15 results on '"S Van Linthout"'
Search Results
2. Circulating miRNAs in Hypertrophic Cardiomyopathy: A Long and Bumpy Road.
- Author
-
van der Meer P and Van Linthout S
- Subjects
- Humans, Heart Failure, Cardiomyopathy, Hypertrophic genetics, MicroRNAs genetics
- Abstract
Competing Interests: Disclosures The University Medical Center Groningen, which employs Dr van der Meer, received consultancy fees and/or grants from Novartis, Pharmacosmos, Vifor Pharma, AstraZeneca, Pfizer, Pharma Nord, BridgeBio, Novo Nordisk, and Ionis. The other authors report no conflicts.
- Published
- 2023
- Full Text
- View/download PDF
3. Early Treatment of Coxsackievirus B3-Infected Animals With Soluble Coxsackievirus-Adenovirus Receptor Inhibits Development of Chronic Coxsackievirus B3 Cardiomyopathy.
- Author
-
Pinkert S, Dieringer B, Klopfleisch R, Savvatis K, Van Linthout S, Pryshliak M, Tschöpe C, Klingel K, Kurreck J, Beling A, and Fechner H
- Subjects
- Animals, Cardiomyopathies pathology, Cardiomyopathies physiopathology, Cardiomyopathies virology, Chronic Disease, Coxsackievirus Infections pathology, Coxsackievirus Infections physiopathology, Coxsackievirus Infections virology, Disease Models, Animal, Enterovirus B, Human pathogenicity, Fibrosis, Male, Mice, Myocarditis pathology, Myocarditis virology, Myocardium pathology, Recombinant Fusion Proteins adverse effects, Ventricular Function, Left, Viral Load, Antiviral Agents administration & dosage, Cardiomyopathies drug therapy, Coxsackievirus Infections drug therapy, Enterovirus B, Human drug effects, Immunoconjugates administration & dosage, Immunoglobulin G administration & dosage, Myocarditis drug therapy, Receptors, Virus administration & dosage
- Abstract
Background: Coxsackie-B-viruses (CVB) are frequent causes of acute myocarditis and dilated cardiomyopathy, but an effective antiviral therapy is still not available. Previously, we and others have demonstrated that treatment with an engineered sCAR-Fc (soluble coxsackievirus-adenovirus receptor fused to the carboxyl-terminus of human IgG) efficiently neutralizes CVB3 and inhibits the development of cardiac dysfunction in mice with acute CVB3-induced myocarditis. In this study, we analyzed the potential of sCAR-Fc for treatment of chronic CVB3-induced myocarditis in an outbred NMRI mouse model., Methods: NMRI mice were infected with the CVB3 strain 31-1-93 and treated with a sCAR-Fc expressing adeno-associated virus 9 vector 1, 3, and 7 days after CVB3 infection. Chronic myocarditis was analyzed on day 28 after infection., Results: Initial investigations showed that NMRI mice develop pronounced chronic myocarditis between day 18 and day 28 after infection with the CVB3 strain 31-1-93. Chronic cardiac infection was characterized by inflammation and fibrosis as well as persistence of viral genomes in the heart tissue and by cardiac dysfunction. Treatment of NMRI mice resulted in a distinct reduction of cardiac inflammation and fibrosis and almost complete elimination of virus RNA from the heart by day 28 after infection. Moreover, hemodynamic measurement revealed improved cardiac contractility and diastolic relaxation in treated mice compared with mice treated with a control vector (mean±SD; maximal pressure, 81.9±9.2 versus 69.4±8.6 mm Hg, P =0.02; left ventricular ejection fraction, 68.9±8.5 versus 54.2±11.5%, P =0.02; dP/dt
max , 7275.2±1674 versus 4432.6±1107 mm Hg/s, P =0.004; dP/dtmin , -4046.9±776 versus -3146.3±642 mm Hg/s, P =0.046). The therapeutic potential of sCAR-Fc is limited, however, since postponed start of sCAR-Fc treatment either 3 or 7 days after infection could not attenuate myocardial injury., Conclusions: Early therapeutic employment of sCAR-Fc, initiated at the beginning of the primary viremia, inhibits the development of chronic CVB3-induced myocarditis and improves the cardiac function to a level equivalent to that of uninfected animals.- Published
- 2019
- Full Text
- View/download PDF
4. Management of Myocarditis-Related Cardiomyopathy in Adults.
- Author
-
Tschöpe C, Cooper LT, Torre-Amione G, and Van Linthout S
- Subjects
- Animals, Cardiomyopathies diagnosis, Cardiomyopathies etiology, Cardiomyopathies physiopathology, Disease Progression, Heart Failure diagnosis, Heart Failure etiology, Heart Failure physiopathology, Humans, Myocarditis diagnosis, Myocarditis etiology, Myocarditis physiopathology, Recovery of Function, Risk Factors, Treatment Outcome, Cardiomyopathies therapy, Heart Failure therapy, Myocarditis therapy
- Abstract
Myocarditis is generally a mild and self-limited consequence of systemic infection of cardiotropic viruses. However, patients can develop a temporary or permanent impairment of cardiac function including acute cardiomyopathy with hemodynamic compromise or severe arrhythmias. In this setting, specific causes of inflammation are associated with variable risks of death and transplantation. Recent translational studies suggest that treatments tailored to specific causes of myocarditis may impact clinical outcomes when added to guideline-directed medical care. This review summarizes recent advances in translational research that influence the utility of endomyocardial biopsy for the management of inflammatory cardiomyopathies. Emerging therapies for myocarditis based on these mechanistic hypotheses are entering clinical trials and may add to the benefits of established heart failure treatment.
- Published
- 2019
- Full Text
- View/download PDF
5. Pathogenic Role of the Damage-Associated Molecular Patterns S100A8 and S100A9 in Coxsackievirus B3-Induced Myocarditis.
- Author
-
Müller I, Vogl T, Pappritz K, Miteva K, Savvatis K, Rohde D, Most P, Lassner D, Pieske B, Kühl U, Van Linthout S, and Tschöpe C
- Subjects
- Adult, Animals, Calgranulin A deficiency, Calgranulin A genetics, Calgranulin B genetics, Case-Control Studies, Chemokine CXCL2 metabolism, Coxsackievirus Infections diagnosis, Coxsackievirus Infections genetics, Coxsackievirus Infections virology, Disease Models, Animal, Enterovirus B, Human genetics, Female, Fibrosis, Host-Pathogen Interactions, Humans, Macrophages metabolism, Macrophages virology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Middle Aged, Myocarditis diagnosis, Myocarditis genetics, Myocarditis virology, Myocytes, Cardiac pathology, Myocytes, Cardiac virology, Neutrophil Infiltration, Oxidative Stress, RAW 264.7 Cells, RNA Interference, RNA, Messenger genetics, Receptor for Advanced Glycation End Products metabolism, Signal Transduction, Transfection, Ventricular Function, Left, Calgranulin A metabolism, Calgranulin B metabolism, Coxsackievirus Infections metabolism, Enterovirus B, Human pathogenicity, Myocarditis metabolism, Myocytes, Cardiac metabolism
- Abstract
Background: The alarmins S100A8 and S100A9 are damage-associated molecular patterns, which play a pivotal role in cardiovascular diseases, inflammation, and viral infections. We aimed to investigate their role in Coxsackievirus B3 (CVB3)-induced myocarditis., Methods and Results: S100A8 and S100A9 mRNA expression was 13.0-fold ( P =0.012) and 5.1-fold ( P =0.038) higher in endomyocardial biopsies from patients with CVB3-positive myocarditis compared with controls, respectively. Elimination of CVB3 led to a downregulation of these alarmins. CVB3-infected mice developed an impaired left ventricular function and displayed an increased left ventricular S100A8 and S100A9 protein expression versus controls. In contrast, CVB3-infected S100A9 knockout mice, which are also a complete knockout for S100A8 on protein level, showed an improved left ventricular function, which was associated with a reduced cardiac inflammatory and oxidative response, and lower CVB3 copy number compared with wild-type CVB3 mice. Exogenous application of S100A8 to S100A9 knockout CVB3 mice induced a severe myocarditis similar to wild-type CVB3 mice. In CVB3-infected HL-1 cells, S100A8 and S100A9 enhanced oxidative stress and CVB3 copy number compared with unstimulated infected cells. In CVB3-infected RAW macrophages, both alarmins increased MIP-2 (macrophage inflammatory protein-2) chemokine expression, which was reduced in CVB3 S100A8 knockdown versus scrambled siRNA CVB3 cells., Conclusions: S100A8 and S100A9 aggravate CVB3-induced myocarditis and might serve as therapeutic targets in inflammatory cardiomyopathies., (© 2017 American Heart Association, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
6. NOD2 (Nucleotide-Binding Oligomerization Domain 2) Is a Major Pathogenic Mediator of Coxsackievirus B3-Induced Myocarditis.
- Author
-
Tschöpe C, Müller I, Xia Y, Savvatis K, Pappritz K, Pinkert S, Lassner D, Heimesaat MM, Spillmann F, Miteva K, Bereswill S, Schultheiss HP, Fechner H, Pieske B, Kühl U, and Van Linthout S
- Subjects
- Animals, Apoptosis, Apoptosis Regulatory Proteins metabolism, CARD Signaling Adaptor Proteins, Case-Control Studies, Caspase 1 metabolism, Cell Line, Coxsackievirus Infections immunology, Coxsackievirus Infections prevention & control, Coxsackievirus Infections virology, Disease Models, Animal, Enterovirus B, Human genetics, Enterovirus B, Human immunology, Genetic Predisposition to Disease, Host-Pathogen Interactions, Humans, Interleukin-1beta metabolism, Male, Mice, Inbred C57BL, Mice, Knockout, Myocarditis immunology, Myocarditis prevention & control, Myocarditis virology, Myocardium immunology, Myocardium pathology, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Nod2 Signaling Adaptor Protein deficiency, Nod2 Signaling Adaptor Protein genetics, Phenotype, RNA Interference, Signal Transduction, Transfection, Up-Regulation, Coxsackievirus Infections metabolism, Enterovirus B, Human metabolism, Myocarditis metabolism, Myocardium metabolism, Nod2 Signaling Adaptor Protein metabolism
- Abstract
Background: The cytoplasmatic pattern recognition receptor, NOD2 (nucleotide-binding oligomerization domain 2), belongs to the innate immune system and is among others responsible for the recognition of single-stranded RNA. With Coxsackievirus B3 (CVB3) being a single-stranded RNA virus, and the recent evidence that the NOD2 target, NLRP3 (NOD-like receptor family, pyrin domain containing 3) is of importance in the pathogenesis of CVB3-induced myocarditis, we aimed to unravel the role of NOD2 in CVB3-induced myocarditis., Methods and Results: Endomyocardial biopsy NOD2 mRNA expression was higher in CVB3-positive patients compared with patients with myocarditis but without evidence of persistent CVB3 infection. Left ventricular NOD2 mRNA expression was also induced in CVB3-induced myocarditis versus healthy control mice. NOD2 knockdown
(-/- ) mice were rescued from the detrimental CVB3-mediated effects as shown by a reduced cardiac inflammation (less cardiac infiltrates and suppression of proinflammatory cytokines), cardiac fibrosis, apoptosis, lower CAR (Coxsackievirus and adenovirus receptor) expression and CVB3 copy number, and an improved left ventricular function in NOD2-/- CVB3 mice compared with wild-type CVB3 mice. In agreement, NOD2-/- decreased the CVB3-induced inflammatory response, CVB3 copy number, and apoptosis in vitro. NOD2-/- was further associated with a reduction in CVB3-induced NLRP3 expression and activity as evidenced by lower ASC (apoptosis-associated speck-like protein containing a CARD) expression, caspase 1 activity, or IL-1β (interleukin-1β) protein expression under in vivo and in vitro CVB3 conditions., Conclusions: NOD2 is an important mediator in the viral uptake and inflammatory response during the pathogenesis of CVB3 myocarditis., (© 2017 American Heart Association, Inc.)- Published
- 2017
- Full Text
- View/download PDF
7. High-density lipoproteins reduce endothelial-to-mesenchymal transition.
- Author
-
Spillmann F, Miteva K, Pieske B, Tschöpe C, and Van Linthout S
- Subjects
- Actins metabolism, Antigens, CD metabolism, Cadherins metabolism, Cell Shape drug effects, Cells, Cultured, Collagen metabolism, Endothelial Cells metabolism, Endothelial Cells pathology, Fibrosis, Humans, Phenotype, Phosphatidylinositol 3-Kinase metabolism, RNA Interference, Scavenger Receptors, Class B genetics, Scavenger Receptors, Class B metabolism, Signal Transduction drug effects, Transfection, Transforming Growth Factor beta1 pharmacology, Endothelial Cells drug effects, Epithelial-Mesenchymal Transition drug effects, Lipoproteins, HDL pharmacology
- Abstract
Objective: Endothelial-to-mesenchymal transition is an inflammation-induced process by which endothelial cells can transdifferentiate into fibroblasts. Based on the endothelial-protective and antifibrotic effects of high-density lipoproteins (HDL), we aimed to investigate whether HDL can reduce endothelial-to-mesenchymal transition., Approach and Results: Therefore, human aortic endothelial cells were stimulated with the profibrotic factor transforming growth factor (TGF)-β1 in the presence or absence of HDL. Their impact on the transition of endothelial cells to mesenchymal-like cells was analyzed. Phase contrast microscopy demonstrated that HDL abrogated the TGF-β1-induced spindle-shape morphology in human aortic endothelial cells. Furthermore, HDL decreased the TGF-β1-mediated induction of α-smooth muscle actin expression and concomitant loss in endothelial cadherin expression, as shown by immunofluorescence staining and flow cytometry. In addition, HDL decreased the TGF-β1-induced collagen deposition in human aortic endothelial cells involving the scavenger receptor class B, type 1 and downstream phosphatidyl inositol-3-kinase following the findings that the HDL-mediated reduction was abrogated by scavenger receptor class B, type 1 siRNA knockdown and phosphatidyl inositol-3-kinase inhibition, respectively. The HDL-mediated reduction in endothelial-to-mesenchymal transition was associated with an induction of the inhibitory Smad, Smad 7., Conclusions: We provide the first in vitro evidence that the endothelial-protective and antifibrotic effects of HDL include the reduction in endothelial-to-mesenchymal transition., (© 2015 American Heart Association, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
8. Lack in treatment options for virus-induced inflammatory cardiomyopathy: can iPS-derived cardiomyocytes close the gap?
- Author
-
Van Linthout S, Tschöpe C, and Schultheiss HP
- Subjects
- Humans, Antiviral Agents therapeutic use, Enterovirus B, Human isolation & purification, Enterovirus Infections drug therapy, Models, Cardiovascular, Myocarditis drug therapy, Myocytes, Cardiac pathology, Pluripotent Stem Cells pathology
- Published
- 2014
- Full Text
- View/download PDF
9. Nerve growth factor promotes cardiac repair following myocardial infarction.
- Author
-
Meloni M, Caporali A, Graiani G, Lagrasta C, Katare R, Van Linthout S, Spillmann F, Campesi I, Madeddu P, Quaini F, and Emanueli C
- Subjects
- Aged, Animals, Apoptosis, Autopsy, Case-Control Studies, Cells, Cultured, Coronary Circulation, Disease Models, Animal, Endothelial Cells pathology, Female, Forkhead Box Protein O3, Forkhead Transcription Factors metabolism, Humans, Male, Mice, Middle Aged, Myocardial Infarction genetics, Myocardial Infarction metabolism, Myocardial Infarction pathology, Myocardial Infarction physiopathology, Myocardium pathology, Neovascularization, Physiologic, Nerve Growth Factor genetics, Proto-Oncogene Proteins c-akt metabolism, Proto-Oncogene Proteins c-kit metabolism, RNA, Messenger biosynthesis, Rats, Rats, Wistar, Receptor, trkA metabolism, Recombinant Proteins biosynthesis, Signal Transduction, Stem Cell Factor metabolism, Stem Cells metabolism, Stem Cells pathology, Time Factors, Transfection, Ventricular Function, Left, Endothelial Cells metabolism, Genetic Therapy, Myocardial Infarction therapy, Myocardium metabolism, Nerve Growth Factor biosynthesis, Regeneration genetics
- Abstract
Rationale: Nerve growth factor (NGF) promotes angiogenesis and cardiomyocyte survival, which are both desirable for postinfarction myocardial healing. Nonetheless, the NGF potential for cardiac repair has never been investigated., Objective: To define expression and localization of NGF and its high-affinity receptor TrkA (tropomyosin-related receptor A) in the human infarcted heart and to investigate the cardiac roles of both endogenous and engineered NGF using a mouse model of myocardial infarction (MI)., Methods and Results: Immunostaining for NGF and TrkA was performed on heart samples from humans deceased of MI or unrelated pathologies. To study the post-MI functions of endogenous NGF, a NGF-neutralizing antibody (Ab-NGF) or nonimmune IgG (control) was given to MI mice. To investigate the NGF therapeutic potential, human NGF gene or control (empty vector) was delivered to the murine periinfarct myocardium. Results indicate that NGF is present in the infarcted human heart. Both cardiomyocytes and endothelial cells (ECs) possess TrkA, which suggests NGF cardiovascular actions in humans. In MI mice, Ab-NGF abrogated native reparative angiogenesis, increased EC and cardiomyocyte apoptosis and worsened cardiac function. Conversely, NGF gene transfer ameliorated EC and cardiomyocyte survival, promoted neovascularization and improved myocardial blood flow and cardiac function. The prosurvival/proangiogenic Akt/Foxo pathway mediated the therapeutic benefits of NGF transfer. Moreover, NGF overexpression increased stem cell factor (the c-kit receptor ligand) expression, which translated in higher myocardial abundance of c-kit(pos) progenitor cells in NGF-engineered hearts., Conclusions: NGF elicits pleiotropic beneficial actions in the post-MI heart. NGF should be considered as a candidate for therapeutic cardiac regeneration.
- Published
- 2010
- Full Text
- View/download PDF
10. Human apolipoprotein A-I gene transfer reduces the development of experimental diabetic cardiomyopathy.
- Author
-
Van Linthout S, Spillmann F, Riad A, Trimpert C, Lievens J, Meloni M, Escher F, Filenberg E, Demir O, Li J, Shakibaei M, Schimke I, Staudt A, Felix SB, Schultheiss HP, De Geest B, and Tschöpe C
- Subjects
- Animals, Apolipoprotein A-I genetics, Cardiomyopathies therapy, Genetic Vectors genetics, Humans, Lipids blood, Lipoproteins, HDL, Rats, Rats, Sprague-Dawley, Streptozocin, Thiobarbituric Acid Reactive Substances analysis, Apolipoprotein A-I administration & dosage, Cardiomyopathies prevention & control, Diabetes Mellitus, Experimental complications, Diabetes Mellitus, Experimental therapy, Genetic Therapy methods
- Abstract
Background: The hallmarks of diabetic cardiomyopathy are cardiac oxidative stress, intramyocardial inflammation, cardiac fibrosis, and cardiac apoptosis. Given the antioxidative, antiinflammatory, and antiapoptotic potential of high-density lipoprotein (HDL), we evaluated the hypothesis that increased HDL via gene transfer (GT) with human apolipoprotein (apo) A-I, the principal apolipoprotein of HDL, may reduce the development of diabetic cardiomyopathy., Methods and Results: Intravenous GT with 3x10(12) particles/kg of the E1E3E4-deleted vector Ad.hapoA-I, expressing human apoA-I, or Ad.Null, containing no expression cassette, was performed 5 days after streptozotocin (STZ) injection. Six weeks after apoA-I GT, HDL cholesterol levels were increased by 1.6-fold (P<0.001) compared with diabetic controls injected with the Ad.Null vector (STZ-Ad.Null). ApoA-I GT and HDL improved LV contractility in vivo and cardiomyocyte contractility ex vivo, respectively. Moreover, apoA-I GT was associated with decreased cardiac oxidative stress and reduced intramyocardial inflammation. In addition, compared with STZ-Ad.Null rats, cardiac fibrosis and glycogen accumulation were reduced by 1.7-fold and 3.1-fold, respectively (P<0.05). Caspase 3/7 activity was decreased 1.2-fold (P<0.05), and the ratio of Bcl-2 to Bax was upregulated 1.9-fold (P<0.005), translating to 2.1-fold (P<0.05) reduced total number of cardiomyocytes with apoptotic characteristics and 3.0-fold (P<0.005) reduced damaged endothelial cells compared with STZ-Ad.Null rats. HDL supplementation ex vivo reduced hyperglycemia-induced cardiomyocyte apoptosis by 3.4-fold (P<0.005). The apoA-I GT-mediated protection was associated with a 1.6-, 1.6-, and 2.4-fold induction of diabetes-downregulated phospho to Akt, endothelial nitric oxide synthase, and glycogen synthase kinase ratio, respectively (P<0.005)., Conclusions: ApoA-I GT reduced the development of streptozotocin-induced diabetic cardiomyopathy.
- Published
- 2008
- Full Text
- View/download PDF
11. Human ApoA-I transfer attenuates transplant arteriosclerosis via enhanced incorporation of bone marrow-derived endothelial progenitor cells.
- Author
-
Feng Y, Jacobs F, Van Craeyveld E, Brunaud C, Snoeys J, Tjwa M, Van Linthout S, and De Geest B
- Subjects
- Animals, Cholesterol, HDL metabolism, Coronary Artery Disease etiology, Disease Models, Animal, Female, Humans, Hypercholesterolemia physiopathology, Mice, Mice, Transgenic, Stem Cells physiology, Tunica Intima physiopathology, Apolipoprotein A-I genetics, Apolipoprotein A-I physiology, Coronary Artery Disease physiopathology, Endothelial Cells physiology, Heart Transplantation adverse effects, Heart Transplantation physiology
- Abstract
Objective: Transplant arteriosclerosis is the leading cause of graft failure and death in patients with heart transplantation. Endothelial progenitor cells (EPCs) contribute to endothelial regeneration in allografts. We investigated whether increased HDL cholesterol induced by adenoviral human apoA-I (AdA-I) transfer increases number and function of EPCs, promotes incorporation of EPCs in Balb/c allografts transplanted paratopically in C57BL/6 ApoE-/- mice, and attenuates transplant arteriosclerosis., Methods and Results: EPC number in ApoE-/- mice was increased after AdA-I transfer as evidenced by 1.5-fold (P<0.01) higher Flk-1 Sca-1-positive cells and 1.4-fold (P<0.01) higher DiI-acLDL isolectin-positive spleen cells. In addition, HDL enhanced EPC function in vitro. Incorporation of bone marrow-derived EPCs was 5.8-fold (P<0.01) higher at day 21 after transplantation in AdA-I-treated apoE-/- mice compared with control mice. Enhanced endothelial regeneration in AdA-I-treated apoE-/- mice as evidenced by a 2.6-fold (P<0.01) increase of CD31-positive endothelial cells resulted in a 1.4-fold (P=0.059) reduction of neointima and a 3.9-fold (P<0.01) increase of luminal area., Conclusions: Human apoA-I transfer increases the number of circulating EPCs, enhances their incorporation into allografts, promotes endothelial regeneration, and attenuates neointima formation in a murine model of transplant arteriosclerosis.
- Published
- 2008
- Full Text
- View/download PDF
12. Cardioprotective mechanisms of the kallikrein-kinin system in diabetic cardiopathy.
- Author
-
Spillmann F, Van Linthout S, Schultheiss HP, and Tschöpe C
- Subjects
- Animals, Calcium metabolism, Endothelium, Vascular pathology, Extracellular Matrix physiology, Heart Diseases etiology, Humans, Rats, Diabetes Complications physiopathology, Glucose metabolism, Heart Diseases physiopathology, Kallikrein-Kinin System physiology
- Abstract
Purpose of Review: Multiple pathogenic mechanisms contribute to the development of diabetic cardiopathy, including intramyocardial inflammation, cardiac fibrosis, abnormal intracellular Ca handling, microangiopathy and endothelial dysfunction. Moreover, the cardiac kallikrein-kinin system is thought to be altered under diabetic conditions and an improvement of this peptide system, e.g. by gene therapeutic approaches, has also been associated with an amelioration of the diabetic heart. In this review, we will discuss the hypothesis that the stimulation of the kallikrein-kinin system could be a promising target for the treatment of diabetic cardiopathy., Recent Findings: The kallikrein-kinin system has cardioprotective properties, which may be particularly important under diabetic conditions. For example, its potential for endothelium-dependent vasodilation, and for improvement of glucose transport and utilization, make bradykinin an important mediator for reducing the consequences of diabetes-related oxidative stress on both the myocardium and vessels., Summary: The different synergistic cardioprotective effects of the kallikrein-kinin system in the diabetic heart suggest that the stimulation of the kallikrein-kinin system might open new avenues for the treatment of diabetic cardiopathy.
- Published
- 2006
- Full Text
- View/download PDF
13. Nitric oxide-releasing aspirin derivative, NCX 4016, promotes reparative angiogenesis and prevents apoptosis and oxidative stress in a mouse model of peripheral ischemia.
- Author
-
Emanueli C, Van Linthout S, Salis MB, Monopoli A, Del Soldato P, Ongini E, and Madeddu P
- Subjects
- Animals, Apoptosis physiology, Aspirin antagonists & inhibitors, Down-Regulation physiology, Gene Expression Regulation drug effects, Male, Mice, Mice, Inbred Strains, Muscle, Skeletal blood supply, Muscle, Skeletal chemistry, Muscle, Skeletal drug effects, Muscle, Skeletal pathology, Nitrites blood, Oxidative Stress physiology, Reperfusion Injury blood, Reperfusion Injury pathology, Vascular Endothelial Growth Factor A genetics, Apoptosis drug effects, Aspirin analogs & derivatives, Aspirin metabolism, Aspirin pharmacology, Disease Models, Animal, Neovascularization, Physiologic drug effects, Nitric Oxide metabolism, Oxidative Stress drug effects, Reperfusion Injury metabolism
- Abstract
Background: Recently, nitric oxide (NO) donors have been developed that mimic the physiological intracellular release of NO. We evaluated whether one of these new compounds, consisting of aspirin coupled to an NO-releasing moiety (NCX 4016), would protect limbs from supervening arterial occlusion., Methods and Results: Mice were assigned to receive regular chow or chow containing NCX 4016 or aspirin (both at 300 mumol/kg body weight, daily) throughout the 3-week experimental period. One week after randomization, they underwent surgical excision of the left femoral artery. Limb blood flow recovery (laser Doppler flowmetry) was accelerated by NCX 4016 as compared with aspirin or vehicle (P<0.05). In controls, histological analysis revealed a 35% increase in the capillary density of ischemic muscles compared with contralateral ones, indicative of spontaneous angiogenesis. Neovascularization was enhanced by NCX 4016 (91%; P<0.05 versus vehicle), but not by aspirin (51%; P=NS versus vehicle). Furthermore, NCX 4016 reduced endothelial cell (EC) apoptosis (4.3+/-1.0 versus 8.7+/-2.0 in aspirin and 12.6+/-3.3 ECs/1000 cap in vehicle; P<0.05 for either comparison) as well as caspase-3 mRNA levels in ischemic muscles ([caspase-3/GAPDH]*100 = 0.09+/-0.04 versus 2.30+/-0.44 in aspirin and 2.30+/-0.32 in vehicle; P<0.01 for either comparison). Nitrite levels and the ratio of reduced to oxidized glutathione were selectively increased in ischemic muscles by NCX 4016. Vascular endothelial growth factor-A expression was reduced by aspirin, with this effect being blunted by NCX 4016., Conclusions: Pretreatment with the new oral NO-releasing aspirin derivative stimulates reparative angiogenesis and prevents apoptosis and oxidative stress, thereby alleviating the consequences of supervening arterial occlusion.
- Published
- 2004
- Full Text
- View/download PDF
14. Akt/protein kinase B and endothelial nitric oxide synthase mediate muscular neovascularization induced by tissue kallikrein gene transfer.
- Author
-
Emanueli C, Salis MB, Van Linthout S, Meloni M, Desortes E, Silvestre JS, Clergue M, Figueroa CD, Gadau S, Condorelli G, and Madeddu P
- Subjects
- Adenoviridae genetics, Animals, Blotting, Western, Cell Division, Endothelial Cells cytology, Endothelium, Vascular cytology, Enzyme Induction drug effects, Exudates and Transudates, Genes, Dominant, Genetic Vectors pharmacology, Humans, Mice, Microcirculation, Muscle, Skeletal metabolism, NG-Nitroarginine Methyl Ester pharmacology, Neovascularization, Physiologic genetics, Nitric Oxide Synthase biosynthesis, Nitric Oxide Synthase genetics, Nitric Oxide Synthase Type II, Nitric Oxide Synthase Type III, Phosphorylation, Phosphoserine analysis, Polymerase Chain Reaction, Protein Processing, Post-Translational, Proto-Oncogene Proteins c-akt, RNA, Messenger biosynthesis, RNA, Messenger genetics, Tissue Kallikreins genetics, Transduction, Genetic, Vascular Endothelial Growth Factor A antagonists & inhibitors, Vascular Endothelial Growth Factor Receptor-2 analysis, Muscle, Skeletal blood supply, Neovascularization, Physiologic drug effects, Nitric Oxide Synthase physiology, Protein Serine-Threonine Kinases physiology, Proto-Oncogene Proteins physiology, Tissue Kallikreins physiology
- Abstract
Background: Angiogenesis gene therapy with human tissue kallikrein (hTK) has shown promise for ischemic disease. The present study was undertaken to (1) assess an optimal gene transfer modality, (2) clarify hTK angiogenic pathways, and (3) discount possible side effects., Methods and Results: The hTK gene was transferred to murine adductors by increasing doses of an adenovirus (Ad.hTK). Heterologous protein production was evaluated by ELISA and immunohistochemistry. Structural and functional characteristics of hTK-induced neovascularization were assessed. Muscular endothelial nitric oxide synthase (eNOS) and vascular endothelial growth factor (VEGF)-A mRNA and protein content were evaluated by real-time polymerase chain reaction and Western blotting. The ability of hTK to phosphorylate-activate Akt/protein kinase B (Akt-B) and VEGF receptor 2 (VEGF-R2) was also determined. Implication of the aforementioned mechanisms in Ad.hTK-induced neovascularization was challenged by blocking Akt-B with a dominant-negative Akt construct; NOS with N(G)-nitro-L-arginine methyl ester; and VEGF-A with neutralizing antibody, VEGF-R2 antagonist, or Ad carrying soluble VEGF-R1 gene. We found that 10(7) PFU Ad.hTK led to peak increases in capillary and arteriole density. Newly developed arterioles persisted for up to 8 weeks. Ad.hTK did not change microvascular permeability. Ad.hTK upregulated eNOS mRNA and protein and activated Akt-B through Ser-473 phosphorylation. Inhibitory studies documented that these biochemical events were instrumental to Ad.hTK-induced neovascularization. In contrast, Ad.hTK neither affected VEGF-A and VEGF-R2 levels nor increased VEGF-R2 phosphorylation. Consistently, Ad.hTK-induced neovascularization was not disturbed by any of the different approaches used to block VEGF-A., Conclusions: Our findings provide new information on the pathway involved in hTK-induced neoangiogenesis and represent an advancement toward clinical applications with Ad.hTK.
- Published
- 2004
- Full Text
- View/download PDF
15. The Arg123-Tyr166 central domain of human ApoAI is critical for lecithin:cholesterol acyltransferase-induced hyperalphalipoproteinemia and HDL remodeling in transgenic mice.
- Author
-
Holvoet P, De Geest B, Van Linthout S, Lox M, Danloy S, Raes K, and Collen D
- Subjects
- Amino Acid Sequence genetics, Animals, Apolipoprotein A-I blood, Apolipoproteins blood, Chimera, Cholesterol, HDL blood, Gene Transfer Techniques, Genotype, Humans, Lipoproteins blood, Mice, Mice, Transgenic genetics, Apolipoprotein A-I genetics, Hyperlipoproteinemias chemically induced, Hyperlipoproteinemias genetics, Lipoproteins, HDL blood, Phosphatidylcholine-Sterol O-Acyltransferase blood, Phosphatidylcholine-Sterol O-Acyltransferase genetics
- Abstract
High density lipoprotein (HDL) metabolism and lecithin:cholesterol acyltransferase (LCAT)-induced HDL remodeling were investigated in transgenic mice expressing human apolipoprotein (apo) AI or an apoAI/apoAII chimera in which the Arg123-Tyr166 domain of apoAI was substituted with the Ser12-Ala75 domain of apoAII. Expression of apoAI and of the apoAI/apoAII chimera resulted in a respective 3. 5-fold and 2.9-fold increase of HDL cholesterol. Human LCAT gene transfer into apoAI-transgenic mice resulted in a 5.1-fold increase of endogenous LCAT activity. This increase was associated with a 2. 4-fold increase of the cholesterol ester-to-free cholesterol ratio of HDL, a shift from HDL(3) to HDL(2), and a 2.4-fold increase of HDL cholesterol levels. Agarose gel electrophoresis revealed that human LCAT gene transfer into human apoAI-transgenic mice resulted in an increase of pre-beta-HDL and of pre-alpha-HDL. In contrast, human LCAT gene transfer did not affect cholesterol levels and HDL distribution profile in mice expressing the apoAI/apoAII chimera. Mouse LCAT did not "see" a difference between wild-type and mutant human apoAI, whereas human LCAT did, thus localizing the species-specific interaction in the central domain of apoAI. In conclusion, the Arg123-Tyr166 central domain of apoAI is not critical for in vivo lipoprotein association. It is, however, critical for LCAT-induced hyperalphalipoproteinemia and HDL remodeling independent of the lipid-binding properties of apoAI.
- Published
- 2000
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.