Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Hypertension ; 80(11): 2265-2279, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37503660

RESUMO

Screening of compounds comprising 8-substituted guanine revealed that 8-aminoguanosine and 8-aminoguanine cause diuresis/natriuresis/glucosuria, yet decrease potassium excretion. Subsequent investigations demonstrated that 8-aminoguanosine's effects are mediated by its metabolite 8-aminoguanine. The mechanism by which 8-aminoguanine causes diuresis/natriuresis/glucosuria involves inhibition of PNPase (purine nucleoside phosphorylase), which increases renal interstitial inosine levels. Additional evidence suggests that inosine, via indirect or direct adenosine A2B receptor activation, increases renal medullary blood flow which enhances renal excretory function. Likely, 8-aminoguanine has pleiotropic actions that also alter renal excretory function. Indeed, the antikaliuretic effects of 8-aminoguanine are independent of PNPase inhibition. 8-Aminoguanine is an endogenous molecule; nitrosative stress leads to production of biomolecules containing 8-nitroguanine moieties. Degradation of these biomolecules releases 8-nitroguanosine and 8-nitro-2'-deoxyguanosine which are converted to 8-aminoguanine. Also, guanosine and guanine per se may contribute to 8-aminoguanine formation. 8-Aminoinosine, 8-aminohypoxanthine, and 8-aminoxanthine likewise induce diuresis/natriuresis/glucosuria, yet do not reduce potassium excretion. Thus, there are several pharmacologically active 8-aminopurines with nuanced effects on renal excretory function. Chronic treatment with 8-aminoguanine attenuates hypertension in deoxycorticosterone/salt rats, prevents strokes, and increases lifespan in Dahl salt-sensitive rats on a high salt diet and attenuates the metabolic syndrome in rats; 8-aminoguanosine retards progression of pulmonary hypertension in rats and anemia and organ damage in sickle cell mice. 8-Aminoguanine reverses age-associated lower urinary tract dysfunction and retinal degeneration. 8-Aminopurines represent a new class of agents (and potentially endogenous factors) that have beneficial effects on the cardiovascular system and kidneys and may turn back the clock in age-associated diseases.


Assuntos
Sistema Cardiovascular , Guanina , Ratos , Camundongos , Animais , Ratos Endogâmicos Dahl , Guanina/metabolismo , Guanina/farmacologia , Natriurese , Sistema Cardiovascular/metabolismo , Potássio , Inosina/farmacologia
2.
Artigo em Inglês | MEDLINE | ID: mdl-36473034

RESUMO

Vascular and lung injury are well established complications associated with hemolytic disorders, and hemolysis associated pulmonary hypertension (PH) has emerged as the most serious complication of sickle cell disease. The causal relationship between intravascular hemolysis and the development of PH is still under investigation. Previously we have shown that repetitive administration of hemolyzed autologous blood causes PH in rats. Dimethyl sulfoxide (DMSO), a widely used solvent and anti-inflammatory agent, induces hemolysis in vivo. We hypothesized that repetitive administration of DMSO would induce PH in rats. We also examined hemolysis-induced release of adenosine deaminase (ADA) and arginase from red blood cells, which may amplify hemolysis-mediated vascular injury. Acute administration of DMSO (1.5ml/30 min into the right atrium) induced intravascular hemolysis and pulmonary vasoconstriction. DMSO-induced increase in right ventricular peak systolic pressure (RVPSP) was associated with increased release of ADA. Notably, the acute increase in RVPSP was attenuated by administration of an adenosine A2A receptor agonist or by pretreatment of animals with ADA inhibitor erythro-9-(2-hydroxy-3-nonyl) adenine (EHNA). Repetitive administration of DMSO for 10 days produced anemia, hemoglobinuria, hemoglobinemia, splenomegaly, and development of PH. Histopathological analysis revealed pulmonary vascular remodeling. The presented data describe a new model of hemolysis induced PH, suggesting that hemolysis is mechanistically related to pulmonary hypertension, and pointing to a potential pathogenic role that adenosine deaminase and accelerated adenosine metabolism may play in hemolysis associated pulmonary hypertension.


Assuntos
Dimetil Sulfóxido , Hipertensão Pulmonar , Animais , Humanos , Ratos , Adenosina Desaminase , Dimetil Sulfóxido/farmacologia , Hipertensão Pulmonar/induzido quimicamente
3.
Int J Mol Sci ; 21(1)2019 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-31877978

RESUMO

Pulmonary arterial hypertension (PAH) is a debilitating and progressive disease that predominantly develops in women. Over the past 15 years, cumulating evidence has pointed toward dysregulated metabolism of sex hormones in animal models and patients with PAH. 17ß-estradiol (E2) is metabolized at positions C2, C4, and C16, which leads to the formation of metabolites with different biological/estrogenic activity. Since the first report that 2-methoxyestradiol, a major non-estrogenic metabolite of E2, attenuates the development and progression of experimental pulmonary hypertension (PH), it has become increasingly clear that E2, E2 precursors, and E2 metabolites exhibit both protective and detrimental effects in PH. Furthermore, both experimental and clinical data suggest that E2 has divergent effects in the pulmonary vasculature versus right ventricle (estrogen paradox in PAH). The estrogen paradox is of significant clinical relevance for understanding the development, progression, and prognosis of PAH. This review updates experimental and clinical findings and provides insights into: (1) the potential impacts that pathways of estradiol metabolism (EMet) may have in PAH; (2) the beneficial and adverse effects of estrogens and their precursors/metabolites in experimental PH and human PAH; (3) the co-morbidities and pathological conditions that may alter EMet and influence the development/progression of PAH; (4) the relevance of the intracrinology of sex hormones to vascular remodeling in PAH; and (5) the advantages/disadvantages of different approaches to modulate EMet in PAH. Finally, we propose the three-tier-estrogen effects in PAH concept, which may offer reconciliation of the opposing effects of E2 in PAH and may provide a better understanding of the complex mechanisms by which EMet affects the pulmonary circulation-right ventricular interaction in PAH.


Assuntos
Estradiol/metabolismo , Ventrículos do Coração , Hipertensão Arterial Pulmonar , Remodelação Vascular , Animais , Modelos Animais de Doenças , Feminino , Ventrículos do Coração/metabolismo , Ventrículos do Coração/patologia , Ventrículos do Coração/fisiopatologia , Humanos , Masculino , Hipertensão Arterial Pulmonar/metabolismo , Hipertensão Arterial Pulmonar/patologia , Hipertensão Arterial Pulmonar/fisiopatologia
4.
Pulm Circ ; 8(3): 2045894018791557, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30003836

RESUMO

Pulmonary hypertension (PH) is emerging as a serious complication associated with hemolytic disorders, and plexiform lesions (PXL) have been reported in patients with sickle cell disease (SCD). We hypothesized that repetitive hemolysis per se induces PH and angioproliferative vasculopathy and evaluated a new mechanism for hemolysis-associated PH (HA-PH) that involves the release of adenosine deaminase (ADA) and purine nucleoside phosphorylase (PNP) from erythrocytes. In healthy rats, repetitive administration of hemolyzed autologous blood (HAB) for 10 days produced reversible pulmonary parenchymal injury and vascular remodeling and PH. Moreover, the combination of a single dose of Sugen-5416 (SU, 200 mg/kg) and 10-day HAB treatment resulted in severe and progressive obliterative PH and formation of PXL (Day 26, right ventricular peak systolic pressure (mmHg): 26.1 ± 1.1, 41.5 ± 0.5 and 85.1 ± 5.9 in untreated, HAB treated and SU+HAB treated rats, respectively). In rats, repetitive administration of HAB increased plasma ADA activity and reduced urinary adenosine levels. Similarly, SCD patients had higher plasma ADA and PNP activity and accelerated adenosine, inosine, and guanosine metabolism than healthy controls. Our study provides evidence that hemolysis per se leads to the development of angioproliferative PH. We also report the development of a rat model of HA-PH that closely mimics pulmonary vasculopathy seen in patients with HA-PH. Finally, this study suggests that in hemolytic diseases released ADA and PNP may increase the risk of PH, likely by abolishing the vasoprotective effects of adenosine, inosine and guanosine. Further characterization of this new rat model of hemolysis-induced angioproliferative PH and additional studies of the role of purines metabolism in HA-PH are warranted.

6.
J Pharmacol Exp Ther ; 356(2): 333-40, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26585572

RESUMO

Heart failure with preserved ejection fraction (HFpEF) is prevalent and often accompanied by metabolic syndrome. Current treatment options are limited. Here, we test the hypothesis that combined A1/A2B adenosine receptor blockade is beneficial in obese ZSF1 rats, an animal model of HFpEF with metabolic syndrome. The combined A1/A2B receptor antagonist 3-[4-(2,6-dioxo-1,3-dipropyl-7H-purin-8-yl)-1-bicyclo[2.2.2]octanyl]propanoic acid (BG9928) was administered orally (10 mg/kg/day) to obese ZSF1 rats (n = 10) for 24 weeks (from 20 to 44 weeks of age). Untreated ZSF1 rats (n = 9) served as controls. After 24 weeks of administration, BG9928 significantly lowered plasma triglycerides (in mg/dl: control group, 4351 ± 550; BG9928 group, 2900 ± 551) without adversely affecting plasma cholesterol or activating renin release. BG9928 significantly decreased 24-hour urinary glucose excretion (in mg/kg/day: control group, 823 ± 179; BG9928 group, 196 ± 80) and improved oral glucose tolerance, polydipsia, and polyuria. BG9928 significantly augmented left ventricular diastolic function in association with a reduction in cardiac vasculitis and cardiac necrosis. BG9928 significantly reduced 24-hour urinary protein excretion (in mg/kg/day: control group, 1702 ± 263; BG9928 group, 1076 ± 238), and this was associated with a reduction in focal segmental glomerulosclerosis, tubular atrophy, tubular dilation, and deposition of proteinaceous material in the tubules. These findings show that, in a model of HFpEF with metabolic syndrome, A1/A2B receptor inhibition improves hyperlipidemia, exerts antidiabetic actions, reduces HFpEF, improves cardiac histopathology, and affords renal protection. We conclude that chronic administration of combined A1/A2B receptor antagonists could be beneficial in patients with HFpEF, in particular those with comorbidities such as obesity, diabetes, and dyslipidemias.


Assuntos
Antagonistas do Receptor A1 de Adenosina/administração & dosagem , Antagonistas do Receptor A2 de Adenosina/administração & dosagem , Insuficiência Cardíaca/tratamento farmacológico , Rim/fisiologia , Volume Sistólico/efeitos dos fármacos , Xantinas/administração & dosagem , Animais , Insuficiência Cardíaca/metabolismo , Rim/efeitos dos fármacos , Masculino , Ratos , Ratos Zucker , Receptor A1 de Adenosina/metabolismo , Receptor A2B de Adenosina/metabolismo , Volume Sistólico/fisiologia , Resultado do Tratamento
7.
Hypertension ; 65(1): 238-49, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25368027

RESUMO

UNLABELLED: Because the effects of dipeptidyl peptidase 4 (DPP4) inhibitors on blood pressure are controversial, we examined the long-term effects of sitagliptin (80 mg/kg per day) on blood pressure (radiotelemetry) in spontaneously hypertensive rats (SHR), Wistar-Kyoto rats, and Zucker Diabetic-Sprague Dawley rats (metabolic syndrome model). In SHR, chronic (3 weeks) sitagliptin significantly increased systolic, mean, and diastolic blood pressures by 10.3, 9.2, and 7.9 mm Hg, respectively, a response abolished by coadministration of BIBP3226 (2 mg/kg per day; selective Y1-receptor antagonist). Sitagliptin also significantly increased blood pressure in SHR treated with hydralazine (vasodilator; 25 mg/kg per day) or enalapril (angiotensin-converting enzyme inhibitor; 10 mg/kg per day). In Wistar-Kyoto rats, chronic sitagliptin slightly decreased systolic, mean, and diastolic blood pressures (-1.8, -1.1, and -0.4 mm Hg, respectively). In Zucker Diabetic-Sprague Dawley rats, chronic sitagliptin decreased systolic, mean, and diastolic blood pressures by -7.7, -5.8, and -4.3 mm Hg, respectively, and did not alter the antihypertensive effects of chronic enalapril. Because DPP4 inhibitors impair the metabolism of neuropeptide Y1-36 (NPY1-36; Y1-receptor agonist) and glucagon-like peptide (GLP)-1(7-36)NH2 (GLP-1 receptor agonist), we examined renovascular responses to NPY1-36 and GLP-1(7-36)NH2 in isolated perfused SHR and Zucker Diabetic-Sprague Dawley kidneys pretreated with norepinephrine (to induce basal tone). In Zucker Diabetic-Sprague Dawley kidneys, NPY1-36 and GLP-1(7-36)NH2 exerted little, if any, effect on renovascular tone. In contrast, in SHR kidneys, both NPY1-36 and GLP-1(7-36)NH2 elicited potent and efficacious vasoconstriction. IN CONCLUSION: (1) The effects of DPP4 inhibitors on blood pressure are context dependent; (2) The context-dependent effects of DPP4 inhibitors are due in part to differential renovascular responses to DPP4's most important substrates (NPY1­36 and GLP-1(7­36)NH2) [corrected]; (3) Y1 receptor antagonists may prevent the prohypertensive and possibly augment the antihypertensive effects of DPP4 inhibitors.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Dipeptidil Peptidase 4/metabolismo , Inibidores da Dipeptidil Peptidase IV/farmacologia , Hipertensão/tratamento farmacológico , Pirazinas/farmacologia , Triazóis/farmacologia , Animais , Determinação da Pressão Arterial , Dipeptidil Peptidase 4/efeitos dos fármacos , Modelos Animais de Doenças , Peptídeo 1 Semelhante ao Glucagon , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Masculino , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Fosfato de Sitagliptina , Resultado do Tratamento
8.
Pulm Circ ; 3(2): 294-314, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24015330

RESUMO

Most subtypes of pulmonary arterial hypertension (PAH) are characterized by a greater susceptibility to disease among females, although females with PAH appear to live longer after diagnosis. While this "estrogen paradoxȍ of enhanced female survival despite increased female susceptibility remains a mystery, recent progress has begun to shed light upon the interplay of sex hormones, the pathogenesis of pulmonary hypertension, and the right ventricular response to stress. For example, emerging data in humans and experimental models suggest that estrogens or differential sex hormone metabolism may modify disease risk among susceptible subjects, and that estrogens may interact with additional local factors such as serotonin to enhance the potentially damaging chronic effects of estrogens on the pulmonary vasculature. Regardless, it remains unclear why not all estrogenic compounds behave equally, nor why estrogens appear to be protective in certain settings but detrimental in others. The contribution of androgens and other compounds, such as dehydroepiandrosterone, to pathogenesis and possibly treatment must be considered as well. In this review, we will discuss the recent understandings on how estrogens, estrogen metabolism, dehydroepiandrosterone, and additional susceptibility factors may all contribute to the pathogenesis or potentially to the treatment of pulmonary hypertension, by evaluating current human, cell-based, and experimental model data.

10.
Am J Physiol Renal Physiol ; 302(4): F466-76, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22114202

RESUMO

Adenosine A(1) receptor antagonists have diuretic/natriuretic activity and may be useful for treating sodium-retaining diseases, many of which are associated with increased renal sympathetic tone. Therefore, it is important to determine whether A(1) receptor antagonists alter renal sympathetic neurotransmission. In isolated, perfused rat kidneys, renal vasoconstriction induced by renal sympathetic nerve simulation was attenuated by 1) 1,3-dipropyl-8-p-sulfophenylxanthine (xanthine analog that is a nonselective adenosine receptor antagonist, but is cell membrane impermeable and thus does not block intracellular phosphodiesterases), 2) xanthine amine congener (xanthine analog that is a selective A(1) receptor antagonist), 3) 1,3-dipropyl-8-cyclopentylxanthine (xanthine analog that is a highly selective A(1) receptor antagonist), and 4) FK453 (nonxanthine analog that is a highly selective A(1) receptor antagonist). In contrast, FR113452 (enantiomer of FK453 that does not block A(1) receptors), MRS-1754 (selective A(2B) receptor antagonist), and VUF-5574 (selective A(3) receptor antagonist) did not alter responses to renal sympathetic nerve stimulation, and ZM-241385 (selective A(2A) receptor antagonist) enhanced responses. Antagonism of A(1) receptors did not alter renal spillover of norepinephrine. 2-Chloro-N(6)-cyclopentyladenosine (highly selective A(1) receptor agonist) increased renal vasoconstriction induced by exogenous norepinephrine, an effect that was blocked by 1,3-dipropyl-8-cyclopentylxanthine, U73122 (phospholipase C inhibitor), GF109203X (protein kinase C inhibitor), PP1 (c-src inhibitor), wortmannin (phosphatidylinositol 3-kinase inhibitor), and OSU-03012 (3-phosphoinositide-dependent protein kinase-1 inhibitor). These results indicate that adenosine formed during renal sympathetic nerve stimulation enhances the postjunctional effects of released norepinephrine via coincident signaling and contributes to renal sympathetic neurotransmission. Likely, the coincident signaling pathway is: phospholipase C → protein kinase C → c-src → phosphatidylinositol 3-kinase → 3-phosphoinositide-dependent protein kinase-1.


Assuntos
Adenosina/metabolismo , Rim/inervação , Receptor A1 de Adenosina/metabolismo , Sistema Nervoso Simpático/fisiologia , Transmissão Sináptica/fisiologia , Animais , Rim/irrigação sanguínea , Rim/efeitos dos fármacos , Masculino , Antagonistas de Receptores Purinérgicos P1/farmacologia , Ratos , Ratos Sprague-Dawley , Sistema Nervoso Simpático/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Vasoconstrição/efeitos dos fármacos , Vasoconstrição/fisiologia
11.
J Cardiovasc Pharmacol ; 56(5): 475-83, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20881615

RESUMO

2-Methoxyestradiol (2ME) is a major nonestrogenic metabolite of estradiol. Our previous studies suggest that 2ME, in several models of cardiac and/or vascular injury, strongly inhibits cardiac and vascular remodeling. Furthermore, our most recent study shows that in male rats, 2ME attenuates the development and retards the progression of monocrotaline (MCT)-induced pulmonary arterial hypertension (PAH), and in female rats, 2ME eliminates the exacerbation of PAH and the increased mortality due to ovariectomy. The current standard of care of patients with PAH includes treatment with an endothelin receptor antagonist (eg, bosentan) or a phosphodiesterase5 inhibitor (eg, sildenafil). Moreover, combination therapy is often prescribed. Therefore, in the present study, we compared the efficacy of 2ME (10 µg · kg(-1) · h(-1), 2ME-10) to the effects of bosentan (200 mg/kg; BOS), sildenafil (50 mg/kg; SIL), and their respective combinations with 2ME-10 (2ME + BOS and 2ME + SIL groups, respectively). Treatments were initiated 12 days after administration of MCT (60 mg/kg). Twenty-eight days after MCT administration, right ventricular peak systolic pressure was measured and morphometric analysis was conducted. 2ME exhibited beneficial effects in pulmonary hypertensive animals and had efficacy comparable to that of BOS and SIL. Importantly, combination treatments had favorable effects on survival, vascular remodeling, and inflammatory response, and the 2ME + SIL combination was significantly more efficacious than any other treatment. These results indicate that 2ME is effective in experimental PAH and suggests that 2ME may provide additional therapeutic benefit over existing drugs used for the treatment of pulmonary hypertension.


Assuntos
Anti-Hipertensivos/uso terapêutico , Estradiol/análogos & derivados , Hipertensão Pulmonar/tratamento farmacológico , Piperazinas/uso terapêutico , Sulfonamidas/uso terapêutico , Sulfonas/uso terapêutico , Vasodilatadores/uso terapêutico , 2-Metoxiestradiol , Animais , Anti-Hipertensivos/farmacologia , Bosentana , Sinergismo Farmacológico , Quimioterapia Combinada , Estradiol/farmacologia , Estradiol/uso terapêutico , Feminino , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/patologia , Masculino , Monocrotalina , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/patologia , Piperazinas/farmacologia , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/patologia , Purinas/farmacologia , Purinas/uso terapêutico , Ratos , Ratos Sprague-Dawley , Citrato de Sildenafila , Sulfonamidas/farmacologia , Sulfonas/farmacologia , Vasodilatadores/farmacologia
12.
J Cardiovasc Pharmacol ; 56(6): 696-708, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20881610

RESUMO

Severe pulmonary arterial hypertension (PAH) is characterized by clustered proliferation of endothelial cells (ECs) in the lumina of small size pulmonary arteries resulting in concentric obliteration of the lumina and formation of complex vascular structures known as plexiform lesions. This debilitating disease occurs more frequently in women, yet both animal studies in classical models of PAH and limited clinical data suggest protective effects of estrogens: the estrogen paradox in pulmonary hypertension. Little is known about the role of estrogens in PAH, but one line of evidence strongly suggests that the vascular protective effects of 17ß-estradiol (estradiol; E2) are mediated largely by its downstream metabolites. Estradiol is metabolized to 2-hydroxyestradiol (2HE) by CYP1A1/CYP1B1, and 2HE is converted to 2-methoxyestradiol (2ME) by catechol-O-methyl transferase. 2ME is extensively metabolized to 2-methoxyestrone, a metabolite that lacks biologic activity, but which may be converted back to 2ME. 2ME has no estrogenic activity, and its effects are mediated by estrogen receptors­independent mechanism(s). Notably, in systemic and pulmonary vascular ECs, smooth muscle cells, and fibroblasts, 2ME exerts stronger antimitotic effects than E2 itself. E2 and 2ME, despite having similar effects on other cardiovascular cells, have opposing effects on ECs; that is, in ECs, E2 is promitogenic, proangiogenic, and antiapoptotic, whereas 2ME is antimitogenic, antiangiogenic, and proapoptotic. This may have significant ramifications in severe PAH that involves uncontrolled proliferation of monoclonal apoptosis-resistant ECs. Based on its cellular effects, 2ME should be expected to attenuate the progression of disease and provide protection in severe PAH. In contrast, E2, due to its mitogenic, angiogenic, and antiapoptotic effects (otherwise desirable in normal quiescent ECs), may even adversely affect endothelial remodeling in PAH, and this may be even more significant if the E2's effects on injured endothelium are not opposed by 2ME (eg, in the event of reduced E2 conversion to 2ME due to hypoxia, inflammation, drugs, environmental factors, or genetic polymorphism of metabolizing enzymes). This review focuses on the effects of estrogens and their metabolites on pulmonary vascular pathobiology and the development of experimental PAH and offers potential explanation for the estrogen paradox in PAH. Furthermore, we propose that unbalanced estradiol metabolism may lead to the development of PAH. Recent animal data and studies in patients with PAH support this concept.


Assuntos
Estradiol/metabolismo , Estrogênios/metabolismo , Hipertensão Pulmonar/metabolismo , Doenças Vasculares/metabolismo , Animais , Humanos , Hipertensão Pulmonar/etiologia , Pneumopatias/etiologia , Pneumopatias/metabolismo , Doenças Vasculares/etiologia , Resistência Vascular/fisiologia
13.
Vascul Pharmacol ; 51(2-3): 190-7, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19540933

RESUMO

Pulmonary hypertension (PH) is a common and life-threatening complication of pulmonary fibrosis. Estradiol (E2) is protective in experimental PH, and its non-estrogenic metabolite 2-methoxyestradiol (2ME) prevents the development and retards the progression of monocrotaline-induced PH in male and female rats. However, the effects of E2 and 2ME on pulmonary fibrosis and associated PH have not been examined. Therefore, we compared the growth inhibitory effects of E2 and 2ME in human lung fibroblasts (hLFs) and pulmonary vascular smooth muscle cells (hPASMCs), and we investigated the effects of estrogen deficiency and 2ME on bleomycin-induced pulmonary fibrosis and PH. Intact and ovariectomized (OVX) female Sprague-Dawley rats were administered intratracheally either saline or bleomycin (15IU/kg), and a subset of OVX bleomycin-treated rats received 2ME (10microg/kg/h) for 21days. Estradiol had only limited inhibitory effects on growth in hPASMCs and no effect in hLFs, whereas 2ME exhibited strong and concentration-dependent (1-10microM) antimitogenic effects in both cell types. Bleomycin caused lung injury/PH (significantly increased lung and right ventricle (RV) weights, RV peak systolic pressure (RVPSP), and RV/left ventricle + septum ratio (RV/LV + S); caused medial hypertrophy and adventitial widening of pulmonary arteries; induced marked focal/diffuse fibrosis with diffuse infiltration of inflammatory (ED1+) cells; and resulted in 30% mortality). OVX exacerbated the disease and increased mortality (to 75%); whereas 2ME tended to reduce mortality (55.5%) and in surviving animals reduced RVPSP and RV/LV + S ratio, and attenuated vascular remodeling, pulmonary inflammation and fibrosis. This study suggests that 2ME may have protective effects in bleomycin-induced PH and fibrosis. Further investigation of 2ME in pulmonary fibrosis and PH is warranted.


Assuntos
Estradiol/análogos & derivados , Estrogênios/deficiência , Hipertensão Pulmonar/tratamento farmacológico , Fibrose Pulmonar/tratamento farmacológico , 2-Metoxiestradiol , Análise de Variância , Animais , Antimitóticos/administração & dosagem , Antimitóticos/farmacologia , Arteríolas/efeitos dos fármacos , Arteríolas/patologia , Bleomicina , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Estradiol/administração & dosagem , Estradiol/farmacologia , Feminino , Fibroblastos/efeitos dos fármacos , Humanos , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/mortalidade , Pulmão/irrigação sanguínea , Pulmão/patologia , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Tamanho do Órgão/efeitos dos fármacos , Ovariectomia , Artéria Pulmonar/citologia , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/mortalidade , Fibrose Pulmonar/patologia , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
14.
Med Hypotheses ; 72(6): 713-9, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19237250

RESUMO

Hemolysis-associated pulmonary hypertension (HA-PH) is a serious clinical complication of various hemolytic disorders, and pulmonary hypertension (PH) is considered the greatest risk factor for death in patients with a hemolytic disorder. It is now well established that hemolysis causes the release of soluble hemoglobin and arginase from injured erythrocytes into plasma. This leads to nitric oxide (NO) deficiency, oxidative stress and a state of endothelial dysfunction that is associated with clinical development of PH. We challenge this concept and propose that in addition to the NO-arginase pathway, the adenosine deaminase-adenosine pathway plays a significant role in HA-PH and that modulation of this pathway may offer protective/therapeutic effects in HA-PH. Our preliminary data suggest that in HA-PH adenosine deaminase (ADA) is released from injured erythrocytes into plasma and that metabolic conversion of adenosine (ADO) to inosine by ADA reduces extracellular ADO levels. Adenosine, mainly via activation of adenosine A(2A) receptors, mediates a number of biological responses that may reduce hemolysis-induced vasculopathy and the risk of PH. Hypoxia is the strongest stimulus for ADO synthesis, and this increased ADO production counteracts some of the tissue/vascular injury caused by hypoxia itself. Unfortunately, under hypoxic conditions (anemia, vasoconstriction, and vaso-occlusion) in HA-PH, this "ADO negative-feed back" is abolished and the vascular protective effects of ADO are severely diminished by ADA released from injured erythrocytes. We hypothesize that in hemolytic anemia the repetitive release of ADA increases the risk for vaso-occlusive events and PH. We also propose that increase in extracellular ADO levels or activation of adenosine A(2A) receptors attenuates HA-PH, and we suggest further preclinical and clinical investigation of ADA inhibitors and ADO agonists in HA-PH.


Assuntos
Adenosina Desaminase/metabolismo , Adenosina/metabolismo , Hemólise , Hipertensão Pulmonar/metabolismo , Modelos Biológicos , Transdução de Sinais , Humanos
15.
Vascul Pharmacol ; 48(4-6): 174-83, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18373958

RESUMO

Our previous studies show that 2-methoxyestradiol, a non-estrogenic metabolite of estradiol (E2), attenuates the development and retards the progression of pulmonary hypertension (PH) in male rats, and in female rats prevents the exacerbation of PH and eliminates mortality due to ovariectomy. Recent studies suggest that 2-ethoxyestradiol (2-EE), a synthetic analog of 2-ME, is an even more potent antimitogen than 2-ME. The goals of this study were: 1) to compare the effects of E2, 2-ME and 2-EE on proliferation of human pulmonary artery endothelial (hPAEC) and smooth muscle cells (hPASMC) and lung fibroblasts (hLF); 2) to examine the effects of 2-ME, its metabolic precursor 2-hydroxyestradiol (2-HE) and 2-EE on isoproterenol (ISO)-induced cardiac hypertrophy in male rats; and 3) to investigate in male rats the effects of 2-EE (10 mug/kg/h via osmotic pump) on the development of monocrotaline (MCT; 60 mg/kg i.p.)-induced PH. E2 had biphasic effects on growth (stimulation at low and inhibition at high concentrations) in hPAEC and mild growth inhibitory effects in hPASMC and hLF (1-10 muM). In contrast, in all three pulmonary cell lines, 2-ME and 2-EE inhibited cell growth with 2-EE being ten times more potent than 2-ME. In ISO-induced cardiac hypertrophy, 2-ME, 2-HE and 2-EE similarly reduced (~-50%) left (LV) and right (RV) ventricular hypertrophy and fibrosis (hydroxyproline content). In animals with MCT-induced PH, treatment with 2-EE for 28 days significantly decreased the elevated RV peak systolic pressure and reduced RV/LV+septum weight ratio, strongly inhibited vascular remodeling (media hypertrophy and adventitia widening), markedly reduced inflammatory responses, and eliminated MCT-induced (63%) mortality. This study provides the first evidence that 2-ethoxyestradiol strongly inhibits vascular remodeling in PH and suggests that anti-proliferative agents, including synthetic analogs of estradiol metabolites may be protective in PH.


Assuntos
Antimitóticos , Estradiol/análogos & derivados , Hipertensão Pulmonar/tratamento farmacológico , Monocrotalina , Doenças Vasculares/tratamento farmacológico , Agonistas Adrenérgicos beta , Animais , Pressão Sanguínea/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Estradiol/farmacologia , Fibroblastos/efeitos dos fármacos , Humanos , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/fisiopatologia , Hipertrofia Ventricular Esquerda/induzido quimicamente , Hipertrofia Ventricular Esquerda/tratamento farmacológico , Hipertrofia Ventricular Esquerda/patologia , Hipertrofia Ventricular Direita/induzido quimicamente , Hipertrofia Ventricular Direita/tratamento farmacológico , Hipertrofia Ventricular Direita/patologia , Isoproterenol , Masculino , Miócitos de Músculo Liso/efeitos dos fármacos , Tamanho do Órgão/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Análise de Sobrevida , Doenças Vasculares/induzido quimicamente , Doenças Vasculares/fisiopatologia
16.
Clin Exp Pharmacol Physiol ; 35(5-6): 594-600, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18177483

RESUMO

1. Because chronic activation of the renal sympathetic nervous system promotes sodium and water retention, it is conceivable that long-term exposure of the kidney to the sympathetic neurotransmitter noradrenaline upregulates the expression of key renal epithelial transport systems. 2. To test this hypothesis, we used immunoblotting of renal cortical and medullary tissue to investigate the abundance of major transport systems expressed along the renal tubule in response to long-term (15 days) infusions of noradrenaline (600 ng/min) in rats. 3. Mean arterial blood pressure and heart rate were significantly elevated in rats receiving chronic infusions of noradrenaline (128 +/- 10 mmHg and 492 +/- 16 b.p.m., respectively) compared with animals treated with saline only (89 +/- 3 mmHg and 376 +/- 14 b.p.m., respectively). 4. Chronic infusions of noradrenaline also increased the protein abundance of the cortical Na(+)/H(+) exchanger isoform 3 (NHE-3; 2.5-fold; P = 0.0142), the cortical sodium-bicarbonate cotransporter NBC-1 (2.5-fold; P = 0.0067), the bumetanide-sensitive sodium-potassium-chloride cotransporter BSC-1/NKCC2 in the inner stripe of outer medulla (threefold; P = 0.0020) and aquaporin-2 in the inner medulla (twofold; P = 0.0039). 5. In contrast, noradrenaline did not significantly affect expression of the thiazide-sensitive Na(+)-Cl(-) cotransporter in the cortex, Na(+)/K(+)-ATPase-alpha(1) in the cortex and inner stripe of the outer or inner medulla, the inwardly rectifying K(+) channel (ROMK-1) in the inner stripe of the outer medulla or aquaporin-1 in the cortex or inner medulla. Noradrenaline did significantly, but modestly (less than twofold), increase aquaporin-1 in the inner stripe of the outer medulla. 6. We conclude that noradrenaline-induced increases in the expression of NHE-3, NBC-1, BSC-1 and aquaporin-2 are likely to play an important role in the regulation of salt and water transport by noradrenaline in the kidney and may explain, at least in part, the altered renal sodium and water handling associated with overactivation of the sympathetic system.


Assuntos
Aquaporina 2/biossíntese , Rim/efeitos dos fármacos , Norepinefrina/farmacologia , Simportadores de Sódio-Bicarbonato/biossíntese , Trocadores de Sódio-Hidrogênio/biossíntese , Simportadores de Cloreto de Sódio-Potássio/biossíntese , Regulação para Cima/efeitos dos fármacos , Animais , Pressão Sanguínea/efeitos dos fármacos , Esquema de Medicação , Frequência Cardíaca/efeitos dos fármacos , Rim/metabolismo , Masculino , Norepinefrina/administração & dosagem , Canais de Potássio Corretores do Fluxo de Internalização/biossíntese , Ratos , Ratos Sprague-Dawley , Receptores de Droga/biossíntese , Simportadores de Cloreto de Sódio/biossíntese , Trocador 3 de Sódio-Hidrogênio , ATPase Trocadora de Sódio-Potássio/biossíntese , Membro 1 da Família 12 de Carreador de Soluto , Organismos Livres de Patógenos Específicos
17.
Ren Fail ; 29(7): 891-902, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17994459

RESUMO

Our previous studies indicate that prolonged caffeine consumption exacerbates renal failure in nephropathy associated with the metabolic syndrome. Reduced activity of the antioxidant defense system and beneficial effects of antioxidant therapy have been reported in diabetic rats and humans. The purpose of this study was to examine the early renal effects of caffeine consumption and the effects of concomitant antioxidant therapy in young obese, diabetic ZSF1 rats. Eleven-week-old male ZSF1 rats were randomized to drink tap water, caffeine (0.1%), tempol (1 mmol/L), or a solution containing caffeine and tempol for nine weeks. Caffeine significantly reduced body weight and glycosuria (weeks 2-9), improved glucose tolerance (week 9), had no effect on elevated plasma triglycerides, plasma cholesterol (week 9) and blood pressure (week 9), and significantly increased plasma cholesterol level (weeks 5 and 9). Yet, as early as after two weeks, caffeine greatly augmented proteinuria and increased renal vascular resistance (RVR) and heart rate (HR: week 9). Tempol had no effects on metabolic status and development of proteinuria, did not alter caffeine-induced metabolic changes and early proteinuria, and attenuated caffeine-induced increase in HR and RVR. Immunohistochemical analysis revealed significant glomerular and interstitial inflammation, proliferation, and fibrosis in control animals. Caffeine augmented the influx of glomerular and interstitial macrophages (ED1+ cells) influx, glomerular and tubular proliferative response, and glomerular collagen IV content. Tempol abolished the exacerbation of renal inflammation, proliferation, and fibrosis induced by caffeine. In conclusion, in nephropathy associated with the metabolic syndrome, caffeine--most likely through the interaction with adenosine receptors and interference with anti-inflammatory and/or glomerular hemodynamic effects of adenosine--augments proteinuria and stimulates some of the key proliferative mechanisms involved in glomerular remodeling and sclerosis. Tempol does not prevent early renal injury (i.e., proteinuria) induced by caffeine, yet abolishes late renal inflammatory, proliferative, and fibrotic change induced by chronic caffeine consumption in obese ZSF1 rats.


Assuntos
Cafeína/toxicidade , Nefropatias Diabéticas/induzido quimicamente , Rim/efeitos dos fármacos , Obesidade/complicações , Animais , Peso Corporal/efeitos dos fármacos , Colágeno Tipo IV/análise , Óxidos N-Cíclicos/farmacologia , Hemodinâmica/efeitos dos fármacos , Rim/patologia , Lipídeos/sangue , Masculino , Antígeno Nuclear de Célula em Proliferação/análise , Ratos , Receptor A2A de Adenosina/efeitos dos fármacos , Receptor A2A de Adenosina/fisiologia , Receptor A2B de Adenosina/efeitos dos fármacos , Receptor A2B de Adenosina/fisiologia , Renina/sangue , Marcadores de Spin
18.
Transpl Int ; 20(3): 277-90, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17291221

RESUMO

We developed methods for prolonged (12 h), sterile, normothermic perfusion of rat kidneys and screened compounds for renal preservation including: mitochondrial transition pore inhibitor (decylubiquinone); caspase inhibitor (Z-VAD); peroxisome proliferator-activated receptor-alpha (PPARalpha) agonists (gemfibrozil, WY-14643); antioxidants (trolox, luteolin, quercetin); growth factors (HGF, PDGF, EGF, IGF-1, VEGF, transferrin); calpain inhibitor (Z-Val-Phe-CHO); calmodulin inhibitor (W7); K(ATP) opener (minoxidil, minoxidil sulfate); PARP inhibitor (3-aminobenzamide); calcium channel blocker (verapamil); V(2) agonist (DDAVP); diuretics (acetazolamide, hydrochlorothiazide, furosemide, mannitol); peroxisome proliferator-activated receptor-beta agonist (L-165041); dopamine agonist (dopamine); essential fatty acid (linolenic acid); beta-NAD; urea; uric acid; and aldosterone. In pilot studies, only PPARalpha agonists and mannitol provided promising results. Accordingly, these agents were investigated further. Fifteen rat kidneys were perfused for 12 h with L-15 media at 37 degrees C in the absence or presence of mannitol, gemfibrozil, gemfibrozil + mannitol or WY-14643. Chronic perfusion in untreated kidneys caused destruction of glomerular and tubular architecture (light and electron microscopy), disappearance of Na(+)-K(+)-ATPase-alpha(1) (Western blotting), and apoptosis (Apoptag staining). Gemfibrozil and WY-14643 marginally improved some biomarkers of renal preservation. However, the combination of gemfibrozil with mannitol markedly improved all parameters of renal preservation. We conclude that PPARalpha agonists, particularly when combined with mannitol, protect organs from normothermic, perfusion-induced damage.


Assuntos
Genfibrozila/farmacologia , Rim/fisiologia , Manitol/farmacologia , Preservação de Órgãos/métodos , PPAR alfa/agonistas , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Interações Medicamentosas , Técnicas In Vitro , Rim/efeitos dos fármacos , Rim/patologia , Rim/ultraestrutura , Masculino , Microscopia Eletrônica , Perfusão , Ratos , Ratos Sprague-Dawley , ATPase Trocadora de Sódio-Potássio/análise
19.
J Pharmacol Exp Ther ; 321(2): 799-809, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17314200

RESUMO

Stimulation of adenylyl cyclase causes cellular efflux of cAMP, and cAMP (unlike adenosine) is stable in blood. Therefore, it is conceivable that cAMP could function as a circulating adenosine prohormone by local target-organ conversion of distally released cAMP to adenosine via the sequential actions of ectophosphodiesterase and ecto-5'-nucleotidase (cAMP==> AMP==> adenosine; called the cAMP-adenosine pathway). A possible specific representation of this general concept is the pancreatohepatorenal cAMP-adenosine mechanism. The pancreas secretes glucagon into the portal circulation, and glucagon is a stimulant of hepatic adenylyl cyclase. Therefore, we hypothesize that the pancreas, via glucagon, stimulates hepatic cAMP production, which provides circulating cAMP for conversion to adenosine in the kidney via the cAMP-adenosine pathway. In normal rats, intravenous cAMP increased urinary and renal interstitial (assessed by renal microdialysis) cAMP and adenosine. Intraportal infusions of glucagon increased plasma cAMP 10-fold, it did not affect plasma adenosine, and it increased urinary and renal interstitial cAMP and adenosine. Local renal interstitial blockade (by adding inhibitors directly to the microdialysis perfusate) of ectophosphodiesterase (using 3-isobutyl-1-methylxanthine or 1,3-dipropyl-8-p-sulfophenylxanthine) or ecto-5'-nucleotidase (using alpha,beta-methyleneadenosine-5'-diphosphate) prevented the cAMP-induced and glucagon-induced increases in renal interstitial adenosine, but not cAMP. In ZSF1 rats with the metabolic syndrome, an oral glucose load increased plasma glucagon and urinary cAMP and adenosine excretion. We conclude that circulating cAMP is a substrate for local conversion to adenosine via the cAMP-adenosine pathway. A specific manifestation of this is the pancreatohepatorenal cAMP-adenosine mechanism (pancreas==> portal glucagon==> liver==> circulating cAMP==> kidney==> local cAMP-adenosine pathway).


Assuntos
Adenosina/urina , AMP Cíclico/metabolismo , Rim/metabolismo , Fígado/metabolismo , Pâncreas/fisiologia , Monofosfato de Adenosina/urina , Animais , AMP Cíclico/administração & dosagem , Glucagon/farmacologia , Glucose/farmacologia , Inosina/urina , Masculino , Microdiálise , Probenecid/farmacologia , Ratos
20.
J Cardiovasc Pharmacol ; 49(1): 56-63, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17261964

RESUMO

The metabolic syndrome is a main cause for cardiovascular disease and for the accelerating epidemic of chronic renal failure. Previous studies show that 2-hydroxyestradiol (2-HE), an estradiol metabolite with little estrogenic activity, decreases obesity and arterial blood pressure and attenuates the development of renal disease in young, obese, diabetic ZSF1 rats. In humans, however, diabetic renal disease is more frequent and severe in older patients. In vivo, 2-HE is readily converted to 2-methoxyestradiol (2-ME), an estradiol metabolite with no estrogenic activity. Accordingly, one purpose of this study was to determine whether 2-ME would provide benefit in aged rats with a very severe form of diabetic renal disease. Another objective was to determine whether synthetic analogs of estradiol metabolites might be beneficial in diabetic renal disease. To achieve these objectives we examined the effects of 2-ME and its analog 2-ethoxyestradiol (2-EE) in aged (35-week-old), obese ZSF1 rats. Animals were treated for 9 weeks with vehicle (PEG-400, 0.5 microL per hour), 2-ME or 2-EE (18 microg/kg per hour). Metabolic and renal function were measured at weeks 0, 3, 6, and 9, and renal hemodynamics and excretory function were assessed at week 9. Aged ZSF1 rats had elevated levels of glycosylated hemoglobin; increased renal cortical expression of proliferating cell nuclear antigen (PCNA), nuclear factor kappa B (NF-kappaB), and vascular endothelial growth factor (VEGF); glycosuria, hypertension; and proteinuria. 2-ME and 2-EE did not affect obesity or hypertension and had variable effects on glucose homeostasis, yet they attenuated proteinuria; increased renal blood flow and glomerular filtration; and reduced renal cortical expression of PCNA, NFkappaB, and VEGF. We conclude that 2ME and 2EE are strikingly renoprotective even in aged animals with severe diabetic renal disease. The present study warrants further investigation of 2-ME and analogs of estradiol metabolites for treatment of kidney disease associated with the metabolic syndrome.


Assuntos
Envelhecimento/efeitos dos fármacos , Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/etiologia , Estradiol/análogos & derivados , Obesidade/tratamento farmacológico , 2-Metoxiestradiol , Animais , Diabetes Mellitus/tratamento farmacológico , Diabetes Mellitus/fisiopatologia , Nefropatias Diabéticas/fisiopatologia , Progressão da Doença , Estradiol/farmacologia , Masculino , Ratos , Moduladores de Tubulina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA