Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 200
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 509(7502): 641-4, 2014 May 29.
Article in English | MEDLINE | ID: mdl-24747400

ABSTRACT

Despite the introduction of antiproliferative drug-eluting stents, coronary heart disease remains the leading cause of death in the United States. In-stent restenosis and bypass graft failure are characterized by excessive smooth muscle cell (SMC) proliferation and concomitant myointima formation with luminal obliteration. Here we show that during the development of myointimal hyperplasia in human arteries, SMCs show hyperpolarization of their mitochondrial membrane potential (ΔΨm) and acquire a temporary state with a high proliferative rate and resistance to apoptosis. Pyruvate dehydrogenase kinase isoform 2 (PDK2) was identified as a key regulatory protein, and its activation proved necessary for relevant myointima formation. Pharmacologic PDK2 blockade with dichloroacetate or lentiviral PDK2 knockdown prevented ΔΨm hyperpolarization, facilitated apoptosis and reduced myointima formation in injured human mammary and coronary arteries, rat aortas, rabbit iliac arteries and swine (pig) coronary arteries. In contrast to several commonly used antiproliferative drugs, dichloroacetate did not prevent vessel re-endothelialization. Targeting myointimal ΔΨm and alleviating apoptosis resistance is a novel strategy for the prevention of proliferative vascular diseases.


Subject(s)
Aorta/injuries , Arteries/injuries , Constriction, Pathologic/prevention & control , Dichloroacetic Acid/pharmacology , Dichloroacetic Acid/therapeutic use , Tunica Intima/drug effects , Tunica Intima/pathology , Angioplasty, Balloon/adverse effects , Animals , Aorta/drug effects , Aorta/pathology , Apoptosis/drug effects , Arteries/drug effects , Arteries/pathology , Cell Proliferation/drug effects , Constriction, Pathologic/pathology , Coronary Vessels/drug effects , Coronary Vessels/injuries , Coronary Vessels/pathology , Disease Models, Animal , Enzyme Activation/drug effects , Gene Knockdown Techniques , Humans , Hyperplasia/drug therapy , Hyperplasia/pathology , Iliac Artery/drug effects , Iliac Artery/injuries , Iliac Artery/pathology , Mammary Arteries/drug effects , Mammary Arteries/injuries , Mammary Arteries/pathology , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Rabbits , Rats , Secondary Prevention , Stents/adverse effects , Swine , Tunica Intima/injuries
2.
Radiology ; 280(3): 826-36, 2016 09.
Article in English | MEDLINE | ID: mdl-27332865

ABSTRACT

Purpose To quantitatively determine the limit of detection of marrow stromal cells (MSC) after cardiac cell therapy (CCT) in swine by using clinical positron emission tomography (PET) reporter gene imaging and magnetic resonance (MR) imaging with cell prelabeling. Materials and Methods Animal studies were approved by the institutional administrative panel on laboratory animal care. Seven swine received 23 intracardiac cell injections that contained control MSC and cell mixtures of MSC expressing a multimodality triple fusion (TF) reporter gene (MSC-TF) and bearing superparamagnetic iron oxide nanoparticles (NP) (MSC-TF-NP) or NP alone. Clinical MR imaging and PET reporter gene molecular imaging were performed after intravenous injection of the radiotracer fluorine 18-radiolabeled 9-[4-fluoro-3-(hydroxyl methyl) butyl] guanine ((18)F-FHBG). Linear regression analysis of both MR imaging and PET data and nonlinear regression analysis of PET data were performed, accounting for multiple injections per animal. Results MR imaging showed a positive correlation between MSC-TF-NP cell number and dephasing (dark) signal (R(2) = 0.72, P = .0001) and a lower detection limit of at least approximately 1.5 × 10(7) cells. PET reporter gene imaging demonstrated a significant positive correlation between MSC-TF and target-to-background ratio with the linear model (R(2) = 0.88, P = .0001, root mean square error = 0.523) and the nonlinear model (R(2) = 0.99, P = .0001, root mean square error = 0.273) and a lower detection limit of 2.5 × 10(8) cells. Conclusion The authors quantitatively determined the limit of detection of MSC after CCT in swine by using clinical PET reporter gene imaging and clinical MR imaging with cell prelabeling. (©) RSNA, 2016 Online supplemental material is available for this article.


Subject(s)
Genes, Reporter , Heart/diagnostic imaging , Mesenchymal Stem Cell Transplantation , Molecular Imaging/methods , Multimodal Imaging/methods , Animals , Fluorine Radioisotopes , Guanine/analogs & derivatives , Magnetic Resonance Imaging , Positron Emission Tomography Computed Tomography , Radiopharmaceuticals , Swine
3.
Radiology ; 280(3): 815-25, 2016 09.
Article in English | MEDLINE | ID: mdl-27308957

ABSTRACT

Purpose To use multimodality reporter-gene imaging to assess the serial survival of marrow stromal cells (MSC) after therapy for myocardial infarction (MI) and to determine if the requisite preclinical imaging end point was met prior to a follow-up large-animal MSC imaging study. Materials and Methods Animal studies were approved by the Institutional Administrative Panel on Laboratory Animal Care. Mice (n = 19) that had experienced MI were injected with bone marrow-derived MSC that expressed a multimodality triple fusion (TF) reporter gene. The TF reporter gene (fluc2-egfp-sr39ttk) consisted of a human promoter, ubiquitin, driving firefly luciferase 2 (fluc2), enhanced green fluorescent protein (egfp), and the sr39tk positron emission tomography reporter gene. Serial bioluminescence imaging of MSC-TF and ex vivo luciferase assays were performed. Correlations were analyzed with the Pearson product-moment correlation, and serial imaging results were analyzed with a mixed-effects regression model. Results Analysis of the MSC-TF after cardiac cell therapy showed significantly lower signal on days 8 and 14 than on day 2 (P = .011 and P = .001, respectively). MSC-TF with MI demonstrated significantly higher signal than MSC-TF without MI at days 4, 8, and 14 (P = .016). Ex vivo luciferase activity assay confirmed the presence of MSC-TF on days 8 and 14 after MI. Conclusion Multimodality reporter-gene imaging was successfully used to assess serial MSC survival after therapy for MI, and it was determined that the requisite preclinical imaging end point, 14 days of MSC survival, was met prior to a follow-up large-animal MSC study. (©) RSNA, 2016 Online supplemental material is available for this article.


Subject(s)
Genes, Reporter , Mesenchymal Stem Cell Transplantation/methods , Molecular Imaging , Multimodal Imaging , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/therapy , Animals , Female , Luciferases, Firefly/metabolism , Luminescent Measurements , Mice , Mice, Nude , Positron-Emission Tomography , Transfection
4.
Arterioscler Thromb Vasc Biol ; 35(9): 1945-53, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26183619

ABSTRACT

OBJECTIVE: Despite advances in stent technology for vascular interventions, in-stent restenosis (ISR) because of myointimal hyperplasia remains a major complication. APPROACH AND RESULTS: We investigated the regulatory role of microRNAs in myointimal hyperplasia/ISR, using a humanized animal model in which balloon-injured human internal mammary arteries with or without stenting were transplanted into Rowett nude rats, followed by microRNA profiling. miR-21 was the only significantly upregulated candidate. In addition, miR-21 expression was increased in human tissue samples from patients with ISR compared with coronary artery disease specimen. We systemically repressed miR-21 via intravenous fluorescein-tagged-locked nucleic acid-anti-miR-21 (anti-21) in our humanized myointimal hyperplasia model. As expected, suppression of vascular miR-21 correlated dose dependently with reduced luminal obliteration. Furthermore, anti-21 did not impede reendothelialization. However, systemic anti-miR-21 had substantial off-target effects, lowering miR-21 expression in liver, heart, lung, and kidney with concomitant increase in serum creatinine levels. We therefore assessed the feasibility of local miR-21 suppression using anti-21-coated stents. Compared with bare-metal stents, anti-21-coated stents effectively reduced ISR, whereas no significant off-target effects could be observed. CONCLUSION: This study demonstrates the efficacy of an anti-miR-coated stent for the reduction of ISR.


Subject(s)
Antibodies, Antinuclear/pharmacology , Coated Materials, Biocompatible , Coronary Restenosis/prevention & control , Gene Expression Regulation , Graft Occlusion, Vascular/prevention & control , MicroRNAs/genetics , Animals , Cell Proliferation/drug effects , Coronary Restenosis/genetics , Coronary Restenosis/metabolism , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Coronary Vessels/ultrastructure , Disease Models, Animal , Drug-Eluting Stents , Female , Graft Occlusion, Vascular/genetics , Graft Occlusion, Vascular/metabolism , Humans , Male , MicroRNAs/biosynthesis , MicroRNAs/immunology , Microscopy, Electron, Scanning , Middle Aged , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/ultrastructure , Neointima/metabolism , Neointima/pathology , Prosthesis Design , Rats , Rats, Nude
5.
Transpl Int ; 29(12): 1337-1348, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27614085

ABSTRACT

Bronchiolitis obliterans syndrome (BOS) is a main cause of allograft dysfunction and mortality after lung transplantation (LTx). A better understanding of BOS pathogenesis is needed to overcome this treatment-refractory complication. Orthotopic tracheal transplantation using human bronchus was performed in Brown Norway (BN) and nude (RNU) rats. Allografts were recovered in both strains at Day 7 (BN7 , n = 6; RNU7 , n = 7) or Day 28 (BN28 , n = 6; RNU28 , n = 6). Immune response of the host against the bronchial graft was assessed. Human samples from BOS patients were used to compare with the histological features of the animal model. Obstruction of the allograft lumen associated with significant infiltration of CD3+ and CD68+ cells was observed in the BN group on Day 28. Immune response from type 1 T-helper cells against the tracheal xenograft was higher in BN animals compared to nude animals on Days 7 and 28 (P < 0.001 and P = 0.035). Xenoreactive antibodies were significantly higher at Day 7 (IgM) and Day 28 (IgG) in the BN group compared to RNU (respectively, 37.6 ± 6.5 vs. 5.8 ± 0.7 mean fluorescence, P = 0.039; and 22.4 ± 3.8 vs. 6.9 ± 1.6 mean fluorescence, P = 0.011). Immunocompetent animals showed a higher infiltration of S100A4+ cells inside the bronchial wall after 28 days, associated with cartilage damage ranging from invasion to complete destruction. In vitro expression of S100A4 by human fibroblasts was higher when stimulated by mononuclear cells (MNCs) from BN rats than from RNU (2.9 ± 0.1 vs. 2.4 ± 0.1 mean fluorescence intensity, P = 0.005). Similarly, S100A4 was highly expressed in response to human MNCs compared to stimulation by T-cell-depleted human MNCs (4.3 ± 0.2 vs. 2.7 ± 0.1 mean fluorescence intensity, P < 0.001). Obliterative bronchiolitis has been induced in a new xenotransplant model in which chronic airway obstruction was associated with immune activation against the xenograft. Cartilage infiltration by S100A4+ cells might be stimulated by T cells.


Subject(s)
Bronchi/transplantation , Bronchiolitis Obliterans/etiology , Trachea/transplantation , Transplantation, Heterologous , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , CD3 Complex/metabolism , Disease Models, Animal , Enzyme-Linked Immunospot Assay , Graft Rejection/pathology , Graft Survival , Humans , Immune System , Liver Transplantation , Postoperative Period , Random Allocation , Rats , Rats, Inbred BN , S100 Calcium-Binding Protein A4/metabolism , Treatment Outcome
6.
J Mol Cell Cardiol ; 84: 13-23, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25871831

ABSTRACT

Valvular and vascular calcification are common causes of cardiovascular morbidity and mortality. Developing effective treatments requires understanding the molecular underpinnings of these processes. Shear stress is thought to play a role in inhibiting calcification. Furthermore, NOTCH1 regulates vascular and valvular endothelium, and human mutations in NOTCH1 can cause calcific aortic valve disease. Here, we determined the genome-wide impact of altering shear stress and NOTCH signaling on human aortic valve endothelium. mRNA-sequencing of primary human aortic valve endothelial cells (HAVECs) with or without knockdown of NOTCH1, in the presence or absence of shear stress, revealed NOTCH1-dependency of the atherosclerosis-related gene connexin 40 (GJA5), and numerous repressors of endochondral ossification. Among these, matrix gla protein (MGP) is highly expressed in aortic valve and vasculature, and inhibits soft tissue calcification by sequestering bone morphogenetic proteins (BMPs). Altering NOTCH1 levels affected MGP mRNA and protein in HAVECs. Furthermore, shear stress activated NOTCH signaling and MGP in a NOTCH1-dependent manner. NOTCH1 positively regulated endothelial MGP in vivo through specific binding motifs upstream of MGP. Our studies suggest that shear stress activates NOTCH1 in primary human aortic valve endothelial cells leading to downregulation of osteoblast-like gene networks that play a role in tissue calcification.


Subject(s)
Aortic Valve Stenosis/genetics , Aortic Valve/pathology , Calcinosis/genetics , Calcium-Binding Proteins/metabolism , Endothelium, Vascular/metabolism , Extracellular Matrix Proteins/metabolism , Gene Regulatory Networks , Receptor, Notch1/metabolism , Aortic Valve Stenosis/pathology , Calcinosis/pathology , Chromatin Immunoprecipitation , Cluster Analysis , DNA/metabolism , Endothelial Cells/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression Regulation , Genome, Human , Humans , Protein Binding , Rheology , Sequence Analysis, RNA , Signal Transduction/genetics , Stress, Mechanical , Matrix Gla Protein
7.
Circulation ; 127(25): 2503-13, 2013 Jun 25.
Article in English | MEDLINE | ID: mdl-23697910

ABSTRACT

BACKGROUND: Lung transplantation and heart-lung transplantation represent surgical options for treatment of medically refractory idiopathic pulmonary arterial hypertension. The effect of the lung allocation score on wait-list and transplantation outcomes in patients with idiopathic pulmonary arterial hypertension is poorly described. METHODS AND RESULTS: Adults diagnosed with idiopathic pulmonary arterial hypertension and listed for transplantation in the 80 months before and after the lung allocation score algorithm was implemented (n=1430) were identified in the United Network for Organ Sharing thoracic registry. Patients were stratified by organ listed and pre- and post-lung allocation score era. The cumulative incidences of transplantation and mortality for wait-listed patients in both eras were appraised with competing outcomes analysis. Posttransplantation survival was assessed with the Kaplan-Meier method. These analyses were repeated in propensity-matched subgroups. Cox proportional hazards analysis evaluated the effect of prelisting and pretransplantation characteristics on mortality. We found that patients in the post-lung allocation score era had significantly worse comorbidities; nevertheless, both lung transplantation and heart-lung transplantation candidates in this era enjoyed lower wait-list mortality and a higher incidence of transplantation in unmatched and propensity-matched analyses. On multivariable analysis, heart-lung transplantation and double-lung transplantation were associated with improved survival from the time of wait-listing, as was being listed at a medium- to high-volume institution. Donor/recipient sex matching predicted posttransplantation survival. CONCLUSIONS: The incidence of transplantation has increased while wait-list mortality has decreased in patients with idiopathic pulmonary arterial hypertension wait-listed for transplantation in the post-lung allocation score era. Both heart-lung transplantation and double-lung transplantation are predictive of survival in transplantation candidates with idiopathic pulmonary arterial hypertension, as is being listed at a medium- to high-volume institution. Donor/recipient sex matching is associated with better posttransplantation survival.


Subject(s)
Health Care Rationing/trends , Heart Transplantation , Hypertension, Pulmonary/mortality , Hypertension, Pulmonary/surgery , Lung Transplantation , Tissue and Organ Procurement/methods , Waiting Lists/mortality , Adult , Algorithms , Familial Primary Pulmonary Hypertension , Female , Heart Transplantation/statistics & numerical data , Humans , Incidence , Kaplan-Meier Estimate , Lung Transplantation/statistics & numerical data , Male , Middle Aged , Proportional Hazards Models , Registries , Retrospective Studies , Survival Rate , Time Factors , Treatment Outcome
8.
Circulation ; 127(16): 1677-91, 2013 Apr 23.
Article in English | MEDLINE | ID: mdl-23519760

ABSTRACT

BACKGROUND: Cardiotoxicity is a leading cause for drug attrition during pharmaceutical development and has resulted in numerous preventable patient deaths. Incidents of adverse cardiac drug reactions are more common in patients with preexisting heart disease than the general population. Here we generated a library of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) from patients with various hereditary cardiac disorders to model differences in cardiac drug toxicity susceptibility for patients of different genetic backgrounds. METHODS AND RESULTS: Action potential duration and drug-induced arrhythmia were measured at the single cell level in hiPSC-CMs derived from healthy subjects and patients with hereditary long QT syndrome, familial hypertrophic cardiomyopathy, and familial dilated cardiomyopathy. Disease phenotypes were verified in long QT syndrome, hypertrophic cardiomyopathy, and dilated cardiomyopathy hiPSC-CMs by immunostaining and single cell patch clamp. Human embryonic stem cell-derived cardiomyocytes (hESC-CMs) and the human ether-a-go-go-related gene expressing human embryonic kidney cells were used as controls. Single cell PCR confirmed expression of all cardiac ion channels in patient-specific hiPSC-CMs as well as hESC-CMs, but not in human embryonic kidney cells. Disease-specific hiPSC-CMs demonstrated increased susceptibility to known cardiotoxic drugs as measured by action potential duration and quantification of drug-induced arrhythmias such as early afterdepolarizations and delayed afterdepolarizations. CONCLUSIONS: We have recapitulated drug-induced cardiotoxicity profiles for healthy subjects, long QT syndrome, hypertrophic cardiomyopathy, and dilated cardiomyopathy patients at the single cell level for the first time. Our data indicate that healthy and diseased individuals exhibit different susceptibilities to cardiotoxic drugs and that use of disease-specific hiPSC-CMs may predict adverse drug responses more accurately than the standard human ether-a-go-go-related gene test or healthy control hiPSC-CM/hESC-CM screening assays.


Subject(s)
Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Hypertrophic, Familial/genetics , Drug Evaluation, Preclinical/methods , Drug-Related Side Effects and Adverse Reactions/genetics , Genetic Predisposition to Disease , Induced Pluripotent Stem Cells/cytology , Long QT Syndrome/genetics , Myocytes, Cardiac/drug effects , Action Potentials/drug effects , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Hypertrophic, Familial/pathology , Cell Differentiation , Cell Line/drug effects , Cell Line/physiology , Cell Size , Cisapride/toxicity , Embryoid Bodies/drug effects , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Gene Expression Profiling , HEK293 Cells/drug effects , HEK293 Cells/physiology , Humans , In Vitro Techniques , Ion Channels/biosynthesis , Ion Channels/genetics , Kidney/cytology , Kidney/embryology , Long QT Syndrome/pathology , Myocytes, Cardiac/physiology , Myofibrils/ultrastructure , Nicorandil/toxicity , Patch-Clamp Techniques , Quinazolines/toxicity , Verapamil/toxicity
9.
Circulation ; 128(11 Suppl 1): S3-13, 2013 Sep 10.
Article in English | MEDLINE | ID: mdl-24030418

ABSTRACT

BACKGROUND: Drug-induced arrhythmia is one of the most common causes of drug development failure and withdrawal from market. This study tested whether human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) combined with a low-impedance microelectrode array (MEA) system could improve on industry-standard preclinical cardiotoxicity screening methods, identify the effects of well-characterized drugs, and elucidate underlying risk factors for drug-induced arrhythmia. hiPSC-CMs may be advantageous over immortalized cell lines because they possess similar functional characteristics as primary human cardiomyocytes and can be generated in unlimited quantities. METHODS AND RESULTS: Pharmacological responses of beating embryoid bodies exposed to a comprehensive panel of drugs at 65 to 95 days postinduction were determined. Responses of hiPSC-CMs to drugs were qualitatively and quantitatively consistent with the reported drug effects in literature. Torsadogenic hERG blockers, such as sotalol and quinidine, produced statistically and physiologically significant effects, consistent with patch-clamp studies, on human embryonic stem cell-derived cardiomyocytes hESC-CMs. False-negative and false-positive hERG blockers were identified accurately. Consistent with published studies using animal models, early afterdepolarizations and ectopic beats were observed in 33% and 40% of embryoid bodies treated with sotalol and quinidine, respectively, compared with negligible early afterdepolarizations and ectopic beats in untreated controls. CONCLUSIONS: We found that drug-induced arrhythmias can be recapitulated in hiPSC-CMs and documented with low impedance MEA. Our data indicate that the MEA/hiPSC-CM assay is a sensitive, robust, and efficient platform for testing drug effectiveness and for arrhythmia screening. This system may hold great potential for reducing drug development costs and may provide significant advantages over current industry standard assays that use immortalized cell lines or animal models.


Subject(s)
Arrhythmias, Cardiac/chemically induced , Cardiovascular Agents/adverse effects , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Tissue Array Analysis/methods , Action Potentials/drug effects , Action Potentials/physiology , Adolescent , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/physiopathology , Cells, Cultured , Drug Evaluation, Preclinical/methods , Electric Impedance , Humans , Induced Pluripotent Stem Cells/physiology , Male , Microelectrodes , Myocytes, Cardiac/physiology
10.
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
11.
Circ Res ; 110(2): 312-24, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22116819

ABSTRACT

RATIONALE: Marfan syndrome (MFS) is a systemic connective tissue disorder notable for the development of aortic root aneurysms and the subsequent life-threatening complications of aortic dissection and rupture. Underlying fibrillin-1 gene mutations cause increased transforming growth factor-ß (TGF-ß) signaling. Although TGF-ß blockade prevents aneurysms in MFS mouse models, the mechanisms through which excessive TGF-ß causes aneurysms remain ill-defined. OBJECTIVE: We investigated the role of microRNA-29b (miR-29b) in aneurysm formation in MFS. METHODS AND RESULTS: Using quantitative polymerase chain reaction, we discovered that miR-29b, a microRNA regulating apoptosis and extracellular matrix synthesis/deposition genes, is increased in the ascending aorta of Marfan (Fbn1(C1039G/+)) mice. Increased apoptosis, assessed by increased cleaved caspase-3 and caspase-9, enhanced caspase-3 activity, and decreased levels of the antiapoptotic proteins, Mcl-1 and Bcl-2, were found in the Fbn1(C1039G/+) aorta. Histological evidence of decreased and fragmented elastin was observed exclusively in the Fbn1(C1039G/+) ascending aorta in association with repressed elastin mRNA and increased matrix metalloproteinase-2 expression and activity, both targets of miR-29b. Evidence of decreased activation of nuclear factor κB, a repressor of miR-29b, and a factor suppressed by TGF-ß, was also observed in Fbn1(C1039G/+) aorta. Furthermore, administration of a nuclear factor κB inhibitor increased miR-29b levels, whereas TGF-ß blockade or losartan effectively decreased miR-29b levels in Fbn1(C1039G/+) mice. Finally, miR-29b blockade by locked nucleic acid antisense oligonucleotides prevented early aneurysm development, aortic wall apoptosis, and extracellular matrix deficiencies. CONCLUSIONS: We identify increased miR-29b expression as key to the pathogenesis of early aneurysm development in MFS by regulating aortic wall apoptosis and extracellular matrix abnormalities.


Subject(s)
Aorta/metabolism , Aortic Aneurysm/metabolism , Marfan Syndrome/metabolism , MicroRNAs/metabolism , Age Factors , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Aorta/pathology , Aortic Aneurysm/genetics , Aortic Aneurysm/pathology , Aortic Aneurysm/prevention & control , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Cells, Cultured , Disease Models, Animal , Elastin/genetics , Elastin/metabolism , Female , Fibrillin-1 , Fibrillins , Genetic Therapy/methods , Losartan/pharmacology , Male , Marfan Syndrome/complications , Marfan Syndrome/genetics , Marfan Syndrome/pathology , Marfan Syndrome/therapy , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , NF-kappa B/metabolism , Oligonucleotides, Antisense/administration & dosage , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/metabolism , Up-Regulation
12.
Circ Res ; 111(7): 882-93, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22821929

ABSTRACT

RATIONALE: Induced pluripotent stem cells (iPSCs) hold great promise for the development of patient-specific therapies for cardiovascular disease. However, clinical translation will require preclinical optimization and validation of large-animal iPSC models. OBJECTIVE: To successfully derive endothelial cells from porcine iPSCs and demonstrate their potential utility for the treatment of myocardial ischemia. METHODS AND RESULTS: Porcine adipose stromal cells were reprogrammed to generate porcine iPSCs (piPSCs). Immunohistochemistry, quantitative PCR, microarray hybridization, and angiogenic assays confirmed that piPSC-derived endothelial cells (piPSC-ECs) shared similar morphological and functional properties as endothelial cells isolated from the autologous pig aorta. To demonstrate their therapeutic potential, piPSC-ECs were transplanted into mice with myocardial infarction. Compared with control, animals transplanted with piPSC-ECs showed significant functional improvement measured by echocardiography (fractional shortening at week 4: 27.2±1.3% versus 22.3±1.1%; P<0.001) and MRI (ejection fraction at week 4: 45.8±1.3% versus 42.3±0.9%; P<0.05). Quantitative protein assays and microfluidic single-cell PCR profiling showed that piPSC-ECs released proangiogenic and antiapoptotic factors in the ischemic microenvironment, which promoted neovascularization and cardiomyocyte survival, respectively. Release of paracrine factors varied significantly among subpopulations of transplanted cells, suggesting that transplantation of specific cell populations may result in greater functional recovery. CONCLUSIONS: In summary, this is the first study to successfully differentiate piPSCs-ECs from piPSCs and demonstrate that transplantation of piPSC-ECs improved cardiac function after myocardial infarction via paracrine activation. Further development of these large animal iPSC models will yield significant insights into their therapeutic potential and accelerate the clinical translation of autologous iPSC-based therapy.


Subject(s)
Cell Transplantation , Endothelium, Vascular/cytology , Endothelium, Vascular/transplantation , Heart/physiopathology , Microfluidic Analytical Techniques , Myocardial Infarction/therapy , Paracrine Communication/physiology , Pluripotent Stem Cells/cytology , Animals , Cell Differentiation/physiology , Cell Survival/physiology , Cells, Cultured , Echocardiography , Endothelium, Vascular/physiology , Female , Magnetic Resonance Imaging , Mice , Mice, SCID , Models, Animal , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/pathology , Neovascularization, Physiologic , Pluripotent Stem Cells/physiology , Swine , Swine, Miniature
13.
Circ Res ; 111(12): 1494-503, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-22967807

ABSTRACT

RATIONALE: Molecular imaging has proven to be a vital tool in the characterization of stem cell behavior in vivo. However, the integration of reporter genes has typically relied on random integration, a method that is associated with unwanted insertional mutagenesis and positional effects on transgene expression. OBJECTIVE: To address this barrier, we used genome editing with zinc finger nuclease (ZFN) technology to integrate reporter genes into a safe harbor gene locus (PPP1R12C, also known as AAVS1) in the genome of human embryonic stem cells and human induced pluripotent stem cells for molecular imaging. METHODS AND RESULTS: We used ZFN technology to integrate a construct containing monomeric red fluorescent protein, firefly luciferase, and herpes simplex virus thymidine kinase reporter genes driven by a constitutive ubiquitin promoter into a safe harbor locus for fluorescence imaging, bioluminescence imaging, and positron emission tomography imaging, respectively. High efficiency of ZFN-mediated targeted integration was achieved in both human embryonic stem cells and induced pluripotent stem cells. ZFN-edited cells maintained both pluripotency and long-term reporter gene expression. Functionally, we successfully tracked the survival of ZFN-edited human embryonic stem cells and their differentiated cardiomyocytes and endothelial cells in murine models, demonstrating the use of ZFN-edited cells for preclinical studies in regenerative medicine. CONCLUSION: Our study demonstrates a novel application of ZFN technology to the targeted genetic engineering of human pluripotent stem cells and their progeny for molecular imaging in vitro and in vivo.


Subject(s)
Deoxyribonucleases/genetics , Embryonic Stem Cells/enzymology , Genetic Engineering/methods , Genome, Human/genetics , Induced Pluripotent Stem Cells/enzymology , RNA Editing/genetics , Zinc Fingers/genetics , Animals , Cell Differentiation/genetics , Cells, Cultured , Deoxyribonucleases/administration & dosage , Embryonic Stem Cells/cytology , Embryonic Stem Cells/transplantation , Gene Targeting/methods , Genes, Reporter/physiology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/transplantation , Mice , Optical Imaging/methods
14.
Circulation ; 126(11 Suppl 1): S20-8, 2012 Sep 11.
Article in English | MEDLINE | ID: mdl-22965984

ABSTRACT

BACKGROUND: Human cardiac progenitor cells (hCPCs) are a promising cell source for regenerative repair after myocardial infarction. Exploitation of their full therapeutic potential may require stable genetic modification of the cells ex vivo. Safe genetic engineering of stem cells, using facile methods for site-specific integration of transgenes into known genomic contexts, would significantly enhance the overall safety and efficacy of cellular therapy in a variety of clinical contexts. METHODS AND RESULTS: We used the phiC31 site-specific recombinase to achieve targeted integration of a triple fusion reporter gene into a known chromosomal context in hCPCs and human endothelial cells. Stable expression of the reporter gene from its unique chromosomal integration site resulted in no discernible genomic instability or adverse changes in cell phenotype. Namely, phiC31-modified hCPCs were unchanged in their differentiation propensity, cellular proliferative rate, and global gene expression profile when compared with unaltered control hCPCs. Expression of the triple fusion reporter gene enabled multimodal assessment of cell fate in vitro and in vivo using fluorescence microscopy, bioluminescence imaging, and positron emission tomography. Intramyocardial transplantation of genetically modified hCPCs resulted in significant improvement in myocardial function 2 weeks after cell delivery, as assessed by echocardiography (P=0.002) and MRI (P=0.001). We also demonstrated the feasibility and therapeutic efficacy of genetically modifying differentiated human endothelial cells, which enhanced hind limb perfusion (P<0.05 at day 7 and 14 after transplantation) on laser Doppler imaging. CONCLUSIONS: The phiC31 integrase genomic modification system is a safe, efficient tool to enable site-specific integration of reporter transgenes in progenitor and differentiated cell types.


Subject(s)
Fetal Stem Cells/transplantation , Genetic Therapy/methods , Hindlimb/blood supply , Ischemia/surgery , Mutagenesis, Insertional/methods , Myocardial Infarction/surgery , Animals , Cell Differentiation , Cell Division , Chromosomes, Human, Pair 19/genetics , Endothelial Cells/cytology , Female , Fetal Heart/cytology , Fetal Stem Cells/cytology , Fetal Stem Cells/metabolism , Gene Expression Regulation , Genes, Reporter , Humans , Integrases , Intracellular Signaling Peptides and Proteins , Ischemia/physiopathology , Luciferases, Firefly/genetics , Luminescent Proteins/genetics , Magnetic Resonance Imaging , Mice , Mice, SCID , Proteins/genetics , Random Allocation , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Thymidine Kinase/genetics , Transgenes , Vesicular Transport Proteins , Viral Proteins/genetics , Virus Integration , Red Fluorescent Protein
15.
J Cell Sci ; 124(Pt 17): 3029-37, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21878509

ABSTRACT

Human embryonic stem cells (hESCs) can serve as a universal cell source for emerging cell or tissue replacement strategies, but immune rejection of hESC derivatives remains an unsolved problem. Here, we sought to describe the mechanisms of rejection for naïve hESCs and upon HLA class I (HLA I) knockdown (hESC(KD)). hESCs were HLA I-positive but negative for HLA II and co-stimulatory molecules. Transplantation of naïve hESC into immunocompetent Balb/c mice induced substantial T helper cell 1 and 2 (Th1 and Th2) responses with rapid cell death, but hESCs survived in immunodeficient SCID-beige recipients. Histology revealed mainly macrophages and T cells, but only scattered natural killer (NK) cells. A surge of hESC-specific antibodies against hESC class I, but not class II antigens, was observed. Using HLA I RNA interference and intrabody technology, HLA I surface expression of hESC(KD) was 88%-99% reduced. T cell activation after hESC(KD) transplantation into Balb/c was significantly diminished, antibody production was substantially alleviated, the levels of graft-infiltrating immune cells were reduced and the survival of hESC(KD) was prolonged. Because of their very low expression of stimulatory NK ligands, NK-susceptibility of naïve hESCs and hESC(KD) was negligible. Thus, HLA I recognition by T cells seems to be the primary mechanism of hESC recognition, and T cells, macrophages and hESC-specific antibodies participate in hESC killing.


Subject(s)
Embryonic Stem Cells/immunology , Embryonic Stem Cells/transplantation , Graft Rejection/immunology , HLA Antigens/genetics , HLA Antigens/immunology , Animals , Embryonic Stem Cells/cytology , Gene Knockdown Techniques/methods , Graft Rejection/genetics , Humans , Male , Mice , Mice, Inbred BALB C , Mice, SCID , Transplantation, Heterologous
16.
Nat Methods ; 7(3): 197-9, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20139967

ABSTRACT

Owing to the risk of insertional mutagenesis, viral transduction has been increasingly replaced by nonviral methods to generate induced pluripotent stem cells (iPSCs). We report the use of 'minicircle' DNA, a vector type that is free of bacterial DNA and capable of high expression in cells, for this purpose. Here we use a single minicircle vector to generate transgene-free iPSCs from adult human adipose stem cells.


Subject(s)
DNA, Circular/genetics , Genetic Vectors , Induced Pluripotent Stem Cells/metabolism , Adult , Humans , Transfection
17.
Arterioscler Thromb Vasc Biol ; 32(1): 92-102, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22034515

ABSTRACT

OBJECTIVE: Clinical trials of bone marrow-derived stem cell therapy for the heart have yielded variable results. The basic mechanism(s) that underlies their potential efficacy remains unknown. In the present study, we evaluated the survival kinetics, transcriptional response, and functional outcome of intramyocardial bone marrow mononuclear cell (BMMC) transplantation for cardiac repair in a murine myocardial infarction model. METHODS AND RESULTS: We used bioluminescence imaging and high-throughput transcriptional profiling to evaluate the in vivo survival kinetics and gene expression changes of transplanted BMMCs after their engraftment into ischemic myocardium. Our results demonstrate short-lived survival of cells following transplant, with less than 1% of cells surviving by 6 weeks posttransplantation. Moreover, transcriptomic analysis of BMMCs revealed nonspecific upregulation of various cell regulatory genes, with a marked downregulation of cell differentiation and maturation pathways. BMMC therapy caused limited improvement of heart function as assessed by echocardiography, invasive hemodynamics, and positron emission tomography. Histological evaluation of cell fate further confirmed findings of the in vivo cell tracking and transcriptomic analysis. CONCLUSIONS: Collectively, these data suggest that BMMC therapy, in its present iteration, may be less efficacious than once thought. Additional refinement of existing cell delivery protocols should be considered to induce better therapeutic efficacy.


Subject(s)
Hematopoietic Stem Cell Transplantation , Myocardial Ischemia/therapy , Animals , Cell Survival , Echocardiography , Female , Gene Expression Profiling , Genes, Reporter , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Male , Mice , Mice, Transgenic , Myocardial Ischemia/genetics , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Positron-Emission Tomography , Time Factors
18.
Nat Med ; 12(3): 361-5, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16491087

ABSTRACT

There is a crucial need for alternatives to native vein or artery for vascular surgery. The clinical efficacy of synthetic, allogeneic or xenogeneic vessels has been limited by thrombosis, rejection, chronic inflammation and poor mechanical properties. Using adult human fibroblasts extracted from skin biopsies harvested from individuals with advanced cardiovascular disease, we constructed tissue-engineered blood vessels (TEBVs) that serve as arterial bypass grafts in long-term animal models. These TEBVs have mechanical properties similar to human blood vessels, without relying upon synthetic or exogenous scaffolding. The TEBVs are antithrombogenic and mechanically stable for 8 months in vivo. Histological analysis showed complete tissue integration and formation of vasa vasorum. The endothelium was confluent and positive for von Willebrand factor. A smooth muscle-specific alpha-actin-positive cell population developed within the TEBV, suggesting regeneration of a vascular media. Electron microscopy showed an endothelial basement membrane, elastogenesis and a complex collagen network. These results indicate that a completely biological and clinically relevant TEBV can be assembled exclusively from an individual's own cells.


Subject(s)
Arteries/growth & development , Blood Vessel Prosthesis , Blood Vessels/cytology , Blood Vessels/growth & development , Tissue Engineering , Adult , Animals , Blood Vessel Prosthesis Implantation , Blood Vessels/transplantation , Cells, Cultured , Dogs , Humans , Primates , Rats , Rats, Nude , Time Factors
19.
J Biol Chem ; 286(37): 32697-704, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21719696

ABSTRACT

Derivation of patient-specific induced pluripotent stem cells (iPSCs) opens a new avenue for future applications of regenerative medicine. However, before iPSCs can be used in a clinical setting, it is critical to validate their in vivo fate following autologous transplantation. Thus far, preclinical studies have been limited to small animals and have yet to be conducted in large animals that are physiologically more similar to humans. In this study, we report the first autologous transplantation of iPSCs in a large animal model through the generation of canine iPSCs (ciPSCs) from the canine adipose stromal cells and canine fibroblasts of adult mongrel dogs. We confirmed pluripotency of ciPSCs using the following techniques: (i) immunostaining and quantitative PCR for the presence of pluripotent and germ layer-specific markers in differentiated ciPSCs; (ii) microarray analysis that demonstrates similar gene expression profiles between ciPSCs and canine embryonic stem cells; (iii) teratoma formation assays; and (iv) karyotyping for genomic stability. Fate of ciPSCs autologously transplanted to the canine heart was tracked in vivo using clinical positron emission tomography, computed tomography, and magnetic resonance imaging. To demonstrate clinical potential of ciPSCs to treat models of injury, we generated endothelial cells (ciPSC-ECs) and used these cells to treat immunodeficient murine models of myocardial infarction and hindlimb ischemia.


Subject(s)
Adipose Tissue/metabolism , Gene Expression Regulation , Induced Pluripotent Stem Cells/metabolism , Stem Cell Transplantation , Adipose Tissue/cytology , Animals , Disease Models, Animal , Dogs , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression Profiling , Humans , Induced Pluripotent Stem Cells/cytology , Male , Mice , Mice, SCID , Myocardial Ischemia/therapy , Oligonucleotide Array Sequence Analysis , Stromal Cells/cytology , Stromal Cells/metabolism , Transplantation, Autologous , Transplantation, Heterologous
20.
Circulation ; 124(11 Suppl): S27-34, 2011 Sep 13.
Article in English | MEDLINE | ID: mdl-21911815

ABSTRACT

BACKGROUND: Although stem cell therapy has provided a promising treatment for myocardial infarction, the low survival of the transplanted cells in the infarcted myocardium is possibly a primary reason for failure of long-term improvement. Therefore, the development of novel prosurvival strategies to boost stem cell survival will be of significant benefit to this field. METHODS AND RESULTS: Cardiac progenitor cells (CPCs) were isolated from transgenic mice, which constitutively express firefly luciferase and green fluorescent protein. The CPCs were transduced with individual lentivirus carrying the precursor of miR-21, miR-24, and miR-221, a cocktail of these 3 microRNA precursors, or green fluorescent protein as a control. After challenge in serum free medium, CPCs treated with the 3 microRNA cocktail showed significantly higher viability compared with untreated CPCs. After intramuscular and intramyocardial injections, in vivo bioluminescence imaging showed that microRNA cocktail-treated CPCs survived significantly longer after transplantation. After left anterior descending artery ligation, microRNA cocktail-treated CPCs boost the therapeutic efficacy in terms of functional recovery. Histological analysis confirmed increased myocardial wall thickness and CPC engraftment in the myocardium with the microRNA cocktail. Finally, we used bioinformatics analysis and experimental validation assays to show that Bim, a critical apoptotic activator, is an important target gene of the microRNA cocktail, which collectively can bind to the 3'UTR region of Bim and suppress its expression. CONCLUSIONS: We have demonstrated that a microRNA prosurvival cocktail (miR-21, miR-24, and miR-221) can improve the engraftment of transplanted cardiac progenitor cells and therapeutic efficacy for treatment of ischemic heart disease.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Graft Survival/physiology , MicroRNAs/genetics , MicroRNAs/therapeutic use , Myocardial Infarction/therapy , Stem Cell Transplantation/methods , Stem Cells/cytology , Animals , Cell Differentiation/physiology , Cells, Cultured , Female , Injections, Intramuscular , Lentivirus , Mice , Mice, SCID , Mice, Transgenic , Models, Animal , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Stem Cells/physiology , Transduction, Genetic , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL