Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Pharmaceutics ; 14(12)2022 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-36559167

RESUMEN

AIM: The influence of the physiochemical properties of dendrimer nanoparticles on cardiac contractility and hemodynamics are not known. Herein, we investigated (a) the effect of polyamidoamine (PAMAM) dendrimer generation (G7, G6, G5, G4 and G3) and surface chemistry (-NH2, -COOH and -OH) on cardiac function in mammalian hearts following ischemia-reperfusion (I/R) injury, and (b) determined if any PAMAM-induced cardiotoxicity could be mitigated by Angiotensin-(1-7) (Ang-(1-7), a cardioprotective agent. METHODS: Hearts isolated from male Wistar rats underwent regional I/R and/or treatment with different PAMAM dendrimers, Ang-(1-7) or its MAS receptors antagonists. Thirty minutes of regional ischemia through ligation of the left anterior descending coronary artery was followed by 30 min of reperfusion. All treatments were initiated 5 min prior to reperfusion and maintained during the first 10 min of reperfusion. Cardiac function parameters for left ventricular contractility, hemodynamics and vascular dynamics data were acquired digitally, whereas cardiac enzymes and infarct size were used as measures of cardiac injury. RESULTS: Treatment of isolated hearts with increasing doses of G7 PAMAM dendrimer progressively exacerbated recovery of cardiac contractility and hemodynamic parameters post-I/R injury. Impairment of cardiac function was progressively less on decreasing dendrimer generation with G3 exhibiting little or no cardiotoxicity. Cationic PAMAMs (-NH2) were more toxic than anionic (-COOH), with neutral PAMAMs (-OH) exhibiting the least cardiotoxicity. Cationic G7 PAMAM-induced cardiac dysfunction was significantly reversed by Ang-(1-7) administration. These cardioprotective effects of Ang-(1-7) were significantly revoked by administration of the MAS receptor antagonists, A779 and D-Pro7-Ang-(1-7). CONCLUSIONS: PAMAM dendrimers can impair the recovery of hearts from I/R injury in a dose-, dendrimer-generation-(size) and surface-charge dependent manner. Importantly, PAMAM-induced cardiotoxicity could be mitigated by Ang-(1-7) acting through its MAS receptor. Thus, this study highlights the activation of Ang-(1-7)/Mas receptor axis as a novel strategy to overcome dendrimer-induced cardiotoxicity.

2.
Front Pharmacol ; 12: 701390, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34408653

RESUMEN

Diabetes mellitus is a major debilitating disease whose global incidence is progressively increasing with currently over 463 million adult sufferers and this figure will likely reach over 700 million by the year 2045. It is the complications of diabetes such as cardiovascular, renal, neuronal and ocular dysfunction that lead to increased patient morbidity and mortality. Of these, cardiovascular complications that can result in stroke and cardiomyopathies are 2- to 5-fold more likely in diabetes but the underlying mechanisms involved in their development are not fully understood. Emerging research suggests that members of the Epidermal Growth Factor Receptor (EGFR/ErbB/HER) family of tyrosine kinases can have a dual role in that they are beneficially required for normal development and physiological functioning of the cardiovascular system (CVS) as well as in salvage pathways following acute cardiac ischemia/reperfusion injury but their chronic dysregulation may also be intricately involved in mediating diabetes-induced cardiovascular pathologies. Here we review the evidence for EGFR/ErbB/HER receptors in mediating these dual roles in the CVS and also discuss their potential interplay with the Renin-Angiotensin-Aldosterone System heptapeptide, Angiotensin-(1-7), as well the arachidonic acid metabolite, 20-HETE (20-hydroxy-5, 8, 11, 14-eicosatetraenoic acid). A greater understanding of the multi-faceted roles of EGFR/ErbB/HER family of tyrosine kinases and their interplay with other key modulators of cardiovascular function could facilitate the development of novel therapeutic strategies for treating diabetes-induced cardiovascular complications.

3.
Adv Drug Deliv Rev ; 178: 113908, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34390777

RESUMEN

Drug delivery systems or vectors are usually needed to improve the bioavailability and effectiveness of a drug through improving its pharmacokinetics/pharmacodynamics at an organ, tissue or cellular level. However, emerging technologies with sensitive readouts as well as a greater understanding of physiological/biological systems have revealed that polymeric drug delivery systems are not biologically inert but can have innate or intrinsic biological actions. In this article, we review the emerging multiple innate biological/toxicological properties of naked polyamidoamine (PAMAM) dendrimer delivery systems in the absence of any drug cargo and discuss their correlation with the defined physicochemical properties of PAMAMs in terms of molecular size (generation), architecture, surface charge and chemistry. Further, we assess whether any of the reported intrinsic biological actions of PAMAMs such as their antimicrobial activity or their ability to sequester glucose and modulate key protein interactions or cell signaling pathways, can be exploited clinically such as in the treatment of diabetes and its complications.


Asunto(s)
Dendrímeros/metabolismo , Sistema de Administración de Fármacos con Nanopartículas/química , Disponibilidad Biológica , Dendrímeros/química , Humanos , Tamaño de la Partícula
4.
J Drug Target ; 28(7-8): 683-699, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32700580

RESUMEN

The COVID-19 pandemic is caused by the severe acute-respiratory-syndrome-coronavirus-2 that uses ACE2 as its receptor. Drugs that raise serum/tissue ACE2 levels include ACE inhibitors (ACEIs) and angiotensin-II receptor blockers (ARBs) that are commonly used in patients with hypertension, cardiovascular disease and/or diabetes. These comorbidities have adverse outcomes in COVID-19 patients that might result from pharmacotherapy. Increasing ACE2 could potentially increase the risk of infection, severity or mortality in COVID-19 or it might be protective as it forms angiotensin-(1-7) which exhibits anti-inflammatory/anti-oxidative effects and prevents diabetes- and/or hypertension-induced end-organ damage. Thus, there existed clinical uncertainty. Here, we review studies implicating 15 classes of drugs in increasing ACE2 levels in vivo and the available literature on the clinical safety of these drugs in COVID-19 patients. Further, in a re-analysis of clinical data from a meta-analysis of 9 studies, we show that ACEIs/ARBs usage was not associated with an increased risk of all-cause mortality. Literature suggests that ACEIs/ARBs usage generally appears to be clinically safe though their use in severe COVID-19 patients might increase the risk of acute renal injury. For definitive clarity, further clinical and mechanistic studies are needed in assessing the safety of all classes of ACE2 raising medications.


Asunto(s)
Infecciones por Coronavirus/tratamiento farmacológico , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/tratamiento farmacológico , Antagonistas de Receptores de Angiotensina/efectos adversos , Antagonistas de Receptores de Angiotensina/farmacología , Enzima Convertidora de Angiotensina 2 , Inhibidores de la Enzima Convertidora de Angiotensina/efectos adversos , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Betacoronavirus/aislamiento & purificación , COVID-19 , Enfermedades Cardiovasculares/complicaciones , Enfermedades Cardiovasculares/tratamiento farmacológico , Infecciones por Coronavirus/mortalidad , Infecciones por Coronavirus/fisiopatología , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/fisiopatología , Humanos , Pandemias , Peptidil-Dipeptidasa A/efectos de los fármacos , Neumonía Viral/mortalidad , Neumonía Viral/fisiopatología , Factores de Riesgo , SARS-CoV-2 , Tratamiento Farmacológico de COVID-19
5.
Int J Nanomedicine ; 15: 4393-4405, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32606684

RESUMEN

AIM: The effects of polyamidoamine (PAMAM) dendrimers on the mammalian heart are not completely understood. In this study, we have investigated the effects of a sixth-generation cationic dendrimer (G6 PAMAM) on cardiac function in control and diabetic rat hearts following ischemia-reperfusion (I/R) injury. METHODS: Isolated hearts from healthy non-diabetic (Ctr) male Wistar rats were subjected to ischemia and reperfusion (I/R). LV contractility and hemodynamics data were computed digitally whereas cardiac damage following I/R injury was assessed by measuring cardiac enzymes. For ex vivo acute exposure experiments, G6 PAMAM was administered during the first 10 mins of reperfusion in Ctr animals. In chronic in vivo studies, nondiabetic rats (Ctr) received either vehicle or daily i.p. injections of G6 PAMAM (40 mg/kg) for 4 weeks. Diabetic (D) animals received either vehicle or daily i.p. injections of G6 PAMAM (10, 20 or 40 mg/kg) for 4 weeks. The impact of G6 PAMAM on pacing-postconditioning (PPC) was also studied in Ctr and D rats. RESULTS: In ex vivo studies, acute administration of G6 PAMAM to isolated Ctr hearts during reperfusion dose-dependently impaired recovery of cardiac hemodynamics and vascular dynamics parameters following I/R injury. Chronic daily i.p. injections of G6 PAMAM significantly (P<0.01) impaired recovery of cardiac function following I/R injury in nondiabetic animals but this was not generally observed in diabetic animals except for CF which was impaired by about 50%. G6 PAMAM treatment completely blocked the protective effects of PPC in the Ctr animals. CONCLUSION: Acute ex vivo or chronic in vivo treatment with naked G6 PAMAM dendrimer can significantly compromise recovery of non-diabetic hearts from I/R injury and can further negate the beneficial effects of PPC. Our findings are therefore extremely important in the nanotoxicological evaluation of G6 PAMAM dendrimers for potential clinical applications in physiological and pathological settings.


Asunto(s)
Dendrímeros/toxicidad , Corazón/fisiopatología , Mamíferos/fisiología , Daño por Reperfusión Miocárdica/fisiopatología , Nanopartículas/toxicidad , Poliaminas/administración & dosificación , Recuperación de la Función/efectos de los fármacos , Animales , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/fisiopatología , Corazón/efectos de los fármacos , Hemodinámica/efectos de los fármacos , Poscondicionamiento Isquémico , Masculino , Contracción Miocárdica/efectos de los fármacos , Miocardio/enzimología , Ratas Wistar
6.
PLoS One ; 14(11): e0224163, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31675376

RESUMEN

The angiotensin-(1-7) [Ang-(1-7)]/MAS1 receptor signaling axis is a key endogenous anti-inflammatory signaling pathway. However, the mechanisms by which its mediates the anti-inflammatory effects are not completely understood. Using an allergic murine model of asthma, we investigated whether Ang-1(1-7)/MAS1 receptor axis a): inhibits allergic inflammation via modulation of Src-dependent transactivation of the epidermal growth factor receptor (EGFR) and downstream signaling effectors such as ERK1/2, and b): directly inhibits neutrophil and/or eosinophil chemotaxis ex vivo. Ovalbumin (OVA)-induced allergic inflammation resulted in increased phosphorylation of Src kinase, EGFR, and ERK1/2. In addition, OVA challenge increased airway cellular influx, perivascular and peribronchial inflammation, fibrosis, goblet cell hyper/metaplasia and airway hyperresponsiveness (AHR). Treatment with Ang-(1-7) inhibited phosphorylation of Src kinase, EGFR, ERK1/2, the cellular and histopathological changes and AHR. Ang-(1-7) treatment also inhibited neutrophil and eosinophil chemotaxis ex vivo. These changes were reversed following pre-treatment with A779. These data show that the anti-inflammatory actions of Ang-(1-7)/ MAS1 receptor axis are mediated, at least in part, via inhibition of Src-dependent transactivation of EGFR and downstream signaling molecules such as ERK1/2. This study therefore shows that inhibition of the Src/EGRF/ERK1/2 dependent signaling pathway is one of the mechanisms by which the Ang-(1-7)/ MAS1 receptor axis mediates it anti-inflammatory effects in diseases such as asthma.


Asunto(s)
Angiotensina I/metabolismo , Asma/metabolismo , Receptores ErbB/metabolismo , Fragmentos de Péptidos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Hipersensibilidad Respiratoria/metabolismo , Transducción de Señal , Familia-src Quinasas/metabolismo , Animales , Western Blotting , Líquido del Lavado Bronquioalveolar/citología , Quimiotaxis de Leucocito , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Proto-Oncogenes Mas , Hipersensibilidad Respiratoria/patología
7.
Nanomedicine ; 18: 78-89, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30844576

RESUMEN

We investigated whether chronic administration of nano-sized polyamidoamine (PAMAM) dendrimers can have beneficial effects on diabetes-induced vascular dysfunction by inhibiting the epidermal growth factor receptor (EGFR)-ERK1/2-Rho kinase (ROCK)-a pathway known to be critical in the development of diabetic vascular complications. Daily administration of naked PAMAMs for up to 4 weeks to streptozotocin-induced diabetic male Wistar rats inhibited EGFR-ERK1/2-ROCK signaling and improved diabetes-induced vascular remodeling and dysfunction in a dose, generation (G6 > G5) and surface chemistry-dependent manner (cationic > anionic > neutral). PAMAMs, AG1478 (a selective EGFR inhibitor), or anti-EGFR siRNA also inhibited vascular EGFR-ERK1/2-ROCK signaling in vitro. These data showed that naked PAMAM dendrimers have the propensity to modulate key (e.g. EGFR) cell signaling cascades with associated pharmacological consequences in vivo that are dependent on their physicochemical properties. Thus, PAMAMs, alone or in combination with vasculoprotective agents, may have a beneficial role in the potential treatment of diabetes-induced vascular complications.


Asunto(s)
Dendrímeros/administración & dosificación , Diabetes Mellitus Experimental/fisiopatología , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Nanopartículas/administración & dosificación , Transducción de Señal , Remodelación Vascular , Quinasas Asociadas a rho/metabolismo , Animales , Glucemia/metabolismo , Peso Corporal , Dendrímeros/química , Diabetes Mellitus Experimental/sangre , Glucosa/toxicidad , Masculino , Arterias Mesentéricas/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Nanopartículas/química , Norepinefrina/farmacología , Tamaño de la Partícula , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Remodelación Vascular/efectos de los fármacos , Vasoconstricción/efectos de los fármacos
8.
Sci Rep ; 7(1): 9919, 2017 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-28855674

RESUMEN

The molecular mechanisms underlying asthma pathogenesis are poorly characterized. In this study, we investigated (1) whether Src mediates epidermal growth factor receptor (EGFR) transactivation; (2) if ERK1/2, PI3Kδ/Akt and NF-κB are signaling effectors downstream of Src/EGFR activation; and (3) if upstream inhibition of Src/EGFR is more effective in downregulating the allergic inflammation than selective inhibition of downstream signaling pathways. Allergic inflammation resulted in increased phosphorylation of EGFR, Akt, ERK1/2 and IκB in the lung tissues from ovalbumin (OVA)-challenged BALB/c mice. Treatment with inhibitors of Src (SU6656) or EGFR (AG1478) reduced EGFR phosphorylation and downstream signaling which resulted in the inhibition of the OVA-induced inflammatory cell influx in bronchoalveolar lavage fluid (BALF), perivascular and peribronchial inflammation, fibrosis, goblet cell hyper/metaplasia and airway hyper-responsiveness. Treatment with pathway-selective inhibitors for ERK1/2 (PD89059) and PI3Kδ/Akt (IC-87114) respectively, or an inhibitor of NF-κB (BAY11-7085) also reduced the OVA-induced asthmatic phenotype but to a lesser extent compared to Src/EGFR inhibition. Thus, Src via EGFR transactivation and subsequent downstream activation of multiple pathways regulates the allergic airway inflammatory response. Furthermore, a broader upstream inhibition of Src/EGFR offers an attractive therapeutic alternative in the treatment of asthma relative to selectively targeting the individual downstream signaling effectors.


Asunto(s)
Asma/genética , FN-kappa B/metabolismo , Ovalbúmina/efectos adversos , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Activación Transcripcional , Animales , Asma/inducido químicamente , Asma/tratamiento farmacológico , Asma/metabolismo , Líquido del Lavado Bronquioalveolar/inmunología , Regulación hacia Abajo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Indoles/administración & dosificación , Indoles/farmacología , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Endogámicos BALB C , Fosforilación/efectos de los fármacos , Quinazolinas/administración & dosificación , Quinazolinas/farmacología , Transducción de Señal , Sulfonamidas/administración & dosificación , Sulfonamidas/farmacología , Tirfostinos/administración & dosificación , Tirfostinos/farmacología
9.
Biomed Res Int ; 2017: 5084961, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28299329

RESUMEN

Molecular mechanisms of the beneficial effects of angiotensin-(1-7), Ang-(1-7), in diabetes-related complications, including erectile dysfunction, remain unclear. We examined the effect of diabetes and/or Ang-(1-7) treatment on vascular reactivity and cyclic guanosine monophosphate (cGMP) phosphodiesterase (PDE) in corpus cavernosum. Male Wistar rats were grouped as (1) control, (2) diabetic (streptozotocin, STZ, treated), (3) control + Ang-(1-7), and (4) diabetic + Ang-(1-7). Following 3 weeks of Ang-(1-7) treatment subsequent to induction of diabetes, rats were sacrificed. Penile cavernosal tissue was isolated to measure vascular reactivity, PDE gene expression and activity, and levels of p38MAP kinase, nitrites, and cGMP. Carbachol-induced vasorelaxant response after preincubation of corpus cavernosum with PE was significantly attenuated in diabetic rats, and Ang-(1-7) markedly corrected the diabetes-induced impairment. Gene expression and activity of PDE and p38MAP kinase were significantly increased in cavernosal tissue of diabetic rats, and Ang-(1-7) markedly attenuated STZ-induced effects. Ang-(1-7) significantly increased the levels of nitrite and cGMP in cavernosal tissue of control and diabetic rats. Cavernosal tissue of diabetic rats had significantly reduced cGMP levels and Ang-(1-7) markedly prevented the STZ-induced cGMP depletion. This study demonstrates that attenuation of diabetes-induced PDE activity might be one of the key mechanisms in the beneficial effects of Ang-(1-7).


Asunto(s)
3',5'-GMP Cíclico Fosfodiesterasas/metabolismo , Angiotensina I/metabolismo , Diabetes Mellitus Experimental/metabolismo , Pene/irrigación sanguínea , Pene/metabolismo , Fragmentos de Péptidos/metabolismo , Animales , GMP Cíclico/metabolismo , Regulación hacia Abajo , Disfunción Eréctil/metabolismo , Regulación Enzimológica de la Expresión Génica , Masculino , Nitritos/química , Ratas , Ratas Wistar , Factores de Riesgo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
10.
Peptides ; 90: 10-16, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28192151

RESUMEN

Angiotensin-(1-7) [Ang-(1-7)] exhibits blood pressure lowering actions, inhibits cell growth, and reduces tissue inflammation and fibrosis which may functionally antagonize an activated Ang II-AT1 receptor axis. Since the vascular actions of Ang-(1-7) and the associated receptor/signaling pathways vary in different vascular beds, the current study established the vasorelaxant properties of the heptapeptide in the renal artery of male Wistar male rats. Ang-(1-7) produced an endothelium-dependent vasodilator relaxation of isolated renal artery segments pre-contracted by a sub-maximal concentration of phenylephrine (PE) (3×10-7M). Ang-(1-7) induced vasodilation of the rat renal artery with an ED50 of 3±1nM and a maximal response of 42±5% (N=10). The two antagonists (10-5M each) for the AT7/Mas receptor (MasR) [D-Pro7]-Ang-(1-7) and [D-Ala7]-Ang-(1-7) significantly reduced the maximal response to 12±1% and 18±3%, respectively. Surprisingly, the AT2R receptor antagonist PD123319, the AT1R antagonist losartan and B2R antagonist HOE140 (10-6M each) also significantly reduced Ang-(1-7)-induced relaxation to 12±2%, 22±3% and 14±7%, respectively. Removal of the endothelium or addition of the soluble guanylate cyclase (sGC) inhibitor ODQ (10-5M) essentially abolished the vasorelaxant response to Ang-(1-7) (10±4% and 10±2%, P <0.05). Finally, the NOS inhibitor LNAME (10-4M) reduced the response to 13±2% (p<0.05), but the cyclooxygenase inhibitor indomethacin failed to block the Ang-(1-7) response. We conclude that Ang-(1-7) exhibits potent vasorelaxant actions in the isolated renal artery that are dependent on an intact endothelium and the apparent stimulation of a NO-sGC pathway. Moreover, Ang-(1-7)-dependent vasorelaxation was sensitive to antagonists against the AT7/Mas, AT1, AT2 and B2 receptor subtypes.


Asunto(s)
Angiotensina I/metabolismo , Fragmentos de Péptidos/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Receptores de Bradiquinina/metabolismo , Arteria Renal/metabolismo , Angiotensina I/administración & dosificación , Antagonistas de Receptores de Angiotensina/administración & dosificación , Animales , Presión Sanguínea/efectos de los fármacos , Antagonistas de los Receptores de Bradiquinina/administración & dosificación , Guanilato Ciclasa/metabolismo , Humanos , Imidazoles/administración & dosificación , Losartán/administración & dosificación , NG-Nitroarginina Metil Éster/administración & dosificación , Óxido Nítrico/metabolismo , Fragmentos de Péptidos/administración & dosificación , Fenilefrina/administración & dosificación , Proto-Oncogenes Mas , Piridinas/administración & dosificación , Ratas , Arteria Renal/efectos de los fármacos , Arteria Renal/patología , Transducción de Señal/efectos de los fármacos , Vasodilatación/genética
11.
Int J Pharm ; 514(2): 353-363, 2016 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-27032566

RESUMEN

The in vivo impact of two generation 6 cationic polyamidoamine (PAMAM) dendrimers on cellular signaling via extracellular-regulated kinase 1/2 (ERK1/2) and p38 mitogen-activated protein kinase (p38 MAPK), as well as their relationship to epidermal growth factor receptor (EGFR), were studied in the normal and/or diabetic rat kidney. A single 10mg/kg/i.p administration of Polyfect (PF; with an intact branching architecture) or Superfect (SF; with a fragmented branching architecture) modulated renal ERK1/2 and p38 MAPK phosphorylation in a dendrimer-specific and animal model-dependent manner. AG1478 treatment (a selective EGFR inhibitor) confirmed that renal ERK1/2 and p38 MAPK signaling was downstream of EGFR. Surprisingly, both PAMAMs induced hyperphosphorylation of ERK1/2 and p38 MAPK (at 1 or 5mg/kg) despite inhibiting EGFR phosphorylation in the diabetic kidney. PAMAMs did not alter renal morphology but their effects on p38 MAPK and EGFR phosphorylation were reversed by ex vivo treatment of kidneys with the anti-oxidant, Tempol. Thus, PAMAMs can intrinsically modulate signaling of mitogen-activated protein kinases (MAPKs) depending on the type of dendrimer (fragmented vs intact branching architecture) and animal model (normal vs diabetic) used and likely occurs via an EGFR-independent and oxidative-stress dependent mechanism. These findings might have important toxicological implications for PAMAM-based delivery systems.


Asunto(s)
Dendrímeros/farmacología , Diabetes Mellitus Experimental/metabolismo , Riñón/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Animales , Glucemia , Óxidos N-Cíclicos/farmacología , Diabetes Mellitus Experimental/patología , Relación Dosis-Respuesta a Droga , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Riñón/patología , Masculino , Fosforilación/efectos de los fármacos , Quinazolinas/farmacología , Ratas , Marcadores de Spin , Tirfostinos/farmacología
12.
Mol Pharm ; 13(5): 1575-86, 2016 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-26985693

RESUMEN

The effects of naked polyamidoamine (PAMAM) dendrimers on renin-angiotensin system (RAS) signaling via Angiotensin (Ang) II-mediated transactivation of the epidermal growth factor receptor (EGFR) and the closely related family member ErbB2 (HER2) were investigated. In primary aortic vascular smooth muscle cells, a cationic fifth-generation (G5) PAMAM dendrimer dose- and time-dependently inhibited Ang II/AT1 receptor-mediated transactivation of EGFR and ErbB2 as well as their downstream signaling via extracellular-regulated kinase 1/2 (ERK1/2). Inhibition even occurred at noncytotoxic concentrations at short (1 h) exposure times and was dependent on dendrimer generation (G7 > G6 > G5 > G4) and surface group chemistry (amino > carboxyl > hydroxyl). Mechanistically, the cationic G5 PAMAM dendrimer inhibited Ang II-mediated transactivation of EGFR and ErbB2 via inhibition of the nonreceptor tyrosine kinase Src. This novel, early onset, intrinsic biological action of PAMAM dendrimers as inhibitors of the Ang II/AT1/Src/EGFR-ErbB2/ERK1/2 signaling pathway could have important toxicological and pharmacological implications.


Asunto(s)
Angiotensina II/farmacología , Dendrímeros/farmacología , Receptores ErbB/metabolismo , Poliaminas/farmacología , Polímeros/farmacología , Receptor ErbB-2/metabolismo , Activación Transcripcional/efectos de los fármacos , Animales , Línea Celular , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Ratas , Receptor de Angiotensina Tipo 1/metabolismo , Transducción de Señal/efectos de los fármacos , Familia-src Quinasas/metabolismo
13.
Am J Physiol Heart Circ Physiol ; 310(1): H104-12, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26519026

RESUMEN

The aim of the present study was to investigate the role of the ANG-(1-7) receptor (Mas) and nitric oxide (NO) in pacing postconditiong (PPC)-mediated cardioprotection against ischemia-reperfusion injury. Cardiac contractility and hemodynamics were assessed using a modified Langendorff system, cardiac damage was assessed by measuring infarct size and creatinine kinase levels, and levels of phosphorylated and total endothelial NO synthase (eNOS) were determined by Western blot analysis. Isolated hearts were subjected to 30 min of regional ischemia, produced by fixed position ligation of the left anterior descending coronary artery, followed by 30 min of reperfusion (n = 6). Hearts were also subjected to PPC (three cycles of 30 s of left ventricular pacing alternated with 30 s of right atrial pacing) and/or treated during reperfusion with ANG-(1-7), N(G)-nitro-l-arginine methyl ester, or the Mas antagonist (d-Ala7)-ANG I/II (1-7). The PPC-mediated improvement in cardiac contractility and hemodyanamics, cardiac damage, and eNOS phosphorylation were significantly attenuated upon treatment with (d-Ala7)-ANG I/II (1-7) or N(G)-nitro-l-arginine methyl ester. Treatment with ANG-(1-7) improved cardiac function and reduced infarct size and creatinine kinase levels; however, the effects of ANG-(1-7) were not additive with PPC. In conclusion, these data provide novel insights into the cardioprotective mechanisms of PPC in that they involve the Mas receptor and eNOS and further suggest a potential therapeutic role for ANG-(1-7) in cardiac ischemic injury.


Asunto(s)
Angiotensina I/metabolismo , Estimulación Cardíaca Artificial/métodos , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/metabolismo , Óxido Nítrico/metabolismo , Fragmentos de Péptidos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Función Ventricular Izquierda , Angiotensina I/farmacología , Animales , Creatina Quinasa/metabolismo , Inhibidores Enzimáticos/farmacología , Hemodinámica , Preparación de Corazón Aislado , Masculino , Contracción Miocárdica , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/patología , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fragmentos de Péptidos/farmacología , Fosforilación , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/efectos de los fármacos , Ratas Wistar , Receptores Acoplados a Proteínas G/efectos de los fármacos , Transducción de Señal , Factores de Tiempo , Función Ventricular Izquierda/efectos de los fármacos
14.
PLoS One ; 10(11): e0141657, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26536590

RESUMEN

Transactivation of the epidermal growth factor receptor (EGFR or ErbB) family members, namely EGFR and ErbB2, appears important in the development of diabetes-induced vascular dysfunction. Angiotensin-(1-7) [Ang-(1-7)] can prevent the development of hyperglycemia-induced vascular complications partly through inhibiting EGFR transactivation. Here, we investigated whether Ang-(1-7) can inhibit transactivation of ErbB2 as well as other ErbB receptors in vivo and in vitro. Streptozotocin-induced diabetic rats were chronically treated with Ang-(1-7) or AG825, a selective ErbB2 inhibitor, for 4 weeks and mechanistic studies performed in the isolated mesenteric vasculature bed as well as in cultured vascular smooth muscle cells (VSMCs). Ang-(1-7) or AG825 treatment inhibited diabetes-induced phosphorylation of ErbB2 receptor at tyrosine residues Y1221/22, Y1248, Y877, as well as downstream signaling via ERK1/2, p38 MAPK, ROCK, eNOS and IkB-α in the mesenteric vascular bed. In VSMCs cultured in high glucose (25 mM), Ang-(1-7) inhibited src-dependent ErbB2 transactivation that was opposed by the selective Mas receptor antagonist, D-Pro7-Ang-(1-7). Ang-(1-7) via Mas receptor also inhibited both Angiotensin II- and noradrenaline/norephinephrine-induced transactivation of ErbB2 and/or EGFR receptors. Further, hyperglycemia-induced transactivation of ErbB3 and ErbB4 receptors could be attenuated by Ang-(1-7) that could be prevented by D-Pro7-Ang-(1-7) in VSMC. These data suggest that Ang-(1-7) via its Mas receptor acts as a pan-ErbB inhibitor and might represent a novel general mechanism by which Ang-(1-7) exerts its beneficial effects in many disease states including diabetes-induced vascular complications.


Asunto(s)
Angiotensina I/farmacología , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Receptores ErbB/genética , Regulación de la Expresión Génica/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Proteínas Proto-Oncogénicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Activación Transcripcional/efectos de los fármacos , Animales , Western Blotting , Células Cultivadas , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patología , Receptores ErbB/antagonistas & inhibidores , Glucosa/metabolismo , Masculino , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Fosforilación , Proto-Oncogenes Mas , Ratas , Ratas Wistar , Transducción de Señal , Vasodilatadores/farmacología
15.
PLoS One ; 10(7): e0132215, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26167903

RESUMEN

Cationic polyamidoamine (PAMAM) dendrimers are branch-like spherical polymers being investigated for a variety of applications in nanomedicine including nucleic acid drug delivery. Emerging evidence suggests they exhibit intrinsic biological and toxicological effects but little is known of their interactions with signal transduction pathways. We previously showed that the activated (fragmented) generation (G) 6 PAMAM dendrimer, Superfect (SF), stimulated epidermal growth factor receptor (EGFR) tyrosine kinase signaling-an important signaling cascade that regulates cell growth, survival and apoptosis- in cultured human embryonic kidney (HEK 293) cells. Here, we firstly studied the in vitro effects of Polyfect (PF), a non-activated (intact) G6 PAMAM dendrimer, on EGFR tyrosine kinase signaling via extracellular-regulated kinase 1/2 (ERK1/2) and p38 mitogen-activated protein kinase (MAPK) in cultured HEK 293 cells and then compared the in vivo effects of a single administration (10mg/kg i.p) of PF or SF on EGFR signaling in the kidneys of normal and diabetic male Wistar rats. Polyfect exhibited a dose- and time-dependent inhibition of EGFR, ERK1/2 and p38 MAPK phosphorylation in HEK-293 cells similar to AG1478, a selective EGFR inhibitor. Administration of dendrimers to non-diabetic or diabetic animals for 24h showed that PF inhibited whereas SF stimulated EGFR phosphorylation in the kidneys of both sets of animals. PF-mediated inhibition of EGFR phosphorylation as well as SF or PF-mediated apoptosis in HEK 293 cells could be significantly reversed by co-treatment with antioxidants such as tempol implying that both these effects involved an oxidative stress-dependent mechanism. These results show for the first time that SF and PF PAMAM dendrimers can differentially modulate the important EGFR signal transduction pathway in vivo and may represent a novel class of EGFR modulators. These findings could have important clinical implications for the use of PAMAM dendrimers in nanomedicine.


Asunto(s)
Dendrímeros/farmacología , Receptores ErbB/efectos de los fármacos , Poliaminas/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Técnicas In Vitro , Riñón/efectos de los fármacos , Riñón/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratas , Ratas Wistar
16.
J Drug Target ; 23(6): 506-18, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26114862

RESUMEN

The epidermal growth factor receptors, EGFR and EGFR2 (ErbB2), appear important mediators of diabetes-induced vascular dysfunction. We investigated whether targeted dual inhibition of EGFR and ErbB2 with Lapatinib would be effective in treating diabetes-induced vascular dysfunction in a rat model of type 1 diabetes. In streptozotocin-induced diabetes, chronic 4-week oral or acute, ex vivo, administration of Lapatinib prevented the development of vascular dysfunction as indicated by the attenuation of the hyper-reactivity of the diabetic mesenteric vascular bed (MVB) to norephinephrine without correcting hyperglycemia. Chronic in vivo or acute ex vivo Lapatinib treatment also significantly attenuated diabetes-induced increases in phosphorylation of EGFR, ErbB2, ERK1/2, AKT, ROCK2 and IkB-alpha as well as normalized the reduced levels of phosphorylated FOXO3A, and eNOS (Ser1177) in the diabetic MVB. Similar results were observed in vascular smooth muscle cells (VSMCs) cultured in high glucose (25 mM) treated with Lapatinib or small interfering RNA (siRNA) targeting the ErbB2 receptor. Lapatinib also prevented high glucose-induced apoptosis in VSMC. Thus, Lapatinib corrects hyperglycemia-induced apoptosis and vascular dysfunction with concomitant reversal of diabetes or high glucose-induced signaling changes in EGFR/ErbB2 and downstream signaling pathways implying that targeted dual inhibition of EGFR/ErbB2 might be an effective vasculoprotective treatment strategy in diabetic patients.


Asunto(s)
Apoptosis/efectos de los fármacos , Angiopatías Diabéticas/tratamiento farmacológico , Angiopatías Diabéticas/fisiopatología , Receptores ErbB/antagonistas & inhibidores , Terapia Molecular Dirigida/métodos , Quinazolinas/uso terapéutico , Receptor ErbB-2/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Animales , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatología , Angiopatías Diabéticas/inducido químicamente , Angiopatías Diabéticas/metabolismo , Receptores ErbB/metabolismo , Hiperglucemia/metabolismo , Hiperglucemia/patología , Lapatinib , Masculino , Músculo Liso Vascular/metabolismo , Fosforilación/efectos de los fármacos , Cultivo Primario de Células , Inhibidores de Proteínas Quinasas/uso terapéutico , Quinazolinas/farmacología , ARN Interferente Pequeño/farmacología , Ratas , Receptor ErbB-2/metabolismo
17.
J Diabetes Res ; 2014: 142154, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25309930

RESUMEN

Angiotensin-(1-7) [Ang-(1-7)] may have beneficial effects in diabetes mellitus-induced erectile dysfunction (DMIED) but its molecular actions in the diabetic corpus cavernosum (CC) are not known. We characterized the effects of diabetes and/or chronic in vivo administration of Ang-(1-7) on vascular reactivity in the rat corpus cavernosum (CC) and on protein expression levels of potential downstream effectors of the renin-angiotensin-aldosterone system (RAAS) such as angiotensin-converting enzyme (ACE), ACE2, Rho kinases 1 and 2 (ROCK1 and ROCK2), and omega-hydroxylase, the cytochrome-P450 enzyme that metabolizes arachidonic acid to form the vasoconstrictor, 20-hydroxyeicosatetraenoic acid. Streptozotocin-treated rats were chronicically administered Ang-(1-7) with or without A779, a Mas receptor antagonist, during weeks 4 to 6 of diabetes. Ang-(1-7) reversed diabetes-induced abnormal reactivity to vasoactive agents (endothelin-1, phenylepherine, and carbachol) in the CC without correcting hyperglycemia. Six weeks of diabetes led to elevated ACE, ROCK1, ROCK 2, and omega-hydroxylase and a concomitant decrease in ACE2 protein expression levels that were normalized by Ang-(1-7) treatment but not upon coadministration of A779. These data are supportive of the notion that the beneficial effects of Ang-(1-7) in DMIED involve counterregulation of diabetes-induced changes in ACE, ACE2, Rho kinases, and omega-hydroxylase proteins in the diabetic CC via a Mas receptor-dependent mechanism.


Asunto(s)
Angiotensina I/uso terapéutico , Citocromo P-450 CYP4A/metabolismo , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Pene/efectos de los fármacos , Fragmentos de Péptidos/uso terapéutico , Peptidil-Dipeptidasa A/metabolismo , Quinasas Asociadas a rho/metabolismo , Enzima Convertidora de Angiotensina 2 , Animales , Glucemia/análisis , Peso Corporal , Diabetes Mellitus Experimental/tratamiento farmacológico , Modelos Animales de Enfermedad , Disfunción Eréctil/tratamiento farmacológico , Masculino , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/metabolismo , Ratas , Ratas Wistar , Receptores Acoplados a Proteínas G/metabolismo , Estreptozocina
18.
J Diabetes Res ; 2013: 427693, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24066305

RESUMEN

AIMS: We evaluated the effects of RU28318 (RU), a selective mineralocorticoid receptor (MR) antagonist, Captopril (Capt), an angiotensin converting enzyme inhibitor, and Losartan (Los), an angiotensin receptor blocker, alone or in combination with ischemia/reperfusion- (I/R-) induced cardiac dysfunction in hearts obtained from normal and diabetic rats. METHODS: Isolated hearts were perfused for 30 min and then subjected to 30 min of global ischemia (I) followed by a period of 30 min of reperfusion (R). Drugs were administered for 30 min either before or after ischemia. Drug regimens tested were RU, Capt, Los, RU + Capt, RU + Los, Capt + Los, and RU + Capt + Los (Triple). Recovery of cardiac hemodynamics was evaluated. RESULTS: Recovery of cardiac function was up to 5-fold worse in hearts obtained from diabetic animals compared to controls. Treatment with RU was generally better in preventing or reversing ischemia-induced cardiac dysfunction in normal hearts compared to treatment with Capt or Los alone. In diabetic hearts, RU was generally similarly effective as Capt or Los treatment. CONCLUSIONS: RU treatment locally might be considered as an effective therapy or preventative measure in cardiac I/R injury. Importantly, RU was the most effective at improving -dP/dt (a measure of diastolic function) when administered to diabetic hearts after ischemia.


Asunto(s)
Antagonistas de Receptores de Angiotensina/uso terapéutico , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Captopril/uso terapéutico , Losartán/uso terapéutico , Antagonistas de Receptores de Mineralocorticoides/uso terapéutico , Isquemia Miocárdica/tratamiento farmacológico , Espironolactona/análogos & derivados , Antagonistas de Receptores de Angiotensina/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Captopril/farmacología , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/fisiopatología , Quimioterapia Combinada , Corazón/efectos de los fármacos , Corazón/fisiopatología , Losartán/farmacología , Masculino , Antagonistas de Receptores de Mineralocorticoides/farmacología , Isquemia Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/prevención & control , Ratas , Ratas Wistar , Espironolactona/farmacología , Espironolactona/uso terapéutico
19.
PLoS One ; 8(6): e67813, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23826343

RESUMEN

Diabetes mellitus leads to vascular complications but the underlying signalling mechanisms are not fully understood. Here, we examined the role of ErbB2 (HER2/Neu), a transmembrane receptor tyrosine kinase of the ErbB/EGFR (epidermal growth factor receptor) family, in mediating diabetes-induced vascular dysfunction in an experimental model of type 1 diabetes. Chronic treatment of streptozotocin-induced diabetic rats (1 mg/kg/alt diem) or acute, ex-vivo (10(-6), 10(-5) M) administration of AG825, a specific inhibitor of ErbB2, significantly corrected the diabetes-induced hyper-reactivity of the perfused mesenteric vascular bed (MVB) to the vasoconstrictor, norephinephrine (NE) and the attenuated responsiveness to the vasodilator, carbachol. Diabetes led to enhanced phosphorylation of ErbB2 at multiple tyrosine (Y) residues (Y1221/1222, Y1248 and Y877) in the MVB that could be attenuated by chronic AG825 treatment. Diabetes- or high glucose-mediated upregulation of ErbB2 phosphorylation was coupled with activation of Rho kinases (ROCKs) and ERK1/2 in MVB and in cultured vascular smooth muscle cells (VSMC) that were attenuated upon treatment with either chronic or acute AG825 or with anti-ErbB2 siRNA. ErbB2 likley heterodimerizes with EGFR, as evidenced by increased co-association in diabetic MVB, and further supported by our finding that ERK1/2 and ROCKs are common downstream effectors since their activation could also be blocked by AG1478. Our results show for the first time that ErbB2 is an upstream effector of ROCKs and ERK1/2 in mediating diabetes-induced vascular dysfunction. Thus, potential strategies aimed at modifying actions of signal transduction pathways involving ErbB2 pathway may prove to be beneficial in treatment of diabetes-induced vascular complications.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Angiopatías Diabéticas/patología , Sistema de Señalización de MAP Quinasas , Receptor ErbB-2/metabolismo , Quinasas Asociadas a rho/metabolismo , Animales , Células Cultivadas , Angiopatías Diabéticas/etiología , Angiopatías Diabéticas/metabolismo , Masculino , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Ratas , Ratas Wistar , Activación Transcripcional , Vasoconstrictores/farmacología , Vasodilatadores/farmacología
20.
Fertil Steril ; 100(1): 226-33, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23541316

RESUMEN

OBJECTIVE: To investigate diabetes mellitus (DM)-induced oxidative DNA damage, putative involvement of angiotensin (Ang) II, and possible modulatory effects of Ang-(1-7) in rat corpus cavernosum (CC). DESIGN: In vivo study. SETTING: Research laboratory. ANIMAL(S): Adult male Wistar rats. INTERVENTION(S): Streptozotocin-induced diabetic rats received either captopril, losartan (both 300 mg/L in drinking water), or Ang-(1-7) (576 µg/kg/d IP) for a 3-week period immediately before sacrifice at 6 weeks of DM. MAIN OUTCOME MEASURE(S): Histopathological changes in CC were examined in Masson's trichrome-stained tissue sections. Oxidative stress was evaluated by measuring total oxidant status and antioxidant status. The DNA damage was estimated by measuring 8-hydroxy-2'-deoxyguanosine by immunohistochemistry and ELISA. RESULT(S): The CC smooth muscle degeneration was observed in association with an increase in total oxidant status and a decrease in total antioxidant status in rats with DM. Oxidative DNA damage was significantly increased in both cytoplasm and nuclei of CC in DM. Treatment with captopril, losartan, or Ang-(1-7) inhibited these changes in rats with DM. CONCLUSION(S): The data indicate that Ang II signaling is involved in DM-induced structural changes and oxidative DNA damage in the CC and that modulation of the signaling by captopril, losartan, and Ang-(1-7) restores the effects of DM. Thus, Ang-(1-7)/MAS1 axis may be a novel therapeutic target for erectile dysfunction in DM.


Asunto(s)
Angiotensina II/fisiología , Angiotensina I/fisiología , Daño del ADN/fisiología , Diabetes Mellitus Experimental/metabolismo , Estrés Oxidativo/fisiología , Pene/fisiología , Fragmentos de Péptidos/fisiología , Angiotensina I/uso terapéutico , Animales , Diabetes Mellitus Experimental/patología , Masculino , Pene/patología , Fragmentos de Péptidos/uso terapéutico , Proto-Oncogenes Mas , Distribución Aleatoria , Ratas , Ratas Wistar , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...