Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Heart Fail Rev ; 23(5): 759-772, 2018 09.
Article in English | MEDLINE | ID: mdl-29737434

ABSTRACT

Anti-apoptotic therapy for cardiomyocytes could be an effective strategy for preventing or treating heart failure. Notably, however, morphological evidence definitively demonstrating cardiomyocyte apoptosis has been very rare in actual heart diseases such as acute myocardial infarction and heart failure. By contrast, within the postinfarction heart, interstitial noncardiomyocytes such as granulation tissue cells do die via apoptosis to form scar tissue. Blockade of this apoptosis improves survival and mitigates ventricular remodeling and dysfunction during the chronic stage. Possible mechanisms to explain this benefit might be preservation of infarcted wall thickness and preservation of myofibroblasts, which could promote infarct shrinkage; both would reduce wall stress through Laplace's law. On the other hand, autophagy is an intracellular degradation mechanism that compensates for energy insufficiency by digesting and recycling intracellular components, and is often observed in cardiomyocytes within failing hearts with various origins including postinfarction. Starvation strongly induces and activates autophagic degeneration within cardiomyocytes. When that activation is inhibited, the starved animals suffer from heart failure. Promoting autophagy through caloric restriction or several reagents not only reduces the acute infarct size but also mitigates postinfarction cardiac remodeling and dysfunction during chronic stages. Moreover, augmenting autophagy by the treatment with resveratrol or exercise can bring about reverse remodeling in failing hearts with a large old myocardial infarction. In conclusion, we propose two strategies for managing postinfarction heart failure through control of cell death/degeneration: (1) anti-apoptosis in granulation tissue noncardiomyocytes; and (2) pro-autophagy in salvaged cardiomyocytes.


Subject(s)
Heart Failure/prevention & control , Myocardial Infarction/complications , Myocardium/pathology , Myocytes, Cardiac/pathology , Animals , Apoptosis , Autophagy , Disease Progression , Heart Failure/metabolism , Heart Failure/pathology , Humans , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/metabolism , Myocytes, Cardiac/metabolism
2.
Cardiovasc Res ; 96(3): 456-65, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22952253

ABSTRACT

AIMS: Active autophagy has recently been reported in doxorubicin-induced cardiotoxicity; here we investigated its pathophysiological role. METHODS AND RESULTS: Acute cardiotoxicity was induced in green fluorescent protein-microtubule-associated protein 1 light chain 3 (GFP-LC3) transgenic mice by administering two intraperitoneal injections of 10 mg/kg doxorubicin with a 3 day interval. A starvation group was deprived of food for 48 h before each injection to induce autophagy in advance. Doxorubicin treatment caused left ventricular dilatation and dysfunction within 6 days. Cardiomyocyte autophagy appeared to be activated in the doxorubicin group, based on LC3, p62, and cathepsin D expression, while it seemed somewhat diminished by starvation prior to doxorubicin treatment. Unexpectedly, however, myocardial ATP levels were reduced in the doxorubicin group, and this reduction was prevented by earlier starvation. Electron microscopy revealed that the autophagic process was indeed initiated in the doxorubicin group, as shown by the increased lysosomes, but was not completed, i.e. autophagolysosome formation was rare. Starvation prior to doxorubicin treatment partly restored autophagosome formation towards control levels. Autophagic flux assays in both in vivo and in vitro models confirmed that doxorubicin impairs completion of the autophagic process in cardiomyocytes. The activities of both AMP-activated protein kinase and the autophagy-initiating kinase unc-51-like kinase 1 (ULK1) were found to be decreased by doxorubicin, and these were restored by prior starvation. CONCLUSION: Prior starvation mitigates acute doxorubicin cardiotoxicity; the underlying mechanism may be, at least in part, restoration and further augmentation of myocardial autophagy, which is impaired by doxorubicin, probably through inactivation of AMP-activated protein kinase and ULK1.


Subject(s)
Antibiotics, Antineoplastic , Autophagy/drug effects , Doxorubicin , Heart Failure/prevention & control , Hypertrophy, Left Ventricular/prevention & control , Myocytes, Cardiac/pathology , Starvation/complications , Ventricular Dysfunction, Left/prevention & control , AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , Animals , Autophagy-Related Protein-1 Homolog , Cathepsin D/metabolism , Cells, Cultured , Energy Metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Heart Failure/chemically induced , Heart Failure/metabolism , Heart Failure/pathology , Heart Failure/physiopathology , Hypertrophy, Left Ventricular/chemically induced , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Mice , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Myocytes, Cardiac/metabolism , Protein Serine-Threonine Kinases/metabolism , Rats , Starvation/metabolism , Stroke Volume , Time Factors , Ventricular Dysfunction, Left/chemically induced , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology , Ventricular Function, Left , Ventricular Pressure
3.
Circ Heart Fail ; 5(2): 274-85, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22343033

ABSTRACT

BACKGROUND: We investigated the effects of asialoerythropoietin (asialoEPO), a nonerythrogenic erythropoietin derivative, on 3 murine models of heart failure with different etiologies. METHODS AND RESULTS: Doxorubicin (15 mg/kg) induced heart failure within 2 weeks (toxic cardiomyopathy). Treatment with asialoEPO (6.9 µg/kg) for 2 weeks thereafter attenuated the associated left ventricular dysfunction and dilatation. In addition, the asialoEPO-treated heart showed less myocardial fibrosis, inflammation, and oxidative damage, and diminished atrophic cardiomyocyte degeneration, which was accompanied by restored expression of GATA-4 and sarcomeric proteins. Mice with large 6-week-old myocardial infarctions exhibited marked left ventricular dysfunction with adverse remodeling (ischemic cardiomyopathy). AsialoEPO treatment for 4 weeks significantly mitigated progression of the dysfunction and remodeling and reduced myocardial fibrosis, inflammation, and oxidative damage. Finally, 25-week-old δ-sarcoglycan-deficient mice (genetic cardiomyopathy) were treated with asialoEPO for 5 weeks. AsialoEPO mitigated the progressive cardiac remodeling and dysfunction through cardiomyocyte hypertrophy, and upregulated expression of GATA-4 and sarcomeric proteins. AsialoEPO appears to act by altering the activity of the downstream erythropoietin receptor signals extracellular signal-regulated protein kinase, Akt, signal transducer, and activator of transcription 3 and 5 in a model-specific manner. CONCLUSIONS: The findings suggest that asialoEPO exerts broad cardioprotective effects through distinct mechanisms depending on the model, which are independent of the erythrogenic action. This compound may be promising for the treatment of heart failure of various etiologies.


Subject(s)
Asialoglycoproteins/therapeutic use , Erythropoietin/analogs & derivatives , Heart Failure/drug therapy , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects , Animals , Asialoglycoproteins/administration & dosage , Disease Models, Animal , Dose-Response Relationship, Drug , Erythropoietin/administration & dosage , Erythropoietin/therapeutic use , Follow-Up Studies , Heart Failure/physiopathology , Mice , Mice, Inbred C57BL , Treatment Outcome , Ventricular Function, Left/physiology
4.
Cardiovasc Res ; 91(2): 330-9, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21406597

ABSTRACT

AIMS: Autophagy is activated in cardiomyocytes in ischaemic heart disease, but its dynamics and functional roles remain unclear after myocardial infarction. We observed the dynamics of cardiomyocyte autophagy and examined its role during postinfarction cardiac remodelling. METHODS AND RESULTS: Myocardial infarction was induced in mice by ligating the left coronary artery. During both the subacute and chronic stages (1 and 3 weeks postinfarction, respectively), autophagy was found to be activated in surviving cardiomyocytes, as demonstrated by the up-regulated expression of microtubule-associated protein-1 light chain 3-II (LC3-II), p62 and cathepsin D, and by electron microscopic findings. Activation of autophagy, specifically the digestion step, was prominent in cardiomyocytes 1 week postinfarction, especially in those bordering the infarct area, while the formation of autophagosomes was prominent 3 weeks postinfarction. Bafilomycin A1 (an autophagy inhibitor) significantly aggravated postinfarction cardiac dysfunction and remodelling. Cardiac hypertrophy was exacerbated in this group and was accompanied by augmented ventricular expression of atrial natriuretic peptide. In these hearts, autophagic findings (i.e. expression of LC3-II and the presence of autophagosomes) were diminished, and activation of AMP-activated protein kinase was enhanced. Treatment with rapamycin (an autophagy enhancer) brought about opposite outcomes, including mitigation of cardiac dysfunction and adverse remodelling. A combined treatment with bafilomycin A1 and rapamycin offset each effect on cardiomyocyte autophagy and cardiac remodelling in the postinfarction heart. CONCLUSION: These findings suggest that cardiomyocyte autophagy is an innate mechanism that protects against progression of postinfarction cardiac remodelling, implying that augmenting autophagy could be a therapeutic strategy.


Subject(s)
Autophagy , Heart Ventricles/pathology , Hypertrophy, Left Ventricular/etiology , Myocardial Infarction/complications , Myocytes, Cardiac/pathology , Ventricular Function, Left , Ventricular Remodeling , AMP-Activated Protein Kinases/metabolism , Analysis of Variance , Animals , Atrial Natriuretic Factor/metabolism , Autophagy/drug effects , Blotting, Western , Cathepsin D/metabolism , Disease Models, Animal , Enzyme Activation , Fluorescent Antibody Technique , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Heart Ventricles/physiopathology , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Macrolides/pharmacology , Mice , Microscopy, Electron , Microtubule-Associated Proteins/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Phosphorylation , Sirolimus/pharmacology , Time Factors , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects
5.
Am J Physiol Heart Circ Physiol ; 300(6): H2261-71, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21421825

ABSTRACT

Ischemia is known to potently stimulate autophagy in the heart, which may contribute to cardiomyocyte survival. In vitro, transfection with small interfering RNAs targeting Atg5 or Lamp-2 (an autophagy-related gene necessary, respectively, for the initiation and digestion step of autophagy), which specifically inhibited autophagy, diminished survival among cultured cardiomyocytes subjected to anoxia and significantly reduced their ATP content, confirming an autophagy-mediated protective effect against anoxia. We next examined the dynamics of cardiomyocyte autophagy and the effects of manipulating autophagy during acute myocardial infarction in vivo. Myocardial infarction was induced by permanent ligation of the left coronary artery in green fluorescent protein-microtubule-associated protein 1 light chain 3 (GFP-LC3) transgenic mice in which GFP-LC3 aggregates to be visible in the cytoplasm when autophagy is activated. Autophagy was rapidly (within 30 min after coronary ligation) activated in cardiomyocytes, and autophagic activity was particularly strong in salvaged cardiomyocytes bordering the infarcted area. Treatment with bafilomycin A1, an autophagy inhibitor, significantly increased infarct size (31% expansion) 24 h postinfarction. Interestingly, acute infarct size was significantly reduced (23% reduction) in starved mice showing prominent autophagy before infarction. Treatment with bafilomycin A1 reduced postinfarction myocardial ATP content, whereas starvation increased myocardial levels of amino acids and ATP, and the combined effects of bafilomycin A1 and starvation on acute infarct size offset one another. The present findings suggest that autophagy is an innate and potent process that protects cardiomyocytes from ischemic death during acute myocardial infarction.


Subject(s)
Autophagy/physiology , Coronary Occlusion/complications , Myocardial Infarction/etiology , Myocardial Infarction/physiopathology , Myocardial Ischemia/physiopathology , Animals , Autophagy/drug effects , Autophagy-Related Protein 5 , Cells, Cultured , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Lysosomal-Associated Membrane Protein 2/antagonists & inhibitors , Lysosomal-Associated Membrane Protein 2/metabolism , Macrolides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Models, Animal , Myocardial Infarction/metabolism , Myocardial Ischemia/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , RNA, Small Interfering/pharmacology
6.
J Cardiovasc Pharmacol ; 57(4): 416-23, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21242812

ABSTRACT

Granulocyte colony-stimulating factor (G-CSF) is a potent angiogenic factor. We hypothesized that G-CSF-immersed gelatin hydrogel microspheres (G-CSF-GHMs) injected into the ischemic legs might continuously release a small amount of G-CSF to locally stimulate angiogenesis without unfavorable systemic effects. Just after ligation of the right femoral artery of BALB/c mice, recombinant human G-CSF (100-µg/kg)-immersed GHM was injected into the right hindlimb muscles; the controls included a saline-injected group, an intramuscularly injected G-CSF group, a subcutaneously injected G-CSG group, and an empty GHM-injected group. Eight weeks later, improvement of blood perfusion to the ischemic limb was significantly augmented in the G-CSF-GHM group compared with any of the control groups. Despite there being no increase in the serum concentration of G-CSF, in peripheral granulocytes, or in circulating endothelial progenitor cells, not only capillary but also arteriolar density was significantly increased in this group. Next, we started treatment with G-CSF-GHM 4 weeks after ligation to examine whether the treatment is effective if performed during the chronic stage of ischemia. The late treatment was also found to effectively improve blood flow in the ischemic leg. In conclusion, G-CSF-GHM administration is suggested to be a promising and readily usable approach to treating peripheral artery disease, applicable even during the chronic stage.


Subject(s)
Granulocyte Colony-Stimulating Factor/pharmacology , Ischemia/drug therapy , Microspheres , Peripheral Arterial Disease/drug therapy , Animals , Disease Models, Animal , Gelatin/chemistry , Granulocyte Colony-Stimulating Factor/administration & dosage , Hindlimb/blood supply , Hindlimb/drug effects , Humans , Hydrogels , Injections, Intramuscular , Ischemia/pathology , Male , Mice , Mice, Inbred BALB C , Peripheral Arterial Disease/pathology , Recombinant Proteins , Time Factors
7.
J Am Coll Cardiol ; 56(23): 1949-58, 2010 Nov 30.
Article in English | MEDLINE | ID: mdl-21109120

ABSTRACT

OBJECTIVES: We examined the effect of asialoerythropoietin (asialoEPO), a nonerythrogenic derivative of erythropoietin (EPO), on renal dysfunction-associated heart failure. BACKGROUND: Although EPO is known to exert beneficial effects on cardiac function, the clinical benefits in patients with chronic kidney disease are controversial. It remains to be addressed whether previously reported outcomes were the result of relief of the anemia, adverse effects of EPO, or direct cardiovascular effects. METHODS: Mice underwent 5/6 nephrectomy to cause renal dysfunction. Eight weeks later, when renal dysfunction was established, anemia and cardiac dysfunction and remodeling were apparent. Mice were then assigned to receive saline (control), recombinant human erythropoietin (rhEPO) at 5,000 IU (714 pmol)/kg, or asialoEPO at 714 pmol/kg, twice/week for 4 weeks. RESULTS: Although only rhEPO relieved the nephrectomy-induced anemia, both rhEPO and asialoEPO significantly and similarly mitigated left ventricular dilation and dysfunction. The hearts of rhEPO- or asialoEPO-treated mice showed less hypertrophy, reflecting decreases in cardiomyocyte hypertrophy and degenerative subcellular changes, as well as significant attenuation of fibrosis, leukocyte infiltration, and oxidative deoxyribonucleic acid damage. These phenotypes were accompanied by restored expression of GATA-4, sarcomeric proteins, and vascular endothelial growth factor and decreased inflammatory cytokines and lipid peroxidation. Finally, myocardial activation was observed of extracellular signal-regulated protein kinase and signal transducer and activator of transcription pathways in the treated mice. CONCLUSIONS: EPO receptor signaling exerts direct cardioprotection in an animal model of renal dysfunction-associated heart failure, probably by mitigating degenerative, pro-fibrosis, inflammatory, and oxidative processes but not through relief of anemia.


Subject(s)
Anemia/complications , Asialoglycoproteins/therapeutic use , Erythropoietin/analogs & derivatives , Erythropoietin/therapeutic use , Heart Failure/etiology , Receptors, Erythropoietin/metabolism , Renal Insufficiency/complications , Signal Transduction , Anemia/drug therapy , Anemia/metabolism , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Heart Failure/metabolism , Heart Failure/prevention & control , Male , Mice , Mice, Inbred C57BL , Oxidative Stress , Recombinant Proteins , Renal Insufficiency/drug therapy , Renal Insufficiency/metabolism
8.
Am J Physiol Heart Circ Physiol ; 299(2): H372-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20495150

ABSTRACT

Anemia may accelerate angiogenesis in ischemic organs through its ability to augment tissue hypoxia-induced generation of several known angiogenic factors and to increase erythropoietin levels, which are also potently angiogenic. We examined the effect of controlled phlebotomy (bloodletting) on blood flow in a mouse ischemic leg model. We ligated the right femoral artery of BALB/c mice. In the phlebotomy group, 200 microl of blood were drawn from the tail vein once a week. After 4 wk, blood flow in the ischemic leg was significantly better in the phlebotomy group (flow ratio of the ischemic to nonischemic leg, 0.87 + or - 0.04) than the control group (0.59 + or - 0.05, P < 0.05), and capillary density was significantly higher. Repeated phlebotomy increased serum erythropoietin levels as well as the expression of hypoxia-inducible transcription factor-1alpha and vascular endothelial growth factor and both the expression and activity of Akt and endothelial nitric oxide synthase (eNOS) in ischemic legs. Treatment with wortmannin or N(omega)-nitro-l-arginine methyl ester significantly attenuated the phlebotomy-induced improvement of blood flow. In addition, fluorescence-activated cell sorting analysis revealed an increase in circulating peripheral endothelial progenitor cells in the phlebotomy group, and treatment with AMD3100, a specific inhibitor of the chemokine receptor CXCR4, blocked the beneficial effect of phlebotomy. These findings suggest that repeated phlebotomy improves blood flow in ischemic legs through an angiogenic action that involves the Akt/eNOS pathway, endothelial progenitor cell mobilization, and their complicated cross talk. An adequately controlled phlebotomy might be one method by which to induce therapeutic angiogenesis.


Subject(s)
Capillaries/physiopathology , Ischemia/therapy , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Phlebotomy , Androstadienes/pharmacology , Animals , Benzylamines , Capillaries/drug effects , Capillaries/metabolism , Capillaries/pathology , Cyclams , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Enzyme Inhibitors/pharmacology , Erythropoietin/blood , Femoral Artery/surgery , Heterocyclic Compounds/pharmacology , Hindlimb , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ischemia/metabolism , Ischemia/pathology , Ischemia/physiopathology , Ligation , Male , Mice , Mice, Inbred BALB C , NG-Nitroarginine Methyl Ester/pharmacology , Neovascularization, Physiologic/drug effects , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism , Regional Blood Flow , Signal Transduction , Stem Cells/metabolism , Stem Cells/pathology , Time Factors , Vascular Endothelial Growth Factor A/metabolism , Wortmannin
9.
Am J Pathol ; 176(2): 687-98, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20035047

ABSTRACT

Activation of Fas signaling is a key mediator of doxorubicin cardiotoxicity, which involves both cardiomyocyte apoptosis and myocardial inflammation. In this study, acute cardiotoxicity was induced in mice by doxorubicin, and some mice simultaneously received an intramuscular injection of adenoviral vector encoding mouse soluble Fas (sFas) gene (Ad.CAG-sFas), an inhibitor of Fas/Fas ligand interaction. Two weeks later, left ventricular dilatation and dysfunction were apparent in the LacZ-treated control group, but both were significantly mitigated in the sFas-treated group. The in situ nick-end labeling-positive rate were similar in the two groups, and although electron microscopy revealed cardiomyocyte degeneration, no apoptotic structural features and no activation of caspases were detected, suggesting an insignificant role of apoptosis in this model. Instead, sFas treatment reversed doxorubicin-induced down-regulation of GATA-4 and attenuated ubiquitination of myosin heavy chain and troponin I to preserve these sarcomeric proteins. In addition, doxorubicin-induced significant leukocyte infiltration, fibrosis, and oxidative damage to the myocardium, all of which were largely reversed by sFas treatment. sFas treatment also suppressed doxorubicin-induced p53 overexpression, phosphorylation of c-Jun N-terminal kinase, c-Jun, and inhibitor of nuclear factor-kappaB, as well as production of cyclooxygenase-2 and monocyte chemoattractant protein-1, and it restored extracellular signal-regulated kinase activation. Therefore, sFas gene therapy prevents the progression of doxorubicin-induced acute cardiotoxicity, with accompanying attenuation of the cardiomyocyte degeneration, inflammation, fibrosis, and oxidative damage caused by Fas signaling.


Subject(s)
Apoptosis/physiology , Doxorubicin , Genetic Therapy/methods , Heart Diseases/chemically induced , Heart Diseases/therapy , fas Receptor/genetics , Animals , Apoptosis/genetics , DNA Damage/genetics , Echocardiography , Endomyocardial Fibrosis/genetics , Endomyocardial Fibrosis/pathology , Endomyocardial Fibrosis/prevention & control , Heart Diseases/genetics , Heart Diseases/pathology , Inflammation/genetics , Inflammation/pathology , Lac Operon , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardium/pathology , Myocardium/ultrastructure , Oxidative Stress/genetics , Oxidative Stress/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Solubility , fas Receptor/antagonists & inhibitors
10.
Am J Physiol Heart Circ Physiol ; 297(4): H1504-13, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19684189

ABSTRACT

The small leucine-rich proteoglycan decorin is a natural inhibitor of transforming growth factor-beta (TGF-beta) and exerts antifibrotic effects in heart and to stimulate skeletal muscle regeneration. We investigated decorin's chronic effects on postinfarction cardiac remodeling and dysfunction. Myocardial infarction (MI) was induced in mice by left coronary artery ligation. An adenoviral vector encoding human decorin (Ad. CAG-decorin) was then injected into the hindlimbs on day 3 post-MI (control, Ad.CAG-LacZ). Four weeks post-MI, the decorin-treated mice showed significant mitigation of the left ventricular dilatation and dysfunction seen in control mice. Although infarct size did not differ between the two groups, the infarcted wall thickness was greater and the segmental length of the infarct was smaller in decorin-treated mice. In addition, cellular components, including myofibroblasts and blood vessels, were more abundant within the infarcted area in decorin-treated mice, and fibrosis was significantly reduced in both the infarcted and noninfarcted areas of the left ventricular wall. Ten days post-MI, there was greater cell proliferation and less apoptosis among granulation tissue cells in the infarcted areas of decorin-treated mice. The treatment, however, did not affect proliferation and apoptosis of salvaged cardiomyocytes. Although decorin gene therapy did not affect TGF-beta1 expression in the infarcted heart, it inhibited Smad2/3 activation (downstream mediators of TGF-beta signaling). In summary, postinfarction decorin gene therapy mitigated cardiac remodeling and dysfunction by altering infarct tissue noncardiomyocyte dynamics and preventing cardiac fibrosis, accompanying inhibition of Smad2/3 activation.


Subject(s)
Adenoviridae/genetics , Extracellular Matrix Proteins/biosynthesis , Genetic Therapy , Genetic Vectors , Hypertrophy, Left Ventricular/prevention & control , Myocardial Infarction/therapy , Myocytes, Cardiac/metabolism , Proteoglycans/biosynthesis , Ventricular Dysfunction, Left/prevention & control , Ventricular Remodeling , Animals , Apoptosis , Cell Proliferation , Chronic Disease , Decorin , Disease Models, Animal , Extracellular Matrix Proteins/genetics , Fibrosis , HeLa Cells , Humans , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocytes, Cardiac/pathology , Proteoglycans/genetics , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Time Factors , Transduction, Genetic , Transfection , Transforming Growth Factor beta1/metabolism , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/physiopathology
11.
Autophagy ; 5(7): 1034-6, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19587530

ABSTRACT

To examine the functional significance and detailed morphological characteristics of starvation-induced autophagy in the adult heart, we starved green fluorescent protein (GFP)-microtubule-associated protein 1 light chain 3 (LC3) transgenic mice for up to 3 days. Electron microscopy revealed that, after as little as 12 hours of starvation, round and homogenously electron-dense lipid droplet-like vacuoles appeared in cardiomyocytes. These were determined to be lysosomes based on cathepsin D immunopositivity and acid phosphatase activity. The number of these lysosomes increased with starvation time, and typical autolysosomes with intracellular organelles destined for degradation appeared and increased in number at later times during the starvation period. Myocardial expression of the autophagy-related proteins LC3-II, cathepsin D and ubiquitin increased, while myocardial ATP content decreased, as the starvation interval proceeded. Treatment with bafilomycin A(1), an autophagy inhibitor, did not affect cardiac function in normally fed mice, but it significantly depressed cardiac function and caused significant left ventricular dilatation in the mice starved for 3 days. Cardiomyocytes from starved mice treated with bafilomycin A(1) showed marked accumulation of lysosomes, and the myocardial amino acid content, which increased during starvation in normally fed mice, as well as the myocardial ATP content, were severely reduced, which likely contributed to the cardiac dysfunction. The present findings suggest autophagy plays a critical role in the maintenance of cardiac function during starvation in the adult.


Subject(s)
Autophagy/physiology , Myocardium/metabolism , Starvation/metabolism , Animals , Lysosomes/metabolism , Lysosomes/ultrastructure , Macrolides/pharmacology , Mice , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Myocardium/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Vacuoles/metabolism , Vacuoles/ultrastructure
12.
Med Mol Morphol ; 42(2): 92-101, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19536616

ABSTRACT

To address whether adult rat ventricular cardiomyocytes (ARVCs) exposed to oxidant stress die via apoptosis (secondarily by necrosis) or primarily by necrosis, we exposed ARVCs to hydrogen peroxide (H2O2; 0.1-100 microM) for up to 24 h and then compared them with isoproterenol-induced apoptotic and Triton X-induced necrotic controls. Cellular shrinkage preceded plasma membrane disruption, reflected by trypan blue uptake in ARVCs exposed to lower concentrations of H2O2 (<1 microM; an apoptotic pattern), but the order was reversed in cells exposed to higher concentrations of H2O2 (>1 microM; a necrotic pattern). DNA fragmentation, caspase-3 activation, mitochondrial membrane potential preservation, and ATP preservation were all apparent in ARVCs treated with low H2O2 (0.5 microM), but not in those treated with high H2O2 (10 microM). In addition, electron microscopy revealed unique morphology in H2O2-treated ARVCs; i.e., the nuclei had a homogeneous ground glass-like appearance that was never accompanied by chromatin condensation. Apparently, high concentrations of H2O2 caused primary necrosis in ARVCs, whereas low concentrations induced biochemically comparable apoptosis, although the latter did not satisfy the morphological criteria of apoptosis. These findings caution against the use of oxidant stress, H2O2 in particular, as an inducer of apoptosis in ARVCs.


Subject(s)
Apoptosis , Hydrogen Peroxide/pharmacology , Myocytes, Cardiac/cytology , Oxidants/pharmacology , Oxidative Stress , Adenosine Triphosphate/metabolism , Animals , Caspase 3/metabolism , Cells, Cultured , DNA Fragmentation , Heart Ventricles/cytology , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/ultrastructure , Models, Animal , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/ultrastructure , Necrosis , Rats , Rats, Sprague-Dawley
13.
J Am Coll Cardiol ; 53(25): 2378-88, 2009 Jun 23.
Article in English | MEDLINE | ID: mdl-19539150

ABSTRACT

OBJECTIVES: We hypothesized that erythropoietin (EPO)-immersed gelatin hydrogel microspheres (GHM) injected into ischemic legs might continuously release a small amount of EPO to locally stimulate angiogenesis without unfavorable systemic effects. BACKGROUND: EPO is a potent angiogenic factor, but its use for relieving ischemic organs is limited because of the untoward systemic erythrogenic effect and its short half-life in plasma. METHODS: The right femoral arteries of BALB/c mice were ligated. Recombinant human EPO (5,000 IU/kg)-immersed GHM was injected into the right hind limb muscles (n = 12); the control groups included a saline-injected group (n = 12), an EPO-injected group (n = 8), and an empty GHM-injected group (n = 8). RESULTS: Eight weeks later, improvement of blood perfusion to the ischemic limb was significantly augmented in the EPO-GHM group compared with any of the control groups. There was no increase in the hemoglobin level, nor was there any increase in endothelial progenitor cells. However, capillary and arteriolar densities were significantly increased in this group. Although the treatment did not affect the levels of vascular endothelial growth factor or interleukin-1 beta, it up-regulated the EPO receptor and matrix metalloproteinase-2 and activated the downstream signaling of Akt and also endothelial nitric oxide synthase in ischemic limbs, which might have been associated with the evident angiogenic and arteriogenic effects in the present system. CONCLUSIONS: The present drug delivery system is suggested to have potential as a novel noninvasive therapy for ischemic peripheral artery disease.


Subject(s)
Erythropoietin/administration & dosage , Ischemia/drug therapy , Lower Extremity/blood supply , Peripheral Vascular Diseases/drug therapy , Adult Stem Cells/drug effects , Angiogenesis Inducing Agents/metabolism , Animals , Cells, Cultured , Delayed-Action Preparations , Endothelial Cells/drug effects , Hydrogels/therapeutic use , Ischemia/metabolism , Male , Mice , Mice, Inbred BALB C , Microspheres , Myocytes, Smooth Muscle/drug effects , Neovascularization, Physiologic/drug effects , Peripheral Vascular Diseases/metabolism , Receptors, Erythropoietin/metabolism , Recombinant Proteins , Regional Blood Flow/drug effects
14.
Am J Pathol ; 174(5): 1705-14, 2009 May.
Article in English | MEDLINE | ID: mdl-19342365

ABSTRACT

To examine the functional significance and morphological characteristics of starvation-induced autophagy in the adult heart, we made green fluorescent protein-microtubule-associated protein 1-light chain 3 (LC3) transgenic mice starve for up to 3 days. Electron microscopy revealed round, homogenous, electron-dense lipid droplet-like vacuoles that initially appeared in cardiomyocytes as early as 12 hours after starvation; these vacuoles were identified as lysosomes based on cathepsin D-immunopositive reactivity and acid phosphatase activity. The increase in the number of lysosomes depended on the starvation interval; typical autophagolysosomes with intracellular organelles also appeared, and their numbers increased at the later phases of starvation. Myocardial expression of autophagy-related proteins, LC3-II, cathepsin D, and ubiquitin, increased, whereas both myocardial ATP content and starvation integral decreased. Treatment with bafilomycin A1, an autophagy inhibitor, did not affect cardiac function in normally fed mice but significantly depressed cardiac function and caused significant left ventricular dilatation in mice starved for 3 days. The cardiomyocytes were occupied with markedly accumulated lysosomes in starved mice treated with bafilomycin A1, and both the myocardial amino acid content, which was increased during starvation, and the myocardial ATP content were severely decreased, potentially contributing to cardiac dysfunction. The present findings suggest a critical role of autophagy in the maintenance of cardiac function during starvation in the adult.


Subject(s)
Autophagy , Microtubule-Associated Proteins/physiology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/ultrastructure , Adenosine Triphosphate/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Body Weight/drug effects , Cathepsin D/metabolism , Enzyme Inhibitors/pharmacology , Fluorescent Antibody Technique , Green Fluorescent Proteins/genetics , Heart Function Tests , Immunoenzyme Techniques , Lysosomes/metabolism , Lysosomes/ultrastructure , Macrolides/pharmacology , Mice , Mice, Transgenic , Myocytes, Cardiac/metabolism , Proton-Translocating ATPases/antagonists & inhibitors , Rats , Ubiquitin/metabolism , Vacuoles/metabolism , Vacuoles/ultrastructure , Ventricular Dysfunction, Left/metabolism
15.
Am J Physiol Heart Circ Physiol ; 296(3): H616-26, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19151252

ABSTRACT

We hypothesized that therapy, composed of antiapoptotic soluble Fas (sFas) gene transfer, combined with administration of the cardioprotective cytokine granulocyte colony-stimulating factor (G-CSF), would markedly mitigate cardiac remodeling and dysfunction following myocardial infarction (MI). On the 3rd day after MI induced by ligating the left coronary artery in mice, four different treatments were initiated: saline injection (Group C, n = 26); G-CSF administration (Group G, n = 27); adenoviral transfer of sFas gene (Group F, n = 26); and the latter two together (Group G+F, n = 26). Four weeks post-MI, Group G+F showed better survival than Group C (96 vs. 65%, P < 0.05) and the best cardiac function among the four groups. In Group G, the infarct scar was smaller and less fibrotic, whereas in Group F the scar was thicker, without a reduction in area, and contained abundant myofibroblasts and vascular cells; Group G+F showed both phenotypes. G-CSF exerted a beneficial effect on infarct tissue dynamics through antifibrotic and proliferative effects on granulation tissue; however, it also exerts an adverse proapoptotic effect that leads to thinning of the infarct scar. sFas appeared to offset the latter drawback. In vitro study using cultured myofibroblasts derived from the infarct tissue revealed that G-CSF increased proliferating activity of those cells accompanying activation of Akt and signal transducer and activator of transcription 3, while accelerating Fas-mediated apoptosis with increasing Bax-to-Bcl-2 ratio. The results suggest that combined use of G-CSF administration and sFas gene therapy is a potentially powerful tool against post-MI heart failure.


Subject(s)
Apoptosis , Cardiotonic Agents/pharmacology , Gene Transfer Techniques , Genetic Therapy , Granulocyte Colony-Stimulating Factor/pharmacology , Heart Failure/therapy , Myocardial Infarction/therapy , Myocardium/pathology , fas Receptor/genetics , Adenoviridae/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Proliferation/drug effects , Cells, Cultured , Combined Modality Therapy , Disease Models, Animal , Fibroblasts/drug effects , Fibroblasts/pathology , Fibrosis , Genetic Vectors , Granulation Tissue/drug effects , Granulation Tissue/pathology , Heart Failure/etiology , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Lenograstim , Male , Mice , Mice, Inbred C57BL , Myocardial Contraction/drug effects , Myocardial Infarction/complications , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Recombinant Proteins/pharmacology , Regeneration/drug effects , Regeneration/genetics , Time Factors , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/therapy , Ventricular Remodeling/drug effects , bcl-2-Associated X Protein/metabolism , fas Receptor/metabolism
16.
Autophagy ; 5(1): 90-2, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19001873

ABSTRACT

We induced autophagy in isolated adult rat ventricular cardiomyocytes by incubating them in glucose-free medium supplemented with mannitol for up to 4 days. The upregulation of LC3 and vacuoles containing partially degraded subcellular organelles were readily apparent in glucose-starved cells. Most dead cells in both groups showed features of necrosis, although the survival rate was significantly lower among glucose-starved cells than among the controls. In contrast, the rate of apoptosis was about the same in both groups. Two inhibitors of autophagy, 3-methyladenine (3-MA) and leupeptin, significantly reduced the viability of both control and glucose-starved cells in a dose-dependent manner and caused specific morphological alterations: 3-MA reduced the number of autophagic vacuoles, whereas leupeptin greatly increased their number and size. Conversely, rapamycin, an enhancer of autophagy, improved the survival of glucose-starved cells. The reduction in cellular ATP caused by glucose depletion was exacerbated by the inhibitors but mitigated by rapamycin, suggesting that inhibition of autophagy may accelerate energy depletion, leading to necrosis. Our findings suggest that in cardiomyocytes, autophagy is a compensatory, prosurvival response to stress, and that autophagic death is an unsuccessful outcome brought on by necrosis.


Subject(s)
Autophagy , Glucose/deficiency , Myocytes, Cardiac/cytology , Animals , Cell Separation , Cell Shape , Culture Media , Models, Biological , Myocytes, Cardiac/ultrastructure , Rats
17.
Am J Physiol Heart Circ Physiol ; 295(4): H1599-607, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18708438

ABSTRACT

Autophagy is simultaneously a mode of programmed cell death and an important physiological process for cell survival, but its pathophysiological significance in cardiac myocytes remains largely unknown. We induced autophagy in isolated adult rat ventricular cardiomyocytes (ARVCs) by incubating them in glucose-free, mannitol-supplemented medium for up to 4 days. Ultrastructurally, intracellular vacuoles containing degenerated subcellular organelles (e.g., mitochondria) were markedly apparent in the glucose-starved cells. Microtubule-associated protein-1 light chain 3 was significantly upregulated among the glucose-starved ARVCs than among the controls. After 4 days, glucose-starved ARVCs showed a significantly worse survival rate (19+/-5.2%) than the controls (55+/-8.3%, P<0.005). Most dead ARVCs in both groups showed features of necrosis, and the rate of apoptosis did not differ between the groups. Two inhibitors of autophagy, 3-methyladenine (3-MA) and leupeptin, significantly and dose-dependently reduced the viability of both control and glucose-starved ARVCs and caused specific morphological alterations; 3-MA reduced autophagic findings, whereas leupeptin greatly increased the numbers and the sizes of vacuoles that contained incompletely digested organelles. The knockdown of the autophagy-related genes with small interfering RNA also reduced the glucose-starved ARVCs viability, but rapamycin, an autophagy enhancer, improved it. Reductions in the ATP content of ARVCs caused by glucose depletion were exacerbated by the inhibitors while attenuated by rapamycin, suggesting that autophagy inhibition might accelerate energy depletion, leading to necrosis. Taken together, our findings suggest that autophagy in cardiomyocytes reflects a prosurvival, compensatory response to stress and that autophagic cardiomyocyte death represents an unsuccessful outcome due to necrosis.


Subject(s)
Autophagy , Cell Shape , Glucose/deficiency , Myocytes, Cardiac/ultrastructure , Vacuoles/ultrastructure , Adenine/analogs & derivatives , Adenine/pharmacology , Adenosine Triphosphate/metabolism , Animals , Autophagy/drug effects , Autophagy/genetics , Cell Shape/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Leupeptins/pharmacology , Male , Microtubule-Associated Proteins/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Necrosis , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Sirolimus/pharmacology , Time Factors
18.
J Neurosci Res ; 86(13): 2820-8, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18618669

ABSTRACT

Adenosine, a modulator of neuronal function in the mammalian central nervous system, exerts a neuroprotective effect via the adenosine A(1) receptor; however, its effect on neural stem cells (NSCs) remains unclear. Because adenosine is released in response to pathological conditions and NSCs play a key role in neuroregeneration, we tested the hypothesis that adenosine is capable of stimulating NSC proliferation. We demonstrated that NSCs dominantly express adenosine A(1) and A(2B) receptors. Adenosine and the adenosine A(1) receptor agonist cyclopentyladenosine (CPA) increased proliferation of NSCs, and this CPA-induced cell proliferation was attenuated by the A(1) antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPA). CPA also induced phosphorylation of extracellular signal-regulated kinase (ERK), mitogen-activated protein kinase/ERK kinase (MEK), and Akt, and their phosphorylation was inhibited by DPCPA. In addition, CPA-induced cell proliferation was inhibited by MEK and Akt inhibitors. These results suggest that activation of adenosine A(1) receptor-stimulated proliferation of NSCs occurs via MEK/ERK and Akt signaling pathways.


Subject(s)
Cell Proliferation , Neurons/cytology , Receptor, Adenosine A1/metabolism , Signal Transduction/physiology , Stem Cells/cytology , Animals , Blotting, Western , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Inbred C57BL , Neurons/metabolism , Oncogene Protein v-akt/metabolism , Phosphorylation , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism
19.
Circ Res ; 103(1): 98-106, 2008 Jul 03.
Article in English | MEDLINE | ID: mdl-18519944

ABSTRACT

Although recanalization of the infarct-related artery late after myocardial infarction (MI) is known to reduce both cardiac remodeling and mortality, the mechanisms responsible are not yet fully understood. We compared infarcted rat hearts in which the infarct-related coronary artery was opened 24 hours after infarction (late reperfusion [LR] group) with those having a permanently occluded artery. Left ventricular dilatation and dysfunction were significantly mitigated in the LR group 1, 2, and 4 weeks post-MI. Attributable, in large part, to the greater number of cells present, the infarcted wall was significantly thicker in the LR group, which likely reduced wall stress and mitigated cardiac dysfunction. Granulation tissue cell proliferation was increased to a greater degree in the LR group 4 days post-MI, whereas the incidence of apoptosis was significantly lower throughout the subacute stage (4 days, 1 week, and 2 weeks post-MI), further suggesting preservation of granulation tissue cells contributes to the thick, cell-rich scar. Functionally, myocardial debris was more rapidly removed from the infarcted areas in the LR group during subacute stages, and stouter collagen was more rapidly synthesized in those areas. Direct acceleration of Fas-mediated apoptosis by hypoxia was confirmed in vitro using infarct tissue-derived myofibroblasts. In salvaged cardiomyocytes, degenerative changes, but not apoptosis, were mitigated in the LR group, accompanied by restoration of GATA-4 and sarcomeric protein expression. Along with various mechanisms proposed earlier, the present findings appear to provide an additional pathophysiological basis for the benefits of late reperfusion.


Subject(s)
Myocardial Infarction/physiopathology , Myocardial Reperfusion , Myocytes, Cardiac , Ventricular Remodeling , Animals , Apoptosis , Carrier Proteins/metabolism , Collagen/biosynthesis , Cytoskeletal Proteins , Fibroblasts/metabolism , Fibroblasts/pathology , GATA4 Transcription Factor/metabolism , Humans , Male , Myocardial Infarction/metabolism , Myocardial Infarction/mortality , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Myocardial Reperfusion/methods , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats , Rats, Wistar , Time Factors
20.
Clin Exp Pharmacol Physiol ; 35(11): 1288-93, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18505441

ABSTRACT

1. In the present study, we examined the effects of inhibiting transforming growth factor (TGF)-beta in a mouse model of diabetic nephropathy. 2. An adenovirus harbouring the gene encoding soluble TGF-beta type II receptor (Ad.CAG-sTbetaRII), a competitive inhibitor of TGF-beta, was injected into hindlimb muscles (systemic delivery) of mice 5 weeks after the induction of diabetes with streptozotocin. The control group was injected with an adenovirus encoding the LacZ gene (Ad-LacZ). 3. Five weeks after administration, anti-TGF-beta gene therapy was found to have had no effect on renal function, albuminuria or glucose metabolism in mice with diabetic nephropathy. Nonetheless, this gene therapy did significantly reduce fibrosis in both glomeruli and renal tubules. These effects were accompanied by attenuation of the increased expression of alpha-smooth muscle actin normally seen in kidneys of diabetic mice and better preservation of glomerular cell numbers, although the thickness of the glomerular capillary basement membrane was unchanged. The plasma concentration of soluble TGF-beta type II receptor peaked on Day 7 after treatment, but was undetectable by Day 14. Moreover, a second treatment with Ad.CAG-sTbetaRII failed to prolong the interval of gene product expression in the blood. 4. The present anti-TGF-beta gene therapy showed a significant antifibrotic effect in a model of diabetic nephropathy, but failed to improve renal function. The inadequacy of the observed effect is likely due to the relatively short interval of gene product expression. This problem will have to be overcome if gene therapies for slowly progressing diseases, like diabetic nephropathy, are to be realised.


Subject(s)
Adenoviridae/genetics , Diabetic Nephropathies/genetics , Diabetic Nephropathies/therapy , Disease Models, Animal , Genetic Vectors/administration & dosage , Protein Serine-Threonine Kinases/administration & dosage , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/administration & dosage , Receptors, Transforming Growth Factor beta/genetics , Animals , Chickens , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/therapy , Diabetic Nephropathies/pathology , Female , Genetic Therapy/methods , Genetic Vectors/genetics , Humans , Mice , Mice, Inbred C57BL , Rabbits , Receptor, Transforming Growth Factor-beta Type II , Solubility , Transforming Growth Factor beta/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...