Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 348
1.
Int J Mol Sci ; 23(22)2022 Nov 16.
Article En | MEDLINE | ID: mdl-36430668

Psoralen, a major furocoumarin component of the Fructus Psoralen (FP), in combination with ultraviolet radiation, cures abnormal pigmentation disorder. In a previous study, we synthesized a series of linear furocoumarins with different substituents, out of which 5-((diethylamino)methyl)-3-phenyl-7H-furo [3,2-g] chromen-7-one (encoded as 5D3PC) showed better pigmenting effect than others in B16 cells. In this study, we examined the mechanism underlying the melanogenic effect of 5D3PC both in vivo and in vitro. To examine the pigmentation effect, the B16 and human melanocyte cell lines, PIG1 and PIG3V melanocytes were incubated with 5D3PC. In animal experiments, C57BL/6 mice received 5% hydroquinone and were administrated with 5D3PC for 30 days. 5D3PC upregulated the melanin synthesis and tyrosinase in B16 cell, PIG1 and PIG3V. The expression level of tyrosinase (TYR), tyrosinase-related protein-1 (TRP-1) and tyrosinase-related protein-2 (TRP-2), microphthalmia-associated transcription factor (MITF), cyclic adenosine monophosphate (cAMP), phosphorylation of cAMP-responsive element binding protein (p-CREB), phosphorylation of p38 mitogen-activated protein kinase (MAPK), c- phosphorylation of Jun N-terminal kinase (p-JNK) was significantly higher in 5D3PC-treated B16 cells. The oral administration of 5D3PC attenuated the depigmentation of the C57BL/6 vitiligo mice model by increasing the numbers of melanin-containing hair follicles, melanogenic protein, and melanogenesis-relative genes expression in skin tissues.


Furocoumarins , Melanins , Animals , Humans , Mice , Cyclic AMP/metabolism , Furocoumarins/pharmacology , Melanins/biosynthesis , Melanins/metabolism , Mice, Inbred C57BL , Monophenol Monooxygenase , Signal Transduction , Ultraviolet Rays , Cyclic AMP-Dependent Protein Kinases/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism
2.
Neurotoxicology ; 91: 218-227, 2022 07.
Article En | MEDLINE | ID: mdl-35643327

The inhibition of the excessive release of glutamate in the brain has emerged as a promising new option for developing therapeutic strategies for neurodegenerative disorders. This study investigated the effect and mechanism of lappaconitine, a diterpenoid alkaloid found in species of Aconitum, on glutamate release in rat cerebral cortex nerve terminals (synaptosomes). Here, we report that in the rat cortical synaptosomal preparation, lappaconitine reduced the K+ channel blocker 4-aminopyridine (4-AP)-evoked Ca2+-dependent release of glutamate. The inhibitory effect of lappaconitine on the evoked glutamate release was blocked by the vesicular transporter inhibitor bafilomycin A1 and calcium-chelating agent ethylene glycol tetraacetic acid (EGTA), but was unaffected by exposure to the glutamate transporter inhibitor dl-threo-beta-benzyloxyaspartate (dl-TBOA). The depolarization-induced elevation of cytosolic calcium concentration ([Ca2+]c) was inhibited by lappaconitine, while the 4-AP-mediated depolarization of the synaptosomal membrane potential was not affected. The inhibition of glutamate release by lappaconitine was markedly decreased in synaptosomes pretreated with the Cav2.3 (R-type) channel blocker SNX-482 or the protein kinase A inhibitor H89. Nevertheless, the lappaconitine-mediated inhibition of glutamate release was not abolished by the intracellular Ca2+-release inhibitors dantrolene and CGP37157. Lappaconitine also significantly decreased the 4-AP-induced phosphorylation of PKA and SNAP-25, a presynaptic substrate for PKA. Our data suggest that lappaconitine reduces Ca2+ influx through R-type Ca2+ channels, subsequently reducing the protein kinase A cascade to inhibit the evoked glutamate release from rat cerebral cortex nerve terminals.


Aconitine , Calcium , Cyclic AMP-Dependent Protein Kinases , Glutamic Acid , 4-Aminopyridine/metabolism , 4-Aminopyridine/pharmacology , Aconitine/analogs & derivatives , Aconitine/pharmacology , Animals , Calcium/metabolism , Cerebral Cortex/metabolism , Cyclic AMP-Dependent Protein Kinases/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Glutamic Acid/metabolism , Presynaptic Terminals/metabolism , Rats , Rats, Sprague-Dawley , Synaptosomes
3.
Int J Mol Sci ; 23(3)2022 Jan 25.
Article En | MEDLINE | ID: mdl-35163281

Calycosin, a bioactive isoflavonoid isolated from root extracts of Astragalus membranaceus, has been reported to inhibit melanogenesis, the mechanism of which remains undefined. In this study, we interrogated the mechanistic basis by which calycosin inhibits melanin production in two model systems, i.e., B16F10 melanoma cells and zebrafish embryos. Calycosin was effective in protecting B16F10 cells from α-melanocyte-stimulating hormone (α-MSH)-induced melanogenesis and tyrosinase activity. This anti-melanogenic effect was accompanied by decreased expression levels of microphthalmia-associated transcription factor (MITF), a key protein controlling melanin synthesis, and its target genes tyrosinase and tyrosinase-related protein-2 (TRP-2) in calycosin-treated cells. Mechanistically, we obtained the first evidence that calycosin-mediated MITF downregulation was attributable to its ability to block signaling pathways mediated by cAMP response element-binding protein (CREB) and p38 MAP kinase. The protein kinase A (PKA) inhibitor H-89 and p38 inhibitor SB203580 validated the premise that calycosin inhibits melanin synthesis and tyrosinase activity by regulating the PKA/CREB and p38 MAPK signaling pathways. Moreover, the in vivo anti-melanogenic efficacy of calycosin was manifested by its ability to suppress body pigmentation and tyrosinase activity in zebrafish embryos. Together, these data suggested the translational potential of calycosin to be developed as skin-lightening cosmeceuticals.


Isoflavones/pharmacology , Melanins/metabolism , Animals , Astragalus propinquus/metabolism , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Isoflavones/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Melanoma/drug therapy , Melanoma/metabolism , Microphthalmia-Associated Transcription Factor/metabolism , Phosphorylation/drug effects , Plant Extracts/pharmacology , Plant Roots , Signal Transduction/drug effects , Zebrafish/metabolism , alpha-MSH/pharmacology , p38 Mitogen-Activated Protein Kinases/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Biomed Pharmacother ; 147: 112651, 2022 Mar.
Article En | MEDLINE | ID: mdl-35063859

Abnormal melanin synthesis upon UV exposure causes excessive oxidative stress, which leads to skin hyperpigmentation disorders such as freckles, melisma, and age spots. The present study investigated the anti-melanogenic effects of decursin and the underlying mechanism using multiple approaches. Decursin exhibited no cytotoxicity and significantly reduced intracellular tyrosinase activity and melanin content in B16F10 melanoma cells. Decursin also inhibited the expression of melanogenic enzymes such as tyrosinase and tyrosinase-related protein (TRP)- 1, but not TRP-2. Mechanistically, decursin suppressed melanin synthesis through cAMP-dependent protein kinase (PKA)/cAMP response element-binding protein (CREB)-dependent downregulation of microphthalmia-associated transcription factor (MITF), a master transcription factor in melanogenesis. Further, decursin exerted anti-melanogenic effects by downregulating the p38 signaling pathway and upregulating extracellular signal-regulated kinase (ERK) and phosphatidylinositol 3 kinase (PI3K)/protein kinase B (Akt)/glycogen synthesis kinase-3ß (GSK-3ß) cascades. in silico analysis showed that decursin formed specific interactions with residues of upstream regulators of MITF and exhibited optimal pharmacokinetic profiles, including permeability and skin sensitization. Finally, the anti-melanogenic effects of decursin were confirmed ex vivo in 3D human skin models, suggesting its applicability as a protective agent against hyperpigmentation.


Benzopyrans/pharmacology , Butyrates/pharmacology , Melanins/metabolism , Microphthalmia-Associated Transcription Factor/drug effects , CREB-Binding Protein/drug effects , Cell Line , Cell Survival/drug effects , Cyclic AMP-Dependent Protein Kinases/drug effects , Glycogen Synthase Kinase 3 beta/drug effects , Humans , Mitogen-Activated Protein Kinases/drug effects , Phosphatidylinositol 3-Kinases/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Signal Transduction/drug effects , Skin/drug effects
5.
Platelets ; 32(4): 469-478, 2021 May 19.
Article En | MEDLINE | ID: mdl-32379559

This study evaluated the antiplatelet effect of the plant carotenoid, astaxanthin (ASTX) in rats fed either control or high cholesterol plus cholic acid diet (HCCD) and possible underlying mechanisms. Adult male Wistar rats were divided into four groups (n = 8/each), namely, control (fed normal diet), control + ASTX (10 mg/kg/day), HCCD-fed rats, and HCCD + ASTX-treated rats. Diets and treatments were orally administered daily for 30 days. In both control and HCCD-fed rats, ASTX significantly increased fecal levels of triglycerides and cholesterol, reduced platelet count, prolonged bleeding time, and inhibited platelet aggregation. It also reduced platelet levels of reactive oxygen species (ROS) and Bcl-2; thromboxane B2 (TXB2) release; and the expression of P2Y12, P-selectin, and CD36 receptors. Moreover, the activity NF-κB p65 and Akt was inhibited. Concomitantly, it increased the protein levels of cleaved caspase-3 and vasodilator-stimulated phosphoprotein (p-VASP) as well as intracellular levels of cAMP. However, in HCCD-fed rats, the effects of ASTX were associated with reduced serum levels of ox-LDL-c and fasting plasma glucose levels. In conclusion, antiplatelet effects of ASTX involve ROS scavenging, inhibiting NF-κB activity, down-regulating P2Y12 expression, and increasing intracellular levels of cAMP that are attributed to its antioxidant, hypolipidemic, and anti-inflammatory effects.


Cyclic AMP-Dependent Protein Kinases/drug effects , Fibrinolytic Agents/therapeutic use , NF-kappa B/metabolism , Receptors, Purinergic P2Y12/metabolism , Animals , Down-Regulation , Fibrinolytic Agents/pharmacology , Humans , Male , Rats , Rats, Wistar , Xanthophylls/pharmacology , Xanthophylls/therapeutic use
6.
J Neurosci ; 41(1): 193-210, 2021 01 06.
Article En | MEDLINE | ID: mdl-33172978

Oral squamous cell carcinoma (OSCC) is one of the most painful cancers, which interferes with orofacial function including talking and eating. We report that legumain (Lgmn) cleaves protease-activated receptor-2 (PAR2) in the acidic OSCC microenvironment to cause pain. Lgmn is a cysteine protease of late endosomes and lysosomes that can be secreted; it exhibits maximal activity in acidic environments. The role of Lgmn in PAR2-dependent cancer pain is unknown. We studied Lgmn activation in human oral cancers and oral cancer mouse models. Lgmn was activated in OSCC patient tumors, compared with matched normal oral tissue. After intraplantar, facial or lingual injection, Lgmn evoked nociception in wild-type (WT) female mice but not in female mice lacking PAR2 in NaV1.8-positive neurons (Par2Nav1.8), nor in female mice treated with a Lgmn inhibitor, LI-1. Inoculation of an OSCC cell line caused mechanical and thermal hyperalgesia that was reversed by LI-1. Par2Nav1.8 and Lgmn deletion attenuated mechanical allodynia in female mice with carcinogen-induced OSCC. Lgmn caused PAR2-dependent hyperexcitability of trigeminal neurons from WT female mice. Par2 deletion, LI-1, and inhibitors of adenylyl cyclase or protein kinase A (PKA) prevented the effects of Lgmn. Under acidified conditions, Lgmn cleaved within the extracellular N terminus of PAR2 at Asn30↓Arg31, proximal to the canonical trypsin activation site. Lgmn activated PAR2 by biased mechanisms in HEK293 cells to induce Ca2+ mobilization, cAMP formation, and PKA/protein kinase D (PKD) activation, but not ß-arrestin recruitment or PAR2 endocytosis. Thus, in the acidified OSCC microenvironment, Lgmn activates PAR2 by biased mechanisms that evoke cancer pain.SIGNIFICANCE STATEMENT Oral squamous cell carcinoma (OSCC) is one of the most painful cancers. We report that legumain (Lgmn), which exhibits maximal activity in acidic environments, cleaves protease-activated receptor-2 (PAR2) on neurons to produce OSCC pain. Active Lgmn was elevated in OSCC patient tumors, compared with matched normal oral tissue. Lgmn evokes pain-like behavior through PAR2 Exposure of pain-sensing neurons to Lgmn decreased the current required to generate an action potential through PAR2 Inhibitors of adenylyl cyclase and protein kinase A (PKA) prevented the effects of Lgmn. Lgmn activated PAR2 to induce calcium mobilization, cAMP formation, and activation of protein kinase D (PKD) and PKA, but not ß-arrestin recruitment or PAR2 endocytosis. Thus, Lgmn is a biased agonist of PAR2 that evokes cancer pain.


Cancer Pain/chemically induced , Carcinoma, Squamous Cell/complications , Cysteine Endopeptidases , Mouth Neoplasms/complications , Receptor, PAR-2/agonists , Aged , Aged, 80 and over , Animals , Arrestin/metabolism , Cancer Pain/psychology , Cyclic AMP-Dependent Protein Kinases/drug effects , Cysteine Endopeptidases/administration & dosage , Endocytosis/drug effects , Enzyme Activation/drug effects , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Protein Kinase C/drug effects , Protein Kinase Inhibitors/pharmacology , Receptor, PAR-2/genetics , Tumor Microenvironment/drug effects
7.
Diabet Med ; 38(2): e14385, 2021 02.
Article En | MEDLINE | ID: mdl-32794582

AIMS: To test whether oral administration of D/L-3-hydroxybutyrate as a sodium salt inhibits lipolysis and intracellular lipid signalling, in particular, hormone-sensitive lipase, and whether D/L-3-hydroxybutyrate alters endogenous glucose production. METHODS: We studied six young men in a randomized, controlled, crossover study after ingestion of Na-D/L-3-hydroxybutyrate (hyperketotic condition) or saline (placebo control). We quantified lipolysis and endogenous glucose production using [9,10-3 H]-palmitate and [3-3H]glucose tracers, and adipose tissue biopsies were collected to investigate key lipolytic enzymes. RESULTS: After ingestion, D/L-3-hydroxybutyrate increased by more than 2.5 mmol/l, free fatty acid concentrations decreased by >70%, and palmitate rate of appearance was halved. Protein kinase A phosphorylation of perilipin was reduced and hormone-sensitive lipase 660 phosphorylation in adipose tissue biopsies was 70-80% decreased in the hyperketotic condition and unchanged in the control. Compared to the control, endogenous glucose production was reduced by close to 20% (P<0.05) after 3-hydroxybutyrate ingestion. CONCLUSION: We conclude that oral D/L-Na-3-hydroxybutyrate increases D/L-3-hydroxybutyrate concentrations within half an hour, decreases free fatty acid concentrations, lowers lipolysis and endogenous glucose production, and dephosphorylates hormone-sensitive lipase. Collectively these phenomena may be viewed as an orchestrated feedback loop, controlling endogenous glucose production, lipolysis and ketogenesis. Such effects would be beneficial in insulin-resistant states. (www.clinicaltrials.gov ID number: NCT02917252).


3-Hydroxybutyric Acid/pharmacology , Gluconeogenesis/drug effects , Lipolysis/drug effects , Sterol Esterase/drug effects , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adult , Cross-Over Studies , Cyclic AMP-Dependent Protein Kinases/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Feedback, Physiological , Humans , Male , Perilipin-1/drug effects , Perilipin-1/metabolism , Phosphorylation/drug effects , Random Allocation , Sterol Esterase/metabolism , Young Adult
8.
J Nat Prod ; 83(9): 2718-2726, 2020 09 25.
Article En | MEDLINE | ID: mdl-32881504

Campafungin A is a polyketide that was recognized in the Candida albicans fitness test due to its antiproliferative and antihyphal activity. Its mode of action was hypothesized to involve inhibition of a cAMP-dependent PKA pathway. The originally proposed structure appeared to require a polyketide assembled in a somewhat unusual fashion. However, structural characterization data were never formally published. This background stimulated a reinvestigation in which campafungin A and three closely related minor constituents were purified from fermentations of a strain of the ascomycete fungus Plenodomus enteroleucus. Labeling studies, along with extensive NMR analysis, enabled assignment of a revised structure consistent with conventional polyketide synthetic machinery. The structure elucidation of campafungin A and new analogues encountered in this study, designated here as campafungins B, C, and D, is presented, along with a proposed biosynthetic route. The antimicrobial spectrum was expanded to methicillin-resistant Staphylococcus aureus, Candida tropicalis, Candida glabrata, Cryptococcus neoformans, Aspergillus fumigatus, and Schizosaccharomyces pombe, with MICs ranging as low as 4-8 µg mL-1 in C. neoformans. Mode-of-action studies employing libraries of C. neoformans mutants indicated that multiple pathways were affected, but mutants in PKA/cAMP pathways were unaffected, indicating that the mode of action was distinct from that observed in C. albicans.


Candida albicans/drug effects , Cryptococcus neoformans/drug effects , Hyphae/drug effects , Hyphae/growth & development , Anti-Bacterial Agents/pharmacology , Antifungal Agents/pharmacology , Ascomycota/chemistry , Ascomycota/metabolism , Bacteria/drug effects , Cyclic AMP-Dependent Protein Kinases/drug effects , Fermentation , Fungi/drug effects , Microbial Sensitivity Tests , Molecular Structure , Polyketides/pharmacology , Signal Transduction/drug effects
9.
Metabolism ; 107: 154228, 2020 06.
Article En | MEDLINE | ID: mdl-32289346

OBJECTIVE: Mangiferin (MF), a xanthonoid derived from Mangifera indica, has shown therapeutic effects on various human diseases including cancer, diabetes, and obesity. Nonetheless, the influence of MF on non-shivering thermogenesis and its underlying mechanism in browning remains unclear. Here, our aim was to investigate the effects of MF on browning and its molecular mechanisms in murine C3H10T1/2 mesenchymal stem cells (MSCs). MATERIALS/METHODS: To determine the function of MF on browning, murine C3H10T1/2 MSCs were treated with MF in an adipogenic differentiation cocktail and the thermogenic and correlated metabolic responses were assessed using MF-mediated signalling. Human adipose-derived MSCs were differentiated and treated with MF to confirm its role in thermogenic induction. RESULTS: MF treatment induced the expression of a brown-fat signature, UCP1, and reduced triglyceride (TG) in C3H10T1/2 MSCs. MF also induced the expression of major thermogenesis regulators: PGC1α, PRDM16, and PPARγ and up-regulated the expression of beiging markers CD137, HSPB7, TBX1, and COX2 in both murine C3H10T1/2 MSCs and human adipose-derived mesenchymal stem cells (hADMSC). We also observed that MF treatment increased the mitochondrial DNA and improved mitochondrial homeostasis by regulating mitofission-fusion plasticity via suppressing PINK1-PRKN-mediated mitophagy. Furthermore, MF treatment improved mitochondrial respiratory function by increasing mitochondrial oxygen consumption and expression of oxidative-phosphorylation (OXPHOS)-related proteins. Chemical-inhibition and gene knockdown experiments revealed that ß3-AR-dependent PKA-p38 MAPK-CREB signalling is crucial for MF-mediated brown-fat formation via suppression of mitophagy in C3H10T1/2 MSCs. CONCLUSIONS: MF promotes the brown adipocyte phenotype by suppressing mitophagy, which is regulated by PKA-p38MAPK-CREB signalling in C3H10T1/2 MSCs. Thus, we propose that MF may be a good browning inducer that can ameliorate obesity.


Adipose Tissue, Brown/drug effects , Mitophagy/drug effects , Mitophagy/genetics , Protein Kinases/genetics , Ubiquitin-Protein Ligases/genetics , Xanthones/pharmacology , Adipocytes, Brown/drug effects , Animals , Cell Differentiation/drug effects , Cyclic AMP-Dependent Protein Kinases/drug effects , Gene Knockdown Techniques , Humans , Mesenchymal Stem Cells/drug effects , Mice , Mice, Inbred C3H , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , Protein Kinases/drug effects , Thermogenesis/drug effects , Thermogenesis/genetics , Ubiquitin-Protein Ligases/drug effects , p38 Mitogen-Activated Protein Kinases/drug effects
10.
Arq Neuropsiquiatr ; 78(1): 21-27, 2020 01.
Article En | MEDLINE | ID: mdl-32074185

OBJECTIVE: The phytohormone abscisic acid (ABA) as a signaling molecule exists in various types of organisms from early multicellular to animal cells and tissues. It has been demonstrated that ABA has an antinociceptive effect in rodents. The present study was designed to assess the possible role of PKA and phosphorylated ERK (p-ERK) on the antinociceptive effects of intrathecal (i.t.) ABA in male Wistar rats. METHODS: The animals were cannulated intrathecally and divided into different experimental groups (n=6‒7): Control (no surgery), vehicle (received ABA vehicle), ABA-treated groups (received ABA in doses of 10 or 20 µg/rat), ABA plus H.89 (PKA inhibitor)-treated group which received the inhibitor 15 min prior to the ABA injection. Tail-flick and hot-plate tests were used as acute nociceptive stimulators to assess ABA analgesic effects. p-ERK was evaluated in the dorsal portion of the spinal cord using immunoblotting. RESULTS: Data showed that a microinjection of ABA (10 and 20 µg/rat, i.t.) significantly increased the nociceptive threshold in tail flick and hot plate tests. The application of PKA inhibitor (H.89, 100 nM/rat) significantly inhibited ABA-induced analgesic effects. Expression of p-ERK was significantly decreased in ABA-injected animals, which were not observed in the ABA+H.89-treated group. CONCLUSIONS: Overall, i.t. administration of ABA (10 µg/rat) induced analgesia and p-ERK down-expression likely by involving the PKA-dependent mechanism.


Abscisic Acid/pharmacology , Analgesics/pharmacology , Cyclic AMP-Dependent Protein Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/drug effects , Plant Growth Regulators/pharmacology , Spinal Cord/metabolism , Animals , Blotting, Western , Cyclic AMP-Dependent Protein Kinases/analysis , Extracellular Signal-Regulated MAP Kinases/analysis , Intracellular Signaling Peptides and Proteins/pharmacology , Male , Rats, Wistar , Reference Values , Reproducibility of Results , Spinal Cord/drug effects , Time Factors
11.
Arq. neuropsiquiatr ; 78(1): 21-27, Jan. 2020. graf
Article En | LILACS | ID: biblio-1088987

Abstract Objective: The phytohormone abscisic acid (ABA) as a signaling molecule exists in various types of organisms from early multicellular to animal cells and tissues. It has been demonstrated that ABA has an antinociceptive effect in rodents. The present study was designed to assess the possible role of PKA and phosphorylated ERK (p-ERK) on the antinociceptive effects of intrathecal (i.t.) ABA in male Wistar rats. Methods: The animals were cannulated intrathecally and divided into different experimental groups (n=6‒7): Control (no surgery), vehicle (received ABA vehicle), ABA-treated groups (received ABA in doses of 10 or 20 µg/rat), ABA plus H.89 (PKA inhibitor)-treated group which received the inhibitor 15 min prior to the ABA injection. Tail-flick and hot-plate tests were used as acute nociceptive stimulators to assess ABA analgesic effects. p-ERK was evaluated in the dorsal portion of the spinal cord using immunoblotting. Results: Data showed that a microinjection of ABA (10 and 20 µg/rat, i.t.) significantly increased the nociceptive threshold in tail flick and hot plate tests. The application of PKA inhibitor (H.89, 100 nM/rat) significantly inhibited ABA-induced analgesic effects. Expression of p-ERK was significantly decreased in ABA-injected animals, which were not observed in the ABA+H.89-treated group. Conclusions: Overall, i.t. administration of ABA (10 µg/rat) induced analgesia and p-ERK down-expression likely by involving the PKA-dependent mechanism.


Resumo Objetivo: O ácido fito-hormônio abscísico (ABA) existe como molécula sinalizadora em vários tipos de organismos, de multicelulares a células e tecidos animais. Foi demonstrado que o ABA tem efeito antinociceptivo em roedores. O presente estudo foi desenhado para avaliar o possível papel da PKA e da ERK fosforilada (p-ERK) nos efeitos antinociceptivos do ABA intratecal (i.t.) em ratos Wistar machos. Métodos: Os animais foram canulados por via i.t. e divididos em diferentes grupos experimentais (n=6‒7): controle (sem cirurgia), veículo (veículo ABA recebido), grupos tratados com ABA (recebeu ABA em doses de 10 ou 20 µg/rato), grupo tratado com ABA mais H.89 (inibidor de PKA) que recebeu o inibidor 15 minutos antes da injeção de ABA. Os testes de movimento da cauda e placa quente foram utilizados como estimuladores nociceptivos agudos para avaliar os efeitos analgésicos da ABA. A p-ERK foi avaliada na porção dorsal da medula espinhal por imunotransferência. Resultados: A microinjeção de ABA (10 e 20 µg/rato, i.t.) aumentou significativamente o limiar nociceptivo nos testes de movimento da cauda e placa quente. A aplicação de inibidor de PKA (H.89, 100 nM/rato) inibiu significativamente os efeitos analgésicos induzidos por ABA. A expressão de p-ERK diminuiu significativamente em animais injetados com ABA que não foram observados no grupo tratado com ABA+H.89. Conclusões: No geral, a administração i.t. de ABA (10 µg/rato) induziu a analgesia e expressão negativa de p-ERK provavelmente envolvendo mecanismo dependente de PKA.


Animals , Male , Plant Growth Regulators/pharmacology , Spinal Cord/metabolism , Abscisic Acid/pharmacology , Cyclic AMP-Dependent Protein Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/drug effects , Analgesics/pharmacology , Reference Values , Spinal Cord/drug effects , Time Factors , Blotting, Western , Reproducibility of Results , Rats, Wistar , Cyclic AMP-Dependent Protein Kinases/analysis , Extracellular Signal-Regulated MAP Kinases/analysis , Intracellular Signaling Peptides and Proteins/pharmacology
12.
Respir Physiol Neurobiol ; 273: 103334, 2020 02.
Article En | MEDLINE | ID: mdl-31689533

BACKGROUND: Hypoxia reportedly increases free radical generation in the body, causing oxidative stress and inhibiting ß2-AR signaling. The present study correlates the prophylactic potential of quercetin and salbutamol in ameliorating fluid clearing capacity of lungs by re-sensitizing ß2-AR signaling under hypoxia. METHODS: Male SD rats supplemented orally with quercetin (50 mg/Kg BW), and salbutamol (2 mg/Kg BW) were exposed to hypobaric hypoxia at 7620 m for 6 h. Western blotting and ELISA quantitated NFĸB and related genes and GPCR pathway proteins. The binding affinities of drugs with receptor were determined by SPR spectroscopy and further confirmed insilico. RESULTS: Quercetin and salbutamol pre-treatment significantly up-regulated the expressions of ß2-AR, GPR-1, GPR-10, GCSα, cAMP content, and down-regulated GRK-2, ß-arrestin, ROS, NFκB (p < 0.001), thus, enhancing alveolar fluid clearance (AFC). SPR and insilico findings revealed a higher binding affinity of ß2-AR with quercetin over salbutamol. CONCLUSION: Results indicated quercetin to be a better prophylactic that augmented AFC in rats exposed to hypoxia by attenuating inflammation and stimulating ß2-AR.


Adrenergic beta-2 Receptor Agonists/pharmacology , Albuterol/pharmacology , Antioxidants/pharmacology , Cyclic AMP-Dependent Protein Kinases/drug effects , Gene Expression/drug effects , Hypoxia/metabolism , Inflammation/drug therapy , Pulmonary Alveoli/drug effects , Quercetin/pharmacology , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction/drug effects , Adrenergic beta-2 Receptor Agonists/administration & dosage , Albuterol/administration & dosage , Animals , Antioxidants/administration & dosage , Body Fluids/drug effects , Disease Models, Animal , Hypoxia/drug therapy , Male , Quercetin/administration & dosage , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, beta-2/drug effects , Up-Regulation
13.
Neurosci Lett ; 714: 134612, 2020 01 01.
Article En | MEDLINE | ID: mdl-31698025

The effects of 4NO2PDPMe and 4APDPMe, which are thalidomide (Tha) analogs that act as selective phosphodiesterase (PDE-4) inhibitors, on estrous behavior (lordosis and proceptive behaviors) and on uterine contraction were studied in ovariectomized (OVX) estrogen-primed Sprague Dawley (SD) and in intact non-pregnant Wistar rats, respectively. We found that intracerebroventricular (ICV) infusion of either 4NO2PDPMe or 4APDPMe (20 to 80 µg) stimulated intense lordosis and proceptive behavior in response to mounts from a sexually active male, within the first 4 h after infusion, and persisting for up to 24 h. Inhibitors of the progesterone receptor (RU486, administered subcutaneously), the estrogen receptor (tamoxifen, ICV), the adenylate cyclase (AC)/ cyclic AMP (cAMP)/protein kinase A (PKA) pathway (administered ICV), and the mitogen activated protein kinase (MAPK) pathway (administered ICV) significantly decreased lordosis and proceptive behavior induced by Tha analogs. Uterine contractility studies showed that Tha analogs inhibited both the K+- and the Ca2+-induced tonic contractions in rat uterus. Tha analogs were equally effective, but 4APDPMe was more potent than 4NO2PDPMe. These results strongly suggest the central role of cAMP in both processes, sexual behavior, and uterine relaxation, and suggest that Tha analogs may also act as Ca2+-channel blockers.


Cyclic AMP/metabolism , Phosphodiesterase 4 Inhibitors/pharmacology , Phthalimides/pharmacology , Propionates/pharmacology , Sexual Behavior, Animal/drug effects , Thalidomide/analogs & derivatives , Uterine Contraction/drug effects , Adenylyl Cyclases/drug effects , Adenylyl Cyclases/metabolism , Animals , Calcium , Cyclic AMP-Dependent Protein Kinases/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Dideoxyadenosine/pharmacology , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Estrogens/pharmacology , Estrus , Female , In Vitro Techniques , Infusions, Intraventricular , Injections, Subcutaneous , Lordosis , Luteolytic Agents/pharmacology , Mifepristone/pharmacology , Ovariectomy , Potassium , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Progesterone , Tamoxifen/pharmacology , Thalidomide/pharmacology , Uterine Contraction/metabolism , Uterus/drug effects
14.
Aging Cell ; 19(1): e13055, 2020 01.
Article En | MEDLINE | ID: mdl-31668016

Intraneuronal accumulation of wild-type tau plays a key role in Alzheimer's disease, while the mechanisms underlying tauopathy and memory impairment remain unclear. Here, we report that overexpressing full-length wild-type human tau (hTau) in mouse hippocampus induces learning and memory deficits with remarkably reduced levels of multiple synapse- and memory-associated proteins. Overexpressing hTau inhibits the activity of protein kinase A (PKA) and decreases the phosphorylation level of cAMP-response element binding protein (CREB), GluA1, and TrkB with reduced BDNF mRNA and protein levels both in vitro and in vivo. Simultaneously, overexpressing hTau increased PKAR2α (an inhibitory subunit of PKA) in nuclear fraction and inactivated proteasome activity. With an increased association of PKAR2α with PA28γ (a nuclear proteasome activator), the formation of PA28γ-20S proteasome complex remarkably decreased in the nuclear fraction, followed by a reduced interaction of PKAR2α with 20S proteasome. Both downregulating PKAR2α by shRNA and upregulating proteasome by expressing PA28γ rescued hTau-induced PKA inhibition and CREB dephosphorylation, and upregulating PKA improved hTau-induced cognitive deficits in mice. Together, these data reveal that intracellular tau accumulation induces synapse and memory impairments by inhibiting PKA/CREB/BDNF/TrkB and PKA/GluA1 signaling, and deficit of PA28γ-20S proteasome complex formation contributes to PKAR2α elevation and PKA inhibition.


CREB-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/drug effects , Neuronal Plasticity/genetics , tau Proteins/metabolism , Humans , Phosphorylation , Signal Transduction , Synapses/metabolism
15.
Biol Psychiatry ; 86(2): 131-142, 2019 07 15.
Article En | MEDLINE | ID: mdl-31076080

BACKGROUND: The basolateral amygdala (BLA) has been widely implicated in the pathophysiology of major depressive disorder. A-kinase anchoring protein 150 (AKAP150) directs kinases and phosphatases to synaptic glutamate receptors, controlling synaptic transmission and plasticity. However, the role of the AKAP150 in the BLA in major depressive disorder remains poorly understood. METHODS: Depressive-like behaviors in C57BL/6J mice were developed by chronic restraint stress (CRS). Mice received either intra-BLA injection of lentivirus-expressing Akap5 short hairpin RNA or Ht-31, a peptide to disrupt the interaction of AKAP150 and protein kinase A (PKA), followed by depressive-like behavioral tests. Alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid glutamate receptor (AMPAR)-mediated miniature excitatory postsynaptic currents were recorded by whole-cell patch-clamp techniques. RESULTS: Chronic stress exposure induced depressive-like behaviors, which were accompanied by an increase in total and synaptic AKAP150 expression in the BLA. Accordingly, CRS facilitated the association of AKAP150 with PKA, but not of calcineurin in the BLA. Intra-BLA infusion of lentivirus-expressing Akap5 short hairpin RNA or Ht-31 prevented depressive-like behaviors and normalized phosphorylation of serine 845 and surface expression of AMPAR subunit 1 (GluA1) in the BLA of CRS mice. Finally, blockage of AKAP150-PKA complex signaling rescued the changes in AMPAR-mediated miniature excitatory postsynaptic currents in depressive-like mice. CONCLUSIONS: These results suggest that AKAP150-PKA directly modulates BLA neuronal synaptic strength, and that AKAP150-PKA-GluA1 streamline signaling complex is responsible for CRS-induced disruption of synaptic AMPAR-mediated transmission and depressive-like behaviors in mice.


A Kinase Anchor Proteins/genetics , Basolateral Nuclear Complex/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Depression/genetics , Depression/psychology , Stress, Psychological/genetics , Stress, Psychological/psychology , A Kinase Anchor Proteins/drug effects , Animals , Cyclic AMP-Dependent Protein Kinases/drug effects , Depression/etiology , Hindlimb Suspension/psychology , Mice , Mice, Inbred C57BL , Proteins/pharmacology , Receptors, AMPA/biosynthesis , Receptors, AMPA/genetics , Restraint, Physical , Stress, Psychological/complications , Swimming/psychology , Synaptic Transmission
16.
J Neurosci ; 39(8): 1484-1504, 2019 02 20.
Article En | MEDLINE | ID: mdl-30541912

Serotonin (5-hydroxytryptamine, 5-HT) receptor agonists are neuroprotective in CNS injury models. However, the neuroprotective functional implications and synaptic mechanism of 8-hydroxy-2- (di-n-propylamino) tetralin (8-OH-DPAT), a serotonin receptor (5-HT1A) agonist, in an adult male Wistar rat model of chronic glaucoma model remain unknown. We found that ocular hypertension decreased 5-HT1A receptor expression in rat retinas because the number of retinal ganglion cells (RGCs) was significantly reduced in rats with induced ocular hypertension relative to that in control retinas and 8-OH-DPAT enhanced the RGC viability. The protective effects of 8-OH-DPAT were blocked by intravitreal administration of the selective 5-HT1A antagonist WAY-100635 or the selective GABAA receptor antagonist SR95531. Using patch-clamp techniques, spontaneous and miniature GABAergic IPSCs (sIPSCs and mIPSCs, respectively) of RGCs in rat retinal slices were recorded. 8-OH-DPAT significantly increased the frequency and amplitude of GABAergic sIPSCs and mIPSCs in ON- and OFF-type RGCs. Among the signaling cascades mediated by the 5-HT1A receptor, the role of cAMP-protein kinase A (PKA) signaling was investigated. The 8-OH-DPAT-induced changes at the synaptic level were enhanced by PKA inhibition by H-89 and blocked by PKA activation with bucladesine. Furthermore, the density of phosphorylated PKA (p-PKA)/PKA was significantly increased in glaucomatous retinas and 8-OH-DPAT significantly decreased p-PKA/PKA expression, which led to the inhibition of PKA phosphorylation upon relieving neurotransmitter GABA release. These results showed that the activation of 5-HT1A receptors in retinas facilitated presynaptic GABA release functions by suppressing cAMP-PKA signaling and decreasing PKA phosphorylation, which could lead to the de-excitation of RGC circuits and suppress excitotoxic processes in glaucoma.SIGNIFICANCE STATEMENT We found that serotonin (5-HT) receptors in the retina (5-HT1A receptors) were downregulated after intraocular pressure elevation. Patch-clamp recordings demonstrated differences in the frequencies of miniature GABAergic IPSCs (mIPSCs) in ON- and OFF-type retinal ganglion cells (RGCs) and RGCs in normal and glaucomatous retinal slices. Therefore, phosphorylated protein kinase A (PKA) inhibition upon release of the neurotransmitter GABA was eliminated by 8-hydroxy-2- (di-n-propylamino) tetralin (8-OH-DPAT), which led to increased levels of GABAergic mIPSCs in ON- and OFF-type RGCs, thus enhancing RGC viability and function. These protective effects were blocked by the GABAA receptor antagonist SR95531 or the 5-HT1A antagonist WAY-100635. This study identified a novel mechanism by which activation of 5-HT1A receptors protects damaged RGCs via the cAMP-PKA signaling pathway that modulates GABAergic presynaptic activity.


Cyclic AMP-Dependent Protein Kinases/physiology , Cyclic AMP/physiology , Eye Proteins/metabolism , Glaucoma/metabolism , Receptor, Serotonin, 5-HT1A/physiology , Retinal Ganglion Cells/metabolism , gamma-Aminobutyric Acid/metabolism , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , Bucladesine/pharmacology , Cyclic AMP-Dependent Protein Kinases/drug effects , Isoquinolines/pharmacology , Male , Ocular Hypertension/metabolism , Phosphorylation/drug effects , Piperazines/pharmacology , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Protein Processing, Post-Translational/drug effects , Pyridines/pharmacology , Rats , Rats, Wistar , Second Messenger Systems/drug effects , Second Messenger Systems/physiology , Serotonin 5-HT1 Receptor Agonists/pharmacology , Serotonin 5-HT1 Receptor Antagonists/pharmacology , Sulfonamides/pharmacology
17.
Pflugers Arch ; 471(2): 365-380, 2019 02.
Article En | MEDLINE | ID: mdl-30291431

Carbocisteine (CCis), a mucoactive agent, is widely used to improve respiratory diseases. This study demonstrated that CCis increases ciliary bend angle (CBA) by 30% and ciliary beat frequency (CBF) by 10% in mouse airway ciliary cells. These increases were induced by an elevation in intracellular pH (pHi; the pHi pathway) and a decrease in the intracellular Cl- concentration ([Cl-]i; the Cl- pathway) stimulated by CCis. The Cl- pathway, which is independent of CO2/HCO3-, increased CBA by 20%. This pathway activated Cl- release via activation of Cl- channels, leading to a decrease in [Cl-]i, and was inhibited by Cl- channel blockers (5-nitro-2-(3-phenylpropylamino) benzoic acid and CFTR(inh)-172). Under the CO2/HCO3--free condition, the CBA increase stimulated by CCis was mimicked by the Cl--free NO3- solution. The pHi pathway, which depends on CO2/HCO3-, increased CBF and CBA by 10%. This pathway activated HCO3- entry via Na+/HCO3- cotransport (NBC), leading to a pHi elevation, and was inhibited by 4,4'-diisothiocyano-2,2'-stilbenedisulfonic acid. The effects of CCis were not affected by a protein kinase A inhibitor (1 µM PKI-A) or Ca2+-free solution. Thus, CCis decreased [Cl-]i via activation of Cl- channels including CFTR, increasing CBA by 20%, and elevated pHi via NBC activation, increasing CBF and CBA by 10%.


Chlorides/metabolism , Cilia/metabolism , Respiratory System/metabolism , Animals , Bicarbonates/metabolism , Calcium/metabolism , Cilia/drug effects , Cyclic AMP-Dependent Protein Kinases/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Hydrogen-Ion Concentration , Mice , Protein Kinase Inhibitors/pharmacology , Sodium/metabolism
18.
J Appl Oral Sci ; 26: e20170231, 2018 May 14.
Article En | MEDLINE | ID: mdl-29768523

We previously reported that elevated extracellular calcium (Ca2+) levels increase bone morphogenetic protein 2 expression in human dental pulp (hDP) cells. However, it is unknown whether extracellular Ca2+ affects the expression of other growth factors such as fibroblast growth factor 2 (FGF2). The present study aimed to examine the effect of extracellular Ca2+ on FGF2 gene expression in hDP and immortalized mouse dental papilla (mDP) cells. Cells were stimulated with 10 mM CaCl2 in the presence or absence of cell signaling inhibitors. FGF2 gene expression was assessed using real-time polymerase chain reaction. The phosphorylation status of signaling molecules was examined by Western blotting. Extracellular Ca2+ increased FGF2 gene expression in mDP and hDP cells. Gene expression of the calcium-sensing receptor and G protein-coupled receptor family C group 6 member A, both of which are extracellular Ca2+ sensors, was not detected. Ca2+-mediated Fgf2 expression was reduced by pretreatment with the protein kinase A (PKA) inhibitor H-89 or extracellular signal-regulated kinase (ERK) 1/2 inhibitor PD98059 but not by pretreatment with the protein kinase C inhibitor GF-109203X or p38 inhibitor SB203580. Extracellular Ca2+ increased PKA activity and ERK1/2 phosphorylation. Ca2+-induced PKA activity decreased by pretreatment with PD98059. These findings indicate that elevated extracellular Ca2+ levels led to increased Fgf2 expression through ERK1/2 and PKA in mDP cells and that this mechanism may be useful for designing regenerative therapies for dentin.


Calcium/pharmacology , Cyclic AMP-Dependent Protein Kinases/drug effects , Dental Papilla/drug effects , Fibroblast Growth Factor 2/drug effects , Gene Expression/drug effects , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 3/drug effects , Animals , Blotting, Western , Calcium Chloride/pharmacology , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/analysis , Enzyme-Linked Immunosorbent Assay , Fibroblast Growth Factor 2/analysis , Fibroblast Growth Factor 2/genetics , MAP Kinase Signaling System/drug effects , Mice , Mitogen-Activated Protein Kinase 1/analysis , Mitogen-Activated Protein Kinase 3/analysis , Real-Time Polymerase Chain Reaction , Reproducibility of Results , Time Factors
19.
J. appl. oral sci ; 26: e20170231, 2018. graf
Article En | LILACS, BBO | ID: biblio-893679

Abstract We previously reported that elevated extracellular calcium (Ca2+) levels increase bone morphogenetic protein 2 expression in human dental pulp (hDP) cells. However, it is unknown whether extracellular Ca2+ affects the expression of other growth factors such as fibroblast growth factor 2 (FGF2). Objective: The present study aimed to examine the effect of extracellular Ca2+ on FGF2 gene expression in hDP and immortalized mouse dental papilla (mDP) cells. Materials and Methods: Cells were stimulated with 10 mM CaCl2 in the presence or absence of cell signaling inhibitors. FGF2 gene expression was assessed using real-time polymerase chain reaction. The phosphorylation status of signaling molecules was examined by Western blotting. Results: Extracellular Ca2+ increased FGF2 gene expression in mDP and hDP cells. Gene expression of the calcium-sensing receptor and G protein-coupled receptor family C group 6 member A, both of which are extracellular Ca2+ sensors, was not detected. Ca2+-mediated Fgf2 expression was reduced by pretreatment with the protein kinase A (PKA) inhibitor H-89 or extracellular signal-regulated kinase (ERK) 1/2 inhibitor PD98059 but not by pretreatment with the protein kinase C inhibitor GF-109203X or p38 inhibitor SB203580. Extracellular Ca2+ increased PKA activity and ERK1/2 phosphorylation. Ca2+-induced PKA activity decreased by pretreatment with PD98059. Conclusions: These findings indicate that elevated extracellular Ca2+ levels led to increased Fgf2 expression through ERK1/2 and PKA in mDP cells and that this mechanism may be useful for designing regenerative therapies for dentin.


Animals , Mice , Gene Expression/drug effects , Calcium/pharmacology , Fibroblast Growth Factor 2/drug effects , Cyclic AMP-Dependent Protein Kinases/drug effects , Mitogen-Activated Protein Kinase 1/drug effects , Dental Papilla/drug effects , Mitogen-Activated Protein Kinase 3/drug effects , Time Factors , Calcium Chloride/pharmacology , Enzyme-Linked Immunosorbent Assay , Cells, Cultured , Blotting, Western , Reproducibility of Results , Fibroblast Growth Factor 2/analysis , Fibroblast Growth Factor 2/genetics , Cyclic AMP-Dependent Protein Kinases/analysis , Mitogen-Activated Protein Kinase 1/analysis , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 3/analysis , Real-Time Polymerase Chain Reaction
20.
Med Sci Monit ; 23: 5462-5471, 2017 Nov 17.
Article En | MEDLINE | ID: mdl-29146892

BACKGROUND The aim of this study was to explore the role of intermedin and its mechanism in cholesterol efflux of macrophage THP-1 and RAW264.7 cell lines in atherosclerosis (AS). MATERIAL AND METHODS ApoE-/- mice were fed with a high-fat diet, and the concentrations of total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C), and high-density lipoprotein cholesterol (HDL-C) were measured. The lipidoses of the aortic sinus were analyzed by hematoxylin and eosin staining, and the cAMP level was detected by enzyme-linked immunosorbent assay (ELISA). The expressions of ATP-binding cassette transporter (ABCA1) were tested by real-time PCR and Western blot analysis. RESULTS IMD decreased serum TC and LDL-C, and increased serum HDL-C level in apoE-/- mice and attenuated AS plaque areas. In vitro, IMD increased intracellular cAMP concentration in a dose-dependent manner in THP-1 and RAW264.7 cell lines, which enhanced the expression of ABCA1 and increased cholesterol efflux rate. However, this effect was inhibited by PKAI in the RAW 264.7 cell line but not in the THP-1 cell line. CONCLUSIONS IMD can ameliorate the development of AS in ApoE-/- mice and regulate cholesterol balance in the RAW264.7 cell line through the cAMP-PKA pathway.


Atherosclerosis/drug therapy , Neuropeptides/pharmacology , Pyrrolizidine Alkaloids/metabolism , Pyrrolizidine Alkaloids/pharmacokinetics , ATP Binding Cassette Transporter 1/metabolism , ATP-Binding Cassette Transporters/metabolism , Animals , Apolipoproteins E/metabolism , Atherosclerosis/metabolism , Biological Transport , Cholesterol/metabolism , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/metabolism , Cyclic AMP-Dependent Protein Kinases/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Diet, High-Fat , Humans , Macrophages/metabolism , Mice , Mice, Knockout , Neuropeptides/metabolism , Plaque, Atherosclerotic/drug therapy , RAW 264.7 Cells/drug effects , RAW 264.7 Cells/physiology , THP-1 Cells/drug effects , THP-1 Cells/physiology , Triglycerides/blood
...