Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(24)2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38139359

ABSTRACT

The serine-threonine kinase Akt plays a fundamental role in cell survival, metabolism, proliferation, and migration. To keep these essential processes under control, Akt activity and stability must be tightly regulated; otherwise, life-threatening conditions might prevail. Although it is well understood that phosphorylation regulates Akt activity, much remains to be known about how its stability is maintained. Here, we characterize BAG5, a chaperone regulator, as a novel Akt-interactor and substrate that attenuates Akt stability together with Hsp70. BAG5 switches monoubiquitination to polyubiquitination of Akt and increases its degradation caused by Hsp90 inhibition and Hsp70 overexpression. Akt interacts with BAG5 at the linker region that joins the first and second BAG domains and phosphorylates the first BAG domain. The Akt-BAG5 complex is formed in serum-starved conditions and dissociates in response to HGF, coincident with BAG5 phosphorylation. BAG5 knockdown attenuated Akt degradation and facilitated its activation, whereas the opposite effect was caused by BAG5 overexpression. Altogether, our results indicate that Akt stability and signaling are dynamically regulated by BAG5, depending on growth factor availability.


Subject(s)
Molecular Chaperones , Proto-Oncogene Proteins c-akt , HSP70 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Ubiquitination , HEK293 Cells , Humans , Animals , Mice
2.
Mol Biol Rep ; 50(9): 7797-7814, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37486442

ABSTRACT

Anxiety and depression disorders are highly prevalent neurological disorders (NDs) that impact up to one in three individuals during their lifetime. Addressing these disorders requires reducing their frequency and impact, understanding molecular causes, implementing prevention strategies, and improving treatments. Cyclic nucleotide monophosphates (cNMPs) like cyclic adenosine monophosphate (cAMP), cyclic guanosine monophosphate (cGMP), cyclic uridine monophosphate (cUMP), and cyclic cytidine monophosphate (cCMP) regulate the transcription of genes involved in neurotransmitters and neurological functions. Evidence suggests that cNMP pathways, including cAMP/cGMP, cAMP response element binding protein (CREB), and Protein kinase A (PKA), play a role in the physiopathology of anxiety and depression disorders. Plant and mushroom-based compounds have been used in traditional and modern medicine due to their beneficial properties. Bioactive compound metabolism can activate key pathways and yield pharmacological outcomes. This review focuses on the molecular mechanisms of bioactive compounds from plants and mushrooms in modulating cNMP pathways. Understanding these processes will support current treatments and aid in the development of novel approaches to reduce the prevalence of anxiety and depression disorders, contributing to improved outcomes and the prevention of associated complications.


Subject(s)
Depression , Nucleotides, Cyclic , Humans , Nucleotides, Cyclic/metabolism , Nucleotides, Cyclic/pharmacology , Depression/drug therapy , Cyclic GMP/metabolism , Cyclic AMP/metabolism , Plants/metabolism , Anxiety/drug therapy
3.
Int J Mol Sci ; 23(11)2022 May 27.
Article in English | MEDLINE | ID: mdl-35682723

ABSTRACT

Angiotensin II (Ang II) is a critical regulator of insulin signaling in the cardiovascular system and metabolic tissues. However, in adipose cells, the regulatory role of Ang II on insulin actions remains to be elucidated. The effect of Ang II on insulin-induced insulin receptor (IR) phosphorylation, Akt activation, and glucose uptake was examined in 3T3-L1 adipocytes. In these cells, Ang II specifically inhibited insulin-stimulated IR and insulin receptor substrate-1 (IRS-1) tyrosine-phosphorylation, Akt activation, and glucose uptake in a time-dependent manner. These inhibitory actions were associated with increased phosphorylation of the IR at serine residues. Interestingly, Ang II-induced serine-phosphorylation of IRS was not detected, suggesting that Ang II-induced desensitization begins from IR regulation itself. PKC inhibition by BIM I restored the inhibitory effect of Ang II on insulin actions. We also found that Ang II promoted activation of several PKC isoforms, including PKCα/ßI/ßII/δ, and its association with the IR, particularly PKCßII, showed the highest interaction. Finally, we also found a similar regulatory effect of Ang II in isolated adipocytes, where insulin-induced Akt phosphorylation was inhibited by Ang II, an effect that was prevented by PKC inhibitors. These results suggest that Ang II may lead to insulin resistance through PKC activation in adipocytes.


Subject(s)
Angiotensin II , Receptor, Insulin , Adipocytes/metabolism , Angiotensin II/metabolism , Angiotensin II/pharmacology , Glucose/metabolism , Insulin/metabolism , Insulin Receptor Substrate Proteins/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Insulin/metabolism , Serine/metabolism
4.
Metabolites ; 11(2)2021 Feb 05.
Article in English | MEDLINE | ID: mdl-33562475

ABSTRACT

Experimental evidence in mice models has demonstrated that a high regulator of G-protein signaling 2 (RSG2) protein levels precede an insulin resistance state. In the same context, a diet rich in saturated fatty acids induces an increase in RGS2 protein expression, which has been associated with decreased basal metabolism in mice; however, the above has not yet been analyzed in humans. For this reason, in the present study, we examined the association between RGS2 expression and insulin resistance state. The incubation with palmitic acid (PA), which inhibits insulin-mediated Akt Ser473 phosphorylation, resulted in the increased RGS2 expression in human umbilical vein endothelial-CS (HUVEC-CS) cells. The RGS2 overexpression without PA was enough to inhibit insulin-mediated Akt Ser473 phosphorylation in HUVEC-CS cells. Remarkably, the platelet RGS2 expression levels were higher in type 2 diabetes mellitus (T2DM) patients than in healthy donors. Moreover, an unbiased principal component analysis (PCA) revealed that RGS2 expression level positively correlated with glycated hemoglobin (HbA1c) and negatively with age and high-density lipoprotein cholesterol (HDL) in T2DM patients. Furthermore, PCA showed that healthy subjects segregated from T2DM patients by having lower levels of HbA1c and RGS2. These results demonstrate that RGS2 overexpression leads to decreased insulin signaling in a human endothelial cell line and is associated with poorly controlled diabetes.

5.
Article in English | MEDLINE | ID: mdl-31920979

ABSTRACT

In the present study, we determined the cellular regulators of ERK1/2 and Akt signaling pathways in response to human CRF1 receptor (CRF1R) activation in transfected COS-7 cells. We found that Pertussis Toxin (PTX) treatment or sequestering Gßγ reduced CRF1R-mediated activation of ERK1/2, suggesting the involvement of a Gi-linked cascade. Neither Gs/PKA nor Gq/PKC were associated with ERK1/2 activation. Besides, CRF induced EGF receptor (EGFR) phosphorylation at Tyr1068, and selective inhibition of EGFR kinase activity by AG1478 strongly inhibited the CRF1R-mediated phosphorylation of ERK1/2, indicating the participation of EGFR transactivation. Furthermore, CRF-induced ERK1/2 phosphorylation was not altered by pretreatment with batimastat, GM6001, or an HB-EGF antibody indicating that metalloproteinase processing of HB-EGF ligands is not required for the CRF-mediated EGFR transactivation. We also observed that CRF induced Src and PYK2 phosphorylation in a Gßγ-dependent manner. Additionally, using the specific Src kinase inhibitor PP2 and the dominant-negative-SrcYF-KM, it was revealed that CRF-stimulated ERK1/2 phosphorylation depends on Src activation. PP2 also blocked the effect of CRF on Src and EGFR (Tyr845) phosphorylation, further demonstrating the centrality of Src. We identified the formation of a protein complex consisting of CRF1R, Src, and EGFR facilitates EGFR transactivation and CRF1R-mediated signaling. CRF stimulated Akt phosphorylation, which was dependent on Gi/ßγ subunits, and Src activation, however, was only slightly dependent on EGFR transactivation. Moreover, PI3K inhibitors were able to inhibit not only the CRF-induced phosphorylation of Akt, as expected, but also ERK1/2 activation by CRF suggesting a PI3K dependency in the CRF1R ERK signaling. Finally, CRF-stimulated ERK1/2 activation was similar in the wild-type CRF1R and the phosphorylation-deficient CRF1R-Δ386 mutant, which has impaired agonist-dependent ß-arrestin-2 recruitment; however, this situation may have resulted from the low ß-arrestin expression in the COS-7 cells. When ß-arrestin-2 was overexpressed in COS-7 cells, CRF-stimulated ERK1/2 phosphorylation was markedly upregulated. These findings indicate that on the base of a constitutive CRF1R/EGFR interaction, the Gi/ßγ subunits upstream activation of Src, PYK2, PI3K, and transactivation of the EGFR are required for CRF1R signaling via the ERK1/2-MAP kinase pathway. In contrast, Akt activation via CRF1R is mediated by the Src/PI3K pathway with little contribution of EGFR transactivation.

6.
Cell Signal ; 28(1): 53-9, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26475209

ABSTRACT

Palmitic acid is a negative regulator of insulin activity. At the molecular level, palmitic acid reduces insulin stimulated Akt Ser473 phosphorylation. Interestingly, we have found that incubation with palmitic acid of human umbilical vein endothelial cells induced a biphasic effect, an initial transient elevation followed by a sustained reduction of SERCA pump protein levels. However, palmitic acid produced a sustained inhibition of SERCA pump ATPase activity. Insulin resistance state appeared before there was a significant reduction of SERCA2 expression. The mechanism by which palmitic acid impairs insulin signaling may involve endoplasmic reticulum stress, because this fatty acid induced activation of both PERK, an ER stress marker, and JNK, a kinase associated with insulin resistance. None of these effects were observed by incubating HUVEC-CS cells with palmitoleic acid. Importantly, SERCA2 overexpression decreased the palmitic acid-induced insulin resistance state. All these results suggest that SERCA pump might be the target of palmitic acid to induce the insulin resistance state in a human vascular endothelial cell line. Importantly, these data suggest that HUVEC-CS cells respond to palmitic acid-exposure with a compensatory overexpression of SERCA pump within the first hour, which eventually fades out and insulin resistance prevails.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Endothelial Cells/drug effects , Fatty Acids, Monounsaturated/pharmacology , Insulin Resistance/physiology , Palmitic Acid/pharmacology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Apoptosis/drug effects , Cell Line , Endoplasmic Reticulum/metabolism , Endothelial Cells/metabolism , Humans
7.
Regul Pept ; 186: 62-76, 2013 Sep 10.
Article in English | MEDLINE | ID: mdl-23820308

ABSTRACT

The primary goal was to determine agonist-specific regulation of CRF2(a) receptor function. Exposure of human retinoblastoma Y79 cells to selective (UCN2, UCN3 or stresscopins) and non-selective (UCN1 or sauvagine) agonists prominently desensitized CRF2(a) receptors in a rapid, concentration-dependent manner. A considerably slower rate and smaller magnitude of desensitization developed in response to the weak agonist CRF. CRF1 receptor desensitization stimulated by CRF, cortagine or stressin1-A had no effect on CRF2(a) receptor cyclic AMP signaling. Conversely, desensitization of CRF2(a) receptors by UCN2 or UCN3 did not cross-desensitize Gs-coupled CRF1 receptor signaling. In transfected HEK293 cells, activation of CRF2(a) receptors by UCN2, UCN3 or CRF resulted in receptor phosphorylation and internalization proportional to agonist potency. Neither protein kinase A nor casein kinases mediated CRF2(a) receptor phosphorylation or desensitization. Exposure of HEK293 or U2OS cells to UCN2 or UCN3 (100nM) produced strong ßarrestin2 translocation and colocalization with membrane CRF2(a) receptors while CRF (1µM) generated only weak ßarrestin2 recruitment. ßarrestin2 did not internalize with the receptor, however, indicating that transient CRF2(a) receptor-arrestin complexes dissociate at or near the cell membrane. Since deletion of the ßarrestin2 gene upregulated Gs-coupled CRF2(a) receptor signaling in MEF cells, a ßarrestin2 mechanism restrains Gs-coupled CRF2(a) receptor signaling activated by urocortins. We further conclude that the rate and extent of homologous CRF2(a) receptor desensitization are governed by agonist-specific mechanisms affecting GRK phosphorylation, ßarrestin2 recruitment, and internalization thereby producing unique signal transduction profiles that differentially affect the stress response.


Subject(s)
Arrestins/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Second Messenger Systems , Amphibian Proteins/pharmacology , Amphibian Proteins/physiology , Cell Line, Tumor , Colforsin/pharmacology , Corticotropin-Releasing Hormone/pharmacology , Corticotropin-Releasing Hormone/physiology , Cyclic AMP/metabolism , HEK293 Cells , Humans , Peptide Hormones/pharmacology , Peptide Hormones/physiology , Phosphorylation , Protein Processing, Post-Translational , Protein Transport , Receptors, Corticotropin-Releasing Hormone/agonists , Urocortins/pharmacology , Urocortins/physiology , beta-Arrestins
8.
Neuropharmacology ; 62(2): 705-14, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22122881

ABSTRACT

The reasons for differences in vulnerability or resilience to the development of posttraumatic stress disorder (PTSD) are unclear. Here we review key genetic diatheses and molecular targets especially signaling pathways that mediate responses to trauma and severe stress and their potential contribution to the etiology of PTSD. Sensitization of glucocorticoid receptor (GR) signaling and dysregulation of GR modulators FKBP5, STAT5B, Bcl-2, and Bax have been implicated in PTSD pathophysiology. Furthermore, Akt, NFκB, MKP-1, and p11, which are G protein-coupled receptor (GPCR) pathway molecules, can promote or prevent sustained high anxiety- and depressive-like behavior following severe stress. Agonist-induced activation of the corticotropin releasing factor CRF(1) receptor is crucial for survival in the context of serious danger or trauma, but persistent CRF(1) receptor hypersignaling when a threatening or traumatic situation is no longer present is maladaptive. CRF(1) receptor single nucleotide polymorphisms (SNPs) can confer susceptibility or resilience to childhood trauma while a SNP for the PAC1 receptor, another class B1 GPCR, has been linked genetically to PTSD. GRK3 phosphorylation of the CRF(1) receptor protein and subsequent binding of ßarrestin2 rapidly terminate Gs-coupled CRF(1) receptor signaling by homologous desensitization. A deficient GRK-ßarrestin2 mechanism would result in excessive CRF(1) receptor signaling thereby contributing to PTSD and co-morbid posttraumatic depression. Clinical trials are needed to assess if small molecule CRF(1) receptor antagonists are effective prophylactic agents when administered immediately after trauma. ßarrestin2-biased agonists for CRF receptors and possibly other GPCRs implicated in PTSD, however, may prove to be novel pharmacotherapy with greater selectivity and therapeutic efficacy. This article is part of a Special Issue entitled 'Post-Traumatic Stress Disorder'.


Subject(s)
Receptors, Corticotropin-Releasing Hormone/metabolism , Receptors, Glucocorticoid/metabolism , Signal Transduction/physiology , Stress Disorders, Post-Traumatic/metabolism , Humans , Phosphorylation , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Glucocorticoid/antagonists & inhibitors , Signal Transduction/drug effects , Stress Disorders, Post-Traumatic/drug therapy
9.
Biochem Pharmacol ; 79(5): 733-45, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-19879250

ABSTRACT

To investigate the potential interactions between the angiotensin II (Ang II) and insulin signaling systems, regulation of IRS-1 phosphorylation and insulin-induced Akt activation by Ang II were examined in clone 9 (C9) hepatocytes. In these cells, Ang II specifically inhibited activation of insulin-induced Akt Thr(308) and its immediate downstream substrate GSK-3alpha/beta in a time-dependent fashion, with approximately 70% reduction at 15 min. These inhibitory actions were associated with increased IRS-1 phosphorylation of Ser(636)/Ser(639) that was prevented by selective blockade of EGFR tyrosine kinase activity with AG1478. Previous studies have shown that insulin-induced phosphorylation of IRS-1 on Ser(636)/Ser(639) is mediated mainly by the PI3K/mTOR/S6K-1 sequence. Studies with specific inhibitors of PI3K (wortmannin) and mTOR (rapamycin) revealed that Ang II stimulates IRS-1 phosphorylation of Ser(636)/Ser(639) via the PI3K/mTOR/S6K-1 pathway. Both inhibitors blocked the effect of Ang II on insulin-induced activation of Akt. Studies using the specific MEK inhibitor, PD98059, revealed that ERK1/2 activation also mediates Ang II-induced S6K-1 and IRS-1 phosphorylation, and the impairment of Akt Thr(308) and GSK-3alpha/beta phosphorylation. Further studies with selective inhibitors showed that PI3K activation was upstream of ERK, suggesting a new mechanism for Ang II-induced impairment of insulin signaling. These findings indicate that Ang II has a significant role in the development of insulin resistance by a mechanism that involves EGFR transactivation and the PI3K/ERK1/2/mTOR-S6K-1 pathway.


Subject(s)
Angiotensin II/pharmacology , ErbB Receptors/genetics , Insulin/metabolism , Signal Transduction/drug effects , Vasoconstrictor Agents/pharmacology , Animals , Cell Line , Enzyme Inhibitors/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Insulin Resistance/physiology , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines , Rats , Receptor, Insulin/metabolism , Serine/genetics , Serine/metabolism , Threonine/genetics , Threonine/metabolism , Transcriptional Activation , Tyrphostins/pharmacology
10.
Ann N Y Acad Sci ; 1179: 120-43, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19906236

ABSTRACT

Markers of hyperactive central corticotropin releasing factor (CRF) systems and CRF-related single nucleotide polymorphisms (SNPs) have been identified in patients with anxiety and depressive disorders. Designing more effective antagonists may now be guided by data showing that small molecules bind to transmembrane domains. Specifically, CRF(1) receptor antagonists have been developed as novel anxiolytic and antidepressant treatments. Because CRF(1) receptors become rapidly desensitized by G protein-coupled receptor kinase (GRK) and beta-arrestin mechanisms in the presence of high agonist concentrations, neuronal hypersecretion of synaptic CRF alone may be insufficient to account for excessive central CRF neurotransmission in stress-induced affective pathophysiology. In addition to desensitizing receptor function, GRK phosphorylation and beta-arrestin binding can shift a G protein-coupled receptor (GPCR) to signal selectively via the extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK-MAPK) or Akt pathways independent of G proteins. Also, Epac-dependent CRF(1) receptor signaling via the ERK-MAPK pathway has been found to potentiate brain-derived neurotrophic factor (BDNF)-stimulated TrkB signaling. Thus, genetic or acquired abnormalities in GRK and beta-arrestin function may be involved in the pathophysiology of stress-induced anxiety and depression.


Subject(s)
Anxiety/physiopathology , Depression/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Signal Transduction/physiology , Stress, Psychological/metabolism , Anxiety Disorders/drug therapy , Anxiety Disorders/metabolism , Depression/drug therapy , Humans , Hypothalamo-Hypophyseal System/metabolism , Models, Biological , Pituitary-Adrenal System/metabolism , Receptors, Corticotropin-Releasing Hormone/agonists , Signal Transduction/drug effects , Stress, Psychological/drug therapy
11.
Mol Cell Endocrinol ; 302(2): 128-39, 2009 Apr 29.
Article in English | MEDLINE | ID: mdl-19150387

ABSTRACT

Angiotensin II (Ang II), the major effector hormone of the renin-angiotensin system (RAS), has an important role in the regulation of vascular and renal homeostasis. Clinical and pharmacological studies have recently shown that Ang II is a critical promoter of insulin resistance and diabetes mellitus type 2. Ang II exerts its actions on insulin-sensitive tissues such as liver, muscle and adipose tissue where it has effects on the insulin receptor (IR), insulin receptor substrate (IRS) proteins and the downstream effectors PI3K, Akt and GLUT4. The molecular mechanisms involved have not been completely identified, but the role of serine/threonine phosphorylation of the IR and IRS-1 proteins in desensitization of insulin action has been well established. The purpose of this review is to highlight recent advances in the understanding of Ang II actions which lead to the development of insulin resistance and its implications for diabetes.


Subject(s)
Angiotensin II/physiology , Insulin Resistance , Diabetes Mellitus/etiology , Humans , Renin-Angiotensin System
12.
Am J Physiol Regul Integr Comp Physiol ; 293(1): R209-22, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17363685

ABSTRACT

The primary goal was to test the hypothesis that agonist-induced corticotropin-releasing factor type 1 (CRF(1)) receptor phosphorylation is required for beta-arrestins to translocate from cytosol to the cell membrane. We also sought to determine the relative importance to beta-arrestin recruitment of motifs in the CRF(1) receptor carboxyl terminus and third intracellular loop. beta-Arrestin-2 translocated significantly more rapidly than beta-arrestin-1 to agonist-activated membrane CRF(1) receptors in multiple cell lines. Although CRF(1) receptors internalized with agonist treatment, neither arrestin isoform trafficked with the receptor inside the cell, indicating that CRF(1) receptor-arrestin complexes dissociate at or near the cell membrane. Both arrestin and clathrin-dependent mechanisms were involved in CRF(1) receptor internalization. To investigate molecular determinants mediating the robust beta-arrestin-2-CRF(1) receptor interaction, mutagenesis was performed to remove potential G protein-coupled receptor kinase phosphorylation sites. Truncating the CRF(1) receptor carboxyl terminus at serine-386 greatly reduced agonist-dependent phosphorylation but only partially impaired beta-arrestin-2 recruitment. Removal of a serine/threonine cluster in the third intracellular loop also significantly reduced CRF(1) receptor phosphorylation but did not alter beta-arrestin-2 recruitment. Phosphorylation was abolished in a CRF(1) receptor possessing both mutations. Surprisingly, this mutant still recruited beta-arrestin-2. These mutations did not alter membrane expression or cAMP signaling of CRF(1) receptors. Our data reveal the involvement of at least the following two distinct receptor regions in beta-arrestin-2 recruitment: 1) a carboxyl-terminal motif in which serine/threonine residues must be phosphorylated and 2) an intracellular loop motif configured by agonist-induced changes in CRF(1) receptor conformation. Deficient beta-arrestin-2-CRF(1) receptor interactions could contribute to the pathophysiology of affective disorders by inducing excessive CRF(1) receptor signaling.


Subject(s)
Anxiety/physiopathology , Arrestins/physiology , Receptors, Corticotropin-Releasing Hormone/metabolism , Stress, Psychological/physiopathology , Amino Acid Sequence , Cell Line , Cells, Cultured , Cyclic AMP/physiology , Epitopes/genetics , Humans , Microscopy, Confocal , Molecular Sequence Data , Mutation/physiology , Phosphorylation , Protein Transport , Receptors, Corticotropin-Releasing Hormone/agonists , Receptors, G-Protein-Coupled/physiology , Transfection , beta-Arrestin 1 , beta-Arrestin 2 , beta-Arrestins
13.
Mol Pharmacol ; 68(2): 356-64, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15905421

ABSTRACT

In rat hepatic C9 cells, angiotensin II (Ang II)-induced activation of angiotensin type 1 (AT(1)) receptors (AT(1)-Rs) stimulates extracellular signal-regulated kinase (ERK) 1/2 phosphorylation via transactivation of the endogenous epidermal growth factor (EGF) receptor (EGF-R) by a protein kinase C (PKC) delta/Src/Pyk2-dependent pathway. This leads to phosphorylation of the EGF-R as well as its subsequent internalization. On the other hand, EGF-induced activation of the EGF-R in C9 cells was found to cause phosphorylation of the AT(1)-R. This was prevented by selective inhibition of the intrinsic tyrosine kinase activity of the EGF-R by AG1478 [4-(3'-chloroanilino)-6,7-dimethoxy-quinazoline] and was reduced by inhibition of PKC and phosphoinositide 3-kinase. EGF-induced AT(1)-R phosphorylation was associated with a decrease in membrane-associated AT(1)-Rs and a reduced inositol phosphate response to Ang II. Agonist activation of endogenous AT(1)-Rs and EGF-Rs induced the formation of a multireceptor complex containing both the AT(1)-R and the transactivated EGF-R. The dependence of these responses on caveolin was indicated by the finding that cholesterol depletion of C9 cells abolished Ang II-induced inositol phosphate production, activation of Akt/PKB and ERK1/2, and AT(1)-R internalization. Confocal microscopy demonstrated that caveolin-1 was endogenously phosphorylated and was distributed on the plasma membrane in patches that undergo redistribution during Ang II stimulation. Agonist-induced phosphorylation and association of caveolin 1 with the AT(1)-R was observed, consistent with a scaffolding role of caveolin during transactivation of the EGF-R by Ang II. The EGF-induced AT(1)-R/caveolin association was abolished by AG1478, suggesting that activation of the EGF-R promotes the association of caveolin and the AT(1)-R.


Subject(s)
ErbB Receptors/agonists , ErbB Receptors/metabolism , Receptor, Angiotensin, Type 1/agonists , Receptor, Angiotensin, Type 1/metabolism , Angiotensin II/metabolism , Angiotensin II/pharmacology , Animals , Cell Line , Dose-Response Relationship, Drug , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , Humans , Rats , Signal Transduction/drug effects , Signal Transduction/physiology
14.
Mol Pharmacol ; 67(1): 184-94, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15465928

ABSTRACT

Protein kinase C (PKC) isoforms are important transducers of signals from G protein-coupled receptors (GPCRs) to diverse cellular targets, including extracellular signal-regulated kinases 1 and 2 (ERK1/2). Clone 9 rat hepatocytes (C9 cells) express receptors for angiotensin II (Ang II) type 1, lysophosphatidic acid (LPA), and epidermal growth factor (EGF), and their stimulation causes transient ERK1/2 phosphorylation through transactivation of the epidermal growth factor receptor (EGF-R). Inhibition of PKC by Go6983 [2-[1-(3-dimethylaminopropyl)-5-methoxyindol-3-yl]-3-(1H-indol-3-yl)maleimide], or PKC depletion by prolonged phorbol 12-myristate 13-acetate (PMA) treatment, attenuated ERK1/2 activation by Ang II and PMA, but not by LPA and EGF. In contrast, another PKC inhibitor, Go6976 [12-(2-cyanoethyl)-6,7,12,13-tetrahydro-13-methyl-5-oxo-5H-indolo(2,3-a)pyrrolo(3,4-c)-carbazole], enhanced basal and agonist-stimulated phosphorylation of ERK1/2, which was not caused by alteration in receptor binding and internalization, stimulation of inositol phosphate production, or activation of Pyk2 and Src tyrosine kinases. However, Go6976 enhanced agonist-induced tyrosine phosphorylation of the EGF receptor, possibly through inhibition of protein tyrosine phosphatase (PTP), because the PTP inhibitor sodium orthovanadate mimicked the effects of Go6976. Selective blockade of EGF-R kinase by AG1478 [4-(3-chloroanilino)6,7-dimethoxyquinazoline] abolished the ERK1/2 activation induced by Go6976. Similar experiments were conducted in human embryonic kidney 293 cells, which express receptors for LPA and EGF but exhibit no significant cross-communication between them. Although Go6976 caused a significant increase in EGF-induced tyrosine phosphorylation of the EGF-R and subsequent ERK1/2 activation, it had no such effects on LPA-induced responses. In Chinese hamster ovary cells, which express receptors for LPA but not for EGF, Go6976 also had no significant effect on LPA-induced ERK1/2 activation. These data indicate that Go6976 potentiates agonist-induced ERK1/2 activation through stimulation of tyrosine phosphorylation of the EGF-R.


Subject(s)
Carbazoles/pharmacology , Enzyme Inhibitors/pharmacology , ErbB Receptors/metabolism , Indoles/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Phosphotyrosine/metabolism , Protein Kinase C/antagonists & inhibitors , Angiotensin II/pharmacology , Animals , CHO Cells , Cell Line , Cricetinae , Enzyme Activation , Epithelial Cells/drug effects , Epithelial Cells/enzymology , ErbB Receptors/drug effects , Humans , Kidney , Liver/cytology , Phosphorylation , Rats
15.
Biochem Pharmacol ; 68(9): 1833-44, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15450949

ABSTRACT

The human corticotropin-releasing factor (hCRF) receptors CRF1 and CRF2(a) couple to the Gs protein. It has been postulated that CRF receptors may also signal through phospholipase C (PLC). To test this hypothesis, binding and signaling properties were determined for both receptor subtypes stably expressed in human embryonic kidney 293 (HEK293) and human SK-N-MC neuroblastoma cells. CRF receptors were highly expressed and strongly coupled to Gs in HEK293 and SK-N-MC cells. However, when the calcium mobilization pathway was investigated, marked differences were observed. In SK-N-MC cells, neither CRF receptor stimulated calcium mobilization in the fluorometric imaging plate reader (FLIPR) assay, whereas activation of orexin type 1 and 2 receptors stably expressed in SK-N-MC cells revealed robust calcium responses. In contrast, intracellular calcium was strongly mobilized by agonist stimulation of hCRF1 and hCRF2(a) receptors in HEK293 cells. In HEK293 cells, potency rank orders for calcium and cAMP responses were identical for both receptors, despite a rightward shift of the dose-response curves. Complete inhibition of calcium signaling of both hCRF1 and hCRF2(a) receptors was observed in the presence of the PLC inhibitor U-73,122 whereas ryanodine, an inhibitor of calcium release channels and the protein kinase A inhibitor Rp-cAMPS were ineffective. Finally, CRF agonists produced a small but significant stimulation of inositol 1,4,5-triphosphate (IP3) accumulation in hCRF1-and hCRF2(a)-transfected HEK293 cells. These data clearly show that phospholipase C-mediated signaling of CRF receptors is dependent upon the cellular background and that in HEK293 cells human CRF receptors robustly respond in the FLIPR format.


Subject(s)
Calcium Signaling/physiology , Receptors, Corticotropin-Releasing Hormone/physiology , Type C Phospholipases/physiology , Calcium/metabolism , Cell Line , Corticotropin-Releasing Hormone/metabolism , Cyclic AMP , Humans , Kidney , Neuroblastoma/pathology , Receptors, Corticotropin-Releasing Hormone/metabolism , Tumor Cells, Cultured
16.
Mol Endocrinol ; 18(8): 2035-48, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15143154

ABSTRACT

Stimulation of the angiotensin II (Ang II) type 1 receptor (AT1-R) causes phosphorylation of extracellularly regulated kinases 1 and 2 (ERK1/2) via epidermal growth factor receptor (EGF-R) transactivation-dependent or -independent pathways in Ang II target cells. Here we examined the mechanisms involved in agonist-induced EGF-R transactivation and subsequent ERK1/2 phosphorylation in clone 9 (C9) hepatocytes, which express endogenous AT1-R, and COS-7 and human embryonic kidney (HEK) 293 cells transfected with the AT1-R. Ang II-induced ERK1/2 activation was attenuated by inhibition of Src kinase and of matrix metalloproteinases (MMPs) in C9 and COS-7 cells, but not in HEK 293 cells. Agonist-mediated MMP activation in C9 cells led to shedding of heparin-binding EGF (HB-EGF) and stimulation of ERK1/2 phosphorylation. Blockade of HB-EGF action by neutralizing antibody or its selective inhibitor, CRM197, attenuated ERK1/2 activation by Ang II. Consistent with its agonist action, HB-EGF stimulation of these cells caused marked phosphorylation of the EGF-R and its adapter molecule, Shc, as well as ERK1/2 and its dependent protein, p90 ribosomal S6 kinase, in a manner similar to that elicited by Ang II or EGF. Although the Tyr319 residue of the AT1-R has been proposed to be an essential regulator of EGF-R transactivation, stimulation of wild-type and mutant (Y319F) AT1-R expressed in COS-7 cells caused EGF-R transactivation and subsequent ERK1/2 phosphorylation through release of HB-EGF in a Src-dependent manner. In contrast, the noninvolvement of MMPs in HEK 293 cells, which may reflect the absence of Src activation by Ang II, was associated with lack of transactivation of the EGF-R. These data demonstrate that the individual actions of Ang II on EGF-R transactivation in specific cell types are related to differential involvement of MMP-dependent HB-EGF release.


Subject(s)
Angiotensin II/pharmacology , Epidermal Growth Factor/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Signal Transduction/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Animals , CSK Tyrosine-Protein Kinase , Cell Line , Chlorocebus aethiops , ErbB Receptors/metabolism , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins , Kinetics , Metalloproteases/antagonists & inhibitors , Metalloproteases/metabolism , Organ Specificity , Phosphorylation/drug effects , Protein Kinase C/metabolism , Protein-Tyrosine Kinases/metabolism , Rats , Receptor, Angiotensin, Type 1 , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1 , Transcriptional Activation/drug effects , src-Family Kinases
17.
J Pharmacol Exp Ther ; 306(2): 794-803, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12734388

ABSTRACT

Protein kinase C (PKC)-mediated desensitization of the corticotropin releasing factor type 1 (CRF1) receptor was investigated in human retinoblastoma Y79 and transfected COS-7 cells. Because stimulation of Y79 cells with CRF resulted in large ( approximately 30-fold) increases in intracellular cAMP accumulation without changing inositol phosphate levels, the CRF1 receptor expressed in retinoblastoma cells couples to Gs, but not to Gq, and predominantly signals via the protein kinase A cascade. Direct activation of PKC by treatment with the phorbol ester phorbol 12-myristate 13-acetate (PMA) or 1,2-dioctanoyl-sn-glycerol (DOG) desensitized CRF1 receptors in Y79 cells, reducing the maximum for CRF- (but not forskolin)-stimulated cAMP accumulation by 56.3 +/- 1.2% and 40.4 +/- 2.1%, respectively (p < 0.001). Pretreating Y79 cells with the PKC inhibitor bisindolylmaleimide I (BIM) markedly inhibited PMA's desensitizing action on CRF-stimulated cAMP accumulation, but did not affect homologous CRF1 receptor desensitization. Retinoblastoma cells were found to express PKCalpha, betaI, betaII, delta, lambda, and RACK1. When alpha and beta isoforms of PKC were down-regulated 80 to 90% by a 48-h PMA exposure, PMA-induced CRF1 receptor desensitization was abolished. In transfected COS-7 cells the magnitude of CRF1 receptor phosphorylation after a 5-min exposure to PMA was 2.32 +/- 0.21-fold greater compared with the basal level. Pretreating COS-7 cells with BIM abolished PMA-induced CRF1 receptor phosphorylation. These studies demonstrate that protein kinase C (possibly alpha and beta isoforms) has an important role in the phosphorylation and heterologous desensitization of the CRF1 receptor.


Subject(s)
Isoenzymes/metabolism , Protein Kinase C/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Acclimatization/physiology , Animals , COS Cells , Cyclic AMP/metabolism , Diglycerides/pharmacology , Dose-Response Relationship, Drug , Down-Regulation , Enzyme Inhibitors/pharmacology , Gene Expression , Humans , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Receptors, Corticotropin-Releasing Hormone/drug effects , Stress, Physiological/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Time Factors , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...