Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Cardiovasc Pharmacol ; 67(1): 47-56, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26322923

RESUMO

AIMS: We recently reported that immunosuppression with FTY720 improves cardiac function and extends longevity in Hypomorphic ApoE mice deficient in scavenger receptor Type-BI expression, also known as the HypoE/SR-BI(­/­) mouse model of diet-induced coronary atherosclerosis and myocardial infarction (MI). In this study, we tested the impact of FTY720 on cardiac dysfunction in HypoE/SR-BI(­/­) mice that survive MI and subsequently develop chronic heart failure. METHODS/RESULTS: HypoE/SR-BI(­/­) mice were bred to Mx1-Cre transgenic mice, and offspring were fed a high-fat diet (HFD) for 3.5 weeks to provoke hyperlipidemia, coronary atherosclerosis, and recurrent MIs. In contrast to our previous study, hyperlipidemia was rapidly reversed by inducible Cre-mediated gene repair of the HypoE allele and switching mice to a normal chow diet. Mice that survived the period of HFD were subsequently given oral FTY720 in drinking water or not, and left ventricular (LV) function was monitored using serial echocardiography for up to 15 weeks. In untreated mice, LV performance progressively deteriorated. Although FTY720 treatment did not initially prevent a decline of heart function among mice 6 weeks after Cre-mediated gene repair, it almost completely restored normal LV function in these mice by 15 weeks. Reversal of heart failure did not result from reduced atherosclerosis as the burden of aortic and coronary atherosclerosis actually increased to similar levels in both groups of mice. Rather, FTY720 caused systemic immunosuppression as assessed by reduced numbers of circulating T and B lymphocytes. In contrast, FTY720 did not enhance the loss of T cells or macrophages that accumulated in the heart during the HFD feeding period, but it did enhance the loss of B cells soon after plasma lipid lowering. Moreover, FTY720 potently reduced the expression of matrix metalloproteinase-2 and genes involved in innate immunity-associated inflammation in the heart. CONCLUSIONS: Our data demonstrate that immunosuppression with FTY720 prevents postinfarction myocardial remodeling and chronic heart failure.


Assuntos
Apolipoproteínas E/deficiência , Doença da Artéria Coronariana/tratamento farmacológico , Cloridrato de Fingolimode/uso terapêutico , Imunossupressores/uso terapêutico , Infarto do Miocárdio/tratamento farmacológico , Receptores Depuradores Classe B/biossíntese , Animais , Doença da Artéria Coronariana/metabolismo , Doença da Artéria Coronariana/mortalidade , Dieta Hiperlipídica/efeitos adversos , Regulação da Expressão Gênica , Camundongos , Camundongos Transgênicos , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/mortalidade , Taxa de Sobrevida/tendências
2.
J Cardiovasc Pharmacol ; 63(5): 406-11, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24805144

RESUMO

Cardiovascular disease is the leading cause of death in Western countries. A major limitation of current treatments is the inability to efficiently repair or replace dead myocardium. Recently, stem cell-based therapies have been explored as an avenue to circumvent current therapeutic limitations. Overall, these therapies seem to result in small improvements in the contractile function of the heart. The exact mechanism(s) of action that underlie these improvements remain unknown, and it is believed that paracrine effects play a significant role. Previously, we had reported that an extract derived from bone marrow cells, in the absence of any live cell, contained cardioprotective soluble factors. In this study, we identify IL-15 as a putative cardioprotectant within the bone marrow cells paracrine profile. Using an in vitro culture system, we assessed the ability of IL-15 to protect cardiomyocytes under hypoxic conditions. For the first time, we have identified IL-15 receptors on the surface of cardiomyocytes and delineated the signaling system by which hypoxic cardiomyocytes may be protected from cellular death and rescued from oxidative stress with IL-15 treatment.


Assuntos
Interleucina-15/fisiologia , Miócitos Cardíacos/fisiologia , Transdução de Sinais/fisiologia , Animais , Hipóxia Celular , Sobrevivência Celular/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Receptores de Interleucina-15/efeitos dos fármacos , Receptores de Interleucina-15/metabolismo , Fator de Transcrição STAT3/fisiologia , Transdução de Sinais/efeitos dos fármacos
3.
J Cardiovasc Pharmacol ; 63(2): 132-143, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24508946

RESUMO

FTY720, an analogue of sphingosine-1-phosphate, is cardioprotective during acute injury. Whether long-term FTY720 affords cardioprotection is unknown. Here, we report the effects of oral FTY720 on ischemia/reperfusion injury and in hypomorphic apoE mice deficient in SR-BI receptor expression (ApoeR61(h/h)/SRB1(-/- mice), a model of diet-induced coronary atherosclerosis and heart failure. We added FTY720 (0.3 mg·kg(-1)·d(-1)) to the drinking water of C57BL/6J mice. After ex vivo cardiac ischemia/reperfusion injury, these mice had significantly improved left ventricular (LV) developed pressure and reduced infarct size compared with controls. Subsequently, ApoeR61(h/h)/SRB1(-/-) mice fed a high-fat diet for 4 weeks were treated or not with oral FTY720 (0.05 mg·kg(-1)·d(-1)). This sharply reduced mortality (P < 0.02) and resulted in better LV function and less LV remodeling compared with controls without reducing hypercholesterolemia and atherosclerosis. Oral FTY720 reduced the number of blood lymphocytes and increased the percentage of CD4+Foxp3+ regulatory T cells (Tregs) in the circulation, spleen, and lymph nodes. FTY720-treated mice exhibited increased TGF-ß and reduced IFN-γ expression in the heart. Also, CD4 expression was increased and strongly correlated with molecules involved in natural Treg activity, such as TGF-ß and GITR. Our data suggest that long-term FTY720 treatment enhances LV function and increases longevity in mice with heart failure. These benefits resulted not from atheroprotection but from systemic immunosuppression and a moderate reduction of inflammation in the heart.


Assuntos
Apolipoproteínas E/genética , Doença da Artéria Coronariana/tratamento farmacológico , Infarto do Miocárdio/tratamento farmacológico , Propilenoglicóis/farmacologia , Esfingosina/análogos & derivados , Animais , Cardiotônicos/administração & dosagem , Cardiotônicos/farmacologia , Doença da Artéria Coronariana/fisiopatologia , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Cloridrato de Fingolimode , Imunossupressores/administração & dosagem , Imunossupressores/farmacologia , Inflamação/tratamento farmacológico , Inflamação/etiologia , Inflamação/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infarto do Miocárdio/fisiopatologia , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Propilenoglicóis/administração & dosagem , Esfingosina/administração & dosagem , Esfingosina/farmacologia , Taxa de Sobrevida , Linfócitos T Reguladores/metabolismo , Fatores de Tempo , Fator de Crescimento Transformador beta/metabolismo , Função Ventricular Esquerda/efeitos dos fármacos
4.
Am J Physiol Heart Circ Physiol ; 300(5): H1753-61, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21335477

RESUMO

Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid that promotes cardiomyocyte survival and contributes to ischemic preconditioning. S1P lyase (SPL) is a stress-activated enzyme responsible for irreversible S1P catabolism. We hypothesized that SPL contributes to oxidative stress by depleting S1P pools available for cardioprotective signaling. Accordingly, we evaluated SPL inhibition as a strategy for reducing cardiac ischemia-reperfusion (I/R) injury. We measured SPL expression and enzyme activity in murine hearts. Basal SPL activity was low in wild-type cardiac tissue but was activated in response to 50 min of ischemia (n = 5, P < 0.01). Hearts of heterozygous SPL knockout mice exhibited reduced SPL activity, elevated S1P levels, smaller infarct size, and increased functional recovery after I/R compared with littermate controls (n = 5, P < 0.01). The small molecule tetrahydroxybutylimidazole (THI) is a Federal Drug Administration-approved food additive that inhibits SPL. When given overnight at 25 mg/l in drinking water, THI raised S1P levels and reduced SPL activity (n = 5, P < 0.01). THI reduced infarct size and enhanced hemodynamic recovery in response to 50 min of ischemia and to 40 min of reperfusion in ex vivo hearts (n = 7, P < .01). These data correlated with an increase in MAP kinase-interacting serine/threonine kinase 1, eukaryotic translation initiation factor 4E, and ribosomal protein S6 phosphorylation levels after I/R, suggesting that SPL inhibition enhances protein translation. Pretreatment with an S1P1 and S1P3 receptor antagonist partially reversed the effects of THI. These results reveal, for the first time, that SPL is an ischemia-induced enzyme that can be targeted as a novel strategy for preventing cardiac I/R injury.


Assuntos
Aldeído Liases/antagonistas & inibidores , Aldeído Liases/fisiologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Aldeído Liases/genética , Animais , Inibidores Enzimáticos/uso terapêutico , Lisofosfolipídeos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Mutação/genética , Miocárdio/metabolismo , Estresse Oxidativo/fisiologia , Esfingosina/análogos & derivados , Esfingosina/metabolismo
5.
Am J Physiol Heart Circ Physiol ; 298(3): H1022-8, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20061542

RESUMO

The lipid mediator sphingosine 1-phosphate (S1P) confers survival benefits in cardiomyocytes and isolated hearts subjected to oxidative stress. High-density lipoprotein (HDL) is a major carrier of S1P in the serum, but whether HDL-associated S1P directly mediates survival in a preparation composed exclusively of cardiomyocytes has not been demonstrated. Accordingly, we tested the hypothesis that signal activation and survival during simulated ischemia-reperfusion injury in response to HDL require lipoprotein-associated S1P. As a model, we used adult mouse cardiomyocytes subjected to hypoxia-reoxygenation. Cells were treated or not with autologous mouse HDL, which significantly increased myocyte viability as measured by trypan blue exclusion. This survival effect was abrogated by the S1P(1) and SIP(3) receptor antagonist VPC 23019. The selective S1P(3) antagonist CAY10444, the G(i) antagonist pertussis toxin, the MEK (MAPK/ERK) kinase inhibitor PD-98059, and the phosphoinositide-3 kinase inhibitor wortmannin also inhibited the prosurvival effect of HDL. We observed that HDL activated both Akt (protein kinase B) and the MEK1/2-ERK1/2 pathway and also stimulated phosphorylation of glycogen synthase kinase-3beta. ERK1/2 activation was through an S1P(1) subtype receptor-G(i) protein-dependent pathway, whereas the activation of Akt was inhibited by CAY10444, indicating mediation by S1P(3) subtype receptors. We conclude that HDL, via its cargo of S1P, can directly protect cardiomyocytes against simulated oxidative injury in the absence of vascular effects and that prosurvival signal activation is dependent on both S1P(1) and S1P(3) subtype receptors.


Assuntos
Lipoproteínas HDL/farmacologia , Lisofosfolipídeos/fisiologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miócitos Cardíacos/efeitos dos fármacos , Esfingosina/análogos & derivados , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , MAP Quinase Quinase 1/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 3 Ativada por Mitógeno/fisiologia , Miócitos Cardíacos/patologia , Estresse Oxidativo/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais/fisiologia , Esfingosina/fisiologia
6.
J Cardiovasc Pharmacol ; 56(6): 659-68, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20881608

RESUMO

There is an increase in reactive oxygen and nitrogen species in cardiomyocytes during myocardial ischemia/reperfusion injury. This leads to oxidative DNA damage and activation of nuclear repair enzymes such as poly(ADP-ribose) polymerase-1 (PARP-1). PARP-1 activation promotes DNA repair under normal conditions. However, excessive activation of PARP-1 leads to cell death. We report that PARP-1 enzymatic activity is directly inhibited by minocycline, and we propose that one mechanism of minocycline cardioprotection is the result of PARP-1 inhibition. Using cultured adult rat cardiac myocytes, we evaluated the mechanism of minocycline protection in which PARP-1 activation was induced by simulated ischemia/reperfusion injury using oxygen­glucose deprivation.We found an increase in reactive oxygen species production, PARP-1 activation, and PARP-1-mediated cell death after simulated ischemia/reperfusion. Cell death was significantly reduced by the PARP inhibitors 3, 4-dihydro-5-[4-(1-piperidinyl)butoxy]-1(2H)-isoquinolinone (10 µM) and PJ-34 (500 nM) or by minocycline (500 nM). Cellular NAD(+) depletion and poly(ADP-ribose) formation, which are biochemical markers of PARP-1 activation, were also blocked by minocycline. Finally, simulated ischemia/reperfusion led to induction of the mitochondrial permeability transition, which was prevented by minocycline. Therefore, we propose that the protective effect of minocycline on cardiac myocyte survival is the result of inhibition of PARP-1 activity.


Assuntos
Cardiotônicos/farmacologia , Minociclina/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Inibidores de Poli(ADP-Ribose) Polimerases , Traumatismo por Reperfusão/prevenção & controle , Animais , Cardiotônicos/uso terapêutico , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Masculino , Minociclina/uso terapêutico , Miócitos Cardíacos/enzimologia , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/metabolismo , Ratos , Traumatismo por Reperfusão/enzimologia
7.
J Cardiovasc Pharmacol ; 55(3): 219-26, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20375713

RESUMO

The DNA-damaging agent N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) causes cardiomyocyte death as a result of energy loss from excessive activation of poly-(ADP) ribose polymerase-1 (PARP-1) resulting in depletion of its substrates nicotinamide adenine dinucleotide (NAD) and ATP. Previously we showed that the chemotherapeutic agent vincristine (VCR) is cardioprotective. Here we tested the hypothesis that VCR inhibits MNNG-induced PARP activation. Adult mouse cardiomyocytes were incubated with 100 micromol/L MNNG with or without concurrent VCR (20 micromol/L) for 2 to 4 hours. Cardiomyocyte survival was measured using the trypan blue exclusion assay. Western blots were used to measure signaling responses. MNNG-induced cardiomyocyte damage was time- and concentration-dependent. MNNG activated PARP-1 and depleted NAD and ATP. VCR completely protected cardiomyocytes from MNNG-induced cell damage and maintained intracellular levels of NAD and ATP. VCR increased phosphorylation of the prosurvival signals Akt, GSK-3beta, Erk1/2, and p70S6 kinase. VCR delayed PARP activation as evidenced by Western blot and by immunofluorescence staining of poly (ADP)-ribose, but without directly inhibiting PARP-1 itself. Known PARP-1 inhibitors also protected cardiomyocytes from MNNG-induced death. Repletion of ATP, NAD, pyruvate, and glutamine had effects similar to PARP-1 inhibitors. We conclude that VCR protects cardiomyocytes from MNNG toxicity by regulating PARP-1 activation, intracellular energy metabolism, and prosurvival signaling.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Poli(ADP-Ribose) Polimerases/efeitos dos fármacos , Vincristina/farmacologia , Animais , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Metabolismo Energético/efeitos dos fármacos , Masculino , Metilnitronitrosoguanidina/administração & dosagem , Metilnitronitrosoguanidina/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/metabolismo , Fosforilação/efeitos dos fármacos , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
8.
Cardiology ; 115(2): 155-62, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20016174

RESUMO

Established doxorubicin cardiomyopathy is a lethal disease. When congestive heart failure develops, mortality is approximately 50%. Extensive research has been done to understand the mechanism and pathophysiology of doxorubicin cardiomyopathy, and considerable knowledge and experience has been gained. Unfortunately, no effective treatment for established doxorubicin cardiomyopathy is presently available. Extensive research has been done and is being done to discover preventive treatments. However an effective and clinically applicable preventive treatment is yet to be discovered.


Assuntos
Antibióticos Antineoplásicos/efeitos adversos , Cardiomiopatias/induzido quimicamente , Doxorrubicina/efeitos adversos , Cardiomiopatias/diagnóstico , Cardiomiopatias/patologia , Cardiomiopatias/terapia , Humanos , Miocárdio/patologia
9.
Am J Physiol Heart Circ Physiol ; 297(4): H1429-35, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19648253

RESUMO

Exogenous sphingosine 1-phosphate (S1P) is an effective cardioprotectant against ischemic injury. We have investigated the hypothesis that S1P is also an important endogenous cardioprotectant released during both ischemic preconditioning (IPC) and ischemic postconditioning (IPOST). IPC of ex vivo rat hearts was instituted by two cycles of 3 min ischemia-5 min reperfusion prior to 40 min of index ischemia and then 40 min of reperfusion. IPC resulted in 70% recovery of left ventricular developed pressure (LVDP) upon reperfusion and a small infarct size (10%). VPC23019 (VPC), a specific antagonist of S1P(1 and 3) G protein-coupled receptors (GPCRs), when present during preconditioning blocked protection afforded by two cycles of IPC. VPC also blocked preconditioning of isolated rat cardiac myocytes subjected to hypoxia-reoxygenation injury. Increased release of S1P from myocytes in response to IPC was also demonstrated. These data indicate that S1P is released from myocytes in response to IPC and protects by binding to S1P GPCRs. In the ex vivo heart, if a third cycle of IPC was added to increase release of endogenous mediators, then the need for any individual mediator (e.g., S1P) was diminished and VPC had little effect. The adenosine antagonist 8-(p-sulfophenyl)-theophylline (8-SPT) likewise inhibited protection by two cycles but not three cycles of IPC, but VPC plus 8-SPT inhibited protection by three cycles of IPC. Similar to IPC, IPOST induced by four postindex ischemia cycles of 15 s reperfusion-15 s ischemia resulted in 66% recovery of LVDP and a 7% infarct size. When VPC was present during postconditioning and reperfusion, LVDP only recovered by 26% and the infarct size increased to 27%. Adding an additional cycle of IPOST reduced the inhibitory effect of VPC and 8-SPT individually, but not their combined effect. These studies reveal that S1P is an important mediator of both IPC and IPOST that is released along with adenosine during each cycle of IPC or IPOST.


Assuntos
Precondicionamento Isquêmico Miocárdico , Lisofosfolipídeos/metabolismo , Infarto do Miocárdio/prevenção & controle , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miocárdio/metabolismo , Esfingosina/análogos & derivados , Função Ventricular Esquerda , Adenosina/metabolismo , Animais , Animais Recém-Nascidos , Células Cultivadas , Técnicas In Vitro , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Perfusão , Antagonistas de Receptores Purinérgicos P1 , Ratos , Ratos Sprague-Dawley , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Receptores de Lisoesfingolipídeo/metabolismo , Receptores Purinérgicos P1/metabolismo , Recuperação de Função Fisiológica , Transdução de Sinais , Esfingosina/metabolismo , Teofilina/análogos & derivados , Teofilina/farmacologia , Fatores de Tempo , Função Ventricular Esquerda/efeitos dos fármacos , Pressão Ventricular
10.
J Cardiovasc Pharmacol ; 53(6): 486-94, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19433984

RESUMO

We examined the ability of sphingosine-1-phosphate (S1P) to desensitize extracellular signal-related kinase (ERK), a mitogen-activated protein kinase linked to antiapoptotic responses in the heart. In isolated adult mouse cardiomyocytes, S1P (10 nM-5 microM) induced ERK phosphorylation in a time- and dose-dependent manner. S1P stimulation of ERK was completely inhibited by an S1P1/3 subtype receptor antagonist (VPC23019), by a Gi protein inhibitor (pertussis toxin) and by a mitogen-activated protein kinase/ERK kinase inhibitor (PD98059). A selective S1P3 receptor antagonist (CAY10444) had no effect on S1P-induced ERK activation. The selective S1P1 agonist SEW2871 also induced ERK phosphorylation. Activation of ERK by restimulation with 100 nM S1P was suppressed after 1 hour of preincubation with 100 nM S1P but recovered fully the next day, suggesting receptor recycling. Similar results were obtained in protein kinase C epsilon-null cardiomyocytes. Treatment with the nonselective S1P receptor agonist FTY720 for 1 hour also reduced phospho-ERK expression in response to subsequent S1P stimulation. In contrast to S1P, some desensitization to FTY720 persisted after overnight exposure. Cell death induced by hypoxia/reoxygenation was reduced by pretreatment with exogenous S1P. This enhanced survival was abrogated by pretreatment with PD98059, VPC23019, or pertussis toxin. Thus, exogenous S1P induces rapid and reversible S1P1-mediated ERK phosphorylation. S1P-induced adult mouse cardiomyocyte survival requires ERK activation mediated via an S1P1-Gi pathway.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Miócitos Cardíacos/metabolismo , Receptores de Lisoesfingolipídeo/fisiologia , Animais , Morte Celular/efeitos dos fármacos , Hipóxia Celular , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ativação Enzimática , Técnicas In Vitro , Lisofosfolipídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/efeitos dos fármacos , Fosforilação , Proteína Quinase C/genética , Receptores de Lisoesfingolipídeo/agonistas , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Transdução de Sinais , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Tiazolidinas/farmacologia
11.
Biochem Biophys Res Commun ; 373(4): 555-60, 2008 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-18590705

RESUMO

Our aim was to test the hypothesis that the vinca alkaloid vincristine could prevent doxorubicin-induced cardiomyocyte death and to identify the mechanisms involved. Adult mouse cardiac myocytes were incubated for 24 h with doxorubicin, with and without concurrent vincristine. Trypan blue exclusion showed that 50-60% of myocytes treated with doxorubicin alone survived. Concurrent vincristine treatment increased survival to 85%. Treatment with doxorubicin+vincristine activated the prosurvival signal Akt and diminished cytochrome C release. The PI3K/Akt inhibitor LY294002 and the MEK/ERK inhibitor PD98059 augmented doxorubicin cardiotoxicity and attenuated salvage during concurrent vincristine treatment, indicating that the mechanism of vincristine cardioprotection involves activation of specific survival signals. Vincristine retarded the onset of apoptosis in association with a delay in poly(ADP) ribose polymerase activation. Vincristine also exhibited greater protection than the antioxidant MPG. These novel findings may have clinical implications for the prevention of doxorubicin cardiomyopathy.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Citoproteção , Doxorrubicina/toxicidade , Miócitos Cardíacos/efeitos dos fármacos , Vincristina/farmacologia , Animais , Apoptose/efeitos dos fármacos , Cromonas/farmacologia , Citocromos c/metabolismo , Flavonoides/farmacologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfolinas/farmacologia , Miócitos Cardíacos/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Poli(ADP-Ribose) Polimerases , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo
12.
Cardiovasc Res ; 74(1): 56-63, 2007 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-17320845

RESUMO

OBJECTIVES: Activation of sphingosine kinase (SphK), which has two known isoforms, is responsible for the synthesis of sphingosine 1-phosphate (S1P), a cell survival factor. We tested the following hypotheses: 1] cardiac myocytes null for the SphK1 gene are more vulnerable to the stress of hypoxia+glucose deprivation; 2] the monoganglioside GM-1, which activates SphK via protein kinase C epsilon, is ineffective in SphK1-null myocytes; 3] S1P generated by SphK activation requires cellular export to be cardioprotective. METHODS: We cultured adult mouse cardiac myocytes from wildtype and SphK1-null mice (deletion of exons 3-6) and measured cell viability by trypan blue exclusion. RESULTS: In wildtype adult mouse cardiomyocytes subjected to 4 h of hypoxic stress+glucose deprivation, cell viability was significantly higher than in SphK1-null cardiomyocytes. SphK1-null cells also displayed more mitochondrial cytochrome C release. Cell death induced by hypoxia+glucose deprivation was substantially prevented by pretreatment with exogenous S1P in both wildtype and SphK1-null myocytes, but S1P was effective at a lower concentration in wildtype cells. Hence, the absence of the Sphk1 gene did not affect receptor coupling or downstream signal transduction. Pretreatment for 1 h with 1 microM of the monoganglioside GM-1 increased survival in wildtype cells, but not in SphK1-null myocytes. Thus, activation of SphK1 by GM-1 leads to cell survival. In wildtype cells, enhanced survival produced by GM-1 was abrogated by pretreatment either with 300 nM of the S1P(1) receptor-selective antagonist VPC23019 or with 100 ng/ml of pertussis toxin for 16 h before exposure to hypoxia+glucose deprivation. CONCLUSION: As the effect of GM-1 is blocked both at the receptor and the G-protein (Gi) levels, we conclude that S1P generated by GM-1 treatment must be exported from the cell and acts in a paracrine or autocrine manner to couple with its cognate receptor.


Assuntos
Lisofosfolipídeos/metabolismo , Isquemia Miocárdica/metabolismo , Miócitos Cardíacos/enzimologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Esfingosina/análogos & derivados , Animais , Apoptose/efeitos dos fármacos , Biomarcadores/análise , Western Blotting/métodos , Hipóxia Celular , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Citocromos c/análise , Citocromos c/metabolismo , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Ginsenosídeos/farmacologia , Glucose/metabolismo , Hipoglicemiantes/farmacologia , Lisofosfolipídeos/antagonistas & inibidores , Lisofosfolipídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias Cardíacas/metabolismo , Miócitos Cardíacos/patologia , Toxina Pertussis/farmacologia , Fosfotransferases (Aceptor do Grupo Álcool)/análise , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Esfingosina/antagonistas & inibidores , Esfingosina/metabolismo , Esfingosina/farmacologia
13.
Med Sci Monit Basic Res ; 19: 126-32, 2013 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-23567658

RESUMO

BACKGROUND: We investigated the hypothesis that postconditioning by FTY720 (FTY) in isolated perfused mouse hearts is independent of the sphingosine 1-phosphate (S1P) pathway. MATERIAL AND METHODS: Ex vivo hearts were exposed to postconditioning (POST) by either ischemia or FTY720. Protection against ischemia/reperfusion (IR) injury was measured by recovery of left ventricular developed pressure (LVDP) and infarct size. RESULTS: FTY effectively postconditioned (POST) ex vivo hearts against ischemia/reperfusion (IR) injury as measured by recovery of LVDP and a low infarct size. FTY protection, unlike S1P but like sphingosine (Sph), was insensitive to inhibition of S1P G-Protein Coupled Receptors (GPCRs) or inhibition of PI3 kinase. Protection by FTY and Sph was however blocked by inhibitors of PKA and PKG. Thus, FTY follows the same cardioprotective pathway as Sph. This was further supported by studies of FTY POST in knockout (KO) mice lacking the SphK2 form of Sph kinase that is needed for phosphorylation of FTY to an S1P analog. In the absence of SphK2, FTY (and Sph) POST was still cardioprotective. This differed from the effect of SphK2 KO on protection by ischemic POST (IPOST). IPOST was not effective in KO hearts. To see if the GPCR signaling pathway to protection is normal in KO hearts, we looked at POST by GPCR agonists S1P and adenosine. Both provided effective protection even in KO hearts suggesting that the problem with IPOST in KO hearts is a low level of S1P available for release during IPOST. Thus, pharmacologic POST with FTY or Sph, like adenosine and S1P, is unaffected in the KO. CONCLUSIONS: FTY720 administered in vivo might behave in a dual manner showing both S1P-like effects and sphingosine-like effects. It appears that the latter may have been overlooked and may be the more important in aging hearts.


Assuntos
Imunossupressores/farmacologia , Pós-Condicionamento Isquêmico/métodos , Lisofosfolipídeos/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Propilenoglicóis/farmacologia , Esfingosina/análogos & derivados , Adenosina/metabolismo , Algoritmos , Animais , Cloridrato de Fingolimode , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Perfusão , Fosforilação , Traumatismo por Reperfusão , Esfingosina/metabolismo , Esfingosina/farmacologia
14.
Oxid Med Cell Longev ; 2011: 961059, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21904650

RESUMO

Sphingosine kinase (SphK) exhibits two isoforms, SphK1 and SphK2. Both forms catalyze the synthesis of sphingosine 1-phosphate (S1P), a sphingolipid involved in ischemic preconditioning (IPC). Since the ratio of SphK1:SphK2 changes dramatically with aging, it is important to assess the role of SphK2 in IR injury and IPC. Langendorff mouse hearts were subjected to IR (30 min equilibration, 50 min global ischemia, and 40 min reperfusion). IPC consisted of 2 min of ischemia and 2 min of reperfusion for two cycles. At baseline, there were no differences in left ventricular developed pressure (LVDP), ± dP/dtmax, and heart rate between SphK2 null (KO) and wild-type (WT) hearts. In KO hearts, SphK2 activity was undetectable, and SphK1 activity was unchanged compared to WT. Total SphK activity was reduced by 53%. SphK2 KO hearts subjected to IR exhibited significantly more cardiac damage (37 ± 1% infarct size) compared with WT (28 ± 1% infarct size); postischemic recovery of LVDP was lower in KO hearts. IPC exerted cardioprotection in WT hearts. The protective effect of IPC against IR was diminished in KO hearts which had much higher infarction sizes (35 ± 2%) compared to the IPC/IR group in control hearts (12 ± 1%). Western analysis revealed that KO hearts had substantial levels of phosphorylated p38 which could predispose the heart to IR injury. Thus, deletion of the SphK2 gene sensitizes the myocardium to IR injury and diminishes the protective effect of IPC.


Assuntos
Precondicionamento Isquêmico Miocárdico , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Animais , Camundongos , Camundongos Knockout , Traumatismo por Reperfusão Miocárdica/genética , Fosforilação , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
15.
FEBS Lett ; 584(1): 159-65, 2010 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-19913538

RESUMO

Alkaline incubation of NADH results in the formation of a very potent inhibitor of lactate dehydrogenase. High resolution mass spectroscopy along with NMR characterization clearly showed that the inhibitor is derived from attachment of a glycolic acid moiety to the 4-position of the dihydronicotinamide ring of NADH. The very potent inhibitor is competitive with respect to NADH. The inhibitor added in submicromolar concentrations to cardiomyocytes protects them from damage caused by hypoxia/reoxygenation stress. In isolated mouse hearts, addition of the inhibitor results in a substantial reduction of myocardial infarct size caused by global ischemia/reperfusion injury.


Assuntos
Cardiotônicos/farmacologia , Inibidores Enzimáticos/farmacologia , L-Lactato Desidrogenase/antagonistas & inibidores , Infarto do Miocárdio/prevenção & controle , Traumatismo por Reperfusão Miocárdica/complicações , Miocárdio/enzimologia , NAD/farmacologia , Animais , Cardiotônicos/química , Inibidores Enzimáticos/química , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/etiologia , NAD/química
16.
J Mol Cell Cardiol ; 43(1): 85-91, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17512943

RESUMO

Cardiac fibroblasts are critical for the maintenance of extracellular matrix deposition and turnover in the normal heart and are key mediators of inflammatory and fibrotic myocardial remodeling in the injured and failing heart. Sphingosine kinase (SphK) activation is a well-recognized determinant of cell fate in cardiac myocytes and other cells, but SphK responses have not previously been studied in cardiac fibroblasts. Initially we found that total SphK activity is over 10-fold higher in cardiac fibroblasts than in adult mouse cardiac myocytes. SphK is composed of two major isoforms, SphK-1 and SphK-2. In fibroblasts isolated from SphK-1 knockout mice, SphK activity was greatly reduced indicating that SphK-1 is the major isoform expressed in these cells. To determine whether SphK regulates cell proliferation and the proinflammatory protein inducible nitric oxide synthase (iNOS), we exposed cultured cardiac fibroblasts to the cytokine interleukin-1beta (IL-1beta) and/or hypoxia. Both hypoxia and IL-1beta alone and in combination enhanced fibroblast SphK activity. In wild-type fibroblasts, hypoxia induced proliferation, but in SphK-1 null fibroblasts this response was blunted even in the presence of serum. In contrast, we found that iNOS expression and NO production were enhanced in SphK-1 null fibroblasts during hypoxia. In wild-type fibroblasts, IL-1beta was only a weak inducer of iNOS and of NO accumulation and hypoxia alone had no significant effect on iNOS activation. However, IL-1beta in combination with hypoxia extensively stimulated iNOS and NO production, and this stimulation was enhanced in SphK-1 null fibroblasts. We conclude that activation of endogenous SphK-1 serves a dual regulatory function: it is required for optimal cardiac fibroblast proliferation but is a negative modulator of proinflammatory responses during hypoxia.


Assuntos
Fibroblastos/metabolismo , Hipóxia , Interleucina-1beta/metabolismo , Miocárdio/citologia , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Animais , Proliferação de Células , Células Cultivadas , Fibroblastos/enzimologia , Fibroblastos/imunologia , Interleucina-1beta/farmacologia , Isoenzimas/genética , Camundongos , Camundongos Knockout , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo
17.
Biochem Biophys Res Commun ; 363(2): 257-62, 2007 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-17880922

RESUMO

We investigated the ability of pyrroloquinoline quinone (PQQ) to confer resistance to acute oxidative stress in freshly isolated adult male rat cardiomyocytes. Fluorescence microscopy was used to detect generation of reactive oxygen species (ROS) and mitochondrial membrane potential (Deltapsi(m)) depolarization induced by hydrogen peroxide. H(2)O(2) caused substantial cell death, which was significantly reduced by preincubation with PQQ. H(2)O(2) also caused an increase in cellular ROS levels as detected by the fluorescent indicators CM-H2XRos and dihydroethidium. ROS levels were significantly reduced by a superoxide dismutase mimetic Mn (III) tetrakis (4-benzoic acid) porphyrin chloride (MnTBAP) or by PQQ treatment. Cyclosporine-A, which inhibits mitochondrial permeability transition, prevented H(2)O(2)-induced Deltapsi(m) depolarization, as did PQQ and MnTBAP. Our results provide direct evidence that PQQ reduces oxidative stress, mitochondrial dysfunction, and cell death in isolated adult rat cardiomyocytes. These findings provide new insight into the mechanisms of PQQ action in the heart.


Assuntos
Mitocôndrias Cardíacas/fisiologia , Miócitos Cardíacos/fisiologia , Estresse Oxidativo/fisiologia , Cofator PQQ/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Animais , Células Cultivadas , Citoproteção/efeitos dos fármacos , Citoproteção/fisiologia , Relação Dose-Resposta a Droga , Masculino , Mitocôndrias Cardíacas/efeitos dos fármacos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Cofator PQQ/administração & dosagem , Ratos
18.
Am J Physiol Heart Circ Physiol ; 293(5): H3150-8, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17766476

RESUMO

Sphingosine 1-phosphate (S1P) is a biologically active lysophospholipid that serves as a key regulator of cellular differentiation and survival. Immune stimuli increase S1P synthesis and secretion by mast cells and platelets, implicating this molecule in tissue responses to injury and inflammation. Binding of S1P to G(i) protein-coupled receptors activates phosphatidylinositol 3-kinase and Akt in a variety of tissues. To elucidate the mechanisms by which S1P enhances adult cardiac myocyte survival during hypoxia, we used a mouse cell culture system in which S1P(1) receptors were observed to transduce signals from exogenous S1P, an S1P(1) receptor antibody with agonist properties, and the pharmacological agents FTY720 and SEW2871. S1P(1) receptor mRNA and protein were abundantly expressed by adult mouse cardiac myocytes. S1P-S1P(1) receptor axis enhancement of myocyte survival during hypoxia was abolished by phosphatidylinositol 3-kinase inhibition. S1P(1) receptor function was closely associated with activation of Akt, inactivation of GSK-3beta, and reduction of cytochrome c release from heart mitochondria. These observations highlight the importance of S1P(1) receptors on ventricular myocytes as mediators of inducible resistance against cellular injury during severe hypoxic stress.


Assuntos
Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Transdução de Sinais/fisiologia , Animais , Hipóxia Celular/fisiologia , Sobrevivência Celular , Células Cultivadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL
19.
J Mol Cell Cardiol ; 43(3): 327-36, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17662302

RESUMO

Vincristine is a chemotherapeutic agent that disrupts microtubules. We noted that paclitaxel (Taxol), which stabilizes microtubules, protected cultured adult mouse cardiac myocytes from oxidative stress induced by H(2)O(2). We hypothesized that vincristine, which disrupts microtubules, should have the opposite effect. To our surprise, we found that pretreatment with concentrations of vincristine ranging from 30 to 120 micromol/L for 60 min preserved myocyte viability and morphology after incubation with 30 micromol/L of H(2)O(2) for 35 min as measured by trypan blue exclusion. The cardioprotective effects of vincristine were also observed during prolonged hypoxia. With continuous exposure to vincristine, survival lasted for as long as 24 h, but longer periods of exposure up to 42 h resulted in extensive cell death. Despite microtubule disruption evidenced on deconvolution microscopy, vincristine activated a prosurvival pathway resulting in increased phosphorylation of Akt, ERK and GSK-3beta and in reduced cytochrome C release into the cytosol. Pharmacological inhibitors of Akt and Erk attenuated the cardioprotective effect of vincristine. We conclude that short-term pretreatment with vincristine exerts dramatic protective effects in cultured adult mouse myocytes subjected to acute oxidative stress. Despite causing microtubule disruption, vincristine initiates a prosurvival signaling pathway. As vincristine and doxorubicin are often used in conjunction to treat patients, it is possible that vincristine could be used to modify the cardiotoxicity of doxorubicin.


Assuntos
Cardiotônicos/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Vincristina/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Ventrículos do Coração/citologia , Peróxido de Hidrogênio/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microtúbulos/efeitos dos fármacos , Oxidantes/farmacologia , Fatores de Tempo
20.
Biochem J ; 369(Pt 3): 485-96, 2003 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-12371906

RESUMO

Enhanced synthesis of a specific matrix metalloproteinase, MMP-2, has been demonstrated in experimental models of ventricular failure and in cardiac extracts from patients with ischaemic cardiomyopathy. Cultured neonatal rat cardiac fibroblasts and myocytes were used to analyse the determinants of MMP-2 synthesis, including the effects of hypoxia. Culture of rat cardiac fibroblasts for 24 h in 1% oxygen enhanced MMP-2 synthesis by more than 5-fold and augmented the MMP-2 synthetic responses of these cells to endothelin-1, angiotensin II and interleukin 1beta. A series of MMP-2 promoter-luciferase constructs were used to map the specific enhancer element(s) that drive MMP-2 transcription in cardiac cells. Deletion studies mapped a region of potent transactivating function within the 91 bp region from -1433 to -1342 bp, the activity of which was increased by hypoxia. Oligonucleotides from this region were cloned in front of a heterologous simian-virus-40 (SV40) promoter and mapped the enhancer activity to a region between -1410 and -1362 bp that included a potential activating protein 1 (AP-1)-binding sequence, C(-1394)CTGACCTCC. Site-specific mutagenesis of the core TGAC sequence (indicated in bold) eliminated the transactivating activity within the -1410 to -1362 bp sequence. Electrophoretic mobility shift assays (EMSAs) using the -1410 to -1362 bp oligonucleotide and rat cardiac fibroblast nuclear extracts demonstrated specific nuclear-protein binding that was eliminated by cold competitor oligonucleotide, but not by the AP-1-mutated oligonucleotide. Antibody-supershift EMSAs of nuclear extracts from normoxic rat cardiac fibroblasts demonstrated Fra1 and JunB binding to the -1410 to -1362 bp oligonucleotide. Nuclear extracts isolated from hypoxic rat cardiac fibroblasts contained Fra1, JunB and also included FosB. Co-transfection of cardiac fibroblasts with Fra1-JunB and FosB-JunB expression plasmids led to significant increases in transcriptional activity. These studies demonstrate that a functional AP-1 site mediates MMP-2 transcription in cardiac cells through the binding of distinctive Fra1-JunB and FosB-JunB heterodimers. The synthesis of MMP-2 is widely considered, in contrast with many members of the MMP gene family, to be independent of the AP-1 transcriptional complex. This report is the first demonstration that defined members of the Fos and Jun transcription-factor families specifically regulate this gene under conditions relevant to critical pathophysiological processes.


Assuntos
Metaloproteinase 2 da Matriz/genética , Miócitos Cardíacos/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Fator de Transcrição AP-1/metabolismo , Angiotensina II/farmacologia , Animais , Sítios de Ligação , Hipóxia Celular , Células Cultivadas , Dimerização , Endotelina-1/farmacologia , Fibroblastos/metabolismo , Interleucina-1/farmacologia , Metaloproteinase 2 da Matriz/efeitos dos fármacos , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinases da Matriz Associadas à Membrana , Metaloendopeptidases/metabolismo , Mutagênese Sítio-Dirigida , Miócitos Cardíacos/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Fator de Transcrição AP-1/genética , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA