Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Biol Reprod ; 110(4): 684-697, 2024 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-38145487

RESUMO

The protein kinase A (PKA) signaling pathway, which mediates protein phosphorylation, is important for sperm motility and male fertility. This process relies on A-kinase anchoring proteins that organize PKA and its signalosomes within specific subcellular compartments. Previously, it was found that the absence of A-kinase anchoring protein 3 (AKAP3) leads to multiple morphological abnormalities in mouse sperm. But how AKAP3 regulates sperm motility is yet to be elucidated. AKAP3 has two amphipathic domains, here named dual and RI, in its N-terminus. These domains are responsible for binding regulatory subunits I alpha (RIα) and II alpha (RIIα) of PKA and for RIα only, respectively. Here, we generated mutant mice lacking the dual and RI domains of AKAP3. It was found that the deletion of these domains caused male mouse infertile, accompanied by mild defects in the fibrous sheath of sperm tails. Additionally, the levels of serine/threonine phosphorylation of PKA substrates and tyrosine phosphorylation decreased in the mutant sperm, which exhibited a defect in hyperactivation under capacitation conditions. The protein levels of PKA subunits remained unchanged. But, interestingly, the regulatory subunit RIα was mis-localized from principal piece to midpiece of sperm tail, whereas this was not observed for RIIα. Further protein-protein interaction assays revealed a preference for AKAP3 to bind RIα over RIIα. Collectively, our findings suggest that AKAP3 is important for sperm hyperactivity by regulating type-I PKA signaling pathway mediated protein phosphorylation via its dual and RI domains.


Assuntos
Proteínas de Ancoragem à Quinase A , Proteína Quinase Tipo I Dependente de AMP Cíclico , Motilidade dos Espermatozoides , Animais , Masculino , Camundongos , Proteínas de Ancoragem à Quinase A/genética , Proteínas de Ancoragem à Quinase A/metabolismo , Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Fertilidade/genética , Sêmen/metabolismo , Transdução de Sinais/fisiologia , Motilidade dos Espermatozoides/genética , Espermatozoides/metabolismo , Capacitação Espermática/genética
2.
J Virol ; 95(13): e0025121, 2021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-33853963

RESUMO

Several types of widespread human papillomaviruses (HPVs) may induce the transformation of infected cells, provoking the development of neoplasms. Two main genera of HPVs are classified as mucosatropic alphapapillomaviruses and cutaneotropic betapapillomaviruses (α- and ß-HPVs, respectively), and they both include high-risk cancer-associated species. The absence of antiviral drugs has driven investigations into the details of the molecular mechanisms of the HPV life cycle. HPV replication depends on the viral helicase E1 and the transcription factor E2. Their biological activities are controlled by numerous cellular proteins, including protein kinases. Here, we report that ubiquitously expressed cyclic AMP-dependent protein kinase A (PKA) differentially regulates the replication of α-HPV11, α-HPV18, and ß-HPV5. PKA stimulates the replication of both α-HPVs studied but has a more profound effect on the replication of high-risk α-HPV18. However, the replication of ß-HPV5 is inhibited by activated PKA in human primary keratinocytes and U2OS cells. We show that the activation of PKA signaling by different pharmacological agents induces the rapid proteasomal degradation of the HPV5 E2 protein, which in turn leads to the downregulation of E2-dependent transcription. In contrast, PKA-stimulated induction of HPV18 replication is the result of the downregulation of the E8^E2 transcript encoding a potent viral transcriptional inhibitor together with the rapid upregulation of E1 and E2 protein levels. IMPORTANCE Several types of human papillomaviruses (HPVs) are causative agents of various types of epithelial cancers. Here, we report that ubiquitously expressed cyclic AMP-dependent protein kinase A (PKA) differentially regulates the replication of various types of HPVs during the initial amplification and maintenance phases of the viral life cycle. The replication of the skin cancer-related pathogen HPV5 is suppressed, whereas the replication of the cervical cancer-associated pathogen HPV18 is activated, in response to elevated PKA activity. To inhibit HPV5 replication, PKA targets the viral transcriptional activator E2, inducing its rapid proteasomal degradation. PKA-dependent stimulation of HPV18 replication relies on the downregulation of another E2 gene product, E8^E2, which encodes a potent transcriptional repressor. Our findings highlight, for the first time, protein kinase-related mechanistic differences in the regulation of the replication of mucosal and cutaneous HPV types.


Assuntos
Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , Papillomavirus Humano 18/crescimento & desenvolvimento , Proteínas Oncogênicas Virais/metabolismo , Replicação Viral/fisiologia , Linhagem Celular Tumoral , DNA Helicases/metabolismo , Genoma Viral/genética , Papillomavirus Humano 18/classificação , Humanos , Infecções por Papillomavirus/patologia , Fatores de Transcrição/metabolismo
3.
Int J Mol Sci ; 21(9)2020 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-32357495

RESUMO

Protein kinase A (PKA) are tetramers of two catalytic and two regulatory subunits, docked at precise intracellular sites to provide localized phosphorylating activity, triggered by cAMP binding to regulatory subunits and subsequent dissociation of catalytic subunits. It is unclear whether in the brain PKA dissociated subunits may also be found. PKA catalytic subunit was examined in various mouse brain areas using immunofluorescence, equilibrium binding and western blot, to reveal its location in comparison to regulatory subunits type RI and RII. In the cerebral cortex, catalytic subunits colocalized with clusters of RI, yet not all RI clusters were bound to catalytic subunits. In stria terminalis, catalytic subunits were in proximity to RI but separated from them. Catalytic subunits clusters were also present in the corpus striatum, where RII clusters were detected, whereas RI clusters were absent. Upon cAMP addition, the distribution of regulatory subunits did not change, while catalytic subunits were completely released from regulatory subunits. Unpredictably, catalytic subunits were not solubilized; instead, they re-targeted to other binding sites within the tissue, suggesting local macromolecular reorganization. Hence, the interactions between catalytic and regulatory subunits of protein kinase A consistently vary in different brain areas, supporting the idea of multiple interaction patterns.


Assuntos
Encéfalo/enzimologia , Proteína Quinase Tipo II Dependente de AMP Cíclico/metabolismo , Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , Animais , Córtex Cerebral/enzimologia , Corpo Estriado/enzimologia , AMP Cíclico/metabolismo , Proteína Quinase Tipo I Dependente de AMP Cíclico/genética , Proteína Quinase Tipo II Dependente de AMP Cíclico/genética , Feminino , Masculino , Camundongos , Especificidade de Órgãos , Núcleos Septais/enzimologia
4.
Circulation ; 136(24): 2337-2355, 2017 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-29051185

RESUMO

BACKGROUND: The nitric oxide-sensitive guanylyl cyclase/cGMP-dependent protein kinase type I signaling pathway can afford protection against the ischemia/reperfusion injury that occurs during myocardial infarction. Reportedly, voltage and Ca2+-activated K+ channels of the BK type are stimulated by cGMP/cGMP-dependent protein kinase type I, and recent ex vivo studies implicated that increased BK activity favors the survival of the myocardium at ischemia/reperfusion. It remains unclear, however, whether the molecular events downstream of cGMP involve BK channels present in cardiomyocytes or in other cardiac cell types. METHODS: Gene-targeted mice with a cardiomyocyte- or smooth muscle cell-specific deletion of the BK (CMBK or SMBK knockouts) were subjected to the open-chest model of myocardial infarction. Infarct sizes of the conditional mutants were compared with litter-matched controls, global BK knockout, and wild-type mice. Cardiac damage was assessed after mechanical conditioning or pharmacological stimulation of the cGMP pathway and by using direct modulators of BK. Long-term outcome was studied with respect to heart functions and cardiac fibrosis in a chronic myocardial infarction model. RESULTS: Global BK knockouts and CMBK knockouts, in contrast with SMBK knockouts, exhibited significantly larger infarct sizes compared with their respective controls. Ablation of CMBK resulted in higher serum levels of cardiac troponin I and elevated amounts of reactive oxygen species, lower phosphorylated extracellular receptor kinase and phosphorylated AKT levels and an increase in myocardial apoptosis. Moreover, CMBK was required to allow beneficial effects of both nitric oxide-sensitive guanylyl cyclase activation and inhibition of the cGMP-degrading phosphodiesterase-5, ischemic preconditioning, and postconditioning regimens. To this end, after 4 weeks of reperfusion, fibrotic tissue increased and myocardial strain echocardiography was significantly compromised in CMBK-deficient mice. CONCLUSIONS: Lack of CMBK channels renders the heart more susceptible to ischemia/reperfusion injury, whereas the pathological events elicited by ischemia/reperfusion do not involve BK in vascular smooth muscle cells. BK seems to permit the protective effects triggered by cinaciguat, riociguat, and different phosphodiesterase-5 inhibitors and beneficial actions of ischemic preconditioning and ischemic postconditioning by a mechanism stemming primarily from cardiomyocytes. This study establishes mitochondrial CMBK channels as a promising target for limiting acute cardiac damage and adverse long-term events that occur after myocardial infarction.


Assuntos
Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Infarto do Miocárdio/tratamento farmacológico , Miocárdio/patologia , Miócitos Cardíacos/fisiologia , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Benzoatos/uso terapêutico , Cardiotônicos/uso terapêutico , Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , Modelos Animais de Doenças , Humanos , Precondicionamento Isquêmico , Canais de Potássio Ativados por Cálcio de Condutância Alta/genética , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infarto do Miocárdio/fisiopatologia , Óxido Nítrico/metabolismo , Pirazóis/uso terapêutico , Pirimidinas/uso terapêutico , Traumatismo por Reperfusão/fisiopatologia
5.
Eur J Pharmacol ; 794: 201-208, 2017 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-27894809

RESUMO

Cyclic adenosine monophosphate (cAMP) regulates many vital functions such as metabolism, proliferation, differentiation and death. Depending on cell types and stimulators, cAMP could either promote or attenuate cell death. cAMP signal can be transduced by protein kinase A (PKA) and/or exchange protein directly activated by cAMP (EPAC). In CML cells, cAMP may suppress their proliferation and enhance their differentiation. However, the role of cAMP on DNA damaging agent toxicity and the mechanism involved has not been studied. In this study, we studied the effect of cAMP on the sensitivity of CML cells to DNA damaging agents. We observed that forskolin (FSK) and dibutyryl-cAMP (DBcAMP) decreased cisplatin and etoposide-induced cell death in K562 cells. Moreover, PKA activator prevented K562 cells from DNA damaging agent-induced cell death while EPAC activator had no effect. Furthermore, we found that the PKA subtype, PKAIA, was involved in cAMP-attenuated resistance in K562 cells. Taken together, our results suggest that increased cAMP level confers CML cells to acquire a novel mechanism against DNA damaging agent toxicity via PKAIA. Thus, PKAIA inhibitor may be helpful in overcoming the resistance to DNA damaging agents in CML cells.


Assuntos
Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , AMP Cíclico/farmacologia , Dano ao DNA , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Bucladesina/farmacologia , Morte Celular/efeitos dos fármacos , Cisplatino/farmacologia , Colforsina/farmacologia , Etoposídeo/farmacologia , Humanos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Células K562 , Transdução de Sinais/efeitos dos fármacos
6.
Proc Natl Acad Sci U S A ; 113(28): 7786-91, 2016 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-27357676

RESUMO

Scaffolding proteins organize the information flow from activated G protein-coupled receptors (GPCRs) to intracellular effector cascades both spatially and temporally. By this means, signaling scaffolds, such as A-kinase anchoring proteins (AKAPs), compartmentalize kinase activity and ensure substrate selectivity. Using a phosphoproteomics approach we identified a physical and functional connection between protein kinase A (PKA) and Gpr161 (an orphan GPCR) signaling. We show that Gpr161 functions as a selective high-affinity AKAP for type I PKA regulatory subunits (RI). Using cell-based reporters to map protein-protein interactions, we discovered that RI binds directly and selectively to a hydrophobic protein-protein interaction interface in the cytoplasmic carboxyl-terminal tail of Gpr161. Furthermore, our data demonstrate that a binary complex between Gpr161 and RI promotes the compartmentalization of Gpr161 to the plasma membrane. Moreover, we show that Gpr161, functioning as an AKAP, recruits PKA RI to primary cilia in zebrafish embryos. We also show that Gpr161 is a target of PKA phosphorylation, and that mutation of the PKA phosphorylation site affects ciliary receptor localization. Thus, we propose that Gpr161 is itself an AKAP and that the cAMP-sensing Gpr161:PKA complex acts as cilium-compartmentalized signalosome, a concept that now needs to be considered in the analyzing, interpreting, and pharmaceutical targeting of PKA-associated functions.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Células HEK293 , Humanos , Luciferases de Renilla , Camundongos , Fosforilação , Peixe-Zebra
7.
J Mol Cell Cardiol ; 91: 215-27, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26773602

RESUMO

The balanced signaling between the two cyclic nucleotides (cNs) cAMP and cGMP plays a critical role in regulating cardiac contractility. Their degradation is controlled by distinctly regulated phosphodiesterase isoenzymes (PDEs), which in turn are also regulated by these cNs. As a result, PDEs facilitate communication between the ß-adrenergic and Nitric Oxide (NO)/cGMP/Protein Kinase G (PKG) signaling pathways, which regulate the synthesis of cAMP and cGMP respectively. The phenomena in which the cAMP and cGMP pathways influence the dynamics of each other are collectively referred to as cN cross-talk. However, the cross-talk response and the individual roles of each PDE isoenzyme in shaping this response remain to be fully characterized. We have developed a computational model of the cN cross-talk network that mechanistically integrates the ß-adrenergic and NO/cGMP/PKG pathways via regulation of PDEs by both cNs. The individual model components and the integrated network model replicate experimentally observed activation-response relationships and temporal dynamics. The model predicts that, due to compensatory interactions between PDEs, NO stimulation in the presence of sub-maximal ß-adrenergic stimulation results in an increase in cytosolic cAMP accumulation and corresponding increases in PKA-I and PKA-II activation; however, the potentiation is small in magnitude compared to that of NO activation of the NO/cGMP/PKG pathway. In a reciprocal manner, ß-adrenergic stimulation in the presence of sub-maximal NO stimulation results in modest cGMP elevation and corresponding increase in PKG activation. In addition, we demonstrate that PDE2 hydrolyzes increasing amounts of cAMP with increasing levels of ß-adrenergic stimulation, and hydrolyzes increasing amounts of cGMP with decreasing levels of NO stimulation. Finally, we show that PDE2 compensates for inhibition of PDE5 both in terms of cGMP and cAMP dynamics, leading to cGMP elevation and increased PKG activation, while maintaining whole-cell ß-adrenergic responses similar to that prior to PDE5 inhibition. By defining and quantifying reactions comprising cN cross-talk, the model characterizes the cross-talk response and reveals the underlying mechanisms of PDEs in this non-linear, tightly-coupled reaction system.


Assuntos
AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Redes Reguladoras de Genes , Modelos Cardiovasculares , Miocárdio/enzimologia , Miócitos Cardíacos/enzimologia , Diester Fosfórico Hidrolases/genética , Animais , Simulação por Computador , Proteína Quinase Tipo I Dependente de AMP Cíclico/genética , Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , Proteína Quinase Tipo II Dependente de AMP Cíclico/genética , Proteína Quinase Tipo II Dependente de AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/genética , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Regulação da Expressão Gênica , Isoenzimas/genética , Isoenzimas/metabolismo , Contração Miocárdica , Miocárdio/citologia , Miócitos Cardíacos/citologia , Óxido Nítrico/metabolismo , Diester Fosfórico Hidrolases/metabolismo , Transdução de Sinais
8.
Zhen Ci Yan Jiu ; 41(6): 497-501, 2016 Dec 25.
Artigo em Chinês | MEDLINE | ID: mdl-29071891

RESUMO

OBJECTIVE: To observe the effect of electroacupuncture (EA) at "Zusanli"(ST 36) on Ghrelin/cAMP/PKA expression in the jejunum in rats with spleen qi deficiency syndrome, so as to reveal its underlying mechanism in improving energy metabolism. METHODS: Forty male SD rats were randomly divided into 4 groups:normal group, spleen qi deficiency syndrome (model) group, EA group and non-acupoint group (n=10 in each group).The model of spleen qi deficiency syndrome was established by improper diet and overstrain. EA (2 Hz/15 Hz, 0.5 mA) was applied to bilateral "Zusanli" (ST 36) in the EA group and non-acupoint in non-acupoint group for 20 min, once a day for 6 days. The pathologic changes of the jejunum tissue were detected by H&E staining. Ghrelin, ATP and cAMP levels in jejunum tissue were determined by ELISA. The expression levels of PKA protein in jejunum tissue were determined by Western blot. RESULTS: H&E staining showed that the intestinal villi of the model group were swelling, shortening and thickening, with a damaged or broken top-part in the model group, and basically restored to normal after EA treatment. ELISA results showed that the contents of Ghrelin, ATP and cAMP in the jejunum tissue were significantly lower in the model group than in the normal group (P<0.05), while significantly higher in the EA group than in the model group (P<0.05). Western blot results showed that the expression of PKA protein in the jejunum tissue was significantly lower in the model group than in the normal group (P<0.05), and significantly higher in the EA group than in the model group and non-acupoint group (P<0.05). CONCLUSIONS: EA at ST 36 can improve the morphological changes in the jejunum of spleen qi deficiency rats, which may be associated with its effects in increasing Ghrelin, ATP and cAMP contents, and up-regulating PKA expression, leading to an increase of energy metabolism and spleen qi at last.


Assuntos
Pontos de Acupuntura , Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Eletroacupuntura , Grelina/metabolismo , Jejuno/metabolismo , Qi , Baço/fisiopatologia , Esplenopatias/terapia , Animais , Proteína Quinase Tipo I Dependente de AMP Cíclico/genética , Modelos Animais de Doenças , Grelina/genética , Humanos , Jejuno/enzimologia , Masculino , Ratos , Ratos Sprague-Dawley , Baço/metabolismo , Esplenopatias/genética , Esplenopatias/metabolismo , Esplenopatias/fisiopatologia
9.
ACS Chem Biol ; 10(6): 1502-10, 2015 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-25765284

RESUMO

A-Kinase Anchoring Proteins (AKAPs) coordinate complex signaling events by serving as spatiotemporal modulators of cAMP-dependent protein kinase activity in cells. Although AKAPs organize a plethora of diverse pathways, their cellular roles are often elusive due to the dynamic nature of these signaling complexes. AKAPs can interact with the type I or type II PKA holoenzymes by virtue of high-affinity interactions with the R-subunits. As a means to delineate AKAP-mediated PKA signaling in cells, we sought to develop isoform-selective disruptors of AKAP signaling. Here, we report the development of conformationally constrained peptides named RI-STapled Anchoring Disruptors (RI-STADs) that target the docking/dimerization domain of the type 1 regulatory subunit of PKA. These high-affinity peptides are isoform-selective for the RI isoforms, can outcompete binding by the classical AKAP disruptor Ht31, and can selectively displace RIα, but not RIIα, from binding the dual-specific AKAP149 complex. Importantly, these peptides are cell-permeable and disrupt Type I PKA-mediated phosphorylation events in the context of live cells. Hence, RI-STAD peptides are versatile cellular tools to selectively probe anchored type I PKA signaling events.


Assuntos
Proteínas de Ancoragem à Quinase A/antagonistas & inibidores , Proteína Quinase Tipo II Dependente de AMP Cíclico/antagonistas & inibidores , Proteína Quinase Tipo I Dependente de AMP Cíclico/antagonistas & inibidores , Peptídeos/química , Inibidores de Proteínas Quinases/química , Subunidades Proteicas/antagonistas & inibidores , Proteínas de Ancoragem à Quinase A/química , Proteínas de Ancoragem à Quinase A/metabolismo , Sequência de Aminoácidos , Sítios de Ligação/efeitos dos fármacos , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular , Proteína Quinase Tipo I Dependente de AMP Cíclico/química , Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , Proteína Quinase Tipo II Dependente de AMP Cíclico/química , Proteína Quinase Tipo II Dependente de AMP Cíclico/metabolismo , Humanos , Cinética , Dados de Sequência Molecular , Peptídeos/farmacologia , Fosforilação , Ligação Proteica/efeitos dos fármacos , Conformação Proteica , Inibidores de Proteínas Quinases/farmacologia , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo
10.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 34(3): 317-23, 2014 Mar.
Artigo em Chinês | MEDLINE | ID: mdl-24758084

RESUMO

OBJECTIVE: To explore the potential molecular mechanisms for Bushen Tiaojing Recipe (BTR) improving the endocrine function of ovarian granular cells by observing the effect of BTR containing serum on follicle stimulating hormone/cyclic adenosine monophosphate-protein kinase A (FSH/ cAMP-PKA) pathway in in vitro cultured human ovarian granular cells. METHODS: The primary ovarian granular cells collected from in vitro fertilization-embryo transfer patients were cultured for 24 h. The human and rat serum containing different concentrations of BTR (low, medium, high dose), and their normal serums were co-incubated with ovarian granular cells for 48 h respectively, and then they were divided into the low, medium, high dose BTR groups and the control group. The levels of estradiol (E2), progesterone (P), and cyclic adenosine monophosphate (cAMP) in the culture medium were measured by radioimmunoassay. The protein expression of FSHR in ovarian granular cells was detected by Western Blot. The mRNA expression of follicle stimulating hormone receptor (FSHR) and P450 aromatase (P450arom) in ovarian granular cells were detected by Real-time PCR. RESULTS: In human BTR containing serum groups: Compared with control group, the levels of E2 and cAMP in the culture medium were higher (both P < 0.05) in the medium and high dose BTR groups; the levels of P in the culture medium decreased in the medium dose BTR group (P < 0.01). The protein and mRNA expression of FSHR in ovarian granular cells increased (all P < 0.01), the mRNA expressions of P450arom in ovarian granular cells were higher (P < 0.05, P< 0.01) in the medium and high dose BTR groups. In rat BTR containing serum groups: Compared with the control group, the levels of E2 in the culture medium were higher (all P < 0.01), cAMP in the culture medium were higher (P < 0.05, P < 0.01) in the medium and high dose BTR group; the levels of P in the culture medium decreased in the medium dose BTR group (P < 0.01). The protein and mRNA expression of FSHR in ovarian granular cells were higher (all P < 0.01), the mRNA expression of P450arom in ovarian granular cells increased in the medium and high dose BTR groups (P < 0.05, P < 0.01). CONCLUSION: BTR could possibly improve the endocrine function of ovarian granular cells by regulating main effector molecules FSHR, cAMP, P450arom, and E2 in FSH/cAMP-PKA pathway of ovarian granular cells.


Assuntos
Medicamentos de Ervas Chinesas/farmacologia , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/metabolismo , Células Cultivadas , Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , Feminino , Hormônio Foliculoestimulante/metabolismo , Células da Granulosa/citologia , Humanos , Soro/química , Transdução de Sinais/efeitos dos fármacos
11.
Am J Respir Cell Mol Biol ; 51(2): 242-50, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24601788

RESUMO

Alveolar macrophages (AMs) represent the first line of innate immune defense in the lung. AMs use pattern recognition receptors (PRRs) to sense pathogens. The best studied PRR is Toll-like receptor (TLR)4, which detects LPS from gram-negative bacteria. The lipid mediator prostaglandin (PG)E2 dampens AM immune responses by inhibiting the signaling events downstream of PRRs. We examined the effect of PGE2 on TLR4 expression in rat AMs. Although PGE2 did not reduce the mRNA levels of TLR4, it decreased TLR4 protein levels. The translation inhibitor cycloheximide reduced TLR4 protein levels with similar kinetics as PGE2, and its effects were not additive with those of the prostanoid, suggesting that PGE2 inhibits TLR at the translational level. The action of PGE2 could be mimicked by the direct stimulator of cAMP formation, forskolin, and involved E prostanoid receptor 2 ligation and cAMP-dependent activation of unanchored type I protein kinase A. Cells pretreated with PGE2 for 24 hours exhibited decreased TNF-α mRNA and protein levels in response to LPS stimulation. Knockdown of TLR4 protein by small interfering RNA to the levels achieved by PGE2 treatment likewise decreased TNF-α mRNA and protein in response to LPS, establishing the functional significance of this PGE2 effect. We provide the first evidence of a lipid mediator acting through its cognate G protein-coupled receptor to affect PRR translation. Because PGE2 is produced in abundance at sites of infection, its inhibitory effects on AM TLR4 expression have important implications for host defense in the lung.


Assuntos
Dinoprostona/metabolismo , Macrófagos Alveolares/metabolismo , Receptor 4 Toll-Like/metabolismo , Transcrição Gênica , Adenilil Ciclases/metabolismo , Animais , AMP Cíclico/metabolismo , Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , Regulação para Baixo , Feminino , Humanos , Imunidade Inata , Mediadores da Inflamação/metabolismo , Macrófagos Alveolares/imunologia , Interferência de RNA , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Receptores de Prostaglandina E/metabolismo , Transdução de Sinais , Fatores de Tempo , Receptor 4 Toll-Like/genética , Transfecção , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Células U937
12.
J Cell Sci ; 127(Pt 1): 216-29, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24190886

RESUMO

Knowledge about the molecular structure of protein kinase A (PKA) isoforms is substantial. In contrast, the dynamics of PKA isoform activity in living primary cells has not been investigated in detail. Using a high content screening microscopy approach, we identified the RIIß subunit of PKA-II to be predominantly expressed in a subgroup of sensory neurons. The RIIß-positive subgroup included most neurons expressing nociceptive markers (TRPV1, NaV1.8, CGRP, IB4) and responded to pain-eliciting capsaicin with calcium influx. Isoform-specific PKA reporters showed in sensory-neuron-derived F11 cells that the inflammatory mediator PGE2 specifically activated PKA-II but not PKA-I. Accordingly, pain-sensitizing inflammatory mediators and activators of PKA increased the phosphorylation of RII subunits (pRII) in subgroups of primary sensory neurons. Detailed analyses revealed basal pRII to be regulated by the phosphatase PP2A. Increase of pRII was followed by phosphorylation of CREB in a PKA-dependent manner. Thus, we propose RII phosphorylation to represent an isoform-specific readout for endogenous PKA-II activity in vivo, suggest RIIß as a novel nociceptive subgroup marker, and extend the current model of PKA-II activation by introducing a PP2A-dependent basal state.


Assuntos
Capsaicina/farmacologia , Nociceptividade/efeitos dos fármacos , Proteína Fosfatase 2/genética , Células Receptoras Sensoriais/efeitos dos fármacos , Animais , Biomarcadores/metabolismo , Peptídeo Relacionado com Gene de Calcitonina/genética , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Cálcio/metabolismo , Colforsina/farmacologia , AMP Cíclico/metabolismo , Subunidade RIIbeta da Proteína Quinase Dependente de AMP Cíclico/genética , Subunidade RIIbeta da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Proteína Quinase Tipo I Dependente de AMP Cíclico/genética , Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , Ciclosporina/farmacologia , Dinoprostona/farmacologia , Regulação da Expressão Gênica , Masculino , Canal de Sódio Disparado por Voltagem NAV1.8/genética , Canal de Sódio Disparado por Voltagem NAV1.8/metabolismo , Fosforilação , Cultura Primária de Células , Proteína Fosfatase 2/metabolismo , Ratos , Ratos Sprague-Dawley , Células Receptoras Sensoriais/citologia , Células Receptoras Sensoriais/metabolismo , Transdução de Sinais , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo
13.
Ukr Biokhim Zh (1999) ; 85(4): 90-3, 2013.
Artigo em Ucraniano | MEDLINE | ID: mdl-24319977

RESUMO

It is known from literature about antioxidant, anti-inflammatory, membrane protective and adreniregulatory properties of N-acetylethanolamines, but data concerning their participation in regulation of steroidogenesis are insufficient. In order to study the influence of a synthetic analogue of endogenous canabinoid anandamide - metanandamide - on the intensity of steroidogenesis the influence of different concentrations of the drug on the contents of 11-hydroxicorticosteroides (11-HCS) in the culture medium after incubation of adrenal tissue in rats of both sexes was investigated. The quantitative determination of 11-HCS was conducted by fluorometric micromethod. It was shown that the incubation of tissue sections with metanandamide leads to a reduction of 11-HCS in males and an increase of their content in females. It was concluded that the inhibition of corticosteroid secretion and synthesis in males may be due to reduction of cAMP and inhibition of cAMP-dependent protein kinase A (PKA) under the effect of metanandamide. The opposite and dose-dependent effects of the preparation in females may be connected with the estrogen influence on the mechanisms of drug effect realization.


Assuntos
Corticosteroides/biossíntese , Córtex Suprarrenal/efeitos dos fármacos , Ácidos Araquidônicos/farmacologia , Etanolaminas/farmacologia , Córtex Suprarrenal/metabolismo , Corticosteroides/agonistas , Corticosteroides/antagonistas & inibidores , Animais , AMP Cíclico/metabolismo , Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Feminino , Fluorometria , Masculino , Microtomia , Ratos , Ratos Wistar , Fatores Sexuais , Técnicas de Cultura de Tecidos
14.
Mol Pharmacol ; 83(5): 1057-65, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23455313

RESUMO

Acute promyelocytic leukemia (APL) is characterized by granulopoietic differentiation arrest at the promyelocytic stage. In most cases, this defect can be overcome by treatment with all-trans-retinoic acid (ATRA), leading to complete clinical remission. Cyclic AMP signaling has a key role in retinoid treatment efficacy: it enhances ATRA-induced maturation in ATRA-sensitive APL cells (including NB4 cells) and restores it in some ATRA-resistant cells (including NB4-LR1 cells). We show that the two cell types express identical levels of the Cα catalytic subunit and comparable global cAMP-dependent protein kinase A (PKA) enzyme activity. However, the maturation-resistant NB4-LR1 cells have a PKA isozyme switch: compared with the NB4 cells, they have decreased content of the juxtanuclearly located PKA regulatory subunit IIα and PKA regulatory subunit IIß, and a compensatory increase of the generally cytoplasmically distributed PKA-RIα. Furthermore, the PKA regulatory subunit II exists mainly in the less cAMP-responsive nonautophosphorylated state in the NB4-LR1 cells. By the use of isozyme-specific cAMP analog pairs, we show that both PKA-I and PKA-II must be activated to achieve maturation in NB4-LR1 as well as NB4 cells. Therefore, special attention should be paid to activating not only PKA-I but also PKA-II in attempts to enhance ATRA-induced APL maturation in a clinical setting.


Assuntos
Antineoplásicos/farmacologia , Proteína Quinase Tipo II Dependente de AMP Cíclico/metabolismo , Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , Leucemia Promielocítica Aguda/tratamento farmacológico , Leucemia Promielocítica Aguda/patologia , Tretinoína/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , AMP Cíclico/metabolismo , Citoplasma/efeitos dos fármacos , Citoplasma/metabolismo , Humanos , Isoenzimas/metabolismo , Leucemia Promielocítica Aguda/enzimologia
15.
Cell Death Dis ; 4: e516, 2013 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-23449452

RESUMO

We show that cyclic AMP (cAMP) elevating agents protect blasts from patients with acute promyelocytic leukemia (APL) against death induced by first-line anti-leukemic anthracyclines like daunorubicin (DNR). The cAMP effect was reproduced in NB4 APL cells, and shown to depend on activation of the generally cytoplasmic cAMP-kinase type I (PKA-I) rather than the perinuclear PKA-II. The protection of both NB4 cells and APL blasts was associated with (inactivating) phosphorylation of PKA site Ser118 of pro-apoptotic Bad and (activating) phosphorylation of PKA site Ser133 of the AML oncogene CREB. Either event would be expected to protect broadly against cell death, and we found cAMP elevation to protect also against 2-deoxyglucose, rotenone, proteasome inhibitor and a BH3-only mimetic. The in vitro findings were mirrored by the findings in NSG mice with orthotopic NB4 cell leukemia. The mice showed more rapid disease progression when given cAMP-increasing agents (prostaglandin E2 analog and theophylline), both with and without DNR chemotherapy. The all-trans retinoic acid (ATRA)-induced terminal APL cell differentiation is a cornerstone in current APL treatment and is enhanced by cAMP. We show also that ATRA-resistant APL cells, believed to be responsible for treatment failure with current ATRA-based treatment protocols, were protected by cAMP against death. This suggests that the beneficial pro-differentiating and non-beneficial pro-survival APL cell effects of cAMP should be weighed against each other. The results suggest also general awareness toward drugs that can affect bone marrow cAMP levels in leukemia patients.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , AMP Cíclico/metabolismo , Daunorrubicina/farmacologia , 1-Metil-3-Isobutilxantina/farmacologia , Animais , Antibióticos Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , AMP Cíclico/agonistas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , Proteína Quinase Tipo II Dependente de AMP Cíclico/antagonistas & inibidores , Proteína Quinase Tipo II Dependente de AMP Cíclico/genética , Proteína Quinase Tipo II Dependente de AMP Cíclico/metabolismo , Daunorrubicina/uso terapêutico , Dinoprostona/análogos & derivados , Dinoprostona/farmacologia , Dinoprostona/uso terapêutico , Progressão da Doença , Células HL-60 , Humanos , Leucemia Promielocítica Aguda/tratamento farmacológico , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Teofilina/farmacologia , Teofilina/uso terapêutico , Transplante Heterólogo , Tretinoína/farmacologia , Tretinoína/uso terapêutico , Proteína de Morte Celular Associada a bcl/metabolismo
16.
J Biol Chem ; 287(52): 43789-97, 2012 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-23115245

RESUMO

Protein kinase A-anchoring proteins (AKAPs) provide spatio-temporal specificity for the omnipotent cAMP-dependent protein kinase (PKA) via high affinity interactions with PKA regulatory subunits (PKA-RI, RII). Many PKA-RII-AKAP complexes are heavily tethered to cellular substructures, whereas PKA-RI-AKAP complexes have remained largely undiscovered. Here, using a cAMP affinity-based chemical proteomics strategy in human heart and platelets, we uncovered a novel, ubiquitously expressed AKAP, termed small membrane (sm)AKAP due to its specific localization at the plasma membrane via potential myristoylation/palmitoylation anchors. In vitro binding studies revealed specificity of smAKAP for PKA-RI (K(d) = 7 nM) over PKA-RII (K(d) = 53 nM) subunits, co-expression of smAKAP with the four PKA R subunits revealed an even more exclusive specificity of smAKAP for PKA-RIα/ß in the cellular context. Applying the singlet oxygen-generating electron microscopy probe miniSOG indicated that smAKAP is tethered to the plasma membrane and is particularly dense at cell-cell junctions and within filopodia. Our preliminary functional characterization of smAKAP provides evidence that, like PKA-RII, PKA-RI can be tightly tethered by a novel repertoire of AKAPs, providing a new perspective on spatio-temporal control of cAMP signaling.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Membrana Celular/metabolismo , Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Lipoilação/fisiologia , Sistemas do Segundo Mensageiro/fisiologia , Proteínas de Ancoragem à Quinase A/genética , Animais , Membrana Celular/genética , AMP Cíclico/genética , Proteína Quinase Tipo I Dependente de AMP Cíclico/genética , Feminino , Humanos , Junções Intercelulares/genética , Junções Intercelulares/metabolismo , Masculino , Camundongos , Ligação Proteica , Pseudópodes/genética , Pseudópodes/metabolismo
17.
PLoS One ; 7(9): e37835, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23028418

RESUMO

Many studies have shown that cyclic adenosine-5'-monophosphate (cAMP)-dependent protein kinase A (PKA) and G-protein-coupled receptor 3 (GPR3) are crucial for controlling meiotic arrest in oocytes. However, it is unclear how gonadotropins modulate these factors to regulate oocyte maturation, especially by gap junctional communication (GJC). Using an in vitro meiosis-arrested mouse cumulus-oocyte complex (COC) culture model, we showed that there is a close relationship between follicle-stimulating hormone (FSH) and the PKA type I (PKAI) and GPR3. The effect of FSH on oocyte maturation was biphasic, initially inhibitory and then stimulatory. During FSH-induced maturation, rapid cAMP surges were observed in both cumulus cells and oocyte. Most GJC between cumulus cells and oocyte ceased immediately after FSH stimulation and recommenced after the cAMP surge. FSH-induced maturation was blocked by PKAI activator 8-AHA-cAMP. Levels of PKAI regulatory subunits and GPR3 decreased and increased, respectively, after FSH stimulation. In the presence of the GJC inhibitor carbenoxolone (CBX), FSH failed to induce the meiotic resumption and the changes in PKAI, GPR3 and cAMP surge in oocyte were no longer detected. Furthermore, GPR3 was upregulated by high cAMP levels, but not by PKAI activation. When applied after FSH stimulation, the specific phosphodiesterase 3A (PDE3A) inhibitor cilostamide immediately blocked meiotic induction, regardless of when it was administered. PKAI activation inhibited mitogen-activated protein kinase (MAPK) phosphorylation in the oocytes of COCs, which participated in the initiation of FSH-induced meiotic maturation in vitro. Just before FSH-induced meiotic maturation, cAMP, PKAI, and GPR3 returned to basal levels, and PDE3A activity and MAPK phosphorylation increased markedly. These experiments show that FSH induces a transient increase in cAMP levels and regulates GJC to control PKAI and GPR3 activities, thereby creating an inhibitory phase. After PDE3A and MAPK activities increase, meiosis resumes.


Assuntos
Comunicação Celular/efeitos dos fármacos , Células do Cúmulo/metabolismo , Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , Hormônio Foliculoestimulante/farmacologia , Junções Comunicantes/metabolismo , Meiose/efeitos dos fármacos , Oócitos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Células do Cúmulo/citologia , Células do Cúmulo/efeitos dos fármacos , AMP Cíclico/análogos & derivados , AMP Cíclico/metabolismo , AMP Cíclico/farmacologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/metabolismo , Ativação Enzimática/efeitos dos fármacos , Feminino , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Oócitos/citologia , Oócitos/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
18.
Biochem J ; 448(3): 307-20, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22957732

RESUMO

PDK1 (phosphoinositide-dependent protein kinase 1) phosphorylates and activates PKA (cAMP-dependent protein kinase) in vitro. Docking of the HM (hydrophobic motif) in the C-terminal tail of the PKA catalytic subunits on to the PIF (PDK1-interacting fragment) pocket of PDK1 is a critical step in this activation process. However, PDK1 regulation of PKA in vivo remains controversial. Saccharomyces cerevisiae contains three PKA catalytic subunits, TPK1, TPK2 and TPK3. We demonstrate that Pkh [PKB (protein kinase B)-activating kinase homologue] protein kinases phosphorylate the activation loop of each Tpk in vivo with various efficiencies. Pkh inactivation reduces the interaction of each catalytic subunit with the regulatory subunit Bcy1 without affecting the specific kinase activity of PKA. Comparative analysis of the in vitro interaction and phosphorylation of Tpks by Pkh1 shows that Tpk1 and Tpk2 interact with Pkh1 through an HM-PIF pocket interaction. Unlike Tpk1, mutagenesis of the activation loop site in Tpk2 does not abolish in vitro phosphorylation, suggesting that Tpk2 contains other, as yet uncharacterized, Pkh1 target sites. Tpk3 is poorly phosphorylated on its activation loop site, and this is due to the weak interaction of Tpk3 with Pkh1 because of the atypical HM found in Tpk3. In conclusion, the results of the present study show that Pkh protein kinases contribute to the divergent regulation of the Tpk catalytic subunits.


Assuntos
Domínio Catalítico/fisiologia , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiologia , Saccharomyces cerevisiae/enzimologia , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Motivos de Aminoácidos/fisiologia , Sequência de Aminoácidos , Ativação Enzimática/fisiologia , Isoenzimas/metabolismo , Dados de Sequência Molecular , Mutagênese , Fosforilação/genética
19.
PLoS One ; 7(7): e40860, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22815846

RESUMO

BACKGROUND: The unconventional motor protein, myosin Va, is crucial for the development of the mouse neuromuscular junction (NMJ) in the early postnatal phase. Furthermore, the cooperative action of protein kinase A (PKA) and myosin Va is essential to maintain the adult NMJ. We here assessed the involvement of myosin Va and PKA in NMJ recovery during muscle regeneration. METHODOLOGY/PRINCIPAL FINDINGS: To address a putative role of myosin Va and PKA in the process of muscle regeneration, we used two experimental models the dystrophic mdx mouse and Notexin-induced muscle degeneration/regeneration. We found that in both systems myosin Va and PKA type I accumulate beneath the NMJs in a fiber maturation-dependent manner. Morphologically intact NMJs were found to express stable nicotinic acetylcholine receptors and to accumulate myosin Va and PKA type I in the subsynaptic region. Subsynaptic cAMP signaling was strongly altered in dystrophic muscle, particularly in fibers with severely subverted NMJ morphology. CONCLUSIONS/SIGNIFICANCE: Our data show a correlation between the subsynaptic accumulation of myosin Va and PKA type I on the one hand and NMJ regeneration status and morphology, AChR stability and specificity of subsynaptic cAMP handling on the other hand. This suggests an important role of myosin Va and PKA type I for the maturation of NMJs in regenerating muscle.


Assuntos
Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Miosina Tipo V/metabolismo , Junção Neuromuscular/enzimologia , Junção Neuromuscular/fisiopatologia , Regeneração , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , AMP Cíclico/metabolismo , Venenos Elapídicos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Distrofia Muscular Animal/patologia , Junção Neuromuscular/efeitos dos fármacos , Junção Neuromuscular/patologia , Estabilidade Proteica/efeitos dos fármacos , Receptores Colinérgicos/metabolismo , Regeneração/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Sinapses/patologia
20.
Sci Signal ; 5(210): ra12, 2012 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-22317922

RESUMO

Macrophage ingestion of the yeast Candida albicans requires its recognition by multiple receptors and the activation of diverse signaling programs. Synthesis of the lipid mediator prostaglandin E(2) (PGE(2)) and generation of cyclic adenosine monophosphate (cAMP) also accompany this process. Here, we characterized the mechanisms underlying PGE(2)-mediated inhibition of phagocytosis and filamentous actin (F-actin) polymerization in response to ingestion of C. albicans by alveolar macrophages. PGE(2) suppressed phagocytosis and F-actin formation through the PGE(2) receptors EP2 and EP4, cAMP, and activation of types I and II protein kinase A. Dephosphorylation and activation of the actin depolymerizing factor cofilin-1 were necessary for these inhibitory effects of PGE(2). PGE(2)-dependent activation of cofilin-1 was mediated by the protein phosphatase activity of PTEN (phosphatase and tensin homolog deleted on chromosome 10), with which it directly associated. Because enhanced production of PGE(2) accompanies many immunosuppressed states, the PTEN-dependent pathway described here may contribute to impaired antifungal defenses.


Assuntos
Candida albicans/imunologia , Candidíase/imunologia , Cofilina 1/imunologia , Dinoprostona/imunologia , Tolerância Imunológica , Macrófagos Alveolares/imunologia , PTEN Fosfo-Hidrolase/imunologia , Fagocitose/imunologia , Actinas/imunologia , Actinas/metabolismo , Animais , Candida albicans/metabolismo , Candidíase/metabolismo , Células Cultivadas , Cofilina 1/metabolismo , AMP Cíclico/imunologia , AMP Cíclico/metabolismo , Proteína Quinase Tipo I Dependente de AMP Cíclico/imunologia , Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , Proteína Quinase Tipo II Dependente de AMP Cíclico/imunologia , Proteína Quinase Tipo II Dependente de AMP Cíclico/metabolismo , Dinoprostona/biossíntese , Feminino , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/microbiologia , PTEN Fosfo-Hidrolase/metabolismo , Fosforilação/imunologia , Ratos , Ratos Wistar , Receptores de Prostaglandina E Subtipo EP2/imunologia , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Receptores de Prostaglandina E Subtipo EP4/imunologia , Receptores de Prostaglandina E Subtipo EP4/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...