Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
Stem Cell Reports ; 12(3): 611-623, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30773486

ABSTRACT

The transplantation of human embryonic stem cell (hESC)-derived insulin-producing ß cells for the treatment of diabetes is finally approaching the clinical stage. However, even with state-of-the-art differentiation protocols, a significant percentage of undefined non-endocrine cell types are still generated. Most importantly, there is the potential for carry-over of non-differentiated cell types that may produce teratomas. We sought to modify hESCs so that their differentiated progeny could be selectively devoid of tumorigenic cells and enriched for cells of the desired phenotype (in this case, ß cells). Here we report the generation of a modified hESC line harboring two suicide gene cassettes, whose expression results in cell death in the presence of specific pro-drugs. We show the efficacy of this system at enriching for ß cells and eliminating tumorigenic ones both in vitro and in vivo. Our approach is innovative inasmuch as it allows for the preservation of the desired cells while eliminating those with the potential to develop teratomas.


Subject(s)
Carcinogenesis/pathology , Human Embryonic Stem Cells/pathology , Insulin-Secreting Cells/pathology , Animals , Carcinogenesis/genetics , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Teratoma/genetics , Teratoma/pathology
2.
Methods Mol Biol ; 1871: 253-264, 2019.
Article in English | MEDLINE | ID: mdl-30276744

ABSTRACT

Diabetes mellitus is caused by either loss of pancreatic islets ß-cells (Type 1 Diabetes, T1D), insufficient insulin release in the islet ß-cells coupled with insulin resistance in target tissues (Type 2 Diabetes, T2D), or impaired insulin release (genetic forms of diabetes and, possibly, T1D subtypes). The investigation of the islet proteome could elucidate facets of the pathogenesis of diabetes. Enzymatically isolated and cultured (EIC) islets are frequently used to investigate biochemical signaling pathways that could trigger ß-cell changes and death in diabetes. However, they cannot fully reflect the natural protein composition and disease process of in vivo islets due to the stress from isolation procedures and in vitro culture. The laser capture microdissection method employs a high-energy laser source to separate the desired cells from the remaining tissue section in an environment which is well conserved and close to the natural condition. Here, we describe a label-free proteomic workflow of laser capture microdissected (LCM) human islets from fresh-frozen pancreas sections of cadaveric donors to obtain an accurate and unbiased profile of the pancreatic islet proteome. The workflow includes preparation of frozen tissue section, staining and dehydration, LCM islets collection, islet protein digestion, label-free Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS), database search, and statistical analysis.


Subject(s)
Chromatography, Liquid , Islets of Langerhans/metabolism , Proteome , Proteomics , Tandem Mass Spectrometry , Chromatography, Liquid/methods , Computational Biology/methods , Data Analysis , Databases, Protein , Frozen Sections , Humans , Immunohistochemistry , Islets of Langerhans/cytology , Laser Capture Microdissection , Proteome/metabolism , Proteomics/methods , Tandem Mass Spectrometry/methods
3.
Cell Rep ; 22(9): 2408-2420, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29490276

ABSTRACT

Treatment of human pancreatic non-endocrine tissue with Bone Morphogenetic Protein 7 (BMP-7) leads to the formation of glucose-responsive ß-like cells. Here, we show that BMP-7 acts on extrainsular cells expressing PDX1 and the BMP receptor activin-like kinase 3 (ALK3/BMPR1A). In vitro lineage tracing indicates that ALK3+ cell populations are multipotent. PDX1+/ALK3+ cells are absent from islets but prominently represented in the major pancreatic ducts and pancreatic duct glands. We identified the purinergic receptor P2Y1 (P2RY1) as a surrogate surface marker for PDX1. Sorted P2RY1+/ALK3bright+ cells form BMP-7-expandable colonies characterized by NKX6.1 and PDX1 expression. Unlike the negative fraction controls, these colonies can be differentiated into multiple pancreatic lineages upon BMP-7 withdrawal. RNA-seq further corroborates the progenitor-like nature of P2RY1+/ALK3bright+ cells and their multilineage differentiation potential. Our studies confirm the existence of progenitor cells in the adult human pancreas and suggest a specific anatomical location within the ductal and glandular networks.


Subject(s)
Bone Morphogenetic Protein 7/pharmacology , Bone Morphogenetic Protein Receptors, Type I/metabolism , Pancreas, Exocrine/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Adult , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cell Lineage/genetics , Cell Proliferation/drug effects , Cells, Cultured , DNA Replication/drug effects , Homeodomain Proteins/metabolism , Humans , Stem Cells/drug effects , Trans-Activators/metabolism
4.
J Proteomics ; 150: 149-159, 2017 01 06.
Article in English | MEDLINE | ID: mdl-27620696

ABSTRACT

The etiology of Type 1 Diabetes (T1D) remains elusive. Enzymatically isolated and cultured (EIC) islets cannot fully reflect the natural protein composition and disease process of in vivo islets, because of the stress from isolation procedures. In order to study islet protein composition in conditions close to the natural environment, we performed proteomic analysis of EIC islets, and laser capture microdissected (LCM) human islets and acinar tissue from fresh-frozen pancreas sections of three cadaveric donors. 1104 and 706 proteins were identified from 6 islets equivalents (IEQ) of LCM islets and acinar tissue, respectively. The proteomic profiles of LCM islets were reproducible within and among cadaveric donors. The endocrine hormones were only detected in LCM islets, whereas catalytic enzymes were significantly enriched in acinar tissue. Furthermore, high overlap (984 proteins) and similar function distribution were found between LCM and EIC islets proteomes, except that EIC islets had more acinar contaminants and stress-related signal transducer activity proteins. The comparison among LCM islets, LCM acinar tissue and EIC islets proteomes indicates that LCM combined with proteomic methods enables accurate and unbiased profiling of islet proteome from frozen pancreata. This paves the way for proteomic studies on human islets during the progression of T1D. SIGNIFICANCE: The etiological agent triggering autoimmunity against beta cells in Type 1 diabetes (T1D) remains obscure. The in vitro models available (enzymatically isolated and cultured islets, EIC islets) do not accurately reflect what happens in vivo due to lack of the natural environment where islets exist and the preparation-induced changes in cell physiology. The importance of this study is that we investigated the feasibility of laser capture microdissection (LCM) for the isolation of intact islets from frozen cadaveric pancreatic tissue sections. We compared the protein profile of LCM islets (9 replicates from 3 cadaveric donors) with that of both LCM acinar tissues (6 replicates from the same 3 cadaveric donor as LCM islets) and EIC islets (at least 4 replicates for each sample with the same islets equivalents) by using proteomics techniques with advanced instrumentation, nanoLC-Q Exactive HF Orbitrap mass spectrometry (nano LC-MS/MS). The results demonstrate that the LCM method is reliable in isolating islets with an intact environment. LCM-based islet proteomics is a feasible approach to obtain good proteome coverage for assessing the pathology of T1D using cadaveric pancreatic samples, even from very small sample amounts. Future applications of this LCM-based proteomic method may help us understand the pathogenesis of T1D and identify potential biomarkers for T1D diagnosis at an early stage.


Subject(s)
Frozen Sections , Islets of Langerhans/metabolism , Laser Capture Microdissection , Pancreas/metabolism , Proteome/analysis , Cadaver , Cell Separation/methods , Cells, Cultured , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Humans , Islets of Langerhans/pathology , Pancreas/pathology , Primary Cell Culture , Proteome/metabolism , Proteomics/methods , Specimen Handling/methods
5.
PLoS One ; 11(6): e0157245, 2016.
Article in English | MEDLINE | ID: mdl-27285580

ABSTRACT

BACKGROUND: Allogeneic human islet transplantation is an effective therapy for the treatment of patients with Type 1 Diabetes (T1D). The low number of islet transplants performed worldwide and the different transplantation protocols used limit the identification of the most effective therapeutic options to improve the efficacy of this approach. METHODS: We present a retrospective analysis on the data collected from 44 patients with T1D who underwent islet transplantation at our institute between 2000 and 2007. Several variables were included: recipient demographics and immunological characteristics, donor and transplant characteristics, induction protocols, and additional medical treatment received. Immunosuppression was induced with anti-CD25 (Daclizumab), alone or in association with anti-tumor necrosis factor alpha (TNF-α) treatments (Etanercept or Infliximab), or with anti-CD52 (Alemtuzumab) in association with anti-TNF-α treatments (Etanercept or Infliximab). Subsets of patients were treated with Filgrastim for moderate/severe neutropenia and/or Exenatide for post prandial hyperglycemia. RESULTS: The analysis performed indicates a negative association between graft survival (c-peptide level ≥ 0.3 ng/ml) and islet infusion volume, with the caveat that, the progressive reduction of infusion volumes over the years has been paralleled by improved immunosuppressive protocols. A positive association is instead suggested between graft survival and administration of Exenatide and Filgrastim, alone or in combination. CONCLUSION: This retrospective analysis may be of assistance to further improve long-term outcomes of protocols for transplant of islets and other organs.


Subject(s)
Filgrastim/therapeutic use , Graft Survival/drug effects , Hematologic Agents/therapeutic use , Hypoglycemic Agents/therapeutic use , Islets of Langerhans Transplantation/methods , Islets of Langerhans/drug effects , Peptides/therapeutic use , Venoms/therapeutic use , Adult , Aged , Antibodies, Monoclonal, Humanized/therapeutic use , Daclizumab , Exenatide , Humans , Hyperglycemia/drug therapy , Hyperglycemia/etiology , Immunoglobulin G/therapeutic use , Immunosuppressive Agents/therapeutic use , Islets of Langerhans/physiology , Islets of Langerhans Transplantation/adverse effects , Middle Aged , Neutropenia/drug therapy , Neutropenia/etiology , Retrospective Studies , Transplantation, Homologous
6.
J Diabetes Res ; 2015: 737586, 2015.
Article in English | MEDLINE | ID: mdl-26347378

ABSTRACT

The nonobese diabetic (NOD) mouse represents a well-established experimental model analogous to human type 1 diabetes mellitus (T1D) as it is characterized by progressive autoimmune destruction of pancreatic ß-cells. Experiments were designed to investigate the impact of moderate-intensity training on T1D immunomodulation and inflammation. Under a chronic exercise regime, NOD mice were trained on a treadmill for 12 weeks (12 m/min for 30 min, 5 d/wk) while age-matched, control animals were left untrained. Prior to and upon completion of the training period, fed plasma glucose and immunological soluble factors were monitored. Both groups showed deteriorated glycemic profiles throughout the study although trained mice tended to be more compensated than controls after 10 weeks of training. An exercise-induced weight loss was detected in the trained mice with respect to the controls from week 6. After 12 weeks, IL-6 and MIP-1ß were decreased in the trained animals compared to their baseline values and versus controls, although not significantly. Morphometric analysis of pancreata revealed the presence of larger infiltrates along with decreased α-cells areas in the control mice compared to trained mice. Exercise may exert positive immunomodulation of systemic functions with respect to both T1D and inflammation, but only in a stringent therapeutic window.


Subject(s)
Diabetes Mellitus, Type 1/blood , Hyperglycemia/pathology , Inflammation/pathology , Physical Conditioning, Animal , Animals , Blood Glucose/analysis , Body Weight , Chemokine CCL4/blood , Diabetes Mellitus, Type 1/genetics , Female , Hyperglycemia/therapy , Immunohistochemistry , Inflammation/therapy , Insulin-Secreting Cells/cytology , Interleukin-6/blood , Mice , Mice, Inbred NOD , Pancreas/metabolism
7.
Diabetes ; 64(12): 4123-34, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26307584

ABSTRACT

The exocrine pancreas can give rise to endocrine insulin-producing cells upon ectopic expression of key transcription factors. However, the need for genetic manipulation remains a translational hurdle for diabetes therapy. Here we report the conversion of adult human nonendocrine pancreatic tissue into endocrine cell types by exposure to bone morphogenetic protein 7. The use of this U.S. Food and Drug Administration-approved agent, without any genetic manipulation, results in the neogenesis of clusters that exhibit high insulin content and glucose responsiveness both in vitro and in vivo. In vitro lineage tracing confirmed that BMP-7-induced insulin-expressing cells arise mainly from extrainsular PDX-1(+), carbonic anhydrase II(-) (mature ductal), elastase 3a (acinar)(-) , and insulin(-) subpopulations. The nongenetic conversion of human pancreatic exocrine cells to endocrine cells is novel and represents a safer and simpler alternative to genetic reprogramming.


Subject(s)
Bone Morphogenetic Protein 7/pharmacology , Cell Transdifferentiation/drug effects , Diabetes Mellitus, Experimental/therapy , Insulin-Secreting Cells/drug effects , Pancreas, Exocrine/drug effects , Animals , Biomarkers/metabolism , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein 7/metabolism , C-Peptide/blood , C-Peptide/metabolism , Cell Lineage , Cells, Cultured , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Fluorescent Antibody Technique , Homeodomain Proteins/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/transplantation , Kidney , Male , Mice, Nude , Pancreas, Exocrine/metabolism , Pancreas, Exocrine/pathology , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Trans-Activators/metabolism , Transplantation, Heterologous , Transplantation, Heterotopic
8.
Biotechnol Bioeng ; 112(9): 1916-26, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25786390

ABSTRACT

With a view toward reduction of graft loss, we explored pancreatic islet transplantation within fibrin matrices rendered pro-angiogenic by incorporation of minimal doses of vascular endothelial growth factor-A165 and platelet-derived growth factor-BB presented complexed to a fibrin-bound integrin-binding fibronectin domain. Engineered matrices allowed for extended release of pro-angiogenic factors and for their synergistic signaling with extracellular matrix-binding domains in the post-transplant period. Aprotinin addition delayed matrix degradation and prolonged pro-angiogenic factor availability within the graft. Both subcutaneous (SC) and epididymal fat pad (EFP) sites were evaluated. We show that in the SC site, diabetes reversal in mice transplanted with 1,000 IEQ of syngeneic islets was not observed for islets transplanted alone, while engineered matrices resulted in a diabetes median reversal time (MDRT) of 38 days. In the EFP site, the MDRT with 250 IEQ of syngeneic islets within the engineered matrices was 24 days versus 86 days for islets transplanted alone. Improved function of engineered grafts was associated with enhanced and earlier (by day 7) angiogenesis. Our findings show that by engineering the transplant site to promote prompt re-vascularization, engraftment and long-term function of islet grafts can be improved in relevant extrahepatic sites.


Subject(s)
Fibrin/chemistry , Islets of Langerhans Transplantation , Islets of Langerhans/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Animals , Becaplermin , Cell Proliferation/drug effects , Humans , Hydrogels/chemistry , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-sis/chemistry , Proto-Oncogene Proteins c-sis/deficiency , Proto-Oncogene Proteins c-sis/pharmacology , Vascular Endothelial Growth Factor A/chemistry
9.
Eur J Immunol ; 44(11): 3307-19, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25113564

ABSTRACT

By restraining T-cell activation and promoting Treg-cell expansion, myeloid-derived suppressor cells (MDSCs) and tolerogenic DCs can control self-reactive and antigraft effector T cells in autoimmunity and transplantation. Their therapeutic use and characterization, however, is limited by their scarce availability in the peripheral blood of tumor-free donors. In the present study, we describe and characterize a novel population of human myeloid suppressor cells, named fibrocytic MDSC, which are differentiated from umbilical cord blood precursors by 4-day culture with FDA-approved cytokines (recombinant human-GM-CSF and recombinant human-G-CSF). This MDSC subset, characterized by the expression of MDSC-, DC-, and fibrocyte-associated markers, promotes Treg-cell expansion and induces normoglycemia in a xenogeneic mouse model of Type 1 diabetes. In order to exert their protolerogenic function, fibrocytic MDSCs require direct contact with activated T cells, which leads to the production and secretion of IDO. This new myeloid subset may have an important role in the in vitro and in vivo production of Treg cells for the treatment of autoimmune diseases, and in either the prevention or control of allograft rejection.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Lymphocyte Activation/immunology , Myeloid Cells/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation/immunology , Cell Line , Cell Proliferation , Diabetes Mellitus, Type 1/immunology , Female , Fetal Blood/cytology , Gene Expression Profiling , Graft Rejection/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , HEK293 Cells , Humans , Immune Tolerance , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Myeloid Cells/cytology , Recombinant Proteins/pharmacology , T-Lymphocytes, Regulatory/transplantation
10.
Pancreas ; 43(8): 1317-22, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25036907

ABSTRACT

OBJECTIVES: The effects of glucocorticoid during culture on human islet cells have been controversial. Exendin-4 (EX) enhances the insulin secretion and significantly improves clinical outcomes in islet cell transplantation. In this study, we examined the effects of glucocorticoids and EX on human islet cells during pretransplant culture. METHODS: Methylprednisolone (MP) and/or EX were added to the standard culture medium for clinical islet cell transplantation. Islets were cultured for 24 hours with 3 different conditions (control, no additives; MP alone; and MP + EX). ß-Cell fractional viability, cellular composition, multiple cytokine/chemokine production, multiple phosphorylation proteins, and glucose-induced insulin secretion were evaluated. RESULTS: Viable ß-cell survival in MP and MP + EX group was significantly higher than in the control group. Exendin-4 prevented MP-induced reduction of insulin secretion. Methylprednisolone supplementation to the culture medium decreased cytokine and chemokine production. Moreover, extracellular signal-regulated kinase 1/2 phosphorylation was significantly increased by MP and MP + EX. CONCLUSIONS: Glucocorticoid supplementation into culture media significantly decreased the cytokine/chemokine production and increased the extracellular signal-regulated kinase 1/2 phosphorylation, resulting in the improvement of human ß-cell survival. In addition, EX maintained the insulin secretion suppressed by MP. The supplementation of MP and EX together could be a useful strategy to create suitable human islets for transplantation.


Subject(s)
Cell Culture Techniques/methods , Culture Media/pharmacology , Insulin/metabolism , Islets of Langerhans/cytology , Cell Survival , Chemokines/metabolism , Cytokines/metabolism , Exenatide , Glucose/pharmacology , Humans , Inflammation Mediators , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , MAP Kinase Signaling System/drug effects , Methylprednisolone/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Peptides/pharmacology , Phosphoproteins/analysis , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Venoms/pharmacology
11.
Genom Data ; 2: 389-92, 2014 Dec.
Article in English | MEDLINE | ID: mdl-26484135

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) have been shown to control self-reactive and anti-graft effector T-cells in autoimmunity and transplantation, but their therapeutic use is limited by their scarce availability in the peripheral blood of tumor-free donors. We isolated and characterized a novel population of myeloid suppressor cells, named fibrocytic MDSC (f-MDSC), which are differentiated from umbilical cord blood (UCB) precursors (Zoso et al., 2014). This MDSC subset promotes regulatory T-cell expansion and induces normoglycemia in a xenogeneic model of type 1 diabetes. Here we describe in details the experimental design and the bioinformatics analyses of the gene expression dataset used to investigate the molecular mechanisms at the base of MDSC tolerogenic and suppressive properties. We also provide an R code to easily access the data and perform the quality controls and basic analyses relevant to this dataset. Raw and pre-processed data are available at Gene Expression Omnibus under accession GSE52376.

12.
Stem Cells Transl Med ; 3(3): 277-89, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24375542

ABSTRACT

The possibility of using human embryonic stem (hES) cell-derived ß cells as an alternative to cadaveric islets for the treatment of type 1 diabetes is now widely acknowledged. However, current differentiation methods consistently fail to generate meaningful numbers of mature, functional ß cells. In order to address this issue, we set out to explore the role of oxygen modulation in the maturation of pancreatic progenitor (PP) cells differentiated from hES cells. We have previously determined that oxygenation is a powerful driver of murine PP differentiation along the endocrine lineage of the pancreas. We hypothesized that targeting physiological oxygen partial pressure (pO2) levels seen in mature islets would help the differentiation of PP cells along the ß-cell lineage. This hypothesis was tested both in vivo (by exposing PP-transplanted immunodeficient mice to a daily hyperbaric oxygen regimen) and in vitro (by allowing PP cells to mature in a perfluorocarbon-based culture device designed to carefully adjust pO2 to a desired range). Our results show that oxygen modulation does indeed contribute to enhanced maturation of PP cells, as evidenced by improved engraftment, segregation of α and ß cells, body weight maintenance, and rate of diabetes reversal in vivo, and by elevated expression of pancreatic endocrine makers, ß-cell differentiation yield, and insulin production in vitro. Our studies confirm the importance of oxygen modulation as a key variable to consider in the design of ß-cell differentiation protocols and open the door to future strategies for the transplantation of fully mature ß cells.


Subject(s)
Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/therapy , Embryonic Stem Cells/drug effects , Immunocompromised Host , Insulin-Secreting Cells/drug effects , Oxygen/pharmacology , Animals , Cell Culture Techniques , Cell Differentiation , Diabetes Mellitus, Experimental/pathology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/immunology , Fluorocarbons/pharmacology , Glucagon-Secreting Cells/cytology , Glucagon-Secreting Cells/drug effects , Glucagon-Secreting Cells/immunology , Glucose/metabolism , Glucose/pharmacology , Graft Survival , Humans , Insulin/biosynthesis , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/transplantation , Mice , Mice, Nude , Oxygen Consumption/physiology
13.
Stem Cells ; 31(10): 2047-60, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23873634

ABSTRACT

Regenerative medicine is transitioning into clinical programs using stem/progenitor cell therapies for repair of damaged organs. We summarize those for liver and pancreas, organs that share endodermal stem cell populations, biliary tree stem cells (hBTSCs), located in peribiliary glands. They are precursors to hepatic stem/progenitors in canals of Hering and to committed progenitors in pancreatic duct glands. They give rise to maturational lineages along a radial axis within bile duct walls and a proximal-to-distal axis starting at the duodenum and ending with mature cells in the liver or pancreas. Clinical trials have been ongoing for years assessing effects of determined stem cells (fetal-liver-derived hepatic stem/progenitors) transplanted into the hepatic artery of patients with various liver diseases. Immunosuppression was not required. Control subjects, those given standard of care for a given condition, all died within a year or deteriorated in their liver functions. Subjects transplanted with 100-150 million hepatic stem/progenitor cells had improved liver functions and survival extending for several years. Full evaluations of safety and efficacy of transplants are still in progress. Determined stem cell therapies for diabetes using hBTSCs remain to be explored but are likely to occur following ongoing preclinical studies. In addition, mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs) are being used for patients with chronic liver conditions or with diabetes. MSCs have demonstrated significant effects through paracrine signaling of trophic and immunomodulatory factors, and there is limited evidence for inefficient lineage restriction into mature parenchymal or islet cells. HSCs' effects are primarily via modulation of immune mechanisms.


Subject(s)
Hepatitis/therapy , Mesenchymal Stem Cell Transplantation , Pancreatitis/therapy , Cell Differentiation , Cell Lineage , Hepatitis/immunology , Humans , Liver/embryology , Liver/immunology , Liver/pathology , Mesenchymal Stem Cells/physiology , Pancreas/embryology , Pancreas/immunology , Pancreas/pathology , Pancreatitis/immunology , Stem Cell Niche
14.
Stem Cells ; 31(9): 1966-79, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23847135

ABSTRACT

Peribiliary glands (PBGs) in bile duct walls, and pancreatic duct glands (PDGs) associated with pancreatic ducts, in humans of all ages, contain a continuous, ramifying network of cells in overlapping maturational lineages. We show that proximal (PBGs)-to-distal (PDGs) maturational lineages start near the duodenum with cells expressing markers of pluripotency (NANOG, OCT4, and SOX2), proliferation (Ki67), self-replication (SALL4), and early hepato-pancreatic commitment (SOX9, SOX17, PDX1, and LGR5), transitioning to PDG cells with no expression of pluripotency or self-replication markers, maintenance of pancreatic genes (PDX1), and expression of markers of pancreatic endocrine maturation (NGN3, MUC6, and insulin). Radial-axis lineages start in PBGs near the ducts' fibromuscular layers with stem cells and end at the ducts' lumens with cells devoid of stem cell traits and positive for pancreatic endocrine genes. Biliary tree-derived cells behaved as stem cells in culture under expansion conditions, culture plastic and serum-free Kubota's Medium, proliferating for months as undifferentiated cells, whereas pancreas-derived cells underwent only approximately 8-10 divisions, then partially differentiated towards an islet fate. Biliary tree-derived cells proved precursors of pancreas' committed progenitors. Both could be driven by three-dimensional conditions, islet-derived matrix components and a serum-free, hormonally defined medium for an islet fate (HDM-P), to form spheroids with ultrastructural, electrophysiological and functional characteristics of neoislets, including glucose regulatability. Implantation of these neoislets into epididymal fat pads of immunocompromised mice, chemically rendered diabetic, resulted in secretion of human C-peptide, regulatable by glucose, and able to alleviate hyperglycemia in hosts. The biliary tree-derived stem cells and their connections to pancreatic committed progenitors constitute a biological framework for life-long pancreatic organogenesis.


Subject(s)
Biliary Tract/cytology , Cell Lineage , Organogenesis , Pancreas/cytology , Pancreas/growth & development , Stem Cells/cytology , Adult , Animals , Antigens, Neoplasm/metabolism , Biomarkers/metabolism , Cell Adhesion Molecules/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Experimental/therapy , Electrophysiological Phenomena , Epithelial Cell Adhesion Molecule , Gene Expression Regulation , Humans , Hyperglycemia/therapy , Islets of Langerhans/cytology , Islets of Langerhans/physiology , Islets of Langerhans/ultrastructure , Islets of Langerhans Transplantation , Mice , Organogenesis/genetics , Pancreatic Ducts/cytology , Phenotype , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Spheroids, Cellular/ultrastructure , Stem Cell Niche/genetics , Stem Cells/metabolism
15.
PLoS One ; 8(1): e55064, 2013.
Article in English | MEDLINE | ID: mdl-23383059

ABSTRACT

microRNAs (miRNAs) play an important role in pancreatic development and adult ß-cell physiology. Our hypothesis is based on the assumption that each islet cell type has a specific pattern of miRNA expression. We sought to determine the profile of miRNA expression in α-and ß-cells, the main components of pancreatic islets, because this analysis may lead to a better understanding of islet gene regulatory pathways. Highly enriched (>98%) subsets of human α-and ß-cells were obtained by flow cytometric sorting after intracellular staining with c-peptide and glucagon antibody. The method of sorting based on intracellular staining is possible because miRNAs are stable after fixation. MiRNA expression levels were determined by quantitative high throughput PCR-based miRNA array platform screening. Most of the miRNAs were preferentially expressed in ß-cells. From the total of 667 miRNAs screened, the Significant Analysis of Microarray identified 141 miRNAs, of which only 7 were expressed more in α-cells (α-miRNAs) and 134 were expressed more in ß-cells (ß-miRNAs). Bioinformatic analysis identified potential targets of ß-miRNAs analyzing the Beta Cell Gene Atlas, described in the T1Dbase, the web platform, supporting the type 1 diabetes (T1D) community. cMaf, a transcription factor regulating glucagon expression expressed selectively in α-cells (TFα) is targeted by ß-miRNAs; miR-200c, miR-125b and miR-182. Min6 cells treated with inhibitors of these miRNAs show an increased expression of cMaf RNA. Conversely, over expression of miR-200c, miR-125b or miR-182 in the mouse alpha cell line αTC6 decreases the level of cMAF mRNA and protein. MiR-200c also inhibits the expression of Zfpm2, a TFα that inhibits the PI3K signaling pathway, at both RNA and protein levels.In conclusion, we identified miRNAs differentially expressed in pancreatic α- and ß-cells and their potential transcription factor targets that could add new insights into different aspects of islet biology and pathophysiology.


Subject(s)
Glucagon-Secreting Cells/metabolism , Insulin-Secreting Cells/metabolism , MicroRNAs/genetics , Transcriptome , Adult , Animals , Cell Line , Computational Biology , Humans , Mice , Middle Aged , Rats
16.
Cell Transplant ; 22(9): 1723-33, 2013.
Article in English | MEDLINE | ID: mdl-23068091

ABSTRACT

Conventional culture vessels are not designed for physiological oxygen (O2) delivery. Both hyperoxia and hypoxia-commonly observed when culturing cells in regular plasticware-have been linked to reduced cellular function and death. Pancreatic islets, used for the clinical treatment of diabetes, are especially sensitive to sub- and supraphysiological O2 concentrations. A result of current culture standards is that a high percentage of islet preparations are never transplanted because of cell death and loss of function in the 24-48 h postisolation. Here, we describe a new culture system designed to provide quasiphysiological oxygenation to islets in culture. The use of dishes where islets rest atop a perfluorocarbon (PFC)-based membrane, coupled with a careful adjustment of environmental O2 concentration to target the islet physiological pO2 range, resulted in dramatic gains in viability and function. These observations underline the importance of approximating culture conditions as closely as possible to those of the native microenvironment, and fill a widely acknowledged gap in our ability to preserve islet functionality in vitro. As stem cell-derived insulin-producing cells are likely to suffer from the same limitations as those observed in real islets, our findings are especially timely in the context of current efforts to define renewable sources for transplantation.


Subject(s)
Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Fluorocarbons , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/physiology , Oxygen/administration & dosage , Oxygen/metabolism , Animals , Cell Death/physiology , Cell Survival/physiology , Female , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Islets of Langerhans/cytology , Mice , Mice, Nude
17.
PLoS One ; 7(12): e50265, 2012.
Article in English | MEDLINE | ID: mdl-23227162

ABSTRACT

Two major hurdles need to be surmounted for cell therapy for diabetes: (i) allo-immune rejection of grafted pancreatic islets, or stem/precursor cell-derived insulin-secreting cells; and (ii) continuing auto-immunity against the diabetogenic endogenous target antigen. Nanotherapeutics offer a novel approach to overcome these problems and here we ask if creation of "stealth" islets encapsulated within a thin cage of pegylated material of 100-200 nanometers thick provides a viable option for islet transplantation. The aims of this study were to test islet viability and functionality following encapsulation within the pegylated cage, and functional efficacy in vivo in terms of graft-derived control of normoglycemia in diabetic mice. We first demonstrated that pegylation of the islet surface, plus or minus nanoparticles, improved long-term islet viability in vitro compared to non-pegylated (naked) control islets. Moreover, pegylation of the islets with nanoparticles was compatible with glucose-stimulated insulin secretion and insulin biogenesis. We next looked for functionality of the created "stealth" DBA/2 (H-2(d)) islets in vivo by comparing glycemic profiles across 4 groups of streptozotozin-induced diabetic C57BL/6 (H-2(b)) recipients of (i) naked islets; (ii) pegylated islets; (iii) pegylated islets with nanoparticles (empty); and (iv) pegylated islets with nanoparticles loaded with a cargo of leukemia inhibitory factor (LIF), a factor both promotes adaptive immune tolerance and regulates pancreatic ß cell mass. Without any other treatment, normoglycemia was lost after 17 d (+/-7.5 d) in control group. In striking contrast, recipients in groups (ii), (iii), and (iv) showed long-term (>100 d) normoglycemia involving 30%; 43%, and 57% of the recipients in each respective group. In conclusion, construction of "stealth" islets by pegylation-based nanotherapeutics not only supports islet structure and function, but also effectively isolates the islets from immune-mediated destruction. The added value of nanoparticles to deliver immune modulators plus growth factors such as LIF expands the potential of this novel therapeutic approach to cell therapy for diabetes.


Subject(s)
Blood Glucose/metabolism , Major Histocompatibility Complex/immunology , Nanomedicine , Pancreas Transplantation , Pancreas/immunology , Polyethylene Glycols/administration & dosage , Animals , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Microscopy, Electron, Scanning
18.
Regen Med ; 7(6 Suppl): 41-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23210810

ABSTRACT

Diabetes mellitus represents a global epidemic affecting over 350 million patients worldwide and projected by the WHO to surpass the 500 million patient mark within the next two decades. Besides Type 1 and Type 2 diabetes mellitus, the study of the endocrine compartment of the pancreas is of great translational interest, as strategies aimed at restoring its mass could become therapies for glycemic dysregulation, drug-related diabetes following diabetogenic therapies, or hyperglycemic disturbances following the treatment of cancer and nesidioblastosis. Such strategies generally fall under one of the 'three Rs': replacement (islet transplantation and stem cell differentiation); reprogramming (e.g., from the exocrine compartment of the pancreas); and regeneration (replication and induction of endogenous stem cells). As the latter has been extensively reviewed in recent months by us and others, this article focuses on emerging reprogramming and replacement approaches.


Subject(s)
Cellular Reprogramming/genetics , Diabetes Mellitus/therapy , Regenerative Medicine/methods , Stem Cell Transplantation , Animals , Humans
19.
Stem Cells Transl Med ; 1(1): 59-63, 2012 Jan.
Article in English | MEDLINE | ID: mdl-23197641

ABSTRACT

Mesenchymal stem cells (MSCs) have already made their mark in the young field of regenerative medicine. Easily derived from many adult tissues, their therapeutic worth has already been validated for a number of conditions. Unlike embryonic stem cells, neither their procurement nor their use is deemed controversial. Here we review the potential use of MSCs for the treatment of type 1 diabetes mellitus, a devastating chronic disease in which the insulin-producing cells of the pancreas (the ß-cells) are the target of an autoimmune process. It has been hypothesized that stem cell-derived ß-cells may be used to replenish the islet mass in diabetic patients, making islet transplantation (a form of cell therapy that has already proven effective at clinically restoring normoglycemia) available to millions of prospective patients. Here we review the most current advances in the design and application of protocols for the differentiation of transplantable ß-cells, with a special emphasis in analyzing MSC potency according to their tissue of origin. Although no single method appears to be ripe enough for clinical trials yet, recent progress in reprogramming (a biotechnological breakthrough that relativizes the thus far insurmountable barriers between embryonal germ layers) bodes well for the rise of MSCs as a potential weapon of choice to develop personalized therapies for type 1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1/surgery , Insulin-Secreting Cells/transplantation , Islets of Langerhans Transplantation , Mesenchymal Stem Cell Transplantation , Regenerative Medicine , Animals , Cell Differentiation , Cell Lineage , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/physiopathology , Humans , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Regeneration
20.
Cold Spring Harb Protoc ; 2012(9): 962-8, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22949710

ABSTRACT

Our knowledge of organ ontogeny is largely based on loss-of-function (knockout) or gain-of-function (transgenesis) approaches. However, developmental modulators such as proteins, mRNAs, microRNAs(miRNAs), small interfering RNAs, and other small molecules may complement the above DNA-modifying technologies in a much more direct way. Unfortunately, their use is often limited by the ability of these compounds to cross the placenta and reach physiologically relevant concentrations when administered systemically to the mother. The design of safe and effective techniques to deliver these compounds into the embryo is therefore an area of great scientific potential. In this article we report a new method for introducing developmental modulators into murine embryos by means of direct injection into the heart. Unlike other reported methods that require surgical exposure of the uterus, our percutaneous ultrasound-guided approach allows for the intracardial injection of mouse embryos as early as embryonic day 10.5 (e10.5) and throughout gestation in a minimally invasive manner that largely preserves embryo viability. This system offers a critical advantage over in vitro settings because the effects of any given treatment can be observed without disturbing the native environment of the developing organ. Procedures are described for the delivery and detection of transducible proteins as well as morpholinos designed to block the expression of specific miRNAs within the living embryo.


Subject(s)
Developmental Biology/methods , Heart/drug effects , Heart/embryology , Injections/methods , Animals , Mice , Morpholinos/administration & dosage , Proteins/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...