1
|
Bahar ME, Kim HJ, Kim DR. Targeting the RAS/RAF/MAPK pathway for cancer therapy: from mechanism to clinical studies. Signal Transduct Target Ther 2023; 8:455. [PMID: 38105263 PMCID: PMC10725898 DOI: 10.1038/s41392-023-01705-z] [Citation(s) in RCA: 203] [Impact Index Per Article: 101.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/03/2023] [Accepted: 11/12/2023] [Indexed: 12/19/2023] Open
Abstract
Metastatic dissemination of solid tumors, a leading cause of cancer-related mortality, underscores the urgent need for enhanced insights into the molecular and cellular mechanisms underlying metastasis, chemoresistance, and the mechanistic backgrounds of individuals whose cancers are prone to migration. The most prevalent signaling cascade governed by multi-kinase inhibitors is the mitogen-activated protein kinase (MAPK) pathway, encompassing the RAS-RAF-MAPK kinase (MEK)-extracellular signal-related kinase (ERK) pathway. RAF kinase is a primary mediator of the MAPK pathway, responsible for the sequential activation of downstream targets, such as MEK and the transcription factor ERK, which control numerous cellular and physiological processes, including organism development, cell cycle control, cell proliferation and differentiation, cell survival, and death. Defects in this signaling cascade are associated with diseases such as cancer. RAF inhibitors (RAFi) combined with MEK blockers represent an FDA-approved therapeutic strategy for numerous RAF-mutant cancers, including melanoma, non-small cell lung carcinoma, and thyroid cancer. However, the development of therapy resistance by cancer cells remains an important barrier. Autophagy, an intracellular lysosome-dependent catabolic recycling process, plays a critical role in the development of RAFi resistance in cancer. Thus, targeting RAF and autophagy could be novel treatment strategies for RAF-mutant cancers. In this review, we delve deeper into the mechanistic insights surrounding RAF kinase signaling in tumorigenesis and RAFi-resistance. Furthermore, we explore and discuss the ongoing development of next-generation RAF inhibitors with enhanced therapeutic profiles. Additionally, this review sheds light on the functional interplay between RAF-targeted therapies and autophagy in cancer.
Collapse
Affiliation(s)
- Md Entaz Bahar
- Department of Biochemistry and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University, College of Medicine, Jinju, South Korea
| | - Hyun Joon Kim
- Department of Anatomy and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University, College of Medicine, Jinju, South Korea
| | - Deok Ryong Kim
- Department of Biochemistry and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University, College of Medicine, Jinju, South Korea.
| |
Collapse
|
2
|
Takaguri A, Ishizaka R, Maki S, Satoh K. The role of tribbles homolog 2 in vascular smooth muscle cell proliferation. Cell Biol Int 2023; 47:787-795. [PMID: 36626273 DOI: 10.1002/cbin.11982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/09/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023]
Abstract
Tribbles homolog 2 (TRIB2) functions as an adapter protein that regulates signal transductions involved in a variety of cellular functions, including tumorigenesis. However, the role of TRIB2 in the proliferation of vascular smooth muscle cells (VSMCs) and the underlying expression mechanisms remain unclear. The present study investigated the role of TRIB2 in VSMC proliferation and revealed that TRIB2 expression increases following vascular injury and platelet-derived growth factor (PDGF)-BB-stimulated VSMCs. We found that pretreatment with diphenyleneiodonium (a nicotinamide adenine dinucleotide phosphate oxidase inhibitor), U0126 (an inhibitor of mitogen-activated protein kinase kinase 1 [MEK1]), or siRNA targeting the gene encoding early growth response 1 (EGR-1) significantly inhibits PDGF-BB-induced TRIB2 expression in VSMCs. Furthermore, TRIB2 knockdown significantly inhibits PDGF-BB-induced proliferation of VSMCs but does not affect the phosphorylation of AKT. However, phosphorylation of ERK1 and expression of proliferating cell nuclear antibody are significantly suppressed in VSMCs by PDGF-BB stimulation. Thus, PDGF-BB-induced TRIB2 expression is mediated by ROS/ERK/EGR-1 pathways and plays a critical role in VSMC proliferation via modulation of ERK activity. We propose TRIB2 as a promising therapeutic target for the prevention of neointima formation and vascular disease.
Collapse
Affiliation(s)
- Akira Takaguri
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, Sapporo, Teine-ku, Japan
| | - Rena Ishizaka
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, Sapporo, Teine-ku, Japan
| | - Shota Maki
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, Sapporo, Teine-ku, Japan
| | - Kumi Satoh
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, Sapporo, Teine-ku, Japan
| |
Collapse
|
3
|
Fricke AL, Mühlhäuser WWD, Reimann L, Zimmermann JP, Reichenbach C, Knapp B, Peikert CD, Heberle AM, Faessler E, Schäuble S, Hahn U, Thedieck K, Radziwill G, Warscheid B. Phosphoproteomics Profiling Defines a Target Landscape of the Basophilic Protein Kinases AKT, S6K, and RSK in Skeletal Myotubes. J Proteome Res 2023; 22:768-789. [PMID: 36763541 DOI: 10.1021/acs.jproteome.2c00505] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Phosphorylation-dependent signal transduction plays an important role in regulating the functions and fate of skeletal muscle cells. Central players in the phospho-signaling network are the protein kinases AKT, S6K, and RSK as part of the PI3K-AKT-mTOR-S6K and RAF-MEK-ERK-RSK pathways. However, despite their functional importance, knowledge about their specific targets is incomplete because these kinases share the same basophilic substrate motif RxRxxp[ST]. To address this, we performed a multifaceted quantitative phosphoproteomics study of skeletal myotubes following kinase inhibition. Our data corroborate a cross talk between AKT and RAF, a negative feedback loop of RSK on ERK, and a putative connection between RSK and PI3K signaling. Altogether, we report a kinase target landscape containing 49 so far unknown target sites. AKT, S6K, and RSK phosphorylate numerous proteins involved in muscle development, integrity, and functions, and signaling converges on factors that are central for the skeletal muscle cytoskeleton. Whereas AKT controls insulin signaling and impinges on GTPase signaling, nuclear signaling is characteristic for RSK. Our data further support a role of RSK in glucose metabolism. Shared targets have functions in RNA maturation, stability, and translation, which suggests that these basophilic kinases establish an intricate signaling network to orchestrate and regulate processes involved in translation.
Collapse
Affiliation(s)
- Anna L Fricke
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany.,Biochemistry II, Theodor Boveri-Institute, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Wignand W D Mühlhäuser
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Lena Reimann
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Johannes P Zimmermann
- Biochemistry II, Theodor Boveri-Institute, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Christa Reichenbach
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Bettina Knapp
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Christian D Peikert
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Alexander M Heberle
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, 6020 Innsbruck, Austria
| | - Erik Faessler
- Jena University Language & Information Engineering (JULIE) Lab, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Sascha Schäuble
- Jena University Language & Information Engineering (JULIE) Lab, Friedrich Schiller University Jena, 07743 Jena, Germany.,Systems Biology and Bioinformatics Unit, Leibniz Institute for Natural Product Research and Infection Biology─Leibniz-HKI, 07745 Jena, Germany
| | - Udo Hahn
- Jena University Language & Information Engineering (JULIE) Lab, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Kathrin Thedieck
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, 6020 Innsbruck, Austria.,Department of Pediatrics, Section Systems Medicine of Metabolism and Signaling, University of Groningen, University Medical Center Groningen, Groningen 9700 RB, The Netherlands.,Department for Neuroscience, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg 26129, Germany
| | - Gerald Radziwill
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| | - Bettina Warscheid
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany.,Biochemistry II, Theodor Boveri-Institute, Biocenter, University of Würzburg, 97074 Würzburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
4
|
Takaguri A, Sasano J, Akihiro O, Satoh K. The role of circadian clock gene BMAL1 in vascular proliferation. Eur J Pharmacol 2020; 872:172924. [PMID: 31958455 DOI: 10.1016/j.ejphar.2020.172924] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 01/10/2020] [Accepted: 01/13/2020] [Indexed: 12/18/2022]
Abstract
Brain and muscle Arnt-like protein-1 (BMAL1), a component of the molecular clock, is implicated in the development of cardiovascular diseases, including atherosclerosis and abdominal aortic aneurysms. However, the role of BMAL1 in vascular proliferation associated with vascular remodeling is unknown. In the present study, we investigated the mechanisms underlying BMAL1 expression in vascular smooth muscle cells (VSMCs) and the role of BMAL1 in VSMC proliferation. BMAL1 expression significantly increased in injured carotid arteries in C57BL/6J mice and platelet-derived growth factor (PDGF)-BB-stimulated VSMC cultures. Pretreatment with diphenyleneiodonium (an NADPH oxidase inhibitor) and U0126 or PD98059 (MEK Inhibitors) inhibited PDGF-BB-induced BMAL1 expression in a dose-dependent manner in VSMCs. In addition, the knockdown of early growth factor protein-1 (Egr-1) significantly inhibited PDGF-BB-induced BMAL1 mRNA or protein expression in VSMCs, and the knockdown of BMAL1 significantly decreased PDGF-BB-induced cell proliferation and extracellular signal-regulated kinase (ERK) phosphorylation but not Akt phosphorylation in VSMCs. The results demonstrate that PDGF-BB up-regulates BMAL1 expression through reactive oxygen species/ERK/Egr-1 pathways and that BMAL1 is involved in PDGF-BB-induced cell proliferation partially through ERK in VSMCs. Thus, BMAL1 may be a novel therapeutic target for the treatment of atherosclerosis including vascular remodeling.
Collapse
Affiliation(s)
- Akira Takaguri
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 7-15-4-1 Maeda, Teine-ku, Sapporo, 006-8590, Japan
| | - Jun Sasano
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 7-15-4-1 Maeda, Teine-ku, Sapporo, 006-8590, Japan
| | - Oomiya Akihiro
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 7-15-4-1 Maeda, Teine-ku, Sapporo, 006-8590, Japan
| | - Kumi Satoh
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 7-15-4-1 Maeda, Teine-ku, Sapporo, 006-8590, Japan.
| |
Collapse
|
5
|
Doherty L, Yu J, Wang X, Hankenson KD, Kalajzic I, Sanjay A. A PDGFRβ-PI3K signaling axis mediates periosteal cell activation during fracture healing. PLoS One 2019; 14:e0223846. [PMID: 31665177 PMCID: PMC6821073 DOI: 10.1371/journal.pone.0223846] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 09/30/2019] [Indexed: 12/21/2022] Open
Abstract
Insufficient and delayed fracture healing remain significant public health problems with limited therapeutic options. Phosphoinositide 3-kinase (PI3K) signaling, a major pathway involved in regulation of fracture healing, promotes proliferation, migration, and differentiation of osteoprogenitors. We have recently reported that knock-in mice with a global increase in PI3K signaling (gCblYF) show enhanced femoral fracture healing characterized by an extraordinary periosteal response to injury. Interestingly, of all growth factor receptors involved in fracture healing, PI3K directly binds only to PDGFR. Given these findings, we hypothesized a PDGFR-PI3K interaction is necessary for mediating robust periosteal cell activation following fracture. In this study, we isolated primary periosteal cells from gCblYF mice to analyze cross-talk between the PDGFRβ and PI3K signaling pathways. We found PDGFRβ signaling contributes to robust Akt phosphorylation in periosteal cells in comparison with other growth factor signaling pathways. Additionally, we performed femoral fractures on gCblYF mice with a conditional removal of PDGFRβ in mesenchymal progenitors using inducible alpha smooth muscle actin (αSMA) CreERT2 mice. Our studies showed that depletion of PDGFRβ signaling within these progenitors in the early phase of fracture healing significantly abrogates PI3K-mediated periosteal activation and proliferation three days after fracture. Combined, these results suggest that PDGFRβ signaling through PI3K is necessary for robust periosteal activation in the earliest phases of fracture healing.
Collapse
Affiliation(s)
- Laura Doherty
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut, United States of America
| | - Jungeun Yu
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut, United States of America
| | - Xi Wang
- Department of Reconstructive Sciences, UConn Health, Farmington, Connecticut, United States of America
| | - Kurt D. Hankenson
- Department of Orthopaedic Surgery, School of Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, UConn Health, Farmington, Connecticut, United States of America
| | - Archana Sanjay
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
6
|
Electrochemically Reduced Water Delays Mammary Tumors Growth in Mice and Inhibits Breast Cancer Cells Survival In Vitro. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:4753507. [PMID: 30402124 PMCID: PMC6196883 DOI: 10.1155/2018/4753507] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 07/25/2018] [Accepted: 09/16/2018] [Indexed: 12/30/2022]
Abstract
Electrochemical reduced water (ERW) has been proposed to have beneficial effects on human health due to its rich content of H2 and the presence of platinum nanoparticles with antioxidant effects. Many studies have demonstrated that ERW scavenging properties are able to reduce the damage caused by oxidative stress in different experimental models. Although few in vivo studies have been reported, it has been demonstrated that ERW may display anticancer effects by induction of tumor cells apoptosis and reduction of both angiogenesis and inflammation. In this study, we show that ERW treatment of MCF-7, MDA-MB-453, and mouse (TUBO) breast cancer cells inhibited cell survival in a time-dependent fashion. ERW decreased ErbB2/neu expression and impaired pERK1/ERK2 and AKT phosphorylation in breast cancer cells. In addition, ERW treatment induced apoptosis of breast cancer cell lines independently of the status of p53 and ER and PR receptors. Our in vivo results showed that ERW treatment of transgenic BALB-neuT mice delayed the development of mammary tumors compared to the control. In addition, ERW induced a significant prolongation of tumor-free survival and a reduction in tumor multiplicity. Overall, these results suggest a potential beneficial role of ERW in inhibiting cancer cells growth.
Collapse
|
7
|
Hejmej A, Bilinska B. The effects of flutamide on cell-cell junctions in the testis, epididymis, and prostate. Reprod Toxicol 2018; 81:1-16. [PMID: 29958919 DOI: 10.1016/j.reprotox.2018.06.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 06/15/2018] [Accepted: 06/20/2018] [Indexed: 12/12/2022]
Abstract
In this review, we summarize recent findings on the effect of the anti-androgen flutamide on cell-cell junctions in the male reproductive system. We outline developmental aspects of flutamide action on the testis, epididymis, and prostate, and describe changes in junction protein expression and organization of junctional complexes in the adult boar following prenatal and postnatal exposure. We also discuss findings on the mechanisms by which flutamide induces alterations in cell-cell junctions in reproductive tissues of adult males, with special emphasis on cytoplasmic effects. Based on the results from in vivo and in vitro studies in the rat, we propose that flutamide affects the expression of junction proteins and junction complex structure not only by inhibiting androgen receptor activity, but equally important by modulating protein kinase-dependent signaling in testicular cells. Additionally, results from studies on prostate cancer cell lines point to a role for the cellular molecular outfit in response to flutamide.
Collapse
Affiliation(s)
- Anna Hejmej
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Barbara Bilinska
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
8
|
Gibbs WS, Garrett SM, Beeson CC, Schnellmann RG. Identification of dual mechanisms mediating 5-hydroxytryptamine receptor 1F-induced mitochondrial biogenesis. Am J Physiol Renal Physiol 2018; 314:F260-F268. [PMID: 29046298 PMCID: PMC5866450 DOI: 10.1152/ajprenal.00324.2017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 10/17/2017] [Accepted: 10/17/2017] [Indexed: 02/07/2023] Open
Abstract
Our laboratory recently made the novel observation that 5-hydroxytryptamine 1F (5-HT1F) receptor activation induces mitochondrial biogenesis (MB), the production of new, functional mitochondria, in vitro and in vivo. We sought to determine the mechanism linking the 5-HT1F receptor to MB in renal proximal tubule cells. Using LY344864 , a selective 5-HT1F receptor agonist, we determined that the 5-HT1F receptor is coupled to Gαi/o and induces MB through Gβγ-dependent activation of Akt, endothelial nitric oxide synthase (eNOS), cyclic guanosine-monophosphate (cGMP), protein kinase G (PKG), and peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α). We also report that the 5-HT1F receptor signals through a second, Gβγ-dependent pathway that is linked by Akt phosphorylation of Raf. In contrast to the activated Akt pathway, Raf phosphorylation reduced extracellular signal regulated kinases (ERK1/2) and foxhead box O3a (FOXO3a) phosphorylation, suppressing an inhibitory MB pathway. These results demonstrate that the 5-HT1F receptor regulates MB through Gβγ-dependent dual mechanisms that activate a stimulatory MB pathway, Akt/eNOS/cGMP/PKG/PGC-1α, while simultaneously repressing an inhibitory MB pathway, Raf/MEK/ERK/FOXO3a. Novel mechanisms of MB provide the foundation for new chemicals that induce MB to treat acute and chronic organ injuries.
Collapse
Affiliation(s)
- Whitney S Gibbs
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina , Charleston, South Carolina
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona , Tucson, Arizona
| | - Sara M Garrett
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina , Charleston, South Carolina
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina , Charleston, South Carolina
| | - Craig C Beeson
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina , Charleston, South Carolina
| | - Rick G Schnellmann
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona , Tucson, Arizona
- Southern Arizona Veterans Affairs Health Care System, Tucson, Arizona
| |
Collapse
|
9
|
Zheng X, Goodwin AF, Tian H, Jheon AH, Klein OD. Ras Signaling Regulates Stem Cells and Amelogenesis in the Mouse Incisor. J Dent Res 2017. [PMID: 28644741 DOI: 10.1177/0022034517717255] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The role of Ras signaling during tooth development is poorly understood. Ras proteins-which are activated by many upstream pathways, including receptor tyrosine kinase cascades-signal through multiple effectors, such as the mitogen-activated protein kinase (MAPK) and PI3K pathways. Here, we utilized the mouse incisor as a model to study how the MAPK and PI3K pathways regulate dental epithelial stem cells and amelogenesis. The rodent incisor-which grows continuously throughout the life of the animal due to the presence of epithelial and mesenchymal stem cells-provides a model for the study of ectodermal organ renewal and regeneration. Utilizing models of Ras dysregulation as well as inhibitors of the MAPK and PI3K pathways, we found that MAPK and PI3K regulate dental epithelial stem cell activity, transit-amplifying cell proliferation, and enamel formation in the mouse incisor.
Collapse
Affiliation(s)
- X Zheng
- 1 Department of Stomatology, Peking University Third Hospital, Beijing, China.,2 Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
| | - A F Goodwin
- 2 Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
| | - H Tian
- 2 Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
| | - A H Jheon
- 2 Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
| | - O D Klein
- 2 Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA.,3 Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
10
|
Tang H, Xue G. Major Physiological Signaling Pathways in the Regulation of Cell Proliferation and Survival. Handb Exp Pharmacol 2017; 249:13-30. [PMID: 28233182 DOI: 10.1007/164_2017_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Multiple signaling pathways regulate cell proliferation and survival and are therefore important for maintaining homeostasis of development. The balance between cell growth and death is achieved through orchestrated signal transduction pathways mediated by complex functional interactions between signaling axes, among which, PI3K/Akt and Ras/MAPK as well as JAK/STAT play a dominant role in promoting cell proliferation, differentiation, and survival. In clinical cancer therapies, drug resistance is the major challenge that occurs in almost all targeted therapeutic strategies. Recent advances in research have suggested that the intrinsic pro-survival signaling crosstalk is the driving force in acquired resistance to a targeted therapy, which may be abolished by interfering with the cross-reacting network.
Collapse
Affiliation(s)
- Huifang Tang
- Department of Pharmacology, Zhejiang University, School of Basic Medical Sciences, Hangzhou, 310058, China.
| | - Gongda Xue
- Department of Biomedicine, University Hospital Basel, Basel, 4031, Switzerland
| |
Collapse
|
11
|
Fritsche-Guenther R, Witzel F, Kempa S, Brummer T, Sers C, Blüthgen N. Effects of RAF inhibitors on PI3K/AKT signalling depend on mutational status of the RAS/RAF signalling axis. Oncotarget 2016; 7:7960-9. [PMID: 26799289 PMCID: PMC4884967 DOI: 10.18632/oncotarget.6959] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 11/25/2015] [Indexed: 01/20/2023] Open
Abstract
Targeted therapies within the RAS/RAF/MEK/ERK signalling axis become increasingly popular, yet cross-talk and feedbacks in the signalling network lead to unexpected effects. Here we look systematically into how inhibiting RAF and MEK with clinically relevant inhibitors result in changes in PI3K/AKT activation. We measure the signalling response using a bead-based ELISA, and use a panel of three cell lines, and isogenic cell lines that express mutant forms of the oncogenes KRAS and BRAF to interrogate the effects of the MEK and RAF inhibitors on signalling. We find that treatment with the RAF inhibitors have opposing effects on AKT phosphorylation depending on the mutational status of two important oncogenes, KRAS and BRAF. If these two genes are in wildtype configuration, RAF inhibitors reduce AKT phosphorylation. In contrast, if BRAF or KRAS are mutant, RAF inhibitors will leave AKT phosphorylation unaffected or lead to an increase of AKT phosphorylation. Down-regulation of phospho-AKT by RAF inhibitors also extends to downstream transcription factors, and correlates with apoptosis induction. Our results show that oncogenes rewire signalling such that targeted therapies can have opposing effects on parallel pathways, which depend on the mutational status of the cell.
Collapse
Affiliation(s)
- Raphaela Fritsche-Guenther
- Max-Delbrück-Center for Molecular Medicin (MDC) Berlin Buch, The Berlin Institute for Medical Systems Biology (BIMSB), 13125 Berlin, Germany
| | - Franziska Witzel
- Institute of Pathology, Molecular Tumor Pathology, Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany.,Institute for Theoretical Biology, Charité - Universitätsmedizin Berlin, 10115 Berlin, Germany
| | - Stefan Kempa
- Max-Delbrück-Center for Molecular Medicin (MDC) Berlin Buch, The Berlin Institute for Medical Systems Biology (BIMSB), 13125 Berlin, Germany
| | - Tilman Brummer
- Institute of Molecular Medicine and Cell Research and Centre for Biological Signalling Studies BIOSS, Albert-Ludwigs University Freiburg, 79104 Freiburg, Germany
| | - Christine Sers
- Institute of Pathology, Molecular Tumor Pathology, Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Nils Blüthgen
- Institute of Pathology, Molecular Tumor Pathology, Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany.,Institute for Theoretical Biology, Charité - Universitätsmedizin Berlin, 10115 Berlin, Germany.,Integrative Research Institute for the Life Sciences, Humboldt University Berlin, 10099 Berlin, Germany
| |
Collapse
|
12
|
Optogenetic clustering of CNK1 reveals mechanistic insights in RAF and AKT signalling controlling cell fate decisions. Sci Rep 2016; 6:38155. [PMID: 27901111 PMCID: PMC5128868 DOI: 10.1038/srep38155] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 11/04/2016] [Indexed: 12/27/2022] Open
Abstract
Scaffold proteins such as the multidomain protein CNK1 orchestrate the signalling network by integrating and controlling the underlying pathways. Using an optogenetic approach to stimulate CNK1 uncoupled from upstream effectors, we identified selective clusters of CNK1 that either stimulate RAF-MEK-ERK or AKT signalling depending on the light intensity applied. OptoCNK1 implemented in MCF7 cells induces differentiation at low light intensity stimulating ERK activity whereas stimulation of AKT signalling by higher light intensity promotes cell proliferation. CNK1 clustering in response to increasing EGF concentrations revealed that CNK1 binds to RAF correlating with ERK activation at low EGF dose. At higher EGF dose active AKT binds to CNK1 and phosphorylates and inhibits RAF. Knockdown of CNK1 protects CNK1 from this AKT/RAF crosstalk. In C2 skeletal muscle cells CNK1 expression is induced with the onset of differentiation. Hence, AKT-bound CNK1 counteracts ERK stimulation in differentiated but not in proliferating cells. Ectopically expressed CNK1 facilitates C2 cell differentiation and knockdown of CNK1 impaired the transcriptional network underlying C2 cell differentiation. Thus, CNK1 expression, CNK1 clustering and the thereto related differential signalling processes decide on proliferation and differentiation in a cell type- and cell stage-dependent manner by orchestrating AKT and RAF signalling.
Collapse
|
13
|
Abstract
Gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) constitute a heterogeneous group of tumours associated with variable clinical presentations, growth rates, and prognoses. To improve the management of GEP-NENs, the WHO developed a classification system that enables tumours to be graded based on markers of cell proliferation in biopsy specimens. Indeed, histopathology has been a mainstay in the diagnosis of GEP-NENs, and the WHO grading system facilitates therapeutic decision-making; however, considerable intratumoural heterogeneity, predominantly comprising regional variations in proliferation rates, complicates the evaluation of tumour biology. The use of molecular imaging modalities to delineate the most-aggressive cell populations is becoming more widespread. In addition, molecular profiling is increasingly undertaken in the clinical setting, and genomic studies have revealed a number of chromosomal alterations in GEP-NENs, although the 'drivers' of neoplastic development have not been identified. Thus, our molecular understanding of GEP-NENs remains insufficient to inform on patient prognosis or selection for treatments, and the WHO classification continues to form the basis for management of this disease. Nevertheless, our increasing understanding of the molecular genetics and biology of GEP-NENs has begun to expose flaws in the WHO classification. We describe the current understanding of the molecular characteristics of GEP-NENs, and discuss how advances in molecular profiling measurements, including assays of circulating mRNAs, are likely to influence the management of these tumours.
Collapse
|
14
|
Chojnacka K, Zarzycka M, Hejmej A, Mruk DD, Gorowska E, Kotula-Balak M, Klimek M, Bilinska B. Hydroxyflutamide affects connexin 43 via the activation of PI3K/Akt-dependent pathway but has no effect on the crosstalk between PI3K/Akt and ERK1/2 pathways at the Raf-1 kinase level in primary rat Sertoli cells. Toxicol In Vitro 2016; 31:146-57. [DOI: 10.1016/j.tiv.2015.09.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 09/23/2015] [Accepted: 09/29/2015] [Indexed: 02/07/2023]
|
15
|
Crosstalk between SDF-1/CXCR4 and SDF-1/CXCR7 in cardiac stem cell migration. Sci Rep 2015; 5:16813. [PMID: 26578388 PMCID: PMC4649491 DOI: 10.1038/srep16813] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 10/20/2015] [Indexed: 12/20/2022] Open
Abstract
Stromal cell-derived factor 1 (SDF-1) is a chemokine that can be expressed in injured cardiomyocytes after myocardial infarction (MI). By combining with its receptor CXCR4, SDF-1 induced stem and progenitor cells migration. CXCR7, a novel receptor for SDF-1, has been identified recently. We aimed to explore the roles of SDF-1/CXCR4 and SDF-1/CXCR7 pathway and their crosstalk in CSCs migration. In the present study, CXCR4 and CXCR7 expression were identified in CSCs. Transwell assay showed that SDF-1 caused CSCs migration in a dose- and time-dependent manner, which could be significantly suppressed by CXCR4 or CXCR7 siRNA. Phospho-ERK, phospho-Akt and Raf-1 significantly elevated in CSCs with SDF-1 stimulation. Knockdown of CXCR4 or CXCR7 significantly decreased phospho-ERK or phospho-Akt, respectively, and eventually resulted in the inhibition of CSCs migration. Moreover, western blot showed that MK2206 (Akt inhibitor) increased the expression of phospho-ERK and Raf-1, whereas PD98059 (ERK inhibitor) had no effect on phospho-Akt and Raf-1. GW5074 (Raf-1 inhibitor) upregulated the expression of phospho-ERK, but had no effect on phospho-Akt. The present study indicated that SDF-1/CXCR7/Akt and SDF-1/CXCR4/ERK pathway played important roles in CSCs migration. Akt phosphorylation inhibited Raf-1 activity, which in turn dephosphorylated ERK and negatively regulated CSCs migration.
Collapse
|
16
|
Zhou J, Du T, Li B, Rong Y, Verkhratsky A, Peng L. Crosstalk Between MAPK/ERK and PI3K/AKT Signal Pathways During Brain Ischemia/Reperfusion. ASN Neuro 2015; 7:7/5/1759091415602463. [PMID: 26442853 PMCID: PMC4601130 DOI: 10.1177/1759091415602463] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The epidermal growth factor receptor (EGFR) is linked to the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) and Raf/mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK1/2) signaling pathways. During brain ischemia/reperfusion, EGFR could be transactivated, which stimulates these intracellular signaling cascades that either protect cells or potentiate cell injury. In the present study, we investigated the activation of EGFR, PI3K/AKT, and Raf/MAPK/ERK1/2 during ischemia or reperfusion of the brain using the middle cerebral artery occlusion model. We found that EGFR was phosphorylated and transactivated during both ischemia and reperfusion periods. During ischemia, the activity of PI3K/AKT pathway was significantly increased, as judged from the strong phosphorylation of AKT; this activation was suppressed by the inhibitors of EGFR and Zn-dependent metalloproteinase. Ischemia, however, did not induce ERK1/2 phosphorylation, which was dependent on reperfusion. Coimmunoprecipitation of Son of sevenless 1 (SOS1) with EGFR showed increased association between the receptor and SOS1 in ischemia, indicating the inhibitory node downstream of SOS1. The inhibitory phosphorylation site of Raf-1 at Ser259, but not its stimulatory phosphorylation site at Ser338, was phosphorylated during ischemia. Furthermore, ischemia prompted the interaction between Raf-1 and AKT, while both the inhibitors of PI3K and AKT not only abolished AKT phosphorylation but also restored ERK1/2 phosphorylation. All these findings suggest that Raf/MAPK/ERK1/2 signal pathway is inhibited by AKT via direct phosphorylation and inhibition at Raf-1 node during ischemia. During reperfusion, we observed a significant increase of ERK1/2 phosphorylation but no change in AKT phosphorylation. Inhibitors of reactive oxygen species and phosphatase and tensin homolog restored AKT phosphorylation but abolished ERK1/2 phosphorylation, suggesting that the reactive oxygen species-dependent increase in phosphatase and tensin homolog activity in reperfusion period relieves ERK1/2 from inhibition of AKT.
Collapse
Affiliation(s)
- Jing Zhou
- Laboratory of Metabolic Brain Diseases, Institute of Metabolic Disease Research and Drug Development, China Medical University, Shenyang, P. R. China
| | - Ting Du
- Laboratory of Metabolic Brain Diseases, Institute of Metabolic Disease Research and Drug Development, China Medical University, Shenyang, P. R. China
| | - Baoman Li
- Laboratory of Metabolic Brain Diseases, Institute of Metabolic Disease Research and Drug Development, China Medical University, Shenyang, P. R. China
| | - Yan Rong
- Laboratory of Metabolic Brain Diseases, Institute of Metabolic Disease Research and Drug Development, China Medical University, Shenyang, P. R. China
| | - Alexei Verkhratsky
- Faculty of Life Science, The University of Manchester, UK Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain University of Nizhny Novgorod, Russia
| | - Liang Peng
- Laboratory of Metabolic Brain Diseases, Institute of Metabolic Disease Research and Drug Development, China Medical University, Shenyang, P. R. China
| |
Collapse
|
17
|
Hellesøy M, Lorens JB. Cellular context-mediated Akt dynamics regulates MAP kinase signaling thresholds during angiogenesis. Mol Biol Cell 2015; 26:2698-711. [PMID: 26023089 PMCID: PMC4501366 DOI: 10.1091/mbc.e14-09-1378] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 05/18/2015] [Indexed: 01/01/2023] Open
Abstract
This study examines the temporal regulation of Akt in endothelial cells during formation of capillary-like networks induced by cell–cell contact with vascular smooth muscle cells (vSMCs) and vSMC-associated VEGF. Heterotypic cell–cell interaction between mural and endothelial cells alters Akt kinase protein dynamics, which regulates angiogenesis. The formation of new blood vessels by sprouting angiogenesis is tightly regulated by contextual cues that affect angiogeneic growth factor signaling. Both constitutive activation and loss of Akt kinase activity in endothelial cells impair angiogenesis, suggesting that Akt dynamics mediates contextual microenvironmental regulation. We explored the temporal regulation of Akt in endothelial cells during formation of capillary-like networks induced by cell–cell contact with vascular smooth muscle cells (vSMCs) and vSMC-associated VEGF. Expression of constitutively active Akt1 strongly inhibited network formation, whereas hemiphosphorylated Akt1 epi-alleles with reduced kinase activity had an intermediate inhibitory effect. Conversely, inhibition of Akt signaling did not affect endothelial cell migration or morphogenesis in vSMC cocultures that generate capillary-like structures. We found that endothelial Akt activity is transiently blocked by proteasomal degradation in the presence of SMCs during the initial phase of capillary-like structure formation. Suppressed Akt activity corresponded to the increased endothelial MAP kinase signaling that was required for angiogenic endothelial morphogenesis. These results reveal a regulatory principle by which cellular context regulates Akt protein dynamics, which determines MAP kinase signaling thresholds necessary drive a morphogenetic program during angiogenesis.
Collapse
Affiliation(s)
- Monica Hellesøy
- Department of Biomedicine, University of Bergen, N-5009 Bergen, Norway
| | - James B Lorens
- Department of Biomedicine, University of Bergen, N-5009 Bergen, Norway Center for Cancer Biomarkers, University of Bergen, N-5009 Bergen, Norway
| |
Collapse
|
18
|
Chen S, Liu B, Kong D, Li S, Li C, Wang H, Sun Y. Atorvastatin calcium inhibits phenotypic modulation of PDGF-BB-induced VSMCs via down-regulation the Akt signaling pathway. PLoS One 2015; 10:e0122577. [PMID: 25874930 PMCID: PMC4398430 DOI: 10.1371/journal.pone.0122577] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 02/16/2015] [Indexed: 11/30/2022] Open
Abstract
Plasticity of vascular smooth muscle cells (VSMCs) plays a central role in the onset and progression of proliferative vascular diseases. In adult tissue, VSMCs exist in a physiological contractile-quiescent phenotype, which is defined by lack of the ability of proliferation and migration, while high expression of contractile marker proteins. After injury to the vessel, VSMC shifts from a contractile phenotype to a pathological synthetic phenotype, associated with increased proliferation, migration and matrix secretion. It has been demonstrated that PDGF-BB is a critical mediator of VSMCs phenotypic switch. Atorvastatin calcium, a selective inhibitor of 3-hydroxy-3-methyl-glutaryl l coenzyme A (HMG-CoA) reductase, exhibits various protective effects against VSMCs. In this study, we investigated the effects of atorvastatin calcium on phenotype modulation of PDGF-BB-induced VSMCs and the related intracellular signal transduction pathways. Treatment of VSMCs with atorvastatin calcium showed dose-dependent inhibition of PDGF-BB-induced proliferation. Atorvastatin calcium co-treatment inhibited the phenotype modulation and cytoskeleton rearrangements and improved the expression of contractile phenotype marker proteins such as α-SM actin, SM22α and calponin in comparison with PDGF-BB alone stimulated VSMCs. Although Akt phosphorylation was strongly elicited by PDGF-BB, Akt activation was attenuated when PDGF-BB was co-administrated with atorvastatin calcium. In conclusion, atorvastatin calcium inhibits phenotype modulation of PDGF-BB-induced VSMCs and activation of the Akt signaling pathway, indicating that Akt might play a vital role in the modulation of phenotype.
Collapse
Affiliation(s)
- Shuang Chen
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Baoqin Liu
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang Liaoning, China
| | - Dehui Kong
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang Liaoning, China
| | - Si Li
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang Liaoning, China
| | - Chao Li
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang Liaoning, China
| | - Huaqin Wang
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang Liaoning, China
| | - Yingxian Sun
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China
- * E-mail:
| |
Collapse
|
19
|
Fernández-Mariño AI, Cidad P, Zafra D, Nocito L, Domínguez J, Oliván-Viguera A, Köhler R, López-López JR, Pérez-García MT, Valverde MÁ, Guinovart JJ, Fernández-Fernández JM. Tungstate-targeting of BKαβ1 channels tunes ERK phosphorylation and cell proliferation in human vascular smooth muscle. PLoS One 2015; 10:e0118148. [PMID: 25659150 PMCID: PMC4320054 DOI: 10.1371/journal.pone.0118148] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 01/05/2015] [Indexed: 02/07/2023] Open
Abstract
Despite the substantial knowledge on the antidiabetic, antiobesity and antihypertensive actions of tungstate, information on its primary target/s is scarce. Tungstate activates both the ERK1/2 pathway and the vascular voltage- and Ca2+-dependent large-conductance BKαβ1 potassium channel, which modulates vascular smooth muscle cell (VSMC) proliferation and function, respectively. Here, we have assessed the possible involvement of BKαβ1 channels in the tungstate-induced ERK phosphorylation and its relevance for VSMC proliferation. Western blot analysis in HEK cell lines showed that expression of vascular BKαβ1 channels potentiates the tungstate-induced ERK1/2 phosphorylation in a Gi/o protein-dependent manner. Tungstate activated BKαβ1 channels upstream of G proteins as channel activation was not altered by the inhibition of G proteins with GDPβS or pertussis toxin. Moreover, analysis of Gi/o protein activation measuring the FRET among heterologously expressed Gi protein subunits suggested that tungstate-targeting of BKαβ1 channels promotes G protein activation. Single channel recordings on VSMCs from wild-type and β1-knockout mice indicated that the presence of the regulatory β1 subunit was essential for the tungstate-mediated activation of BK channels in VSMCs. Moreover, the specific BK channel blocker iberiotoxin lowered tungstate-induced ERK phosphorylation by 55% and partially reverted (by 51%) the tungstate-produced reduction of platelet-derived growth factor (PDGF)-induced proliferation in human VSMCs. Our observations indicate that tungstate-targeting of BKαβ1 channels promotes activation of PTX-sensitive Gi proteins to enhance the tungstate-induced phosphorylation of ERK, and inhibits PDGF-stimulated cell proliferation in human vascular smooth muscle.
Collapse
Affiliation(s)
- Ana Isabel Fernández-Mariño
- Laboratori de Fisiologia Molecular i Canalopaties, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | - Pilar Cidad
- Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain
| | - Delia Zafra
- Institute for Research in Biomedicine (IRB Barcelona) and Department of Biochemistry and Molecular Biology, University of Barcelona, and Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), Barcelona, Spain
| | - Laura Nocito
- Institute for Research in Biomedicine (IRB Barcelona) and Department of Biochemistry and Molecular Biology, University of Barcelona, and Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), Barcelona, Spain
| | - Jorge Domínguez
- Institute for Research in Biomedicine (IRB Barcelona) and Department of Biochemistry and Molecular Biology, University of Barcelona, and Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), Barcelona, Spain
| | - Aida Oliván-Viguera
- Aragon Institute of Health Sciences I+CS/IIS and Fundación Agencia Aragonesa para la Investigación y Desarrollo (ARAID), Zaragoza, Spain
| | - Ralf Köhler
- Aragon Institute of Health Sciences I+CS/IIS and Fundación Agencia Aragonesa para la Investigación y Desarrollo (ARAID), Zaragoza, Spain
| | - José R. López-López
- Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain
| | - María Teresa Pérez-García
- Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain
| | - Miguel Ángel Valverde
- Laboratori de Fisiologia Molecular i Canalopaties, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | - Joan J. Guinovart
- Institute for Research in Biomedicine (IRB Barcelona) and Department of Biochemistry and Molecular Biology, University of Barcelona, and Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), Barcelona, Spain
| | - José M. Fernández-Fernández
- Laboratori de Fisiologia Molecular i Canalopaties, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
- * E-mail:
| |
Collapse
|
20
|
Nurminskaya M, Beazley KE, Smith EP, Belkin AM. Transglutaminase 2 promotes PDGF-mediated activation of PDGFR/Akt1 and β-catenin signaling in vascular smooth muscle cells and supports neointima formation. J Vasc Res 2015; 51:418-28. [PMID: 25612735 DOI: 10.1159/000369461] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 10/25/2014] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Phenotypic switch of vascular smooth muscle cells (VSMCs) accompanies neointima formation and associates with vascular diseases. Platelet-derived growth factor (PDGF)-induced activation of PDGFR/Akt1 and β-catenin signaling pathways in VSMCs has been implicated in vessel occlusion. Transglutaminase 2 (TG2) regulates these pathways and its levels are increased in the neointima. OBJECTIVE The aim of this study was to evaluate the role of TG2 in PDGF/β-catenin signaling cross-talk and assess its contribution to neointima. METHODS Aortic VSMCs from wild-type and TG2 knockout mice were tested in vitro for levels of VSMC markers, proliferation, migration and PDGF-induced activation of PDGFR/Akt1 and β-catenin pathways. Neointima in these mice was studied ex vivo in coronary vessels using a heart slice model and in vivo using a carotid artery ligation model. RESULTS Genetic deletion of TG2 attenuated the PDGF-induced phenotypic switch of aortic VSMCs, reduced their proliferation and migration rates, and inhibited PDGF-induced activation of PDGFR/Akt1 and β-catenin pathways in both ex vivo and in vivo neointima models. Importantly, genetic deletion of TG2 also markedly attenuated vessel occlusion. CONCLUSIONS TG2 promotes neointima formation by mediating the PDGF-induced activation of the PDGFR/Akt1 and β-catenin pathways in VSMCs. This study identifies TG2 as a potential therapeutic target for blocking neointima in blood vessels.
Collapse
Affiliation(s)
- Maria Nurminskaya
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Md., USA
| | | | | | | |
Collapse
|
21
|
Adapala NS, Barbe MF, Tsygankov AY, Lorenzo JA, Sanjay A. Loss of Cbl-PI3K interaction enhances osteoclast survival due to p21-Ras mediated PI3K activation independent of Cbl-b. J Cell Biochem 2015; 115:1277-89. [PMID: 24470255 DOI: 10.1002/jcb.24779] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 01/24/2014] [Indexed: 01/14/2023]
Abstract
Cbl family proteins, Cbl and Cbl-b, are E3 ubiquitin ligases and adaptor proteins, which play important roles in bone-resorbing osteoclasts. Loss of Cbl in mice decreases osteoclast migration, resulting in delayed bone development where as absence of Cbl-b decreases bone volume due to hyper-resorptive osteoclasts. A major structural difference between Cbl and Cbl-b is tyrosine 737 (in YEAM motif) only on Cbl, which upon phosphorylation interacts with the p85 subunit of phosphatidylinositol-3 Kinase (PI3K). In contrast to Cbl(-/-) and Cbl-b(-/-) , mice lacking Cbl-PI3K interaction due to a Y737F (tyrosine to phenylalanine, YF) mutation showed enhanced osteoclast survival, but defective bone resorption. To investigate whether Cbl-PI3K interaction contributes to distinct roles of Cbl and Cbl-b in osteoclasts, mice bearing CblY737F mutation in the Cbl-b(-/-) background (YF/YF;Cbl-b(-/-) ) were generated. The differentiation and survival were augmented similarly in YF/YF and YF/YF;Cbl-b(-/-) osteoclasts, associated with enhanced PI3K signaling suggesting an exclusive role of Cbl-PI3K interaction, independent of Cbl-b. In addition to PI3K, the small GTPase Ras also regulates osteoclast survival. In the absence of Cbl-PI3K interaction, increased Ras GTPase activity and Ras-PI3K binding were observed and inhibition of Ras activation attenuated PI3K mediated osteoclast survival. In contrast to differentiation and survival, increased osteoclast activity observed in Cbl-b(-/-) mice persisted even after introduction of the resorption-defective YF mutation in YF/YF;Cbl-b(-/-) mice. Hence, Cbl and Cbl-b play mutually exclusive roles in osteoclasts. Whereas Cbl-PI3K interaction regulates differentiation and survival, bone resorption is predominantly regulated by Cbl-b in osteoclasts.
Collapse
Affiliation(s)
- Naga Suresh Adapala
- Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, Connecticut, 06032
| | | | | | | | | |
Collapse
|
22
|
Yuan H, Chen R, Wu L, Chen Q, Hu A, Zhang T, Wang Z, Zhu X. The regulatory mechanism of neurogenesis by IGF-1 in adult mice. Mol Neurobiol 2014; 51:512-22. [PMID: 24777577 DOI: 10.1007/s12035-014-8717-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 04/14/2014] [Indexed: 12/22/2022]
Abstract
Growth factors like insulin-like growth factor 1 (IGF-1) is reported to mediate neurogenesis in the subgranular zone (SGZ) and the subventricular zone (SVZ) of the adult mammalian brain, but its regulatory mechanism remains unclear. We generated transgenic mice overexpressing IGF-1 specifically in neural stem cells (NSCs) and assessed the effect of IGF-1 on neurogenesis in adult mice NSCs. Overexpression of IGF-1 could stimulate the expression of phospho-Akt and phospho-ERK1/2 while inducing proliferation and differentiation of NSCs in the SGZ and SVZ. The MEK/ERK inhibitor U0126 could inhibit ERK1/2 phosphorylation, further inhibiting the proliferation of NSCs in the SGZ and SVZ but had no effect on the phosphorylation of Akt. By contrast, The PI3K/Akt inhibitor LY294002 inhibited phosphorylation of Akt and differentiation of NSCs in the SGZ and SVZ, resulting in no change in the proliferation of NSCs and ERK1/2 phosphorylation. These results demonstrate that IGF-1 upregulates the proliferation of NSCs by triggering MEK/ERK pathway signaling in the adult mice SGZ and SVZ. Meanwhile, IGF-1 also induces differentiation of NSCs via the PI3K/Akt pathway in adult mice. However, we found no evidence of crosstalk between the PI3K/Akt and MEK/ERK pathways in adult mice NSCs. Our work provides new experimental evidence of the involvement of the PI3K/Akt and MEK/ERK pathways in the proliferation and differentiation of the NSCs of adult mice.
Collapse
Affiliation(s)
- Honghua Yuan
- Research Center for Neurobiology, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Resveratrol inhibits phenotype modulation by platelet derived growth factor-bb in rat aortic smooth muscle cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:572430. [PMID: 24738020 PMCID: PMC3964901 DOI: 10.1155/2014/572430] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 01/08/2014] [Accepted: 01/27/2014] [Indexed: 02/07/2023]
Abstract
Dedifferentiated vascular smooth muscle cells (VSMCs) are phenotypically modulated from the contractile state to the active synthetic state in the vessel wall. In this study, we investigated the effects of resveratrol on phenotype modulation by dedifferentiation and the intracellular signal transduction pathways of platelet derived growth factor-bb (PDGF-bb) in rat aortic vascular smooth muscle cells (RAOSMCs). Treatment of RAOSMCs with resveratrol showed dose-dependent inhibition of PDGF-bb-stimulated proliferation. Resveratrol treatment inhibited this phenotype change and disassembly of actin filaments and maintained the expression of contractile phenotype-related proteins such as calponin and smooth muscle actin-alpha in comparison with only PDGF-bb stimulated RAOSMC. Although PDGF stimulation elicited strong and detectable Akt and mTOR phosphorylations lasting for several hours, Akt activation was much weaker when PDGF was used with resveratrol. In contrast, resveratrol only slightly inhibited phosphorylations of 42/44 MAPK and p38 MAPK. In conclusion, RAOSMC dedifferentiation, phenotype, and proliferation rate were inhibited by resveratrol via interruption of the balance of Akt, 42/44MAPK, and p38MAPK pathway activation stimulated by PDGF-bb.
Collapse
|
24
|
Yue L, Haroun S, Parent JL, de Brum-Fernandes AJ. Prostaglandin D(2) induces apoptosis of human osteoclasts through ERK1/2 and Akt signaling pathways. Bone 2014; 60:112-21. [PMID: 24345643 DOI: 10.1016/j.bone.2013.12.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 12/06/2013] [Accepted: 12/09/2013] [Indexed: 11/20/2022]
Abstract
In a recent study we have shown that prostaglandin D2 (PGD2) induces human osteoclast (OC) apoptosis through the activation of the chemoattractant receptor homologous molecule expressed on T-helper type 2 cell (CRTH2) receptor and the intrinsic apoptotic pathway. However, the molecular mechanisms underlying this response remain elusive. The objective of this study is to investigate the intracellular signaling pathways mediating PGD2-induced OC apoptosis. OCs were generated by in vitro differentiation of human peripheral blood mononuclear cells (PBMCs), and then treated with or without the selective inhibitors of mitogen-activated protein kinase-extracellular signal-regulated kinase (ERK) kinase, (MEK)-1/2, phosphatidylinositol3-kinase (PI3K) and NF-κB/IκB kinase-2 (IKK2) prior to the treatments of PGD2 as well as its agonists and antagonists. Fluorogenic substrate assay and immunoblotting were performed to determine the caspase-3 activity and key proteins involved in Akt, ERK1/2 and NF-κB signaling pathways. Treatments with both PGD2 and a CRTH2 agonist decreased ERK1/2 (Thr202/Tyr204) and Akt (Ser473) phosphorylation, whereas both treatments increased β-arrestin-1 phosphorylation (Ser412) in the presence of naproxen, which was used to eliminate endogenous prostaglandin production. In the absence of naproxen, treatment with a CRTH2 antagonist increased both ERK1/2 and Akt phosphorylations, and reduced the phosphorylation of β-arrestin-1. Treatment of OCs with a selective MEK-1/2 inhibitor increased caspase-3 activity and OC apoptosis induced by both PGD2 and a CRTH2 agonist. Moreover, a CRTH2 antagonist diminished the selective MEK-1/2 inhibitor-induced increase in caspase-3 activity in the presence of endogenous prostaglandins. In addition, treatment of OCs with a selective PI3K inhibitor decreased ERK1/2 (Thr202/Tyr204) phosphorylation caused by PGD2, whereas increased ERK1/2 (Thr202/Tyr204) phosphorylation by a CRTH2 antagonist was attenuated with a PI3K inhibitor treatment. The DP receptor was not implicated in any of the parameters evaluated. Treatment of OCs with PGD2 as well as its receptor agonists and antagonists did not alter the phosphorylation of RelA/p65 (Ser536). Moreover, the caspase-3 activity was not altered in OCs treated with a selective IKK2/NF-κB inhibitor. In conclusion, endogenous or exogenous PGD2 induces CRTH2-dependent apoptosis in human differentiated OCs; β-arrestin-1, ERK1/2, and Akt, but not IKK2/NF-κB are probably implicated in the signaling pathways of this receptor in the model studied.
Collapse
Affiliation(s)
- Li Yue
- Department of Pharmacology, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, Quebec J1H 5N4, Canada; Division of Rheumatology, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, Quebec J1H 5N4, Canada.
| | - Sonia Haroun
- Division of Rheumatology, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, Quebec J1H 5N4, Canada.
| | - Jean-Luc Parent
- Department of Pharmacology, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, Quebec J1H 5N4, Canada; Division of Rheumatology, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, Quebec J1H 5N4, Canada.
| | - Artur J de Brum-Fernandes
- Department of Pharmacology, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, Quebec J1H 5N4, Canada; Division of Rheumatology, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, Quebec J1H 5N4, Canada.
| |
Collapse
|
25
|
Mitogenesis of vascular smooth muscle cell stimulated by platelet-derived growth factor-bb is inhibited by blocking of intracellular signaling by epigallocatechin-3-O-gallate. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:827905. [PMID: 24307927 PMCID: PMC3836374 DOI: 10.1155/2013/827905] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 08/19/2013] [Accepted: 09/09/2013] [Indexed: 01/04/2023]
Abstract
Epigallocatechin gallate (EGCG) is known to exhibit antioxidant, antiproliferative, and antithrombogenic effects and reduce the risk of cardiovascular diseases. Key events in the development of cardiovascular disease are hypertrophy and hyperplasia according to vascular smooth muscle cell proliferation. In this study, we investigated whether EGCG can interfere with PDGF-bb stimulated proliferation, cell cycle distribution, and the gelatinolytic activity of MMP and signal transduction pathways on RAOSMC when it was treated in two different ways-cotreatment with PDGF-bb and pretreatment of EGCG before addition of PDGF-bb. Both cotreated and pretreated EGCG significantly inhibited PDGF-bb induced proliferation, cell cycle progression of the G0/G1 phase, and the gelatinolytic activity of MMP-2/9 on RAOSMC. Also, EGCG blocked PDGF receptor-β (PDGFR-β) phosphorylation on PDGF-bb stimulated RAOSMC under pretreatment with cells as well as cotreatment with PDGF-bb. The downstream signal transduction pathways of PDGFR-β, including p42/44 MAPK, p38 MAPK, and Akt phosphorylation, were also inhibited by EGCG in a pattern similar to PDGFR-β phosphorylation. These findings suggest that EGCG can inhibit PDGF-bb stimulated mitogenesis by indirectly and directly interrupting PDGF-bb signals and blocking the signaling pathway via PDGFR-β phosphorylation. Furthermore, EGCG may be used for treatment and prevention of cardiovascular disease through blocking of PDGF-bb signaling.
Collapse
|
26
|
Foster JS, Fish LM, Phipps JE, Bruker CT, Lewis JM, Bell JL, Solomon A, Kestler DP. Odontogenic ameloblast-associated protein (ODAM) inhibits growth and migration of human melanoma cells and elicits PTEN elevation and inactivation of PI3K/AKT signaling. BMC Cancer 2013; 13:227. [PMID: 23648148 PMCID: PMC3651709 DOI: 10.1186/1471-2407-13-227] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 04/25/2013] [Indexed: 12/17/2022] Open
Abstract
Background The Odontogenic Ameloblast-associated Protein (ODAM) is expressed in a wide range of normal epithelial, and neoplastic tissues, and we have posited that ODAM serves as a novel prognostic biomarker for breast cancer and melanoma. Transfection of ODAM into breast cancer cells yields suppression of cellular growth, motility, and in vivo tumorigenicity. Herein we have extended these studies to the effects of ODAM on cultured melanoma cell lines. Methods The A375 and C8161 melanoma cell lines were stably transfected with ODAM and assayed for properties associated with tumorigenicity including cell growth, motility, and extracellular matrix adhesion. In addition, ODAM–transfected cells were assayed for signal transduction via AKT which promotes cell proliferation and survival in many neoplasms. Results ODAM expression in A375 and C8161 cells strongly inhibited cell growth and motility in vitro, increased cell adhesion to extracellular matrix, and yielded significant cytoskeletal/morphologic rearrangement. Furthermore, AKT activity was downregulated by ODAM expression while an increase was noted in expression of the PTEN (phosphatase and tensin homolog on chromosome 10) tumor suppressor gene, an antagonist of AKT activation. Increased PTEN in ODAM-expressing cells was associated with increases in PTEN mRNA levels and de novo protein synthesis. Silencing of PTEN expression yielded recovery of AKT activity in ODAM-expressing melanoma cells. Similar PTEN elevation and inhibition of AKT by ODAM was observed in MDA-MB-231 breast cancer cells while ODAM expression had no effect in PTEN-deficient BT-549 breast cancer cells. Conclusions The apparent anti-neoplastic effects of ODAM in cultured melanoma and breast cancer cells are associated with increased PTEN expression, and suppression of AKT activity. This association should serve to clarify the clinical import of ODAM expression and any role it may serve as an indicator of tumor behavior.
Collapse
Affiliation(s)
- James S Foster
- Department of Medicine, Human Immunology and Cancer Program, University of Tennessee Health Sciences Center-Knoxville, Knoxville, TN 37920, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Strauch AR, Hariharan S. Dynamic Interplay of Smooth Muscle α-Actin Gene-Regulatory Proteins Reflects the Biological Complexity of Myofibroblast Differentiation. BIOLOGY 2013; 2:555-86. [PMID: 24832798 PMCID: PMC3960882 DOI: 10.3390/biology2020555] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 03/01/2013] [Accepted: 03/06/2013] [Indexed: 01/06/2023]
Abstract
Myofibroblasts (MFBs) are smooth muscle-like cells that provide contractile force required for tissue repair during wound healing. The leading agonist for MFB differentiation is transforming growth factor β1 (TGFβ1) that induces transcription of genes encoding smooth muscle α-actin (SMαA) and interstitial collagen that are markers for MFB differentiation. TGFβ1 augments activation of Smad transcription factors, pro-survival Akt kinase, and p38 MAP kinase as well as Wingless/int (Wnt) developmental signaling. These actions conspire to activate β-catenin needed for expression of cyclin D, laminin, fibronectin, and metalloproteinases that aid in repairing epithelial cells and their associated basement membranes. Importantly, β-catenin also provides a feed-forward stimulus that amplifies local TGFβ1 autocrine/paracrine signaling causing transition of mesenchymal stromal cells, pericytes, and epithelial cells into contractile MFBs. Complex, mutually interactive mechanisms have evolved that permit several mammalian cell types to activate the SMαA promoter and undergo MFB differentiation. These molecular controls will be reviewed with an emphasis on the dynamic interplay between serum response factor, TGFβ1-activated Smads, Wnt-activated β-catenin, p38/calcium-activated NFAT protein, and the RNA-binding proteins, Purα, Purβ, and YB-1, in governing transcriptional and translational control of the SMαA gene in injury-activated MFBs.
Collapse
Affiliation(s)
- Arthur Roger Strauch
- Department of Physiology & Cell Biology and the Ohio State Biochemistry Program, the Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA.
| | - Seethalakshmi Hariharan
- Department of Physiology & Cell Biology and the Ohio State Biochemistry Program, the Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA.
| |
Collapse
|
28
|
Weiss GA, Rossi MR, Khushalani NI, Lo K, Gibbs JF, Bharthuar A, Cowell JK, Iyer R. Evaluation of phosphatidylinositol-3-kinase catalytic subunit (PIK3CA) and epidermal growth factor receptor (EGFR) gene mutations in pancreaticobiliary adenocarcinoma. J Gastrointest Oncol 2013; 4:20-9. [PMID: 23450128 PMCID: PMC3562624 DOI: 10.3978/j.issn.2078-6891.2012.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2012] [Accepted: 02/21/2012] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Phosphatidylinositol-3-kinase (PI3K) activation involves the epidermal growth factor receptor (EGFR) and plays an important role in cell survival signaling in pancreaticobiliary cancer. EGFR gene mutations have been correlated with clinical response to EGFR inhibitors in patients with advanced non-small cell lung cancer. This study examined the prevalence of PIK3CA and EGFR mutations in pancreaticobiliary cancer where erlotinib, an EGFR inhibitor, is approved for therapy. METHODS Thirty patients who underwent pancreatectomy for pancreaticobiliary carcinoma were identified. Genomic DNA was extracted from formalin fixed paraffin embedded tumor and adjacent normal tissue, and exons 9 and 20 (for the PIK3CA gene) and exons 18-21 (for the EGFR gene) were amplified by PCR and sequenced. Literature review on EGFR and/or PIK3CA mutations in pancreaticobiliary adenocarcinomas was conducted. RESULTS No mutations in either PIK3CA or EGFR genes were identified. The study identified one synonymous single nucleotide polymorphism (SNP) (rs1050171) in the coding region of EGFR. A previously unreported change, suspected to be a SNP, was observed in intron 18 of EGFR (IVS18+15, C>T). Review of the literature showed EGFR mutation rate of 2% and 10.5% in pancreatic and biliary tract carcinomas, respectively. PIK3CA mutations were found in 3.6% and 11.7% of pancreatic and biliary tract carcinomas, respectively. CONCLUSIONS A low prevalence of EGFR or PIK3CA mutations exists in pancreatic cancer (<5%), indicating that mutation screening may not be as useful in determining prognosis or response to targeted inhibition.
Collapse
Affiliation(s)
- Guy A. Weiss
- Department of Medicine, University at Buffalo, Buffalo, New York, USA
| | - Michael R. Rossi
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | - Ken Lo
- Epigenetics, Roche Nimblegen, Inc., Madison, Wisconsin, USA
| | | | | | - John K. Cowell
- GHSU Cancer Center, Medical College of Georgia, Georgia Health Sciences University, Augusta, Georgia, USA
| | - Renuka Iyer
- Roswell Park Cancer Inst, Buffalo, New York, USA
| |
Collapse
|
29
|
Takano N, Tsuruma K, Ohno Y, Shimazawa M, Hara H. Bimatoprost protects retinal neuronal damage via Akt pathway. Eur J Pharmacol 2013; 702:56-61. [PMID: 23395963 DOI: 10.1016/j.ejphar.2013.01.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 01/17/2013] [Accepted: 01/29/2013] [Indexed: 12/27/2022]
Abstract
Worldwide, prostaglandin analogs, such as bimatoprost, have become the major therapeutic class for medical treatment of glaucoma because of their efficacy and generally well tolerated systemic safety profile. However, the detailed mechanism of the direct action of bimatoprost on retinal ganglion cells (RGC) has rarely been understood. Thus, in this study, we elucidated the mechanism of the protective effects of bimatoprost on RGC against oxidative stress. To examine the protective effects of bimatoprost, cultured RGC with various concentrations of bimatoprost (in both free acid and amide form) were exposed to l-buthionin-(S,R)-sulfoximine (BSO) plus glutamate or serum depletion in vitro and intravitreal injection of N-methyl-D-aspartate (NMDA) was used to induce retinal damage in vivo. To elucidate the protective mechanism of bimatoprost, we used western blot analysis to investigate the phosphorylation of Akt and extracellular signal-regulated kinase (ERK). Bimatoprost significantly reduced BSO plus glutamate- and serum deprivation-induced death in concentration-dependent manners. Bimatoprost induced activation of Akt and ERK, and a phosphatidylinositol 3-kinase inhibitor, LY294002, attenuated the protective effect of bimatoprost. On the other hand, a mitogen-activated protein kinase kinase inhibitor, U0126, exhibited protective effect unexpectedly. Moreover, ERK was more phosphorylated by attenuation of Akt activity in cultured RGC. In an in vivo study, bimatoprost reduced NMDA-induced RGC death. Taken together, these findings indicate that bimatoprost has protective effects on in vitro and in vivo retinal damage, suggesting that the mechanism underlying may be via the Akt pathway, which may modulate the ERK pathway.
Collapse
Affiliation(s)
- Norihito Takano
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan
| | | | | | | | | |
Collapse
|
30
|
Deng L, Shi B, Zhuang Y, Chu J, Shi X, Zhang S, Guo M. Performance and mechanism of neuroleukin in the growth and survival of sertoli cell-induced neurons in a coculture system. Cell Transplant 2013; 23:381-94. [PMID: 23394468 DOI: 10.3727/096368913x663578] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Sertoli cells (SCs), which are recognized as the "nurse cells" of the testis due to their important biofunctions, have been used in cotransplantation with neurons in cell therapy. However, it is not clear whether SCs influence neuronal communication and survival. In this study, we showed that approximately 60% of cortical neural stem cells (NSCs) cocultured with SCs differentiated into mature neurons. In addition, the neurite outgrowth and neuronal survival rates were significantly enhanced in the coculture system compared with differentiated neurons induced by a differentiation medium. The neuroleukin (NLK) secretion of SCs was also identified at the RNA and protein level, and the roles of NLK in neuromorphology and physiological regulation were systematically investigated for the first time. These results not only highlight the significance of paracrine regulation of NSCs by SCs but also confirm the role NLK plays in the differentiation and survival of NSCs. Finally, we proposed a possible hypothesis for the mechanism of NLK in the growth and survival of SC-induced neurons based on Western blotting results, which is that NLK secreted by SCs activates the Ras/Raf/MEK/Erk, Jak/Stat, and PI3K/Akt pathways, but not the NF-κB pathway, in neurons resulting in their growth and survival.
Collapse
Affiliation(s)
- Lei Deng
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, P.R. China
| | | | | | | | | | | | | |
Collapse
|
31
|
Benelli R, Venè R, Minghelli S, Carlone S, Gatteschi B, Ferrari N. Celecoxib induces proliferation and Amphiregulin production in colon subepithelial myofibroblasts, activating erk1–2 signaling in synergy with EGFR. Cancer Lett 2013; 328:73-82. [DOI: 10.1016/j.canlet.2012.09.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Revised: 09/12/2012] [Accepted: 09/12/2012] [Indexed: 02/04/2023]
|
32
|
Huang W, Zhao Y, Zhu X, Cai Z, Wang S, Yao S, Qi Z, Xie P. Fluoxetine upregulates phosphorylated-AKT and phosphorylated-ERK1/2 proteins in neural stem cells: evidence for a crosstalk between AKT and ERK1/2 pathways. J Mol Neurosci 2012; 49:244-9. [PMID: 22674052 DOI: 10.1007/s12031-012-9822-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 05/24/2012] [Indexed: 01/17/2023]
Abstract
Fluoxetine is a widely used antidepressant drug which inhibits the reuptake of serotonin in the central nervous system (CNS). Recent studies have shown that fluoxetine can promote neurogenesis and improve the survival rate of neurons. However, whether fluoxetine modulates the neuroprotection of neural stem cells (NSCs) needs to be elucidated. In this study, we demonstrated that 50 μM fluoxetine significantly upregulated expression of the phosphorylated-AKT and ERK1/2 proteins in NSCs derived from rats. Besides, expression of phosphorylated-AKT and phosphorylated-ERK1/2 in fluoxetine-treated NSCs was effectively blocked (P<0.05) by both PI3-K inhibitor (LY294002) and MEK inhibitor (PD98059). It was, therefore, concluded that the crosstalk between PI3K/AKT and MAPK/ERK pathways involved AKT and ERK1/2 phosphorylation by fluoxetine treatment. This study points to a novel role of fluoxetine in neuroprotection as an antidepressant drug and also unravels the crosstalk mechanism between the two signaling pathways.
Collapse
Affiliation(s)
- Wen Huang
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Banerjee A, Panettieri R. Vitamin D modulates airway smooth muscle function in COPD. Curr Opin Pharmacol 2012; 12:266-74. [PMID: 22365730 DOI: 10.1016/j.coph.2012.01.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Revised: 01/24/2012] [Accepted: 01/26/2012] [Indexed: 11/30/2022]
Abstract
COPD is a disease manifested as persistent airflow obstruction with an enhanced inflammatory response in the airways and lungs to noxious particles and gases which evokes symptoms of dyspnea on exertion, cough and mucus production. Airway smooth muscle plays a central role in the COPD diathesis and is implicated in many aspects of COPD pathogenesis. Vitamin D deficiency has been associated with COPD severity and studies suggest a role for Vitamin D as a treatment for COPD. In this review, we describe the effects of 1,25-dihydroxyvitamin D on airway smooth muscle function, including agonist-induced shortening, secretion of inflammatory mediators, and myocyte hypertrophy and hyperplasia.
Collapse
Affiliation(s)
- Audreesh Banerjee
- Department of Medicine, Airways Biology Initiative, Division of Pulmonary, Allergy and Critical Care Medicine, Hospital of University of Pennsylvania, Philadelphia, PA, United States
| | | |
Collapse
|
34
|
Harbrecht BG, Nweze I, Smith JW, Zhang B. Insulin inhibits hepatocyte iNOS expression induced by cytokines by an Akt-dependent mechanism. Am J Physiol Gastrointest Liver Physiol 2012; 302:G116-22. [PMID: 22038823 PMCID: PMC3345958 DOI: 10.1152/ajpgi.00114.2011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Hepatocyte inducible nitric oxide synthese (iNOS) expression is a tightly controlled pathway that mediates hepatic inflammation and hepatocyte injury in a variety of disease states. We have shown that cyclic adenosine monophosphate (cAMP) regulates cytokine-induced hepatocyte iNOS expression through mechanisms that involve protein kinase B/Akt. We hypothesized that insulin, which activates Akt signaling in hepatocytes, as well as signaling through p38 and MAPK p42/p44, would regulate iNOS expression during inflammation. In primary rat hepatocytes, insulin inhibited cytokine-stimulated nitrite accumulation and iNOS expression in a dose-dependent manner. Inhibition of MAPK p42/p44 with PD98059 had no effect on iNOS activation, whereas SB203580 to block p38 reversed insulin's inhibitory effect. However, insulin did not increase p38 activation and inhibition of p38 signaling with a dominant negative p38 plasmid had no effect on cytokine- or insulin-mediated effects on iNOS. We found that SB203580 blocked insulin-induced Akt activation. Inhibition of Akt signaling with LY294002 or a dominant negative Akt plasmid increased cytokine-stimulated nitrite production and iNOS protein expression and blocked the inhibitory effects of insulin. NF-κB induces iNOS expression and can be regulated by Akt, but insulin had no effect on cytokine-mediated IκBα levels or NF-κB p65 translocation. Our data demonstrate that insulin inhibits cytokine-stimulated hepatocyte iNOS expression and does so through effects on Akt-mediated signaling.
Collapse
Affiliation(s)
- Brian G. Harbrecht
- Department of Surgery and the Price Institute for Surgical Research, University of Louisville School of Medicine, Louisville, Kentucky
| | - Ikenna Nweze
- Department of Surgery and the Price Institute for Surgical Research, University of Louisville School of Medicine, Louisville, Kentucky
| | - Jason W. Smith
- Department of Surgery and the Price Institute for Surgical Research, University of Louisville School of Medicine, Louisville, Kentucky
| | - Baochun Zhang
- Department of Surgery and the Price Institute for Surgical Research, University of Louisville School of Medicine, Louisville, Kentucky
| |
Collapse
|
35
|
Castellano E, Downward J. RAS Interaction with PI3K: More Than Just Another Effector Pathway. Genes Cancer 2011; 2:261-74. [PMID: 21779497 DOI: 10.1177/1947601911408079] [Citation(s) in RCA: 519] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
RAS PROTEINS ARE SMALL GTPASES KNOWN FOR THEIR INVOLVEMENT IN ONCOGENESIS: around 25% of human tumors present mutations in a member of this family. RAS operates in a complex signaling network with multiple activators and effectors, which allows them to regulate many cellular functions such as cell proliferation, differentiation, apoptosis, and senescence. Phosphatidylinositol 3-kinase (PI3K) is one of the main effector pathways of RAS, regulating cell growth, cell cycle entry, cell survival, cytoskeleton reorganization, and metabolism. However, it is the involvement of this pathway in human tumors that has attracted most attention. PI3K has proven to be necessary for RAS-induced transformation in vitro, and more importantly, mice with mutations in the PI3K catalytic subunit p110α that block its ability to interact with RAS are highly resistant to endogenous oncogenic KRAS-induced lung tumorigenesis and HRAS-induced skin carcinogenesis. These animals also have a delayed development of the lymphatic vasculature. Many PI3K inhibitors have been developed that are now in clinical trials. However, it is a complex pathway with many feedback loops, and interactions with other pathways make the results of its inhibition hard to predict. Combined therapy with another RAS-regulated pathway such as RAF/MEK/ERK may be the most effective way to treat cancer, at least in animal models mimicking the human disease. In this review, we will summarize current knowledge about how RAS regulates one of its best-known effectors, PI3K.
Collapse
Affiliation(s)
- Esther Castellano
- Signal Transduction Laboratory, Cancer Research UK London Research Institute, London, UK
| | | |
Collapse
|
36
|
Wang Y, Xing KY, Lou MF. Regulation of cytosolic phospholipase A2 (cPLA2alpha) and its association with cell proliferation in human lens epithelial cells. Invest Ophthalmol Vis Sci 2011; 52:8231-40. [PMID: 21896865 PMCID: PMC3208028 DOI: 10.1167/iovs.11-7542] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Revised: 07/25/2011] [Accepted: 08/18/2011] [Indexed: 12/13/2022] Open
Abstract
PURPOSE To investigate the molecular mechanism for cytosolic phospholipase A2 (cPLA(2)α) regulation and its association to platelet-derived growth factor (PDGF)-induced cell proliferation. METHODS cPLA(2)α was examined using human lens epithelial (HLE) B3 cells. Reactive oxygen species (ROS) generation induced by PDGF was analyzed by luminescence assay. Cell proliferation was measured by cell counting and by BrdU assay. Human cPLA(2)α gene was cloned via RT-PCR followed by site-directed mutagenesis to construct HLE B3 cells expressing either inactive cPLA(2)α enzyme with S228A mutation (S228A), or cPLA(2)α truncated at the calcium-binding C2 domain (C2D). Activity of cPLA(2)α was measured by arachidonic acid (AA) release from cell membranes using [(3)H]-arachidonic acid prelabeled cells. The effect of intracellular calcium level on cPLA(2)α function was examined by treating cells with ionomycin (calcium influx), thapsgargin (endoplasmic reticulum [ER] calcium store release) or 1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid tetrakis (BAPTA; calcium chelator). Activation of extracellular signal-regulated kinases (ERK), JNK, p38, or Akt was detected by Western blot analysis using specific antibodies. RESULTS S228A mutant showed suppressed PDGF-induced reactive oxygen species generation, ERK and JNK activation (no effect on p38 or Akt), and cell proliferation in comparison with the vector alone (Vec) control. Calcium-binding C2 domain cells lost the ability of membrane translocation and activation of cPLA(2)α. PDGF cell signaling was calcium-dependent, and the calcium was supplied either from the external flux or endoplasmic reticulum store. However, enrichment of cellular calcium not only augmented PDGF function, but also demonstrated a cPLA(2)α-dependent calcium-signaling cascade that led to cell proliferation. CONCLUSIONS cPLA(2)α is regulated by calcium mobilization and mitogen-activated protein kinases (MAPK) activation. Both PDGF mitogenic action and calcium signaling are cPLA(2)α-dependent.
Collapse
Affiliation(s)
- Yin Wang
- From the School of Veterinary Medicine and Biomedical Sciences, and
- the Redox Biology Center, University of Nebraska-Lincoln, Lincoln, Nebraska; and
| | - Kui-Yi Xing
- From the School of Veterinary Medicine and Biomedical Sciences, and
- the Redox Biology Center, University of Nebraska-Lincoln, Lincoln, Nebraska; and
| | - Marjorie F. Lou
- From the School of Veterinary Medicine and Biomedical Sciences, and
- the Redox Biology Center, University of Nebraska-Lincoln, Lincoln, Nebraska; and
- the Department of Ophthalmology, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
37
|
Chen WJ, Chen YH, Lin KH, Ting CH, Yeh YH. Cilostazol promotes vascular smooth muscles cell differentiation through the cAMP response element-binding protein-dependent pathway. Arterioscler Thromb Vasc Biol 2011; 31:2106-13. [PMID: 21680899 DOI: 10.1161/atvbaha.111.230987] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Cilostazol, a potent type 3 phosphodiesterase inhibitor, has recently been found to reduce neointimal formation by inhibiting vascular smooth muscle cell (VSMC) proliferation. The aim of this study is to investigate whether cilostazol exerts an action on phenotypic modulation of VSMCs, another important process in the pathogenesis of neointimal formation. METHODS AND RESULTS Cilostazol may convert VSMCs from a serum-induced dedifferentiation state to a differentiated state, as indicated by a spindle-shaped morphology and an increase in the expression of smooth muscle cell differentiation marker contractile proteins. The upregulation of contractile proteins by cilostazol involves the cAMP/protein kinase A (PKA) signaling pathway, because the cAMP analog mimicked and specific cAMP/PKA inhibitors opposed the effect of cilostazol. Furthermore, cilostazol-activated cAMP response element (CRE)-binding protein (CREB), including phosphorylation at Ser133 and its nuclear translocation. Deletion and mutational analysis of the contractile protein promoters along with chromatin immunoprecipitation using anti-CREB antibody showed that CRE is essential for cilostazol-induced contractile protein expression. Transfection of dominant-negative CREB (mutated Ser133) plasmid in VSMCs blocked cilostazol-stimulated contractile protein expression. In vivo, cilostazol upregulated contractile proteins and induced the activation of CREB in the neointima of balloon-injured arteries. CONCLUSIONS Cilostazol promotes VSMC differentiation through the cAMP/PKA/CREB signaling cascade.
Collapse
Affiliation(s)
- Wei-Jan Chen
- First Cardiovascular Division, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Tao-Yuan, Taiwan.
| | | | | | | | | |
Collapse
|
38
|
Li J, Liu J, Song J, Wang X, Weiss HL, Townsend CM, Gao T, Evers BM. mTORC1 inhibition increases neurotensin secretion and gene expression through activation of the MEK/ERK/c-Jun pathway in the human endocrine cell line BON. Am J Physiol Cell Physiol 2011; 301:C213-26. [PMID: 21508335 DOI: 10.1152/ajpcell.00067.2011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The mammalian target of rapamycin (mTOR) signaling exists in two complexes: mTORC1 and mTORC2. Neurotensin (NT), an intestinal hormone secreted by enteroendocrine (N) cells in the small bowel, has important physiological effects in the gastrointestinal tract. The human endocrine cell line BON abundantly expresses the NT gene and synthesizes and secretes NT in a manner analogous to that of N cells. Here, we demonstrate that the inhibition of mTORC1 by rapamycin (mTORC1 inhibitor), torin1 (both mTORC1 and mTORC2 inhibitor) or short hairpin RNA-mediated knockdown of mTOR, regulatory associated protein of mTOR (RAPTOR), and p70 S6 kinase (p70S6K) increased basal NT release via upregulating NT gene expression in BON cells. c-Jun activity was increased by rapamycin or torin1 or p70S6K knockdown. c-Jun overexpression dramatically increased NT promoter activity, which was blocked by PD98059, an mitogen-activated protein kinase kinase (MEK) inhibitor. Furthermore, overexpression of MEK1 or extracellular signal-regulated kinase 1 (ERK1) increased c-Jun expression and NT promoter activity. More importantly, PD98059 blocked rapamycin- or torin1-enhanced NT secretion. Consistently, rapamycin and torin1 also increased NT gene expression in Hep3B cells, a human hepatoma cell line that, similar to BON, expresses high levels of NT. Phosphorylation of c-Jun and ERK1/2 was also increased by rapamycin and torin1 in Hep3B cells. Finally, we showed activation of mTOR in BON cells treated with amino acids, high glucose, or serum and, concurrently, the attenuation of ERK1/2 and c-Jun phosphorylation and NT secretion. Together, mTORC1, as a nutrient sensor, negatively regulates NT secretion via the MEK/ERK/c-Jun signaling pathway. Our results identify a physiological link between mTORC1 and MEK/ERK signaling in controlling intestinal hormone gene expression and secretion.
Collapse
Affiliation(s)
- Jing Li
- Department of Surgery, University of Kentucky, Lexington, Kentucky 40536, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Muslin AJ. Akt2: a critical regulator of cardiomyocyte survival and metabolism. Pediatr Cardiol 2011; 32:317-22. [PMID: 21279637 DOI: 10.1007/s00246-010-9879-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 12/17/2010] [Indexed: 01/09/2023]
Abstract
Akt proteins are serine/threonine protein kinases that participate in several important intracellular signal transduction cascades. Akt1 and Akt2 are expressed in cardiomyocytes, and both are activated by the action of a variety of growth factors and extracellular ligands. In work with genetically modified mice that had targeted disruption of the genes encoding Akt1 or Akt2, findings showed that Akt1 specifically regulated the physiologic growth of cardiomyocytes that occurred in response to exercise training. In contrast, Akt2 does not regulate physiologic growth but instead regulates glucose metabolism in response to insulin stimulation in cardiomyocytes. Furthermore, Akt2 plays a critical role in antagonizing cardiomyocyte apoptosis that occurs in response to a variety of stimuli, including pathologic remodeling after experimental myocardial infarction. In addition, the protein tribbles 3 (TRB3), an Akt antagonist, was found to be expressed in cardiomyocytes and to be induced by stimuli that cause endoplasmic reticulum stress. Endoplasmic reticulum stress-mediated antagonism of Akt signaling in cardiomyocytes was dependent on TRB3 induction. Finally, myocardial infarction caused endoplasmic reticulum stress in the infarct border zone that was associated with TRB3 induction. These results demonstrate the differential roles of Akt family members and the importance of Akt2 in cardiomyocyte survival.
Collapse
Affiliation(s)
- Anthony J Muslin
- Center for Cardiovascular Research, Washington University School of Medicine, 660 South Euclid Avenue, Box 8086, St. Louis, MO 63110, USA.
| |
Collapse
|
40
|
Kittilson JD, Jones E, Sheridan MA. ERK, Akt, and STAT5 are Differentially Activated by the Two Growth Hormone Receptor Subtypes of a Teleost Fish (Oncorhynchus Mykiss). Front Endocrinol (Lausanne) 2011; 2:30. [PMID: 22649371 PMCID: PMC3355939 DOI: 10.3389/fendo.2011.00030] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 08/26/2011] [Indexed: 11/29/2022] Open
Abstract
Previously, we found that the teleost fish, rainbow trout, possesses two growth hormone receptor (GHR) subtypes that display distinct ligand-binding and agonist-induced regulation features. In this study, we used Chinese hamster ovary-K1 cells stably transfected individually with the two trout GHR subtypes, GHR1 and GHR2, to elucidate receptor-effector pathway linkages. Growth hormone (GH) stimulated rapid (5-10 min) phosphorylation of ERK, Akt, JAk2, and STAT5 in both GHR1- and GHR2-expressing cells; however; STAT5 was activated to a greater extent through GHR1 than through GHR2, whereas ERK and Akt were activated to a greater through GHR2 than through GHR1. Although blockade of the ERK pathway had no effect on the activation of Akt, inhibition of PI3K-Akt partially prevented activation of ERK, suggesting cross-talk between the ERK and PI3K-Akt pathways. JAK2 inhibition completely blocked activation of ERK, Akt, and STAT5, suggesting that all of these pathways link to GHR1 and GHR2 via JAK2. These findings establish important receptor-effector pathway linkages and suggest that the GHR subtypes of teleost fish may be functionally distinct.
Collapse
Affiliation(s)
| | - Evan Jones
- Department of Biological Sciences, North Dakota State UniversityFargo, ND, USA
| | - Mark A. Sheridan
- Department of Biological Sciences, North Dakota State UniversityFargo, ND, USA
- *Correspondence: Mark A. Sheridan, Department of Biological Sciences, North Dakota State University, P.O. Box 6050, Department 2715, Fargo, ND 58108-6050, USA. e-mail:
| |
Collapse
|
41
|
Zhang B, Li S, Harbrecht BG. Akt-mediated signaling is induced by cytokines and cyclic adenosine monophosphate and suppresses hepatocyte inducible nitric oxide synthase expression independent of MAPK P44/42. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2010; 1813:73-9. [PMID: 20934465 DOI: 10.1016/j.bbamcr.2010.10.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2009] [Revised: 09/29/2010] [Accepted: 10/01/2010] [Indexed: 11/26/2022]
Abstract
Cyclic AMP inhibits the expression of nitric oxide synthase (Harbrecht et al., 1995 [1]) in hepatocytes but the mechanism for this effect is incompletely understood. Cyclic AMP can activate several intracellular signaling pathways in hepatocytes including Protein Kinase A (PKA), cAMP regulated guanine nucleotide exchange factors (cAMP-GEFs), and calcium-mediated Protein Kinases. There is considerable overlap and cross-talk between many of these signaling pathways, however, and how these cascades regulate hepatocyte iNOS is not known. We hypothesized that Akt mediates the effect of cAMP on hepatocyte iNOS expression. Hepatocytes cultured with cytokines and dbcAMP increased Akt phosphorylation up to 2h of culture. Akt phosphorylation was inhibited by the PI3K inhibitor LY294002 (10μM), farnyltranferase inhibitor FTI-276, or transfection with a dominant negative Akt. The cyclic AMP-induced suppression of cytokine-stimulated iNOS was partially reversed by LY294002 and FTI-276. LY294002 also increased NFκB nucleus translocation by Western blot analysis in nuclear extracts. Cyclic AMP increased phosphorylation of Raf1 at serine 259 which was blocked by LY294002 and associated with decreased MAPK P44/42 phosphorylation. However, inhibition of MAPK P44/42 signaling with PD98059 failed to suppress cytokine-induced hepatocyte iNOS expression and did not enhance the inhibitory effect of dbcAMP on iNOS production. A constitutively active MAPK P44/42 plasmid had no effect on cytokine-stimulated NO production. These data demonstrate that dbcAMP regulates hepatocyte iNOS expression through an Akt-mediated signaling mechanism that is independent of MAPK P44/42.
Collapse
Affiliation(s)
- Baochun Zhang
- University of Louisville, Department of Surgery, Louisville, KY 40292, USA.
| | | | | |
Collapse
|
42
|
Yang XS, Liu S, Liu YJ, Liu JW, Liu TJ, Wang XQ, Yan Q. Overexpression of fucosyltransferase IV promotes A431 cell proliferation through activating MAPK and PI3K/Akt signaling pathways. J Cell Physiol 2010; 225:612-9. [PMID: 20506505 DOI: 10.1002/jcp.22250] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Lewis Y (LeY) is a carbohydrate tumor-asssociated antigen. The majority of cancer cells derived from epithelial tissue express LeY type difucosylated oligosaccharide. Fucosyltransferase IV (FUT4) is an essential enzyme that catalyzes the synthesis of LeY oligosaccharide. Our previous studies have shown that FUT4 overexpression promotes A431 cell proliferation, but the mechanism is still largely unknown. Herein, we investigated the role of the mitogen-activated protein kinases (MAPKs) and phosphoinositide-3 kinase (PI3K)/Akt signaling pathways on FUT4-induced cell proliferation. Results show that overexpression of FUT4 increases the phosphorylation of ERK1/2, p38 MAPK, and PI3K/Akt. Inhibitors of PI3K (LY294002 and Wortmannin) prevented the phosphorylation of ERK1/2, p38 MAPK, and Akt PI3K). Moreover, phosphorylation of Akt is abolished by inhibitors of ERK1/2 (PD98059) and p38 MAPK (SB203580). These data suggested that FUT4 not only activates MAPK and PI3K/Akt signals, but also promotes the crosstalk among these signaling pathways. In addition, FUT4-induced stimulation of cell proliferation correlates with increased cell cycle progression by promoting cells into S-phase. The mechanism involves in increased expression of cyclin D1, cyclin E, CDK 2, CDK 4, and pRb, and decreased level of cyclin-dependent kinases inhibitors p21 and p27, which are blocked by the inhibitors of upstream signal molecules, MAPK and PI3K/Akt. In conclusion, these studies suggest that FUT4 regulates A431 cell growth through controlling cell cycle progression via MAPK and PI3K/Akt signaling pathways.
Collapse
Affiliation(s)
- Xue-Song Yang
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
43
|
Shear stress activates Akt during vascular smooth muscle cell reorientation. Biotechnol Appl Biochem 2010; 55:85-90. [PMID: 20055757 DOI: 10.1042/ba20090258] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Vascular intervention procedures can lead to endothelial damage and expose the underlying VSMCs (vascular smooth muscle cells) to shear stress. Although shear stress has been implicated in the proliferation and migration of VSMCs, the molecular mechanism(s) underlying these events are not well understood. In the present study, we examined the effect of shear stress on VSMC reorientation and the activation of Akt (also called protein kinase B) pathway signalling. Cells were subjected to a shear of 9.8 dynes/cm2 (1 dyne=10-5 N) for 0 min, 5 min, 15 min, 30 min, 1 h, 4 h and 24 h. Shear stress caused the VSMCs to realign at an angle that was approximately 45 degrees relative to the shear force vector after 24 h. Immunoblotting demonstrated that the phosphorylations of Akt and Akt-related signalling proteins [mTOR (mammalian target of rapamycin), PTEN (phosphatase and tensin homologue deleted on chromosome 10) and p70S6k (p70 S6-kinase)] were increased after shear stimulation. These results indicate that the activation of the Akt pathway signalling is closely correlated with shear-induced VSMC reorientation.
Collapse
|
44
|
Castellano E, Downward J. Role of RAS in the regulation of PI 3-kinase. Curr Top Microbiol Immunol 2010; 346:143-69. [PMID: 20563706 DOI: 10.1007/82_2010_56] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Ras proteins are key regulators of signalling cascades, controlling many processes such as proliferation, differentiation and apoptosis. Mutations in these proteins or in their effectors, activators and regulators are associated with pathological conditions, particularly the development of various forms of human cancer. RAS proteins signal through direct interaction with a number of effector enzymes, one of the best characterized being type I phosphatidylinositol (PI) 3-kinases. Although the ability of RAS to control PI 3-kinase has long been well established in cultured cells, evidence for a role of the interaction of endogenous RAS with PI 3-kinase in normal and malignant cell growth in vivo has only been obtained recently. Mice with mutations in the PI 3-kinase catalytic p110a isoform that block its ability to interact with RAS are highly resistant to endogenous KRAS oncogene induced lung tumourigenesis and HRAS oncogene induced skin carcinogenesis. Cells from these mice show proliferative defects and selective disruption of signalling from certain growth factors to PI 3-kinase, while the mice also display delayed development of the lymphatic vasculature. The interaction of RAS with p110a is thus required in vivo for some normal growth factor signalling and also for RAS-driven tumour formation. RAS family members were among the first oncogenes identified over 40 years ago. In the late 1960s, the rat-derived Harvey and Kirsten murine sarcoma retroviruses were discovered and subsequently shown to promote cancer formation through related oncogenes, termed RAS (from rat sarcoma virus). The central role of RAS proteins in human cancer is highlighted by the large number of tumours in which they are activated by mutation: approximately 20% of human cancers carry a mutation in RAS proteins. Because of the complex signalling network in which RAS operates, with multiple activators and effectors, each with a different pattern of tissue-specific expression and a distinct set of intracellular functions, one of the critical issues concerns the specific role of each effector in RAS-driven oncogenesis. In this chapter, we summarize current knowledge about how RAS regulates one of its best-known effectors, phosphoinositide 3-kinase (PI3K).
Collapse
Affiliation(s)
- Esther Castellano
- Signal Transduction Laboratory, Cancer Research UK London Research Institute, London, WC2A 3PX, UK
| | | |
Collapse
|
45
|
Torella D, Gasparri C, Ellison GM, Curcio A, Leone A, Vicinanza C, Galuppo V, Mendicino I, Sacco W, Aquila I, Surace FC, Luposella M, Stillo G, Agosti V, Cosentino C, Avvedimento EV, Indolfi C. Differential regulation of vascular smooth muscle and endothelial cell proliferation in vitro and in vivo by cAMP/PKA-activated p85alphaPI3K. Am J Physiol Heart Circ Physiol 2009; 297:H2015-25. [PMID: 19783773 DOI: 10.1152/ajpheart.00738.2009] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
cAMP inhibits proliferation in most cell types, triggering different and sometimes opposing molecular pathways. p85alpha (phosphatidylinositol 3-kinase regulatory subunit) is phosphorylated by cAMP/PKA in certain cell lineages, but its effects on vascular smooth muscle cells (VSMCs) and endothelial cells (ECs) are unknown. In the present study, we evaluated 1) the role of p85alpha in the integration of cAMP/PKA-dependent signaling on the regulation of VSMC and EC growth in vitro; and 2) the effects of PKA-modified p85alpha on neointimal hyperplasia and endothelial healing after balloon injury in vivo. Plasmid constructs carrying wild-type and PKA-modified p85alpha were employed in VSMCs and ECs in vitro and after balloon injury in rat carotid arteries in vivo. cAMP/PKA reduced VSMC proliferation through p85alpha phosphorylation. Transfected PKA-activated p85alpha binds p21ras, reducing ERK1/2 activation and VSMC proliferation in vitro. In contrast, EC proliferation inhibition by cAMP is independent from PKA modification of p85alpha and ERK1/2 inhibition; indeed, PKA-activated p85alpha did not inhibit per se ERK1/2 activation and proliferation in ECs in vitro. Interestingly, cAMP reduced both VSMC and EC apoptotic death through p85alpha phosphorylation. Accordingly, PKA-activated p85alpha triggered Akt activation, reducing both VSMC and EC apoptosis in vitro. Finally, compared with controls, vascular gene transfer of PKA-activated p85alpha significantly reduced neointimal formation after balloon injury in rats, without inhibiting endothelial regeneration of the injured arterial segment. In conclusions, PKA-activated p85alpha integrates cAMP/PKA signaling differently in VSMCs and ECs. By reducing neointimal hyperplasia without inhibiting endothelial regeneration, it exerts a protective effect against restenosis after balloon injury.
Collapse
Affiliation(s)
- Daniele Torella
- Laboratory of Molecular and Cellular Cardiology, Magna Graecia University, Campus S. Venuta, Viale Europa-Germaneto, Catanzaro 88100, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abkhezr M, Keramati AR, Ostad SN, Davoodi J, Ghahremani MH. The time course of Akt and ERK activation on XIAP expression in HEK 293 cell line. Mol Biol Rep 2009; 37:2037-42. [PMID: 19649722 DOI: 10.1007/s11033-009-9658-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2009] [Accepted: 07/21/2009] [Indexed: 10/20/2022]
Abstract
The cell growth is controlled by the interaction of survival and cell growth arrest pathways as well as apoptosis mechanisms which determine the outcome of cell faith as proliferation or apoptosis. In this study, we have studied the activity of survival pathways, i.e., Akt and ERK1/2 with regard to XIAP (inhibitor of apoptosis) in serum starved and stimulated conditions. The HEK-293 cells were cultured in RPMI + 10% FBS. The cells were serum starved by switching to medium with 1% FBS for 24 h and serum stimulated by changing the medium to 10% FBS following serum starvation. The expression of p-Akt, p-ERK, Akt, ERK and XIAP was studied in various time points using western blot. The apoptosis was evaluated by DNA condensation using Hoechst 33258 and Caspase-3 assay. In serum starved condition expression of p-Akt and XIAP is very low. Serum stimulation increases p-Akt and p-ERK within 5 min and sustains a high level for 30 min. The expression of total Akt and ERK1/2 has not changed significantly for 24 h. XIAP expression starts at 6 h after serum stimulation, reaches to maximum level at 12 h and decreases to baseline within 24 h. Furthermore, serum starvation for 24 h does not induced apoptosis and DNA condensation. Taken together, the results indicate that serum activates Akt and ERK pathways earlier than XIAP expression. Furthermore, XIAP expression is low in serum starvation unlike p-ERK which suggests a survival role for ERK in serums starvation. The expression pattern of XIAP indicates induction by Akt and/or ERK activation which requires further studies.
Collapse
Affiliation(s)
- Mousa Abkhezr
- Department of Pharmacology-Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran University, Tehran, Iran
| | | | | | | | | |
Collapse
|
47
|
Abstract
The EL4 murine lymphoma cell line exists in variant phenotypes that differ with respect to responses to the tumor promoter phorbol 12-myristate 13-acetate (PMA1). Previous work showed that "PMA-sensitive" cells, characterized by a high magnitude of PMA-induced Erk activation, express RasGRP, a phorbol ester receptor that directly activates Ras. In "PMA-resistant" and "intermediate" EL4 cell lines, PMA induces Erk activation to lesser extents, but with a greater response in intermediate cells. In the current study, these cell lines were used to examine mechanisms of Raf-1 modulation. Phospho-specific antibodies were utilized to define patterns and kinetics of Raf-1 phosphorylation on several sites. Further studies showed that Akt is constitutively activated to a greater extent in PMA-resistant than in PMA-sensitive cells, and also to a greater extent in resistant than intermediate cells. Akt negatively regulates Raf-1 activation (Ser259), partially explaining the difference between resistant and intermediate cells. Erk activation exerts negative feedback on Raf-1 (Ser289/296/301), thus resulting in earlier termination of the signal in cells with a higher level of Erk activation. RKIP, a Raf inhibitory protein, is expressed at higher levels in resistant cells than in sensitive or intermediate cells. Knockdown of RKIP increases Erk activation and also negative feedback. In conclusion, this study delineates Raf-1 phosphorylation events occurring in response to PMA in cell lines with different extents of Erk activation. Variations in the levels of expression and activation of multiple signaling proteins work in an integrated fashion to modulate the extent and duration of Erk activation.
Collapse
Affiliation(s)
- Shujie Han
- Department of Pharmaceutical Sciences, Washington State University, Pullman, Washington 99164-6534, United States
| | | |
Collapse
|
48
|
Hatakeyama N, Kojima T, Iba K, Murata M, Thi MM, Spray DC, Osanai M, Chiba H, Ishiai S, Yamashita T, Sawada N. IGF-I regulates tight-junction protein claudin-1 during differentiation of osteoblast-like MC3T3-E1 cells via a MAP-kinase pathway. Cell Tissue Res 2008; 334:243-54. [PMID: 18855015 PMCID: PMC2692508 DOI: 10.1007/s00441-008-0690-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2008] [Accepted: 08/27/2008] [Indexed: 12/18/2022]
Abstract
Insulin-like growth factor I (IGF-I) is expressed in many tissues, including bone, and acts on the proliferation and differentiation of osteoblasts as an autocrine/paracrine regulator. Tight-junction proteins have been detected in osteoblasts, and direct cell-to-cell interactions may modulate osteoblast function with respect, for example, to gap junctions. In order to investigate the regulation of expression of tight-junction molecules and of function during bone differentiation, osteoblast-like MC3T3-E1 cells and osteocyte-like MLO-Y4 cells were treated with IGF-I. In both MC3T3-E1 cells and MLO-Y4 cells, the tight-junction molecules occludin, claudin-1, -2, and -6, and the gap-junction molecule connexin 43 (Cx43) were detected by reverse transcription with polymerase chain reaction. In MC3T3-E1 cells but not MLO-Y4 cells, mRNAs of claudin-1, -2, and -6, Cx43, and type I collagen, and proteins of claudin-1 and Cx43 were increased after treatment with IGF-I. Such treatment significantly decreased paracellular permeability in MC3T3-E1 cells. The expression of claudin-1 in MC3T3-E1 cells after IGF-I treatment was mainly upregulated via a mitogen-activated protein (MAP)-kinase pathway and, in part, modulated by a PI3-kinase pathway, whereas Cx43 expression and the mediated gap-junctional intercellular communication protein did not contribute to the upregulation. Furthermore, in MC3T3-E1 cells during wound healing, upregulation of claudin-1 was observed together with an increase of IGF-I and type I collagen. These findings suggest that the induction of tight-junction protein claudin-1 and paracellular permeability during the differentiation of osteoblast-like MC3T3-E1 cells after treatment with IGF-I is regulated via a MAP-kinase pathway, but not with respect to gap junctions.
Collapse
Affiliation(s)
- Naoko Hatakeyama
- N. Hatakeyama, K. Iba, T. Yamashita, Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, S1, W17, Sapporo 060-8556, Japan
- N. Hatakeyama, T. Kojima, M. Murata, M. Osanai, H. Chiba, N. Sawada, Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan, e-mail:
- M. M. Thi, D. C. Spray, Department of Neuroscience, Albert Einstein College of Medicine, Bronx, N.Y., USA
- S. Ishiai, Department of Rehabilitation, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takashi Kojima
- N. Hatakeyama, K. Iba, T. Yamashita, Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, S1, W17, Sapporo 060-8556, Japan
- N. Hatakeyama, T. Kojima, M. Murata, M. Osanai, H. Chiba, N. Sawada, Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan, e-mail:
- M. M. Thi, D. C. Spray, Department of Neuroscience, Albert Einstein College of Medicine, Bronx, N.Y., USA
- S. Ishiai, Department of Rehabilitation, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kousuke Iba
- N. Hatakeyama, K. Iba, T. Yamashita, Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, S1, W17, Sapporo 060-8556, Japan
- N. Hatakeyama, T. Kojima, M. Murata, M. Osanai, H. Chiba, N. Sawada, Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan, e-mail:
- M. M. Thi, D. C. Spray, Department of Neuroscience, Albert Einstein College of Medicine, Bronx, N.Y., USA
- S. Ishiai, Department of Rehabilitation, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masaki Murata
- N. Hatakeyama, K. Iba, T. Yamashita, Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, S1, W17, Sapporo 060-8556, Japan
- N. Hatakeyama, T. Kojima, M. Murata, M. Osanai, H. Chiba, N. Sawada, Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan, e-mail:
- M. M. Thi, D. C. Spray, Department of Neuroscience, Albert Einstein College of Medicine, Bronx, N.Y., USA
- S. Ishiai, Department of Rehabilitation, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Mia M. Thi
- N. Hatakeyama, K. Iba, T. Yamashita, Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, S1, W17, Sapporo 060-8556, Japan
- N. Hatakeyama, T. Kojima, M. Murata, M. Osanai, H. Chiba, N. Sawada, Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan, e-mail:
- M. M. Thi, D. C. Spray, Department of Neuroscience, Albert Einstein College of Medicine, Bronx, N.Y., USA
- S. Ishiai, Department of Rehabilitation, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - David C. Spray
- N. Hatakeyama, K. Iba, T. Yamashita, Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, S1, W17, Sapporo 060-8556, Japan
- N. Hatakeyama, T. Kojima, M. Murata, M. Osanai, H. Chiba, N. Sawada, Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan, e-mail:
- M. M. Thi, D. C. Spray, Department of Neuroscience, Albert Einstein College of Medicine, Bronx, N.Y., USA
- S. Ishiai, Department of Rehabilitation, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Makoto Osanai
- N. Hatakeyama, K. Iba, T. Yamashita, Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, S1, W17, Sapporo 060-8556, Japan
- N. Hatakeyama, T. Kojima, M. Murata, M. Osanai, H. Chiba, N. Sawada, Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan, e-mail:
- M. M. Thi, D. C. Spray, Department of Neuroscience, Albert Einstein College of Medicine, Bronx, N.Y., USA
- S. Ishiai, Department of Rehabilitation, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hideki Chiba
- N. Hatakeyama, K. Iba, T. Yamashita, Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, S1, W17, Sapporo 060-8556, Japan
- N. Hatakeyama, T. Kojima, M. Murata, M. Osanai, H. Chiba, N. Sawada, Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan, e-mail:
- M. M. Thi, D. C. Spray, Department of Neuroscience, Albert Einstein College of Medicine, Bronx, N.Y., USA
- S. Ishiai, Department of Rehabilitation, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Sumio Ishiai
- N. Hatakeyama, K. Iba, T. Yamashita, Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, S1, W17, Sapporo 060-8556, Japan
- N. Hatakeyama, T. Kojima, M. Murata, M. Osanai, H. Chiba, N. Sawada, Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan, e-mail:
- M. M. Thi, D. C. Spray, Department of Neuroscience, Albert Einstein College of Medicine, Bronx, N.Y., USA
- S. Ishiai, Department of Rehabilitation, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Toshihiko Yamashita
- N. Hatakeyama, K. Iba, T. Yamashita, Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, S1, W17, Sapporo 060-8556, Japan
- N. Hatakeyama, T. Kojima, M. Murata, M. Osanai, H. Chiba, N. Sawada, Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan, e-mail:
- M. M. Thi, D. C. Spray, Department of Neuroscience, Albert Einstein College of Medicine, Bronx, N.Y., USA
- S. Ishiai, Department of Rehabilitation, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Norimasa Sawada
- N. Hatakeyama, K. Iba, T. Yamashita, Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, S1, W17, Sapporo 060-8556, Japan
- N. Hatakeyama, T. Kojima, M. Murata, M. Osanai, H. Chiba, N. Sawada, Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan, e-mail:
- M. M. Thi, D. C. Spray, Department of Neuroscience, Albert Einstein College of Medicine, Bronx, N.Y., USA
- S. Ishiai, Department of Rehabilitation, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
49
|
Abstract
The tumor suppressor PTEN was originally identified as a negative regulator of the phosphoinositide 3-kinase (PI3K) signaling, a main regulator of cell growth, metabolism and survival. Yet this function of PTEN is extremely relevant for its tumor-suppressive ability, albeit the recent characterization of many PI3K-independent tumor-suppressive activities. PI3K-mediated PIP(3) production leads to the activation of the canonical AKT-mTORC1 pathway. The implications of this signaling cascade in health and disease have been underscored by the high number of regulators within the pathway whose alterations give rise to different malignancies, including familiar syndromes, metabolic dysfunctions and cancer. Moreover, PI3K is tightly buffered at multiple levels by downstream components, which have turned this signaling pathway literally upside down. PI3K and its downstream components in turn cross-talk with a number of other pathways, thereby leading to a complex network of signals that may have dramatic consequences when perturbed. Here, we review the current status of the PTEN-PI3K signaling pathway with special emphasis on the most recent data on targets and regulation of the PTEN-PI3K axis. This provides novel provocative therapeutic implications based on the targeted modulation of PI3K-cross-talking signals.
Collapse
Affiliation(s)
- A Carracedo
- Cancer Genetics Program, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Harvard Medical School, Boston, MA 02215, USA
| | | |
Collapse
|
50
|
Baljuls A, Schmitz W, Mueller T, Zahedi RP, Sickmann A, Hekman M, Rapp UR. Positive regulation of A-RAF by phosphorylation of isoform-specific hinge segment and identification of novel phosphorylation sites. J Biol Chem 2008; 283:27239-54. [PMID: 18662992 DOI: 10.1074/jbc.m801782200] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In mammals the RAF family of serine/threonine kinases consists of three members, A-, B-, and C-RAF. Activation of RAF kinases involves a complex series of phosphorylations. Although the most prominent phosphorylation sites of B- and C-RAF are well characterized, little is known about regulatory phosphorylation of A-RAF. Using mass spectrometry, we identified here a number of novel in vivo phosphorylation sites in A-RAF. In particular, we found that Ser-432 participates in MEK binding and is indispensable for A-RAF signaling. On the other hand, phosphorylation within the activation segment does not contribute to epidermal growth factor-mediated activation. Furthermore, we show that the potential 14-3-3 binding domains in A-RAF are phosphorylated independently of its activation status. Of importance, we identified a novel regulatory domain in A-RAF (referred to as IH-segment) positioned between amino acids 248 and 267 that contains seven putative phosphorylation sites. Three of these sites, serines 257, 262, and 264, regulate A-RAF activation in a stimulatory manner. The spatial model of the A-RAF fragment, including residues between Ser-246 and Glu-277, revealed a switch of charge at the molecular surface of the IH-region upon phosphorylation, suggesting a mechanism in which the high accumulation of negative charges may lead to an electrostatic destabilization of protein-membrane interaction resulting in depletion of A-RAF from the plasma membrane. Together, we provide here for the first time a detailed analysis of in vivo A-RAF phosphorylation status and demonstrate that regulation of A-RAF by phosphorylation exhibits unique features compared with B- and C-RAF.
Collapse
Affiliation(s)
- Angela Baljuls
- Institute for Medical Radiation and Cell Research, University of Wuerzburg, 97078 Wuerzburg, Germany
| | | | | | | | | | | | | |
Collapse
|