Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
J Mol Cell Cardiol ; 178: 51-58, 2023 05.
Article in English | MEDLINE | ID: mdl-36965701

ABSTRACT

Cardiovascular disease is one of the leading causes of morbidity and mortality worldwide, with myocardial infarctions being amongst the deadliest manifestations. Reduced blood flow to the heart can result in the death of cardiac tissue, leaving affected patients susceptible to further complications and recurrent disease. Further, contemporary management typically involves a pharmacopeia to manage the metabolic conditions contributing to atherosclerotic and hypertensive heart disease, rather than regeneration of the damaged myocardium. With modern healthcare extending lifespan, a larger demographic will be at risk for heart disease, driving the need for novel therapeutics that surpass those currently available in efficacy. Transdifferentiation and cellular reprogramming have been looked to as potential methods for the treatment of diseases throughout the body. Specifically targeting the fibrotic cells in cardiac scar tissue as a source to be reprogrammed into induced cardiomyocytes remains an appealing option. This review aims to highlight the history of and advances in cardiac reprogramming and describe its translational potential as a treatment for cardiovascular disease.


Subject(s)
Heart Diseases , Myocardial Infarction , Humans , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Cellular Reprogramming/genetics , Heart Diseases/metabolism , Myocardial Infarction/metabolism , Technology
2.
Sci Rep ; 12(1): 11416, 2022 07 06.
Article in English | MEDLINE | ID: mdl-35794145

ABSTRACT

Direct cell reprogramming represents a promising new myocardial regeneration strategy involving in situ transdifferentiation of cardiac fibroblasts into induced cardiomyocytes. Adult human cells are relatively resistant to reprogramming, however, likely because of epigenetic restraints on reprogramming gene activation. We hypothesized that modulation of the epigenetic regulator gene p63 could improve the efficiency of human cell cardio-differentiation. qRT-PCR analysis demonstrated significantly increased expression of a panel of cardiomyocyte marker genes in neonatal rat and adult rat and human cardiac fibroblasts treated with p63 shRNA (shp63) and the cardio-differentiation factors Hand2/Myocardin (H/M) versus treatment with Gata4, Mef2c and Tbx5 (GMT) with or without shp63 (p < 0.001). FACS analysis demonstrated that shp63+ H/M treatment of human cardiac fibroblasts significantly increased the percentage of cells expressing the cardiomyocyte marker cTnT compared to GMT treatment with or without shp63 (14.8% ± 1.4% versus 4.3% ± 1.1% and 3.1% ± 0.98%, respectively; p < 0.001). We further demonstrated that overexpression of the p63-transactivation inhibitory domain (TID) interferes with the physical interaction of p63 with the epigenetic regulator HDAC1 and that human cardiac fibroblasts treated with p63-TID+ H/M demonstrate increased cardiomyocyte marker gene expression compared to cells treated with shp63+ H/M (p < 0.05). Whereas human cardiac fibroblasts treated with GMT alone failed to contract in co-culture experiments, human cardiac fibroblasts treated with shp63+ HM or p63-TID+ H/M demonstrated calcium transients upon electrical stimulation and contractility synchronous with surrounding neonatal cardiomyocytes. These findings demonstrate that p63 silencing provides enhanced rat and human cardiac fibroblast transdifferentiation into induced cardiomyocytes compared to a standard reprogramming strategy. p63-TID overexpression may be a useful reprogramming strategy for overcoming epigenetic barriers to human fibroblast cardio-differentiation.


Subject(s)
Myocytes, Cardiac , T-Box Domain Proteins , Animals , Cellular Reprogramming , Epigenesis, Genetic , Fibroblasts/metabolism , Humans , Membrane Proteins/genetics , Myocytes, Cardiac/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Rats , T-Box Domain Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
3.
J Am Heart Assoc ; 10(24): e022659, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34889103

ABSTRACT

Background The conversion of fibroblasts into induced cardiomyocytes may regenerate myocardial tissue from cardiac scar through in situ cell transdifferentiation. The efficiency transdifferentiation is low, especially for human cells. We explored the leveraging of Hippo pathway intermediates to enhance induced cardiomyocyte generation. Methods and Results We screened Hippo effectors Yap (yes-associated protein), Taz (transcriptional activator binding domain), and Tead1 (TEA domain transcription factor 1; Td) for their reprogramming efficacy with cardio-differentiating factors Gata4, Mef2C, and Tbx5 (GMT). Td induced nearly 3-fold increased expression of cardiomyocyte marker cTnT (cardiac troponin T) by mouse embryonic and adult rat fibroblasts versus GMT administration alone (P<0.0001), while Yap and Taz failed to enhance cTnT expression. Serial substitution demonstrated that Td replacement of TBX5 induced the greatest cTnT expression enhancement and sarcomere organization in rat fibroblasts treated with all GMT substitutions (GMTd versus GMT: 17±1.2% versus 5.4±0.3%, P<0.0001). Cell contractility (beating) was seen in 6% of GMTd-treated cells by 4 weeks after treatment, whereas no beating GMT-treated cells were observed. Human cardiac fibroblasts likewise demonstrated increased cTnT expression with GMTd versus GMT treatment (7.5±0.3% versus 3.0±0.3%, P<0.01). Mechanistically, GMTd administration increased expression of the trimethylated lysine 4 of histone 3 (H3K4me3) mark at the promoter regions of cardio-differentiation genes and mitochondrial biogenesis regulator genes in rat and human fibroblast, compared with GMT. Conclusions These data suggest that the Hippo pathway intermediate Tead1 is an important regulator of cardiac reprogramming that increases the efficiency of maturate induced cardiomyocytes generation and may be a vital component of human cardiodifferentiation strategies.


Subject(s)
Fibroblasts , Hippo Signaling Pathway , Myocytes, Cardiac , TEA Domain Transcription Factors , Animals , Cell Transdifferentiation , Fibroblasts/physiology , Mice , Myocytes, Cardiac/physiology , Rats , TEA Domain Transcription Factors/metabolism
4.
Sci Rep ; 11(1): 22605, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34799643

ABSTRACT

Fibroblast reprogramming offers the potential for myocardial regeneration via in situ cell transdifferentiation. We explored a novel strategy leveraging endothelial cell plasticity to enhance reprogramming efficiency. Rat cardiac endothelial cells and fibroblasts were treated with Gata4, Mef2c, and Tbx5 (GMT) to assess the cardio-differentiation potential of these cells. The endothelial cell transdifferentiation factor ETV2 was transiently over-expressed in fibroblasts followed by GMT treatment to assess "trans-endothelial" cardio-differentiation. Endothelial cells treated with GMT generated more cTnT+ cells than did cardiac fibroblasts (13% ± 2% vs 4% ± 0.5%, p < 0.01). Cardiac fibroblasts treated with ETV2 demonstrated increased endothelial cell markers, and when then treated with GMT yielded greater prevalence of cells expressing cardiomyocyte markers including cTnT than did fibroblasts treated with GMT or ETV2 (10.3% ± 0.2% vs 1.7% ± 0.06% and 0.6 ± 0.03, p < 0.01). Rat cardiac fibroblasts treated with GMT + ETV2 demonstrated calcium transients upon electrical stimulation and contractility synchronous with surrounding neonatal cardiomyocytes, whereas cells treated with GMT or ETV2 alone failed to contract in co-culture experiments. Human cardiac fibroblasts treated with ETV2 and then GMT likewise demonstrated greater prevalence of cTnT expression than did cells treated with GMT alone (2.8-fold increase, p < 0.05). Cardiac fibroblast transitioning through a trans-endothelial state appears to enhance cardio-differentiation by enhancing fibroblast plasticity.


Subject(s)
Cell Transdifferentiation , Cellular Reprogramming , Endothelium/metabolism , Fibroblasts/metabolism , Animals , Animals, Newborn , Cell Plasticity , Cell Separation , Coculture Techniques , Endothelial Cells/metabolism , Flow Cytometry , Humans , Myocytes, Cardiac/metabolism , Prevalence , Rats
5.
Biomater Sci ; 8(18): 5061-5070, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32797143

ABSTRACT

Paracrine factors secreted by mesenchymal stem cells (MSCs) have been previously shown to improve cardiac function following acute myocardial infarction (MI). However, cell therapy activates the innate immune response, leading to the rapid elimination of transplanted cells and only short-term therapeutic delivery. Herein, we describe a new strategy to deliver sustained paracrine-mediated MSC therapy to ischemic myocardium. Using an immune evasive, small molecule modified alginate, we encapsulated rat MSC cells in a core-shell hydrogel capsule and implanted them in the pericardial sac of post-MI rats. Encapsulated cells allowed diffusion of reparative paracrine factors at levels similar to non-encapsulated cells in vitro. Encapsulation enabled sustained cell survival with localization over the heart for 2 weeks. The effect of the experimental group on ventricular function and fibrosis was compared with blank (cell free) capsules and unencapsulated MSCs injected into infarcted myocardium. MSC capsules improved post-MI ventricular function ∼2.5× greater than MSC injection. After 4 weeks, post-MI fibrosis was reduced ∼2/3 with MSC capsules, but unchanged with MSC injection. MSC encapsulation with alginate core-shell capsules sustains cell survival and potentiates efficacy of therapy.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Myocardial Infarction , Alginates , Animals , Myocardial Infarction/therapy , Myocardium , Rats
6.
J Am Heart Assoc ; 9(12): e015686, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32500803

ABSTRACT

Background Given known inefficiencies in reprogramming of fibroblasts into mature induced cardiomyocytes (iCMs), we sought to identify small molecules that would overcome these barriers to cardiac cell transdifferentiation. Methods and Results We screened alternative combinations of compounds known to impact cell reprogramming using morphologic and functional cell differentiation assays in vitro. After screening 6 putative reprogramming factors, we found that a combination of the histone deacetylase inhibitor sodium butyrate, the WNT inhibitor ICG-001, and the cardiac growth regulator retinoic acid (RA) maximally enhanced iCM generation from primary rat cardiac fibroblasts when combined with administration of the cardiodifferentiating transcription factors Gata4, Mef2C, and Tbx5 (GMT) compared with GMT administration alone (23±1.5% versus 3.3±0.2%; P<0.0001). Expression of the cardiac markers cardiac troponin T, Myh6, and Nkx2.5 was upregulated as early as 10 days after GMT-sodium butyrate, ICG-001, and RA treatment. Human iCM generation was likewise enhanced when administration of the human cardiac reprogramming factors GMT, Hand2, and Myocardin plus miR-590 was combined with sodium butyrate, ICG-001, and RA compared with GMT, Hand2, and Myocardin plus miR-590 treatment alone (25±1.3% versus 5.7±0.4%; P<0.0001). Rat and human iCMs also more frequently demonstrated spontaneous beating in coculture with neonatal cardiomyocytes with the addition of sodium butyrate, ICG-001, and RA to transcription factor cocktails compared with transcription factor treatment alone. Conclusions The combined administration of histone deacetylase and WNT inhibitors with RA enhances rat and human iCM generation induced by transcription factor administration alone. These findings suggest opportunities for improved translational approaches for cardiac regeneration.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Butyric Acid/pharmacology , Cell Transdifferentiation/drug effects , Cellular Reprogramming Techniques , Cellular Reprogramming/drug effects , Fibroblasts/drug effects , Histone Deacetylase Inhibitors/pharmacology , Myocytes, Cardiac/drug effects , Pyrimidinones/pharmacology , Tretinoin/pharmacology , Animals , Cells, Cultured , Fibroblasts/metabolism , Gene Expression Regulation , Humans , Male , Myocytes, Cardiac/metabolism , Phenotype , Rats, Sprague-Dawley , Transcription Factors/genetics , Transcription Factors/metabolism , Wnt Signaling Pathway/drug effects
7.
J Thorac Cardiovasc Surg ; 156(2): 556-565.e1, 2018 08.
Article in English | MEDLINE | ID: mdl-29716728

ABSTRACT

OBJECTIVE: Reprogramming of fibroblasts into induced cardiomyocytes represents a potential new therapy for heart failure. We hypothesized that inactivation of p63, a p53 gene family member, may help overcome human cell resistance to reprogramming. METHODS: p63 Knockout (-/-) and knockdown murine embryonic fibroblasts (MEFs), p63-/- adult murine cardiac fibroblasts, and human cardiac fibroblasts were assessed for cardiomyocyte-specific feature changes, with or without treatment by the cardiac transcription factors Hand2-Myocardin (HM). RESULTS: Flow cytometry revealed that a significantly greater number of p63-/- MEFs expressed the cardiac-specific marker cardiac troponin T (cTnT) in culture compared with wild-type (WT) cells (38% ± 11% vs 0.9% ± 0.9%, P < .05). HM treatment of p63-/- MEFs increased cTnT expression to 74% ± 3% of cells but did not induce cTnT expression in wild-type murine embryonic fibroblasts. shRNA-mediated p63 knockdown likewise yielded a 20-fold increase in cTnT microRNA expression compared with untreated MEFs. Adult murine cardiac fibroblasts demonstrated a 200-fold increase in cTnT gene expression after inducible p63 knockout and expressed sarcomeric α-actinin as well as cTnT. These p63-/- adult cardiac fibroblasts exhibited calcium transients and electrically stimulated contractions when co-cultured with neonatal rat cardiomyocytes and treated with HM. Increased expression of cTnT and other marker genes was also observed in p63 knockdown human cardiac fibroblasts procured from patients undergoing procedures for heart failure. CONCLUSIONS: Downregulation of p63 facilitates direct cardiac cellular reprogramming and may help overcome the resistance of human cells to reprogramming.


Subject(s)
Cellular Reprogramming/genetics , Fibroblasts/cytology , Gene Silencing/physiology , Myocytes, Cardiac/cytology , Phosphoproteins/genetics , Trans-Activators/genetics , Animals , Cells, Cultured , Humans , Mice , Rats , Troponin T/analysis , Troponin T/metabolism
9.
J Hematol Oncol ; 10(1): 159, 2017 10 03.
Article in English | MEDLINE | ID: mdl-28974232

ABSTRACT

BACKGROUND: The stem cell factor spalt-like transcription factor 4 (SALL4) plays important roles in normal hematopoiesis and also in leukemogenesis. We previously reported that SALL4 exerts its effect by recruiting important epigenetic factors such as DNA methyltransferases DNMT1 and lysine-specific demethylase 1 (LSD1/KDM1A). Both of these proteins are critically involved in mixed lineage leukemia (MLL)-rearranged (MLL-r) leukemia, which has a very poor clinical prognosis. Recently, SALL4 has been further linked to the functions of MLL and its target gene homeobox A9 (HOXA9). However, it remains unclear whether SALL4 is indeed a key player in MLL-r leukemia pathogenesis. METHODS: Using a mouse bone marrow retroviral transduction/ transplantation approach combined with tamoxifen-inducible, CreERT2-mediated Sall4 gene deletion, we studied SALL4 functions in leukemic transformation that was induced by MLL-AF9-one of the most common MLL-r oncoproteins found in patients. In addition, the underlying transcriptional and epigenetic mechanisms were explored using chromatin immunoprecipitation (ChIP) sequencing (ChIP-Seq), mRNA microarray, qRT-PCR, histone modification, co-immunoprecipitation (co-IP), cell cycle, and apoptosis assays. The effects of SALL4 loss on normal hematopoiesis in mice were also investigated. RESULTS: In vitro and in vivo studies revealed that SALL4 expression is critically required for MLL-AF9-induced leukemic transformation and disease progression in mice. Loss of SALL4 in MLL-AF9-transformed cells induced apoptosis and cell cycle arrest at G1. ChIP-Seq assay identified that Sall4 binds to key MLL-AF9 target genes and important MLL-r or non-MLL-r leukemia-related genes. ChIP-PCR assays indicated that SALL4 affects the levels of the histone modification markers H3K79me2/3 and H3K4me3 at MLL-AF9 target gene promoters by physically interacting with DOT1-like histone H3K79 methyltransferase (DOT1l) and LSD1/KDM1A, and thereby regulates transcript expression. Surprisingly, normal Sall4 f/f /CreERT2 mice treated with tamoxifen or vav-Cre-mediated (hematopoietic-specific) Sall4 -/- mice were healthy and displayed no significant hematopoietic defects. CONCLUSIONS: Our findings indicate that SALL4 critically contributes to MLL-AF9-induced leukemia, unraveling the underlying transcriptional and epigenetic mechanisms in this disease and suggesting that selectively targeting the SALL4 pathway may be a promising approach for managing human MLL-r leukemia.


Subject(s)
DNA-Binding Proteins/genetics , Hematopoietic Stem Cells/physiology , Histones/metabolism , Leukemia/genetics , Transcription Factors/genetics , Animals , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/metabolism , Leukemia/pathology , Mice , Mice, Transgenic , Transcription Factors/metabolism
10.
J Thorac Cardiovasc Surg ; 154(5): 1601-1610.e3, 2017 11.
Article in English | MEDLINE | ID: mdl-28711329

ABSTRACT

OBJECTIVE: The administration of a variety of reprogramming factor cocktails has now been shown to reprogram cardiac fibroblasts into induced cardiomyocyte-like cells. However, reductions in ventricular fibrosis observed after reprogramming factor administration seem to far exceed the extent of induced cardiomyocyte-like cell generation in vivo. We investigated whether reprogramming factor administration might primarily play a role in activating antifibrotic molecular pathways. METHODS: Adult rat cardiac fibroblasts were infected with lentivirus encoding the transcription factors Gata4, Mef2c, or Tbx5, all 3 vectors, or a green fluorescent protein control vector. Gene and protein expression assays were performed to identify relevant antifibrotic targets of these factors. The antifibrotic effects of these factors were then investigated in a rat coronary ligation model. RESULTS: Gata4, Mef2c, or Tbx5 administration to rat cardiac fibroblasts in vitro significantly downregulated expression of Snail and the profibrotic factors connective tissue growth factor, collagen1a1, and fibronectin. Of these factors, Gata4 was shown to be the one responsible for the downregulation of the profibrotic factors and Snail (mRNA expression fold change relative to green fluorescent protein for Snail, Gata4: 0.5 ± 0.3, Mef2c: 1.3 ± 1.0, Tbx5: 0.9 ± 0.5, Gata4, Mef2c, or Tbx5: 0.6 ± 0.2, P < .05). Chromatin immunoprecipitation quantitative polymerase chain reaction identified Gata4 binding sites in the Snail promoter. In a rat coronary ligation model, only Gata4 administration alone improved postinfarct ventricular function and reduced the extent of postinfarct fibrosis. CONCLUSIONS: Gata4 administration reduces postinfarct ventricular fibrosis and improves ventricular function in a rat coronary ligation model, potentially as a result of Gata4-mediated downregulation of the profibrotic mediator Snail.


Subject(s)
Cellular Reprogramming/genetics , Fibroblasts/physiology , Fibrosis , GATA4 Transcription Factor , Lentivirus , Myocytes, Cardiac/physiology , Animals , Cellular Reprogramming Techniques , Collagen Type I/analysis , Collagen Type I, alpha 1 Chain , Connective Tissue Growth Factor/analysis , Down-Regulation , Fibronectins/analysis , Fibrosis/metabolism , Fibrosis/prevention & control , GATA4 Transcription Factor/metabolism , GATA4 Transcription Factor/pharmacology , Genetic Vectors , MEF2 Transcription Factors/metabolism , MEF2 Transcription Factors/pharmacology , Rats , Signal Transduction , Snail Family Transcription Factors , T-Box Domain Proteins/metabolism , T-Box Domain Proteins/pharmacokinetics , Zinc Fingers
11.
J Thorac Cardiovasc Surg ; 153(2): 329-339.e3, 2017 02.
Article in English | MEDLINE | ID: mdl-27773576

ABSTRACT

OBJECTIVE: The reprogramming of cardiac fibroblasts into induced cardiomyocyte-like cells improves ventricular function in myocardial infarction models. Only integrating persistent expression vectors have thus far been used to induce reprogramming, potentially limiting its clinical applicability. We therefore tested the reprogramming potential of nonintegrating, acute expression adenoviral (Ad) vectors. METHODS: Ad or lentivirus vectors encoding Gata4 (G), Mef2c (M), and Tbx5 (T) were validated in vitro. Sprague-Dawley rats then underwent coronary ligation and Ad-mediated administration of vascular endothelial growth factor to generate infarct prevascularization. Three weeks later, animals received Ad or lentivirus encoding G, M, or T (AdGMT or LentiGMT) or an equivalent dose of a null vector (n = 11, 10, and 10, respectively). Outcomes were analyzed by echocardiography, magnetic resonance imaging, and histology. RESULTS: Ad and lentivirus vectors provided equivalent G, M, and T expression in vitro. AdGMT and LentiGMT both likewise induced expression of the cardiomyocyte marker cardiac troponin T in approximately 6% of cardiac fibroblasts versus <1% cardiac troponin T expression in AdNull (adenoviral vector that does not encode a transgene)-treated cells. Infarcted myocardium that had been treated with AdGMT likewise demonstrated greater density of cells expressing the cardiomyocyte marker beta myosin heavy chain 7 compared with AdNull-treated animals. Echocardiography demonstrated that AdGMT and LentiGMT both increased ejection fraction compared with AdNull (AdGMT: 21% ± 3%, LentiGMT: 14% ± 5%, AdNull: -0.4% ± 2%; P < .05). CONCLUSIONS: Ad vectors are at least as effective as lentiviral vectors in inducing cardiac fibroblast transdifferentiation into induced cardiomyocyte-like cells and improving cardiac function in postinfarct rat hearts. Short-term expression Ad vectors may represent an important means to induce cardiac cellular reprogramming in humans.


Subject(s)
Cellular Reprogramming Techniques/methods , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , Regeneration , Adenoviridae , Animals , Cell Transdifferentiation , Disease Models, Animal , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Transfer Techniques , Genetic Vectors , Male , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/pharmacology
12.
J Am Heart Assoc ; 5(11)2016 11 10.
Article in English | MEDLINE | ID: mdl-27930352

ABSTRACT

BACKGROUND: Reprogramming of cardiac fibroblasts into induced cardiomyocyte-like cells represents a promising potential new therapy for treating heart disease, inducing significant improvements in postinfarct ventricular function in rodent models. Because reprogramming factors effective in transdifferentiating rodent cells are not sufficient to reprogram human cells, we sought to identify reprogramming factors potentially applicable to human studies. METHODS AND RESULTS: Lentivirus vectors expressing Gata4, Mef2c, and Tbx5 (GMT); Hand2 (H), Myocardin (My), or microRNA (miR)-590 were administered to rat, porcine, and human cardiac fibroblasts in vitro. induced cardiomyocyte-like cell production was then evaluated by assessing expression of the cardiomyocyte marker, cardiac troponin T (cTnT), whereas signaling pathway studies were performed to identify reprogramming factor targets. GMT administration induced cTnT expression in ≈6% of rat fibroblasts, but failed to induce cTnT expression in porcine or human cardiac fibroblasts. Addition of H/My and/or miR-590 to GMT administration resulted in cTNT expression in ≈5% of porcine and human fibroblasts and also upregulated the expression of the cardiac genes, MYH6 and TNNT2. When cocultured with murine cardiomyocytes, cTnT-expressing porcine cardiac fibroblasts exhibited spontaneous contractions. Administration of GMT plus either H/My or miR-590 alone also downregulated fibroblast genes COL1A1 and COL3A1. miR-590 was shown to directly suppress the zinc finger protein, specificity protein 1 (Sp1), which was able to substitute for miR-590 in inducing cellular reprogramming. CONCLUSIONS: These data support porcine studies as a surrogate for testing human cardiac reprogramming, and suggest that miR-590-mediated repression of Sp1 represents an alternative pathway for enhancing human cardiac cellular reprogramming.


Subject(s)
Cell Transdifferentiation/genetics , Cellular Reprogramming Techniques/methods , Fibroblasts/cytology , MicroRNAs/genetics , Myocytes, Cardiac/cytology , Sp1 Transcription Factor/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Flow Cytometry , GATA4 Transcription Factor/genetics , Genetic Vectors , Humans , In Vitro Techniques , MEF2 Transcription Factors/genetics , Nuclear Proteins/genetics , Rats , Swine , T-Box Domain Proteins/genetics , Trans-Activators/genetics
13.
Article in English | MEDLINE | ID: mdl-27554127

ABSTRACT

OPINION STATEMENT: Direct cardiac cellular reprogramming of endogenous cardiac fibroblasts directly into induced cardiomyocytes is a highly feasible, promising therapeutic option for patients with advanced heart failure. The most successful cardiac reprogramming strategy will likely be a multimodal approach involving an optimal combination of cardio-differentiating factors, suppression of fibroblast gene expression, and induction of angiogenic factors.

14.
J Thorac Cardiovasc Surg ; 148(4): 1656-1664.e2, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24755332

ABSTRACT

OBJECTIVE: The in situ reprogramming of cardiac fibroblasts into induced cardiomyocytes by the administration of gene transfer vectors encoding Gata4 (G), Mef2c (M), and Tbx5 (T) has been shown to improve ventricular function in myocardial infarction models. The efficacy of this strategy could, however, be limited by the need for fibroblast targets to be infected 3 times--once by each of the 3 transgene vectors. We hypothesized that a polycistronic "triplet" vector encoding all 3 transgenes would enhance postinfarct ventricular function compared with use of "singlet" vectors. METHODS: After validation of the polycistronic vector expression in vitro, adult male Fischer 344 rats (n=6) underwent coronary ligation with or without intramyocardial administration of an adenovirus encoding all 3 major vascular endothelial growth factor (VEGF) isoforms (AdVEGF-All6A positive), followed 3 weeks later by the administration to AdVEGF-All6A-positive treated rats of singlet lentivirus encoding G, M, or T (1×10(5) transducing units each) or the same total dose of a GMT "triplet" lentivirus vector. RESULTS: Western blots demonstrated that triplet and singlet vectors yielded equivalent GMT transgene expression, and fluorescence activated cell sorting demonstrated that triplet vectors were nearly twice as potent as singlet vectors in generating induced cardiomyocytes from cardiac fibroblasts. Echocardiography demonstrated that GMT triplet vectors were more effective than the 3 combined singlet vectors in enhancing ventricular function from postinfarct baselines (triplet, 37%±10%; singlet, 13%±7%; negative control, 9%±5%; P<.05). CONCLUSIONS: These data have confirmed that the in situ administration of G, M, and T induces postinfarct ventricular functional improvement and that GMT polycistronic vectors enhance the efficacy of this strategy.


Subject(s)
Cell Differentiation/genetics , GATA4 Transcription Factor/genetics , Gene Transfer Techniques , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , Myogenic Regulatory Factors/genetics , T-Box Domain Proteins/genetics , Vascular Endothelial Growth Factor A/genetics , Adenoviridae/genetics , Animals , Blotting, Western , Cell Differentiation/physiology , Fibroblasts/pathology , GATA4 Transcription Factor/physiology , Genetic Vectors , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/physiology , Male , Models, Animal , Myocytes, Cardiac/physiology , Myogenic Regulatory Factors/physiology , Rats , Rats, Inbred F344 , T-Box Domain Proteins/physiology
15.
J Am Heart Assoc ; 1(6): e005652, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23316332

ABSTRACT

BACKGROUND: In situ cellular reprogramming offers the possibility of regenerating functional cardiomyocytes directly from scar fibroblasts, obviating the challenges of cell implantation. We hypothesized that pretreating scar with gene transfer of the angiogenic vascular endothelial growth factor (VEGF) would enhance the efficacy of this strategy. METHODS AND RESULTS: Gata4, Mef2c, and Tbx5 (GMT) administration via lentiviral transduction was demonstrated to transdifferentiate rat fibroblasts into (induced) cardiomyocytes in vitro by cardiomyocyte marker studies. Fisher 344 rats underwent coronary ligation and intramyocardial administration of an adenovirus encoding all 3 major isoforms of VEGF (AdVEGF-All6A(+)) or an AdNull control vector (n=12/group). Lentivirus encoding GMT or a GFP control was administered to each animal 3 weeks later, followed by histologic and echocardiographic analyses. GMT administration reduced the extent of fibrosis by half compared with GFP controls (12 ± 2% vs 24 ± 3%, P<0.01) and reduced the number of myofibroblasts detected in the infarct zone by 4-fold. GMT-treated animals also demonstrated greater density of cardiomyocyte-specific marker beta myosin heavy chain 7(+) cells compared with animals receiving GFP with or without VEGF (P<0.01). Ejection fraction was significantly improved after GMT vs GFP administration (12 ± 3% vs -7 ± 3%, P<0.01). Eight (73%) GFP animals but no GMT animals demonstrated decreased ejection fraction during this interval (P<0.01). Also, improvement in ejection fraction was 4-fold greater in GMT/VEGF vs GMT/null animals (17 ± 2% vs 4 ± 1%, P<0.05). CONCLUSIONS: VEGF administration to infarcted myocardium enhances the efficacy of GMT-mediated cellular reprogramming in improving myocardial function and reducing the extent of myocardial fibrosis compared with the use of GMT or VEGF alone.


Subject(s)
Myocardial Infarction/drug therapy , Neovascularization, Physiologic/physiology , Transcription Factors/administration & dosage , Vascular Endothelial Growth Factor A/administration & dosage , Ventricular Function/physiology , Animals , Drug Therapy, Combination , Echocardiography , Fibrosis/prevention & control , Fluorescent Antibody Technique , GATA4 Transcription Factor/administration & dosage , Gene Transfer Techniques , MEF2 Transcription Factors , Myocardial Infarction/physiopathology , Myocytes, Cardiac/physiology , Myogenic Regulatory Factors/administration & dosage , Neovascularization, Physiologic/drug effects , Rats , Rats, Inbred F344 , T-Box Domain Proteins/administration & dosage , Transfection , Treatment Outcome , Ventricular Function/drug effects
16.
Cardiovasc Revasc Med ; 6(3): 113-20, 2005.
Article in English | MEDLINE | ID: mdl-16275607

ABSTRACT

Extracellular matrix (ECM) scaffolds may be useful as a tissue engineering approach toward myocardial regeneration in the infarcted heart. An appropriate large-animal model for testing the utility of biologically derived ECM in this application is needed. The purpose of this study was to develop such a model for optimal procedural success during and after patch implantation surgery. Myocardial infarction (MI) was created by embolization of the diagonal artery (DA) branch of the left anterior descending coronary artery with collagen suspension. After 4 to 6 weeks, 14 pigs received patch implant (ECM or expanded polytetrafluoroethylene). Six pigs were infarcted in the first DA and seven pigs in the second DA. Electrophysiology study was performed within 3 days before surgery. During surgery, the size and location of the infarct were measured. Infarcted myocardium (1.5-cm diameter) was transmurally excised under partial cardiopulmonary bypass. Patches (3-cm diameter) were sutured to the endomyocardial defect. Four pigs died postoperatively. After 1 month, 10 pigs were euthanized and the locations of patches were examined. Success rate of patch implant in the second DA (85.7%) was higher than the first DA (50%) group. Infarct size in the second DA was smaller than in the first DA (4.6+/-1.2 vs. 10.8+/-2.4 cm(2), P<.05). The second DA was more anteriorly positioned, which enabled easier access from the midsternal thoracotomy. However, the first DA was more laterally located requiring more manipulation of the heart during surgery. Electrophysiology revealed no ventricular tachyarrhythmia in the second DA but 33.3% in the first DA group (P<.05). At necropsy, the endocardial position of the first DA-infarct patches was anteroapical, whereas the second DA-infarct patches were more basolateral and often involved the anterior papillary muscle. The success rate of patch implant was associated with infarction size and location, and may be related to arrhythmic substrate. Experimental MI created by the second DA embolization is a feasible model for investigation of tissue-engineered cardiac patch implantation. This large-animal model is also suitable for study of cell therapy via endocardial catheter-based approaches or open surgical methods.


Subject(s)
Cardiac Surgical Procedures , Disease Models, Animal , Tissue Engineering , Animals , Coronary Angiography , Coronary Vessels/diagnostic imaging , Coronary Vessels/pathology , Coronary Vessels/surgery , Echocardiography , Electrophysiologic Techniques, Cardiac , Extracellular Matrix/transplantation , Female , Male , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/pathology , Myocardial Infarction/surgery , Swine
17.
Circulation ; 112(9 Suppl): I135-43, 2005 Aug 30.
Article in English | MEDLINE | ID: mdl-16159805

ABSTRACT

BACKGROUND: Heart failure remains a significant problem. Tissue-engineered cardiac patches offer potential to treat severe heart failure. We studied an extracellular matrix scaffold for repairing the infarcted left ventricle. METHODS AND RESULTS: Pigs (n=42) underwent left ventricular (LV) infarction. At 6 to 8 weeks, either 4-layer multilaminate urinary bladder-derived extracellular matrix or expanded polytetrafluoroethlyene (ePTFE) was implanted as full-thickness LV wall patch replacement. At 1-week, 1-month, or 3-month intervals, pigs were terminated. After macroscopic examination, samples of tissue were prepared for histology, immunocytochemistry, and analysis of cell proportions by flow cytometry. One-week and 1-month patches were intact with thrombus and inflammation; at 1 month, there was also tissue with spindle-shaped cells in proteoglycan-rich and collagenous matrix. More alpha-smooth muscle actin-positive cells were present in urinary bladder matrix (UBM) than in ePTFE (22.2+/-3.3% versus 8.4+/-2.7%; P=0.04). At 3 months, UBM was bioresorbed, and a collagen-rich vascularized tissue with numerous myofibroblasts was present. Isolated regions of alpha-sarcomeric actin-positive, intensely alpha-smooth muscle actin-immunopositive, and striated cells were observed. ePTFE at 3 months had foreign-body response with necrosis and calcification. Flow cytometry showed similarities of cells from UBM to normal myocardium, whereas ePTFE had limited cardiomyocyte markers. CONCLUSIONS: Appearance of a fibrocellular tissue that included contractile cells accompanied biodegradation of UBM when implanted as an LV-free wall infarction patch. UBM appears superior to synthetic material for cardiac patching and trends toward myocardial replacement at 3 months.


Subject(s)
Extracellular Matrix/transplantation , Heart Failure/surgery , Prostheses and Implants , Tissue Engineering , Absorbable Implants , Animals , Biocompatible Materials , Biomarkers , Female , Flow Cytometry , Heart Failure/etiology , Heart Ventricles/surgery , Male , Materials Testing , Myocardial Infarction/complications , Myocardium/pathology , Polytetrafluoroethylene , Sus scrofa , Urinary Bladder/ultrastructure , Wound Healing
18.
J Vasc Interv Radiol ; 14(1): 33-9, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12525584

ABSTRACT

PURPOSE: Despite careful preoperative assessment, problematic access to the abdominal aorta for transluminal endografting (TE) of abdominal aortic aneurysm (AAA) is sometimes encountered. This study identifies preoperative risk factors predictive of problematic access and determines the impact of problematic access on outcomes. MATERIALS AND METHODS: Three hundred twenty-one consecutive TE procedures for AAA were divided into two groups: group A, which had access problems (n = 74), and group B, which had none (n = 247). RESULTS: Logistic regression analysis of risk factors showed that (i) a pulmonary risk score of 3 (P <.001; odds ratio, 11.2), (ii) a hyperlipidemia score of 3 (P =.004; odds ratio, 2.6), and (iii) a small body (short height with low weight, P =.003; odds ratio, of 4.2) were independent risk factors for problematic access. Outcomes compared included rates of perioperative mortality, aborted procedure, surgical conversion, major complication, limb complication, and endoleak. The perioperative mortality rate was higher in group A (6.8% vs 1.2%; P =.018) and procedures were more likely to be aborted in patients in group A (12.2% vs 0%; P <.001). Rates of surgical conversion, major complication, and limb complication were not different between the two groups. The endoleak rate was higher in group B (10.8% vs 26.7%; P =.004). Logistic regression analysis showed that problematic access was an independent risk factor (P =.004; odds ratio, 12.0) for perioperative mortality. CONCLUSIONS: Moderate to severe chronic obstructive pulmonary disease and hyperlipidemia, both risk factors for atherosclerosis, were related to problematic access in this series. Small body size was another factor related to problematic access. Problematic access was an independent risk factor for perioperative mortality.


Subject(s)
Aortic Aneurysm, Abdominal/surgery , Blood Vessel Prosthesis Implantation/methods , Aged , Aged, 80 and over , Aortic Aneurysm, Abdominal/complications , Aortic Aneurysm, Abdominal/mortality , Blood Vessel Prosthesis Implantation/mortality , Female , Humans , Hyperlipidemias/complications , Logistic Models , Male , Outcome Assessment, Health Care , Postoperative Complications , Pulmonary Disease, Chronic Obstructive/complications , Retrospective Studies , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...