Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
PLoS One ; 18(1): e0277442, 2023.
Article in English | MEDLINE | ID: mdl-36662847

ABSTRACT

BACKGROUND: Recent preclinical studies have demonstrated that bone marrow (BM)-derived Muse cells have a homing mechanism to reach damaged cardiac tissue while also being able to reduce myocardial infarct size and improve cardiac function; however, the potential of BM-Muse cells to foster new blood-vessel formation has not been fully assessed. Up to date, adipose tissue (AT)-derived Muse cells remain to be studied in acute myocardial infarction (AMI). The aim of the present study was to analyze in vitro and in vivo the neovascularization capacity of AT-Muse cells while exploring their biodistribution and differentiation potential in a translational ovine model of AMI. METHODS AND RESULTS: AT-Muse cells were successfully isolated from ovine adipose tissue. In adult sheep, one or more diagonal branches of the left anterior descending coronary artery were permanently ligated for thirty minutes. Sheep were randomized in two groups and treated with intramyocardial injections: Vehicle (PBS, n = 4) and AT-Muse (2x107 AT-Muse cells labeled with PKH26 Red Fluorescent Dye, n = 4). Molecular characterization showed higher expression of angiogenic genes (VEGF, PGF and ANG) and increased number of tube formation in AT-Muse cells group compared to Adipose-derived mesenchymal stromal cells (ASCs) group. At 7 days post-IAM, the AT-Muse group showed significantly more arterioles and capillaries than the Vehicle group. Co-localization of PKH26+ cells with desmin, sarcomeric actin and troponin T implied the differentiation of Muse cells to a cardiac fate; moreover, PKH26+ cells also co-localized with a lectin marker, suggesting a possible differentiation to a vascular lineage. CONCLUSION: Intramyocardially administered AT-Muse cells displayed a significant neovascularization activity and survival capacity in an ovine model of AMI.


Subject(s)
Alprostadil , Myocardial Infarction , Animals , Sheep , Alprostadil/metabolism , Tissue Distribution , Myocardial Infarction/therapy , Myocardial Infarction/metabolism , Adipocytes/metabolism
2.
Comput Struct Biotechnol J ; 20: 3779-3782, 2022.
Article in English | MEDLINE | ID: mdl-35891794

ABSTRACT

Angiogenic gene overexpression has been the main strategy in numerous vascular regenerative gene therapy projects. However, most have failed in clinical trials. CRISPRa technology enhances gene overexpression levels based on the identification of sgRNAs with maximum efficiency and safety. CRISPick and CHOP CHOP are the most widely used web tools for the prediction of sgRNAs. The objective of our study was to analyze the performance of both platforms for the sgRNA design to angiogenic genes (VEGFA, KDR, EPO, HIF-1A, HGF, FGF, PGF, FGF1) involving different human reference genomes (GRCH 37 and GRCH 38). The top 20 ranked sgRNAs proposed by the two tools were analyzed in different aspects. No significant differences were found on the DNA curvature associated with the sgRNA binding sites but the sgRNA predicted on-target efficiency was significantly greater when CRISPick was used. Moreover, the mean ranking variation was greater for the same platform in EPO, EGF, HIF-1A, PGF and HGF, whereas it did not reach statistical significance in KDR, FGF-1 and VEGFA. The rearrangement analysis of the ranking positions was also different between platforms. CRISPick proved to be more accurate in establishing the best sgRNAs in relation to a more complete genome, whereas CHOP CHOP showed a narrower classification reordering.

3.
J Cardiovasc Pharmacol ; 78(3): 346-360, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34516452

ABSTRACT

ABSTRACT: Adult mammalian cardiomyocytes show scarce division ability, which makes the heart ineffective in replacing lost contractile cells after ischemic cardiomyopathy. In the past decades, there have been increasing efforts in the search for novel strategies to regenerate the injured myocardium. Among them, gene therapy is one of the most promising ones, based on recent and emerging studies that support the fact that functional cardiomyocyte regeneration can be accomplished by the stimulation and enhancement of the endogenous ability of these cells to achieve cell division. This capacity can be targeted by stimulating several molecules, such as cell cycle regulators, noncoding RNAs, transcription, and metabolic factors. Therefore, the proposed target, together with the selection of the vector used, administration route, and the experimental animal model used in the development of the therapy would determine the success in the clinical field.


Subject(s)
Cell Proliferation , Genetic Therapy , Myocardial Ischemia/therapy , Myocytes, Cardiac/pathology , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Gene Expression Regulation , Humans , Myocardial Ischemia/genetics , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Myocytes, Cardiac/metabolism , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Recovery of Function , Regeneration , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Int J Cardiovasc Imaging ; 37(5): 1789-1798, 2021 May.
Article in English | MEDLINE | ID: mdl-33442855

ABSTRACT

Transient alterations in ventricular conduction and synchronized cardiac performance have been reported in experimental models of myocardial ischemia. In post-stress 99mTc-sestamibi-gated-SPECT myocardial perfusion imaging (MPI), the time elapsed between tracer injection and image acquisition could influence the detection of ischemic left ventricular mechanical dyssynchrony (LVMD). We aimed at evaluating whether early vs. delayed post-stress MPI improve ischemic LVMD detection using the phase analysis parameters standard deviation (SD) and histogram bandwidth (HB) and to assess the correlation between stress-induced changes in SD and HB and other functional parameters. We prospectively studied 32 control subjects (Group-1) and 60 ischemic patients (Group-2). Stress-induced changes were calculated as stress minus rest (Δ). LVMD was defined as post-stress increases of either SD or HB. Group-2 showed higher ΔSD and ΔHB in early than in delayed images: early ΔSD: 1.63 (- 0.37 to 4.83) vs. delayed ΔSD: - 0.39 (- 3.82 to 1.74); early ΔHB: 2.50 (- 4 to 12) vs. ΔHB delayed: - 4 (- 15.75 to 4), all p < 0.01. ΔSD and ΔHB correlated linearly with ΔLV-ejection-fraction (EF) and ΔLV-end systolic-volume (ESV) in early images, all p < 0.01. Early images detected LVMD in more patients than delayed scans (78% vs. 38%; p < 0.01) All patients with LVEF drop in early post-stress evaluation had LVMD. Early post-stress images improve ischemic LVMD detection. Ischemic LVEF and LVESV changes correlate with ΔSD and ΔHB.


Subject(s)
Myocardial Perfusion Imaging , Ventricular Dysfunction, Left , Electrocardiography , Humans , Predictive Value of Tests , Technetium Tc 99m Sestamibi , Tomography, Emission-Computed, Single-Photon , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/etiology
5.
Cytotherapy ; 22(10): 563-572, 2020 10.
Article in English | MEDLINE | ID: mdl-32723595

ABSTRACT

BACKGROUND AIMS: Peripheral arterial disease (PAD) is a progressive, disabling ailment for which no effective treatment exists. Gene therapy-mediated neovascularization has emerged as a potentially useful strategy. We tested the angiogenic and arteriogenic efficacy and safety of a baculovirus (BV) encoding mutant, oxygen-resistant hypoxia-inducible factor 1-alpha (mHIF-1α), in rabbits with PAD. METHODS: After assessing the transfection efficiency of the BV.mHIF-1α vector and its tubulogenesis potential in vitro, we randomized rabbits with experimental PAD to receive 1 × 109 copies of BV.mHIF-1α or BV.null (n = 6 per group) 7 days after surgery. Two weeks post-treatment, collateralization (digital angiography) and capillary and arteriolar densities (immunohistochemistry) were measured in the posterior limbs. Ischemic damage was evaluated in adductor and gastrocnemius muscle samples. Tracking of viral DNA in injected zones and remote tissues at different time points was performed in additional rabbits using a BV encoding GFP. RESULTS: Angiographically visible collaterals were more numerous in BV.mHIF-1α-treated rabbits (8.12 ± 0.42 vs 6.13 ± 1.15 collaterals/cm2, P < 0.05). The same occurred with arteriolar (27.9 ± 7.0 vs 15.3 ± 4.0 arterioles/mm2) and capillary (341.8 ± 109.9 vs 208.8 ± 87.7 capillaries/mm2, P < 0.05) densities. BV.mHIF-1α-treated rabbits displayed less ischemic muscle damage than BV.null-treated animals. Viral DNA and GFP mRNA were detectable only at 3 and 7 days after injection in hind limbs. Neither the virus nor GFP mRNA was detected in remote tissues. CONCLUSIONS: In rabbits with PAD, BV.mHIF-1α induced neovascularization and reduced ischemic damage, exhibiting a good safety profile at 14 days post-treatment. Complementary studies to evaluate its potential usefulness in the clinic are needed.


Subject(s)
Baculoviridae/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Ischemia/therapy , Muscle, Skeletal/blood supply , Muscle, Skeletal/pathology , Neovascularization, Physiologic , Peripheral Arterial Disease/therapy , Animals , Arterioles , Disease Models, Animal , Gene Expression , Genetic Therapy , Hindlimb/blood supply , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ischemia/pathology , Microvessels/pathology , Peripheral Arterial Disease/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rabbits , Transfection
6.
Am J Physiol Heart Circ Physiol ; 318(4): H994-H1007, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32167779

ABSTRACT

The adult mammalian cardiomyocyte has a very limited capacity to reenter the cell cycle and advance into mitosis. Therefore, diseases characterized by lost contractile tissue usually evolve into myocardial remodeling and heart failure. Analyzing the cardiac transcriptome at different developmental stages in a large mammal closer to the human than laboratory rodents may serve to disclose positive and negative cardiomyocyte cell cycle regulators potentially targetable to induce cardiac regeneration in the clinical setting. Thus we aimed at characterizing the transcriptomic profiles of the early fetal, late fetal, and adult sheep heart by employing RNA-seq technique and bioinformatic analysis to detect protein-encoding genes that in some of the stages were turned off, turned on, or differentially expressed. Genes earlier proposed as positive cell cycle regulators such as cyclin A, cdk2, meis2, meis3, and PCNA showed higher expression in fetal hearts and lower in AH, as expected. In contrast, genes previously proposed as cell cycle inhibitors, such as meis1, p16, and sav1, tended to be higher in fetal than in adult hearts, suggesting that these genes are involved in cell processes other than cell cycle regulation. Additionally, we described Gene Ontology (GO) enrichment of different sets of genes. GO analysis revealed that differentially expressed gene sets were mainly associated with metabolic and cellular processes. The cell cycle-related genes fam64a, cdc20, and cdk1, and the metabolism-related genes pitx and adipoq showed strong differential expression between fetal and adult hearts, thus being potent candidates to be targeted in human cardiac regeneration strategies.NEW & NOTEWORTHY We characterized the transcriptomic profiles of the fetal and adult sheep hearts employing RNAseq technique and bioinformatic analyses to provide sets of transcripts whose variation in expression level may link them to a specific role in cell cycle regulation. It is important to remark that this study was performed in a large mammal closer to humans than laboratory rodents. In consequence, the results can be used for further translational studies in cardiac regeneration.


Subject(s)
Gene Expression Regulation, Developmental , Heart/physiology , Myocardium/metabolism , Regeneration , Transcriptome , Animals , Cyclin A/genetics , Cyclin A/metabolism , Female , Heart/growth & development , Male , Sheep , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Drug Deliv Transl Res ; 9(5): 935-944, 2019 10.
Article in English | MEDLINE | ID: mdl-30859393

ABSTRACT

In rodents with acute myocardial infarction (AMI), high mobility group box 1 (HMGB1) injection has produced controversial results. Given the lack of data in large mammals, we searched the dose that would promote angiogenesis and expression of specific regenerative genes in sheep with AMI (protocol 1) and, subsequently, use this dose to study long-term effects on infarct size and left ventricular (LV) function (protocol 2). Protocol 1: Sheep with AMI received 250 µg (high-dose, n = 7), 25 µg (low-dose, n = 7) HMGB1, or PBS (placebo, n = 7) in 10 intramyocardial injections (0.2 ml each) in the peri-infarct area. Seven days later, only the high-HMGB1-dose group exhibited higher microvascular densities, Ki67-positive cardiomyocytes, and overexpression of VEGF, Ckit, Tbx20, Nkx2.5, and Gata4. Protocol 2: Sheep with AMI received HMGB1 250 µg (n = 6) or PBS (n = 6). At 60 days, HMGB1-treated sheep showed smaller infarcts (8.5 ± 2.11 vs. 12.2 ± 1.97% LV area, P < 0.05, ANOVA-Bonferroni) and higher microvascular density (capillaries, 1798 ± 252 vs. 1266 ± 250/mm2; arterioles, 18.3 ± 3.9 vs. 11.7 ± 2.2/mm2; both P < 0.01). Echocardiographic LV ejection fraction, circumferential shortening, and wall thickening increased from day 3 to 60 with HMGB1 (all P < 0.05). Conclusion: in ovine AMI, high-dose HMGB1 induces angio-arteriogenesis, reduces infarct size, and improves LV function at 2 months post-treatment.


Subject(s)
Cardiotonic Agents/administration & dosage , HMGB1 Protein/administration & dosage , Myocardial Infarction/drug therapy , Animals , Female , Male , Microvessels/drug effects , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , Sheep , Ventricular Function, Left/drug effects
8.
Curr Drug Targets ; 20(2): 241-254, 2019.
Article in English | MEDLINE | ID: mdl-30068271

ABSTRACT

Adult mammalian cardiomyocytes (CMs) exhibit limited proliferative capacity, as cell cycle activity leads to an increase in DNA content, but mitosis and cytokinesis are infrequent. This makes the heart highly inefficient in replacing with neoformed cardiomyocytes lost contractile cells as occurs in diseases such as myocardial infarction and dilated cardiomyopathy. Regenerative therapies based on the implant of stem cells of diverse origin do not warrant engraftment and electromechanical connection of the new cells with the resident ones, a fundamental condition to restore the physiology of the cardiac syncytium. Consequently, there is a growing interest in identifying factors playing relevant roles in the regulation of the CM cell cycle to be targeted in order to induce the resident cardiomyocytes to divide into daughter cells and thus achieve myocardial regeneration with preservation of physiologic syncytial performance. Despite the scientific progress achieved over the last decades, many questions remain unanswered, including how cardiomyocyte proliferation is regulated during heart development in gestation and neonatal life. This can reveal unknown cell cycle regulation mechanisms and molecules that may be manipulated to achieve cardiac self-regeneration. We hereby revise updated data on CM cell cycle regulation, participating molecules and pathways recently linked with the cell cycle, as well as experimental therapies involving them.


Subject(s)
Myocytes, Cardiac/physiology , Regeneration , Animals , Cell Cycle , Cell Proliferation , Gene Regulatory Networks , Humans
9.
Artif Cells Nanomed Biotechnol ; 46(sup3): S717-S724, 2018.
Article in English | MEDLINE | ID: mdl-30289284

ABSTRACT

Diaphragmatic myoblasts (DM) are stem cells of the diaphragm, a muscle displaying high resistance to stress and exhaustion. We hypothesized that DM modified to overexpress connexin-43 (cx43), seeded on aligned poly (l-lactic acid) (PLLA) sheets would decrease infarct size and improve ventricular function in sheep with acute myocardial infarction (AMI). Sheep with AMI received PLLA sheets without DM (PLLA group), sheets with DM (PLLA-DM group), sheets with DM overexpressing cx43 (PLLA-DMcx43) or no treatment (control group, n = 6 per group). Infarct size (cardiac magnetic resonance) decreased ∼25% in PLLA-DMcx43 [from 8.2 ± 0.6 ml (day 2) to 6.5 ± 0.7 ml (day 45), p < .01, ANOVA-Bonferroni] but not in the other groups. Ejection fraction (EF%) (echocardiography) at 3 days post-AMI fell significantly in all groups. At 45 days, PLLA-DM y PLLA-DMcx43 recovered their EF% to pre-AMI values (PLLA-DM: 61.1 ± 0.5% vs. 58.9 ± 3.3%, p = NS; PLLA-DMcx43: 64.6 ± 2.9% vs. 56.9 ± 2.4%, p = NS), but not in control (56.8 ± 2.0% vs. 43.8 ± 1.1%, p < .01) and PLLA (65.7 ± 2.1% vs. 56.6 ± 4.8%, p < .01). Capillary density was higher (p < .05) in PLLA-DMcx43 group than in the remaining groups. In conclusion, PLLA-DMcx43 reduces infarct size in sheep with AMI. PLLA-DMcx43 and PLLA-DM improve ventricular function similarly. Given its safety and feasibility, this novel approach may prove beneficial in the clinic.


Subject(s)
Connexin 43/biosynthesis , Coronary Occlusion , Diaphragm/metabolism , Myoblasts , Myocardial Infarction , Polyesters/chemistry , Tissue Scaffolds/chemistry , Ventricular Function , Animals , Coronary Occlusion/metabolism , Coronary Occlusion/pathology , Coronary Occlusion/physiopathology , Coronary Occlusion/therapy , Diaphragm/pathology , Male , Myoblasts/metabolism , Myoblasts/pathology , Myoblasts/transplantation , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Infarction/therapy , Sheep
10.
Cytotechnology ; 70(2): 651-664, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29143226

ABSTRACT

Diaphragmatic myoblasts (DMs) are precursors of type-1 muscle cells displaying high exhaustion threshold on account that they contract and relax 20 times/min over a lifespan, making them potentially useful in cardiac regeneration strategies. Besides, it has been shown that biomaterials for stem cell delivery improve cell retention and viability in the target organ. In the present study, we aimed at developing a novel approach based on the use of poly (L-lactic acid) (PLLA) scaffolds seeded with DMs overexpressing connexin-43 (cx43), a gap junction protein that promotes inter-cell connectivity. DMs isolated from ovine diaphragm biopsies were characterized by immunohistochemistry and ability to differentiate into myotubes (MTs) and transduced with a lentiviral vector encoding cx43. After confirming cx43 expression (RT-qPCR and Western blot) and its effect on inter-cell connectivity (fluorescence recovery after photobleaching), DMs were grown on fiber-aligned or random PLLA scaffolds. DMs were successfully isolated and characterized. Cx43 mRNA and protein were overexpressed and favored inter-cell connectivity. Alignment of the scaffold fibers not only aligned but also elongated the cells, increasing the contact surface between them. This novel approach is feasible and combines the advantages of bioresorbable scaffolds as delivery method and a cell type that on account of its features may be suitable for cardiac regeneration. Future studies on animal models of myocardial infarction are needed to establish its usefulness on scar reduction and cardiac function.

11.
J Am Heart Assoc ; 5(7)2016 07 06.
Article in English | MEDLINE | ID: mdl-27385426

ABSTRACT

BACKGROUND: Bone marrow mesenchymal stromal cells (BMMSCs) are cardioprotective in acute myocardial infarction (AMI) because of release of paracrine angiogenic and prosurvival factors. Hypoxia-inducible factor 1-α (HIF1-α), rapidly degraded during normoxia, is stabilized during ischemia and upregulates various cardioprotective genes. We hypothesized that BMMSCs engineered to overexpress mutant, oxygen-resistant HIF1-α would confer greater cardioprotection than nontransfected BMMSCs in sheep with AMI. METHODS AND RESULTS: Allogeneic BMMSCs transfected with a minicircle vector encoding mutant HIF1-α (BMMSC-HIF) were injected in the peri-infarct of sheep (n=6) undergoing coronary occlusion. Over 2 months, infarct volume measured by cardiac magnetic resonance (CMR) imaging decreased by 71.7±1.3% (P<0.001), and left ventricular (LV) percent ejection fraction (%EF) increased near 2-fold (P<0.001) in the presence of markedly decreased end-systolic volume. Sheep receiving nontransfected BMMSCs (BMMSC; n=6) displayed less infarct size limitation and percent LVEF improvement, whereas in placebo-treated animals (n=6), neither parameters changed over time. HIF1-α-transfected BMMSCs (BMMSC-HIF) induced angio-/arteriogenesis and decreased apoptosis by HIF1-mediated overexpression of erythropoietin, inducible nitrous oxide synthase, vascular endothelial growth factor, and angiopoietin-1. Cell tracking using paramagnetic iron nanoparticles in 12 additional sheep revealed enhanced long-term retention of BMMSC-HIF. CONCLUSIONS: Intramyocardial delivery of BMMSC-HIF reduced infarct size and improved LV systolic performance compared to BMMSC, attributed to increased neovascularization and cardioprotective effects induced by HIF1-mediated overexpression of paracrine factors and enhanced retention of injected cells. Given the safety of the minicircle vector and the feasibility of BMMSCs for allogeneic application, this treatment may be potentially useful in the clinic.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Myocardial Infarction/therapy , Animals , Disease Models, Animal , Flow Cytometry , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunoblotting , Magnetic Resonance Imaging , Male , Mesenchymal Stem Cell Transplantation/methods , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/pathology , Sheep
12.
Cytotechnology ; 68(4): 665-74, 2016 Aug.
Article in English | MEDLINE | ID: mdl-25432330

ABSTRACT

The adult heart contains a population of cardiac progenitor cells (CPCs). Growing and collecting an adequate number of CPCs demands complex culture media containing growth factors. Since activated macrophages secrete many growth factors, we investigated if activated isolated heart cells seeded on a feeder layer of activated peritoneal macrophages (PM) could result in CPCs and if these, in turn, could exert cardioprotection in rats with myocardial infarction (MI). Heart cells of inbred Wistar rats were isolated by collagenase digestion and cultured on PM obtained 72 h after intraperitoneal injection of 12 ml thioglycollate. Cells (1 × 10(6)) exhibiting CPC phenotype (immunohistochemistry) were injected in the periphery of rat MI 10 min after coronary artery occlusion. Control rats received vehicle. Three weeks later, left ventricular (LV) function (echocardiogram) was assessed, animals were euthanized and the hearts removed for histological studies. Five to six days after seeding heart cells on PM, spherical clusters composed of small bright and spherical cells expressing mostly c-Kit and Sca-1 antigens were apparent. After explant, those clusters developed cobblestone-like monolayers that expressed smooth muscle actin and sarcomeric actin and were successfully transferred for more than ten passages. When injected in the MI periphery, many of them survived at 21 days after coronary ligature, improved LV ejection fraction and decreased scar size as compared with control rats. CPC-derived cells with cardiocyte and smooth muscle phenotypes can be successfully grown on a feeder layer of activated syngeneic PM. These cells decreased scar size and improved heart function in rats with MI.

13.
Arterioscler Thromb Vasc Biol ; 35(1): 184-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25414254

ABSTRACT

OBJECTIVES: Critical limb ischemia complicates peripheral artery disease leading to tissue damage and amputation. We hypothesized that modifying adipose stromal cells (ASCs) to overexpress human vascular endothelial growth factor 165 (VEGF) would limit ischemic muscle damage to a larger extent than nonmodified ASCs. APPROACH AND RESULTS: Rabbits with critical hindlimb ischemia were injected with allogeneic abdominal fat-derived ASCs transfected with plasmid-VEGF165 (ASCs-VEGF; n=10). Additional rabbits received nontransfected ASCs (ASCs; n=10) or vehicle (placebo; n=10). One month later, ASCs-VEGF rabbits exhibited significantly higher density of angiographically visible collaterals and capillaries versus placebo (both P<0.05) but not versus ASCs (both P=NS). Arteriolar density, however, was increased in both ASCs and ASCs-VEGF groups (both P<0.05 versus placebo). ASCs-VEGF and ASCs showed comparable post-treatment improvements in Doppler-assessed peak systolic velocity, blood pressure ratio, and resistance index. Ischemic lesions were found in 40% of the muscle samples in the placebo group, 19% in the ASCs-VEGF group, and 17% in the ASCs groups (both P<0.05 versus placebo, Fisher test). CONCLUSIONS: In a rabbit model of critical limb ischemia, intramuscular injection of ASCs genetically modified to overexpress VEGF increase angiographically visible collaterals and capillary density. However, both modified and nonmodified ASCs increase arteriolar density to a similar extent and afford equal protection against ischemia-induced muscle lesions. These results indicate that modifying ASCs to overexpress VEGF does not enhance the protective effect of ASCs, and that arteriolar proliferation plays a pivotal role in limiting the irreversible tissue damage of critical limb ischemia.


Subject(s)
Adipose Tissue/transplantation , Genetic Therapy/methods , Ischemia/therapy , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Stromal Cells/transplantation , Vascular Endothelial Growth Factor A/biosynthesis , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Arterioles/metabolism , Arterioles/physiopathology , Blood Flow Velocity , Capillaries/metabolism , Capillaries/physiopathology , Cells, Cultured , Collateral Circulation , Disease Models, Animal , Female , Hindlimb , Humans , Ischemia/genetics , Ischemia/metabolism , Ischemia/pathology , Ischemia/physiopathology , Male , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Necrosis , Rabbits , Recovery of Function , Regional Blood Flow , Stromal Cells/metabolism , Time Factors , Transfection , Vascular Endothelial Growth Factor A/genetics
14.
Cytotherapy ; 15(2): 163-70, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23321328

ABSTRACT

BACKGROUND AIMS: Given the close similarity between ovine and human cardiomyocytes, sheep models of myocardial infarction and heart failure are increasingly used in studies of stem cell-mediated heart regeneration. In these studies, mesenchymal stromal cells (MSCs) are frequently employed. To enhance the paracrine effects of these MSCs, ex vivo transfection with genes encoding growth factors has been proposed. Although viral vectors exhibit higher transfection efficiency than plasmids, they entail the risks of uncontrolled transgene expression and immune reactions that preclude repeated administration. Our aim was to optimize the efficiency of plasmid-mediated transfection of ovine MSCs, while preserving cell viability. METHODS: Varying amounts of diverse cationic lipids were used to obtain the reagent-to-DNA mass ratio showing highest luciferase activity. Transfection efficiency (flow cytometry) was tested on plasmid-green fluorescent protein-transfected MSCs at increasing DNA mass. RESULTS: Lipofectamine LTX 5 µL and Plus reagent 4 µL with 2 µg of DNA yielded 42.3 ± 4.7% transfection efficiency, while preserving cell viability. Using these transfection conditions, we transfected MSCs with a plasmid encoding human vascular endothelial growth factor (VEGF) and found high VEGF protein concentrations in the culture supernatant from day 2 (1968 ± 324 pg/mL per µg DNA) through at least day 12 (888 ± 386 pg/mL per µg DNA) after transfection. CONCLUSIONS: Plasmid-mediated transfection of ovine MSCs to over-express paracrine heart-regenerative growth factors is feasible and efficient and overcomes the risks and limitations associated with the use of viral vectors.


Subject(s)
Bone Marrow Cells/cytology , Mesenchymal Stem Cells/cytology , Sheep, Domestic/genetics , Vascular Endothelial Growth Factor A/genetics , Animals , Cell- and Tissue-Based Therapy , Genetic Therapy , Heart/physiology , Humans , Myocytes, Cardiac/metabolism , Paracrine Communication , Plasmids , Regeneration , Transfection/methods , Vascular Endothelial Growth Factor A/metabolism
15.
Catheter Cardiovasc Interv ; 82(6): 899-906, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-22777825

ABSTRACT

OBJECTIVES: We aimed to assess safety and, secondarily, the efficacy of intramyocardial high-dose plasmid-vascular endothelial growth factor (VEGF) 165 (pVEGF165) gene transfer in no-option patients with coronary artery disease (CAD). BACKGROUND: Controlled trials of pVEGF165 in CAD have shown little benefit. One possible reason is shortness of dosage. We have shown in large mammalian models of chronic myocardial ischemia and acute myocardial infarction that intramyocardial pVEGF165 at doses significantly higher than those used in recent phase II trials is safe and efficacious on myocardial perfusion, left ventricular function, and infarct size limitation. METHODS: Using an injection catheter, 10 patients with severe CAD not amenable for revascularization received 10 intramyocardial injections of 0.38 mg (total dose, 3.8 mg) pVEGF165 in zones exhibiting myocardial ischemia, as assessed by combined stress 99mTc-sestamibi single-photon emission computed tomography and stress echocardiography. RESULTS: No serious adverse events related to either VEGF or the injection procedure occurred over the 2-year follow-up. One patient suffered femoral artery thrombosis after a follow-up coronary angiography, successfully resolved with medical treatment. Six patients suffered uncomplicated coronary ischemic events during the second year follow-up. Angina functional class decreased from 2.6 ± 0.2 to 1.2 ± 0.3 (mean ± SEM, P < 0.05), quality of life increased from 56.9 ± 3.2 to 82.6 ± 2.4 (P < 0.05), the summed difference score of myocardial perfusion decreased from 13.4 ± 2 to 7.7 ± 1.8 (P < 0.04), and stress ejection fraction did not change (44.2 ± 3.6% to 47.8 ± 3.1%, P = NS). CONCLUSIONS: High-dose intramyocardial pVEGF165 is safe at 2 years follow-up in patients with severe CAD. The efficacy results observed must be taken cautiously given the uncontrolled, open-label study design.


Subject(s)
Coronary Artery Disease/therapy , Gene Transfer Techniques , Genetic Therapy/methods , Neovascularization, Physiologic , Plasmids , Vascular Endothelial Growth Factor A/biosynthesis , Aged , Argentina , Collateral Circulation , Coronary Angiography , Coronary Artery Disease/diagnosis , Coronary Artery Disease/genetics , Coronary Artery Disease/metabolism , Coronary Artery Disease/physiopathology , Coronary Circulation , Echocardiography, Stress , Female , Genetic Therapy/adverse effects , Humans , Injections , Male , Middle Aged , Myocardial Perfusion Imaging/methods , Radiopharmaceuticals , Severity of Illness Index , Stroke Volume , Technetium Tc 99m Sestamibi , Time Factors , Tomography, Emission-Computed, Single-Photon , Treatment Outcome , Vascular Endothelial Growth Factor A/genetics
16.
Int J Cardiol ; 165(2): 291-8, 2013 May 10.
Article in English | MEDLINE | ID: mdl-21944383

ABSTRACT

BACKGROUND: In reperfused acute myocardial infarction (RAMI), cardioprotective treatments may enhance myocardial salvage and hence reduce the area of necrosis. Based on studies showing that plasmid-mediated vascular endothelial growth factor (pVEGF) gene transfer reduces infarct size by combining angio-arteriogenic and cardiomyogenic effects and that erythropoietin (EPO) exerts anti-apoptotic actions in animal models of AMI, we aimed to assess if their association would reduce infarct size to a larger extent than any of them individually in a large mammalian model of RAMI. METHODS: Adult sheep subjected to 90-minute coronary artery occlusion received upon reperfusion intramyocardial pVEGF 3.8 mg plus intravenous EPO 1000 IU/kg (n=8), pVEGF (n=8), EPO (n=8) or placebo (n=8). RESULTS: Fifteen days after treatment, infarct size was smaller in the 3 treatment groups (pVEGF+EPO: 8 ± 1 %; pVEGF: 16 ± 5 %; EPO: 13 ± 4 %) compared to placebo (25 ± 7 %, p<0.001). However, in the EPO+VEGF group infarct size was significantly smaller than in the groups receiving EPO or VEGF individually (p<0.05). DNA fragmentation, a hallmark of late apoptosis, was significantly lower in sheep receiving EPO. The combined treatment, while not affecting global left ventricular performance, improved regional peri-infarct function and prevented over-time expansion of the post-infarct perfusion defect. CONCLUSIONS: Combined pVEGF and EPO treatment might be clinically useful to enhance the benefits of early revascularization in patients with acute myocardial infarction.


Subject(s)
Erythropoietin/administration & dosage , Gene Transfer Techniques , Myocardial Infarction/drug therapy , Myocardial Reperfusion/methods , Vascular Endothelial Growth Factor A/administration & dosage , Vascular Endothelial Growth Factor A/genetics , Animals , Drug Therapy, Combination , Humans , Male , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Sheep, Domestic , Ventricular Function, Left/drug effects , Ventricular Function, Left/physiology
17.
J Gene Med ; 14(4): 279-87, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21954009

ABSTRACT

BACKGROUND: In large mammalian models of acute myocardial infarction (AMI), plasmid-mediated vascular endothelial growth factor (pVEGF) gene transfer has been shown to induce angio-arteriogenesis, proliferation of myocyte precursors and adult cardiomyocyte mitosis, reducing infarct size at 15 days after coronary artery occlusion. However, it is unknown whether these effects persist at longer follow-up times, nor how they affect cardiac performance. We thus assessed infarct size, left ventricular (LV) function and perfusion in 2-month-old ovine AMI. METHODS: Adult sheep with coronary artery occlusion were randomized to blindly receive ten intramyocardial injections of 3.8 mg of pVEGF or empty plasmid distributed at the infarct border. Three and 60 days later, LV perfusion (single-photon emission computed tomography) and function (stress echocardiography) were assessed. Finally, hemodynamics (LV catheterization), scar size and peri-infarct histology were studied. RESULTS: Infarct size was 30% smaller in pVEGF-treated sheep (23.6 ± 1.9% versus 32.7 ± 2.7% of the LV; p < 0.02). Percentage fractional shortening and wall thickening at the infarct border improved after pVEGF, as did myocardial perfusion and LV wall motion under pharmacological stress. Global LV function did not differ between groups, although the force-frequency response was preserved in pVEGF group and lost in placebo animals. These effects were associated with angio-arteriogenesis and proliferation of cardiomyocyte precursors. CONCLUSIONS: In sheep with AMI, pVEGF gene transfer affords long-term infarct size reduction, yielding regional LV function and perfusion improvement and reducing remodeling progression. These results suggest the potential usefulness of this approach in the clinical setting.


Subject(s)
Coronary Occlusion/therapy , Myocardial Infarction/therapy , Vascular Endothelial Growth Factor A/genetics , Ventricular Function, Left , Animals , Coronary Occlusion/complications , Coronary Occlusion/physiopathology , Gene Transfer Techniques , Humans , Myocardial Infarction/etiology , Myocardial Infarction/physiopathology , Sheep
18.
Article in English | MEDLINE | ID: mdl-23366854

ABSTRACT

UNLABELLED: Arterial stiffening is a common but highly variable disorder. Additionally, excessive arterial pulsatility is associated with various common diseases of aging and hypertension. Fractal dimension (FD) quantifies the time series complexity defined by its geometrical representation. OBJECTIVE: Arterial pressure and diameter time series were evaluated in order to assess the relationship between arterial stiffness and FD. METHODS: Three Corriedale male sheep were operated. Left anterior descending artery (LAD) was dissected and the external arterial diameter was measured trough sonomicrometry. Similarly, a pressure microtransducer was positioned in the upper third of the ascending aorta. Simultaneous pressure and diameter were measured in normal state and under smooth muscle activation. Each time series FD were assessed by the application of Higuchi's method while arterial wall elastic modulus was evaluated by means of the pressure-strain relationship. RESULTS: Coronary stiffness was increased from normal state to phenylephrine state (47.32%, 21.12%, 10.87%) while aortic pressure FD was decreased (2.11%, 2.57%, 6.85%), respectively. CONCLUSION: Acute hypertension induced by phenylephrine produces an increase in the coronary wall elastic modulus with a concomitant decrease in the fractal nature of the aortic pressure, suggesting that coronary stiffening is associated with an unwrinkled aortic pressure.


Subject(s)
Arterial Pressure , Coronary Artery Disease/physiopathology , Coronary Vessels/physiopathology , Models, Cardiovascular , Animals , Computer Simulation , Elastic Modulus , Fractals , Male , Sheep
19.
Int J Clin Exp Med ; 4(4): 258-64, 2011.
Article in English | MEDLINE | ID: mdl-22140597

ABSTRACT

Ovine models of ischemic heart disease and cardiac failure are increasingly used in translational research. However, reliable extrapolation of the results to the clinical setting requires knowing if ovine normal left ventricular (LV) function is comparable to that of humans. We thus assessed for echocardiographic LV dimensions and indexes in a large normal adult sheep population and compared them with standardized values in normal human adults. Bidimensional and tissue Doppler echocardiograms were performed in 69 young adult Corriedale sheep under light sedation. LV dimensions and indexes of systolic and diastolic function were measured. Absolute and body surface areanormalized values were compared to those for normal adult humans and their statistical distribution was assessed. Normalized dimensions (except for end diastolic diameter) as well as ejection fraction and fractional shortening fell within the ranges established by the American Society of Echocardiography and European Association of Echocardiography for normal adult humans. Normalized end diastolic diameter exceeded the upper normal limit but got close to it when correcting for the higher heart mass/body surface area ratio of sheep with respect to humans. Diastolic parameters also fell within normal human ranges except for a slightly lower mitral deceleration time. All values exhibited a Gaussian distribution. We conclude that echocardiographic parameters of systolic and diastolic LV performance in young adult sheep can be reliably extrapolated to the adult human, thus supporting the use of ovine models of human heart disease in translational research.

20.
ILAR J ; 52(1): E16-21, 2011.
Article in English | MEDLINE | ID: mdl-21454923

ABSTRACT

Studies on cardiac regeneration require large mammalian models of dilated cardiomyopathy (DCM) after acute myocardial infarction (AMI), and pig and sheep models are increasingly used in this field of preclinical research. Given the large interindividual variability in ovine left anterior descending artery (LAD) anatomy, protocols based on the coronary arteries to be ligated often lead to significant variation in infarct sizes and hence to heterogeneous results, ranging from no ventricular remodeling to acute, lethal left ventricular (LV) failure. We designed an ovine model of postinfarction DCM based on estimated infarct size rather than on a predetermined menu of coronary artery ligatures. In seven adult sheep we induced an anterolateral AMI of approximately 25% of the LV mass by ligating the branches of the LAD that, by visual inspection, would lead to such an infarct size. In 10 to 12 weeks, LV end-diastolic volume more than doubled and LV end-systolic volume almost tripled. LV ejection fraction decreased dramatically, as did LV percent fractional shortening and LV percent wall thickening. Infarct size (planimetry) was approximately 25% of the LV endocardial surface. We conclude that in sheep, an anterolateral AMI of approximately 25% of the LV mass--regardless of the coronary branches ligated to attain that infarct size--results in a model of postinfarction DCM that may prove useful in preclinical research on myocardial regeneration.


Subject(s)
Cardiomyopathy, Dilated/physiopathology , Myocardial Infarction/physiopathology , Animals , Cardiomyopathy, Dilated/pathology , Coronary Vessel Anomalies/pathology , Coronary Vessel Anomalies/physiopathology , Coronary Vessels/pathology , Coronary Vessels/physiopathology , Myocardial Infarction/pathology , Sheep , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...