Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.304
Filter
1.
Reprod Sci ; 30(4): 1281-1290, 2023 04.
Article in English | MEDLINE | ID: mdl-36207578

ABSTRACT

Tideglusib is considered to be a promising alternative to nonyl alcohol-9 contraceptives. Previous studies have demonstrated that the rapid spermicidal effect of tideglusib at a high concentration (≥ 10 µM) may occur through detergent-like activity; however, the effect of low concentrations of tideglusib (< 5 µM) on sperm is unknown. We explored the intracellular mechanism of tideglusib (< 5 µM) on the immobilization of human sperm by exploring related signaling pathways in human sperm. After treatment with tideglusib (1.25 µM) for 2 h, sperm motility rate decreased to 0, while sperm membrane integrity rate was 70%. Protein tyrosine phosphorylation level and intracellular cyclic adenosine 3,5-monophosphate (cAMP) concentration decreased significantly compared to those in the control group. Isobutylmethylxanthine and 8-Bromo-cAMP relieved the inhibition of spermatozoa tyrosine phosphorylation, while tyrosine phosphorylation of sperm protein in the H89 and CALP1 treatment groups was significantly inhibited, and there was no difference in the tideglusib treatment group. H-89 and CALP1 reduced the level of serine phosphorylation of GSK-3α/ß (Ser21/9), while its level was enhanced by IBMX and 8-Bromo-cAMP. Our results show the existence of the GSK3-cAMP/PKA regulatory loop in human sperm, which may mediate the immobilization effect of tideglusib at low of concentrations (e.g., 1.25 µM) on sperm motility.


Subject(s)
Cyclic AMP , Glycogen Synthase Kinase 3 , Humans , Male , Cyclic AMP/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3/pharmacology , 8-Bromo Cyclic Adenosine Monophosphate/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Sperm Motility/physiology , Semen/metabolism , Spermatozoa/metabolism , Phosphorylation , Tyrosine/metabolism
2.
Sci Rep ; 11(1): 19453, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34593918

ABSTRACT

Angiogenesis is physiologically essential for embryogenesis and development and reinitiated in adult animals during tissue growth and repair. Forming new vessels from the walls of existing vessels occurs as a multistep process coordinated by sprouting, branching, and a new lumenized network formation. However, little is known regarding the molecular mechanisms that form new tubular structures, especially molecules regulating the proper network density of newly formed capillaries. This study conducted microarray analyses in human primary microvascular endothelial cells (HMVECs) plated on Matrigel. The RAPGEF4 gene that encodes exchange proteins directly activated by cAMP 2 (EPAC2) proteins was increased in Matrigel-driven tubulogenesis. Tube formation was suppressed by the overexpression of EPAC2 and enhanced by EPAC2 knockdown in endothelial cells. Endothelial cell morphology was changed to round cell morphology by EPAC2 overexpression, while EPAC2 knockdown showed an elongated cell shape with filopodia-like protrusions. Furthermore, increased EPAC2 inhibited endothelial cell migration, and ablation of EPAC2 inversely enhanced cell mobility. These results suggest that EPAC2 affects the morphology and migration of microvascular endothelial cells and is involved in the termination and proper network formation of vascular tubes.


Subject(s)
Endothelial Cells/drug effects , Endothelium, Vascular/growth & development , Guanine Nucleotide Exchange Factors/metabolism , Morphogenesis , 8-Bromo Cyclic Adenosine Monophosphate/analogs & derivatives , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Cell Movement , Cell Shape , Cells, Cultured , Collagen , Drug Combinations , Guanine Nucleotide Exchange Factors/genetics , HEK293 Cells , Humans , Laminin , Proteoglycans , Pseudopodia
3.
Gen Comp Endocrinol ; 295: 113535, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32535173

ABSTRACT

The steroidogenic acute regulatory (StAR) protein performs the delivery of cholesterol from the outer to inner mitochondrial membrane. This is considered the rate-limiting step of acute steroid production, widely studied in mammals. However, there are only few reports regarding the characterization and expression of StAR protein in non-mammalian vertebrates. In this study, StAR protein sequence of Rhinella arenarum has been characterized and deduced from interrenal and testis cDNA sequences. StAR encodes a 285 amino acid protein with a conserved domain containing putative lipid binding sites. In vitro incubations showed that expression of StAR mRNA in testis, determined by qPCR, and testosterone synthesis determined by radioimmunoassay were stimulated after treatment with hCG and 8Br-cAMP. However, StAR mRNA expression results obtained with hCG show a higher stimulation than those obtained with 8Br-cAMP, even though steroidogenic production is the same with both treatments.


Subject(s)
Anura/metabolism , Phosphoproteins/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/analogs & derivatives , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Amino Acid Sequence , Androgens/biosynthesis , Animals , Chorionic Gonadotropin/pharmacology , Male , Phosphoproteins/chemistry , Phosphoproteins/genetics , Phylogeny , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tissue Distribution
4.
Am J Physiol Lung Cell Mol Physiol ; 318(5): L1074-L1083, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32186399

ABSTRACT

Activation of the inflammasome-caspase-1 axis in lung endothelial cells is emerging as a novel arm of the innate immune response to pneumonia and sepsis caused by Pseudomonas aeruginosa. Increased levels of circulating autacoids are hallmarks of pneumonia and sepsis and induce physiological responses via cAMP signaling in targeted cells. However, it is unknown whether cAMP affects other functions, such as P. aeruginosa-induced caspase-1 activation. Herein, we describe the effects of cAMP signaling on caspase-1 activation using a single cell flow cytometry-based assay. P. aeruginosa infection of cultured lung endothelial cells caused caspase-1 activation in a distinct population of cells. Unexpectedly, pharmacological cAMP elevation increased the total number of lung endothelial cells with activated caspase-1. Interestingly, addition of cAMP agonists augmented P. aeruginosa infection of lung endothelial cells as a partial explanation underlying cAMP priming of caspase-1 activation. The cAMP effect(s) appeared to function as a priming signal because addition of cAMP agonists was required either before or early during the onset of infection. However, absolute cAMP levels measured by ELISA were not predictive of cAMP-priming effects. Importantly, inhibition of de novo cAMP synthesis decreased the number of lung endothelial cells with activated caspase-1 during infection. Collectively, our data suggest that lung endothelial cells rely on cAMP signaling to prime caspase-1 activation during P. aeruginosa infection.


Subject(s)
Caspase 1/genetics , Cyclic AMP/metabolism , Endothelial Cells/metabolism , Pseudomonas aeruginosa/metabolism , Signal Transduction , 1-Methyl-3-isobutylxanthine/pharmacology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Adenine/analogs & derivatives , Adenine/pharmacology , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Animals , Caspase 1/metabolism , Cell Proliferation/drug effects , Colforsin/pharmacology , Cyclic AMP/agonists , Cyclic AMP/antagonists & inhibitors , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Dinoprostone/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/microbiology , Endothelial Cells/pathology , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Inflammasomes/drug effects , Inflammasomes/genetics , Inflammasomes/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Lung/metabolism , Lung/microbiology , Lung/pathology , Primary Cell Culture , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/growth & development , Rats , Rolipram/pharmacology , Single-Cell Analysis
5.
EBioMedicine ; 50: 343-354, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31707150

ABSTRACT

BACKGROUND: Recurrent implantation failure (RIF) remains a critical and challenging problem in assisted reproductive technology mainly due to impaired decidualization. The endocytic and transcytotic activity in the endometrium are crucial for decidualization. The most representative endocytic gene is the C-terminal Eps15 homology domain-containing 1 (EHD1), but whether EHD1-mediated endocytic function is responsible for embryo implantation during decidualization remains unclear. METHODS: A transcriptomic analysis was performed to evaluate the differentially expressed genes between the fertile control and RIF group. The expression and location of EHD1 in endometrial tissues were further examined by IHC, qRT-PCR and Western blotting. The transduction of an EHD1 recombinant adenovirus into human endometrial stromal cells was performed to investigate relevant decidualization marker genes. Additionally, a microarray analysis following the adenovirus-mediated overexpression of EHD1 was conducted to identify EHD1-related changes in HESCs, and the potential molecular mechanisms were further confirmed through immunofluorescence and coimmunoprecipitation analyses. FINDINGS: An RNA-seq analysis demonstrated that EHD1 expression was significantly higher in the mid-secretory endometrium of the RIF group than in that of the fertile control group. The analysis of the menstrual cycle showed that expression of EHD1 increased in the mid-proliferative phase and showed a gradual decrease in the mid-secretory and decidual phases. Furthermore, EHD1 overexpression impaired decidualization by suppressing the expression of prolactin and insulin-like growth factor binding protein-1 and the formation of the cytoskeleton. The mechanistic analysis revealed the EHD1 regulated LRP5/6 protein function through the endocytic pathway, and subsequently suppressed the Wnt4/ß-catenin pathway during decidualization. In addition, a Wnt4 agonist improved an impaired decidualization process. INTERPRETATION: Regulation of the EHD1-Wnt4 pathway might serve as a promising therapeutic strategy for improving endometrial receptivity in RIF women.


Subject(s)
Abortion, Habitual/genetics , Abortion, Habitual/metabolism , Decidua/metabolism , Signal Transduction , Vesicular Transport Proteins/genetics , Wnt4 Protein/metabolism , beta Catenin/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Abortion, Habitual/diagnosis , Adult , Biomarkers , Decidua/drug effects , Decidua/pathology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Female , Fluorescent Antibody Technique , Gene Expression Profiling , Humans , Immunohistochemistry , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Vesicular Transport Proteins/metabolism , Young Adult
6.
Biol Reprod ; 100(5): 1395-1405, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30772912

ABSTRACT

Impaired decidualization has been considered a major cause of infertility in adenomyosis. However, the mechanism remains poorly understood. Recent studies suggest that microRNAs (miRNA) play a crucial role in embryo implantation. The aim of the present study was to identify the role of miR-21 in human endometrial stromal cell (hESC) decidualization in vitro. To explore the roles of miR-21 in decidualization, we detected the expression of miR-21 in the endometrium of fertile control and adenomyosis patients, and analyzed the effects of miR-21 on the biological behaviors of hESC decidualization. The results demonstrated that miR-21 was downregulated in the endometrium of adenomyosis patients compared with the control endometrium. miR-21 effectively promoted the expression of the 8Br-cAMP plus medroxyprogesterone acetate (MPA)-induced hESC decidualization marker genes PRL and IGFBP-1 and morphological transformation through the modulation of KLF12 and NR4A1 expression; conversely, inhibition of miR-21 expression compromised hESC decidualization in vitro. In addition, Luciferase reporter, western blotting, and quantitative real-time PCR (qRT-PCR) assays confirmed that miR-21 interacted with the 3' untranslated region of the transcription factor KLF12 and downregulated KLF12 at the transcriptional and translational levels. KLF12 overexpression abolished miR-21-enhanced 8Br-cAMP plus MPA-induced decidualization. Taken together, these results illustrate that miR-21 promotes endometrial decidualization by inhibiting KLF12, and miR-21 overexpression reverses the poor decidual response of hESCs in patients with adenomyosis in vitro.


Subject(s)
Endometrium/cytology , Kruppel-Like Transcription Factors/metabolism , MicroRNAs/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Adult , Cells, Cultured , Endometrium/physiology , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Kruppel-Like Transcription Factors/genetics , Medroxyprogesterone Acetate/pharmacology , MicroRNAs/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
7.
Asian J Androl ; 21(4): 337-344, 2019.
Article in English | MEDLINE | ID: mdl-30632486

ABSTRACT

To interact with the egg, the spermatozoon must undergo several biochemical and motility modifications in the female reproductive tract, collectively called capacitation. Only capacitated sperm can undergo acrosomal exocytosis, near or on the egg, a process that allows the sperm to penetrate and fertilize the egg. In the present study, we investigated the involvement of cyclic adenosine monophosphate (cAMP)-dependent processes on acrosomal exocytosis. Inhibition of protein kinase A (PKA) at the end of capacitation induced acrosomal exocytosis. This process is cAMP-dependent; however, the addition of relatively high concentration of the membrane-permeable 8-bromo-cAMP (8Br-cAMP, 0.1 mmol l-1) analog induced significant inhibition of the acrosomal exocytosis. The induction of acrosomal exocytosis by PKA inhibition was significantly inhibited by an exchange protein directly activated by cAMP (EPAC) ESI09 inhibitor. The EPAC selective substrate activated AE at relatively low concentrations (0.02-0.1 µmol l-1), whereas higher concentrations (>5 µmol l-1) were inhibitory to the AE induced by PKA inhibition. Inhibition of PKA revealed about 50% increase in intracellular cAMP levels, conditions under which EPAC can be activated to induce the AE. Induction of AE by activating the actin severing-protein, gelsolin, which causes F-actin dispersion, was inhibited by the EPAC inhibitor. The AE induced by PKA inhibition was mediated by phospholipase C activity but not by the Ca2+-channel, CatSper. Thus, inhibition of PKA at the end of the capacitation process induced EPAC/phospholipase C-dependent acrosomal exocytosis. EPAC mediates F-actin depolymerization and/or activation of effectors downstream to F-actin breakdown that lead to acrosomal exocytosis.


Subject(s)
Acrosome Reaction/drug effects , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Exocytosis/drug effects , Guanine Nucleotide Exchange Factors/metabolism , Protein Kinase Inhibitors/pharmacology , Spermatozoa/drug effects , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Acrosome/drug effects , Acrosome/metabolism , Calcimycin/pharmacology , Cyclic AMP/metabolism , Humans , Male , Signal Transduction/drug effects , Spermatozoa/metabolism , Thapsigargin/pharmacology
8.
Pharmacology ; 103(1-2): 76-81, 2019.
Article in English | MEDLINE | ID: mdl-30517935

ABSTRACT

Dextromethorphan (DEX) presynaptically decreases glutamatergic transmission in second-order neurons of the nucleus tractus solitarius (TS). To clarify the inhibitory mechanism of DEX, the present study examined the interaction of DEX with cAMP. The effects of DEX on miniature and TS-evoked excitatory postsynaptic currents (mEPSCs and eEPSCs) were recorded under activation of the cAMP-dependent pathway using the brainstem slices. An increase in cAMP by forskolin counteracted the inhibitory effect of DEX on mEPSCs. Eight-Bromo-cAMP and N-ethylmaleimide also attenuated the DEX effect. However, forskolin had negligible effects on the DEX-induced inhibition of eEPSCs. This suggests that DEX decreases spontaneous glutamate release by inhibiting the cAMP-dependent pathway and synchronous release by another unknown mechanism.


Subject(s)
Cyclic AMP/metabolism , Dextromethorphan/pharmacology , Glutamates/metabolism , Neurons/drug effects , Solitary Nucleus/drug effects , Solitary Nucleus/physiology , Synaptic Transmission/drug effects , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Colforsin/pharmacology , Ethylmaleimide/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Guinea Pigs , Male , Miniature Postsynaptic Potentials/drug effects , Neurons/metabolism , Neurons/physiology , Solitary Nucleus/metabolism , Synaptic Transmission/physiology
9.
Asian Journal of Andrology ; (6): 337-344, 2019.
Article in English | WPRIM (Western Pacific) | ID: wpr-1009691

ABSTRACT

To interact with the egg, the spermatozoon must undergo several biochemical and motility modifications in the female reproductive tract, collectively called capacitation. Only capacitated sperm can undergo acrosomal exocytosis, near or on the egg, a process that allows the sperm to penetrate and fertilize the egg. In the present study, we investigated the involvement of cyclic adenosine monophosphate (cAMP)-dependent processes on acrosomal exocytosis. Inhibition of protein kinase A (PKA) at the end of capacitation induced acrosomal exocytosis. This process is cAMP-dependent; however, the addition of relatively high concentration of the membrane-permeable 8-bromo-cAMP (8Br-cAMP, 0.1 mmol l-1) analog induced significant inhibition of the acrosomal exocytosis. The induction of acrosomal exocytosis by PKA inhibition was significantly inhibited by an exchange protein directly activated by cAMP (EPAC) ESI09 inhibitor. The EPAC selective substrate activated AE at relatively low concentrations (0.02-0.1 μmol l-1), whereas higher concentrations (>5 μmol l-1) were inhibitory to the AE induced by PKA inhibition. Inhibition of PKA revealed about 50% increase in intracellular cAMP levels, conditions under which EPAC can be activated to induce the AE. Induction of AE by activating the actin severing-protein, gelsolin, which causes F-actin dispersion, was inhibited by the EPAC inhibitor. The AE induced by PKA inhibition was mediated by phospholipase C activity but not by the Ca2+-channel, CatSper. Thus, inhibition of PKA at the end of the capacitation process induced EPAC/phospholipase C-dependent acrosomal exocytosis. EPAC mediates F-actin depolymerization and/or activation of effectors downstream to F-actin breakdown that lead to acrosomal exocytosis.


Subject(s)
Humans , Male , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Acrosome/metabolism , Acrosome Reaction/drug effects , Calcimycin/pharmacology , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Exocytosis/drug effects , Guanine Nucleotide Exchange Factors/metabolism , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Spermatozoa/metabolism , Thapsigargin/pharmacology
10.
Cell Physiol Biochem ; 48(6): 2399-2408, 2018.
Article in English | MEDLINE | ID: mdl-30121651

ABSTRACT

BACKGROUND/AIMS: High mobility group box 1 (Hmgb1) is associated with a variety of physiological processes including embryonic development, cell proliferation and differentiation, but little information is available regarding its biological role in decidualization. METHODS: In situ hybridization, real-time PCR, RNA interference, gene overexpression and MTS assay were used to analyze the spatiotemporal expression of Hmgb1 in mouse uterus during the pre-implantation period, and explore its function and regulatory mechanisms during uterine decidualization. RESULTS: Hmgb1 mRNA was obviously observed in uterine epithelium on day 2 and 3 of pregnancy, but its expression was scarcely detected on day 4 of pregnancy. With the onset of embryo implantation, abundant Hmgb1 expression was noted in the subluminal stromal cells around the implanting blastocyst at implantation sites. Meanwhile, the accumulation of Hmgb1 mRNA was visualized in the decidual cells. Hmgb1 advanced the proliferation of uterine stromal cells and induced the expression of prolactin family 8, subfamily a, member 2 (Prl8a2), a reliable differentiation marker for decidualization. In uterine stromal cells, cAMP analogue 8-Br-cAMP up-regulated the expression of Hmgb1, but the up-regulation was abrogated by protein kinase A (PKA) inhibitor H89. Silencing of Hmgb1 by specific siRNA impeded the induction of 8-Br-cAMP on Prl8a2. Further analysis evidenced that Hmgb1 was a critical mediator of Kruppel-like factor 5 (Klf5) function in stromal differentiation. Knockdown of bone morphogenetic protein 2 (Bmp2) prevented the up-regulation of Prl8a2 elicited by Hmgb1 overexpression, whereas addition of exogenous recombinant Bmp2 protein (rBmp2) reversed the repression of Hmgb1 siRNA on Prl8a2 expression. CONCLUSION: Hmgb1 may play an important role during mouse uterine decidualization.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Cell Differentiation/drug effects , HMGB1 Protein/metabolism , Kruppel-Like Transcription Factors/metabolism , Prolactin/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Bone Morphogenetic Protein 2/antagonists & inhibitors , Bone Morphogenetic Protein 2/genetics , Cell Proliferation/drug effects , Cells, Cultured , Embryo Implantation , Female , HMGB1 Protein/antagonists & inhibitors , HMGB1 Protein/genetics , Isoquinolines/pharmacology , Kruppel-Like Transcription Factors/genetics , Mice , Pregnancy , Prolactin/genetics , RNA Interference , RNA, Small Interfering/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/pharmacology , Stromal Cells/cytology , Stromal Cells/metabolism , Sulfonamides/pharmacology , Up-Regulation/drug effects , Uterus/cytology
11.
Diabetes ; 67(10): 1999-2011, 2018 10.
Article in English | MEDLINE | ID: mdl-29986926

ABSTRACT

Glucose metabolism stimulates cell division control protein 42 homolog (Cdc42)-p21-activated kinase (Pak1) activity and initiates filamentous actin (F-actin) cytoskeleton remodeling in pancreatic ß-cells so that cytoplasmic secretory granules can translocate to the plasma membrane where insulin exocytosis occurs. Since glucose metabolism also generates cAMP in ß-cells, the cross talk of cAMP signaling with Cdc42-Pak1 activation might be of fundamental importance to glucose-stimulated insulin secretion (GSIS). Previously, the type-2 isoform of cAMP-regulated guanine nucleotide exchange factor 2 (Epac2) was established to mediate a potentiation of GSIS by cAMP-elevating agents. Here we report that nondiabetic human islets and INS-1 832/13 ß-cells treated with the selective Epac activator 8-pCPT-2'-O-Me-cAMP-AM exhibited Cdc42-Pak1 activation at 1 mmol/L glucose and that the magnitude of this effect was equivalent to that which was measured during stimulation with 20 mmol/L glucose in the absence of 8-pCPT-2'-O-Me-cAMP-AM. Conversely, the cAMP antagonist Rp-8-Br-cAMPS-pAB prevented glucose-stimulated Cdc42-Pak1 activation, thereby blocking GSIS while also increasing cellular F-actin content. Although islets from donors with type 2 diabetes had profound defects in glucose-stimulated Cdc42-Pak1 activation and insulin secretion, these defects were rescued by the Epac activator so that GSIS was restored. Collectively, these findings indicate an unexpected role for cAMP as a permissive or direct metabolic coupling factor in support of GSIS that is Epac2 and Cdc42-Pak1 regulated.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucose/pharmacology , Insulin/metabolism , Islets of Langerhans/metabolism , cdc42 GTP-Binding Protein/metabolism , p21-Activated Kinases/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/analogs & derivatives , 8-Bromo Cyclic Adenosine Monophosphate/chemistry , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Cell Line , Guanine Nucleotide Exchange Factors/metabolism , Humans , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Islets of Langerhans/drug effects , Rats , Thionucleotides/chemistry , Thionucleotides/pharmacology
12.
Endocr J ; 65(8): 833-840, 2018 Aug 27.
Article in English | MEDLINE | ID: mdl-29887570

ABSTRACT

Although curcumin was widely applied as a functional food for different diseases, it was found to reduce serum testosterone level and fertility in male animals by unknown molecular mechanisms. Here in our study, we investigated the possible mechanisms of curcumin-suppressed testosterone production in Leydig cells. Our enzyme immunoassay results showed that curcumin cell-autonomously suppressed ovine luteinizing hormone-stimulated testosterone production in primary Leydig cells and 8-bromo-cyclic adenosine monophosphate (8-br-cAMP)-induced progesterone production in MA-10 cells. Furthermore, our real-time PCR, Western blot, and 22R-OHC/pregnenolone supplementing experiment data demonstrated that curcumin suppressed 8-br-cAMP-induced steroidogenesis in Leydig cells by inhibiting the expression of StAR and Cyp11a1. Interestingly, our Western blot data showed that although curcumin suppressed PKA activity, it did not alter the 8-br-cAMP-induced phosphorylation of CREB. On the contrary, the real-time PCR results showed that curcumin suppressed 8-br-cAMP-induced expression of Nr5a1 and Fos, which are crucial for cAMP-stimulated StAR and Cyp11a1 expression in Leydig cells. Collectively, our data demonstrated that curcumin may suppress cAMP-induced steroidogenesis in mouse Leydig cells by down-regulating Nr5a1/Fos-controlled StAR and Cyp11a1 expression independently of the PKA-CREB signaling pathway.


Subject(s)
Cholesterol Side-Chain Cleavage Enzyme/metabolism , Curcumin/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Down-Regulation/drug effects , Leydig Cells/drug effects , Phosphoproteins/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Cell Line , Leydig Cells/metabolism , Luteinizing Hormone/pharmacology , Male , Mice , Progesterone/biosynthesis , Signal Transduction/physiology , Testosterone/biosynthesis
13.
Cereb Cortex ; 28(4): 1458-1471, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29351585

ABSTRACT

Neurons in primary visual cortex (V1) are more resilient than those in dorsolateral prefrontal cortex (dlPFC) in aging, schizophrenia and Alzheimer's disease. The current study compared glutamate and neuromodulatory actions in macaque V1 to those in dlPFC, and found striking regional differences. V1 neuronal firing to visual stimuli depended on AMPA receptors, with subtle NMDA receptor contributions, while dlPFC depends primarily on NMDA receptors. Neuromodulatory actions also differed between regions. In V1, cAMP signaling increased neuronal firing, and the phosphodiesterase PDE4A was positioned to regulate cAMP effects on glutamate release from axons. HCN channels in V1 were classically located on distal dendrites, and enhanced cell firing. These data contrast with dlPFC, where PDE4A and HCN channels are concentrated in thin spines, and cAMP-HCN signaling gates inputs and weakens firing. These regional differences may explain why V1 neurons are more resilient than dlPFC neurons to the challenges of age and disease.


Subject(s)
Nerve Net/physiology , Neurons/physiology , Prefrontal Cortex/cytology , Synapses/physiology , Visual Cortex/cytology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Cardiovascular Agents/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/ultrastructure , Dendritic Spines/drug effects , Dendritic Spines/ultrastructure , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/ultrastructure , Macaca mulatta , Membrane Potentials/drug effects , Microscopy, Immunoelectron , Nerve Net/drug effects , Neurons/drug effects , Neurons/ultrastructure , Photic Stimulation , Pyrimidines/pharmacology , Signal Transduction/drug effects , Synapses/drug effects , Synapses/ultrastructure
14.
J Cell Physiol ; 233(2): 1612-1626, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28657144

ABSTRACT

Ptn is a pleiotropic growth factor involving in the regulation of cellular proliferation and differentiation, but its biological function in uterine decidualization remains unknown. Here, we showed that Ptn was highly expressed in the decidual cells, and could induce the proliferation of uterine stromal cells and expression of Prl8a2 and Prl3c1 which were two well-established differentiation markers for decidualization, suggesting an important role of Ptn in decidualization. In the uterine stromal cells, progesterone stimulated the expression of Ptn accompanied with an accumulation of intracellular cAMP level. Silencing of Ptn impeded the induction of progesterone and cAMP on the differentiation of uterine stromal cells. Administration of PKA inhibitor H89 resulted in a blockage of progesterone on Ptn expression. Further analysis evidenced that regulation of progesterone and cAMP on Ptn was mediated by C/EBPß. During in vitro decidualization, knockdown of Ptn could weaken the up-regulation of Prl8a2 and Prl3c1 elicited by C/EBPß overexpression, while constitutive activation of Ptn reversed the repressive effects of C/EBPß siRNA on the expression of Prl8a2 and Prl3c1. Meanwhile, Ptn might mediate the regulation of C/EBPß on Hand2 which was a downstream target of Ptn in the differentiation of uterine stromal cells. Attenuation of Ptn or C/EBPß by specific siRNA blocked the stimulation of Hand2 by progesterone and cAMP. Collectively, Ptn may play a vital role in the progesterone-induced decidualization pathway.


Subject(s)
8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Basic Helix-Loop-Helix Transcription Factors/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , Carrier Proteins/metabolism , Cell Differentiation/drug effects , Cytokines/metabolism , Decidua/drug effects , Progesterone/pharmacology , Stromal Cells/drug effects , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , CCAAT-Enhancer-Binding Protein-beta/genetics , Carrier Proteins/genetics , Cell Proliferation/drug effects , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytokines/genetics , Decidua/cytology , Decidua/metabolism , Female , Gene Expression Regulation , Mice , Pregnancy , Prolactin/analogs & derivatives , Prolactin/genetics , Prolactin/metabolism , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Stromal Cells/metabolism , Time Factors , Transfection
15.
Mol Cell Endocrinol ; 461: 79-88, 2018 02 05.
Article in English | MEDLINE | ID: mdl-28859905

ABSTRACT

Atrazine (ATR) alters female reproductive functions in different animal species. Here, we analyzed whether ATR disturbs steroidogenic and ovulatory processes in hormone-stimulated human cumulus granulosa cells and mechanism of its action. Results showed that treatment of human cumulus granulosa cells with 20 µM ATR for 48 h resulted in lower FSH-stimulated estradiol and progesterone production. ATR reduced mRNA levels of aromatase (CYP19A1), steroidogenic acute regulatory protein (STAR) and luteinizing hormone/choriogonadotropin receptor (LHCGR). Addition of hCG 48 h after FSH and ATR treatment did not trigger maximal expression of the ovulatory genes amphiregulin (AREG) and epiregulin (EREG). Mechanistic experiments showed that ATR activated cPDE and decreased cAMP level. Addition of total PDE and specific PDE4 inhibitors, IBMX and rolipram, prevented ATR's action on CYP19A1 and STAR mRNA expression in FSH-stimulated human cumulus granulosa cells. This study suggests that ATR alters steroidogenesis and ovulatory process in human cumulus granulosa cells jeopardizing female reproduction.


Subject(s)
Atrazine/toxicity , Cumulus Cells/metabolism , Cyclic AMP/metabolism , Follicle Stimulating Hormone/pharmacology , Luteinizing Hormone/metabolism , Ovulation/genetics , Phosphoric Diester Hydrolases/metabolism , Steroids/biosynthesis , 1-Methyl-3-isobutylxanthine/pharmacology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Cell Survival/drug effects , Colforsin/pharmacology , Cumulus Cells/drug effects , Estradiol/biosynthesis , Female , Gene Expression Regulation/drug effects , Humans , Ovulation/drug effects , Phosphodiesterase Inhibitors/pharmacology , Progesterone/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rolipram/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics
16.
Neuropharmacology ; 128: 255-268, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29042318

ABSTRACT

The interaction between NMDA receptors and µ-opioid receptors in primary afferent terminals was studied by using NMDA to induce substance P release, measured as neurokinin 1 receptor internalization. In rat spinal cord slices, the µ-opioid receptor agonists morphine, DAMGO and endomorphin-2 inhibited NMDA-induced substance P release, whereas the antagonist CTAP right-shifted the concentration response of DAMGO. In vivo, substance P release induced by intrathecal NMDA after priming with BDNF was inhibited by DAMGO. ω-Conotoxins MVIIC and GVIA inhibited about half of the NMDA-induced substance P release, showing that it was partially mediated by the opening of voltage-gated calcium (Cav) channels. In contrast, DAMGO or ω-conotoxins did not inhibit capsaicin-induced substance P release. In cultured DRG neurons, DAMGO but not ω-conotoxin inhibited NMDA-induced increases in intracellular calcium, indicating that µ-opioid receptors can inhibit NMDA receptor function by mechanisms other than inactivation of Cav channels. Moreover, DAMGO decreased the ω-conotoxin-insensitive component of the substance P release. Potent inhibition by ifenprodil showed that these NMDA receptors have the NR2B subunit. Activators of adenylyl cyclase and protein kinase A (PKA) induced substance P release and this was decreased by the NMDA receptor blocker MK-801 and by DAMGO. Conversely, inhibitors of adenylyl cyclase and PKA, but not of protein kinase C, decreased NMDA-induced substance P release. Hence, these NMDA receptors are positively modulated by the adenylyl cyclase-PKA pathway, which is inhibited by µ-opioid receptors. In conclusion, µ-opioid receptors inhibit NMDA receptor-induced substance P release through Cav channel inactivation and adenylyl cyclase inhibition.


Subject(s)
Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Opioid, mu/metabolism , Spinal Cord/metabolism , Substance P/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Analgesics, Opioid/pharmacology , Animals , Calcium Channel Blockers/pharmacology , Cells, Cultured , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agents/pharmacology , Male , Morphine/pharmacology , Narcotic Antagonists/pharmacology , Neurons/drug effects , Neurons/metabolism , Protein Transport/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Neurokinin-1/metabolism , Spinal Cord/cytology , Spinal Cord/drug effects , omega-Conotoxins/pharmacology
17.
Epilepsia ; 59(2): 449-459, 2018 02.
Article in English | MEDLINE | ID: mdl-29283181

ABSTRACT

OBJECTIVE: γ-Aminobutyric acid (GABA) is the major inhibitory neurotransmitter in adult central nervous system, and profound alterations of GABA receptor functions are linked to temporal lobe epilepsy (TLE). Here we describe the functional relationships between GABA receptors type B (GABAB R) and type A (GABAA R) in human temporal cortex and how TLE affects this aspect of GABAergic signaling. METHODS: Miniature inhibitory postsynaptic currents (mIPSCs) were recorded by patch-clamp techniques from human L5 pyramidal neurons in slices from temporal cortex tissue obtained from surgery. RESULTS: We describe a constitutive functional crosstalk between GABAB Rs and GABAA Rs in human temporal layer 5 pyramidal neurons, which is lost in epileptic tissues. The activation of GABAB Rs by baclofen, in addition to the expected reduction of mIPSC frequency, produced, in cortex of nonepileptic patients, the prolongation of mIPSC rise and decay times, thus increasing the inhibitory net charge associated with a single synaptic event. Block of K+ channels did not prevent the increase of decay time and charge. Protein kinase A (PKA) blocker KT5720 and pertussis toxin inhibited the action of baclofen, whereas 8Br-cAMP mimicked the GABAB R action. The same GABAB R-mediated modulation of GABAA Rs was observed in pyramidal neurons of rat temporal cortex, with both PKA and PKC involved in the process. In cortices from TLE patients and epileptic rats, baclofen lost its ability to modulate mIPSCs. SIGNIFICANCE: Our results highlight the association of TLE with functional changes of GABAergic signaling that may be related to seizure propagation, and suggest that the selective activation of a definite subset of nonpresynaptic GABAB Rs may be therapeutically useful in TLE.


Subject(s)
Epilepsy, Temporal Lobe/metabolism , Neocortex/metabolism , Pyramidal Cells/metabolism , Receptors, GABA-A/metabolism , Receptors, GABA-B/metabolism , Temporal Lobe/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Adolescent , Adult , Animals , Baclofen/pharmacology , Carbazoles/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Disease Models, Animal , Drug Resistant Epilepsy/metabolism , Drug Resistant Epilepsy/physiopathology , Drug Resistant Epilepsy/surgery , Enzyme Inhibitors/pharmacology , Epilepsy/chemically induced , Epilepsy/metabolism , Epilepsy/physiopathology , Epilepsy, Temporal Lobe/physiopathology , Epilepsy, Temporal Lobe/surgery , Female , GABA-B Receptor Agonists/pharmacology , Humans , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Male , Middle Aged , Muscarinic Agonists/toxicity , Neocortex/drug effects , Neocortex/physiopathology , Patch-Clamp Techniques , Pertussis Toxin/pharmacology , Pilocarpine/toxicity , Protein Kinase C/metabolism , Pyramidal Cells/drug effects , Pyrroles/pharmacology , Rats , Temporal Lobe/drug effects , Temporal Lobe/physiopathology
18.
Stem Cell Res Ther ; 8(1): 246, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-29096715

ABSTRACT

BACKGROUND: Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs) are a novel and promising strategy for tissue engineering because of their ability to differentiate into many cell types. We characterized the differentiation of WJ-MSCs into endometrial epithelial cell (EEC)-like and endometrial stromal cell (ESC)-like cells and assessed the effect of 17ß-estradiol and 8-Br-cAMP on the differentiation system. METHODS: WJ-MSCs were treated in two ways to differentiate into EEC-like and ESC-like cells respectively: cocultured with ESCs in control/differentiation medium (17ß-estradiol, growth factors); and cultured in control/differentiation medium (8-Br-cAMP alone or 8-Br-cAMP plus 17ß-estrogen and growth factors). Three signaling pathway inhibitors (SB203580, PD98059, H89) were used to investigate the mechanism of WJ-MSC differentiation into ESC-like cells. Immunofluorescence, western blot and flow cytometry analyses were used to analyze expression of epithelial markers and stromal cell markers. Enzyme-linked immunosorbent assays were used to test the production of secretory proteins associated with the differentiation of ESC-like cells. RESULTS: 17ß-estradiol at 1 µM downregulated vimentin and CD13 and upregulated cytokeratin and CD9 proteins, promoting the differentiation of WJ-MSCs into EEC-like cells in the coculture system. 8-Br-cAMP at 0.5 mM upregulated vimentin and CD13 and downregulated CK and CD9, promoting the differentiation of WJ-MSCs into ESC-like cells. Prolactin (PRL) and insulin-like growth factor-binding protein 1 (IGFBP1) were upregulated and the protein kinase A (PKA) signaling pathway was activated, whereas extracellular signal-regulated (ERK)1/2 and p38 mitogen-activated protein kinase (MAPK) were not affected. CONCLUSIONS: 17ß-estradiol at 1 µM is a good inducer for facilitating the differentiation of WJ-MSCs into EEC-like cells. 8-Br-cAMP plus estrogen and growth factors can induce the differentiation of WJ-MSCs into ESC-like cells. During the differentiation of WJ-MSCs into ESC-like cells, PRL and IGFBP1 were upregulated by the treatment and the PKA signaling pathway was activated, whereas ERK1/2 and p38 MAPK were not affected. These findings suggest a promising approach to the treatment of endometrial damage and other endometrial diseases and suggest new applications for WJ-MSCs in clinical practice.


Subject(s)
Cell Differentiation/drug effects , Epithelial Cells/drug effects , Gene Expression Regulation/drug effects , Mesenchymal Stem Cells/drug effects , Stromal Cells/drug effects , Wharton Jelly/drug effects , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , CD13 Antigens/genetics , CD13 Antigens/metabolism , Cell Proliferation/drug effects , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Endometrium/cytology , Endometrium/drug effects , Endometrium/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Estradiol/pharmacology , Female , Flavonoids/pharmacology , Humans , Imidazoles/pharmacology , Insulin-Like Growth Factor Binding Protein 1/genetics , Insulin-Like Growth Factor Binding Protein 1/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Isoquinolines/pharmacology , Keratins/genetics , Keratins/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Primary Cell Culture , Prolactin/genetics , Prolactin/metabolism , Pyridines/pharmacology , Signal Transduction , Stromal Cells/cytology , Stromal Cells/metabolism , Sulfonamides/pharmacology , Tetraspanin 29/genetics , Tetraspanin 29/metabolism , Umbilical Cord/cytology , Umbilical Cord/drug effects , Umbilical Cord/metabolism , Vimentin/genetics , Vimentin/metabolism , Wharton Jelly/cytology , Wharton Jelly/metabolism
19.
Am J Physiol Cell Physiol ; 313(6): C593-C603, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28855161

ABSTRACT

Red blood cell (RBC)-derived adenosine triphosphate (ATP) has been proposed as an integral component in the regulation of oxygen supply to skeletal muscle. In ex vivo settings RBCs have been shown to release ATP in response to a number of stimuli, including stimulation of adrenergic receptors. Further evidence suggested that ATP release from RBCs was dependent on activation of adenylate cyclase (AC)/cyclic adenosine monophosphate (cAMP)-dependent pathways and involved the pannexin 1 (Panx1) channel. Here we show that RBCs express Panx1 and confirm its absence in Panx1 knockout (-/-) RBCs. However, Panx1-/- mice lack any decrease in exercise performance, challenging the assumptions that Panx1 plays an essential role in increased blood perfusion to exercising skeletal muscle and therefore in ATP release from RBCs. We therefore tested the role of Panx1 in ATP release from RBCs ex vivo in RBC suspensions. We found that stimulation with hypotonic potassium gluconate buffer resulted in a significant increase in ATP in the supernatant, but this was highly correlated with RBC lysis. Next, we treated RBCs with a stable cAMP analog, which did not induce ATP release from wild-type or Panx1-/- mice. Similarly, multiple pharmacological treatments activating AC in RBCs increased intracellular cAMP levels (as measured via mass spectrometry) but did not induce ATP release. The data presented here question the importance of Panx1 for exercise performance and dispute the general assumption that ATP release from RBCs via Panx1 is regulated via cAMP.


Subject(s)
Adenosine Triphosphate/blood , Connexins/blood , Cyclic AMP/blood , Energy Metabolism , Erythrocytes/metabolism , Muscle, Skeletal/metabolism , Nerve Tissue Proteins/blood , Second Messenger Systems , 1-Methyl-3-isobutylxanthine/pharmacology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Adenosine Triphosphate/metabolism , Adenylyl Cyclases/blood , Adult , Animals , Colforsin/pharmacology , Connexins/deficiency , Connexins/genetics , Energy Metabolism/drug effects , Erythrocytes/drug effects , Exercise Tolerance , Female , Genotype , Gluconates/pharmacology , Hemolysis , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle Contraction , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Phenotype , Time Factors , Young Adult
20.
J Mol Cell Cardiol ; 111: 51-60, 2017 10.
Article in English | MEDLINE | ID: mdl-28778766

ABSTRACT

Ovariectomy (OVX) promotes sarcoplasmic reticulum (SR) Ca2+ overload in ventricular myocytes. We hypothesized that the cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) pathway contributes to this Ca2+ dysregulation. Myocytes were isolated from adult female C57BL/6 mice following either OVX or sham surgery (surgery at ≈1mos). Contractions, Ca2+ concentrations (fura-2) and ionic currents were measured simultaneously (37°C, 2Hz) in voltage-clamped myocytes. Intracellular cAMP levels were determined with an enzyme immunoassay; phosphodiesterase (PDE) and adenylyl cyclase (AC) isoform expression was examined with qPCR. Ca2+ currents were similar in myocytes from sham and OVX mice but Ca2+ transients, excitation-contraction (EC)-coupling gain, SR content and contractions were larger in OVX than sham cells. To determine if the cAMP/PKA pathway mediated OVX-induced alterations in EC-coupling, cardiomyocytes were incubated with the PKA inhibitor H-89 (2µM), which abolished baseline differences. While basal intracellular cAMP did not differ, levels were higher in OVX than sham in the presence of a non-selective PDE inhibitor (300µM IBMX), or an AC activator (10µM forskolin). This suggests the production of cAMP by AC and its breakdown by PDE were enhanced by OVX. Consistent with this, mRNA levels for both AC5 and PDE4A were higher in OVX in comparison to sham. Differences in Ca2+ homeostasis and contractions were abolished when sham and OVX cells were dialyzed with patch pipettes containing the same concentration of 8-bromoadenosine-cAMP (50µM). Interestingly, selective inhibition of PDE4 increased Ca2+ current only in OVX cells. Together, these findings suggest that estrogen suppresses SR Ca2+ release and that this is regulated, at least in part, by the cAMP/PKA pathway. These changes in the cAMP/PKA pathway may promote Ca2+ dysregulation and cardiovascular disease when ovarian estrogen levels fall. These results advance our understanding of female-specific cardiomyocyte mechanisms that may affect responses to therapeutic interventions in older women.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Excitation Contraction Coupling , Myocardium/metabolism , Ovariectomy , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Adenylyl Cyclases/metabolism , Animals , Calcium Signaling/drug effects , Cell Size/drug effects , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Excitation Contraction Coupling/drug effects , Female , Isoquinolines/pharmacology , Mice, Inbred C57BL , Models, Biological , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Organ Size/drug effects , Protein Kinase Inhibitors/pharmacology , Rolipram/pharmacology , Sarcoplasmic Reticulum/metabolism , Sulfonamides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...