Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Sci Transl Med ; 12(535)2020 03 18.
Article in English | MEDLINE | ID: mdl-32188720

ABSTRACT

Danon disease (DD) is a rare X-linked autophagic vacuolar myopathy associated with multiorgan dysfunction, including the heart, skeletal muscle, and liver. There are no specific treatments, and most male patients die from advanced heart failure during the second or third decade of life. DD is caused by mutations in the lysosomal-associated membrane protein 2 (LAMP2) gene, a key mediator of autophagy. LAMP2 has three isoforms: LAMP2A, LAMP2B, and LAMP2C. LAMP2B is the predominant isoform expressed in cardiomyocytes. This study evaluates the efficacy of human LAMP2B gene transfer using a recombinant adeno-associated virus 9 carrying human LAMP2B (AAV9.LAMP2B) in a Lamp2 knockout (KO) mouse, a DD model. AAV9.LAMP2B was intravenously injected into 2- and 6-month-old Lamp2 KO male mice to assess efficacy in adolescent and adult phenotypes. Lamp2 KO mice receiving AAV9.LAMP2B demonstrated dose-dependent restoration of human LAMP2B protein in the heart, liver, and skeletal muscle tissue. Impaired autophagic flux, evidenced by increased LC3-II, was abrogated by LAMP2B gene transfer in all tissues in both cohorts. Cardiac function was also improved, and transaminases were reduced in AAV9.LAMP2B-treated KO mice, indicating favorable effects on the heart and liver. Survival was also higher in the older cohort receiving high vector doses. No anti-LAMP2 antibodies were detected in mice that received AAV9.LAMP2B. In summary, LAMP2B gene transfer improves metabolic and physiologic function in a DD murine model, suggesting that a similar therapeutic approach may be effective for treating patients with this highly morbid disease.


Subject(s)
Glycogen Storage Disease Type IIb , Adolescent , Animals , Disease Models, Animal , Glycogen Storage Disease Type IIb/genetics , Humans , Lysosomal-Associated Membrane Protein 2/genetics , Male , Mice , Mice, Knockout , Phenotype
2.
Pediatr Cardiol ; 40(7): 1401-1409, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31367953

ABSTRACT

Integrin receptors enable cells to sense and respond to their chemical and physical environment. As a class of membrane receptors, they provide a dynamic, tightly regulated link between the extracellular matrix or cellular counter-receptors and intracellular cytoskeletal and signaling networks. They enable transmission of mechanical force across the plasma membrane, and particularly for cardiomyocytes, may sense the mechanical load placed on cells. Talins and Kindlins are two families of FERM-domain proteins which bind the cytoplasmic tail of integrins, recruit cytoskeletal and signaling proteins involved in mechano-transduction, and those which synergize to activate integrins, allowing the integrins to physically change and bind to extracellular ligands. In this review, we will discuss the roles of talin and kindlin, particularly as integrin activators, with a focus on cardiac myocytes.


Subject(s)
Integrins/genetics , Membrane Proteins/physiology , Neoplasm Proteins/physiology , Talin/genetics , Animals , Heart/physiology , Humans , Integrins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Signal Transduction , Talin/metabolism
3.
Nat Biomed Eng ; 3(2): 147-157, 2019 02.
Article in English | MEDLINE | ID: mdl-30923642

ABSTRACT

Dilated cardiomyopathy (DCM) is a leading cause of morbidity and mortality worldwide; yet how genetic variation and environmental factors impact DCM heritability remains unclear. Here, we report that compound genetic interactions between DNA sequence variants contribute to the complex heritability of DCM. By using genetic data from a large family with a history of DCM, we discovered that heterozygous sequence variants in the TROPOMYOSIN 1 (TPM1) and VINCULIN (VCL) genes cose-gregate in individuals affected by DCM. In vitro studies of patient-derived and isogenic human-pluripotent-stem-cell-derived cardio-myocytes that were genome-edited via CRISPR to create an allelic series of TPM1 and VCL variants revealed that cardiomyocytes with both TPM1 and VCL variants display reduced contractility and sarcomeres that are less organized. Analyses of mice genetically engineered to harbour these human TPM1 and VCL variants show that stress on the heart may also influence the variable penetrance and expressivity of DCM-associated genetic variants in vivo. We conclude that compound genetic variants can interact combinatorially to induce DCM, particularly when influenced by other disease-provoking stressors.


Subject(s)
Cardiomyopathy, Dilated/genetics , Genetic Predisposition to Disease , Genetic Variation , Animals , Cardiomyopathy, Dilated/physiopathology , Extracellular Matrix/metabolism , Female , Gene Expression Regulation , Humans , Inheritance Patterns/genetics , Male , Mice , Models, Biological , Muscle Contraction/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Pedigree , Pluripotent Stem Cells/metabolism , Up-Regulation/genetics
5.
Proc Natl Acad Sci U S A ; 114(30): E6250-E6259, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28698364

ABSTRACT

Continuous contraction-relaxation cycles of the heart require strong and stable connections of cardiac myocytes (CMs) with the extracellular matrix (ECM) to preserve sarcolemmal integrity. CM attachment to the ECM is mediated by integrin complexes localized at the muscle adhesion sites termed costameres. The ubiquitously expressed cytoskeletal protein talin (Tln) is a component of muscle costameres that links integrins ultimately to the sarcomere. There are two talin genes, Tln1 and Tln2. Here, we tested the function of these two Tln forms in myocardium where Tln2 is the dominant isoform in postnatal CMs. Surprisingly, global deletion of Tln2 in mice caused no structural or functional changes in heart, presumably because CM Tln1 became up-regulated. Tln2 loss increased integrin activation, although levels of the muscle-specific ß1D-integrin isoform were reduced by 50%. With this result, we produced mice that had simultaneous loss of both CM Tln1 and Tln2 and found that cardiac dysfunction occurred by 4 wk with 100% mortality by 6 mo. ß1D integrin and other costameric proteins were lost from the CMs, and membrane integrity was compromised. Given that integrin protein reduction occurred with Tln loss, rescue of the phenotype was attempted through transgenic integrin overexpression, but this could not restore WT CM integrin levels nor improve heart function. Our results show that CM Tln2 is essential for proper ß1D-integrin expression and that Tln1 can substitute for Tln2 in preserving heart function, but that loss of all Tln forms from the heart-muscle cell leads to myocyte instability and a dilated cardiomyopathy.


Subject(s)
Cardiomyopathy, Dilated/genetics , Integrin beta1/metabolism , Myocytes, Cardiac/metabolism , Talin/genetics , Animals , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Mice , Myocardium/pathology , Myocytes, Cardiac/physiology , Talin/metabolism , Talin/physiology
6.
PLoS One ; 12(5): e0177660, 2017.
Article in English | MEDLINE | ID: mdl-28498861

ABSTRACT

Anthracyclines are chemotherapeutic drugs known to induce heart failure in a dose-dependent manner. Mechanisms involved in anthracycline cardiotoxicity are an area of relevant investigation. Caveolins bind, organize and regulate receptors and signaling molecules within cell membranes. Caveolin-3 (Cav-3), integrins and related membrane repair proteins can function as cardioprotective proteins. Expression of these proteins in anthracycline-induced heart failure has not been evaluated. We tested the hypothesis that daunorubicin alters cardioprotective protein expression in the heart. Rabbits were administered daunorubicin (3 mg/kg, IV) weekly, for three weeks or nine weeks. Nine weeks but not three weeks of daunorubicin resulted in progressive reduced left ventricular function. Cav-3 expression in the heart was unchanged at three weeks of daunorubicin and increased in nine week treated rabbits when compared to control hearts. Electron microscopy showed caveolae in the heart were increased and mitochondrial number and size were decreased after nine weeks of daunorubicin. Activated beta-1 (ß1) integrin and the membrane repair protein MG53 were increased after nine weeks of daunorubicin vs. controls with no change at the three week time point. The results suggest a potential pathophysiological role for Cav3, integrins and membrane repair in daunorubicin-induced heart failure.


Subject(s)
Anthracyclines/toxicity , Caveolins/metabolism , Daunorubicin/toxicity , Heart Failure/metabolism , Integrins/metabolism , Animals , Blotting, Western , Cardiotoxicity/blood , Cardiotoxicity/metabolism , Cholesterol/blood , Cholesterol/metabolism , Echocardiography , Heart Failure/chemically induced , Immunohistochemistry , Microscopy, Electron , Myocardium/metabolism , Myocardium/pathology , Rabbits
7.
Circ Heart Fail ; 9(8)2016 08.
Article in English | MEDLINE | ID: mdl-27502369

ABSTRACT

BACKGROUND: The striated muscle costamere, a multiprotein complex at the boundary between the sarcomere and the sarcolemma, plays an integral role in maintaining striated muscle structure and function. Multiple costamere-associated proteins, such as integrins and integrin-interacting proteins, have been identified and shown to play an increasingly important role in the pathogenesis of human cardiomyopathy. Kindlin-2 is an adaptor protein that binds to the integrin ß cytoplasmic tail to promote integrin activation. Genetic deficiency of Kindlin-2 results in embryonic lethality, and knockdown of the Kindlin-2 homolog in Caenorhabditis elegans and Danio rerio suggests that it has an essential role in integrin function and normal muscle structure and function. The precise role of Kindlin-2 in the mammalian cardiac myocyte remains to be determined. METHODS AND RESULTS: The current studies were designed to investigate the role of Kindlin-2 in the mammalian heart. We generated a series of cardiac myocyte-specific Kindlin-2 knockout mice with excision of the Kindlin-2 gene in either developing or adult cardiac myocytes. We found that mice lacking Kindlin-2 in the early developing heart are embryonic lethal. We demonstrate that deletion of Kindlin-2 at late gestation or in adult cardiac myocytes resulted in heart failure and premature death, which were associated with enlargement of the heart and extensive fibrosis. In addition, integrin ß1D protein expression was significantly downregulated in the adult heart. CONCLUSIONS: Kindlin-2 is required to maintain integrin ß1D protein stability. Postnatal loss of Kindlin-2 from cardiac myocytes leads to progressive heart failure, showing the importance of costameric proteins like Kindlin-2 for homeostasis of normal heart function.


Subject(s)
Cytoskeletal Proteins/deficiency , Heart Failure/metabolism , Muscle Proteins/deficiency , Myocytes, Cardiac/metabolism , Age Factors , Animals , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cytoskeletal Proteins/genetics , Disease Progression , Down-Regulation , Fibrosis , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , Gestational Age , Heart Failure/genetics , Heart Failure/pathology , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/pathology , Integrin beta1/genetics , Integrin beta1/metabolism , Mice, Knockout , Muscle Proteins/genetics , Myocytes, Cardiac/pathology , Phenotype
8.
J Mol Cell Cardiol ; 93: 162-74, 2016 04.
Article in English | MEDLINE | ID: mdl-26562414

ABSTRACT

Cardiac fibrosis is one of the major components of the healing mechanism following any injury of the heart and as such may contribute to both systolic and diastolic dysfunction in a range of pathophysiologic conditions. Canonically, it can occur as part of the remodeling process that occurs following myocardial infarction or that follows as a response to pressure overload. Integrins are cell surface receptors which act in both cellular adhesion and signaling. Most importantly, in the context of the continuously contracting myocardium, they are recognized as mechanotransducers. They have been implicated in the development of fibrosis in several organs, including the heart. This review will focus on the involvement of integrins and integrin-related proteins, in cardiac fibrosis, outlining the roles of these proteins in the fibrotic responses in specific cardiac pathologies, discuss some of the common end effectors (angiotensin II, transforming growth factor beta 1 and mechanical stress) through which integrins function and finally discuss how manipulation of this set of proteins may lead to new treatments which could prove useful to alter the deleterious effects of cardiac fibrosis.


Subject(s)
Carrier Proteins/metabolism , Integrins/metabolism , Myocardium/metabolism , Myocardium/pathology , Age Factors , Aging , Angiotensin II/metabolism , Animals , Blood Pressure , Cytokines/metabolism , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/pathology , Epithelial-Mesenchymal Transition , Fibrosis , Humans , Molecular Targeted Therapy , Myocardial Infarction/etiology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Protein Binding , Stress, Mechanical
9.
Circ Res ; 114(3): 572-586, 2014 Jan 31.
Article in English | MEDLINE | ID: mdl-24481847

ABSTRACT

Integrins are heterodimeric, transmembrane receptors that are expressed in all cells, including those in the heart. They participate in multiple critical cellular processes including adhesion, extracellular matrix organization, signaling, survival, and proliferation. Particularly relevant for a contracting muscle cell, integrins are mechanotransducers, translating mechanical to biochemical information. Although it is likely that cardiovascular clinicians and scientists have the highest recognition of integrins in the cardiovascular system from drugs used to inhibit platelet aggregation, the focus of this article will be on the role of integrins specifically in the cardiac myocyte. After a general introduction to integrin biology, the article will discuss important work on integrin signaling, mechanotransduction, and lessons learned about integrin function from a range of model organisms. Then we will detail work on integrin-related proteins in the myocyte, how integrins may interact with ion channels and mediate viral uptake into cells, and also play a role in stem cell biology. Finally, we will discuss directions for future study.


Subject(s)
Integrins/physiology , Myocytes, Cardiac/chemistry , Signal Transduction/physiology , Animals , CD47 Antigen/chemistry , CD47 Antigen/metabolism , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/metabolism , Humans , Muscle Proteins/chemistry , Muscle Proteins/metabolism , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology
10.
J Clin Invest ; 123(10): 4294-308, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24091324

ABSTRACT

Ischemic damage is recognized to cause cardiomyocyte (CM) death and myocardial dysfunction, but the role of cell-matrix interactions and integrins in this process has not been extensively studied. Expression of α7ß1D integrin, the dominant integrin in normal adult CMs, increases during ischemia/reperfusion (I/R), while deficiency of ß1 integrins increases ischemic damage. We hypothesized that the forced overexpression of integrins on the CM would offer protection from I/R injury. Tg mice with CM-specific overexpression of integrin α7ß1D exposed to I/R had a substantial reduction in infarct size compared with that of α5ß1D-overexpressing mice and WT littermate controls. Using isolated CMs, we found that α7ß1D preserved mitochondrial membrane potential during hypoxia/reoxygenation (H/R) injury via inhibition of mitochondrial Ca2+ overload but did not alter H/R effects on oxidative stress. Therefore, we assessed Ca2+ handling proteins in the CM and found that ß1D integrin colocalized with ryanodine receptor 2 (RyR2) in CM T-tubules, complexed with RyR2 in human and rat heart, and specifically bound to RyR2 amino acids 165-175. Integrins stabilized the RyR2 interdomain interaction, and this stabilization required integrin receptor binding to its ECM ligand. These data suggest that α7ß1D integrin modifies Ca2+ regulatory pathways and offers a means to protect the myocardium from ischemic injury.


Subject(s)
Integrins/metabolism , Myocardial Ischemia/metabolism , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Amino Acid Sequence , Animals , Calcium/metabolism , Cell Hypoxia , Cells, Cultured , Humans , Integrins/chemistry , Male , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Myocardial Ischemia/pathology , Myocardial Reperfusion Injury/pathology , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Processing, Post-Translational , Protein Stability , Protein Subunits/metabolism , Rats , Rats, Sprague-Dawley , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/metabolism
11.
J Biol Chem ; 288(6): 4252-64, 2013 Feb 08.
Article in English | MEDLINE | ID: mdl-23266827

ABSTRACT

Integrins are adhesive, signaling, and mechanotransduction proteins. Talin (Tln) activates integrins and links it to the actin cytoskeleton. Vertebrates contain two talin genes, tln1 and tln2. How Tln1 and Tln2 function in cardiac myocytes (CMs) is unknown. Tln1 and Tln2 expression were evaluated in the normal embryonic and adult mouse heart as well as in control and failing human adult myocardium. Tln1 function was then tested in the basal and mechanically stressed myocardium after cardiomyocyte-specific excision of the Tln1 gene. During embryogenesis, both Tln forms are highly expressed in CMs, but in the mature heart Tln2 becomes the main Tln isoform, localizing to the costameres. Tln1 expression is minimal in the adult CM. With pharmacological and mechanical stress causing hypertrophy, Tln1 is up-regulated in CMs and is specifically detected at costameres, suggesting its importance in the compensatory response to CM stress. In human failing heart, CM Tln1 also increases compared with control samples from normal functioning myocardium. To directly test Tln1 function in CMs, we generated CM-specific Tln1 knock-out mice (Tln1cKO). Tln1cKO mice showed normal basal cardiac structure and function but when subjected to pressure overload showed blunted hypertrophy, less fibrosis, and improved cardiac function versus controls. Acute responses of ERK1/2, p38, Akt, and glycogen synthase kinase 3 after mechanical stress were strongly blunted in Tln1cKO mice. Given these results, we conclude that Tln1 and Tln2 have distinct functions in the myocardium. Our data show that reduction of CM Tln1 expression can lead to improved cardiac remodeling following pressure overload.


Subject(s)
Cardiomegaly/metabolism , Myocardium/metabolism , Talin/biosynthesis , Adult , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Female , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Heart Failure/genetics , Heart Failure/metabolism , Heart Failure/pathology , Humans , MAP Kinase Signaling System/genetics , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Stress, Physiological/genetics , Talin/genetics , Up-Regulation/genetics , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Eur J Cell Biol ; 91(3): 180-91, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22306379

ABSTRACT

Talins are adaptor proteins that connect the integrin family of cell adhesion receptors to cytoskeletal actin. Vertebrates express two closely related talins encoded by separate genes, and while it is well established that talin1 plays a key role in cell adhesion and spreading, little is known about the role of talin2. To facilitate such studies, we report the characterisation of 4 new isoform-specific talin mouse monoclonal antibodies that work in Western blotting, immuno-precipitation, immuno-fluorescence and immuno-histochemistry. Using these antibodies, we show that talin1 and talin2 do not form heterodimers, and that they are differentially localised within the cell. Talin1 was concentrated in peripheral focal adhesions while talin2 was observed in both focal and fibrillar adhesions, and knock-down of talin2 compromised fibronectin fibrillogenesis. Although differentiated human macrophages express both isoforms, only talin1 showed discrete staining and was localised to the ring structure of podosomes. However, siRNA-mediated knock-down of macrophage talin2 led to a significant reduction in podosomal matrix degradation. We have also used the antibodies to localise each isoform in tissue sections using both cryostat and paraffin-embedded material. In skeletal muscle talin2 was localised to both myotendinous junctions and costameres while talin1 was restricted to the former structure. In contrast, both isoforms co-localised in kidney with staining of the glomerulus, and the tubular epithelial and interstitial cells of the cortex and medulla. We anticipate that these antibodies will form a valuable resource for future studies on the function of the two major talin isoforms.


Subject(s)
Antibodies, Monoclonal , Fibronectins/metabolism , Macrophages/ultrastructure , Protein Isoforms/analysis , Talin/metabolism , Animals , Antibody Specificity , Focal Adhesions/metabolism , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred BALB C , NIH 3T3 Cells , Protein Isoforms/metabolism , RNA, Small Interfering , Rats
13.
Am J Pathol ; 180(3): 952-962, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22248583

ABSTRACT

How mechanical signals are transmitted in the cardiac myocyte is poorly understood. In this study, we produced a tamoxifen-inducible mouse model in which ß1 integrin could be reduced specifically in the adult cardiomyocyte, so that the function of this integrin could be assessed in the postnatal and mechanically stressed heart. The expression of ß1 integrin was reduced to 35% of control levels, but function remained normal at baseline. With aortic constriction, the knockout mice survived but had a blunted hypertrophic response. Integrin knockout myocytes, in contrast to controls, showed reduced integrin-linked kinase expression both at baseline and after hemodynamic stress; focal adhesion kinase expression was reduced after stress. Alterations in multiple signaling pathways were detected in the integrin knockout group after acute and chronic hemodynamic stress. Most remarkably, when we challenged the knockout mice with short-term loading, the robust responses of several kinases (extracellular signal-regulated kinase 1/2, p38, and Akt) evident in control mice were essentially abolished in the knockout mice. We also found that reduction of myocyte ß1 integrin expression modified adrenergic-mediated signaling through extracellular signal-regulated kinase, p38, and Akt. Reduction of ß1 integrin expression in the mature cardiac myocyte leads to a varied response compared with when this protein is reduced during either the embryonic or perinatal period. These results show that ß1 integrin expression is required for proper mechanotransductive and adrenergic responses of the adult heart.


Subject(s)
Cardiomegaly/etiology , Integrin beta1/physiology , Myocytes, Cardiac/physiology , Signal Transduction/physiology , Animals , Aorta , Cardiomegaly/metabolism , Cell Death , Constriction , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Gene Deletion , Hemodynamics/physiology , Integrin beta1/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Myocytes, Cardiac/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Stress, Mechanical
14.
J Investig Med ; 57(8): 849-55, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19952892

ABSTRACT

Cardiomyopathy is a heart muscle disease caused by decreased contractility of the ventricles leading to heart failure and premature death. Multiple conditions like ischemic heart disease (atherosclerosis), hypertension, diabetes, viral infection, alcohol abuse, obesity and genetic mutations can lead to cardiomyopathy. Single gene mutations in sarcomeric proteins, Z-disk-associated proteins, membrane/associated proteins, intermediate filaments, calcium cycle proteins as well as in modifier genes have been linked to cardiomyopathy. Clinical practice guidelines have been formulated by the American Heart Association and the Heart Failure Association of America on how to genetically evaluate patients with cardiomyopathy. To illustrate the concept that alterations in genes cause cardiovascular disease, this review will focus on two membrane-associated proteins, vinculin and talin. We will discuss the general function of vinculin/metavinulin as well as talin1 and talin2, with emphasis on what is understood about their role in the cardiac myocyte and in whole heart.


Subject(s)
Myocardium/metabolism , Talin/physiology , Vinculin/physiology , Animals , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Humans , Myocardium/chemistry , Myocardium/pathology , Myocytes, Cardiac/chemistry , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Talin/chemistry , Talin/genetics , Vinculin/chemistry , Vinculin/genetics
15.
J Investig Med ; 57(8): 856-60, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19952893

ABSTRACT

How the myocardium undergoes geometric, structural, and molecular alterations that result in an end phenotype as might be seen in patients with dilated cardiomyopathy or after myocardial infarction is still poorly understood. Structural modification of the left ventricle, which occurs during these pathological states, results from long-term changes in loading conditions and is commonly referred to as "remodeling." Remodeling may occur from increased wall stress in the face of hypertensive heart disease, valvular disease, or, perhaps most dramatically, after permanent coronary occlusion. A fundamental derangement of myocyte function is the most common perception for the basis of remodeling, but the role of cells in the heart other than the muscle cell must, of course, be considered. Although studies of the myocyte have been extensive, cardiac fibroblasts have been studied less than myocytes. The fibroblast has a broad range of functions in the myocardium ranging from elaboration and remodeling of the extracellular matrix to communication of a range of signals within the heart, including electrical, chemical, and mechanical ones. Integrins are cell surface receptors that are instrumental in mediating cell-matrix interactions in all cells of the organism, including all types within the myocardium. This review will focus on the role of integrins and related proteins in the remodeling process, with a particular emphasis on the cardiac fibroblast. We will illustrate this function by drawing on 2 unique mouse models with perturbation of proteins linked to integrin function.


Subject(s)
Fibroblasts/physiology , Focal Adhesions/metabolism , Integrins/physiology , Myocytes, Cardiac/physiology , Animals , Fibroblasts/pathology , Focal Adhesions/genetics , Focal Adhesions/pathology , Humans , Integrins/genetics , Myocytes, Cardiac/pathology
16.
Am J Physiol Heart Circ Physiol ; 296(3): H627-38, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19136609

ABSTRACT

Migration and proliferation of cardiac fibroblasts (CFs) play an important role in the myocardial remodeling process. While many factors have been identified that regulate CF growth and migration, less is known about the signaling mechanisms involved in these processes. Here, we utilized Cre-LoxP technology to obtain focal adhesion kinase (FAK)-deficient adult mouse CFs and studied how FAK functioned in modulating cell adhesion, proliferation, and migration of these cells. Treatment of FAK(flox/flox) CFs with Ad/Cre virus caused over 70% reduction of FAK protein levels within a cell population. FAK-deficient CFs showed no changes in focal adhesions, cell morphology, or protein expression levels of vinculin, talin, or paxillin; proline-rich tyrosine kinase 2 (Pyk2) expression and activity were increased. Knockdown of FAK protein in CFs increased PDGF-BB-induced proliferation, while it reduced PDGF-BB-induced migration. Adhesion to fibronectin was not altered. To distinguish between the function of FAK and Pyk2, FAK function was inhibited via adenoviral-mediated overexpression of the natural FAK inhibitor FAK-related nonkinase (FRNK). Ad/FRNK had no effect on Pyk2 expression, inhibited the PDGF-BB-induced migration, but did not change the PDGF-BB-induced proliferation. FAK deficiency had only modest effects on increasing PDGF-BB activation of p38 and JNK MAPKs, with no alteration in the ERK response vs. control cells. These results demonstrate that FAK is required for the PDGF-BB-induced migratory response of adult mouse CFs and suggest that FAK could play an essential role in the wound-healing response that occurs in numerous cardiac pathologies.


Subject(s)
Cell Movement , Cell Proliferation , Fibroblasts/enzymology , Focal Adhesion Kinase 1/metabolism , Myocardium/enzymology , Animals , Becaplermin , Cell Adhesion , Cell Shape , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Kinase 1/deficiency , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 2/metabolism , Focal Adhesions/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/cytology , Paxillin/metabolism , Platelet-Derived Growth Factor/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-sis , Talin/metabolism , Time Factors , Vinculin/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Mol Cell Biol ; 27(21): 7522-37, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17785437

ABSTRACT

Vinculin is a ubiquitously expressed multiliganded protein that links the actin cytoskeleton to the cell membrane. In myocytes, it is localized in protein complexes which anchor the contractile apparatus to the sarcolemma. Its function in the myocardium remains poorly understood. Therefore, we developed a mouse model with cardiac-myocyte-specific inactivation of the vinculin (Vcl) gene by using Cre-loxP technology. Sudden death was found in 49% of the knockout (cVclKO) mice younger than 3 months of age despite preservation of contractile function. Conscious telemetry documented ventricular tachycardia as the cause of sudden death, while defective myocardial conduction was detected by optical mapping. cVclKO mice that survived through the vulnerable period of sudden death developed dilated cardiomyopathy and died before 6 months of age. Prior to the onset of cardiac dysfunction, ultrastructural analysis of cVclKO heart tissue showed abnormal adherens junctions with dissolution of the intercalated disc structure, expression of the junctional proteins cadherin and beta1D integrin were reduced, and the gap junction protein connexin 43 was mislocalized to the lateral myocyte border. This is the first report of tissue-specific inactivation of the Vcl gene and shows that it is required for preservation of normal cell-cell and cell-matrix adhesive structures.


Subject(s)
Adherens Junctions/pathology , Cardiomyopathy, Dilated/pathology , Death, Sudden/pathology , Myocytes, Cardiac/metabolism , Vinculin/deficiency , Vinculin/genetics , Alleles , Animals , Cadherins/metabolism , Cardiomyopathy, Dilated/physiopathology , Connexin 43/metabolism , Heart Conduction System/abnormalities , Heart Conduction System/physiopathology , Hypertrophy , Integrins/metabolism , Male , Mice , Mice, Knockout , Mortality , Myocardium/pathology , Myocardium/ultrastructure , Myocytes, Cardiac/pathology , Organ Specificity , Systole , Tachycardia, Ventricular/pathology
18.
Cardiovasc Res ; 69(3): 574-84, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16253214

ABSTRACT

The concept of myocardial remodeling links an initial pathological insult to a progressive geometric change of the ventricle. Currently, our concepts of the remodeling process have evolved to include not only changes in ventricular size and shape, but cellular and molecular remodeling, particularly as the ventricle evolves towards failure. In recent years, much attention has focused on the role of cell-extracellular matrix (ECM) connections in this process. In this review, we will specifically delineate how cell membrane-linked molecules of three classes: integrins, membrane-type matrix metalloproteinases, and ADAMs (A Disintegrin And Metalloproteinase) might play crucial roles in myocardial remodeling. These molecules are essential for cell-ECM adhesion, cell signaling, matrix modification, and proteolysis of surface receptors. Our goal is to put forth concepts on how they might interrelate to modulate the remodeling process in the heart.


Subject(s)
Extracellular Matrix/enzymology , Heart Failure/enzymology , Integrins/physiology , Matrix Metalloproteinases/metabolism , Myocardium/enzymology , Animals , Disintegrins/metabolism , Extracellular Matrix/pathology , Heart Failure/pathology , Humans , Matrix Metalloproteinases, Membrane-Associated , Myocardium/pathology , Ventricular Remodeling
19.
Am J Physiol Heart Circ Physiol ; 288(1): H227-34, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15345478

ABSTRACT

Long-standing diabetes can result in the development of cardiomyopathy, which can be accompanied by myocardial fibrosis. Although exposure of cultured kidney and skin fibroblasts to high glucose (HG) concentration is known to increase collagen synthesis, little is known about cardiac fibroblasts (CFs). Therefore, we determined the influence of HG conditions on CF functions and the effects of losartan and vitamin E in these responses. We cultured rat CFs in either normal glucose (NG; 5.5 mM) or HG (25 mM) media and assessed changes in protein and collagen synthesis, matrix metalloproteinase (MMP) activity, and levels of mRNA for ANG II type 1 (AT(1)) receptors. Results indicate that HG-level CFs synthesized more protein and collagen, and these effects were not due to changes in osmotic pressure. The addition of ANG II stimulated protein and collagen synthesis in NG-concentration but not HG-concentration CFs. Interestingly, losartan pretreatment blocked the HG- or ANG II-induced increases in both protein and collagen synthesis. HG or ANG II decreased total MMP activity. Decreases in MMP activity were blocked by losartan. AT(1) mRNA levels were upregulated with HG concentration. Vitamin E pretreatment blocked the effects of HG on total protein synthesis and stimulated MMP activity. Results suggest that HG levels may promote fibrosis by increasing CF protein and collagen synthesis and decreasing MMP activity. HG levels may cause these effects via the upregulation of AT(1) receptors, which can be blocked by losartan. However, vitamin E can alter HG concentration-induced changes in CF functions independently of AT(1) mRNA levels.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Antioxidants/pharmacology , Fibroblasts/physiology , Glucose/administration & dosage , Losartan/pharmacology , Myocardium/cytology , Vitamin E/pharmacology , Angiotensin II/pharmacology , Animals , Cells, Cultured , Collagen Type I/metabolism , Dose-Response Relationship, Drug , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibrosis , Glucose/pharmacology , Leucine/metabolism , Male , Matrix Metalloproteinases/metabolism , Osmolar Concentration , Proline/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...