Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 168
Filtrar
1.
J Am Soc Nephrol ; 31(5): 1050-1065, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32291286

RESUMEN

BACKGROUND: Kidney injury associated with cold storage is a determinant of delayed graft function and the long-term outcome of transplanted kidneys, but the underlying mechanism remains elusive. We previously reported a role of protein kinase C-δ (PKCδ) in renal tubular injury during cisplatin nephrotoxicity and albumin-associated kidney injury, but whether PKCδ is involved in ischemic or transplantation-associated kidney injury is unknown. METHODS: To investigate PKCδ's potential role in injury during cold storage-associated transplantation, we incubated rat kidney proximal tubule cells in University of Wisconsin (UW) solution at 4°C for cold storage, returning them to normal culture medium at 37°C for rewarming. We also stored kidneys from donor mice in cold UW solution for various durations, followed by transplantation into syngeneic recipient mice. RESULTS: We observed PKCδ activation in both in vitro and in vivo models of cold-storage rewarming or transplantation. In the mouse model, PKCδ was activated and accumulated in mitochondria, where it mediated phosphorylation of a mitochondrial fission protein, dynamin-related protein 1 (Drp1), at serine 616. Drp1 activation resulted in mitochondrial fission or fragmentation, accompanied by mitochondrial damage and tubular cell death. Deficiency of PKCδ in donor kidney ameliorated Drp1 phosphorylation, mitochondrial damage, tubular cell death, and kidney injury during cold storage-associated transplantation. PKCδ deficiency also improved the repair and function of the renal graft as a life-supporting kidney. An inhibitor of PKCδ, δV1-1, protected kidneys against cold storage-associated transplantation injury. CONCLUSIONS: These results indicate that PKCδ is a key mediator of mitochondrial damage and renal tubular injury in cold storage-associated transplantation and may be an effective therapeutic target for improving renal transplant outcomes.


Asunto(s)
Frío/efectos adversos , Dinaminas/metabolismo , Trasplante de Riñón , Necrosis Tubular Aguda/etiología , Túbulos Renales Proximales/enzimología , Preservación de Órganos/métodos , Proteína Quinasa C-delta/fisiología , Animales , Apoptosis , División Celular , Células Cultivadas , Activación Enzimática , Necrosis Tubular Aguda/enzimología , Túbulos Renales Proximales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/enzimología , Fosforilación , Proteína Quinasa C-delta/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional , Ratas
2.
FASEB J ; 33(3): 4513-4524, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30589566

RESUMEN

Recent studies have shown that autophagy exhibits a renoprotective role in various models of acute kidney injury (AKI). However, its role in vancomycin (Van)-induced AKI remains largely unclarified. This study was the first to indicate that autophagy was rapidly activated in both human kidney-2 cells and renal tissues, and mammalian target of rapamycin (mTOR) was inactivated via the suppression of ERK1/2 and mTOR during Van treatment. Interestingly, for both in vitro and in vivo experiments, the suppression of autophagy via chloroquine and PT-Atg7-KO significantly ameliorated Van-induced kidney injury and renal tubular cell apoptosis. Global gene expression analysis indicated that the expression levels of 6159 genes were induced by Van treatment in the kidney cortical tissues of PT-Atg7 wild-type mice, and 18 of them were notably suppressed in PT-Atg7-KO mice. These 18 genes were further classified as programmed cell death, protein binding, signal transduction, E3 ubiquitin ligase, nucleoside diphosphate kinase activity, and E1-like activating enzyme. Unexpectedly, following Van treatment, PKC-δ expression was found to be highest among the 4 genes related to cell death, which was remarkably suppressed in vitro and in PT-Atg7-KO mice. In addition, Atg7 could induce renal cell apoptosis during Van treatment via binding to PKC-δ. Likewise, the inhibition of PKCδ ameliorated Van-induced apoptosis in human kidney-2 cells and kidney tissues. Furthermore, the data showed that PT-Atg7-KO exerted a renoprotective effect against Van-induced nephrotoxicity, but this effect was lost after injection with myc-tagged PKCδ. Taken altogether, these results indicate that Van induces autophagy by suppressing the activation of the ERK1/2 and mTOR signaling pathway. In addition, Atg7 mediates Van-induced AKI through the activation of PKCδ. In sum, autophagy inhibition may serve as a novel therapeutic target for treating nephrotoxic AKI induced by Van.-Xu, X., Pan, J., Li, H., Li, X., Fang, F., Wu, D., Zhou, Y., Zheng, P., Xiong, L., Zhang, D. Atg7 mediates renal tubular cell apoptosis in vancomycin nephrotoxicity through activation of PKC-δ.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Apoptosis/fisiología , Proteína 7 Relacionada con la Autofagia/fisiología , Autofagia/fisiología , Túbulos Renales/efectos de los fármacos , Proteína Quinasa C-delta/fisiología , Vancomicina/toxicidad , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Proteína 7 Relacionada con la Autofagia/antagonistas & inhibidores , Proteína 7 Relacionada con la Autofagia/deficiencia , Proteína 7 Relacionada con la Autofagia/genética , Línea Celular , Activación Enzimática/efectos de los fármacos , Perfilación de la Expresión Génica , Ontología de Genes , Humanos , Túbulos Renales/metabolismo , Túbulos Renales/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica , Serina-Treonina Quinasas TOR/metabolismo
3.
Mol Cell ; 45(1): 25-37, 2012 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-22244330

RESUMEN

BIK protein is an initiator of mitochondrial apoptosis, and BIK expression is induced by proapoptotic signals, including DNA damage. Here, we demonstrate that 3' end processing and expression of BIK mRNA are controlled by the nuclear PI4,5P(2)-regulated poly(A) polymerase Star-PAP downstream of DNA damage. Nuclear PKCδ is a key mediator of apoptosis, and DNA damage stimulates PKCδ association with the Star-PAP complex where PKCδ is required for Star-PAP-dependent BIK expression. PKCδ binds the PI4,5P(2)-generating enzyme PIPKIα, which is essential for PKCδ interaction with the Star-PAP complex, and PKCδ activity is directly stimulated by PI4,5P(2). Features in the BIK 3' UTR uniquely define Star-PAP specificity and may block canonical PAP activity toward BIK mRNA. This reveals a nuclear phosphoinositide signaling nexus where PIPKIα, PI4,5P(2), and PKCδ regulate Star-PAP control of BIK expression and induction of apoptosis. This pathway is distinct from the Star-PAP-mediated oxidative stress pathway indicating signal-specific regulation of mRNA 3' end processing.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Apoptosis , Proteínas de la Membrana/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Polinucleotido Adenililtransferasa/fisiología , Proteína Quinasa C-delta/fisiología , Proteínas Reguladoras de la Apoptosis/metabolismo , Secuencia de Bases , Daño del ADN , Células HEK293 , Humanos , Proteínas de la Membrana/metabolismo , Proteínas Mitocondriales , Datos de Secuencia Molecular , Nucleotidiltransferasas , Fosforilación , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Polinucleotido Adenililtransferasa/genética , Polinucleotido Adenililtransferasa/metabolismo , Proteína Quinasa C-delta/metabolismo , ARN Mensajero/metabolismo , Transducción de Señal
4.
Int J Mol Sci ; 21(18)2020 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-32911704

RESUMEN

Engagement of integrin αIIbß3 promotes platelet-platelet interaction and stimulates outside-in signaling that amplifies activation. Protein kinase Cδ (PKCδ) is known to play an important role in platelet activation, but its role in outside-in signaling has not been established. In the present study, we determined the role of PKCδ and its signaling pathways in integrin αIIbß3-mediated outside-in signaling in platelets using PKCδ-deficient platelets. Platelet spreading to immobilized fibrinogen resulted in PKCδ phosphorylation, suggesting that αIIbß3 activation caused PKCδ activation. αIIbß3-mediated phosphorylation of Akt was significantly inhibited in PKCδ -/- platelets, indicating a role of PKCδ in outside-in signaling. αIIbß3-mediated PKCδ phosphorylation was inhibited by proline-rich tyrosine kinase 2 (Pyk2) selective inhibitor, suggesting that Pyk2 contributes to the regulation of PKCδ phosphorylation in outside-in signaling. Additionally, Src-family kinase inhibitor PP2 inhibited integrin-mediated Pyk2 and PKCδ phosphorylation. Lastly, platelet spreading was inhibited in PKCδ -/- platelets compared to the wild-type (WT) platelets, and clot retraction from PKCδ -/- platelets was markedly delayed, indicating that PKCδ is involved in the regulation of αIIbß3-dependent interactivities with cytoskeleton elements. Together, these results provide evidence that PKCδ plays an important role in outside-in signaling, which is regulated by Pyk2 in platelets.


Asunto(s)
Plaquetas/metabolismo , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Proteína Quinasa C-delta/metabolismo , Animales , Plaquetas/fisiología , Retracción del Coagulo/fisiología , Femenino , Fibrinógeno/metabolismo , Quinasa 2 de Adhesión Focal/metabolismo , Integrinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , Activación Plaquetaria/fisiología , Adhesividad Plaquetaria/fisiología , Agregación Plaquetaria/fisiología , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/fisiología , Proteína Quinasa C-delta/fisiología , Transducción de Señal/fisiología
5.
J Physiol ; 597(2): 481-498, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30382587

RESUMEN

KEY POINTS: Concurrent 5-HT2A (Q pathway) and 5-HT7 (S pathway) serotonin receptor activation cancels phrenic motor facilitation due to mutual cross-talk inhibition. Spinal protein kinase Cδ (PKCδ) or protein kinase A inhibition restores phrenic motor facilitation with concurrent Q and S pathway activation, demonstrating a key role for these kinases in cross-talk inhibition. Spinal PKCδ inhibition enhances adenosine-dependent severe acute intermittent hypoxia-induced phrenic long-term facilitation (S pathway), consistent with relief of cross-talk inhibition. ABSTRACT: Intermittent spinal serotonin receptor activation elicits long-lasting phrenic motor facilitation (pMF), a form of respiratory motor plasticity. When activated alone, spinal Gq protein-coupled serotonin 2A receptors (5-HT2A ) initiate pMF by a mechanism that requires ERK-MAP kinase signalling and new BDNF protein synthesis (Q pathway). Spinal Gs protein-coupled serotonin 7 (5-HT7 ) and adenosine 2A (A2A ) receptor activation also elicits pMF, but via distinct mechanisms (S pathway) that require Akt signalling and new TrkB protein synthesis. Although studies have shown inhibitory cross-talk interactions between these competing pathways, the underlying cellular mechanisms are unknown. We propose the following hypotheses: (1) concurrent 5-HT2A and 5-HT7 activation undermines pMF; (2) protein kinase A (PKA) and (3) NADPH oxidase mediate inhibitory interactions between Q (5-HT2A ) and S (5-HT7 ) pathways. Selective 5-HT2A (DOI hydrochloride) and 5HT7 (AS-19) agonists were administered intrathecally at C4 (three injections, 5-min intervals) in anaesthetized, vagotomized and ventilated male rats. With either spinal 5-HT2A or 5-HT7 activation alone, phrenic amplitude progressively increased (pMF). In contrast, concurrent 5-HT2A and 5-HT7 activation failed to elicit pMF. The 5-HT2A -induced Q pathway was restored by inhibiting PKA activity (Rp-8-Br-cAMPS). NADPH oxidase inhibition did not prevent cross-talk inhibition. Therefore, we investigated alternative mechanisms to explain Q to S pathway inhibition. Spinal protein kinase C (PKC) inhibition with Gö6983 or PKCδ peptide inhibitor restored the 5-HT7 -induced S pathway to pMF, revealing PKCδ as the relevant isoform. Spinal PKCδ inhibition enhanced the S pathway-dependent form of pMF elicited by severe acute intermittent hypoxia. We suggest that powerful constraints between 5-HT2A and 5-HT7 or A2A receptor-induced pMF are mediated by PKCδ and PKA, respectively.


Asunto(s)
Hipoxia/fisiopatología , Nervio Frénico/fisiología , Proteína Quinasa C-delta/fisiología , Receptor de Serotonina 5-HT2A/fisiología , Receptores de Serotonina/fisiología , Médula Espinal/fisiología , Anfetaminas/farmacología , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Masculino , Proteína Quinasa C-delta/antagonistas & inhibidores , Pirazoles/farmacología , Ratas Sprague-Dawley , Agonistas de Receptores de Serotonina/farmacología , Tetrahidronaftalenos/farmacología
6.
J Am Soc Nephrol ; 28(4): 1131-1144, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27799485

RESUMEN

Nephrotoxicity is a major adverse effect in cisplatin chemotherapy, and renoprotective approaches are unavailable. Recent work unveiled a critical role of protein kinase Cδ (PKCδ) in cisplatin nephrotoxicity and further demonstrated that inhibition of PKCδ not only protects kidneys but enhances the chemotherapeutic effect of cisplatin in tumors; however, the underlying mechanisms remain elusive. Here, we show that cisplatin induced rapid activation of autophagy in cultured kidney tubular cells and in the kidneys of injected mice. Cisplatin also induced the phosphorylation of mammalian target of rapamycin (mTOR), p70S6 kinase downstream of mTOR, and serine/threonine-protein kinase ULK1, a component of the autophagy initiating complex. In vitro, pharmacologic inhibition of mTOR, directly or through inhibition of AKT, enhanced autophagy after cisplatin treatment. Notably, in both cells and kidneys, blockade of PKCδ suppressed the cisplatin-induced phosphorylation of AKT, mTOR, p70S6 kinase, and ULK1 resulting in upregulation of autophagy. Furthermore, constitutively active and inactive forms of PKCδ respectively enhanced and suppressed cisplatin-induced apoptosis in cultured cells. In mechanistic studies, we showed coimmunoprecipitation of PKCδ and AKT from lysates of cisplatin-treated cells and direct phosphorylation of AKT at serine-473 by PKCδin vitro Finally, administration of the PKCδ inhibitor rottlerin with cisplatin protected against cisplatin nephrotoxicity in wild-type mice, but not in renal autophagy-deficient mice. Together, these results reveal a pathway consisting of PKCδ, AKT, mTOR, and ULK1 that inhibits autophagy in cisplatin nephrotoxicity. PKCδ mediates cisplatin nephrotoxicity at least in part by suppressing autophagy, and accordingly, PKCδ inhibition protects kidneys by upregulating autophagy.


Asunto(s)
Apoptosis , Autofagia/fisiología , Enfermedades Renales/patología , Riñón/citología , Proteína Quinasa C-delta/fisiología , Animales , Células Cultivadas , Cisplatino/toxicidad , Enfermedades Renales/inducido químicamente , Masculino , Ratones
7.
J Neurosci ; 35(11): 4614-25, 2015 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-25788678

RESUMEN

As one of the most effective and frequently used chemotherapeutic agents, paclitaxel produces peripheral neuropathy (paclitaxel-induced peripheral neuropathy or PIPN) that negatively affects chemotherapy and persists after cancer therapy. The mechanisms underlying this dose-limiting side effect remain to be fully elucidated. This study aimed to investigate the role of nociceptor protein kinase C (PKC) isoforms in PIPN. Employing multiple complementary approaches, we have identified a subset of PKC isoforms, namely ßII, δ, and ϵ, were activated by paclitaxel in the isolated primary afferent sensory neurons. Persistent activation of PKCßII, PKCδ, and PKCϵ was also observed in the dorsal root ganglion neurons after chronic treatment with paclitaxel in a mouse model of PIPN. Isoform-selective inhibitors of PKCßII, PKCδ, and PKCϵ given intrathecally dose-dependently attenuated paclitaxel-induced mechanical allodynia and heat hyperalgesia. Surprisingly, spinal inhibition of PKCßII and PKCδ, but not PKCϵ, blocked the spontaneous pain induced by paclitaxel. These data suggest that a subset of nociceptor PKC isoforms differentially contribute to spontaneous and evoked pain in PIPN, although it is not clear whether PKCϵ in other regions regulates spontaneous pain in PIPN. The findings can potentially offer new selective targets for pharmacological intervention of PIPN.


Asunto(s)
Nociceptores/fisiología , Paclitaxel/toxicidad , Dolor/enzimología , Proteína Quinasa C beta/fisiología , Proteína Quinasa C-delta/fisiología , Proteína Quinasa C-epsilon/fisiología , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/enzimología , Masculino , Ratones , Ratones Endogámicos ICR , Nociceptores/efectos de los fármacos , Dolor/inducido químicamente , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/fisiología , Proteína Quinasa C beta/antagonistas & inhibidores , Proteína Quinasa C-delta/antagonistas & inhibidores , Proteína Quinasa C-epsilon/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Ratas , Ratas Sprague-Dawley
8.
J Cell Biochem ; 117(3): 647-58, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26284819

RESUMEN

Hypoxia-inducible factor-1α (HIF-1α) is one of the key transcription factors that mediate adaptation to hypoxia. Despite increasing evidence implicating the PKC family as potential modulators of HIF-1α, the molecular mechanisms of PKC isoform-dependent HIF-1α activity under hypoxic conditions have not been systematically elucidated in cancer cell lines. Here, we collectively investigated how each isoform of the PKC family contributes to HIF-1α accumulation in the human cervical cancer cell line HeLa. Among the abundant PKC isoforms, blockade of either PKCα or PKCδ was found to substantially reduce HIF-1α accumulation and transcriptional activity in hypoxic cells. Knockdown of PKCδ resulted in a reduction of HIF-1α mRNA levels, whereas the HIF-1α mRNA level was unchanged regardless of PKCα knockdown. Upon searching for the downstream effectors of these kinases, we found that PKCα controls HIF-1α translation via AKT-mTOR under hypoxic conditions. On the other hand, one of the well-known transcriptional regulation pathways of HIF-1α, nuclear factor-κB (NF-κB) is identified as a downstream effector of PKCδ. Taken together, our findings provide insights into the roles of PKC isoforms as additional, discrete modulators of hypoxia-stimulated HIF-1α accumulation through different signaling pathways.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteína Quinasa C-alfa/fisiología , Proteína Quinasa C-delta/fisiología , Hipoxia de la Célula , Células HeLa , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Isoenzimas/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Biosíntesis de Proteínas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Estabilidad del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
9.
Blood ; 123(6): 931-4, 2014 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-24357731

RESUMEN

Immune complexes consisting of heparin, platelet factor 4 (PF4), and PF4/heparin-reactive antibodies are central to the pathogenesis of heparin-induced thrombocytopenia (HIT). It is as yet unclear what triggers the initial induction of pathogenic antibodies. We identified B cells in peripheral blood of healthy adults that produce PF4/heparin-specific antibodies following in vitro stimulation with proinflammatory molecules containing deoxycytosine-deoxyguanosine (CpG). Similarly, B cells from unmanipulated wild-type mice produced PF4/heparin-specific antibodies following in vitro or in vivo CpG stimulation. Thus, both healthy humans and mice possess preexisting inactive/tolerant PF4/heparin-specific B cells. The findings suggest that breakdown of tolerance leads to PF4/heparin-specific B-cell activation and antibody production in patients developing HIT. Consistent with this concept, mice lacking protein kinase Cδ (PKCδ) that are prone to breakdown of B-cell tolerance produced anti-PF4/heparin antibodies spontaneously. Therefore, breakdown of tolerance can lead to PF4/heparin-specific antibody production, and B-cell tolerance may play an important role in HIT pathogenesis.


Asunto(s)
Formación de Anticuerpos/inmunología , Anticoagulantes/efectos adversos , Linfocitos B/inmunología , Heparina/efectos adversos , Factor Plaquetario 4/metabolismo , Proteína Quinasa C-delta/fisiología , Trombocitopenia/inmunología , Adulto , Animales , Anticoagulantes/metabolismo , Linfocitos B/metabolismo , Linfocitos B/patología , Células Cultivadas , Heparina/metabolismo , Humanos , Tolerancia Inmunológica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor Plaquetario 4/inmunología , Pronóstico , Trombocitopenia/inducido químicamente , Trombocitopenia/metabolismo
10.
Am J Physiol Lung Cell Mol Physiol ; 308(8): L827-36, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25659900

RESUMEN

Pulmonary hypertension (PH) eventually leads to right ventricular (RV) fibrosis and dysfunction that is associated with increased morbidity and mortality. Although angiotensin II plays an important role in RV remodeling associated with hypoxic PH, the molecular mechanisms underlying RV fibrosis in PH largely remain unresolved. We hypothesized that PKC-p38 signaling is involved in RV collagen accumulation in PH and in response to angiotensin II stimulation. Adult male Sprague-Dawley rats were exposed to 3 wk of normoxia or hypoxia (10% FiO2 ) as a model of PH. Hypoxic rats developed RV hypertrophy and fibrosis associated with an increase in PKC ßII and δ protein expression and p38 dephosphorylation in freshly isolated RV cardiac fibroblasts. Further mechanistic studies were performed in cultured primary cardiac fibroblasts stimulated with angiotensin II, a key activator of ventricular fibrosis in PH. Angiotensin II induced a reduction in p38 phosphorylation that was attenuated following chemical inhibition of PKC ßII and δ. Molecular and chemical inhibition of PKC ßII and δ abrogated angiotensin II-induced cardiac fibroblast proliferation and collagen deposition in vitro. The effects of PKC inhibition on proliferation and fibrosis were reversed by chemical inhibition of p38. Conversely, constitutive activation of p38 attenuated angiotensin II-induced increase of cardiac fibroblast proliferation and collagen accumulation. PKC ßII- and δ-dependent inactivation of p38 regulates cardiac fibroblast proliferation and collagen deposition in response to angiotensin II, which suggests that the PKC-p38 signaling in cardiac fibroblasts may be involved and important in the pathophysiology of RV fibrosis in PH.


Asunto(s)
Angiotensina II/fisiología , Hipertensión Pulmonar/enzimología , Hipertrofia Ventricular Derecha/enzimología , Proteína Quinasa C beta/fisiología , Proteína Quinasa C-delta/fisiología , Animales , Hipoxia de la Célula , Proliferación Celular , Células Cultivadas , Colágeno/metabolismo , Activación Enzimática , Fibroblastos/enzimología , Fibrosis , Ventrículos Cardíacos/patología , Hipertensión Pulmonar/complicaciones , Masculino , Ratas Sprague-Dawley , Disfunción Ventricular Derecha/enzimología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Mol Reprod Dev ; 82(10): 797-808, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26202826

RESUMEN

This study tested the function of protein kinase C delta (PKCδ) during fertilization and embryonic development using gene-knockout (Prkcd(-/-)) mice. Fertility analysis revealed that Prkcd(-/-) mating pairs produce significantly fewer pups per litter than wild-type pairs (P < 0.05), and exhibit a high incidence of embryonic loss post-implantation. Both Prkcd(-/-) male as well as Prkcd(-/-) female mice mated to Prkcd(+/+) controls also showed reduced litter sizes, with a selective loss of Prkcd-null pups. Further analysis of the females demonstrated comparable in vitro fertilization outcomes between control and Prkcd(-/-) oocytes fertilized with wild-type sperm. Pregnant Prkcd(-/-) females, however, exhibited a reduced number of total implantations, suggesting a possible disruption in early embryo quality and/or implantation. In turn, male gamete analysis revealed that Prkcd(-/-) sperm demonstrated a decreased capacity to penetrate the zona pellucida (P < 0.05), necessary for successful fertilization. Moreover, we identified phosphorylated PKCδ as a component of the sperm acrosome, indicating a potential role for this kinase in acrosome exocytosis. Therefore, loss of PKCδ disrupts key reproductive functions in both males and females that limit fertility.


Asunto(s)
Desarrollo Embrionario , Fertilización , Proteína Quinasa C-delta/fisiología , Acrosoma/enzimología , Animales , Implantación del Embrión , Femenino , Fertilidad , Fertilización In Vitro , Masculino , Ratones , Ratones Noqueados , Embarazo , Proteína Quinasa C-delta/deficiencia , Proteína Quinasa C-delta/genética
12.
Hepatology ; 58(1): 284-92, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23424156

RESUMEN

UNLABELLED: Taurolithocholate (TLC) acutely inhibits the biliary excretion of multidrug-resistant associated protein 2 (Mrp2) substrates by inducing Mrp2 retrieval from the canalicular membrane, whereas cyclic adenosine monophosphate (cAMP) increases plasma membrane (PM)-MRP2. The effect of TLC may be mediated via protein kinase Cϵ (PKCϵ). Myristoylated alanine-rich C kinase substrate (MARCKS) is a membrane-bound F-actin crosslinking protein and is phosphorylated by PKCs. MARCKS phosphorylation has been implicated in endocytosis, and the underlying mechanism appears to be the detachment of phosphorylated myristoylated alanine-rich C kinase substrate (pMARCKS) from the membrane. The aim of the present study was to test the hypothesis that TLC-induced MRP2 retrieval involves PKCϵ-mediated MARCKS phosphorylation. Studies were conducted in HuH7 cells stably transfected with sodium taurocholate cotransporting polypeptide (HuH-NTCP cells) and in rat hepatocytes. TLC increased PM-PKCϵ and decreased PM-MRP2 in both HuH-NTCP cells and hepatocytes. cAMP did not affect PM-PKCϵ and increased PM-MRP2 in these cells. In HuH-NTCP cells, dominant-negative (DN) PKCϵ reversed TLC-induced decreases in PM-MRP2 without affecting cAMP-induced increases in PM-MRP2. TLC, but not cAMP, increased MARCKS phosphorylation in HuH-NTCP cells and hepatocytes. TLC and phorbol myristate acetate increased cytosolic pMARCKS and decreased PM-MARCKS in HuH-NTCP cells. TLC failed to increase MARCKS phosphorylation in HuH-NTCP cells transfected with DN-PKCϵ, and this suggested PKCϵ-mediated phosphorylation of MARCKS by TLC. In HuH-NTCP cells transfected with phosphorylation-deficient MARCKS, TLC failed to increase MARCKS phosphorylation or decrease PM-MRP2. CONCLUSION: Taken together, these results support the hypothesis that TLC-induced MRP2 retrieval involves TLC-mediated activation of PKCϵ followed by MARCKS phosphorylation and consequent detachment of MARCKS from the membrane.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Proteína Quinasa C-delta/fisiología , Ácido Taurolitocólico/metabolismo , Animales , Línea Celular Tumoral , Humanos , Masculino , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/efectos de los fármacos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Sustrato de la Proteína Quinasa C Rico en Alanina Miristoilada , Ratas
13.
Exp Cell Res ; 319(3): 153-60, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23063429

RESUMEN

Gö6976 is a nonglycosidic indolocarbazole compound widely used as a specific inhibitor of PKCα/ß. In experiments probing for a role of PKCα in human laminin-2-integrin-mediated cell adhesion and spreading of PC12 cells, we observed unexpected enhancements of adhesion, spreading and stress fiber formation to 1 µM Gö6976 with concomitant increase in membrane translocation of PKCδ and autophosphorylation of focal adhesion kinase (FAK). Importantly, enhanced cellular behavior and membrane translocation of PKCδ induced by Gö6976 was retained in siRNA-transfected PC12 cells to knockdown PKCα expression. Gö6976 also induced laminin-dependent cell adhesion in NIH/3T3 and CV-1 fibroblasts, suggesting of a mechanism that may be common to multiple cell-types. A specific inhibitor of PKCδ, rottlerin, completely abrogated Gö6976-dependent increase in PC12 cell adhesion to laminin as well as the activation of small GTPases, Rac1 and Cdc42, that are downstream of PKCδ in adhesion receptor signaling. siRNA knockdown of Rac1 and Cdc42 expression inhibited cell spreading and lamellipodia formation in PC12 cells. Overall, these results suggest that Gö6976 may stimulate membrane recruitment of PKCδ through a mechanism that is independent of PKCα/ß signaling. In addition, the activation of Rac1 and Cdc42 by human laminin-2-integrin-dependent activation of PKCδ/FAK signaling mediates cell spreading and lamellipodia formation in PC12 cells.


Asunto(s)
Carbazoles/farmacología , Membrana Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proteína Quinasa C-delta/fisiología , Animales , Adhesión Celular/efectos de los fármacos , Membrana Celular/metabolismo , Membrana Celular/fisiología , Células Cultivadas , Chlorocebus aethiops , Activación Enzimática/efectos de los fármacos , Células HEK293 , Humanos , Ratones , Células 3T3 NIH , Células PC12 , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C beta , Proteína Quinasa C-alfa/antagonistas & inhibidores , Proteína Quinasa C-delta/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Ratas , Regulación hacia Arriba/efectos de los fármacos
14.
Biochem Biophys Res Commun ; 437(3): 380-5, 2013 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-23827392

RESUMEN

Our previous works have shown that the (NADPH) oxidase (Nox) enzyme, in particular Nox1, plays an important role in oxidative stress and subsequent dopaminergic cell death elicited by paraquat (PQ). In non-neuronal and glial cells, protein kinase C δ (PKCδ) shows the ability to regulate the activity of the Nox system. Herein we aimed to investigate if also in dopaminergic neurons exposed to PQ, PKCδ can regulate Nox1 expression. The chemical inhibitor, rottlerin, and short interference RNA (siRNA) were used to inhibit or selectively knockdown PKCδ, respectively. The studies were performed using the immortalized rat mesencephalic dopaminergic cell line (N27 cells) exposed to PQ, after pre-incubation with rottlerin or transfected with PKCδ-siRNA. We observed that inhibition or knockdown of PKCδ significantly reduced PQ induced Nox1 transcript and protein levels, ROS generation and subsequent dopaminergic cell death. The results suggest that PKCδ plays a role in the regulation of Nox1-mediated oxidative stress elicited by PQ and could have a role in the pathogenesis of Parkinson's disease.


Asunto(s)
Dopamina/fisiología , NADH NADPH Oxidorreductasas/biosíntesis , Neuronas/enzimología , Paraquat/farmacología , Proteína Quinasa C-delta/fisiología , Acetofenonas/farmacología , Animales , Benzopiranos/farmacología , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Línea Celular Transformada , Inducción Enzimática/efectos de los fármacos , Inducción Enzimática/genética , Técnicas de Silenciamiento del Gen , NADH NADPH Oxidorreductasas/genética , NADPH Oxidasa 1 , Neuronas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Proteína Quinasa C-delta/antagonistas & inhibidores , Proteína Quinasa C-delta/biosíntesis , Ratas , Especies Reactivas de Oxígeno/metabolismo
15.
Mol Cell Biochem ; 372(1-2): 169-79, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23010891

RESUMEN

Activation of protein kinase C (PKC) is a critical intracellular signaling triggered by ischemic preconditioning (IPC), but the precise mechanisms underlying the actions of PKC in IPC-mediated cardioprotection remain unclear. Here, we investigated the role of PKC activation on the antioxidant activity by IPC in rabbit hearts. Isolated rabbit hearts were subjected to 60 min of global ischemia by cold cardioplegic arrest (4 °C) and 60 min of reperfusion (37 °C). IPC was induced by three cycles of 2-min ischemia following 3 min of reperfusion (37 °C) before cardioplegic arrest. IPC resulted in a better recovery of mechanical function, increased tissue reduced glutathione-to-oxidized glutathione ratio (GSH/GSSG), superoxide dismutase and catalase content, and decreased tissue malondialdehyde (MDA) content compared to control hearts subjected to 60 min of cardioplegic ischemia and 60 min of reperfusion. IPC also significantly induced activation of nuclear factor erythroid 2-related factor 2 (Nrf2) and the inductions of antioxidant genes heme oxygenase-1 (HO-1) and manganese superoxide dismutase (MnSOD). Injection of phorbol 12-myristate 13 acetate, an activator of PKC, before cardioplegic ischemia induced translocation of PKC-δ and -ε isoforms to membrane fraction, nuclear accumulation of Nrf2, and conferred cardioprotection similar to IPC. Polymyxin B, an inhibitor of PKC, blocked the membrane translocation of PKC-δ and -ε during IPC, inhibited Nrf2 nuclear accumulation, and significantly diminished the IPC-induced cardioprotection when administrated before IPC. These results indicate that the activation of PKC induces the translocation of Nrf2 and the enhancement of endogenous antioxidant defenses in the IPC hearts and suggest that PKC may target Nrf2 to confer cardioprotection.


Asunto(s)
Precondicionamiento Isquémico , Miocardio/enzimología , Factor 2 Relacionado con NF-E2/metabolismo , Proteína Quinasa C-delta/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Animales , Antioxidantes/metabolismo , Soluciones Cardiopléjicas/farmacología , Núcleo Celular/metabolismo , Vasos Coronarios/fisiopatología , Activación Enzimática/efectos de los fármacos , Femenino , Expresión Génica/efectos de los fármacos , Glutatión/metabolismo , Técnicas In Vitro , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , Isoenzimas/fisiología , L-Lactato Deshidrogenasa/metabolismo , Masculino , Malondialdehído/metabolismo , Contracción Miocárdica/efectos de los fármacos , Miocardio/metabolismo , Estrés Oxidativo , Polimixina B/farmacología , Proteína Quinasa C-delta/antagonistas & inhibidores , Proteína Quinasa C-delta/fisiología , Proteína Quinasa C-epsilon/antagonistas & inhibidores , Proteína Quinasa C-epsilon/fisiología , Transporte de Proteínas , Conejos , Superóxido Dismutasa/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Función Ventricular Izquierda , Presión Ventricular
16.
Mediators Inflamm ; 2013: 326041, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24385683

RESUMEN

Thrombin is a key mediator of fibrin deposition, angiogenesis, and proinflammatory processes. Abnormalities in these processes are primary features of rheumatoid arthritis and osteoarthritis. Matrix metalloproteinase-13 (MMP-13) may contribute to the breakdown of articular cartilage during arthritis. However, the role of thrombin in MMP-13 production in chondrocytes is unknown. In this study, we investigated the intracellular signaling pathways involved in thrombin-induced MMP-13 expression in human chondrocytes. We found that stimulation with thrombin led to increased secretion of MMP-13 in cultured human chondrocytes. Further, this thrombin-induced MMP-13 production was reduced after transfection with siRNAs against protease activated receptors 1 and 3 (PAR1 and PAR3), but not with PAR4 siRNA. Treatment with specific inhibitors for PKCδ, c-Src, EGFR, PI3K, Akt, or AP-1 or with the corresponding siRNAs against these signaling proteins also abolished the thrombin-mediated increase in MMP-13 production in chondrocytes. Our results provide evidence that thrombin acts through the PAR1/PAR3 receptors and activates PKCδ and c-Src, resulting in EGFR transactivation and activation of PI3K, Akt, and finally AP-1 on the MMP-13 promoter, thereby contributing to cartilage destruction during arthritis.


Asunto(s)
Condrocitos/enzimología , Metaloproteinasa 13 de la Matriz/genética , Transducción de Señal , Trombina/farmacología , Artritis Reumatoide/etiología , Proteína Tirosina Quinasa CSK , Células Cultivadas , Receptores ErbB/fisiología , Humanos , Osteoartritis/etiología , Fosfatidilinositol 3-Quinasas/fisiología , Proteína Quinasa C-delta/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , ARN Mensajero/análisis , Receptor PAR-1/fisiología , Transducción de Señal/fisiología , Factor de Transcripción AP-1/fisiología , Familia-src Quinasas/fisiología
17.
Diabetologia ; 55(10): 2682-2692, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22820510

RESUMEN

AIMS/HYPOTHESIS: Activation of the G protein-coupled receptor (GPR)40 by long-chain fatty acids potentiates glucose-stimulated insulin secretion (GSIS) from pancreatic beta cells, and GPR40 agonists are in clinical development for type 2 diabetes therapy. GPR40 couples to the G protein subunit Gα(q/11) but the signalling cascade activated downstream is unknown. This study aimed to determine the mechanisms of GPR40-dependent potentiation of GSIS by fatty acids. METHODS: Insulin secretion in response to glucose, oleate or diacylglycerol (DAG) was assessed in dynamic perifusions and static incubations in islets from wild-type (WT) and Gpr40 (-/-) mice. Depolymerisation of filamentous actin (F-actin) was visualised by phalloidin staining and epifluorescence. Pharmacological and molecular approaches were used to ascertain the roles of protein kinase D (PKD) and protein kinase C delta in GPR40-mediated potentiation of GSIS. RESULTS: Oleate potentiates the second phase of GSIS, and this effect is largely dependent upon GPR40. Accordingly, oleate induces rapid F-actin remodelling in WT but not in Gpr40 (-/-) islets. Exogenous DAG potentiates GSIS in both WT and Gpr40 (-/-) islets. Oleate induces PKD phosphorylation at residues Ser-744/748 and Ser-916 in WT but not Gpr40 (-/-) islets. Importantly, oleate-induced F-actin depolymerisation and potentiation of GSIS are lost upon pharmacological inhibition of PKD1 or deletion of Prkd1. CONCLUSIONS/INTERPRETATION: We conclude that the signalling cascade downstream of GPR40 activation by fatty acids involves activation of PKD1, F-actin depolymerisation and potentiation of second-phase insulin secretion. These results provide important information on the mechanisms of action of GPR40, a novel drug target for type 2 diabetes.


Asunto(s)
Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Proteína Quinasa C/fisiología , Receptores Acoplados a Proteínas G/fisiología , Actinas/metabolismo , Animales , Células Cultivadas , Diglicéridos/farmacología , Glucosa/farmacología , Secreción de Insulina , Islotes Pancreáticos/citología , Islotes Pancreáticos/efectos de los fármacos , Ratones , Ratones Noqueados , Modelos Animales , Ácido Oléico/farmacología , Proteína Quinasa C-delta/deficiencia , Proteína Quinasa C-delta/genética , Proteína Quinasa C-delta/fisiología , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/genética , Transducción de Señal/fisiología
18.
J Neuroinflammation ; 9: 82, 2012 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-22540228

RESUMEN

BACKGROUND: The mechanisms of progressive dopaminergic neuronal loss in Parkinson's disease (PD) remain poorly understood, largely due to the complex etiology and multifactorial nature of disease pathogenesis. Several lines of evidence from human studies and experimental models over the last decade have identified neuroinflammation as a potential pathophysiological mechanism contributing to disease progression. Tumor necrosis factor α (TNF) has recently emerged as the primary neuroinflammatory mediator that can elicit dopaminergic cell death in PD. However, the signaling pathways by which TNF mediates dopaminergic cell death have not been completely elucidated. METHODS: In this study we used a dopaminergic neuronal cell model and recombinant TNF to characterize intracellular signaling pathways activated during TNF-induced dopaminergic neurotoxicity. Etanercept and neutralizing antibodies to tumor necrosis factor receptor 1 (TNFR1) were used to block TNF signaling. We confirmed the results from our mechanistic studies in primary embryonic mesencephalic cultures and in vivo using the stereotaxic lipopolysaccharide (LPS) model of nigral dopaminergic degeneration. RESULTS: TNF signaling in dopaminergic neuronal cells triggered the activation of protein kinase Cδ (PKCδ), an isoform of the novel PKC family, by caspase-3 and caspase-8 dependent proteolytic cleavage. Both TNFR1 neutralizing antibodies and the soluble TNF receptor Etanercept blocked TNF-induced PKCδ proteolytic activation. Proteolytic activation of PKCδ was accompanied by translocation of the kinase to the nucleus. Notably, inhibition of PKCδ signaling by small interfering (si)RNA or overexpression of a PKCδ cleavage-resistant mutant protected against TNF-induced dopaminergic neuronal cell death. Further, primary dopaminergic neurons obtained from PKCδ knockout (-/-) mice were resistant to TNF toxicity. The proteolytic activation of PKCδ in the mouse substantia nigra in the neuroinflammatory LPS model was also observed. CONCLUSIONS: Collectively, these results identify proteolytic activation of PKCδ proapoptotic signaling as a key downstream effector of dopaminergic cell death induced by TNF. These findings also provide a rationale for therapeutically targeting PKCδ to mitigate progressive dopaminergic degeneration resulting from chronic neuroinflammatory processes.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Neuronas Dopaminérgicas/enzimología , Encefalitis/enzimología , Encefalitis/patología , Proteína Quinasa C-delta/metabolismo , Receptores de Muerte Celular/fisiología , Transducción de Señal/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Reguladoras de la Apoptosis/toxicidad , Muerte Celular/fisiología , Células Cultivadas , Neuronas Dopaminérgicas/patología , Encefalitis/etiología , Activación Enzimática/fisiología , Ratones , Proteína Quinasa C-delta/fisiología , Proteolisis , Ratas , Factor de Necrosis Tumoral alfa/toxicidad
19.
Blood ; 116(26): 6027-36, 2010 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-20861461

RESUMEN

The neutrophil nicotinamide adenine dinucleotide phosphate-oxidase is a multisubunit enzyme (comprising gp91(phox), p22(phox), p67(phox), p40(phox), p47(phox), and Rac) that plays a vital role in microbial killing. The recent discovery of a chronic granulomatous disease patient who expresses a mutant p40(phox) subunit, together with the development of mouse models of p40(phox) function, indicate phosphatidylinositol 3-phosphate binding to the PX domain of p40(phox) is an important signal for oxidase activation. However, the presence of other conserved residues and domains in p40(phox) suggest further regulatory roles for this protein. To test this, we introduced wild-type and mutated versions of p40(phox) into fully differentiated mouse neutrophils by retroviral transduction of p40(phox)(-/-) bone marrow progenitors and repopulation of the bone marrow compartment in radiation chimaeras. Phosphorylation of p40(phox) on threonine 154, but not serine 315, was required for full oxidase activation in response to formylated bacterial peptide fMLP, serum-opsonized S aureus, and immunoglobulin-opsonized sheep red blood cells. A functional SH3 domain was not required for oxidase activation, and deletion of the entire domain resulted in enhanced oxidase responses. Phosphorylation of threonine 154 in response to S aureus was mediated by protein kinase Cδ and was required for full translocation of p47(phox) to phagosomes. These results define an important new element in the physiological activation of the oxidase.


Asunto(s)
NADPH Oxidasas/metabolismo , Neutrófilos/enzimología , Fosfoproteínas/fisiología , Proteína Quinasa C-delta/fisiología , Infecciones Estafilocócicas/metabolismo , Treonina , Animales , Western Blotting , Médula Ósea/metabolismo , Eritrocitos/metabolismo , Femenino , Masculino , Ratones , Ratones Noqueados/microbiología , N-Formilmetionina Leucil-Fenilalanina/farmacología , Fagosomas/metabolismo , Fosforilación , Especies Reactivas de Oxígeno/metabolismo , Retroviridae/genética , Ovinos , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/patología , Staphylococcus aureus , Irradiación Corporal Total
20.
Proc Natl Acad Sci U S A ; 106(50): 21353-8, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19965374

RESUMEN

Cyclin-dependent kinase 5 (Cdk5) and its activator p35 are critical for radial migration and lamination of cortical neurons. However, how this kinase is regulated by extracellular and intracellular signals during cortical morphogenesis remains unclear. Here, we show that PKCdelta, a member of novel PKC expressing in cortical neurons, could stabilize p35 by direct phosphorylation. PKCdelta attenuated the degradation of p35 but not its mutant derivative, which could not be phosphorylated by PKCdelta. Down-regulation of PKCdelta by in utero electroporation of specific small interference RNA (siRNA) severely impaired the radial migration of cortical neurons. This migration defect was similar to that caused by down-regulation of p35 and could be prevented by cotransfection with the wild-type but not the mutant p35. Furthermore, PKCdelta could be activated by the promigratory factor brain-derived neurotrophic factor (BDNF) and was required for the activation of Cdk5 by BDNF. Both PKCdelta and p35 were required for the promigratory effect of BDNF on cultured newborn neurons. Thus, PKCdelta may promote cortical radial migration through maintaining the proper level of p35 in newborn neurons.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/fisiología , Movimiento Celular , Corteza Cerebral/citología , Proteínas del Tejido Nervioso/metabolismo , Proteína Quinasa C-delta/fisiología , Animales , Células Cultivadas , Quinasa 5 Dependiente de la Ciclina , Regulación hacia Abajo/efectos de los fármacos , Neuronas/citología , Neuronas/metabolismo , Fosforilación , Proteína Quinasa C-delta/genética , Estabilidad Proteica , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda