods , Animals , Cell Differentiation , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans Transplantation/methods , Membranes, Artificial , Mice, Nude , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , Rats, Wistar , Stem Cells/metabolism , Tissue Scaffolds/chemistry
14.
Biochem Biophys Res Commun ; 488(3): 471-476, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28501624

ABSTRACT

Porcine pancreatic stem cells (PSCs) are seed cells with potential use for diabetes treatment. Stem cell differentiation requires strict control of protein turnover and lysosomal digestion of organelles. Autophagy is a highly conserved process that controls the turnover of organelles and proteins within cells and contributes to the balance of cellular components. However, whether autophagy plays roles in PSC differentiation remains unknown. In this study, we successfully induced porcine PSCs into insulin-producing cells and found that autophagy was activated during the second induction stage. Inhibition of autophagy in the second stage resulted in reduced differentiational efficiency and impaired glucose-stimulated insulin secretion. Moreover, the expression of active ß-catenin increased while autophagy was activated but was suppressed when autophagy was inhibited. Therefore, autophagy is essential to the formation of insulin-producing cells, and the effects of autophagy on differentiation may be regulated by canonical Wnt signalling pathway.


Subject(s)
Autophagy , Cell Differentiation , Insulin/biosynthesis , Pancreas/cytology , Pancreas/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Insulin/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Swine , Wnt Signaling Pathway
15.
Biochem Biophys Res Commun ; 479(3): 537-543, 2016 Oct 21.
Article in English | MEDLINE | ID: mdl-27664705

ABSTRACT

Porcine pancreatic stem cells (PSCs) are one kind of the potential cells for treatment of human diabetes. Autophagy is a highly conserved cellular degradation process in which it helps to maintain the balance between the synthesis, degradation and subsequent recycling of cellular components. However, how autophagy contributes to PSCs has not yet been investigated. Here, we established GFP-LC3 transfected porcine PSC lines in which the accumulation of autophagosomes can be efficiently visualized to evaluate the autophagic activity. Moreover, we observed that starved PSCs which showed increased autophagic activity exhibited an increased tendency to proliferate through the results of BrdU, flow cytometry and western blotting. Furthermore, increased expression of active ß-catenin after inducing autophagy indicated that it might be the canonical Wnt signaling that autophagy activated to exert the function on the stimulation of PSCs proliferation. Collectively, these results demonstrated that autophagy stimulated proliferation of PSCs might be regulated by the canonical Wnt signaling pathway. Our results for the first time shed light on a role of autophagy for stimulating the proliferation of porcine PSCs.


Subject(s)
Autophagy , Pancreas/cytology , Stem Cells/cytology , Wnt Signaling Pathway/physiology , Animals , Bromodeoxyuridine/chemistry , Cell Culture Techniques , Cell Proliferation , Cells, Cultured , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Lentivirus/genetics , Microscopy, Fluorescence , Microscopy, Phase-Contrast , Polymerase Chain Reaction , Swine , Wnt Proteins/metabolism , beta Catenin/metabolism
16.
Cell Tissue Res ; 362(2): 379-88, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26085341

ABSTRACT

Pancreatic stem cells (PSCs) transplantation is a potential therapeutic approach to type 1 diabetes mellitus (D1M). However, before clinical use, there are some major hurdles to be faced that need to be comprehensively considered and given some potential solutions in vitro. Human PSCs are difficult to obtain and have a short replicative senescence. As an alternative, we instead established porcine PSCs; as insulin is highly conserved and physiological glucose levels are similar between human and porcine. In order to solve the problems during transplantation therapy, such as the need for an enormous amount of PSCs and good cell survival in overactive autoimmunity induced by reactive oxygen cpecies (ROS) in D1M patients, we utilized Wnt3a overexpression to activate the canonical Wnt signaling pathway in PSCs. We found that the expression of proliferation genes, such as c-Myc, was up-regulated as the downstream of ß-catenin, which promoted the PSCs proliferation and made cell numbers to meet the transplantation needs. We also showed that activation of the Wnt pathway made cells more readily tolerate ROS-caused mitochondria injury and cell apoptosis, thus making cells survive in autoimmune patients. The present study provides a theoretical basis for cell transplantation therapy of diabetes.


Subject(s)
Apoptosis/physiology , Cell Proliferation/physiology , Pancreas/metabolism , Stem Cells/metabolism , Wnt Signaling Pathway/physiology , Wnt3A Protein/metabolism , Animals , Cell Survival , Cells, Cultured , Pancreas/cytology , Swine , beta Catenin/metabolism
17.
Cell Med ; 5(2-3): 63-8, 2013 Nov 10.
Article in English | MEDLINE | ID: mdl-26858867

ABSTRACT

Recently, mouse pancreatic stem cells have been isolated from adult mouse pancreata. However, these pancreatic stem cells could be maintained only under specific culture conditions with lot-limited fetal bovine serum (FBS). For the efficient isolation and maintenance of mouse pancreatic stem cells, it is important to identify culture conditions that can be used independent of the FBS lot. In this study, we evaluated the culture conditions required to maintain mouse pancreatic stem cells. The mouse pancreatic stem cells derived from the pancreas of a newborn mouse, HN#101, were cultured under the following conditions: 1) Dulbecco's modified Eagle's medium (DMEM) with 20% lot-limited FBS, in which mouse pancreatic stem cells could be cultured without changes in morphology and growth activity; 2) complete embryonic stem (ES) cell media; and 3) complete ES cell media on feeder layers of mitomycin C-treated STO cells, which were the same culture conditions used for mouse ES cells. Under culture conditions #1 and #3, the HN#101 cells continued to form a flat "cobblestone" monolayer and continued to divide actively beyond the population doubling level (PDL) 100 without growth inhibition, but this did not occur under culture condition #2. The gene expression profile and differentiated capacity of the HN#101 cells cultured for 2 months under culture condition #3 were similar to those of HN#101 cells at PDL 50. These data suggest that complete ES cell media on feeder layers could be useful for maintaining the undifferentiated state of pancreatic stem cells.

18.
Cell Med ; 5(2-3): 69-73, 2013 Nov 10.
Article in English | MEDLINE | ID: mdl-26858868

ABSTRACT

Mouse pancreatic stem cells have been isolated from mouse pancreata. This study evaluated the efficacy of isolating mouse pancreatic stem cells using mice of different ages. The pancreata of newborn mice, 8-week-old mice, and 24-week-old mice were harvested and digested by using collagenase. The "duct-like" cells in the digested pancreatic tissue were then inoculated into 96-well plates, cloned by limiting dilution, and cultured in DMEM with 20% FBS. Pancreatic stem cells were isolated from the pancreata of all newborn mice, while cells could only be isolated from 10% of the pancreata of 8-week-old mice and could not be isolated from the pancreata of any 24-week-old mice. These data suggest that young mice may have some pancreatic stem cells and that older mice may only have a few pancreatic stem cells. These data also indicate that it is extremely difficult to isolate pancreatic stem cells from older mice, suggesting that future research focus its efforts on finding methods of isolating pancreatic stem cells from adult mice.

19.
Neural Regen Res ; 7(7): 506-10, 2012 Mar 05.
Article in English | MEDLINE | ID: mdl-25745436

ABSTRACT

Pancreatic stem cells were isolated and cultured from aborted human fetal pancreases of gestational age 14-20 weeks. They were seeded at a density of 1 × 10(4) in serum-free media for differentiation into neuron-like cells, expressing ß-tubulin III and glial fibrillary acidic protein. These neuron-like cells displayed a synapse-like morphology and appeared to form a neuronal network. Pancreatic stem cells were also seeded at a density of 1 × 10(5) for differentiation into islet-like cells, expressing insulin and glucagon, with an islet-like morphology. These cells had glucose-stimulated secretion of human insulin and C-peptide. Results suggest that pancreatic stem cells can be differentiated into neuron-like and islet-like cells.

20.
Cell Med ; 3(1-3): 51-61, 2012 Jan.
Article in English | MEDLINE | ID: mdl-28058181

ABSTRACT

Islet transplantation, including ß-cells, has proven to be effective for diabetes in many recent studies; however, this treatment strategy requires sufficient organ donors. One attractive approach for the generation of ß-cells is to utilize the expansion and differentiation of cells from pancreatic stem cells (PSCs), which are closely associated to the ß-cells lineage. In this study, we investigated whether important transcription factors (Pdx-1, Ngn3, NeuroD, and MafA) in islet cells could be efficiently transduced into mouse PSCs (mPSCs) using Sendai virus (SeV) vectors and found that the transduced cells were differentiated into insulin-producing pancreatic ß-cells. The mPSCs transduced with single transcription factors using SeV vectors could not express the insulin-2 mRNA. When combinations of two transcription factors were transduced using the SeV vectors, including combinations of Pdx-1 + NeuroD, Pdx-1 + MafA, and NeuroD + MafA, the expression of insulin-2 mRNA was low but could be detected. When combinations of three or more transcription factors were transduced using SeV vectors, the expression of insulin-2 mRNA could be detected. In particular, the transduction of the combination of PDX-1, NeuroD, and MafA produced the most effective for the expression of insulin-2 mRNA out of all of the different combinations examined. These data suggest that the transduction of transcription factors using SeV vectors facilitates mPSC differentiation into insulin-producing cells and showed the possibility of regenerating ß-cells by using transduced PSCs.

SELECTION OF CITATIONS
SEARCH DETAIL
...