Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Cancers (Basel) ; 13(22)2021 Nov 18.
Article in English | MEDLINE | ID: mdl-34830945

ABSTRACT

Advances in the treatment of pancreatic ductal adenocarcinoma (PDAC) using neoadjuvant chemoradiotherapy, chemotherapy, and immunotherapy have had minimal impact on the overall survival of patients. A general lack of immunogenic features and a complex tumor microenvironment (TME) are likely culprits for therapy refractoriness in PDAC. Induced pluripotent stem cells (iPSCs) should be explored as a means to advance the treatment options for PDAC, by providing representative in vitro models of pancreatic cancer development. In addition, iPSCs could be used for tailor-made cellular immunotherapies or as a source of tumor-associated antigens in the context of vaccination.

2.
Stem Cell Reports ; 16(6): 1468-1477, 2021 06 08.
Article in English | MEDLINE | ID: mdl-33961792

ABSTRACT

Induced pluripotent stem cells (iPSCs) and cancer cells share cellular similarities and transcriptomic profiles. Here, we show that an iPSC-based cancer vaccine, comprised of autologous iPSCs and CpG, stimulated cytotoxic antitumor CD8+ T cell effector and memory responses, induced cancer-specific humoral immune responses, reduced immunosuppressive CD4+ T regulatory cells, and prevented tumor formation in 75% of pancreatic ductal adenocarcinoma (PDAC) mice. We demonstrate that shared gene expression profiles of "iPSC-cancer signature genes" and others are overexpressed in mouse and human iPSC lines, PDAC cells, and multiple human solid tumor types compared with normal tissues. These results support further studies of iPSC vaccination in PDAC in preclinical and clinical models and in other cancer types that have low mutational burdens.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Carcinoma, Pancreatic Ductal/immunology , Induced Pluripotent Stem Cells/immunology , Pancreatic Neoplasms/immunology , T-Lymphocytopenia, Idiopathic CD4-Positive/immunology , Animals , Antineoplastic Agents/therapeutic use , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/metabolism , Cancer Vaccines/therapeutic use , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/therapy , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Immunologic Memory , Induced Pluripotent Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Pancreatic Neoplasms/therapy , T-Lymphocytopenia, Idiopathic CD4-Positive/metabolism , Transcriptome , Xenograft Model Antitumor Assays
3.
Stem Cells Dev ; 29(18): 1179-1189, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32723003

ABSTRACT

Pluripotent stem cells are promising candidates for cell-based regenerative therapies. To avoid rejection of transplanted cells, several approaches are being pursued to reduce immunogenicity of the cells or modulate the recipient's immune response. These include gene editing to reduce the antigenicity of cell products, immunosuppression of the host, or using major histocompatibility complex-matched cells from cell banks. In this context, we have investigated the antigenicity of H-Y antigens, a class of minor histocompatibility antigens encoded by the Y chromosome, to assess whether the gender of the donor affects the cell's antigenicity. In a murine transplant model, we show that the H-Y antigen in undifferentiated embryonic stem cells (ESCs), as well as ESC-derived endothelial cells, provokes T- and B cell responses in female recipients.


Subject(s)
Embryonic Stem Cells/metabolism , Graft Rejection/immunology , H-Y Antigen/metabolism , Animals , Animals, Newborn , B-Lymphocytes/immunology , Female , Immune Tolerance , Immunity , Male , Mice , Mice, Inbred BALB C , Stem Cell Transplantation , Survival Analysis , T-Lymphocytes/immunology
4.
Nat Biomed Eng ; 2(2): 104-113, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29721363

ABSTRACT

Stem-cell-based therapies hold considerable promise for regenerative medicine. However, acute donor-cell death within several weeks after cell delivery remains a critical hurdle for clinical translation. Co-transplantation of stem cells with pro-survival factors can improve cell engraftment, but this strategy has been hampered by the typically short half-lives of the factors and by the use of Matrigel and other scaffolds that are not chemically defined. Here, we report a collagen-dendrimer biomaterial crosslinked with pro-survival peptide analogues that adheres to the extracellular matrix and slowly releases the peptides, significantly prolonging stem cell survival in mouse models of ischaemic injury. The biomaterial can serve as a generic delivery system to improve functional outcomes in cell-replacement therapy.

5.
Cell Stem Cell ; 22(4): 501-513.e7, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29456158

ABSTRACT

Cancer cells and embryonic tissues share a number of cellular and molecular properties, suggesting that induced pluripotent stem cells (iPSCs) may be harnessed to elicit anti-tumor responses in cancer vaccines. RNA sequencing revealed that human and murine iPSCs express tumor-associated antigens, and we show here a proof of principle for using irradiated iPSCs in autologous anti-tumor vaccines. In a prophylactic setting, iPSC vaccines prevent tumor growth in syngeneic murine breast cancer, mesothelioma, and melanoma models. As an adjuvant, the iPSC vaccine inhibited melanoma recurrence at the resection site and reduced metastatic tumor load, which was associated with fewer Th17 cells and increased CD11b+GR1hi myeloid cells. Adoptive transfer of T cells isolated from vaccine-treated tumor-bearing mice inhibited tumor growth in unvaccinated recipients, indicating that the iPSC vaccine promotes an antigen-specific anti-tumor T cell response. Our data suggest an easy, generalizable strategy for multiple types of cancer that could prove highly valuable in clinical immunotherapy.


Subject(s)
Breast Neoplasms/immunology , Cancer Vaccines/immunology , Induced Pluripotent Stem Cells/immunology , Melanoma/immunology , Mesothelioma/immunology , Animals , Breast Neoplasms/therapy , Female , Humans , Induced Pluripotent Stem Cells/cytology , Melanoma/therapy , Mesothelioma/therapy , Mice
6.
Cell Rep ; 20(8): 1978-1990, 2017 Aug 22.
Article in English | MEDLINE | ID: mdl-28834758

ABSTRACT

There is growing interest in using embryonic stem cell (ESC) and induced pluripotent stem cell (iPSC) derivatives for tissue regeneration. However, an increased understanding of human immune responses to stem cell-derived allografts is necessary for maintaining long-term graft persistence. To model this alloimmunity, humanized mice engrafted with human hematopoietic and immune cells could prove to be useful. In this study, an in-depth analysis of graft-infiltrating human lymphocytes and splenocytes revealed that humanized mice incompletely model human immune responses toward allogeneic stem cells and their derivatives. Furthermore, using an "allogenized" mouse model, we show the feasibility of reconstituting immunodeficient mice with a functional mouse immune system and describe a key role of innate immune cells in the rejection of mouse stem cell allografts.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Immunity, Innate/immunology , Pluripotent Stem Cells/metabolism , Transplantation Conditioning/methods , Animals , Disease Models, Animal , Graft Rejection , Humans , Mice
7.
Circ Res ; 117(8): 720-30, 2015 Sep 25.
Article in English | MEDLINE | ID: mdl-26291556

ABSTRACT

RATIONALE: Tissue engineering approaches may improve survival and functional benefits from human embryonic stem cell-derived cardiomyocyte transplantation, thereby potentially preventing dilative remodeling and progression to heart failure. OBJECTIVE: Assessment of transport stability, long-term survival, structural organization, functional benefits, and teratoma risk of engineered heart muscle (EHM) in a chronic myocardial infarction model. METHODS AND RESULTS: We constructed EHMs from human embryonic stem cell-derived cardiomyocytes and released them for transatlantic shipping following predefined quality control criteria. Two days of shipment did not lead to adverse effects on cell viability or contractile performance of EHMs (n=3, P=0.83, P=0.87). One month after ischemia/reperfusion injury, EHMs were implanted onto immunocompromised rat hearts to simulate chronic ischemia. Bioluminescence imaging showed stable engraftment with no significant cell loss between week 2 and 12 (n=6, P=0.67), preserving ≤25% of the transplanted cells. Despite high engraftment rates and attenuated disease progression (change in ejection fraction for EHMs, -6.7±1.4% versus control, -10.9±1.5%; n>12; P=0.05), we observed no difference between EHMs containing viable and nonviable human cardiomyocytes in this chronic xenotransplantation model (n>12; P=0.41). Grafted cardiomyocytes showed enhanced sarcomere alignment and increased connexin 43 expression at 220 days after transplantation. No teratomas or tumors were found in any of the animals (n=14) used for long-term monitoring. CONCLUSIONS: EHM transplantation led to high engraftment rates, long-term survival, and progressive maturation of human cardiomyocytes. However, cell engraftment was not correlated with functional improvements in this chronic myocardial infarction model. Most importantly, the safety of this approach was demonstrated by the lack of tumor or teratoma formation.


Subject(s)
Embryonic Stem Cells/transplantation , Graft Survival , Heart Transplantation/methods , Myocardial Infarction/surgery , Myocytes, Cardiac/transplantation , Papillary Muscles/transplantation , Tissue Engineering/methods , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Line , Cell Survival , Connexin 43/metabolism , Disease Models, Animal , Embryonic Stem Cells/immunology , Embryonic Stem Cells/metabolism , Heart Transplantation/adverse effects , Heterografts , Humans , Immunosuppressive Agents/pharmacology , Male , Myocardial Contraction , Myocardial Infarction/immunology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Papillary Muscles/immunology , Papillary Muscles/metabolism , Papillary Muscles/pathology , Papillary Muscles/physiopathology , Rats, Nude , Rats, Sprague-Dawley , Stroke Volume , Time Factors , Transfection
8.
Curr Protoc Stem Cell Biol ; 32: 4A.8.1-4A.8.17, 2015 Feb 02.
Article in English | MEDLINE | ID: mdl-25640819

ABSTRACT

This unit describes protocols for evaluating the pluripotency of embryonic and induced pluripotent stem cells using a teratoma formation assay. Cells are prepared for injection and transplanted into immunodeficient mice at the gastrocnemius muscle, a site well suited for teratoma growth and surgical access. Teratomas that form from the cell transplants are explanted, fixed in paraformaldehyde, and embedded in paraffin. These preserved samples are sectioned, stained, and analyzed. Pluripotency of a cell line is confirmed by whether the teratoma contains tissues derived from each of the embryonic germ layers: endoderm, mesoderm, and ectoderm. Alternatively, explanted and fixed teratomas can be cryopreserved for immunohistochemistry, which allows for more detailed identification of specific tissue types present in the samples.


Subject(s)
Pluripotent Stem Cells/cytology , Stem Cell Research , Teratoma/pathology , Animals , Cryopreservation , Fluorescent Antibody Technique , Humans , Injections , Mice, Inbred NOD , Mice, SCID , Paraffin Embedding , Tissue Fixation
9.
Circ Res ; 116(7): e40-50, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-25654979

ABSTRACT

RATIONALE: The mechanism of functional restoration by stem cell therapy remains poorly understood. Novel manganese-enhanced MRI and bioluminescence reporter gene imaging were applied to follow myocardial viability and cell engraftment, respectively. Human-placenta-derived amniotic mesenchymal stem cells (AMCs) demonstrate unique immunoregulatory and precardiac properties. In this study, the restorative effects of 3 AMC-derived subpopulations were examined in a murine myocardial injury model: (1) unselected AMCs, (2) ckit(+)AMCs, and (3) AMC-derived induced pluripotent stem cells (MiPSCs). OBJECTIVE: To determine the differential restorative effects of the AMC-derived subpopulations in the murine myocardial injury model using multimodality imaging. METHODS AND RESULTS: SCID (severe combined immunodeficiency) mice underwent left anterior descending artery ligation and were divided into 4 treatment arms: (1) normal saline control (n=14), (2) unselected AMCs (n=10), (3) ckit(+)AMCs (n=13), and (4) MiPSCs (n=11). Cardiac MRI assessed myocardial viability and left ventricular function, whereas bioluminescence imaging assessed stem cell engraftment during a 4-week period. Immunohistological labeling and reverse transcriptase polymerase chain reaction of the explanted myocardium were performed. The unselected AMC and ckit(+)AMC-treated mice demonstrated transient left ventricular functional improvement. However, the MiPSCs exhibited a significantly greater increase in left ventricular function compared with all the other groups during the entire 4-week period. Left ventricular functional improvement correlated with increased myocardial viability and sustained stem cell engraftment. The MiPSC-treated animals lacked any evidence of de novo cardiac differentiation. CONCLUSION: The functional restoration seen in MiPSCs was characterized by increased myocardial viability and sustained engraftment without de novo cardiac differentiation, indicating salvage of the injured myocardium.


Subject(s)
Induced Pluripotent Stem Cells/transplantation , Magnetic Resonance Imaging/methods , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Multimodal Imaging , Myocardial Infarction/therapy , Myocardium/pathology , Animals , Cell Separation/methods , Cell Survival , Coronary Stenosis/complications , Disease Models, Animal , Female , Gene Expression Profiling , Genes, Reporter , Graft Survival , Heterografts , Humans , Ligation , Luminescent Measurements , Male , Manganese , Mesenchymal Stem Cells/chemistry , Mice , Mice, Mutant Strains , Mice, SCID , Myocardial Infarction/etiology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Placenta/cytology , Pregnancy , Proto-Oncogene Proteins c-kit/analysis , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Telomere Homeostasis , Ventricular Function, Left
10.
Sci Rep ; 5: 8081, 2015 Jan 28.
Article in English | MEDLINE | ID: mdl-25628230

ABSTRACT

The development of human induced pluripotent stem cell (iPSC) technology has revolutionized the regenerative medicine field. This technology provides a powerful tool for disease modeling and drug screening approaches. To circumvent the risk of random integration into the host genome caused by retroviruses, non-integrating reprogramming methods have been developed. However, these techniques are relatively inefficient or expensive. The mini-intronic plasmid (MIP) is an alternative, robust transgene expression vector for reprogramming. Here we developed a single plasmid reprogramming system which carries codon-optimized (Co) sequences of the canonical reprogramming factors (Oct4, Klf4, Sox2, and c-Myc) and short hairpin RNA against p53 ("4-in-1 CoMiP"). We have derived human and mouse iPSC lines from fibroblasts by performing a single transfection. Either independently or together with an additional vector encoding for LIN28, NANOG, and GFP, we were also able to reprogram blood-derived peripheral blood mononuclear cells (PBMCs) into iPSCs. Taken together, the CoMiP system offers a new highly efficient, integration-free, easy to use, and inexpensive methodology for reprogramming. Furthermore, the CoMIP construct is color-labeled, free of any antibiotic selection cassettes, and independent of the requirement for expression of the Epstein-Barr Virus nuclear antigen (EBNA), making it particularly beneficial for future applications in regenerative medicine.


Subject(s)
Cellular Reprogramming , Plasmids/metabolism , Pluripotent Stem Cells/cytology , Animals , Cells, Cultured , Codon , Epstein-Barr Virus Nuclear Antigens/genetics , Epstein-Barr Virus Nuclear Antigens/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunity, Innate , Introns , Karyotyping , Kruppel-Like Factor 4 , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Mice , Microscopy, Fluorescence , Nanog Homeobox Protein , Plasmids/genetics , Pluripotent Stem Cells/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Small Interfering/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics
11.
Eur Heart J ; 36(13): 806-16, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25368203

ABSTRACT

AIMS: High-fat diet-induced obesity (DIO) is a major contributor to type II diabetes and micro- and macro-vascular complications leading to peripheral vascular disease (PVD). Metabolic abnormalities of induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) from obese individuals could potentially limit their therapeutic efficacy for PVD. The aim of this study was to compare the function of iPSC-ECs from normal and DIO mice using comprehensive in vitro and in vivo assays. METHODS AND RESULTS: Six-week-old C57Bl/6 mice were fed with a normal or high-fat diet. At 24 weeks, iPSCs were generated from tail tip fibroblasts and differentiated into iPSC-ECs using a directed monolayer approach. In vitro functional analysis revealed that iPSC-ECs from DIO mice had significantly decreased capacity to form capillary-like networks, diminished migration, and lower proliferation. Microarray and ELISA confirmed elevated apoptotic, inflammatory, and oxidative stress pathways in DIO iPSC-ECs. Following hindlimb ischaemia, mice receiving intramuscular injections of DIO iPSC-ECs had significantly decreased reperfusion compared with mice injected with control healthy iPSC-ECs. Hindlimb sections revealed increased muscle atrophy and presence of inflammatory cells in mice receiving DIO iPSC-ECs. When pravastatin was co-administered to mice receiving DIO iPSC-ECs, a significant increase in reperfusion was observed; however, this beneficial effect was blunted by co-administration of the nitric oxide synthase inhibitor, N(ω)-nitro-l-arginine methyl ester. CONCLUSION: This is the first study to provide evidence that iPSC-ECs from DIO mice exhibit signs of endothelial dysfunction and have suboptimal efficacy following transplantation in a hindlimb ischaemia model. These findings may have important implications for future treatment of PVD using iPSC-ECs in the obese population.


Subject(s)
Endothelial Cells/physiology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Induced Pluripotent Stem Cells/physiology , Nitric Oxide/physiology , Obesity/physiopathology , Pravastatin/pharmacology , Analysis of Variance , Animals , Apoptosis/physiology , Cell Differentiation , Diet, High-Fat , Enzyme Inhibitors/pharmacology , Fibroblasts/physiology , Hindlimb/blood supply , Injections, Intramuscular , Ischemia/physiopathology , Ischemia/prevention & control , Mice, Inbred C57BL , Muscle, Skeletal , Muscular Diseases/prevention & control , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress/physiology , Proto-Oncogene Proteins c-akt/metabolism , Random Allocation , Reperfusion Injury/physiopathology , Signal Transduction
12.
Methods Mol Biol ; 1181: 1-13, 2014.
Article in English | MEDLINE | ID: mdl-25070322

ABSTRACT

The ability to induce pluripotency in somatic cells is one of the most important scientific achievements in the fields of stem cell research and regenerative medicine. This technique allows researchers to obtain pluripotent stem cells without the controversial use of embryos, providing a novel and powerful tool for disease modeling and drug screening approaches. However, using viruses for the delivery of reprogramming genes and transcription factors may result in integration into the host genome and cause random mutations within the target cell, thus limiting the use of these cells for downstream applications. To overcome this limitation, various non-integrating techniques, including Sendai virus, mRNA, minicircle, and plasmid-based methods, have recently been developed. Utilizing a newly developed codon optimized 4-in-1 minicircle (CoMiC), we were able to reprogram human adult fibroblasts using chemically defined media and without the need for feeder cells.


Subject(s)
Cellular Reprogramming , Codon/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Genetic Techniques , Plasmids/genetics , Adult , Biopsy , Cell Separation , Collagen/pharmacology , Drug Combinations , Fibroblasts/drug effects , Fluorescent Antibody Technique , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Laminin/pharmacology , Proteoglycans/pharmacology , Skin/pathology , Transfection
13.
Nat Commun ; 5: 3903, 2014 May 30.
Article in English | MEDLINE | ID: mdl-24875164

ABSTRACT

The exact nature of the immune response elicited by autologous-induced pluripotent stem cell (iPSC) progeny is still not well understood. Here we show in murine models that autologous iPSC-derived endothelial cells (iECs) elicit an immune response that resembles the one against a comparable somatic cell, the aortic endothelial cell (AEC). These cells exhibit long-term survival in vivo and prompt a tolerogenic immune response characterized by elevated IL-10 expression. In contrast, undifferentiated iPSCs elicit a very different immune response with high lymphocytic infiltration and elevated IFN-γ, granzyme-B and perforin intragraft. Furthermore, the clonal structure of infiltrating T cells from iEC grafts is statistically indistinguishable from that of AECs, but is different from that of undifferentiated iPSC grafts. Taken together, our results indicate that the differentiation of iPSCs results in a loss of immunogenicity and leads to the induction of tolerance, despite expected antigen expression differences between iPSC-derived versus original somatic cells.


Subject(s)
Cell Differentiation/immunology , Endothelial Cells/immunology , Graft Rejection/immunology , Immune Tolerance/immunology , Induced Pluripotent Stem Cells/transplantation , Self Tolerance/immunology , Animals , Aorta/cytology , Cells, Cultured , Endothelial Cells/cytology , Graft Survival , Granzymes/immunology , Induced Pluripotent Stem Cells/immunology , Interleukin-10/immunology , Mice , Perforin/immunology
15.
Hum Gene Ther ; 25(1): 41-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24090375

ABSTRACT

In this study, we target the hypoxia inducible factor-1 alpha (HIF-1-alpha) pathway by short hairpin RNA interference therapy targeting prolyl hydroxylase-2 (shPHD2). We use the minicircle (MC) vector technology as an alternative for conventional nonviral plasmid (PL) vectors in order to improve neovascularization after unilateral hindlimb ischemia in a murine model. Gene expression and transfection efficiency of MC and PL, both in vitro and in vivo, were assessed using bioluminescence imaging (BLI) and firefly luciferase (Luc) reporter gene. C57Bl6 mice underwent unilateral electrocoagulation of the femoral artery and gastrocnemic muscle injection with MC-shPHD2, PL-shPHD2, or phosphate-buffered saline (PBS) as control. Blood flow recovery was monitored using laser Doppler perfusion imaging, and collaterals were visualized by immunohistochemistry and angiography. MC-Luc showed a 4.6-fold higher in vitro BLI signal compared with PL-Luc. BLI signals in vivo were 4.3×10(5)±3.3×10(5) (MC-Luc) versus 0.4×10(5)±0.3×10(5) (PL-Luc) at day 28 (p=0.016). Compared with PL-shPHD2 or PBS, MC-shPHD2 significantly improved blood flow recovery, up to 50% from day 3 until day 14 after ischemia induction. MC-shPHD2 significantly increased collateral density and capillary density, as monitored by alpha-smooth muscle actin expression and CD31(+) expression, respectively. Angiography data confirmed the histological findings. Significant downregulation of PHD2 mRNA levels by MC-shPHD2 was confirmed by quantitative polymerase chain reaction. Finally, Western blot analysis confirmed significantly higher levels of HIF-1-alpha protein by MC-shPHD2, compared with PL-shPHD2 and PBS. This study provides initial evidence of a new potential therapeutic approach for peripheral artery disease. The combination of HIF-1-alpha pathway targeting by shPHD2 with the robust nonviral MC plasmid improved postischemic neovascularization, making this approach a promising potential treatment option for critical limb ischemia.


Subject(s)
Gene Silencing , Genetic Vectors/genetics , Hindlimb/blood supply , Hindlimb/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Ischemia/genetics , Neovascularization, Physiologic/genetics , RNA, Small Interfering/genetics , Angiography , Animals , Cell Line , Gene Expression , Gene Transfer Techniques , Genes, Reporter , Genetic Therapy , Genetic Vectors/administration & dosage , Genetic Vectors/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Ischemia/metabolism , Ischemia/therapy , Mice , Plasmids/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism
16.
Stem Cells ; 31(11): 2354-63, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24038578

ABSTRACT

RATIONALE: Human embryonic stem cell (hESC) derivatives are attractive candidates for therapeutic use. The engraftment and survival of hESC derivatives as xenografts or allografts require effective immunosuppression to prevent immune cell infiltration and graft destruction. OBJECTIVE: To test the hypothesis that a short-course, dual-agent regimen of two costimulation-adhesion blockade agents can induce better engraftment of hESC derivatives compared to current immunosuppressive agents. METHODS AND RESULTS: We transduced hESCs with a double fusion reporter gene construct expressing firefly luciferase (Fluc) and enhanced green fluorescent protein, and differentiated these cells to endothelial cells (hESC-ECs). Reporter gene expression enabled longitudinal assessment of cell engraftment by bioluminescence imaging. Costimulation-adhesion therapy resulted in superior hESC-EC and mouse EC engraftment compared to cyclosporine therapy in a hind limb model. Costimulation-adhesion therapy also promoted robust hESC-EC and hESC-derived cardiomyocyte survival in an ischemic myocardial injury model. Improved hESC-EC engraftment had a cardioprotective effect after myocardial injury, as assessed by magnetic resonance imaging. Mechanistically, costimulation-adhesion therapy is associated with systemic and intragraft upregulation of T-cell immunoglobulin and mucin domain 3 (TIM3) and a reduced proinflammatory cytokine profile. CONCLUSIONS: Costimulation-adhesion therapy is a superior alternative to current clinical immunosuppressive strategies for preventing the post-transplant rejection of hESC derivatives. By extending the window for cellular engraftment, costimulation-adhesion therapy enhances functional preservation following ischemic injury. This regimen may function through a TIM3-dependent mechanism.


Subject(s)
Antibodies, Monoclonal/pharmacology , Cyclosporine/pharmacology , Embryonic Stem Cells/transplantation , Graft Rejection/prevention & control , Immunoconjugates/pharmacology , Immunosuppressive Agents/pharmacology , Prednisone/pharmacology , Abatacept , Animals , Cardiotonic Agents/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/immunology , Endothelial Cells/immunology , Endothelial Cells/transplantation , Graft Rejection/immunology , Humans , Immune Tolerance , Immunosuppression Therapy/methods , Mice , Mice, Inbred NOD , Mice, SCID , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardial Infarction/surgery , Random Allocation
17.
Liver Transpl ; 18(1): 112-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21987454

ABSTRACT

In Eurotransplant, more than 50% of liver allografts come from extended criteria donors (ECDs). However, not every ECD is the same. The limits of their use are being explored. A continuous scoring system for analyzing donor risk has been developed within the Organ Procurement and Transplantation Network (OPTN), the Donor Risk Index (DRI). The objective of this study was the validation of this donor risk index (DRI) in Eurotransplant. The study was a database analysis of all 5939 liver transplants involving deceased donors and adult recipients from January 1, 2003 to December 31, 2007 in Eurotransplant. Data were analyzed with Kaplan-Meier and Cox regression models. Follow-up data were available for 5723 patients with a median follow up of 2.5 years. The mean DRI was remarkably higher in the Eurotransplant region versus OPTN (1.71 versus 1.45), and this indicated different donor populations. Nevertheless, we were able to validate the DRI for the Eurotransplant region. Kaplan-Meier curves per DRI category showed a significant correlation between the DRI and outcomes (P < 0.001). A multivariate analysis demonstrated that the DRI was the most significant factor influencing outcomes (P < 0.001). Among all donor, transplant, and recipient variables, the DRI was the strongest predictor of outcomes.


Subject(s)
Liver Diseases/surgery , Liver Transplantation/standards , Tissue Donors , Tissue and Organ Procurement/standards , Adult , Aged , Europe , Female , Follow-Up Studies , Humans , Kaplan-Meier Estimate , Liver Transplantation/mortality , Male , Middle Aged , Multivariate Analysis , Proportional Hazards Models , Retrospective Studies , Risk Factors , Treatment Outcome
18.
J R Soc Interface ; 7 Suppl 6: S753-63, 2010 Dec 06.
Article in English | MEDLINE | ID: mdl-20880852

ABSTRACT

Embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) have the ability (i) to duplicate indefinitely while maintaining pluripotency and (ii) to differentiate into cell types of all three embryonic germ layers. These two properties of ESCs and iPSCs make them potentially suitable for tissue engineering and cell replacement therapy for many different diseases, including Parkinson's disease, diabetes and heart disease. However, one critical obstacle in the clinical application of ESCs or iPSCs is the risk of teratoma formation. The emerging field of molecular imaging is allowing researchers to track transplanted ESCs or iPSCs in vivo, enabling early detection of teratomas.


Subject(s)
Embryonic Stem Cells/pathology , Induced Pluripotent Stem Cells/pathology , Teratoma/pathology , Animals , Cell Differentiation , Humans , Mice , Molecular Imaging/methods , Regenerative Medicine , Tissue Engineering
SELECTION OF CITATIONS
SEARCH DETAIL
...