1
|
Yang CM, Yang CC, Hsu WH, Hsiao LD, Tseng HC, Shih YF. Tumor Necrosis Factor-α-Induced C-C Motif Chemokine Ligand 20 Expression through TNF Receptor 1-Dependent Activation of EGFR/p38 MAPK and JNK1/2/FoxO1 or the NF-κB Pathway in Human Cardiac Fibroblasts. Int J Mol Sci 2022; 23:ijms23169086. [PMID: 36012347 PMCID: PMC9409325 DOI: 10.3390/ijms23169086] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 11/16/2022] Open
Abstract
Tumor necrosis factor (TNF)-α is involved in the pathogenesis of cardiac injury, inflammation, and apoptosis. It is a crucial pro-inflammatory cytokine in many heart disorders, including chronic heart failure and ischemic heart disease, contributing to cardiac remodeling and dysfunction. The implication of TNF-α in inflammatory responses in the heart has been indicated to be mediated through the induction of C-C Motif Chemokine Ligand 20 (CCL20). However, the detailed mechanisms of TNF-α-induced CCL20 upregulation in human cardiac fibroblasts (HCFs) are not completely defined. We demonstrated that in HCFs, TNF-α induced CCL20 mRNA expression and promoter activity leading to an increase in the secretion of CCL20. TNF-α-mediated responses were attenuated by pretreatment with TNFR1 antibody, the inhibitor of epidermal growth factor receptor (EGFR) (AG1478), p38 mitogen-activated protein kinase (MAPK) (p38 inhibitor VIII, p38i VIII), c-Jun amino N-terminal kinase (JNK)1/2 (SP600125), nuclear factor kappaB (NF-κB) (helenalin), or forkhead box O (FoxO)1 (AS1841856) and transfection with siRNA of TNFR1, EGFR, p38α, JNK2, p65, or FoxO1. Moreover, TNF-α markedly induced EGFR, p38 MAPK, JNK1/2, FoxO1, and NF-κB p65 phosphorylation which was inhibited by their respective inhibitors in these cells. In addition, TNF-α-enhanced binding of FoxO1 or p65 to the CCL20 promoter was inhibited by p38i VIII, SP600125, and AS1841856, or helenalin, respectively. Accordingly, in HCFs, our findings are the first to clarify that TNF-α-induced CCL20 secretion is mediated through a TNFR1-dependent EGFR/p38 MAPK and JNK1/2/FoxO1 or NF-κB cascade. We demonstrated that TNFR1-derived EGFR transactivation is involved in the TNF-α-induced responses in these cells. Understanding the regulation of CCL20 expression by TNF-α on HCFs may provide a potential therapeutic strategy in cardiac inflammatory disorders.
Collapse
Affiliation(s)
- Chuen-Mao Yang
- Department of Pharmacology, College of Medicine, China Medical University, Taichung 40402, Taiwan
- Ph.D. Program for Biotech Pharmaceutical Industry, China Medical University, Taichung 40402, Taiwan
- Department of Post-Baccalaureate Veterinary Medicine, College of Medical and Health Science, Asia University, Wufeng, Taichung 41354, Taiwan
- Correspondence: ; Tel.: +886-4-22053366 (ext. 2229)
| | - Chien-Chung Yang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital at Tao-Yuan, Kwei-San, Tao-Yuan 33302, Taiwan
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan 33302, Taiwan
| | - Wun-Hsin Hsu
- Department of Pharmacology, College of Medicine, China Medical University, Taichung 40402, Taiwan
| | - Li-Der Hsiao
- Department of Pharmacology, College of Medicine, China Medical University, Taichung 40402, Taiwan
| | - Hui-Ching Tseng
- Department of Pharmacology, College of Medicine, China Medical University, Taichung 40402, Taiwan
| | - Ya-Fang Shih
- Department of Pharmacology, College of Medicine, China Medical University, Taichung 40402, Taiwan
| |
Collapse
|
2
|
Gém JB, Kovács KB, Szalai L, Szakadáti G, Porkoláb E, Szalai B, Turu G, Tóth AD, Szekeres M, Hunyady L, Balla A. Characterization of Type 1 Angiotensin II Receptor Activation Induced Dual-Specificity MAPK Phosphatase Gene Expression Changes in Rat Vascular Smooth Muscle Cells. Cells 2021; 10:3538. [PMID: 34944046 PMCID: PMC8700539 DOI: 10.3390/cells10123538] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 01/03/2023] Open
Abstract
Activation of the type I angiotensin receptor (AT1-R) in vascular smooth muscle cells (VSMCs) plays a crucial role in the regulation of blood pressure; however, it is also responsible for the development of pathological conditions such as vascular remodeling, hypertension and atherosclerosis. Stimulation of the VSMC by angiotensin II (AngII) promotes a broad variety of biological effects, including gene expression changes. In this paper, we have taken an integrated approach in which an analysis of AngII-induced gene expression changes has been combined with the use of small-molecule inhibitors and lentiviral-based gene silencing, to characterize the mechanism of signal transduction in response to AngII stimulation in primary rat VSMCs. We carried out Affymetrix GeneChip experiments to analyze the effects of AngII stimulation on gene expression; several genes, including DUSP5, DUSP6, and DUSP10, were identified as upregulated genes in response to stimulation. Since various dual-specificity MAPK phosphatase (DUSP) enzymes are important in the regulation of mitogen-activated protein kinase (MAPK) signaling pathways, these genes have been selected for further analysis. We investigated the kinetics of gene-expression changes and the possible signal transduction processes that lead to altered expression changes after AngII stimulation. Our data shows that the upregulated genes can be stimulated through multiple and synergistic signal transduction pathways. We have also found in our gene-silencing experiments that epidermal growth factor receptor (EGFR) transactivation is not critical in the AngII-induced expression changes of the investigated genes. Our data can help us understand the details of AngII-induced long-term effects and the pathophysiology of AT1-R. Moreover, it can help to develop potential interventions for those symptoms that are induced by the over-functioning of this receptor, such as vascular remodeling, cardiac hypertrophy or atherosclerosis.
Collapse
Affiliation(s)
- Janka Borbála Gém
- Department of Physiology, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary; (J.B.G.); (K.B.K.); (L.S.); (G.S.); (E.P.); (B.S.); (G.T.); (A.D.T.); (M.S.)
| | - Kinga Bernadett Kovács
- Department of Physiology, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary; (J.B.G.); (K.B.K.); (L.S.); (G.S.); (E.P.); (B.S.); (G.T.); (A.D.T.); (M.S.)
| | - Laura Szalai
- Department of Physiology, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary; (J.B.G.); (K.B.K.); (L.S.); (G.S.); (E.P.); (B.S.); (G.T.); (A.D.T.); (M.S.)
- MTA-SE Laboratory of Molecular Physiology, Hungarian Academy of Sciences and Semmelweis University, 1085 Budapest, Hungary
| | - Gyöngyi Szakadáti
- Department of Physiology, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary; (J.B.G.); (K.B.K.); (L.S.); (G.S.); (E.P.); (B.S.); (G.T.); (A.D.T.); (M.S.)
| | - Edit Porkoláb
- Department of Physiology, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary; (J.B.G.); (K.B.K.); (L.S.); (G.S.); (E.P.); (B.S.); (G.T.); (A.D.T.); (M.S.)
- MTA-SE Laboratory of Molecular Physiology, Hungarian Academy of Sciences and Semmelweis University, 1085 Budapest, Hungary
| | - Bence Szalai
- Department of Physiology, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary; (J.B.G.); (K.B.K.); (L.S.); (G.S.); (E.P.); (B.S.); (G.T.); (A.D.T.); (M.S.)
| | - Gábor Turu
- Department of Physiology, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary; (J.B.G.); (K.B.K.); (L.S.); (G.S.); (E.P.); (B.S.); (G.T.); (A.D.T.); (M.S.)
- MTA-SE Laboratory of Molecular Physiology, Hungarian Academy of Sciences and Semmelweis University, 1085 Budapest, Hungary
| | - András Dávid Tóth
- Department of Physiology, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary; (J.B.G.); (K.B.K.); (L.S.); (G.S.); (E.P.); (B.S.); (G.T.); (A.D.T.); (M.S.)
- MTA-SE Laboratory of Molecular Physiology, Hungarian Academy of Sciences and Semmelweis University, 1085 Budapest, Hungary
- Department of Internal Medicine and Hematology, Semmelweis University, 1085 Budapest, Hungary
| | - Mária Szekeres
- Department of Physiology, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary; (J.B.G.); (K.B.K.); (L.S.); (G.S.); (E.P.); (B.S.); (G.T.); (A.D.T.); (M.S.)
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, 1085 Budapest, Hungary
| | - László Hunyady
- Department of Physiology, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary; (J.B.G.); (K.B.K.); (L.S.); (G.S.); (E.P.); (B.S.); (G.T.); (A.D.T.); (M.S.)
- MTA-SE Laboratory of Molecular Physiology, Hungarian Academy of Sciences and Semmelweis University, 1085 Budapest, Hungary
| | - András Balla
- Department of Physiology, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary; (J.B.G.); (K.B.K.); (L.S.); (G.S.); (E.P.); (B.S.); (G.T.); (A.D.T.); (M.S.)
- MTA-SE Laboratory of Molecular Physiology, Hungarian Academy of Sciences and Semmelweis University, 1085 Budapest, Hungary
| |
Collapse
|
3
|
Fujii A, Masuda T, Iwata M, Tobo T, Wakiyama H, Koike K, Kosai K, Nakano T, Kuramitsu S, Kitagawa A, Sato K, Kouyama Y, Shimizu D, Matsumoto Y, Utsunomiya T, Ohtsuka T, Yamanishi Y, Nakamura M, Mimori K. The novel driver gene ASAP2 is a potential druggable target in pancreatic cancer. Cancer Sci 2021; 112:1655-1668. [PMID: 33605496 PMCID: PMC8019229 DOI: 10.1111/cas.14858] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 02/11/2021] [Accepted: 02/14/2021] [Indexed: 12/13/2022] Open
Abstract
Targeting mutated oncogenes is an effective approach for treating cancer. The 4 main driver genes of pancreatic ductal adenocarcinoma (PDAC) are KRAS, TP53, CDKN2A, and SMAD4, collectively called the "big 4" of PDAC, however they remain challenging therapeutic targets. In this study, ArfGAP with SH3 domain, ankyrin repeat and PH domain 2 (ASAP2), one of the ArfGAP family, was identified as a novel driver gene in PDAC. Clinical analysis with PDAC datasets showed that ASAP2 was overexpressed in PDAC cells based on increased DNA copy numbers, and high ASAP2 expression contributed to a poor prognosis in PDAC. The biological roles of ASAP2 were investigated using ASAP2-knockout PDAC cells generated with CRISPR-Cas9 technology or transfected PDAC cells. In vitro and in vivo analyses showed that ASAP2 promoted tumor growth by facilitating cell cycle progression through phosphorylation of epidermal growth factor receptor (EGFR). A repositioned drug targeting the ASAP2 pathway was identified using a bioinformatics approach. The gene perturbation correlation method showed that niclosamide, an antiparasitic drug, suppressed PDAC growth by inhibition of ASAP2 expression. These data show that ASAP2 is a novel druggable driver gene that activates the EGFR signaling pathway. Furthermore, niclosamide was identified as a repositioned therapeutic agent for PDAC possibly targeting ASAP2.
Collapse
Affiliation(s)
- Atsushi Fujii
- Department of SurgeryKyushu University Beppu HospitalOitaJapan
- Department of Surgery and OncologyGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Takaaki Masuda
- Department of SurgeryKyushu University Beppu HospitalOitaJapan
| | - Michio Iwata
- Department of Bioscience and BioinformaticsFaculty of Computer Science and Systems EngineeringKyushu Institute of TechnologyFukuokaJapan
| | - Taro Tobo
- Department of Clinical Laboratory MedicineKyushu University Beppu HospitalOitaJapan
| | | | - Kensuke Koike
- Department of SurgeryKyushu University Beppu HospitalOitaJapan
| | - Keisuke Kosai
- Department of SurgeryKyushu University Beppu HospitalOitaJapan
| | - Takafumi Nakano
- Department of SurgeryKyushu University Beppu HospitalOitaJapan
| | | | | | - Kuniaki Sato
- Department of SurgeryKyushu University Beppu HospitalOitaJapan
| | - Yuta Kouyama
- Department of SurgeryKyushu University Beppu HospitalOitaJapan
| | - Dai Shimizu
- Department of SurgeryKyushu University Beppu HospitalOitaJapan
| | | | | | - Takao Ohtsuka
- Department of Digestive Surgery, Breast and Thyroid SurgeryKagoshima UniversityKagoshimaJapan
| | - Yoshihiro Yamanishi
- Department of Bioscience and BioinformaticsFaculty of Computer Science and Systems EngineeringKyushu Institute of TechnologyFukuokaJapan
| | - Masafumi Nakamura
- Department of Surgery and OncologyGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Koshi Mimori
- Department of SurgeryKyushu University Beppu HospitalOitaJapan
| |
Collapse
|
4
|
Wang W, Wu J, Mukherjee A, He T, Wang XY, Ma Y, Fang X. Lysophosphatidic acid induces tumor necrosis factor-alpha to regulate a pro-inflammatory cytokine network in ovarian cancer. FASEB J 2020; 34:13935-13948. [PMID: 32851734 DOI: 10.1096/fj.202001136r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 12/13/2022]
Abstract
Epithelial ovarian carcinoma tissues express high levels of tumor necrosis factor-alpha (TNF-α) and other inflammatory cytokines. The underlying mechanism leading to the abnormal TNF-α expression in ovarian cancer remains poorly understood. In the current study, we demonstrated that lysophosphatidic acid (LPA), a lipid mediator present in ascites of ovarian cancer patients, induced expression of TNF-α mRNA and release of TNF-α protein in ovarian cancer cells. LPA also induced expression of interleukin-1β (IL-1β) mRNA but no significant increase in IL-1β protein was detected. LPA enhanced TNF-α mRNA through NF-κB-mediated transcriptional activation. Inactivation of ADAM17, a disintegrin and metalloproteinase, with a specific inhibitor TMI-1 or by shRNA knockdown prevented ovarian cancer cells from releasing TNF-α protein in response to LPA, indicating that LPA-mediated TNF-α production relies on both transcriptional upregulations of the TNF-α gene and the activity of ADAM17, the membrane-associated TNF-α-converting enzyme. Like many other biological responses to LPA, induction of TNF-α by LPA also depended on the transactivation of the epidermal growth factor receptor (EGFR). Interestingly, our results revealed that ADAM17 was also the shedding protease responsible for the transactivation of EGFR by LPA in ovarian cancer cells. To explore the biological outcomes of LPA-induced TNF-α, we examined the effects of a TNF-α neutralizing antibody and recombinant TNF-α soluble receptor on LPA-stimulated expression of pro-tumorigenic cytokines and chemokines overexpressed in ovarian cancer. Blockade of TNF-α signaling significantly reduced the production of IL-8, IL-6, and CXCL1, suggesting a hierarchy of mechanisms contributing to the robust expression of the inflammatory mediators in response to LPA in ovarian cancer cells. In contrast, TNF-α inhibition did not affect LPA-dependent cell proliferation. Taken together, our results establish that the bioactive lipid LPA drives the expression of TNF-α to regulate an inflammatory network in ovarian cancer.
Collapse
Affiliation(s)
- Wei Wang
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - Jinhua Wu
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - Abir Mukherjee
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - Tianhai He
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - Xiang-Yang Wang
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Yibao Ma
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - Xianjun Fang
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
5
|
DeLalio LJ, Billaud M, Ruddiman CA, Johnstone SR, Butcher JT, Wolpe AG, Jin X, Keller TCS, Keller AS, Rivière T, Good ME, Best AK, Lohman AW, Swayne LA, Penuela S, Thompson RJ, Lampe PD, Yeager M, Isakson BE. Constitutive SRC-mediated phosphorylation of pannexin 1 at tyrosine 198 occurs at the plasma membrane. J Biol Chem 2019; 294:6940-6956. [PMID: 30814251 DOI: 10.1074/jbc.ra118.006982] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 02/15/2019] [Indexed: 11/06/2022] Open
Abstract
Pannexin 1 (PANX1)-mediated ATP release in vascular smooth muscle coordinates α1-adrenergic receptor (α1-AR) vasoconstriction and blood pressure homeostasis. We recently identified amino acids 198-200 (YLK) on the PANX1 intracellular loop that are critical for α1-AR-mediated vasoconstriction and PANX1 channel function. We report herein that the YLK motif is contained within an SRC homology 2 domain and is directly phosphorylated by SRC proto-oncogene, nonreceptor tyrosine kinase (SRC) at Tyr198 We demonstrate that PANX1-mediated ATP release occurs independently of intracellular calcium but is sensitive to SRC family kinase (SFK) inhibition, suggestive of channel regulation by tyrosine phosphorylation. Using a PANX1 Tyr198-specific antibody, SFK inhibitors, SRC knockdown, temperature-dependent SRC cells, and kinase assays, we found that PANX1-mediated ATP release and vasoconstriction involves constitutive phosphorylation of PANX1 Tyr198 by SRC. We specifically detected SRC-mediated Tyr198 phosphorylation at the plasma membrane and observed that it is not enhanced or induced by α1-AR activation. Last, we show that PANX1 immunostaining is enriched in the smooth muscle layer of arteries from hypertensive humans and that Tyr198 phosphorylation is detectable in these samples, indicative of a role for membrane-associated PANX1 in small arteries of hypertensive humans. Our discovery adds insight into the regulation of PANX1 by post-translational modifications and connects a significant purinergic vasoconstriction pathway with a previously identified, yet unexplored, tyrosine kinase-based α1-AR constriction mechanism. This work implicates SRC-mediated PANX1 function in normal vascular hemodynamics and suggests that Tyr198-phosphorylated PANX1 is involved in hypertensive vascular pathology.
Collapse
Affiliation(s)
- Leon J DeLalio
- From the Robert M. Berne Cardiovascular Research Center.,Department of Pharmacology
| | - Marie Billaud
- the Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Claire A Ruddiman
- From the Robert M. Berne Cardiovascular Research Center.,Department of Pharmacology
| | | | - Joshua T Butcher
- the Department of Physiology, Augusta University, Augusta, Georgia 30912
| | - Abigail G Wolpe
- From the Robert M. Berne Cardiovascular Research Center.,Department of Cell Biology, and
| | - Xueyao Jin
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia 22908
| | - T C Stevenson Keller
- From the Robert M. Berne Cardiovascular Research Center.,Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia 22908
| | - Alexander S Keller
- From the Robert M. Berne Cardiovascular Research Center.,Department of Pharmacology
| | - Thibaud Rivière
- the Department of Life and Health Sciences, University of Bordeaux, 33000 Bordeaux, France
| | | | - Angela K Best
- From the Robert M. Berne Cardiovascular Research Center
| | - Alexander W Lohman
- the Hotchkiss Brain Institute and.,Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Leigh Anne Swayne
- the Division of Medical Sciences, Centre for Biomedical Research, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Silvia Penuela
- the Departments of Anatomy and Cell Biology and Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario N6A 3K7, Canada, and
| | - Roger J Thompson
- the Hotchkiss Brain Institute and.,Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Paul D Lampe
- the Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| | - Mark Yeager
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia 22908
| | - Brant E Isakson
- From the Robert M. Berne Cardiovascular Research Center, .,Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia 22908
| |
Collapse
|
6
|
Parra-Mercado GK, Fuentes-Gonzalez AM, Hernandez-Aranda J, Diaz-Coranguez M, Dautzenberg FM, Catt KJ, Hauger RL, Olivares-Reyes JA. CRF 1 Receptor Signaling via the ERK1/2-MAP and Akt Kinase Cascades: Roles of Src, EGF Receptor, and PI3-Kinase Mechanisms. Front Endocrinol (Lausanne) 2019; 10:869. [PMID: 31920979 PMCID: PMC6921279 DOI: 10.3389/fendo.2019.00869] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 11/27/2019] [Indexed: 12/28/2022] Open
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 Gβγ 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 Gβγ-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/βγ 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 β-arrestin-2 recruitment; however, this situation may have resulted from the low β-arrestin expression in the COS-7 cells. When β-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/βγ 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.
Collapse
Affiliation(s)
- G. Karina Parra-Mercado
- Laboratory of Signal Transduction, Department of Biochemistry, Center for Research and Advanced Studies of the National Polytechnic Institute, CINVESTAV-IPN, Mexico City, Mexico
| | - Alma M. Fuentes-Gonzalez
- Laboratory of Signal Transduction, Department of Biochemistry, Center for Research and Advanced Studies of the National Polytechnic Institute, CINVESTAV-IPN, Mexico City, Mexico
| | - Judith Hernandez-Aranda
- Laboratory of Signal Transduction, Department of Biochemistry, Center for Research and Advanced Studies of the National Polytechnic Institute, CINVESTAV-IPN, Mexico City, Mexico
| | - Monica Diaz-Coranguez
- Laboratory of Signal Transduction, Department of Biochemistry, Center for Research and Advanced Studies of the National Polytechnic Institute, CINVESTAV-IPN, Mexico City, Mexico
| | | | - Kevin J. Catt
- Section on Hormonal Regulation, Program on Developmental Endocrinology and Genetics, National Institute of Child Health and Human Development, Bethesda, MD, United States
| | - Richard L. Hauger
- Center of Excellence for Stress and Mental Health, VA Healthcare System, San Diego, CA, United States
- Department of Psychiatry, University of California, San Diego, San Diego, CA, United States
| | - J. Alberto Olivares-Reyes
- Laboratory of Signal Transduction, Department of Biochemistry, Center for Research and Advanced Studies of the National Polytechnic Institute, CINVESTAV-IPN, Mexico City, Mexico
- *Correspondence: J. Alberto Olivares-Reyes
| |
Collapse
|
7
|
O'Brien SL, Johnstone EKM, Devost D, Conroy J, Reichelt ME, Purdue BW, Ayoub MA, Kawai T, Inoue A, Eguchi S, Hébert TE, Pfleger KDG, Thomas WG. BRET-based assay to monitor EGFR transactivation by the AT 1R reveals G q/11 protein-independent activation and AT 1R-EGFR complexes. Biochem Pharmacol 2018; 158:232-242. [PMID: 30347205 DOI: 10.1016/j.bcp.2018.10.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 10/17/2018] [Indexed: 01/09/2023]
Abstract
The type 1 angiotensin II (AngII) receptor (AT1R) transactivates the epidermal growth factor receptor (EGFR), which leads to pathological remodeling of heart, blood vessels and kidney. End-point assays are used as surrogates of EGFR activation, however these downstream readouts are not applicable to live cells, in real-time. Herein, we report the use of a bioluminescence resonance energy transfer (BRET)-based assay to assess recruitment of the EGFR adaptor protein, growth factor receptor-bound protein 2 (Grb2), to the EGFR. In a variety of cell lines, both epidermal growth factor (EGF) and AngII stimulated Grb2 recruitment to EGFR. The BRET assay was used to screen a panel of 9 G protein-coupled receptors (GPCRs) and further developed for other EGFR family members (HER2 and HER3); the AT1R was able to transactivate HER2, but not HER3. Mechanistically, AT1R-mediated ERK1/2 activation was dependent on Gq/11 and EGFR tyrosine kinase activity, whereas the recruitment of Grb2 to the EGFR was independent of Gq/11 and only partially dependent on EGFR tyrosine kinase activity. This Gq/11 independence of EGFR transactivation was confirmed using AT1R mutants and in CRISPR cell lines lacking Gq/11. EGFR transactivation was also apparently independent of β-arrestins. Finally, we used additional BRET-based assays and confocal microscopy to provide evidence that both AngII- and EGF-stimulation promoted AT1R-EGFR heteromerization. In summary, we report an alternative approach to monitoring AT1R-EGFR transactivation in live cells, which provides a more direct and proximal view of this process, including the potential for complexes between the AT1R and EGFR.
Collapse
Affiliation(s)
- Shannon L O'Brien
- Receptor Biology Group, The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia 4072, Queensland, Australia
| | - Elizabeth K M Johnstone
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Crawley, Western Australia 6009, Australia
| | - Dominic Devost
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Jacinta Conroy
- Receptor Biology Group, The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia 4072, Queensland, Australia
| | - Melissa E Reichelt
- Receptor Biology Group, The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia 4072, Queensland, Australia
| | - Brooke W Purdue
- Receptor Biology Group, The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia 4072, Queensland, Australia
| | - Mohammed A Ayoub
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Crawley, Western Australia 6009, Australia
| | - Tatsuo Kawai
- Cardiovascular Research Centre, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Satoru Eguchi
- Cardiovascular Research Centre, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Kevin D G Pfleger
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Crawley, Western Australia 6009, Australia; Dimerix Limited, Nedlands, Western Australia 6009, Australia
| | - Walter G Thomas
- Receptor Biology Group, The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia 4072, Queensland, Australia; Centre for Cardiac and Vasculature Biology, The University of Queensland, St Lucia 4072, Queensland, Australia.
| |
Collapse
|
8
|
Blurring Boundaries: Receptor Tyrosine Kinases as functional G Protein-Coupled Receptors. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 339:1-40. [DOI: 10.1016/bs.ircmb.2018.02.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
9
|
Cardiovascular homeostasis dependence on MICU2, a regulatory subunit of the mitochondrial calcium uniporter. Proc Natl Acad Sci U S A 2017; 114:E9096-E9104. [PMID: 29073106 PMCID: PMC5664535 DOI: 10.1073/pnas.1711303114] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hypertension increases the risk for development of abdominal aortic aneurysms, a silent pathology that is prone to rupture and cause sudden cardiac death. Male gender, smoking, and hypertension appear to increase risk for development of abdominal aortic aneurysms by provoking oxidative stress responses in cardiovascular tissues. Here we uncovered unexpected linkages between the calcium-sensing regulatory subunit MICU2 of the mitochondrial calcium uniporter and stress responses. We show that naive Micu2−/− mice had abnormalities of cardiac relaxation but, with modest blood pressure elevation, developed abdominal aortic aneurysms with spontaneous rupture. These findings implicate mitochondrial calcium homeostasis as a critical pathway involved in protecting cardiovascular tissues from oxidative stress. Comparative analyses of transcriptional profiles from humans and mice with cardiovascular pathologies revealed consistently elevated expression of MICU2, a regulatory subunit of the mitochondrial calcium uniporter complex. To determine if MICU2 expression was cardioprotective, we produced and characterized Micu2−/− mice. Mutant mice had left atrial enlargement and Micu2−/− cardiomyocytes had delayed sarcomere relaxation and cytosolic calcium reuptake kinetics, indicating diastolic dysfunction. RNA sequencing (RNA-seq) of Micu2−/− ventricular tissues revealed markedly reduced transcripts encoding the apelin receptor (Micu2−/− vs. wild type, P = 7.8 × 10−40), which suppresses angiotensin II receptor signaling via allosteric transinhibition. We found that Micu2−/− and wild-type mice had comparable basal blood pressures and elevated responses to angiotensin II infusion, but that Micu2−/− mice exhibited systolic dysfunction and 30% lethality from abdominal aortic rupture. Aneurysms and rupture did not occur with norepinephrine-induced hypertension. Aortic tissue from Micu2−/− mice had increased expression of extracellular matrix remodeling genes, while single-cell RNA-seq analyses showed increased expression of genes related to reactive oxygen species, inflammation, and proliferation in fibroblast and smooth muscle cells. We concluded that Micu2−/− mice recapitulate features of diastolic heart disease and define previously unappreciated roles for Micu2 in regulating angiotensin II-mediated hypertensive responses that are critical in protecting the abdominal aorta from injury.
Collapse
|
10
|
Lipoxin A 4 activates ALX/FPR2 receptor to regulate conjunctival goblet cell secretion. Mucosal Immunol 2017; 10:46-57. [PMID: 27072607 PMCID: PMC5063650 DOI: 10.1038/mi.2016.33] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 02/22/2016] [Indexed: 02/04/2023]
Abstract
Conjunctival goblet cells play a major role in maintaining the mucus layer of the tear film under physiological conditions as well as in inflammatory diseases like dry eye and allergic conjunctivitis. Resolution of inflammation is mediated by proresolution agonists such as lipoxin A4 (LXA4) that can also function under physiological conditions. The purpose of this study was to determine the actions of LXA4 on cultured rat conjunctival goblet cell mucin secretion, intracellular [Ca2+] ([Ca2+]i), and identify signaling pathways activated by LXA4. ALX/FPR2 (formyl peptide receptor2) was localized to goblet cells in rat conjunctiva and in cultured goblet cells. LXA4 significantly increased mucin secretion, [Ca2+]i, and extracellular regulated kinase 1/2 (ERK 1/2) activation. These functions were inhibited by ALX/FPR2 inhibitors. Stable analogs of LXA4 increased [Ca2+]i to the same extent as LXA4. Sequential addition of either LXA4 or resolvin D1 followed by the second compound decreased [Ca2+]i of the second compound compared with its initial response. LXA4 activated phospholipases C, D, and A2 and downstream molecules protein kinase C, ERK 1/2, and Ca2+/calmodulin-dependent kinase to increase mucin secretion and [Ca2+]i. We conclude that conjunctival goblet cells respond to LXA4 to maintain the homeostasis of the ocular surface and could be a novel treatment for dry eye diseases.
Collapse
|
11
|
Anguita E, Villalobo A. Src-family tyrosine kinases and the Ca 2+ signal. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1864:915-932. [PMID: 27818271 DOI: 10.1016/j.bbamcr.2016.10.022] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 10/25/2016] [Accepted: 10/30/2016] [Indexed: 01/08/2023]
Abstract
In this review, we shall describe the rich crosstalk between non-receptor Src-family kinases (SFKs) and the Ca2+ transient generated in activated cells by a variety of extracellular and intracellular stimuli, resulting in diverse signaling events. The exchange of information between SFKs and Ca2+ is reciprocal, as it flows in both directions. These kinases are main actors in pathways leading to the generation of the Ca2+ signal, and reciprocally, the Ca2+ signal modulates SFKs activity and functions. We will cover how SFKs participate in the generation of the cytosolic Ca2+ rise upon activation of a series of receptors and the mechanism of clearance of this Ca2+ signal. The role of SFKs modulating Ca2+-translocating channels participating in these events will be amply discussed. Finally, the role of the Ca2+ sensor protein calmodulin on the activity of c-Src, and potentially on other SFKs, will be outlined as well. This article is part of a Special Issue entitled: ECS Meeting edited by Claus Heizmann, Joachim Krebs and Jacques Haiech.
Collapse
Affiliation(s)
- Estefanía Anguita
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, c/ Arturo Duperier 4, E-28029 Madrid, Spain
| | - Antonio Villalobo
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, c/ Arturo Duperier 4, E-28029 Madrid, Spain.
| |
Collapse
|
12
|
Doyle JJ, Doyle AJ, Wilson NK, Habashi JP, Bedja D, Whitworth RE, Lindsay ME, Schoenhoff F, Myers L, Huso N, Bachir S, Squires O, Rusholme B, Ehsan H, Huso D, Thomas CJ, Caulfield MJ, Van Eyk JE, Judge DP, Dietz HC. A deleterious gene-by-environment interaction imposed by calcium channel blockers in Marfan syndrome. eLife 2015; 4. [PMID: 26506064 PMCID: PMC4621743 DOI: 10.7554/elife.08648] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 09/22/2015] [Indexed: 01/01/2023] Open
Abstract
Calcium channel blockers (CCBs) are prescribed to patients with Marfan syndrome for prophylaxis against aortic aneurysm progression, despite limited evidence for their efficacy and safety in the disorder. Unexpectedly, Marfan mice treated with CCBs show accelerated aneurysm expansion, rupture, and premature lethality. This effect is both extracellular signal-regulated kinase (ERK1/2) dependent and angiotensin-II type 1 receptor (AT1R) dependent. We have identified protein kinase C beta (PKCβ) as a critical mediator of this pathway and demonstrate that the PKCβ inhibitor enzastaurin, and the clinically available anti-hypertensive agent hydralazine, both normalize aortic growth in Marfan mice, in association with reduced PKCβ and ERK1/2 activation. Furthermore, patients with Marfan syndrome and other forms of inherited thoracic aortic aneurysm taking CCBs display increased risk of aortic dissection and need for aortic surgery, compared to patients on other antihypertensive agents. DOI:http://dx.doi.org/10.7554/eLife.08648.001 Marfan syndrome is a disorder that affects the body's connective tissues, which maintain the structure of the body and support organs and other tissues. People with Marfan syndrome have connective tissues that can stretch more than those of other people, which put them at increased risk of a life-threatening tear in their aorta (the main artery in the body), muscle weakness and other problems. A cell communication pathway called TGFβ signaling is involved in cell growth and many other important processes. TGFβ signaling is more active in patients with Marfan syndrome due to mutations in a gene called FBN1. Drugs that block TGFβ signaling—which are also used to treat high blood pressure—can reduce the symptoms of the disorder. Unfortunately, not all people with Marfan disease can tolerate these drugs and other medications called calcium channel blockers, which also lower blood pressure, are often used as an alternative. It is thought that calcium channel blockers help reduce stress on blood vessels, but there is little data to show whether these drugs are safe and helpful for patients with Marfan syndrome. Now, Doyle, Doyle et al. studied the effect of two different calcium channel blockers on mice that have a mutation in Fbn1—the mouse equivalent of FBN1—that is similar to those found in humans with Marfan syndrome. The experiments show that the aortas of these mice grew more quickly and were more likely to tear when compared to mice that did not receive these drugs. Many of these aortic tears were fatal. The calcium channel blockers increased the activity of two signaling molecules that are regulated by TGFβ signaling. Treating the Marfan mice with other drugs that lower the activity of these signaling molecules protected the aorta, even if they were also treated with the calcium channel blockers. Doyle, Doyle et al. examined a registry of human patients. This revealed preliminary evidence that aortic tears and aortic repair surgery were more common in patients with Marfan syndrome who had received calcium channel blockers than patients who had been treated with other drugs. Together, these findings suggest that it may be dangerous to treat patients with Marfan syndrome with calcium channel blockers. Additional work will be needed to confirm this risk, to find out if it extends to other similar conditions, and to explore the therapeutic potential of drugs that target the two enzymes. DOI:http://dx.doi.org/10.7554/eLife.08648.002
Collapse
Affiliation(s)
- Jefferson J Doyle
- Howard Hughes Medical Institute and Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, United States.,Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Alexander J Doyle
- Howard Hughes Medical Institute and Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, United States.,William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, United Kingdom
| | - Nicole K Wilson
- Howard Hughes Medical Institute and Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Jennifer P Habashi
- Howard Hughes Medical Institute and Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, United States.,Division of Pediatric Cardiology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Djahida Bedja
- Department of Cardiology, Johns Hopkins University School of Medicine, Baltimore, United States.,Australian School of Advanced Medicine, Macquarie University, Sydney, Australia
| | - Ryan E Whitworth
- Research Triangle Institute International, Durham, United States
| | - Mark E Lindsay
- Thoracic Aortic Center, Departments of Medicine and Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, United States
| | - Florian Schoenhoff
- Department of Cardiovascular Surgery, Inselspital, Bern, Switzerland.,Proteomics Innovation Center in Heart Failure, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Loretha Myers
- Howard Hughes Medical Institute and Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Nick Huso
- Howard Hughes Medical Institute and Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Suha Bachir
- Howard Hughes Medical Institute and Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Oliver Squires
- Howard Hughes Medical Institute and Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Benjamin Rusholme
- Howard Hughes Medical Institute and Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Hamid Ehsan
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, United States
| | - David Huso
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Craig J Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Bethesda, United States
| | - Mark J Caulfield
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, United Kingdom
| | - Jennifer E Van Eyk
- Proteomics Innovation Center in Heart Failure, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Daniel P Judge
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Harry C Dietz
- Howard Hughes Medical Institute and Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, United States.,Division of Pediatric Cardiology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, United States.,Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, United States
| | | | | |
Collapse
|
13
|
Cattaneo F, Guerra G, Parisi M, De Marinis M, Tafuri D, Cinelli M, Ammendola R. Cell-surface receptors transactivation mediated by g protein-coupled receptors. Int J Mol Sci 2014; 15:19700-28. [PMID: 25356505 PMCID: PMC4264134 DOI: 10.3390/ijms151119700] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 09/30/2014] [Accepted: 10/13/2014] [Indexed: 12/17/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are seven transmembrane-spanning proteins belonging to a large family of cell-surface receptors involved in many intracellular signaling cascades. Despite GPCRs lack intrinsic tyrosine kinase activity, tyrosine phosphorylation of a tyrosine kinase receptor (RTK) occurs in response to binding of specific agonists of several such receptors, triggering intracellular mitogenic cascades. This suggests that the notion that GPCRs are associated with the regulation of post-mitotic cell functions is no longer believable. Crosstalk between GPCR and RTK may occur by different molecular mechanism such as the activation of metalloproteases, which can induce the metalloprotease-dependent release of RTK ligands, or in a ligand-independent manner involving membrane associated non-receptor tyrosine kinases, such as c-Src. Reactive oxygen species (ROS) are also implicated as signaling intermediates in RTKs transactivation. Intracellular concentration of ROS increases transiently in cells stimulated with GPCR agonists and their deliberated and regulated generation is mainly catalyzed by enzymes that belong to nicotinamide adenine dinucleotide phosphate (NADPH) oxidase family. Oxidation and/or reduction of cysteine sulfhydryl groups of phosphatases tightly controls the activity of RTKs and ROS-mediated inhibition of cellular phosphatases results in an equilibrium shift from the non-phosphorylated to the phosphorylated state of RTKs. Many GPCR agonists activate phospholipase C, which catalyze the hydrolysis of phosphatidylinositol 4,5-bis-phosphate to produce inositol 1,4,5-triphosphate and diacylglicerol. The consequent mobilization of Ca2+ from endoplasmic reticulum leads to the activation of protein kinase C (PKC) isoforms. PKCα mediates feedback inhibition of RTK transactivation during GPCR stimulation. Recent data have expanded the coverage of transactivation to include Serine/Threonine kinase receptors and Toll-like receptors. Herein, we discuss the main mechanisms of GPCR-mediated cell-surface receptors transactivation and the pathways involved in intracellular responses induced by GPCR agonists. These studies may suggest the design of novel strategies for therapeutic interventions.
Collapse
Affiliation(s)
- Fabio Cattaneo
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, Naples 80131, Italy.
| | - Germano Guerra
- Department of Medicine and Health Sciences, University of Molise, Campobasso 86100, Italy.
| | - Melania Parisi
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, Naples 80131, Italy.
| | - Marta De Marinis
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, Naples 80131, Italy.
| | - Domenico Tafuri
- Department of Sport Science and Wellness, University of Naples Parthenope, Naples 80133, Italy.
| | - Mariapia Cinelli
- Department of Public Health, School of Medicine, University of Naples Federico II, Naples 80131, Italy.
| | - Rosario Ammendola
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, Naples 80131, Italy.
| |
Collapse
|
14
|
Ogawa A, Firth AL, Ariyasu S, Yamadori I, Matsubara H, Song S, Fraidenburg DR, Yuan JXJ. Thrombin-mediated activation of Akt signaling contributes to pulmonary vascular remodeling in pulmonary hypertension. Physiol Rep 2013; 1:e00190. [PMID: 24744867 PMCID: PMC3970741 DOI: 10.1002/phy2.190] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 11/27/2013] [Indexed: 12/11/2022] Open
Abstract
Chronic thromboembolic pulmonary hypertension (CTEPH) has been increasingly recognized as a common source of elevated pulmonary vascular resistance and pulmonary hypertension. It is clear that development of pulmonary thromboemboli is the inciting event for this process, yet it remains unclear why some patients have persistent pulmonary artery occlusion leading to distal pulmonary vascular remodeling and CTEPH. Thrombin, a serine protease, is an integral part of the common coagulation cascade, yet thrombin also has direct cellular effects through interaction with the family of PAR membrane receptors. This study is designed to determine the effects of thrombin on Akt signaling in pulmonary artery smooth muscle cells (PASMC) from normal humans and pulmonary hypertension patients. Thrombin treatment of PASMC resulted in a transient increase in Akt phosphorylation and had similar effects on the downstream targets of the Akt/mTOR pathway. Ca2+ is shown to be required for Akt phosphorylation as well as serum starvation, a distinct effect compared to platelet‐derived growth factor. Thrombin treatment was associated with a rise in intracellular [Ca2+] and enhanced store‐operated calcium entry (SOCE). These effects lead to enhanced proliferation, which is more dramatic in both IPAH and CTEPH PASMC. Enhanced proliferation is also shown to be attenuated by inhibition of Akt/mTOR in CTEPH PASMC. Thrombin has direct effects on PASMC increasing intracellular [Ca2+] and PASMC proliferation, an effect attributed to Akt phosphorylation. The current results implicate the effects of thrombin in the pathogenesis of idiopathic pulmonary arterial hypertension (IPAH) and CTEPH, which may potentially be a novel therapeutic target. Thrombin is known to play an important role in thrombotic events including pulmonary embolism. In this manuscript, we show a direct effect of thrombin on pulmonary artery smooth muscle cells in both normal and diseased states through Akt signaling, which leads to increased store‐operated calcium entry and cellular proliferation. These direct effects of thrombin may play a role in the development and progression of chronic thromboembolic pulmonary hypertension.
Collapse
Affiliation(s)
- Aiko Ogawa
- Department of Clinical Science, National Hospital Organization Okayama Medical CenterTamasu, Kita-kuOkayama, Japan
| | - Amy L Firth
- The Salk Institute of Biological Studies, La Jolla, California
| | - Sanae Ariyasu
- Clinical Pathology, National Hospital Organization Okayama Medical Center, TamasuKita-kuOkayama, Japan
| | - Ichiro Yamadori
- Clinical Pathology, National Hospital Organization Okayama Medical Center, TamasuKita-kuOkayama, Japan
| | - Hiromi Matsubara
- Department of Clinical Science, National Hospital Organization Okayama Medical CenterTamasu, Kita-kuOkayama, Japan
| | - Shanshan Song
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois at Chicago, Chicago, Illinois ; Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois
| | - Dustin R Fraidenburg
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois at Chicago, Chicago, Illinois ; Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois
| | - Jason X-J Yuan
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois at Chicago, Chicago, Illinois ; Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois ; Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
15
|
George AJ, Purdue BW, Gould CM, Thomas DW, Handoko Y, Qian H, Quaife-Ryan GA, Morgan KA, Simpson KJ, Thomas WG, Hannan RD. A functional siRNA screen identifies genes modulating angiotensin II-mediated EGFR transactivation. J Cell Sci 2013; 126:5377-90. [PMID: 24046455 DOI: 10.1242/jcs.128280] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The angiotensin type 1 receptor (AT1R) transactivates the epidermal growth factor receptor (EGFR) to mediate cellular growth, however, the molecular mechanisms involved have not yet been resolved. To address this, we performed a functional siRNA screen of the human kinome in human mammary epithelial cells that demonstrate a robust AT1R-EGFR transactivation. We identified a suite of genes encoding proteins that both positively and negatively regulate AT1R-EGFR transactivation. Many candidates are components of EGFR signalling networks, whereas others, including TRIO, BMX and CHKA, have not been previously linked to EGFR transactivation. Individual knockdown of TRIO, BMX or CHKA attenuated tyrosine phosphorylation of the EGFR by angiotensin II stimulation, but this did not occur following direct stimulation of the EGFR with EGF, indicating that these proteins function between the activated AT1R and the EGFR. Further investigation of TRIO and CHKA revealed that their activity is likely to be required for AT1R-EGFR transactivation. CHKA also mediated EGFR transactivation in response to another G protein-coupled receptor (GPCR) ligand, thrombin, indicating a pervasive role for CHKA in GPCR-EGFR crosstalk. Our study reveals the power of unbiased, functional genomic screens to identify new signalling mediators important for tissue remodelling in cardiovascular disease and cancer.
Collapse
Affiliation(s)
- Amee J George
- School of Biomedical Sciences, The University of Queensland, St. Lucia, Queensland, 4072, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Jang EJ, Seok YM, Arterburn JB, Olatunji LA, Kim IK. GPER-1 agonist G1 induces vasorelaxation through activation of epidermal growth factor receptor-dependent signalling pathway. J Pharm Pharmacol 2013; 65:1488-99. [DOI: 10.1111/jphp.12113] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Accepted: 06/15/2013] [Indexed: 01/07/2023]
Abstract
Abstract
Objectives
The G protein-coupled oestrogen receptor-1 (GPER-1) agonist G1 induces endothelium-dependent relaxation. Activation of the epidermal growth factor (EGF) receptor leads to transduction of signals from the plasma membrane for the release of nitric oxide. We tested the hypothesis that G1 induces endothelium-dependent vasorelaxation through activation of the EGF receptor.
Methods
Rat aortic rings were mounted in organ baths. After pretreatment with various inhibitors, aortic rings contracted with 11,9-epoxymethano-prostaglandin F2α or KCl were subjected to relaxation by G1.
Key findings
G1 induced endothelium-dependent vasorelaxation, which was attenuated by pretreatment with either L-Nω-nitroarginine methyl ester (L-NAME), an inhibitor of nitric oxide synthase, or (3aS,4R,9bR)-4-(6-bromo-1,3-benzodioxol-5-yl)-3a,4,5,9b-tetrahydro-3H-cyclopenta[c]quinoline HB-EGF, heparin-binding EGF-like growth factor, a GPER-1 antagonist. Neither a general oestrogen receptor antagonist, ICI 182 780, nor a selective oestrogen receptor-α antagonist, methyl-piperidino-pyrazole dihydrochloride (MPP), had an effect on G1-induced vasorelaxation. However, pretreatment with EGF receptor blockers, AG1478 or DAPH, resulted in attenuated G1-induced vasorelaxation. In addition, pretreatment with Src inhibitor 4-amino-3-(4-chlorophenyl)-1-(t-butyl)-1H-pyrazolo[3,4-d]pyrimidine, 4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine or Akt inhibitor VIII also resulted in attenuated vascular relaxation induced by the cumulative addition of G1. However, neither phosphatidylinositol-3 kinase inhibitors LY294002 and wortmannin nor an extracellular signal-regulated kinase inhibitor 1,4-diamino-2,3-dicyano-1,4-bis(o-aminophenylmercapto) butadiene monoethanolate had effect on vascular relaxation induced by the cumulative addition of G1.
Conclusions
G1 induces endothelium-dependent vasorelaxation through Src-mediated activation of the EGF receptor and the Akt pathway in rat aorta.
Collapse
Affiliation(s)
- Eun Jin Jang
- Department of Pharmacology, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Young Mi Seok
- Cardiovascular Research Institute, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Jeffrey B Arterburn
- Department of Chemistry and Biochemistry MSC 3C, New Mexico State University, Las Cruces, New Mexico, USA
| | - Lawrence A Olatunji
- Department of Physiology, Cardiovascular and Membrane Physiology, Ilorin, Nigeria
- Basic Medical Sciences Unit, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - In Kyeom Kim
- Department of Pharmacology, Kyungpook National University School of Medicine, Daegu, Republic of Korea
- Cardiovascular Research Institute, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| |
Collapse
|
17
|
Oxidative stress induced by P2X7 receptor stimulation in murine macrophages is mediated by c-Src/Pyk2 and ERK1/2. Biochim Biophys Acta Gen Subj 2013; 1830:4650-9. [PMID: 23711511 DOI: 10.1016/j.bbagen.2013.05.023] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 05/11/2013] [Accepted: 05/15/2013] [Indexed: 12/20/2022]
Abstract
BACKGROUND Activation of ATP-gated P2X7 receptors (P2X7R) in macrophages leads to production of reactive oxygen species (ROS) by a mechanism that is partially characterized. Here we used J774 cells to identify the signaling cascade that couples ROS production to receptor stimulation. METHODS J774 cells and mP2X7-transfected HEK293 cells were stimulated with Bz-ATP in the presence and absence of extracellular calcium. Protein inhibitors were used to evaluate the physiological role of various kinases in ROS production. In addition, phospho-antibodies against ERK1/2 and Pyk2 were used to determine activation of these two kinases. RESULTS ROS generation in either J774 or HEK293 cells (expressing P2X7, NOX2, Rac1, p47phox and p67phox) was strictly dependent on calcium entry via P2X7R. Stimulation of P2X7R activated Pyk2 but not calmodulin. Inhibitors of MEK1/2 and c-Src abolished ERK1/2 activation and ROS production but inhibitors of PI3K and p38 MAPK had no effect on ROS generation. PKC inhibitors abolished ERK1/2 activation but barely reduced the amount of ROS produced by Bz-ATP. In agreement, the amount of ROS produced by PMA was about half of that produced by Bz-ATP. CONCLUSIONS Purinergic stimulation resulted in calcium entry via P2X7R and subsequent activation of the PKC/c-Src/Pyk2/ERK1/2 pathway to produce ROS. This signaling mechanism did not require PI3K, p38 MAPK or calmodulin. GENERAL SIGNIFICANCE ROS is generated in order to kill invading pathogens, thus elucidating the mechanism of ROS production in macrophages and other immune cells allow us to understand how our body copes with microbial infections.
Collapse
|
18
|
Caverzasio J, Biver E, Thouverey C. Predominant role of PDGF receptor transactivation in Wnt3a-induced osteoblastic cell proliferation. J Bone Miner Res 2013; 28:260-70. [PMID: 22927028 DOI: 10.1002/jbmr.1748] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 08/02/2012] [Accepted: 08/20/2012] [Indexed: 12/17/2022]
Abstract
Previous studies have shown that Wnt3a enhances the proliferation and inhibits the osteogenic differentiation of human mesenchymal stem cells (hMSCs). In this study, we investigated the signaling pathways involved in Wnt3a-induced osteoblastic cell proliferation. Experiments with DKK1, a natural antagonist of Lrp5/6, indicated that Wnt/β-catenin did not play a major role in Wnt3a-induced osteoblastic cell proliferation. The use of selective inhibitors of known mitogenic pathways implicates Src family kinases (SFKs) and a protein kinase C (PKC) in this cellular response. Time-dependent analysis of signaling molecules activated by Wnt3a in MC3T3-E1 cells revealed parallel activation of the canonical pathway and of several tyrosine kinases, including SFKs and PDGF receptors (PDGF-Rs). Functional analysis with specific inhibitors suggested a major role of PDGF-Rs in mediating Wnt3a-induced cell proliferation. Further investigation with an si-RNA approach confirmed a predominant role of this receptor in this cellular response. The use of soluble decoy PDGF-Rs that can sequester extracellular PDGFs excluding that part of the increased PDGF receptor phosphorylation by Wnt3a was the result of autocrine production of PDGFs. A selective SFK inhibitor blunted the enhanced PDGF-R phosphorylation and cell proliferation induced by Wnt3a. Studies of initial events involved in the regulation of this pathway suggest a role of dishevelled. In conclusion, data presented in this study indicate that cell proliferation induced by Wnt3a in osteoblastic cells is mediated by a dishevelled-dependent and β-catenin-independent pathway, which involves the transactivation of PDGF receptors.
Collapse
Affiliation(s)
- Joseph Caverzasio
- Service of Bone Diseases, Department of Internal Medicine Specialties, University Hospital of Geneva, Geneva, Switzerland.
| | | | | |
Collapse
|
19
|
Reduction of TRPV1 expression in the trigeminal system by botulinum neurotoxin type-A. Neurobiol Dis 2012; 48:367-78. [DOI: 10.1016/j.nbd.2012.07.010] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 06/30/2012] [Accepted: 07/09/2012] [Indexed: 01/30/2023] Open
|
20
|
Zhou Q, Li G, Deng XY, He XB, Chen LJ, Wu C, Shi Y, Wu KP, Mei LJ, Lu JX, Zhou NM. Activated human hydroxy-carboxylic acid receptor-3 signals to MAP kinase cascades via the PLC-dependent PKC and MMP-mediated EGFR pathways. Br J Pharmacol 2012; 166:1756-73. [PMID: 22289163 DOI: 10.1111/j.1476-5381.2012.01875.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE 3-Hydroxy-octanoate, recently identified as a ligand for, the orphan GPCR, HCA(3), is of particular interest given its ability to treat lipid disorders and atherosclerosis. Here we demonstrate the pathway of HCA(3)-mediated activation of ERK1/2. EXPERIMENTAL APPROACH Using CHO-K1 cells stably expressing HCA(3) receptors and A431 cells, a human epidermoid cell line with high levels of endogenous expression of functional HCA(3) receptors, HCA(3)-mediated activation of ERK1/2 was measured by Western blot. KEY RESULTS HCA(3)-mediated activation of ERK1/2 was rapid, peaking at 5 min, and was Pertussis toxin sensitive. Our data, obtained by time course analyses in combination with different kinase inhibitors, demonstrated that on agonist stimulation, HCA(3) receptors evoked ERK1/2 activation via two distinct pathways, the PLC/PKC pathway at early time points (≤ 2 min) and the MMP/ epidermal growth factor receptor (EGFR) transactivation pathway with a maximum response at 5 min. Furthermore, our present results also indicated that the βγ-subunits of the G(i) protein play a critical role in HCA(3)-activated ERK1/2 phosphorylation, whereas β-arrestins and Src were not required for ERK1/2 activation. CONCLUSIONS AND IMPLICATIONS We have described the molecular mechanisms underlying the coupling of human HCA(3) receptors to the ERK1/2 MAP kinase pathway in CHO-K1 and A431 cells, which implicate the G(i) protein-initiated, PLC/PKC -and platelet-derived growth factor receptor/EGFR transactivation-dependent pathways. These observations may provide new insights into the pharmacological effects and the physiological functions modulated by the HCA(3)-mediated activation of ERK1/2.
Collapse
Affiliation(s)
- Q Zhou
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Science, Wenzhou Medical College, Wenzhou, Zhejiang, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Meents JE, Neeb L, Reuter U. TRPV1 in migraine pathophysiology. Trends Mol Med 2010; 16:153-9. [DOI: 10.1016/j.molmed.2010.02.004] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2009] [Revised: 02/19/2010] [Accepted: 02/19/2010] [Indexed: 11/15/2022]
|
22
|
Fan C, Katsuyama M, Wei H, Xia Q, Liu W, Yabe-Nishimura C. Molecular mechanisms underlying PGF2alpha-induced hypertrophy of vascular smooth muscle cells. YAKUGAKU ZASSHI 2010; 130:211-4. [PMID: 20118645 DOI: 10.1248/yakushi.130.211] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The present review focuses primarily on the studies we made in recent years to improve the understanding of the molecular mechanisms of PGF2alpha-induced hypertrophy of Vascular Smooth Muscle Cells (VSMC). In this review, we will summarize the recent findings in the context of the PGF2alpha signaling pathway in three parts: PGF2alpha binding to its receptor, transactivation of EGF receptor, two independent signaling transduction pathways increasing the expression of NOX1 gene.
Collapse
Affiliation(s)
- Chunyuan Fan
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China.
| | | | | | | | | | | |
Collapse
|
23
|
Arellano-Plancarte A, Hernandez-Aranda J, Catt KJ, Olivares-Reyes JA. Angiotensin-induced EGF receptor transactivation inhibits insulin signaling in C9 hepatic cells. Biochem Pharmacol 2009; 79:733-45. [PMID: 19879250 DOI: 10.1016/j.bcp.2009.10.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2009] [Revised: 10/17/2009] [Accepted: 10/19/2009] [Indexed: 01/28/2023]
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.
Collapse
Affiliation(s)
- Araceli Arellano-Plancarte
- Department of Biochemistry, Center for Research and Advanced Studies of the National Polytechnic Institute, Cinvestav-IPN, A.P. 14-740, Mexico, 07360 D.F., Mexico
| | | | | | | |
Collapse
|
24
|
Resende RR, Adhikari A. Cholinergic receptor pathways involved in apoptosis, cell proliferation and neuronal differentiation. Cell Commun Signal 2009; 7:20. [PMID: 19712465 PMCID: PMC2744676 DOI: 10.1186/1478-811x-7-20] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2009] [Accepted: 08/27/2009] [Indexed: 11/14/2022] Open
Abstract
Acetylcholine (ACh) has been shown to modulate neuronal differentiation during early development. Both muscarinic and nicotinic acetylcholine receptors (AChRs) regulate a wide variety of physiological responses, including apoptosis, cellular proliferation and neuronal differentiation. However, the intracellular mechanisms underlying these effects of AChR signaling are not fully understood. It is known that activation of AChRs increase cellular proliferation and neurogenesis and that regulation of intracellular calcium through AChRs may underlie the many functions of ACh. Intriguingly, activation of diverse signaling molecules such as Ras-mitogen-activated protein kinase, phosphatidylinositol 3-kinase-Akt, protein kinase C and c-Src is modulated by AChRs. Here we discuss the roles of ACh in neuronal differentiation, cell proliferation and apoptosis. We also discuss the pathways involved in these processes, as well as the effects of novel endogenous AChRs agonists and strategies to enhance neuronal-differentiation of stem and neural progenitor cells. Further understanding of the intracellular mechanisms underlying AChR signaling may provide insights for novel therapeutic strategies, as abnormal AChR activity is present in many diseases.
Collapse
Affiliation(s)
- Rodrigo R Resende
- Department of Physics, Institute of Exact Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil.
| | | |
Collapse
|
25
|
Olivares-Reyes JA, Arellano-Plancarte A, Castillo-Hernandez JR. Angiotensin II and the development of insulin resistance: implications for diabetes. Mol Cell Endocrinol 2009; 302:128-39. [PMID: 19150387 DOI: 10.1016/j.mce.2008.12.011] [Citation(s) in RCA: 141] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2008] [Revised: 12/12/2008] [Accepted: 12/13/2008] [Indexed: 12/31/2022]
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.
Collapse
Affiliation(s)
- J Alberto Olivares-Reyes
- Department of Biochemistry, Center for Research and Advanced Studies of the National Polytechnic Institute, CINVESTAV-IPN, Mexico, DF, Mexico.
| | | | | |
Collapse
|
26
|
Yin X, Li B, Chen H, Catt KJ. Differential signaling pathways in angiotensin II- and epidermal growth factor-stimulated hepatic C9 cells. Mol Pharmacol 2008; 74:1223-33. [PMID: 18687808 DOI: 10.1124/mol.108.048504] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Caveolin1 (Cav1) is an important component of the plasmamembrane microdomains, such as caveolae/lipid rafts, that are associated with angiotensin II type 1 (AT(1)) and epidermal growth factor (EGF) receptors in certain cell types. The interactions of Cav1 with other signaling molecules that mediate AT(1) receptor function were analyzed in angiotensin II (Ang II)- and EGF-stimulated hepatic C9 cells. This study demonstrated that cholesterol-rich domains mediate the actions of early upstream signaling molecules such as Src and intracellular Ca(2+) in cells stimulated by Ang II, but not by EGF, and that Cav1 has a scaffolding role in the process of mitogen-activated protein kinase activation. Furthermore, Cav1 phosphorylation by Ang II and EGF was regulated by intracellular Ca(2+) and Src, further indicating reciprocal interactions among Cav1, Src, and intracellular Ca(2+) through the AT(1) receptor. Phosphorylation of Cav1 and the EGF receptor by Ang II, but not of extracellular signal-regulated kinase 1/2, was dependent on intracellular Ca(2+). The phosphatidylinositol 3-kinase inhibitors, 2-(4-morpholinyl)-8-phenyl-1(4H)-benzopyran-4-one hydrochloride (LY294002) and wortmannin, differentially modulated both Cav1 and EGF receptor activation by Ang II through intracellular Ca(2+). These findings further demonstrate the importance of Cav1 in conjunction with the receptor-mediated signaling pathways involved in cell proliferation and survival. It is clear that differential signaling pathways are operative in Ang II- and EGF-stimulated C9 cells and that cholesterol-enriched microdomains are essential components in cellular signaling processes that are dependent on specific agonists and/or cell types.
Collapse
Affiliation(s)
- Xing Yin
- Endocrinology and Reproduction Research Branch, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
27
|
Schauwienold D, Sastre AP, Genzel N, Schaefer M, Reusch HP. The transactivated epidermal growth factor receptor recruits Pyk2 to regulate Src kinase activity. J Biol Chem 2008; 283:27748-27756. [PMID: 18667434 DOI: 10.1074/jbc.m801431200] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
G protein-coupled receptors such as proteinase-activated receptor 1 induce phosphorylation of mitogen-activated protein kinases through multiple pathways including transactivation of receptor tyrosine kinases. In vascular smooth muscle cells, both matrix-metalloproteinase-dependent extracellular shedding of membrane-bound epidermal growth factor (EGF) receptor ligands and activation of the nonreceptor tyrosine kinases Pyk2 and Src contributed to the thrombin-induced ERK1/2 phosphorylation. Surprisingly, disruption of the HB-EGF-mediated extracellular mode of EGF receptor transactivation also prevented the phosphorylation of the nonreceptor tyrosine kinases Pyk2 and Src, locating these kinases downstream of the transactivated EGF receptor. The ionomycin-induced Pyk2 phosphorylation was partially sensitive to AG1478, heparin, or the matrix-metalloproteinase inhibitor BB2116, and the ionomycin-induced EGF receptor phosphorylation was almost completely blocked by these inhibitors of extracellular transactivation. Coimmunoprecipitation experiments revealed that, upon thrombin stimulation, a signaling complex consisting of Pyk2 and Src assembles at the EGF receptor. Reconstitution of the signaling molecules in HEK293 or vascular smooth muscle cells and subsequent determination of the EGF-induced Src kinase activity applying fluorescent sensor proteins demonstrated that a Ca(2+)-independent mode of Pyk2 activation is critical for the activation of Src downstream of the EGF receptor.
Collapse
Affiliation(s)
- Dag Schauwienold
- Abteilung Klinische Pharmakologie, Ruhr-Universität Bochum, 44780 Bochum, Germany
| | - Alejandra Pérez Sastre
- Neurowissenschaftliches Forschungszentrum, Molecular Pharmacology and Cell Biology, Charité-Universitätsmedizin Berlin, 14195 Berlin, Germany
| | - Nadine Genzel
- Neurowissenschaftliches Forschungszentrum, Molecular Pharmacology and Cell Biology, Charité-Universitätsmedizin Berlin, 14195 Berlin, Germany; ImaGenes GmbH, 13125 Berlin, Germany
| | - Michael Schaefer
- Neurowissenschaftliches Forschungszentrum, Molecular Pharmacology and Cell Biology, Charité-Universitätsmedizin Berlin, 14195 Berlin, Germany.
| | - H Peter Reusch
- Abteilung Klinische Pharmakologie, Ruhr-Universität Bochum, 44780 Bochum, Germany
| |
Collapse
|
28
|
Bierhals K, Sondersorg AC, Lin CT, Rosenbaum C, Waldmann H, Wehner F. The ε-Isoform of PKC Mediates the Hypertonic Activation of Cation Channels in Confluent Monolayers of Rat Hepatocytes. Cell Physiol Biochem 2008; 20:397-404. [PMID: 17762167 DOI: 10.1159/000107524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2007] [Indexed: 01/29/2023] Open
Abstract
We were interested whether PKC alpha, delta, epsilon or zeta is the isoform actually employed in the activation of hypertonicity-induced cation channels (HICCs) in primary cultures of rat hepatocytes. Quantitative SDS-page and Western-blot experiments revealed that PKC alpha, delta and epsilon were stimulated by Indolactam V (as a DAG substitute for activation of c and nPKCs) but that only PKC delta and epsilon did respond to hypertonic stress. Furthermore, chelation of intracellular Ca(++) by BAPTA-AM did not alter HICC activation in cable-analysis experiments whereas Indolactam V as well as V8 (an Indolactam derivative specific for PKC delta and epsilon) activated HICC currents under isotonic conditions. Finally, by use of Rottlerin (as an inhibitor exhibiting a slight preference for PKC delta over epsilon) PKC epsilon could be identified as the most likely isoform responsible for the activation of the HICC.
Collapse
Affiliation(s)
- Katrin Bierhals
- Department of Systemic Cell Biology, Max-Planck-Institute for Molecular Physiology, Dortmund, Germany
| | | | | | | | | | | |
Collapse
|
29
|
Sataranatarajan K, Lee MJ, Mariappan MM, Feliers D. PKCdelta regulates the stimulation of vascular endothelial factor mRNA translation by angiotensin II through hnRNP K. Cell Signal 2008; 20:969-77. [PMID: 18295448 DOI: 10.1016/j.cellsig.2008.01.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2007] [Revised: 01/16/2008] [Accepted: 01/17/2008] [Indexed: 01/13/2023]
Abstract
Angiotensin II (Ang II)-induced renal injury is partly mediated by growth factors such as VEGF. We have previously shown that Ang II rapidly increases VEGF protein synthesis in proximal tubular epithelial (MCT) cells by augmenting mRNA translation, which is partly dependent on activation and binding of hnRNP K to 3' untranslated region (UTR) of VEGF mRNA. Regulation of hnRNP K activation by PKCdelta was studied in MCT cells. Transfection with a PKCdelta siRNA inhibited hnRNP K Ser302 phosphorylation and activation, and reduced Ang II stimulation of VEGF synthesis. Inhibition of PKCdelta with röttlerin also prevented binding of hnRNP K to VEGF mRNA and reduced the efficiency of VEGF mRNA translation. In db/db mice at 2 weeks of type 2 diabetes, VEGF expression was increased, which was due not to increase in transcription but to augmented translation of VEGF mRNA. Augmented VEGF expression was associated with increased binding of hnRNP K to VEGF mRNA. c-src and PKCdelta activities and hnRNP K phosphorylation on Ser302 in renal cortex of db/db mice were increased compared to control mice. We conclude: Ang II-induced VEGF mRNA translation is associated with activation of hnRNP K in MCT cells. In the signaling pathway leading to hnRNP K activation induced by Ang II, PKCdelta is downstream of c-src. PKCdelta-mediated phosphorylation of hnRNP K is required for Ang II stimulation of VEGF mRNA translation. In mice with type 2 diabetes, src and PKCdelta activation and hnRNP K phosphorylation correlate with increased VEGF mRNA translation and kidney hypertrophy. 3' UTR events are important in regulation of VEGF expression in models of renal injury.
Collapse
Affiliation(s)
- Kavithalakshmi Sataranatarajan
- O'Brien Kidney Research Center, Department of Medicine/Nephrology, University of Texas Health Science Center, San Antonio, Texas, United States
| | | | | | | |
Collapse
|
30
|
Escano CS, Keever LB, Gutweiler AA, Andresen BT. Angiotensin II Activates Extracellular Signal-Regulated Kinase Independently of Receptor Tyrosine Kinases in Renal Smooth Muscle Cells: Implications for Blood Pressure Regulation. J Pharmacol Exp Ther 2007; 324:34-42. [PMID: 17911376 DOI: 10.1124/jpet.107.126300] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Angiotensin II can cause hypertension through enhanced vasoconstriction of renal vasculature. One proposed mechanism for reduction of angiotensin II-induced hypertension is through inhibition of the mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) mitogen-activated protein kinase cascade. MEK/ERK has been shown to phosphorylate the regulatory subunit of myosin light chain at identical positions as myosin light chain kinase. There are multiple mechanisms proposed regarding angiotensin II-mediated ERK activation. We hypothesized that renal microvascular smooth muscle cells (RmuVSMCs) signal through a unique pathway compared with thoracic aorta smooth muscle cells (TASMCs), which is involved in blood pressure regulation. Use of epidermal growth factor (EGF) and platelet derived growth factor (PDGF) receptor-specific inhibitors 4-(3-chloroanilino)-6,7-dimethoxyquinazoline (AG1478) and 6,7-dimethoxy-3-phenylquinoxaline (AG1296), respectively, demonstrates that angiotensin II activates ERK in TASMCs, but not RmuVSMCs, through transactivation of EGF and PDGF receptors. In addition, inhibition of Src with its specific inhibitor 4-amino-5-(4-chlorophenyl)-7-(t-butyl) pyrazolo[3,4-d]pyrimidine (PP2) abolishes angiotensin II-, but not EGF-or PDGF-, mediated phosphorylation of ERK in RmuVSMCs, yet it has no effect in TASMCs. The physiological significance of transactivation was examined in vivo using anesthetized Wistar-Kyoto rats with 15 mg/kg 2'-amino-3'-methoxyflavone (PD98059), an MEK inhibitor, as well as 20 mg/kg AG1478 and 1.5 mg/kg AG1296 in an acute model of angiotensin II-mediated increase in blood pressure. None of the inhibitors had an effect on basal blood pressure, and only PD98059 reduced angiotensin II-mediated increase in blood pressure. Moreover, in RmuVSMCs, but not TASMCs, angiotensin II localizes phosphorylated ERK to actin filaments. In conclusion, angiotensin II signals through a unique mechanism in the renal vascular bed that may contribute to hypertension.
Collapse
Affiliation(s)
- Crisanto S Escano
- Internal Medicine/Endocrinology, D109 HSC Diabetes Center, One Hospital Dr., Columbia, MO 65212, USA
| | | | | | | |
Collapse
|
31
|
Montiel M, Quesada J, Jiménez E. Activation of calcium-dependent kinases and epidermal growth factor receptor regulate muscarinic acetylcholine receptor-mediated MAPK/ERK activation in thyroid epithelial cells. Cell Signal 2007; 19:2138-46. [PMID: 17643958 DOI: 10.1016/j.cellsig.2007.06.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2007] [Accepted: 06/15/2007] [Indexed: 10/23/2022]
Abstract
We previously showed that stimulation of muscarinic acetylcholine receptors (mAChR) by carbachol (Cch) caused a time- and dose-dependent increase of mitogen-activated protein kinase/extracellular signal-regulated kinases (MAPK/ERK) phosphorylation in thyroid epithelial cells. In this study, we demonstrated that mAChR stimulation also induced a time-dependent increase in the tyrosine phosphorylation of proline-rich tyrosine kinase 2 (Pyk2), which was prevented by pretreatment of thyroid epithelial cells with the specific Src-family tyrosine kinase inhibitor PP2. Besides, phosphorylation of Pyk2 was attenuated by chelation of extracellular Ca(2+) or inhibition of phospholipase C (PLC), and was evoked by thapsigargin, a specific microsomal Ca(2+)-ATPase inhibitor. Incorporation of Pyk2 antisense oligonucleotides in thyroid epithelial cells to down-regulated Pyk2 expression or pretreatment of cells with the Ca(2+)/calmodulin protein kinase II (CaM kinase II) inhibitor KN-62 significantly reduced Cch-induced MAPK/ERK phosphorylation. In addition, Cch-induced MAPK/ERK phosphorylation was partially inhibited by LY294002 and wortmannin, two selective inhibitors of phosphatidylinositol 3-kinase (PI3K), tyrphostin AG1478, a specific inhibitor of epidermal growth factor receptor (EGFR) kinase, and (-)-perillic acid, a post-translational inhibitor of small G-proteins isoprenylation. Taken together, our data suggest that Pyk2, CaM kinase II and Src-family tyrosine kinases are key molecules for the activation of MAPK/ERK cascade through the EGFR/Ras/Raf pathway in thyroid epithelial cells in response to mAChR stimulation.
Collapse
Affiliation(s)
- Mercedes Montiel
- Department of Biochemistry and Molecular Biology, University of Malaga, Boulevard Louis Pasteur 32, 29071 Malaga, Spain
| | | | | |
Collapse
|
32
|
Azar ZM, Mehdi MZ, Srivastava AK. Activation of insulin-like growth factor type-1 receptor is required for H2O2-induced PKB phosphorylation in vascular smooth muscle cells. Can J Physiol Pharmacol 2007; 84:777-86. [PMID: 16998541 DOI: 10.1139/y06-024] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Evidence accumulated in recent years has revealed a potential role for reactive oxygen species (ROS) in the pathophysiology of cardiovascular diseases. However, the precise mechanisms by which ROS contribute to the development of these diseases are not fully established. Previous work from our laboratory has indicated that exogenous hydrogen peroxide (H2O2) activates several signaling protein kinases, such as extracellular signal-regulated kinase 1 and 2 (ERK1/2) and protein kinase B (PKB) in A10 vascular smooth muscle cells (VSMC). However, the upstream elements responsible for this activation remain unclear. Although a role for epidermal growth factor receptor (EGFR) protein tyrosine kinase (PTK) in H2O2-induced ERK1/2 signaling has been suggested, the contribution of this PTK or other receptor or nonreceptor PTKs to PKB activation is not well defined in VSMC. In this study, we used pharmacological inhibitors to investigate the role of receptor and Src-family-PTKs in H2O2-induced PKB phosphorylation. AG1478, a specific inhibitor of EGFR, failed to attenuate the H2O2-induced increase in PKB Ser473 phosphorylation, whereas AG1024, an inhibitor of insulin-like growth factor type1 receptor (IGF-1R)-PTK, almost completely blocked this response. H2O2 treatment also enhanced tyrosine phosphorylation of the IGF-1Rbeta subunit, which was significantly inhibited by AG1024 pretreatment of cells. Furthermore, pharmacological inhibition of Src by PP2 (4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazole(3,4-d) pyrimidine) decreased PKB phosphorylation. Moreover, H2O2-induced PKB phosphorylation was associated with increased tyrosine phosphorylation of c-Src and Pyk2 in an AG1024- and PP2-inhibitable manner. In conclusion, these data provide evidence of the contribution of IGF-1R-PTK in initiating H2O2-evoked PKB phosphorylation in A10 VSMC, with an intermediary role for c-Src and Pyk2 in this process.
Collapse
Affiliation(s)
- Zeina M Azar
- Laboratory of Cell Signaling, Research Centre, Centre hospitalier de l'Université de Montréal (CHUM) - Hôtel-Dieu and Department of Medicine, Université de Montréal, 3850, St. Urbain Street, Rm. 7-135, Montreal, QC H2W 1T7, Canada
| | | | | |
Collapse
|
33
|
Szidonya L, Süpeki K, Karip E, Turu G, Várnai P, Clark AJL, Hunyady L. AT1 receptor blocker-insensitive mutant AT1A angiotensin receptors reveal the presence of G protein-independent signaling in C9 cells. Biochem Pharmacol 2007; 73:1582-92. [PMID: 17284329 DOI: 10.1016/j.bcp.2007.01.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2006] [Revised: 12/28/2006] [Accepted: 01/03/2007] [Indexed: 01/01/2023]
Abstract
Although mutant receptors are highly useful to dissect the signal transduction pathways of receptors, they are difficult to study in physiological target tissues, due to the presence of endogenous receptors. To study AT(1) angiotensin receptors in their physiological environment, we constructed a mutant receptor, which differs only from the AT(1A) receptor in its reduced affinity for candesartan, a biphenylimidazole antagonist. We have determined that the conserved S109Y substitution of the rat AT(1A) receptor eliminates its candesartan binding, without exerting any major effect on its angiotensin II and peptide angiotensin receptor antagonist binding, internalization kinetics, beta-arrestin binding, and potency or efficacy of the inositol phosphate response. To demonstrate the usefulness of this mutant receptor in signal transduction studies, we combined it with substitution of the highly conserved DRY sequence with AAY, which abolishes G protein activation. In rat C9 hepatocytes the S109Y receptor caused ERK activation with the same mechanism as the endogenous AT(1) receptor. After combination with the DRY/AAY mutation G protein-independent ERK activation was detected demonstrating that this approach can be used to study the angiotensin II-stimulated signaling pathways in cells endogenously expressing AT(1) receptors.
Collapse
Affiliation(s)
- László Szidonya
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | | | | | | | | | | | | |
Collapse
|
34
|
Schindler EM, Baumgartner M, Gribben EM, Li L, Efimova T. The role of proline-rich protein tyrosine kinase 2 in differentiation-dependent signaling in human epidermal keratinocytes. J Invest Dermatol 2007; 127:1094-106. [PMID: 17205062 DOI: 10.1038/sj.jid.5700662] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Non-receptor tyrosine kinase proline-rich protein tyrosine kinase 2 (Pyk2) functions as an integrator of multiple signaling pathways involved in the regulation of fundamental cellular processes. Pyk2 expression, regulation, and functions in skin have not been examined. Here we investigated the expression and subcellular localization of Pyk2 in human epidermis and in primary human keratinocytes, and studied the mechanisms of Pyk2 activation by differentiation-inducing stimuli, and the role of Pyk2 as a regulator of keratinocyte differentiation. We demonstrate that Pyk2 is abundantly expressed in skin keratinocytes. Notably, the endogenous Pyk2 protein is predominantly localized in keratinocyte nuclei throughout all layers of healthy human epidermis, and in cultured human keratinocytes. Pyk2 is activated by treatment with keratinocyte-differentiating agents, 12-O-tetradecanoylphorbol-13-acetate and calcium via a mechanism that requires intracellular calcium release and functional protein kinase C (PKC) and Src activities. Particularly, differentiation-promoting PKC delta and PKC eta elicit Pyk2 activation. Our data show that Pyk2 increases promoter activity and endogenous protein levels of involucrin, a marker of keratinocyte terminal differentiation. This regulation is associated with increased expression of Fra-1 and JunD, activator protein-1 transcription factors known to be required for involucrin expression. Altogether, these results provide insights into Pyk2 signaling in epidermis and reveal a novel role for Pyk2 in regulation of keratinocyte differentiation.
Collapse
Affiliation(s)
- Eva M Schindler
- Division of Dermatology, Department of Internal Medicine, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | | | | | | | | |
Collapse
|
35
|
Zhou CH, Qian ZY, Xiang M, He SY. Involvement of Ca2+ in the inhibition by crocetin of angiotensin II-induced ERK1/2 activation in vascular smooth muscle cells. Eur J Pharmacol 2007; 554:85-91. [PMID: 17109849 DOI: 10.1016/j.ejphar.2006.09.069] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2006] [Revised: 09/23/2006] [Accepted: 09/27/2006] [Indexed: 11/15/2022]
Abstract
Crocetin, a carotenoid compound, was isolated from Gardenia jasminoids Ellis. Our recent study shows that crocetin inhibits angiotensin II-induced extracellular signal-regulated kinases 1/2 (ERK1/2) activation and subsequent proliferation in vascular smooth muscle cells (VSMCs). To further explore the mechanism involved, in the present study, we investigated the effect of Ca(2+) in the activation of ERK1/2 and whether Ca(2+) is involved in the suppression by crocetin of angiotensin II-induced ERK1/2 activation. Our findings showed that crocetin pretreatment partially attenuated both the intracellular Ca(2+) mobilization and the extracellular Ca(2+) influx induced by angiotensin II. Moreover, angiotensin II-induced ERK1/2 activation was completely abolished by acetoxymethyl ester of 1,2-bis(2-aminophenoxy)ethane-N,N,N ',N'-tetraacetic acid (BAPTA-AM), an intracellular Ca(2+) chelator, and partially inhibited by EGTA, an extracellular Ca(2+) chelator, or verapamil, an L-type Ca(2+) channel blocker. These findings suggest that Ca(2+) may play an important role in angiotensin II-induced ERK1/2 activation in VSMCs, and Ca(2+)-dependent pathway may be involved in the inhibitory effect by crocetin of angiotensin II-induced ERK1/2 activation.
Collapse
MESH Headings
- Angiotensin II/pharmacology
- Animals
- Animals, Newborn
- Calcium/metabolism
- Calcium/pharmacokinetics
- Calcium/physiology
- Calcium Channel Blockers/pharmacology
- Carotenoids/pharmacology
- Cattle
- Cells, Cultured
- Chelating Agents/pharmacology
- Dose-Response Relationship, Drug
- Egtazic Acid/pharmacology
- Enzyme Activation/drug effects
- Intracellular Fluid/drug effects
- Intracellular Fluid/metabolism
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Time Factors
- Verapamil/pharmacology
- Vitamin A/analogs & derivatives
Collapse
Affiliation(s)
- Cheng-Hua Zhou
- Department of Pharmacology, China Pharmaceutical University, Tongjia Xiang 24, Nanjing 210009, PR China
| | | | | | | |
Collapse
|
36
|
Han HJ, Han JY, Heo JS, Lee SH, Lee MY, Kim YH. ANG II-stimulated DNA synthesis is mediated by ANG II receptor-dependent Ca2+/PKC as well as EGF receptor-dependent PI3K/Akt/mTOR/p70S6K1 signal pathways in mouse embryonic stem cells. J Cell Physiol 2007; 211:618-29. [PMID: 17219409 DOI: 10.1002/jcp.20967] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Effect of angiotensin II (ANG II) on mouse embryonic stem (ES) cell proliferation was examined. ANG II increased [(3)H] thymidine incorporation in a time- (>4 h) and dose- (>10(-9) M) dependent manner. The ANG II-induced increase in [(3)H] thymidine incorporation was blocked by inhibition of ANG II type 1 (AT(1)) receptor but not by ANG II type 2 (AT(2)) receptor, and AT(1) receptor was expressed. ANG II increased inositol phosphates formation and [Ca(2+)](i), and translocated PKC alpha, delta, and zeta to the membrane fraction. Consequently, the inhibition of PLC/PKC suppressed ANG II-induced increase in [(3)H] thymidine incorporation. The inhibition of EGF receptor kinase or tyrosine kinase prevented ANG II-induced increase in [(3)H] thymidine incorporation. ANG II phosphorylated EGF receptor and increased Akt, mTOR, and p70S6K1 phosphorylation blocked by AG 1478 (EGF receptor kinase blocker). ANG II-induced increase in [(3)H] thymidine incorporation was blocked by the inhibition of p44/42 MAPKs but not by p38 MAPK inhibition. Indeed, ANG II phosphorylated p44/42 MAPKs, which was prevented by the inhibition of the PKC and AT(1) receptor. ANG II increased c-fos, c-jun, and c-myc levels. ANG II also increased the protein levels of cyclin D1, cyclin E, cyclin-dependent kinase (CDK) 2, and CDK4 but decreased the p21(cip1/waf1) and p27(kip1), CDK inhibitory proteins. These proteins were blocked by the inhibition of AT(1) receptor, PLC/PKC, p44/42 MAPKs, EGF receptor, or tyrosine kinase. In conclusion, ANG II-stimulated DNA synthesis is mediated by ANG II receptor-dependent Ca(2+)/PKC and EGF receptor-dependent PI3K/Akt/mTOR/p70S6K1 signal pathways in mouse ES cells.
Collapse
Affiliation(s)
- Ho Jae Han
- Department of Veterinary Physiology, Biotherapy Human Resources Center, College of Veterinary Medicine, Chonnam National University, Gwangju, Korea.
| | | | | | | | | | | |
Collapse
|
37
|
Shah BH, Baukal AJ, Chen HD, Shah AB, Catt KJ. Mechanisms of endothelin-1-induced MAP kinase activation in adrenal glomerulosa cells. J Steroid Biochem Mol Biol 2006; 102:79-88. [PMID: 17113976 PMCID: PMC3196343 DOI: 10.1016/j.jsbmb.2006.09.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
G protein-coupled receptors (GPCRs) such as angiotensin II, bradykinin and endothelin-1 (ET-1) are critically involved in the regulation of adrenal function, including aldosterone production from zona glomerulosa cells. Whereas, substantial data are available on the signaling mechanisms of ET-1 in cardiovascular tissues, such information in adrenal glomerulosa cells is lacking. Bovine adrenal glomerulosa (BAG) cells express receptors for endothelin-1 (ET-1) and their stimulation caused phosphorylation of Src (at Tyr416), proline-rich tyrosine kinase (Pyk2 at Tyr402), extracellularly regulated signal kinases (ERK1/2), and their dependent proteins, p90 ribosomal S6 kinase (RSK-1) and CREB. ET-1 elicited these responses predominantly through activation of a G(i)-linked cascade with a minor contribution from the G(q)/PKC pathway. Whereas, selective inhibition of EGF-R kinase with AG1478 caused complete inhibition of EGF-induced ERK/RSK-1/CREB activation, it caused only partial reduction (30-40%) of such ET-1-induced responses. Consistent with this, inhibition of matrix metalloproteinases (MMPs) with GM6001 reduced ERK1/2 activation by ET-1, consistent with partial involvement of the MMP-dependent EGF-R activation in this cascade. Activation of ERK/RSK-1/CREB by both ET-1 and EGF was abolished by inhibition of Src, indicating its central role in ET-1 signaling in BAG cells. Moreover, the signaling characteristics of ET-1 in cultured BAG cells closely resembled those observed in clonal adrenocortical H295R cells. The ET-1-induced proliferation of BAG and H295 R cells was much smaller than that induced by Ang II or FGF. These data demonstrate that ET-1 causes ERK/RSK-1/CREB phosphorylation predominantly through activation of G(i) and Src, with a minor contribution from MMP-dependent EGF-R transactivation.
Collapse
Affiliation(s)
- Bukhtiar H Shah
- Endocrinology and Reproduction Research Branch, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
38
|
Alexander LD, Ding Y, Alagarsamy S, Cui XL, Douglas JG. Arachidonic acid induces ERK activation via Src SH2 domain association with the epidermal growth factor receptor. Kidney Int 2006; 69:1823-32. [PMID: 16598196 DOI: 10.1038/sj.ki.5000363] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Within the kidney, angiotensin II type 2 (AT(2)) receptor mediates phospholipase A(2) (PLA(2)) activation, arachidonic acid release, epidermal growth factor (EGF) receptor transactivation, and mitogen-activated protein kinase activation. Arachidonic acid mimics this transactivation by an undetermined mechanism. The role of c-Src in mediating angiotensin II and arachidonic acid signaling was determined by employing immunocomplex kinase assay, Western blotting analysis, and protein immunoblotting on co-precipitated EGF receptor (EGFR) proteins and agarose conjugates of glutathione S-transferase fusion proteins containing the c-Src homology 2 (SH2) and SH3 domains. Angiotensin II induced extracellular signal-regulated kinase (ERK) activation in primary cultures of rabbit proximal tubule cells via the activation of c-Src and association of the EGFR with the c-Src SH2 domain, effects that were mimicked by arachidonic acid and its inactive analogue eicosatetraynoic acid. Inhibition of PLA(2) by mepacrine and methyl arachidonyl fluorophosphate, AT(2) receptor by PD123319, Src family kinases by, 1-(tert-butyl)-3-(4-chlorophenyl)-4-aminopyrazolo[3,4-d] pyrimidine (PP2) and c-Src by overexpression of a dominant-negative mutant of c-Src abrogated these effects. However, inhibitors of arachidonic acid metabolic pathways did not block these effects. The present work provides a new and novel paradigm for transactivation of a kinase receptor linked to a fatty acid, which may apply to activation of a variety of phospholipases and accompanying arachidonic acid release.
Collapse
Affiliation(s)
- L D Alexander
- Department of Natural Sciences, The University of Michigan-Dearborn, 48128, USA.
| | | | | | | | | |
Collapse
|
39
|
Wu SS, Jácamo RO, Vong SK, Rozengurt E. Differential regulation of Pyk2 phosphorylation at Tyr-402 and Tyr-580 in intestinal epithelial cells: roles of calcium, Src, Rho kinase, and the cytoskeleton. Cell Signal 2006; 18:1932-40. [PMID: 16574377 DOI: 10.1016/j.cellsig.2006.02.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2006] [Revised: 02/21/2006] [Accepted: 02/22/2006] [Indexed: 10/25/2022]
Abstract
The calcium-dependent proline-rich tyrosine kinase Pyk2 is activated by tyrosine phosphorylation, associates with focal adhesion proteins, and has been linked to proliferative and migratory responses in a variety of mesenchymal and epithelial cell types. Full Pyk2 activation requires phosphorylation at functionally distinct sites, including autophosphorylation site Tyr-402 and catalytic domain site Tyr-580, though the mechanisms involved are unclear. The pathways mediating Pyk2 phosphorylation at Tyr-402 and Tyr-580 were therefore investigated. Both sites were rapidly and transiently phosphorylated following cell stimulation by Ang II or LPA. However, only Tyr-580 phosphorylation was rapidly enhanced by intracellular Ca(2+) release, or inhibited by Ca(2+) depletion. Conversely, Tyr-402 phosphorylation was highly sensitive to inhibition of actin stress fibers, or of Rho kinase (ROK), an upstream regulator of stress fiber assembly. Ang II also induced a delayed (30-60 min) secondary phosphorylation peak occurring at Tyr-402 alone. Unlike the homologous focal adhesion kinase (FAK), Pyk2 phosphorylation was sensitive neither to the Src inhibitor PP2, nor to truncation of its N-terminal region, which contains a putative autoinhibitory FERM domain. These results better define the mechanisms involved in Pyk2 activation, demonstrating that autophosphorylation is ROK- and stress fiber-dependent, while transphosphorylation within the kinase domain is Ca(2+)-dependent and Src-independent in intestinal epithelial cells. This contrasts with the tight sequential coupling of phosphorylation seen in FAK activation, and further underlines the differences between these closely related kinases.
Collapse
Affiliation(s)
- Steven S Wu
- Department of Pediatrics, David Geffen School of Medicine, University of California-Los Angeles, 900 Veteran Avenue, Los Angeles, CA 90095, USA.
| | | | | | | |
Collapse
|
40
|
Fan C, Katsuyama M, Yabe-Nishimura C. PKCdelta mediates up-regulation of NOX1, a catalytic subunit of NADPH oxidase, via transactivation of the EGF receptor: possible involvement of PKCdelta in vascular hypertrophy. Biochem J 2006; 390:761-7. [PMID: 15913451 PMCID: PMC1199669 DOI: 10.1042/bj20050287] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
NADPH oxidase is the major source of superoxide production in cardiovascular tissues. We reported previously that PG (prostaglandin) F2alpha caused hypertrophy of vascular smooth muscle cells by induction of NOX1, a catalytic subunit of NADPH oxidase. PGF2alpha-induced NOX1 expression was mediated by transactivation of the EGF (epidermal growth factor) receptor and subsequent activation of ERK (extracellular-signal-regulated kinase) 1/2, PI3K (phosphoinositide 3-kinase) and ATF-1 (activating transcription factor-1), a member of the CREB (cAMP-response-element-binding protein)/ATF family. As the receptor for PGF2alpha is known to activate PKC (protein kinase C), involvement of PKC in up-regulation of NOX1 expression was investigated in A7r5 cells. GF109203x, a non-selective inhibitor of PKC, dose-dependently suppressed the induction of NOX1 mRNA by PGF2alpha. Whereas an inhibitor of the conventional PKC, Gö 6976, and a PKCeta translocation-inhibitor peptide had no effect, an inhibitor of PKCdelta, rottlerin, significantly attenuated the PGF2alpha-induced increase in NOX1 mRNA. Gene silencing of PKCdelta by RNA interference significantly suppressed the PGF2alpha-induced increase in NOX1 mRNA, as well as phosphorylation of the EGF receptor, ERK1/2 and ATF-1. Silencing of the PKCdelta gene also attenuated the PDGF (platelet-derived growth factor)- induced increase in NOX1 mRNA and transactivation of the EGF receptor. Moreover, the augmented synthesis of the protein induced by PGF2alpha or PDGF was abolished by gene silencing of PKCdelta. These results suggest that PKCdelta-mediated transactivation of the EGF receptor is elicited not only by PGF2alpha, but also by PDGF, and that the subsequent activation of ERK1/2 and ATF-1 leads to up-regulation of NOX1 gene expression and ensuing hypertrophy in the vascular cell lineage.
Collapse
Affiliation(s)
- Chun Yuan Fan
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Masato Katsuyama
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Chihiro Yabe-Nishimura
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
- To whom correspondence should be addressed (email )
| |
Collapse
|
41
|
Neithardt A, Farshori MP, Shah FB, Catt KJ, Shah BH. Dependence of GnRH-induced phosphorylation of CREB and BAD on EGF receptor transactivation in GT1-7 neuronal cells. J Cell Physiol 2006; 208:586-93. [PMID: 16741954 DOI: 10.1002/jcp.20697] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The hypothalamic neuropeptide, gonadotropin releasing hormone (GnRH), is a primary regulatory factor in the neuroendocrine control of reproduction. The GnRH decapeptide is released in an episodic manner from hypothalamic GnRH neurons, which are known to express GnRH receptors. Here we examined the signaling pathways by which autocrine GnRH stimulation generates cell survival and proliferative signals in hypothalamic GT1-7 cells. Both GnRH and epidermal growth factor (EGF) caused rapid phosphorylation of cyclic AMP response element binding protein (CREB) and BAD. The selective epidermal growth factor receptor (EGF-R) antagonist, AG1478, attenuates the phosphorylation of these proteins by GnRH and EGF. Inhibition of PKC and Src abolished the stimulatory effects of GnRH, but not that of EGF, consistent with a critical role of these signaling molecules upstream of the EGF-R. All of these effects of GnRH were mimicked by phorbol 12 myristate 13-acetate (PMA). Consistent with the prosurvival and mitogenic effects of phosphoinositide 3-kinase/Akt (P13-K/Akt) downstream of the EGF-R, inhibition of P13-K diminished the activation of these proteins following stimulation with GnRH, EGF, and PMA. Overexpression of dominant negative Akt attenuated agonist-induced phosphorylation of BAD, but not that of ERK1/2 and CREB. Moreover, overexpression of wild-type RSK-1 resulted in enhanced basal as well as agonist-induced phosphorylation of CREB and BAD, indicating a critical role of RSK-1 in activating cytosolic as well as nuclear proteins. These data reveal novel signaling mechanisms of GnRH-induced phosphorylation of CREB and BAD in GT1-7 neurons through transactivation of the EGF-R.
Collapse
Affiliation(s)
- Adrienne Neithardt
- Section on Hormonal Regulation, Endocrinology and Reproduction Research Branch, NICHD, National Institutes of Health, Bethesda, Maryland 20892-4510, USA
| | | | | | | | | |
Collapse
|
42
|
Shah BH, Shah FB, Catt KJ. Role of metalloproteinase-dependent EGF receptor activation in α1-adrenoceptor-stimulated MAP kinase phosphorylation in GT1-7 neurons. J Neurochem 2006; 96:520-32. [PMID: 16336626 DOI: 10.1111/j.1471-4159.2005.03585.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Adrenoceptors (ARs) are involved in the regulation of gonadotropin-releasing hormone (GnRH) release from native and immortalized hypothalamic (GT1-7) neurons. However, the AR-mediated signaling mechanisms and their functional significance in these cells are not known. Stimulation of GT1-7 cells with the alpha1-AR agonist, phenylephrine (Phe), causes phosphorylation of extracellular signal-regulated kinases 1 and 2 (ERK1/2) mitogen-activated protein (MAP) kinases that is mediated by protein kinase C (PKC)-dependent transactivation of the epidermal growth factor receptor (EGF-R). Phe stimulation causes shedding of the soluble ligand, heparin-binding EGF (HB-EGF), as a consequence of matrix metalloproteinase (MMP) activation. Phe-induced phosphorylation of the EGF-R, and subsequently of Shc and ERK1/2, was attenuated by inhibition of MMP or HB-EGF with the selective inhibitor, CRM197, or by a neutralizing antibody. In contrast, phosphorylation of the EGF-R, Shc and ERK1/2 by EGF and HB-EGF was independent of PKC and MMP activity. Moreover, inhibition of Src attenuated ERK1/2 responses by Phe, but not by HB-EGF and EGF, indicating that Src acts upstream of the EGF-R. Consistent with a potential role of reactive oxygen species (ROS), Phe-induced phosphorylation of EGF-R was attenuated by the antioxidant, N-acetylcysteine. These data suggest that activation of the alpha1-AR causes phosphorylation of ERK1/2 through activation of PKC, ROS and Src, and shedding of HB-EGF, which binds to and activates the EGF-R.
Collapse
Affiliation(s)
- Bukhtiar H Shah
- Endocrinology and Reproduction Research Branch, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892-4510, USA.
| | | | | |
Collapse
|
43
|
Baker OJ, Camden JM, Ratchford AM, Seye CI, Erb L, Weisman GA. Differential coupling of the P2Y1 receptor to Galpha14 and Galphaq/11 proteins during the development of the rat salivary gland. Arch Oral Biol 2005; 51:359-70. [PMID: 16336941 DOI: 10.1016/j.archoralbio.2005.10.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2005] [Revised: 10/11/2005] [Accepted: 10/14/2005] [Indexed: 02/06/2023]
Abstract
UNLABELLED In rat submandibular gland (SMG), the P2Y1 receptor (P2Y1R) mediates increases in the intracellular calcium concentration, [Ca2+]i that diminish as the animal ages from 1 to 4-6 weeks. However, P2Y1R mRNA levels do not change with age, suggesting that the age-dependent decrease in the [Ca2+]i response to P2Y1R agonists may be due to alterations in the activity of a component of the P2Y1R signalling pathway. OBJECTIVES To assess whether the decrease in P2Y1R-mediated intracellular calcium signalling in SMG cells as rats age is due to a decrease in P2Y1R coupling to G proteins or to a decrease in the expression of a cognate G protein. DESIGN SMG cells were isolated from Sprague-Dawley rats. P2Y1R function was assessed by measuring 2-MeSADP-induced increases in [Ca2+]i and ERK1/2 activation. P2Y(1)R-mediated activation of G proteins was determined by the [35S]GTPgammaS binding assay. Gq protein expression was determined by RT-PCR, Northern, and Western analysis. RESULTS In SMG cells from 1-week-old rats, two bands (52 and 42kDa) were detected using anti-Galpha14 antibody, whereas in SMG cells from 4- to 6-week-old rats only the 42 kDa band was detected. Furthermore, 2-MeSADP-induced GTPgamma35S binding to Galpha14 and Galphaq/11 decreases in SMG cells from 4- to 6-week-old rats as compared to 1-week-old rats. CONCLUSIONS These findings suggest that the age-dependent decrease in P2Y1R-mediated intracellular calcium signalling in rat SMG cells is due to a loss of 52 kDa Galpha14 and indicate the differential coupling of the P2Y1R to Galpha14 and Galphaq/11 as the gland develops.
Collapse
Affiliation(s)
- Olga J Baker
- Department of Biochemistry, University of Missouri-Columbia, 540E Life Sciences Center, 1201 Rollins Road, Columbia, MO 65211-7310, USA
| | | | | | | | | | | |
Collapse
|
44
|
Zhang X, Huang J, McNaughton PA. NGF rapidly increases membrane expression of TRPV1 heat-gated ion channels. EMBO J 2005; 24:4211-23. [PMID: 16319926 PMCID: PMC1356334 DOI: 10.1038/sj.emboj.7600893] [Citation(s) in RCA: 552] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2005] [Accepted: 11/09/2005] [Indexed: 12/29/2022] Open
Abstract
Nociceptors, or pain-sensitive receptors, are unique among sensory receptors in that their sensitivity is increased by noxious stimulation. This process, called sensitization or hyperalgesia, is mediated by a variety of proinflammatory factors, including bradykinin, ATP and NGF, which cause sensitization to noxious heat stimuli by enhancing the membrane current carried by the heat- and capsaicin-gated ion channel, TRPV1. Several different mechanisms for sensitization of TRPV1 have been proposed. Here we show that NGF, acting on the TrkA receptor, activates a signalling pathway in which PI3 kinase plays a crucial early role, with Src kinase as the downstream element which binds to and phosphorylates TRPV1. Phosphorylation of TRPV1 at a single tyrosine residue, Y200, followed by insertion of TRPV1 channels into the surface membrane, explains most of the rapid sensitizing actions of NGF.
Collapse
Affiliation(s)
- Xuming Zhang
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Jiehong Huang
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Peter A McNaughton
- Department of Pharmacology, University of Cambridge, Cambridge, UK
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK. Tel.: +44 1223 334012; Fax: +44 1223 334040; E-mail:
| |
Collapse
|
45
|
Hunyady L, Catt KJ. Pleiotropic AT1 receptor signaling pathways mediating physiological and pathogenic actions of angiotensin II. Mol Endocrinol 2005; 20:953-70. [PMID: 16141358 DOI: 10.1210/me.2004-0536] [Citation(s) in RCA: 402] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Angiotensin II (Ang II) activates a wide spectrum of signaling responses via the AT1 receptor (AT1R) that mediate its physiological control of blood pressure, thirst, and sodium balance and its diverse pathological actions in cardiovascular, renal, and other cell types. Ang II-induced AT1R activation via Gq/11 stimulates phospholipases A2, C, and D, and activates inositol trisphosphate/Ca2+ signaling, protein kinase C isoforms, and MAPKs, as well as several tyrosine kinases (Pyk2, Src, Tyk2, FAK), scaffold proteins (G protein-coupled receptor kinase-interacting protein 1, p130Cas, paxillin, vinculin), receptor tyrosine kinases, and the nuclear factor-kappaB pathway. The AT1R also signals via Gi/o and G11/12 and stimulates G protein-independent signaling pathways, such as beta-arrestin-mediated MAPK activation and the Jak/STAT. Alterations in homo- or heterodimerization of the AT1R may also contribute to its pathophysiological roles. Many of the deleterious actions of AT1R activation are initiated by locally generated, rather than circulating, Ang II and are concomitant with the harmful effects of aldosterone in the cardiovascular system. AT1R-mediated overproduction of reactive oxygen species has potent growth-promoting, proinflammatory, and profibrotic actions by exerting positive feedback effects that amplify its signaling in cardiovascular cells, leukocytes, and monocytes. In addition to its roles in cardiovascular and renal disease, agonist-induced activation of the AT1R also participates in the development of metabolic diseases and promotes tumor progression and metastasis through its growth-promoting and proangiogenic activities. The recognition of Ang II's pathogenic actions is leading to novel clinical applications of angiotensin-converting enzyme inhibitors and AT1R antagonists, in addition to their established therapeutic actions in essential hypertension.
Collapse
Affiliation(s)
- László Hunyady
- Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | | |
Collapse
|
46
|
Gardner OS, Dewar BJ, Graves LM. Activation of Mitogen-Activated Protein Kinases by Peroxisome Proliferator-Activated Receptor Ligands: An Example of Nongenomic Signaling. Mol Pharmacol 2005; 68:933-41. [PMID: 16020742 DOI: 10.1124/mol.105.012260] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are a subfamily of nuclear hormone receptors that function as ligand-activated transcription factors to regulate lipid metabolism and homeostasis. In addition to their ability to promote gene transcription in a PPAR-dependent manner, ligands for this receptor family have recently been shown to induce mitogen-activated protein kinase (MAPK) phosphorylation. It is noteworthy that the transcriptional changes induced by PPAR ligands can be separated into distinct PPAR- and MAPK-dependent signaling pathways, suggesting that MAPKs alone mediate some of the effects of PPAR agonists in a nongenomic manner. This review will highlight recent studies that elucidate the nongenomic mechanisms of PPAR ligand-induced MAPK phosphorylation. The potential relevance of MAPK signaling in PPAR biology is also discussed.
Collapse
Affiliation(s)
- Olivia S Gardner
- Curriculum in Toxicology, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | |
Collapse
|
47
|
Dangi S, Shapiro P. Cdc2-mediated Inhibition of Epidermal Growth Factor Activation of the Extracellular Signal-regulated Kinase Pathway during Mitosis. J Biol Chem 2005; 280:24524-31. [PMID: 15888452 DOI: 10.1074/jbc.m414079200] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Inhibition of general transcription and translation occurs during mitosis to preserve the high energy requirements needed for the dynamic structural changes that are occurring at this time of the cell cycle. Although the mitotic kinase Cdc2 appears to directly phosphorylate and inhibit key proteins directly involved in transcription and translation, the role of Cdc2 in regulating up-stream growth factor receptor-mediated signal transduction pathways is limited. In the present study, we examined mechanisms involved in uncoupling receptor-mediated activation of the extracellular signal-regulated (ERK) signaling pathway in mitotic cells. Treatment with epidermal growth factor (EGF) failed to activate the ERK pathway in mitotic cells, although partial activation of ERK could be achieved in mitotic cells treated with phorbol 12-myristate 13-acetate (PMA). The discrepancy between EGF and PMA-mediated ERK activation suggested that multiple events in the ERK pathway were regulated during mitosis. We show that Cdc2 inhibits EGF-mediated ERK activation through direct interaction and phosphorylation of several ERK pathway proteins, including the guanine nucleotide exchange factor, Sos-1, and Raf-1 kinase. Inhibition of Cdc2 activity with roscovitine in mitotic cells restored ERK activation by EGF and PMA. Similarly, mitotic inhibition of ERK activity in cells expressing active mutants of H-Ras and Raf-1 kinase could also be reversed following Cdc2 inhibition. In contrast, ERK activation in cells expressing active MEK1 was not inhibited during mitosis or affected by roscovitine. These data suggest that Cdc2 inhibits growth factor receptor-mediated ERK activation during mitosis by primarily targeting signaling proteins that are upstream of MEK1.
Collapse
Affiliation(s)
- Surabhi Dangi
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, USA
| | | |
Collapse
|
48
|
Shah BH, Baukal AJ, Shah FB, Catt KJ. Mechanisms of extracellularly regulated kinases 1/2 activation in adrenal glomerulosa cells by lysophosphatidic acid and epidermal growth factor. Mol Endocrinol 2005; 19:2535-48. [PMID: 15928312 DOI: 10.1210/me.2005-0082] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The regulation of adrenal function, including aldosterone production from adrenal glomerulosa cells, is dependent on a variety of G protein-coupled receptors (GPCRs) and receptor tyrosine kinases (RTKs). In many cell types, GPCR-mediated MAPK activation is mediated through transactivation of RTKs, in particular the epidermal growth factor (EGF) receptor (EGF-R). However, the extent to which this cross-communication between GPCRs and RTKs is operative in the adrenal glomerulosa has not been defined. Bovine adrenal glomerulosa cells express receptors for lysophosphatidic acid (LPA) and EGF. In cultured bovine adrenal glomerulosa cells, LPA, which is predominantly coupled to Gi and partially to Gq/protein kinase C alpha and epsilon, caused phosphorylation of Src (at Tyr416), proline-rich tyrosine kinase (Pyk2 at Tyr402), EGF-R, protein kinase B/Akt, extracellularly regulated signal kinases 1/2, and their dependent protein, p90 ribosomal S6 kinase. Overexpression of dominant negative mutants of Ras or EGF-R, and selective inhibition of EGF-R kinase with AG1478, significantly reduced LPA-induced ERK1/2 phosphorylation. However, this was not impaired by inhibition of matrix metalloproteinase (MMP) and heparin-binding EGF. LPA-induced ERK1/2 activation occurs predominantly through EGF-R transactivation by Gi/Src and partly through activation of protein kinase C, which acts downstream of EGF-R and Ras. In contrast, LPA-induced phosphorylation of Shc and ERK1/2 in clonal hepatocytes (C9 cells) was primarily mediated through MMP-dependent transactivation of the EGF-R. These observations in adrenal glomerulosa and hepatic cells demonstrate that LPA phosphorylates ERK1/2 through EGF-R transactivation in a MMP-dependent or -independent manner in individual target cells. This reflects the ability of GPCRs expressed in cell lines and neoplastic cells to utilize distinct signaling pathways that can elicit altered responses compared with those of native tissues.
Collapse
Affiliation(s)
- Bukhtiar H Shah
- Endocrinology and Reproduction Research Branch, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda Maryland 20892-4510, USA.
| | | | | | | |
Collapse
|
49
|
Abstract
In this issue of Molecular Pharmacology, Oliveras-Reyes et al. (p. 356) describe the agonist-stimulated formation of a caveolin-dependent signalplex that includes both the angiotensin AT(1) receptor and the epidermal growth factor receptor, and probably also a number of other signal transduction intermediates. The signalplex is thought to facilitate the action of protein kinases that mediate angiotensin II-induced transactivation of the epidermal growth factor receptor and activation of extracellular signal-regulated kinase, and epidermal growth factor-induced inositol phosphate accumulation and phosphorylation/desensitization of the AT(1) receptor. This work contributes to an emerging view of the complexity and nonlinearity of signaling via G protein-coupled receptors and receptor tyrosine kinases, and of the importance of membrane compartmentalization to signal transduction.
Collapse
Affiliation(s)
- Kim A Neve
- VA Medical Center (R&D-30), 3710 SW US Veterans Hospital Rd, Portland, OR 97239-2999, USA.
| |
Collapse
|
50
|
Shah BH, Neithardt A, Chu DB, Shah FB, Catt KJ. Role of EGF receptor transactivation in phosphoinositide 3-kinase-dependent activation of MAP kinase by GPCRs. J Cell Physiol 2005; 206:47-57. [PMID: 15920762 DOI: 10.1002/jcp.20423] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Many G protein coupled receptors (GPCRs) cause phosphorylation of MAP kinases through transactivation of the epidermal growth factor receptor (EGF-R), leading to increased cell survival and growth, motility, and migration. Phosphoinositide 3-kinase (PI3K) is one of the important cell survival signaling molecules activated by EGF-R stimulation. However, the extent to which EGF-R transactivation is essential for GPCR agonist-stimulated PI3K activation is not known. Here we examined the mechanism of PI3K activation that elicits GPCR-mediated ERK1/2 activation by pathways dependent and/or independent of EGF-R transactivation in specific cell types. Immortalized hypothalamic neurons (GT1-7 cells) express endogenous gonadotropin-releasing hormone receptors (GnRH-R) and their stimulation causes marked phosphorylation of ERK1/2 and Akt (Ser 473) through transactivation of the EGF-R and recruitment of PI3K. In C9 hepatocytes, agonist activation of AT1 angiotensin II (AT1-R), lysophosphatidic acid (LPA), and EGF receptors caused phosphorylation of Akt through activation of the EGF-R in a PI3K-dependent manner. However, ERK1/2 activation by these agonists in these cells was independent of PI3K activation. In contrast, agonist stimulation of HEK 293 cells stably expressing AT1-R caused ERK1/2 phosphorylation that was independent of EGF-R transactivation but required PI3K activation. LPA signaling in these cells showed partial and complete dependence on EGF-R and PI3K, respectively. These data indicate that GPCR-induced ERK1/2 phosphorylation is dependent or independent of PI3K in specific cell types, and that the involvement of PI3K during ERK1/2 activation is not dependent solely on agonist-induced transactivation of the EGF-R.
Collapse
Affiliation(s)
- Bukhtiar H Shah
- Endocrinology and Reproduction Research Branch, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892-4510, USA.
| | | | | | | | | |
Collapse
|