Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Dev Cell ; 34(4): 387-99, 2015 Aug 24.
Article in English | MEDLINE | ID: mdl-26256209

ABSTRACT

Some organisms, such as adult zebrafish and newborn mice, have the capacity to regenerate heart tissue following injury. Unraveling the mechanisms of heart regeneration is fundamental to understanding why regeneration fails in adult humans. Numerous studies have revealed that nerves are crucial for organ regeneration, thus we aimed to determine whether nerves guide heart regeneration. Here, we show using transgenic zebrafish that inhibition of cardiac innervation leads to reduction of myocyte proliferation following injury. Specifically, pharmacological inhibition of cholinergic nerve function reduces cardiomyocyte proliferation in the injured hearts of both zebrafish and neonatal mice. Direct mechanical denervation impairs heart regeneration in neonatal mice, which was rescued by the administration of neuregulin 1 (NRG1) and nerve growth factor (NGF) recombinant proteins. Transcriptional analysis of mechanically denervated hearts revealed a blunted inflammatory and immune response following injury. These findings demonstrate that nerve function is required for both zebrafish and mouse heart regeneration.


Subject(s)
Cholinergic Neurons/physiology , Heart/innervation , Heart/physiology , Myocytes, Cardiac/cytology , Regeneration , Animals , Animals, Genetically Modified , Animals, Newborn , Cell Proliferation/drug effects , Denervation , Gene Expression Regulation/drug effects , Immunity/drug effects , Immunity/genetics , Inflammation/genetics , Mice , Models, Biological , Molecular Sequence Data , Nerve Growth Factor/pharmacology , Neuregulin-1/pharmacology , Regeneration/drug effects , Synaptic Transmission/drug effects , Vagotomy , Zebrafish
2.
Proc Natl Acad Sci U S A ; 112(23): 7249-54, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-25941360

ABSTRACT

Hypertension increases the pressure load on the heart and is associated with a poorly understood chronic systemic inflammatory state. Interleukin 33 (IL-33) binds to membrane-bound ST2 (ST2L) and has antihypertrophic and antifibrotic effects in the myocardium. In contrast, soluble ST2 appears to act as a decoy receptor for IL-33, blocking myocardial and vascular benefits, and is a prognostic biomarker in patients with cardiovascular diseases. Here we report that a highly local intramyocardial IL-33/ST2 conversation regulates the heart's response to pressure overload. Either endothelial-specific deletion of IL33 or cardiomyocyte-specific deletion of ST2 exacerbated cardiac hypertrophy with pressure overload. Furthermore, pressure overload induced systemic circulating IL-33 as well as systemic circulating IL-13 and TGF-beta1; this was abolished by endothelial-specific deletion of IL33 but not by cardiomyocyte-specific deletion of IL33. Our study reveals that endothelial cell secretion of IL-33 is crucial for translating myocardial pressure overload into a selective systemic inflammatory response.


Subject(s)
Cardiomegaly/physiopathology , Hypertension/physiopathology , Inflammation/physiopathology , Interleukins/physiology , Myocardium/pathology , Animals , Cardiomegaly/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Hypertension/metabolism , Hypertension/pathology , Inflammation/metabolism , Inflammation/pathology , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Interleukins/metabolism , Mice , Mice, Knockout , Receptors, Interleukin/genetics , Receptors, Interleukin/physiology
4.
Circ Res ; 116(5): 804-15, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25477501

ABSTRACT

RATIONALE: Neonatal mice have the capacity to regenerate their hearts in response to injury, but this potential is lost after the first week of life. The transcriptional changes that underpin mammalian cardiac regeneration have not been fully characterized at the molecular level. OBJECTIVE: The objectives of our study were to determine whether myocytes revert the transcriptional phenotype to a less differentiated state during regeneration and to systematically interrogate the transcriptional data to identify and validate potential regulators of this process. METHODS AND RESULTS: We derived a core transcriptional signature of injury-induced cardiac myocyte (CM) regeneration in mouse by comparing global transcriptional programs in a dynamic model of in vitro and in vivo CM differentiation, in vitro CM explant model, as well as a neonatal heart resection model. The regenerating mouse heart revealed a transcriptional reversion of CM differentiation processes, including reactivation of latent developmental programs similar to those observed during destabilization of a mature CM phenotype in the explant model. We identified potential upstream regulators of the core network, including interleukin 13, which induced CM cell cycle entry and STAT6/STAT3 signaling in vitro. We demonstrate that STAT3/periostin and STAT6 signaling are critical mediators of interleukin 13 signaling in CMs. These downstream signaling molecules are also modulated in the regenerating mouse heart. CONCLUSIONS: Our work reveals new insights into the transcriptional regulation of mammalian cardiac regeneration and provides the founding circuitry for identifying potential regulators for stimulating heart regeneration.


Subject(s)
Myocytes, Cardiac/metabolism , Regeneration/physiology , Transcription, Genetic , Animals , Animals, Newborn , Cell Adhesion Molecules/physiology , Cell Cycle , Cell Dedifferentiation/genetics , Cell Differentiation , Cells, Cultured , Culture Media, Serum-Free , DNA Replication , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Heart Ventricles/cytology , Interleukin-13/pharmacology , Interleukin-13/physiology , Interleukin-13 Receptor alpha1 Subunit/antagonists & inhibitors , Interleukin-13 Receptor alpha1 Subunit/genetics , Interleukin-4 Receptor alpha Subunit/antagonists & inhibitors , Interleukin-4 Receptor alpha Subunit/genetics , Mice , Muscle Development , Myocytes, Cardiac/drug effects , RNA Interference , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/physiology , STAT6 Transcription Factor/physiology , Sequence Alignment , Transcription Factors/physiology , Transcriptome
5.
J Mol Cell Cardiol ; 79: 315-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25533939

ABSTRACT

The finding that neonatal mice are able to regenerate myocardium after apical resection has recently been questioned. We determined if heart regeneration is influenced by the size of cardiac resection and whether surgical retraction of the ventricular apex results in an increase in cardiomyocyte cell cycle activity. We performed moderate or large apical ventricular resections on neonatal mice and quantified scar infiltration into the left ventricular wall at 21 days post-surgery. Moderately resected hearts had 15±2% of the wall infiltrated by a collagen scar; significantly greater scar infiltration (23±4%) was observed in hearts with large resections. Resected hearts had higher levels of cardiomyocyte cell cycle activity relative to sham hearts. Surgically retracting the ventricle often resulted in fibrosis and induced cardiomyocyte cell cycle activity that were comparable to that of resected hearts. We conclude that apical resection in neonatal mice induces cardiomyocyte cell cycle activity and neomyogenesis, although scarring can occur. Surgical technique and definition of approach to assessing the extent of regeneration are both critical when using the neonatal mouse apical resection model.


Subject(s)
Cardiac Surgical Procedures , Heart/physiology , Regeneration/physiology , Animals , Animals, Newborn , Cell Cycle , Fibrosis , Heart Ventricles/surgery , Mice , Myocardium/pathology , Myocytes, Cardiac/pathology
6.
Circulation ; 128(2): 152-61, 2013 Jul 09.
Article in English | MEDLINE | ID: mdl-23757312

ABSTRACT

BACKGROUND: Doxorubicin (DOXO) is an effective anthracycline chemotherapeutic, but its use is limited by cumulative dose-dependent cardiotoxicity. Neuregulin-1ß is an ErbB receptor family ligand that is effective against DOXO-induced cardiomyopathy in experimental models but is also proneoplastic. We previously showed that an engineered bivalent neuregulin-1ß (NN) has reduced proneoplastic potential in comparison with the epidermal growth factor-like domain of neuregulin-1ß (NRG), an effect mediated by receptor biasing toward ErbB3 homotypic interactions uncommonly formed by native neuregulin-1ß. Here, we hypothesized that a newly formulated, covalent NN would be cardioprotective with reduced proneoplastic effects in comparison with NRG. METHODS AND RESULTS: NN was expressed as a maltose-binding protein fusion in Escherichia coli. As established previously, NN stimulated antineoplastic or cytostatic signaling and phenotype in cancer cells, whereas NRG stimulated proneoplastic signaling and phenotype. In neonatal rat cardiomyocytes, NN and NRG induced similar downstream signaling. NN, like NRG, attenuated the double-stranded DNA breaks associated with DOXO exposure in neonatal rat cardiomyocytes and human cardiomyocytes derived from induced pluripotent stem cells. NN treatment significantly attenuated DOXO-induced decrease in fractional shortening as measured by blinded echocardiography in mice in a chronic cardiomyopathy model (57.7±0.6% versus 50.9±2.6%, P=0.004), whereas native NRG had no significant effect (49.4±3.7% versus 50.9±2.6%, P=0.813). CONCLUSIONS: NN is a cardioprotective agent that promotes cardiomyocyte survival and improves cardiac function in DOXO-induced cardiotoxicity. Given the reduced proneoplastic potential of NN versus NRG, NN has translational potential for cardioprotection in patients with cancer receiving anthracyclines.


Subject(s)
Cardiotonic Agents/pharmacology , Chemical Engineering/methods , Doxorubicin/toxicity , Myocytes, Cardiac/drug effects , Neuregulin-1/genetics , Neuregulin-1/pharmacology , Amino Acid Sequence , Animals , Animals, Newborn , Cardiotoxins/antagonists & inhibitors , Cardiotoxins/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Doxorubicin/antagonists & inhibitors , Female , Humans , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology , Random Allocation , Rats , Single-Blind Method
7.
Cell ; 153(4): 828-39, 2013 May 09.
Article in English | MEDLINE | ID: mdl-23663781

ABSTRACT

The most common form of heart failure occurs with normal systolic function and often involves cardiac hypertrophy in the elderly. To clarify the biological mechanisms that drive cardiac hypertrophy in aging, we tested the influence of circulating factors using heterochronic parabiosis, a surgical technique in which joining of animals of different ages leads to a shared circulation. After 4 weeks of exposure to the circulation of young mice, cardiac hypertrophy in old mice dramatically regressed, accompanied by reduced cardiomyocyte size and molecular remodeling. Reversal of age-related hypertrophy was not attributable to hemodynamic or behavioral effects of parabiosis, implicating a blood-borne factor. Using modified aptamer-based proteomics, we identified the TGF-ß superfamily member GDF11 as a circulating factor in young mice that declines with age. Treatment of old mice to restore GDF11 to youthful levels recapitulated the effects of parabiosis and reversed age-related hypertrophy, revealing a therapeutic opportunity for cardiac aging.


Subject(s)
Aging , Bone Morphogenetic Proteins/metabolism , Cardiomegaly/metabolism , Growth Differentiation Factors/metabolism , Myocytes, Cardiac/metabolism , Parabiosis , Animals , Blood Pressure , Female , Forkhead Transcription Factors/metabolism , Humans , Hypertrophy, Left Ventricular/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/cytology
8.
Circ Heart Fail ; 4(4): 509-18, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21606214

ABSTRACT

BACKGROUND: Stromal cell-derived factor-1 (SDF-1) is a chemoattractant of stem/progenitor cells, and several studies have shown that SDF-1 may improve ventricular function after infarction. SDF-1 is cleaved by proteases including matrix metalloproteinase-2 (MMP-2) and CD26/dipeptidylpeptidase-4 (DPP-4), which are activated in injured tissues. METHODS AND RESULTS: We investigated the biodistribution and functional roles of SDF-1 in experimental ischemia/reperfusion injury in rats. Radiolabeled SDF-1 given by intracoronary injection was selectively concentrated in ischemic myocardium. The enhanced uptake of SDF-1 in ischemic myocardium was not mediated by its receptor, CXCR4. Mass spectrometry and Western analyses showed that SDF-1 was cleaved by DPP-4 in plasma and myocardium, whereas a bioengineered MMP-2/DPP-4-resistant form of SDF-1, SSDF-1(S4V), was highly stable. A single dose of SSDF-1(S4V) exhibited greater potency for cardioprotection than wild-type SDF-1. SSDF-1(S4V) improved cardiac function in rats even after a 3-hour ischemic period. CONCLUSIONS: These results show that a single dose of protease-resistant SSDF-1(S4V) after myocardial infarction leads to dramatic improvement in angiogenesis and ventricular function even 3 hours after the onset of ischemia, revealing a simple, clinically feasible approach to prevention of heart failure.


Subject(s)
Chemokine CXCL12/pharmacology , Chemokine CXCL12/therapeutic use , Heart/drug effects , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Animals , Capillaries/drug effects , Chemokine CXCL12/administration & dosage , Disease Models, Animal , Dose-Response Relationship, Drug , Heart/physiology , Heart Failure/prevention & control , Injections, Intra-Arterial , Matrix Metalloproteinase 2/metabolism , Myocardium/metabolism , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Rats , Receptors, CXCR4/metabolism , Treatment Outcome
9.
Cell Stem Cell ; 8(4): 389-98, 2011 Apr 08.
Article in English | MEDLINE | ID: mdl-21474103

ABSTRACT

Cell therapy can improve cardiac function in animals and humans after injury, but the mechanism is unclear. We performed cell therapy experiments in genetically engineered mice that permanently express green fluorescent protein (GFP) only in cardiomyocytes after a pulse of 4-OH-tamoxifen. Myocardial infarction diluted the GFP(+) cardiomyocyte pool, indicating refreshment by non-GFP(+) progenitors. Cell therapy with bone marrow-derived c-kit(+) cells, but not mesenchymal stem cells, further diluted the GFP(+) pool, consistent with c-kit(+) cell-mediated augmentation of cardiomyocyte progenitor activity. This effect could not be explained by transdifferentiation to cardiomyocytes by exogenously delivered c-kit(+) cells or by cell fusion. Therapy with c-kit(+) cells but not mesenchymal stem cells improved cardiac function. These findings suggest that stimulation of endogenous cardiogenic progenitor activity is a critical mechanism of cardiac cell therapy.


Subject(s)
Bone Marrow Transplantation , Myocardial Infarction/therapy , Myocytes, Cardiac/cytology , Stem Cells/cytology , Animals , Cell Transdifferentiation , Green Fluorescent Proteins , Mesenchymal Stem Cells , Mice , Myoblasts, Cardiac , Proto-Oncogene Proteins c-kit
10.
J Mol Cell Cardiol ; 50(5): 841-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21316369

ABSTRACT

Therapies selectively targeting ischemic myocardium could be applied by intravenous injection. Here, we report an approach for ischemic tissue-selective targeting based on in vivo screening of random peptide sequences using phage display. We performed in vivo biopanning using a phage library in a rat model of ischemia-reperfusion and identified three peptide motifs, CSTSMLKAC, CKPGTSSYC, and CPDRSVNNC, that exhibited preferential binding to ischemic heart tissue compared to normal heart as well as other control organs. The CSTSMLKAC sequence was capable of mediating selective homing of phage to ischemic heart tissue. The CSTSMLKAC peptide was then made as a fusion protein with Sumo-mCherry and injected intravenously in a mouse model of myocardial ischemia-reperfusion injury; subsequently, bio-distribution of Sumo-mCherry-CSTSMLKAC was measured with quantitative ELISA. The targeting peptide led to a significant increase in homing to ischemic left ventricle compared to tissues from non-ischemic left ventricle, the right ventricle, lung, liver, spleen, skeletal muscle, and brain (all p<0.001). These results indicate that the peptide sequence CSTSMLKAC represents a novel molecular tool that may be useful in targeting ischemic tissue and delivering bioengineered proteins into the injured myocardium by systemic intravenous administration.


Subject(s)
Myocardial Ischemia/drug therapy , Peptide Library , Peptides/therapeutic use , Amino Acid Sequence , Animals , Male , Mice , Peptides/chemistry , Rats , Rats, Sprague-Dawley
11.
J Mol Cell Cardiol ; 48(6): 1290-7, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19913544

ABSTRACT

Inherited mutations cause approximately 30% of all dilated cardiomyopathy cases, with autosomal dominant mutations in the LMNA gene accounting for more than one third of these. The LMNA gene encodes the nuclear envelope proteins lamins A and C, which provide structural support to the nucleus and also play critical roles in transcriptional regulation. Functional deletion of a single allele is sufficient to trigger dilated cardiomyopathy in humans and mice. However, whereas Lmna(-/-) mice develop severe muscular dystrophy and dilated cardiomyopathy and die by 8 weeks of age, heterozygous Lmna(+/-) mice have a much milder phenotype, with changes in ventricular function and morphology only becoming apparent at 1 year of age. Here, we studied 8- to 20-week-old Lmna(+/-) mice and wild-type littermates in a pressure overload model to examine whether increased mechanical load can accelerate or exacerbate myocardial dysfunction in the heterozygotes. While overall survival was similar between genotypes, Lmna(+/-) animals had a significantly attenuated hypertrophic response to pressure overload as evidenced by reduced ventricular mass and myocyte size. Analysis of pressure overload-induced transcriptional changes suggested that the reduced hypertrophy in the Lmna(+/-) mice was accompanied by impaired activation of the mechanosensitive gene Egr-1. In conclusion, our findings provide further support for a critical role of lamins A and C in regulating the cellular response to mechanical stress in cardiomyocytes and demonstrate that haploinsufficiency of lamins A and C alone is sufficient to alter hypertrophic responses and cardiac function in the face of pressure overload in the heart.


Subject(s)
Cardiomyopathy, Dilated/genetics , Lamin Type A/metabolism , Mutation , Animals , Aorta/pathology , Cardiomyopathies , Disease Models, Animal , Echocardiography/methods , Humans , Lamin Type A/genetics , Mice , Mice, Transgenic , Nuclear Envelope/metabolism , Phenotype , Stress, Mechanical
12.
Circ Heart Fail ; 2(6): 684-91, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19919994

ABSTRACT

BACKGROUND: ST2 is an interleukin (IL)-1 receptor family member with membrane-bound (ST2L) and soluble (sST2) isoforms, and sST2 is a biomarker for poor outcome in patients with myocardial infarction (MI). IL-33, the recently discovered ligand for ST2, activates nuclear factor kappaB and thus may regulate apoptotic cell death. We tested the hypothesis that IL-33 is cardioprotective after MI through ST2 signaling. METHODS AND RESULTS: IL-33 protected cultured cardiomyocytes from hypoxia-induced apoptosis, and this cardioprotection was partially inhibited by sST2. IL-33 induced expression of the antiapoptotic factors XIAP, cIAP1, and survivin. To define the cardioprotective role of IL-33 in vivo, we performed a blinded and randomized study of ischemia/reperfusion in rats. IL-33 reduced cardiomyocyte apoptosis, suppressed caspase-3 activity, and increased expression of IAP family member proteins. IL-33 decreased both infarct and fibrosis volumes at 15 days; furthermore, both echocardiographic and hemodynamic studies revealed that IL-33 improved ventricular function. To determine whether cardioprotection by IL-33 is mediated through ST2 signaling, a randomized and blinded study of ST2(-/-) versus wild-type littermate mice was performed in 98 mice subjected to MI. At 4 weeks after MI, IL-33 reduced ventricular dilation and improved contractile function in wild-type mice but not in ST2(-/-) mice. Finally, IL-33 improved survival after MI in wild-type but not in ST2(-/-) mice. CONCLUSIONS: IL-33 prevents cardiomyocyte apoptosis and improves cardiac function and survival after MI through ST2 signaling.


Subject(s)
Apoptosis/drug effects , Cardiotonic Agents/pharmacology , Interleukins/pharmacology , Myocardial Infarction/drug therapy , Myocardial Reperfusion Injury/drug therapy , Myocytes, Cardiac/drug effects , Receptors, Interleukin-1/metabolism , Receptors, Interleukin/metabolism , Signal Transduction/drug effects , Animals , Animals, Newborn , Apoptosis Regulatory Proteins/metabolism , Cell Survival/drug effects , Cells, Cultured , Disease Models, Animal , Fibrosis , Interleukin-1 Receptor-Like 1 Protein , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction/drug effects , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats , Rats, Sprague-Dawley , Receptors, Interleukin/deficiency , Receptors, Interleukin/genetics , Recombinant Proteins/pharmacology , Time Factors
13.
Circ Res ; 101(12): 1328-38, 2007 Dec 07.
Article in English | MEDLINE | ID: mdl-17916779

ABSTRACT

Biomechanical overload induces cardiac hypertrophy and heart failure, and reactive oxygen species (ROS) play a role in both processes. Thioredoxin-Interacting Protein (Txnip) is encoded by a mechanically-regulated gene that controls cell growth and apoptosis in part through interaction with the endogenous dithiol antioxidant thioredoxin. Here we show that Txnip is a critical regulator of the cardiac response to pressure overload. We generated inducible cardiomyocyte-specific and systemic Txnip-null mice (Txnip-KO) using Flp/frt and Cre/loxP technologies. Compared with littermate controls, Txnip-KO hearts had attenuated cardiac hypertrophy and preserved left ventricular (LV) contractile reserve through 4 weeks of pressure overload; however, the beneficial effects were not sustained and Txnip deletion ultimately led to maladaptive LV remodeling at 8 weeks of pressure overload. Interestingly, these effects of Txnip deletion on cardiac performance were not accompanied by global changes in thioredoxin activity or ROS; instead, Txnip-KO hearts had a robust increase in myocardial glucose uptake. Thus, deletion of Txnip plays an unanticipated role in myocardial energy homeostasis rather than redox regulation. These results support the emerging concept that the function of Txnip is not as a simple thioredoxin inhibitor but as a metabolic control protein.


Subject(s)
Blood Pressure/genetics , Cardiomegaly/genetics , Cardiomegaly/physiopathology , Carrier Proteins/genetics , Gene Deletion , Gene Targeting , Thioredoxins/genetics , Animals , Cardiomegaly/metabolism , Carrier Proteins/physiology , Female , Gene Targeting/methods , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Thioredoxins/physiology , Ventricular Remodeling/physiology
14.
Circulation ; 116(15): 1683-92, 2007 Oct 09.
Article in English | MEDLINE | ID: mdl-17875967

ABSTRACT

BACKGROUND: Local delivery of chemotactic factors represents a novel approach to tissue regeneration. However, successful chemokine protein delivery is challenged by barriers including the rapid diffusion of chemokines and cleavage of chemokines by proteases that are activated in injured tissues. Stromal cell-derived factor-1 (SDF-1) is a well-characterized chemokine for attracting stem cells and thus a strong candidate for promoting regeneration. However, SDF-1 is cleaved by exopeptidases and matrix metalloproteinase-2, generating a neurotoxin implicated in some forms of dementia. METHODS AND RESULTS: We designed a new chemokine called S-SDF-1(S4V) that is resistant to matrix metalloproteinase-2 and exopeptidase cleavage but retains chemotactic bioactivity, reducing the neurotoxic potential of native SDF-1. To deliver S-SDF-1(S4V), we expressed and purified fusion proteins to tether the chemokine to self-assembling peptides, which form nanofibers and allow local delivery. Intramyocardial delivery of S-SDF-1(S4V) after myocardial infarction recruited CXCR4+/c-Kit+ stem cells (46+/-7 to 119+/-18 cells per section) and increased capillary density (from 169+/-42 to 283+/-27 per 1 mm2). Furthermore, in a randomized, blinded study of 176 rats with myocardial infarction, nanofiber delivery of the protease-resistant S-SDF-1(S4V) improved cardiac function (ejection fraction increased from 34.0+/-2.5% to 50.7+/-3.1%), whereas native SDF-1 had no beneficial effects. CONCLUSIONS: The combined advances of a new, protease-resistant SDF-1 and nanofiber-mediated delivery promoted recruitment of stem cells and improved cardiac function after myocardial infarction. These data demonstrate that driving chemotaxis of stem cells by local chemokine delivery is a promising new strategy for tissue regeneration.


Subject(s)
Chemokines, CXC/genetics , Matrix Metalloproteinase 2/metabolism , Myocardial Infarction/physiopathology , Neovascularization, Physiologic , Stem Cells/physiology , Animals , Blood Pressure , Carotid Arteries/physiology , Chemokine CXCL12 , Chemokines, CXC/isolation & purification , Chemokines, CXC/pharmacology , Dipeptidyl Peptidase 4/metabolism , Drug Resistance , Hematopoietic Stem Cell Mobilization , Male , Mutation , Peptide Hydrolases/metabolism , Plasmids , Rats , Rats, Sprague-Dawley , Regeneration
15.
Nat Med ; 13(8): 970-4, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17660827

ABSTRACT

An emerging concept is that the mammalian myocardium has the potential to regenerate, but that regeneration might be too inefficient to repair the extensive myocardial injury that is typical of human disease. However, the degree to which stem cells or precursor cells contribute to the renewal of adult mammalian cardiomyocytes remains controversial. Here we report evidence that stem cells or precursor cells contribute to the replacement of adult mammalian cardiomyocytes after injury but do not contribute significantly to cardiomyocyte renewal during normal aging. We generated double-transgenic mice to track the fate of adult cardiomyocytes in a 'pulse-chase' fashion: after a 4-OH-tamoxifen pulse, green fluorescent protein (GFP) expression was induced only in cardiomyocytes, with 82.7% of cardiomyocytes expressing GFP. During normal aging up to one year, the percentage of GFP+ cardiomyocytes remained unchanged, indicating that stem or precursor cells did not refresh uninjured cardiomyocytes at a significant rate during this period of time. By contrast, after myocardial infarction or pressure overload, the percentage of GFP+ cardiomyocytes decreased from 82.8% in heart tissue from sham-treated mice to 67.5% in areas bordering a myocardial infarction, 76.6% in areas away from a myocardial infarction, and 75.7% in hearts subjected to pressure overload, indicating that stem cells or precursor cells had refreshed the cardiomyocytes.


Subject(s)
Aging/physiology , Cell Lineage , Heart Injuries/genetics , Heart Injuries/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/pathology , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cell Proliferation , Gene Expression Profiling , Mice , Mice, Transgenic , Myocytes, Cardiac/metabolism
16.
Circulation ; 114(7): 637-44, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16894033

ABSTRACT

BACKGROUND: Local delivery methods can target therapies to specific tissues and potentially avoid toxicity to other organs. Platelet-derived growth factor can protect the myocardium, but it also plays an important role in promoting pulmonary hypertension. It is not known whether local myocardial delivery of platelet-derived growth factor during myocardial infarction (MI) can lead to sustained cardiac benefit without causing pulmonary hypertension. METHODS AND RESULTS: We performed a randomized and blinded experiment of 127 rats that survived experimental MI or sham surgery. We delivered platelet-derived growth factor (PDGF)-BB with self-assembling peptide nanofibers (NFs) to provide controlled release within the myocardium. There were 6 groups with n > or = 20 in each group: sham, sham+NF, sham+NF/PDGF, MI, MI+NF, and MI+NF/PDGF. Serial echocardiography from 1 day to 3 months showed significant improvement of ventricular fractional shortening, end-systolic dimension, and end-diastolic dimension with local PDGF delivery (P < 0.05 for MI+NF/PDGF versus MI or MI+NF). Catheterization at 4 months revealed improved ventricular function in the controlled delivery group (left ventricular end-diastolic pressure, cardiac index, +dP/dt, -dP/dt, and time constant of exponential decay all P < 0.05 for MI+NF/P versus MI or MI+NF). Infarcted myocardial volume was reduced by NF/PDGF therapy (34.0 +/- 13.3% in MI, 28.9 +/- 12.9% in MI+NF, and 12.0 +/- 5.8% in MI+NF/PDGF; P < 0.001). There was no evidence of pulmonary toxicity from the therapy, with no differences in right ventricular end-systolic pressure, right ventricular dP/dt, bromodeoxyuridine staining, or pulmonary artery medial wall thickness. CONCLUSIONS: Intramyocardial delivery of PDGF by self-assembling peptide NFs leads to long-term improvement in cardiac performance after experimental infarction without apparent pulmonary toxicity. Local myocardial protection may allow prevention of heart failure without systemic toxicity.


Subject(s)
Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/prevention & control , Myocardial Infarction/physiopathology , Platelet-Derived Growth Factor/administration & dosage , Platelet-Derived Growth Factor/pharmacology , Ventricular Function/drug effects , Animals , Drug Delivery Systems , Echocardiography , Hemodynamics/drug effects , Hemodynamics/physiology , Hypertension, Pulmonary/physiopathology , Injections/methods , Lung/drug effects , Lung/physiopathology , Male , Myocardial Infarction/drug therapy , Nanotechnology , Platelet-Derived Growth Factor/adverse effects , Random Allocation , Rats , Rats, Sprague-Dawley , Regional Blood Flow/drug effects , Ventricular Function/physiology
17.
Am J Physiol Heart Circ Physiol ; 290(1): H232-9, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16126817

ABSTRACT

Matrix metalloproteinases (MMPs) are postulated to be necessary for neovascularization during wound healing. MMP-9 deletion alters remodeling postmyocardial infarction (post-MI), but whether and to what degree MMP-9 affects neovascularization post-MI is unknown. Neovascularization was evaluated in wild-type (WT; n = 63) and MMP-9 null (n = 55) mice at 7-days post-MI. Despite similar infarct sizes, MMP-9 deletion improved left ventricular function as evaluated by hemodynamic analysis. Blood vessel quantity and quality were evaluated by three independent studies. First, vessel density was increased in the infarct of MMP-9 null mice compared with WT, as quantified by Griffonia (Bandeiraea) simplicifolia lectin I (GSL-I) immunohistochemistry. Second, preexisting vessels, stained in vivo with FITC-labeled GSL-I pre-MI, were present in the viable but not MI region. Third, a technetium-99m-labeled peptide (NC100692), which selectively binds to activated alpha(v)beta3-integrin in angiogenic vessels, was injected into post-MI mice. Relative NC100692 activity in myocardial segments with diminished perfusion (0-40% nonischemic) was higher in MMP-9 null than in WT mice (383 +/- 162% vs. 250 +/- 118%, respectively; P = 0.002). The unique finding of this study was that MMP-9 deletion stimulated, rather than impaired, neovascularization in remodeling myocardium. Thus targeted strategies to inhibit MMP-9 early post-MI will likely not impair the angiogenic response.


Subject(s)
Gene Deletion , Matrix Metalloproteinase 9/genetics , Myocardial Infarction/physiopathology , Neovascularization, Physiologic/physiology , Animals , Coronary Vessels/pathology , Ligation , Macrophages/pathology , Matrix Metalloproteinase 9/deficiency , Mice , Mice, Knockout , Myocardial Infarction/pathology , Ventricular Remodeling/physiology
18.
J Clin Invest ; 116(1): 237-48, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16357943

ABSTRACT

Endothelial cells can protect cardiomyocytes from injury, but the mechanism of this protection is incompletely described. Here we demonstrate that protection of cardiomyocytes by endothelial cells occurs through PDGF-BB signaling. PDGF-BB induced cardiomyocyte Akt phosphorylation in a time- and dose-dependent manner and prevented apoptosis via PI3K/Akt signaling. Using injectable self-assembling peptide nanofibers, which bound PDGF-BB in vitro, sustained delivery of PDGF-BB to the myocardium at the injected sites for 14 days was achieved. A blinded and randomized study in 96 rats showed that injecting nanofibers with PDGF-BB, but not nanofibers or PDGF-BB alone, decreased cardiomyocyte death and preserved systolic function after myocardial infarction. A separate blinded and randomized study in 52 rats showed that PDGF-BB delivered with nanofibers decreased infarct size after ischemia/reperfusion. PDGF-BB with nanofibers induced PDGFR-beta and Akt phosphorylation in cardiomyocytes in vivo. These data demonstrate that endothelial cells protect cardiomyocytes via PDGF-BB signaling and that this in vitro finding can be translated into an effective in vivo method of protecting myocardium after infarction. Furthermore, this study shows that injectable nanofibers allow precise and sustained delivery of proteins to the myocardium with potential therapeutic benefits.


Subject(s)
Endothelium, Vascular/physiology , Heart/physiology , Platelet-Derived Growth Factor/pharmacology , Animals , Apoptosis/drug effects , Becaplermin , Drug Delivery Systems , Endothelium, Vascular/drug effects , Heart/drug effects , Heart Conduction System , Muscle Cells/cytology , Muscle Cells/drug effects , Muscle Cells/physiology , Myocardium/cytology , Nerve Fibers/drug effects , Nerve Fibers/physiology , Platelet-Derived Growth Factor/administration & dosage , Platelet-Derived Growth Factor/therapeutic use , Proto-Oncogene Proteins c-sis , Rats
19.
Am J Physiol Heart Circ Physiol ; 288(4): H1802-9, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15563531

ABSTRACT

This study compared the effects of rosuvastatin on left ventricular infarct size in mice after permanent coronary occlusion vs. 60 min of ischemia followed by 24 h of reperfusion. Statins can inhibit neutrophil adhesion, increase nitric oxide synthase (NOS) expression, and mobilize progenitor stem cells after ischemic injury. Mice received blinded and randomized administration of rosuvastatin (20 mg.kg(-1).day(-1)) or saline from 2 days before surgery until death. After 60 min of ischemia with reperfusion, infarct size was reduced by 18% (P = 0.03) in mice randomized to receive rosuvastatin (n = 18) vs. saline (n = 22) but was similar after permanent occlusion in rosuvastatin (n = 17) and saline (n = 20) groups (P = not significant). Myocardial infarct size after permanent left anterior descending coronary artery occlusion (n = 6) tended to be greater in NOS3-deficient mice than in the wild-type saline group (33 +/- 4 vs. 23 +/- 2%, P = 0.08). Infarct size in NOS3-deficient mice was not modified by treatment with rosuvastatin (34 +/- 5%, n = 6, P = not significant vs. NOS3-deficient saline group). After 60 min of ischemia-reperfusion, neutrophil infiltration was similar in rosuvastatin and saline groups as was the percentage of CD34(+), Sca-1(+), and c-Kit(+) cells. Left ventricular NOS3 mRNA and protein levels were unchanged by rosuvastatin. Rosuvastatin reduces infarct size after 60 min of ischemia-reperfusion but not after permanent coronary occlusion, suggesting a potential anti-inflammatory effect. Although we were unable to demonstrate that the myocardial protection was due to an effect on neutrophil infiltration, stem cell mobilization, or induction of NOS3, these data suggest that rosuvastatin may be particularly beneficial in myocardial protection after ischemia-reperfusion injury.


Subject(s)
Fluorobenzenes/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/pathology , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Animals , Coronary Disease/drug therapy , Coronary Disease/pathology , Heart Ventricles/pathology , Hematopoietic Stem Cell Mobilization , Ligation , Male , Mice , Mice, Inbred C57BL , Myocardium/pathology , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , RNA, Messenger/analysis , Rosuvastatin Calcium , Stem Cells/drug effects
20.
Circulation ; 109(21): 2581-6, 2004 Jun 01.
Article in English | MEDLINE | ID: mdl-15123525

ABSTRACT

BACKGROUND: Although cellular redox balance plays an important role in mechanically induced cardiac hypertrophy, the mechanisms of regulation are incompletely defined. Because thioredoxin is a major intracellular antioxidant and can also regulate redox-dependent transcription, we explored the role of thioredoxin activity in mechanically overloaded cardiomyocytes in vitro and in vivo. METHODS AND RESULTS: Overexpression of thioredoxin induced protein synthesis in cardiomyocytes (127+/-5% of controls, P<0.01). Overexpression of thioredoxin-interacting protein (Txnip), an endogenous thioredoxin inhibitor, reduced protein synthesis in response to mechanical strain (89+/-5% reduction, P<0.01), phenylephrine (80+/-3% reduction, P<0.01), or angiotensin II (80+/-4% reduction, P<0.01). In vivo, myocardial thioredoxin activity increased 3.5-fold compared with sham controls after transverse aortic constriction (P<0.01). Aortic constriction did not change thioredoxin expression but reduced Txnip expression by 40% (P<0.05). Gene transfer studies showed that cells that overexpress Txnip develop less hypertrophy after aortic constriction than control cells in the same animals (28.1+/-5.2% reduction versus noninfected cells, P<0.01). CONCLUSIONS: Thus, even though thioredoxin is an antioxidant, activation of thioredoxin participates in the development of pressure-overload cardiac hypertrophy, demonstrating the dual function of thioredoxin as both an antioxidant and a signaling protein. These results also support the emerging concept that the thioredoxin inhibitor Txnip is a critical regulator of biomechanical signaling.


Subject(s)
Cardiomegaly/metabolism , Carrier Proteins/physiology , Heart/drug effects , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Thioredoxins/metabolism , Angiotensin II/pharmacology , Animals , Aortic Diseases/complications , Cardiomegaly/etiology , Cardiomegaly/genetics , Carrier Proteins/genetics , Cell Cycle Proteins , Cell Size , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Cells, Cultured/pathology , Constriction, Pathologic/complications , Disease Models, Animal , Genetic Vectors/genetics , Genetic Vectors/pharmacology , Ligation , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Oxidation-Reduction , Phenylephrine/pharmacology , Random Allocation , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species , Signal Transduction , Single-Blind Method , Stress, Mechanical , Thioredoxins/genetics , Transcriptional Activation/drug effects , Transcriptional Activation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...