1
|
Mastos C, Xu X, Keen AC, Halls ML. Signalling of Adrenoceptors: Canonical Pathways and New Paradigms. Handb Exp Pharmacol 2024; 285:147-184. [PMID: 38227198 DOI: 10.1007/164_2023_704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
The concept of G protein-coupled receptors initially arose from studies of the β-adrenoceptor, adenylyl cyclase, and cAMP signalling pathway. Since then both canonical G protein-coupled receptor signalling pathways and emerging paradigms in receptor signalling have been defined by experiments focused on adrenoceptors. Here, we discuss the evidence for G protein coupling specificity of the nine adrenoceptor subtypes. We summarise the ability of each of the adrenoceptors to activate proximal signalling mediators including cAMP, calcium, mitogen-activated protein kinases, and protein kinase C pathways. Finally, we highlight the importance of precise spatial and temporal control of adrenoceptor signalling that is controlled by the localisation of receptors at intracellular membranes and in larger protein complexes.
Collapse
Affiliation(s)
- Chantel Mastos
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Xiaomeng Xu
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Alastair C Keen
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Michelle L Halls
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.
| |
Collapse
|
2
|
Pandey S, Mangmool S, Madreiter-Sokolowski CT, Wichaiyo S, Luangmonkong T, Parichatikanond W. Exendin-4 protects against high glucose-induced mitochondrial dysfunction and oxidative stress in SH-SY5Y neuroblastoma cells through GLP-1 receptor/Epac/Akt signaling. Eur J Pharmacol 2023:175896. [PMID: 37391007 DOI: 10.1016/j.ejphar.2023.175896] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/16/2023] [Accepted: 06/28/2023] [Indexed: 07/02/2023]
Abstract
Mitochondrial dysfunction under diabetic condition leads to the development and progression of neurodegenerative complications. Recently, the beneficial effects of glucagon-like peptide-1 (GLP-1) receptor agonists on diabetic neuropathies have been widely recognized. However, molecular mechanisms underlying the neuroprotective effects of GLP-1 receptor agonists against high glucose (HG)-induced neuronal damages is not completely elucidated. Here, we investigated the underlying mechanisms of GLP-1 receptor agonist treatment against oxidative stress, mitochondrial dysfunction, and neuronal damages under HG-conditions mimicking a diabetic hyperglycemic state in SH-SY5Y neuroblastoma cells. We revealed that treatment with exendin-4, a GLP-1 receptor agonist, not only increased the expression of survival markers, phospho-Akt/Akt and Bcl-2, but also decreased the expression of pro-apoptotic marker, Bax, and reduced the levels of reactive oxygen species (ROS) defense markers (catalase, SOD-2, and HO-1) under HG conditions. The expressions of mitochondrial function associated genes, MCU and UCP3, and mitochondrial fission genes, DRP1 and FIS1, were decreased by exendin-4 compared to non-treated levels, while the protein expression levels of mitochondrial homeostasis regulators, Parkin and PINK1, were enhanced. In addition, blockade of Epac and Akt activities was able to antagonize these neuroprotective effects of exendin-4. Collectively, we demonstrated that stimulation of GLP-1 receptor propagates a neuroprotective cascade against the oxidative stresses and mitochondrial dysfunctions as well as augments survival through the Epac/Akt-dependent pathway. Therefore, the revealed mechanisms underlying GLP-1 receptor pathway by preserving mitochondrial homeostasis would be a therapeutic candidate to alleviate neuronal dysfunctions and delay the progression of diabetic neuropathies.
Collapse
Affiliation(s)
- Sudhir Pandey
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok, 10400, Thailand
| | - Supachoke Mangmool
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Corina T Madreiter-Sokolowski
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, 8010, Austria
| | - Surasak Wichaiyo
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok, 10400, Thailand
| | - Theerut Luangmonkong
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok, 10400, Thailand
| | | |
Collapse
|
3
|
Ramsey A, Akana L, Miyajima E, Douglas S, Gray J, Rowland A, Sharma KD, Xu J, Xie JY, Zhou GL. CAP1 (cyclase-associated protein 1) mediates the cyclic AMP signals that activate Rap1 in stimulating matrix adhesion of colon cancer cells. Cell Signal 2023; 104:110589. [PMID: 36621727 PMCID: PMC9908859 DOI: 10.1016/j.cellsig.2023.110589] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 12/12/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023]
Abstract
We previously reported that CAP1 (Cyclase-Associated Protein 1) regulates matrix adhesion in mammalian cells through FAK (Focal Adhesion Kinase). More recently, we discovered a phosphor-regulation mechanism for CAP1 through the Ser307/Ser309 tandem site that is of critical importance for all CAP1 functions. However, molecular mechanisms underlying the CAP1 function in adhesion and its regulation remain largely unknown. Here we report that Rap1 also facilitates the CAP1 function in adhesion, and more importantly, we identify a novel signaling pathway where CAP1 mediates the cAMP signals, through the cAMP effectors Epac (Exchange proteins directly activated by cAMP) and PKA (Protein Kinase A), to activate Rap1 in stimulating matrix adhesion in colon cancer cells. Knockdown of CAP1 led to opposite adhesion phenotypes in SW480 and HCT116 colon cancer cells, with reduced matrix adhesion and reduced FAK and Rap1 activities in SW480 cells while it stimulated matrix adhesion as well as FAK and Rap1 activities in HCT116 cells. Importantly, depletion of CAP1 abolished the stimulatory effects of the cAMP activators forskolin and isoproterenol, as well as that of Epac and PKA, on matrix adhesion in both cell types. Our results consistently support a required role for CAP1 in the cAMP activation of Rap1. Identification of the key role for CAP1 in linking the major second messenger cAMP to activation of Rap1 in stimulating adhesion, which may potentially also regulate proliferation in other cell types, not only vertically extends our knowledge on CAP biology, but also carries important translational potential for targeting CAP1 in cancer therapeutics.
Collapse
Affiliation(s)
- Auburn Ramsey
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA
| | - Lokesh Akana
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA
| | - Erina Miyajima
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA
| | - Spencer Douglas
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA
| | - Joshua Gray
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA
| | - Alyssa Rowland
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA
| | - Krishna Deo Sharma
- Molecular Biosciences Graduate Program, Arkansas State University, State University, AR 72467, USA
| | - Jianfeng Xu
- Molecular Biosciences Graduate Program, Arkansas State University, State University, AR 72467, USA; Arkansas Biosciences Institute, Arkansas State University, Jonesboro, AR 72401, USA; College of Agriculture, Arkansas State University, State University, AR 72467, USA
| | - Jennifer Y Xie
- Molecular Biosciences Graduate Program, Arkansas State University, State University, AR 72467, USA; Department of Basic Sciences, New York Institute of Technology College of Osteopathic Medicine at Arkansas State University, Jonesboro, AR 72401, USA
| | - Guo-Lei Zhou
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA; Molecular Biosciences Graduate Program, Arkansas State University, State University, AR 72467, USA.
| |
Collapse
|
4
|
Mao P, Huang C, Li Y, Zhao Y, Zhou S, Zhao Z, Mu Y, Wang L, Li F, Zhao AZ. Pharmacological targeting of type phosphodiesterase 4 inhibits the development of acute myeloid leukemia by impairing mitochondrial function through the Wnt/β-catenin pathway. Biomed Pharmacother 2023; 157:114027. [PMID: 36436494 DOI: 10.1016/j.biopha.2022.114027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 11/09/2022] [Accepted: 11/17/2022] [Indexed: 11/27/2022] Open
Abstract
Acute myeloid leukemia (AML) is prone to drug-resistant relapse with a low 5-year survival rate. New therapeutic modalities are sorely needed to provide hope for AML relapse patients. Herein, we demonstrated a specific inhibitor of type 4 phosphodiesterase (PDE4), Zl-n-91, could significantly reduce the proliferation of AML cells, block DNA replication process, and increase AML cell death. Zl-n-91 also impeded the growth of subcutaneous xenograft and prolonged the survival of the MLL-AF9-driven AML model. Bioinformatic analysis revealed that elevated mitochondrial gene signatures inversely correlate with the survival of AML patients; and importantly, Zl-n-91 strongly suppressed the function of mitochondria. In addition, this PDE4 inhibitor induced alterations in multiple signaling pathways, including the reduction of β-catenin activity. Stimulation of the Wnt/β-catenin pathway could attenuate the inhibitory effect of Zl-n-91 on AML cell proliferation as well as mitochondrial function. Taken together, we revealed for the first time that targeting PDE4 activity could attenuate mitochondrial function through a Wnt/β-catenin pathway, which in turn would block the growth of AML cells. Specific PDE4 inhibitors can potentially serve as a new treatment modality for AML patients.
Collapse
Affiliation(s)
- Ping Mao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Xiaoguwei Street, Panyu District, Guangzhou, Guangdong 510006, China
| | - Changhao Huang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Xiaoguwei Street, Panyu District, Guangzhou, Guangdong 510006, China
| | - Yuyu Li
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Xiaoguwei Street, Panyu District, Guangzhou, Guangdong 510006, China
| | - Yuanyi Zhao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Xiaoguwei Street, Panyu District, Guangzhou, Guangdong 510006, China
| | - Sujin Zhou
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Xiaoguwei Street, Panyu District, Guangzhou, Guangdong 510006, China
| | - Zhenggang Zhao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Xiaoguwei Street, Panyu District, Guangzhou, Guangdong 510006, China
| | - Yunping Mu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Xiaoguwei Street, Panyu District, Guangzhou, Guangdong 510006, China
| | - Lina Wang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Xiaoguwei Street, Panyu District, Guangzhou, Guangdong 510006, China
| | - Fanghong Li
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Xiaoguwei Street, Panyu District, Guangzhou, Guangdong 510006, China.
| | - Allan Z Zhao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Xiaoguwei Street, Panyu District, Guangzhou, Guangdong 510006, China.
| |
Collapse
|
5
|
Quantitative live-cell imaging of GPCR downstream signaling dynamics. Biochem J 2022; 479:883-900. [PMID: 35383830 DOI: 10.1042/bcj20220021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 04/03/2022] [Accepted: 04/06/2022] [Indexed: 11/17/2022]
Abstract
G-protein-coupled receptors (GPCRs) play an important role in sensing various extracellular stimuli, such as neurotransmitters, hormones, and tastants, and transducing the input information into the cell. While the human genome encodes more than 800 GPCR genes, only four Gα-proteins (Gαs, Gαi/o, Gαq/11, and Gα12/13) are known to couple with GPCRs. It remains unclear how such divergent GPCR information is translated into the downstream G-protein signaling dynamics. To answer this question, we report a live-cell fluorescence imaging system for monitoring GPCR downstream signaling dynamics. Genetically encoded biosensors for cAMP, Ca2+, RhoA, and ERK were selected as markers for GPCR downstream signaling, and were stably expressed in HeLa cells. GPCR was further transiently overexpressed in the cells. As a proof-of-concept, we visualized GPCR signaling dynamics of 5 dopamine receptors and 12 serotonin receptors, and found heterogeneity between GPCRs and between cells. Even when the same Gα proteins were known to be coupled, the patterns of dynamics in GPCR downstream signaling, including the signal strength and duration, were substantially distinct among GPCRs. These results suggest the importance of dynamical encoding in GPCR signaling.
Collapse
|
6
|
The Influence of Burst-Firing EMF on Forskolin-Induced Pheochromocytoma (PC12) Plasma Membrane Extensions. NEUROSCI 2021. [DOI: 10.3390/neurosci2040028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Previous research has demonstrated that pheochromocytoma (PC12) cells treated with forskolin provides a model for the in vitro examination of neuritogenesis. Exposure to electromagnetic fields (EMFs), especially those which have been designed to mimic biological function, can influence the functions of various biological systems. We aimed to assess whether exposure of PC12 cells treated with forskolin to patterned EMF would produce more plasma membrane extensions (PME) as compared to PC12 cells treated with forskolin alone (i.e., no EMF exposure). In addition, we aimed to determine whether the differences observed between the proportion of PME of PC12 cells treated with forskolin and exposed to EMF were specific to the intensity, pattern, or timing of the applied EMF. Our results showed an overall increase in PME for PC12 cells treated with forskolin and exposed to Burst-firing EMF as compared to PC12 cells receiving forskolin alone. No other patterned EMF investigated were deemed to be effective. Furthermore, intensity and timing of the Burst-firing pattern did not significantly alter the proportion of PME of PC12 cells treated with forskolin and exposed to patterned EMF.
Collapse
|
7
|
Li W, Hu C, Zhang X, Wang B, Li Z, Ling M, Sun S, Guo C, Li D, Liu S. SUZ12 Loss Amplifies the Ras/ERK Pathway by Activating Adenylate Cyclase 1 in NF1-Associated Neurofibromas. Front Oncol 2021; 11:738300. [PMID: 34692515 PMCID: PMC8526866 DOI: 10.3389/fonc.2021.738300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/20/2021] [Indexed: 11/13/2022] Open
Abstract
Patients with germline neurofibromatosis type 1 (NF1) microdeletions frequently exhibit hereditary syndromes such as cardiovascular anomalies and have an increased risk of malignant peripheral nerve sheath tumors (MPNSTs). This study aimed to identify the genes codeleted with SUZ12 that are related to MPNST. We used differential gene expression and enrichment analyses to analyze the SUZ12-mutant and SUZ12-wild-type gene expression profiles in the GSE118186 and GSE66743 datasets in Gene Expression Omnibus (GEO). PPI network analysis combined with MPNST patient survival analysis was used to identify ADCY1, which catalyzes the conversion of ATP to cAMP, as a key gene. Moreover, chromatin immunoprecipitation sequencing (ChIP-Seq) showed that the distribution of H3K27me3 in the ADCY1 promoter region and gene body was significantly reduced in SUZ12-mutant cells. To verify the role of ADCY1 in SUZ12 mutation, we used RNA interference and plasmid transfection to interfere with SUZ12 expression in plexiform neurofibroma (pNF) and MPNST cell lines and then treated the cells with forskolin, IBMX and H89. ERK phosphorylation was accelerated and prolonged after siRNA transfection, especially in ipNF05.5 cells, and the intensity and duration of ERK activation were reduced after SUZ12 overexpression. Importantly, the level of p-ERK was consistent with that of Rap1-GTP. Moreover, H89 completely blocked Rap1 activation and the changes in the p-ERK level after SUZ12 siRNA transfection. In conclusion, our findings suggested that SUZ12 loss potentiates the effects of NF1 mutations by amplifying Ras signaling through the ADCY1/cAMP/Rap1/ERK pathway and that SUZ12 may serve as a therapeutic and prognostic biomarker in NF1-associated neurofibromas.
Collapse
Affiliation(s)
- Weijie Li
- Department of Injury and Repair, and Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Chenhao Hu
- Department of Injury and Repair, and Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Xingnan Zhang
- Department of Injury and Repair, and Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Binbin Wang
- Department of Injury and Repair, and Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Zhen Li
- Department of Injury and Repair, and Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Miao Ling
- Department of Injury and Repair, and Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Shengqiao Sun
- Department of Injury and Repair, and Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Chao Guo
- Department of Injury and Repair, and Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Dezhi Li
- Department of Injury and Repair, and Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Song Liu
- Department of Injury and Repair, and Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,U 1195, Institut national de la santé et de la recherche médicale (INSERM) and University Paris-Sud and University Paris Saclay, Le Kremlin-Bicêtre, France
| |
Collapse
|
8
|
Identification and Characterization of an Affimer Affinity Reagent for the Detection of the cAMP Sensor, EPAC1. Cells 2021; 10:cells10092307. [PMID: 34571955 PMCID: PMC8465552 DOI: 10.3390/cells10092307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 01/15/2023] Open
Abstract
An exchange protein directly activated by cAMP 1 (EPAC1) is an intracellular sensor for cAMP that is involved in a wide variety of cellular and physiological processes in health and disease. However, reagents are lacking to study its association with intracellular cAMP nanodomains. Here, we use non-antibody Affimer protein scaffolds to develop isoform-selective protein binders of EPAC1. Phage-display screens were carried out against purified, biotinylated human recombinant EPAC1ΔDEP protein (amino acids 149–811), which identified five potential EPAC1-selective Affimer binders. Dot blots and indirect ELISA assays were next used to identify Affimer 780A as the top EPAC1 binder. Mutagenesis studies further revealed a potential interaction site for 780A within the EPAC1 cyclic nucleotide binding domain (CNBD). In addition, 780A was shown to co-precipitate EPAC1 from transfected cells and co-localize with both wild-type EPAC1 and a mis-targeting mutant of EPAC1(K212R), predominantly in perinuclear and cytosolic regions of cells, respectively. As a novel EPAC1-selective binder, 780A therefore has the potential to be used in future studies to further understand compartmentalization of the cAMP-EPAC1 signaling system.
Collapse
|
9
|
Pitsava G, Stratakis CA, Faucz FR. PRKAR1A and Thyroid Tumors. Cancers (Basel) 2021; 13:cancers13153834. [PMID: 34359735 PMCID: PMC8345073 DOI: 10.3390/cancers13153834] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/24/2021] [Accepted: 07/27/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary In 2021 it is estimated that there will be 44,280 new cases of thyroid cancer in the United States and the incidence rate is higher in women than in men by almost 3 times. Well-differentiated thyroid cancer is the most common subtype of thyroid cancer and includes follicular (FTC) and papillary (PTC) carcinomas. Over the last decade, researchers have been able to better understand the molecular mechanisms involved in thyroid carcinogenesis, identifying genes including but not limited to RAS, BRAF, PAX8/PPARγ chromosomal rearrangements and others, as well as several tumor genes involved in major signaling pathways regulating cell cycle, differentiation, growth, or proliferation. Patients with Carney complex (CNC) have increased incidence of thyroid tumors, including cancer, yet little is known about this association. CNC is a familial multiple neoplasia and lentiginosis syndrome cause by inactivating mutations in the PRKAR1A gene which encodes the regulatory subunit type 1α of protein kinase A. This work summarizes what we know today about PRKAR1A defects in humans and mice and their role in thyroid tumor development, as the first such review on this issue. Abstract Thyroid cancer is the most common type of endocrine malignancy and the incidence is rapidly increasing. Follicular (FTC) and papillary thyroid (PTC) carcinomas comprise the well-differentiated subtype and they are the two most common thyroid carcinomas. Multiple molecular genetic and epigenetic alterations have been identified in various types of thyroid tumors over the years. Point mutations in BRAF, RAS as well as RET/PTC and PAX8/PPARγ chromosomal rearrangements are common. Thyroid cancer, including both FTC and PTC, has been observed in patients with Carney Complex (CNC), a syndrome that is inherited in an autosomal dominant manner and predisposes to various tumors. CNC is caused by inactivating mutations in the tumor-suppressor gene encoding the cyclic AMP (cAMP)-dependent protein kinase A (PKA) type 1α regulatory subunit (PRKAR1A) mapped in chromosome 17 (17q22–24). Growth of the thyroid is driven by the TSH/cAMP/PKA signaling pathway and it has been shown in mouse models that PKA activation through genetic ablation of the regulatory subunit Prkar1a can cause FTC. In this review, we provide an overview of the molecular mechanisms contributing to thyroid tumorigenesis associated with inactivation of the RRKAR1A gene.
Collapse
Affiliation(s)
- Georgia Pitsava
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institutes of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA;
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Constantine A. Stratakis
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Fabio R. Faucz
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA;
- Correspondence: ; Tel.: +1-301-451-7177
| |
Collapse
|
10
|
Choi KM, Haak AJ, Diaz Espinosa AM, Cummins KA, Link PA, Aravamudhan A, Wood DK, Tschumperlin DJ. GPCR-mediated YAP/TAZ inactivation in fibroblasts via EPAC1/2, RAP2C, and MAP4K7. J Cell Physiol 2021; 236:7759-7774. [PMID: 34046891 DOI: 10.1002/jcp.30459] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 05/06/2021] [Accepted: 05/19/2021] [Indexed: 12/29/2022]
Abstract
Yes-associated protein (YAP) and PDZ-binding motif (TAZ) have emerged as important regulators of pathologic fibroblast activation in fibrotic diseases. Agonism of Gαs-coupled G protein coupled receptors (GPCRs) provides an attractive approach to inhibit the nuclear localization and function of YAP and TAZ in fibroblasts that inhibits or reverses their pathological activation. Agonism of the dopamine D1 GPCR has proven effective in preclinical models of lung and liver fibrosis. However, the molecular mechanisms coupling GPCR agonism to YAP and TAZ inactivation in fibroblasts remain incompletely understood. Here, using human lung fibroblasts, we identify critical roles for the cAMP effectors EPAC1/2, the small GTPase RAP2c, and the serine/threonine kinase MAP4K7 as the essential elements in the downstream signaling cascade linking GPCR agonism to LATS1/2-mediated YAP and TAZ phosphorylation and nuclear exclusion in fibroblasts. We further show that this EPAC/RAP2c/MAP4K7 signaling cascade is essential to the effects of dopamine D1 receptor agonism on reducing fibroblast proliferation, contraction, and extracellular matrix production. Targeted modulation of this cascade in fibroblasts may prove a useful strategy to regulate YAP and TAZ signaling and fibroblast activities central to tissue repair and fibrosis.
Collapse
Affiliation(s)
- Kyoung Moo Choi
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| | - Andrew J Haak
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| | - Ana M Diaz Espinosa
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| | - Katherine A Cummins
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota, USA
| | - Patrick A Link
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| | - Aja Aravamudhan
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| | - David K Wood
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota, USA
| | - Daniel J Tschumperlin
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| |
Collapse
|
11
|
Mehta S, Zhang J. Biochemical Activity Architectures Visualized-Using Genetically Encoded Fluorescent Biosensors to Map the Spatial Boundaries of Signaling Compartments. Acc Chem Res 2021; 54:2409-2420. [PMID: 33949851 DOI: 10.1021/acs.accounts.1c00056] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
All biological processes arise through the coordinated actions of biochemical pathways. How such functional diversity is achieved by a finite cast of molecular players remains a central mystery in biology. Spatial compartmentation-the idea that biochemical activities are organized around discrete spatial domains within cells-was first proposed nearly 40 years ago and has become firmly rooted in our understanding of how biochemical pathways are regulated to ensure specificity. However, directly interrogating spatial compartmentation and its mechanistic origins has only really become possible in the last 20 or so years, following technological advances such as the development of genetically encoded fluorescent biosensors. These powerful molecular tools permit a direct, real-time visualization of dynamic biochemical processes in native biological contexts, and they are essential for probing the spatial regulation of biochemical activities. In this Account, we review our lab's efforts in developing and using biosensors to map the spatial compartmentation of intracellular signaling pathways and illuminate key mechanisms that establish the boundaries of an intricate biochemical activity architecture. We first discuss the role of regulatory fences, wherein the dynamic activation and deactivation of diffusible messengers produce diverse signaling compartments. For example, we used biosensors for the Ca2+ effector calmodulin and its downstream target calcineurin to reveal a spatial gradient of calmodulin that controls the temporal dynamics of calcineurin signaling. Our studies using cyclic adenosine monophosphate (cAMP) biosensors have similarly elucidated fenced cAMP domains generated by competing production and degradation pathways, ranging in size from cell-spanning gradients to nanoscale hotspots. Second, we describe the role played by intracellular membranes in creating unique signaling platforms with distinctive pathway regulation, as revealed through studies using subcellularly targeted fluorescent biosensors. Using biosensors to visualize subcellular extracellular response kinase (ERK) pathway activity, for example, led us to discover a local signaling circuit that mediates distinct plasma membrane ERK dynamics versus global ERK signaling. Similarly, our work developing biosensors to monitor the subcellular mechanistic target of rapamycin complex 1 (mTORC1) signaling allowed us to not only clarify the presence of mTORC1 activity in the nucleus but also identify a novel mechanism governing the activation of mTORC1 in this location. Finally, we detail how molecular assemblies enable the precise spatial tuning of biochemical activity, through investigations enabled by cutting-edge advances in biosensor design. We recently identified liquid-liquid phase separation as a major factor in cAMP compartmentation aided by a new strategy for targeting biosensors to endogenously expressed proteins via genome editing, for instance, and have also been able to directly visualize nanometer-scale protein kinase signalosomes using an entirely new class of biosensors specifically developed for the dynamic super-resolution imaging of live-cell biochemical activities. Our work provides key insights into the molecular logic of spatially regulated signaling and lays the foundation for a broader exploration of biochemical activity architectures across multiple spatial scales.
Collapse
Affiliation(s)
- Sohum Mehta
- Department of Pharmacology, University of California, San Diego, La Jolla, California 92093, United States
| | - Jin Zhang
- Departments of Pharmacology, Bioengineering, and Chemistry and Biochemistry, University of California, San Diego, La Jolla, California 92093, United States
| |
Collapse
|
12
|
Chavkin NW, Leaf EM, Brooks KE, Wallingford MC, Lund SM, Giachelli CM. Adapter Protein RapGEF1 Is Required for ERK1/2 Signaling in Response to Elevated Phosphate in Vascular Smooth Muscle Cells. J Vasc Res 2021; 58:277-285. [PMID: 33951626 PMCID: PMC8478708 DOI: 10.1159/000516044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 03/17/2021] [Indexed: 11/19/2022] Open
Abstract
The sodium-dependent phosphate transporter, SLC20A1, is required for elevated inorganic phosphate (Pi) induced vascular smooth muscle cell (VSMC) matrix mineralization and phenotype transdifferentiation. Recently, elevated Pi was shown to induce ERK1/2 phosphorylation through SLC20A1 by Pi uptake-independent functions in VSMCs, suggesting a cell signaling response to elevated Pi. Previous studies identified Rap1 guanine nucleotide exchange factor (RapGEF1) as an SLC20A1-interacting protein and RapGEF1 promotes ERK1/2 phosphorylation through Rap1 activation. In this study, we tested the hypothesis that RapGEF1 is a critical component of the SLC20A1-mediated Pi-induced ERK1/2 phosphorylation pathway. Co-localization of SLC20A1 and RapGEF1, knockdown of RapGEF1 with siRNA, and small molecule inhibitors of Rap1, B-Raf, and Mek1/2 were investigated. SLC20A1 and RapGEF1 were co-localized in peri-membranous structures in VSMCs. Knockdown of RapGEF1 and small molecule inhibitors against Rap1, B-Raf, and Mek1/2 eliminated elevated Pi-induced ERK1/2 phosphorylation. Knockdown of RapGEF1 inhibited SM22α mRNA expression and blocked elevated Pi-induced downregulation of SM22α mRNA. Together, these data suggest that RapGEF1 is required for SLC20A1-mediated elevated Pi signaling through a Rap1/B-Raf/Mek1/2 cell signaling pathway, thereby promoting ERK1/2 phosphorylation and inhibiting SM22α gene expression in VSMCs.
Collapse
Affiliation(s)
| | - Elizabeth M. Leaf
- Department of Bioengineering, University of Washington, Seattle WA USA
| | - Kadin E. Brooks
- Department of Bioengineering, University of Washington, Seattle WA USA
| | | | - Susan M. Lund
- Department of Bioengineering, University of Washington, Seattle WA USA
| | | |
Collapse
|
13
|
Vega Hissi EG, De Costa Guardamagna AB, Garro AD, Falcon CR, Anderluh M, Tomašič T, Kikelj D, Yaneff A, Davio CA, Enriz RD, Zurita AR. A Potent N-(piperidin-4-yl)-1H-pyrrole-2-carboxamide Inhibitor of Adenylyl Cyclase of G. lamblia: Biological Evaluation and Molecular Modelling Studies. ChemMedChem 2021; 16:2094-2105. [PMID: 33783977 DOI: 10.1002/cmdc.202100037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/26/2021] [Indexed: 11/06/2022]
Abstract
In this work, we report a derivative of N-(piperidin-4-yl)-1H-pyrrole-2-carboxamide as a new inhibitor for adenylyl cyclase of Giardia lamblia which was obtained from a study using structural data of the nucleotidyl cyclase 1 (gNC1) of this parasite. For such a study, we developed a model for this specific enzyme by using homology techniques, which is the first model reported for gNC1 of G. lamblia. Our studies show that the new inhibitor has a competitive mechanism of action against this enzyme. 2-Hydroxyestradiol was used as the reference compound for comparative studies. Results in this work are important from two points of view. on the one hand, an experimentally corroborated model for gNC1 of G. lamblia obtained by molecular modelling is presented; on the other hand, the new inhibitor obtained is an undoubtedly excellent starting structure for the development of new metabolic inhibitors for G. lamblia.
Collapse
Affiliation(s)
- Esteban G Vega Hissi
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700, San Luis, Argentina
| | - Antonella B De Costa Guardamagna
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700, San Luis, Argentina
| | - Adriana D Garro
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700, San Luis, Argentina
| | - Cristian R Falcon
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700, San Luis, Argentina
| | - Marko Anderluh
- Department of Medicinal Chemistry, University of Ljubljana, Faculty of Pharmacy Askerceva, cesta 7, 1000, Ljubljana, Slovenia
| | - Tihomir Tomašič
- Department of Medicinal Chemistry, University of Ljubljana, Faculty of Pharmacy Askerceva, cesta 7, 1000, Ljubljana, Slovenia
| | - Danijel Kikelj
- Department of Medicinal Chemistry, University of Ljubljana, Faculty of Pharmacy Askerceva, cesta 7, 1000, Ljubljana, Slovenia
| | - Agustín Yaneff
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, C1113, AAD, Buenos Aires, Argentina
| | - Carlos A Davio
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, C1113, AAD, Buenos Aires, Argentina
| | - Ricardo D Enriz
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700, San Luis, Argentina
| | - Adolfo R Zurita
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700, San Luis, Argentina
| |
Collapse
|
14
|
Kermath BA, Vanderplow AM, Cahill ME. Dysregulated Prefrontal Cortical RhoA Signal Transduction in Bipolar Disorder with Psychosis: New Implications for Disease Pathophysiology. Cereb Cortex 2021; 30:59-71. [PMID: 31220216 DOI: 10.1093/cercor/bhz070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
While research has identified alterations in dorsolateral prefrontal cortical function as a key factor to the etiology of bipolar disorder, few studies have uncovered robust changes in protein signal transduction pathways in this disorder. Given the direct relevance of protein-based expressional alterations to cellular functions and because many of the key regulatory mechanisms for the disease pathogenesis likely include alterations in protein activity rather than changes in expression alone, the identification of alterations in discrete signal transduction pathways in bipolar disorder would have broad implications for understanding the disease pathophysiology. As prior microarray data point to a previously unrecognized involvement of the RhoA network in bipolar disorder, here we investigate the protein expression and activity of key components of a RhoA signal transduction pathway in dorsolateral prefrontal cortical homogenates from subjects with bipolar disorder. The results of this investigation implicate overactivation of prefrontal cortical RhoA signaling in specific subtypes of bipolar disorder. The specificity of these findings is demonstrated by a lack of comparable changes in schizophrenia; however, our findings do identify convergence between both disorders at the level of activity-mediated actin cytoskeletal regulation. These findings have implications for understanding the altered cortical synaptic connectivity of bipolar disorder.
Collapse
Affiliation(s)
- Bailey A Kermath
- Department of Comparative Biosciences, University of Wisconsin at Madison, Madison, WI, USA
| | - Amanda M Vanderplow
- Department of Comparative Biosciences, University of Wisconsin at Madison, Madison, WI, USA
| | - Michael E Cahill
- Department of Comparative Biosciences, University of Wisconsin at Madison, Madison, WI, USA
| |
Collapse
|
15
|
Braune S, Küpper JH, Jung F. Effect of Prostanoids on Human Platelet Function: An Overview. Int J Mol Sci 2020; 21:ijms21239020. [PMID: 33260972 PMCID: PMC7730041 DOI: 10.3390/ijms21239020] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/19/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022] Open
Abstract
Prostanoids are bioactive lipid mediators and take part in many physiological and pathophysiological processes in practically every organ, tissue and cell, including the vascular, renal, gastrointestinal and reproductive systems. In this review, we focus on their influence on platelets, which are key elements in thrombosis and hemostasis. The function of platelets is influenced by mediators in the blood and the vascular wall. Activated platelets aggregate and release bioactive substances, thereby activating further neighbored platelets, which finally can lead to the formation of thrombi. Prostanoids regulate the function of blood platelets by both activating or inhibiting and so are involved in hemostasis. Each prostanoid has a unique activity profile and, thus, a specific profile of action. This article reviews the effects of the following prostanoids: prostaglandin-D2 (PGD2), prostaglandin-E1, -E2 and E3 (PGE1, PGE2, PGE3), prostaglandin F2α (PGF2α), prostacyclin (PGI2) and thromboxane-A2 (TXA2) on platelet activation and aggregation via their respective receptors.
Collapse
|
16
|
Expression of a novel brain specific isoform of C3G is regulated during development. Sci Rep 2020; 10:18838. [PMID: 33139841 PMCID: PMC7606606 DOI: 10.1038/s41598-020-75813-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 10/01/2020] [Indexed: 12/13/2022] Open
Abstract
Mice lacking C3G (RapGEF1), a ubiquitously expressed protein essential for neuronal differentiation, show multiple defects in brain development. Function of C3G in neurogenesis is poorly defined. Here, we identify brain specific expression of a novel C3G isoform in mice and humans. This isoform has an insert in the Crk-binding region, generating a polypeptide of 175 kDa, unlike the previously known 140 kDa form expressed in all other tissues. In the adult mouse brain, C3G expression is seen in neurons, but was not detectable in GFAP-positive cells. C3G levels were high in the CA3 region of hippocampus and in mitral cells of olfactory bulb. Neural progenitor cells positive for Doublecortin and Nestin, show expression of C3G. During development, C3G is expressed in precursor cells prior to their differentiation into mature neurons or astrocytes. The 175 kDa as well as 140 kDa forms are seen in embryonic mouse brain, while only the 175 kDa variant is seen in post-natal brain. Human cerebral organoids generated from induced pluripotent stem cells predominantly expressed the 140 kDa polypeptides, and the 175 kDa isoform appeared upon maturation. This study describes developmental regulation and neuronal expression of a brain specific isoform of C3G, a molecule essential for normal development of the mammalian brain.
Collapse
|
17
|
Pei J, Xiao Z, Guo Z, Pei Y, Wei S, Wu H, Wang D. Sustained Stimulation of β 2AR Inhibits Insulin Signaling in H9C2 Cardiomyoblast Cells Through the PKA-Dependent Signaling Pathway. Diabetes Metab Syndr Obes 2020; 13:3887-3898. [PMID: 33116735 PMCID: PMC7585860 DOI: 10.2147/dmso.s268028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/05/2020] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION This study aimed to investigate the role of β2 adrenergic receptor (β2AR) in insulin signaling transduction in H9C2 cardiomyoblast cells to understand the formation of the β2AR-insulin receptor (IR) protein complex and its role in insulin-induced Glut4 expression. METHODS H9C2 cells were treated with various protein inhibitors (CGP, β1AR inhibitor CGP20712; ICI, β2AR inhibitor ICI 118,551; PKI, PKA inhibitor myristoylated PKI; PD 0325901, MEK inhibitor; SP600125, JNK inhibitor) with or without insulin or isoproterenol (ISO) before RNA-sequencing (RNA-Seq) and quantitative-PCR (Q-PCR). Yeast two-hybrid, co-immunoprecipitation and His-tag pull-down assay were carried out to investigate the formation of the β2AR-IR protein complex. The intracellular concentrations of cAMP in H9C2 cells were tested by high performance liquid chromatography (HPLC) and the phosphorylation of JNK was tested by Western blot. RESULTS Gene Ontology (GO) analysis revealed that the most significantly enriched processes in the domain of molecular function (MF) were catalytic activity and binding, whereas in the domain of biological processes (BP) were metabolic process and cellular process. Furthermore, the enriched processes in the domain of cellular components (CC) were cell and cell parts. The Kyoto encyclopedia of genes and genomes (KEGG) pathway analysis showed that the most significant pathways that have been altered included the PI3K-Akt and MAPK signaling pathways. Q-PCR, which was performed to verify the gene expression levels exhibited consistent results. In evaluating the signaling pathways, the sustained stimulation of β2AR by ISO inhibited insulin signalling, and the effect was primarily through the cAMP-PKA-JNK pathway and MEK/JNK signaling pathway. Yeast two-hybrid, co-immunoprecipitation and His-tag pull-down assay revealed that β2AR, IR, insulin receptor substrate 1 (IRS1), Grb2-associated binding protein 1 (GAB1) and Grb2 existed in the same protein complex. CONCLUSION The sustained stimulation of β2AR might inhibit insulin signaling transduction through the cAMP-PKA-JNK and MEK/JNK pathways in H9C2 cells.
Collapse
Affiliation(s)
- Jinli Pei
- Key Laboratory of Ministry of Education for Tropical Bioresources, Hainan University, Haikou, Hainan570228, People's Republic of China
- Laboratory of Biotechnology and Molecular Pharmacology, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan570228, People's Republic of China
| | - Zhengpan Xiao
- Key Laboratory of Ministry of Education for Tropical Bioresources, Hainan University, Haikou, Hainan570228, People's Republic of China
- Laboratory of Biotechnology and Molecular Pharmacology, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan570228, People's Republic of China
| | - Ziyi Guo
- Key Laboratory of Ministry of Education for Tropical Bioresources, Hainan University, Haikou, Hainan570228, People's Republic of China
- Laboratory of Biotechnology and Molecular Pharmacology, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan570228, People's Republic of China
| | - Yechun Pei
- Key Laboratory of Ministry of Education for Tropical Bioresources, Hainan University, Haikou, Hainan570228, People's Republic of China
- Laboratory of Biotechnology and Molecular Pharmacology, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan570228, People's Republic of China
| | - Shuangshuang Wei
- Key Laboratory of Ministry of Education for Tropical Bioresources, Hainan University, Haikou, Hainan570228, People's Republic of China
- Laboratory of Biotechnology and Molecular Pharmacology, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan570228, People's Republic of China
| | - Hao Wu
- Key Laboratory of Ministry of Education for Tropical Bioresources, Hainan University, Haikou, Hainan570228, People's Republic of China
- Laboratory of Biotechnology and Molecular Pharmacology, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan570228, People's Republic of China
| | - Dayong Wang
- Key Laboratory of Ministry of Education for Tropical Bioresources, Hainan University, Haikou, Hainan570228, People's Republic of China
- Laboratory of Biotechnology and Molecular Pharmacology, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan570228, People's Republic of China
| |
Collapse
|
18
|
Hoy JJ, Parra NS, Park J, Kuhn S, Iglesias-Bartolome R. Protein kinase A inhibitor proteins (PKIs) divert GPCR-Gαs-cAMP signaling toward EPAC and ERK activation and are involved in tumor growth. FASEB J 2020; 34:13900-13917. [PMID: 32830375 PMCID: PMC7722164 DOI: 10.1096/fj.202001515r] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/30/2020] [Accepted: 08/07/2020] [Indexed: 01/12/2023]
Abstract
The PKA-inhibitor (PKI) family members PKIα, PKIβ, and PKIγ bind with high affinity to PKA and block its kinase activity, modulating the extent, and duration of PKA-mediated signaling events. While PKA is a well-known regulator of physiological and oncogenic events, the role of PKI proteins in these pathways has remained elusive. Here, by measuring activation of the MAPK pathway downstream of GPCR-Gαs-cAMP signaling, we show that the expression levels of PKI proteins can alter the balance of activation of two major cAMP targets: PKA and EPAC. Our results indicate that PKA maintains repressive control over MAPK signaling as well as a negative feedback on cAMP concentration. Overexpression of PKI and its subsequent repression of PKA dysregulates these signaling pathways, resulting in increased intracellular cAMP, and enhanced activation of EPAC and MAPK. We also find that amplifications of PKIA are common in prostate cancer and are associated with reduced progression free survival. Depletion of PKIA in prostate cancer cells leads to reduced migration, increased sensitivity to anoikis and reduced tumor growth. By altering PKA activity PKI can act as a molecular switch, driving GPCR-Gαs-cAMP signaling toward activation of EPAC-RAP1 and MAPK, ultimately modulating tumor growth.
Collapse
Affiliation(s)
- James J. Hoy
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Natalia Salinas Parra
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jeannie Park
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Skyler Kuhn
- Collaborative Bioinformatics Resource, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ramiro Iglesias-Bartolome
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
19
|
Silveira WA, Gonçalves DA, Machado J, Lautherbach N, Lustrino D, Paula-Gomes S, Pereira MG, Miyabara EH, Sandri M, Kettelhut IC, Navegantes LC. cAMP-dependent protein kinase inhibits FoxO activity and regulates skeletal muscle plasticity in mice. FASEB J 2020; 34:12946-12962. [PMID: 32772437 DOI: 10.1096/fj.201902102rr] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 07/16/2020] [Accepted: 07/21/2020] [Indexed: 01/04/2023]
Abstract
Although we have shown that catecholamines suppress the activity of the Ubiquitin-Proteasome System (UPS) and atrophy-related genes expression through a cAMP-dependent manner in skeletal muscle from rodents, the underlying mechanisms remain unclear. Here, we report that a single injection of norepinephrine (NE; 1 mg kg-1 ; s.c) attenuated the fasting-induced up-regulation of FoxO-target genes in tibialis anterior (TA) muscles by the stimulation of PKA/CREB and Akt/FoxO1 signaling pathways. In addition, muscle-specific activation of PKA by the overexpression of PKA catalytic subunit (PKAcat) suppressed FoxO reporter activity induced by (1) a wild-type; (2) a non-phosphorylatable; (3) a non-phosphorylatable and non-acetylatable forms of FoxO1 and FoxO3; (4) downregulation of FoxO protein content, and probably by (5) PGC-1α up-regulation. Consistently, the overexpression of the PKAcat inhibitor (PKI) up-regulated FoxO activity and the content of Atrogin-1 and MuRF1, as well as induced muscle fiber atrophy, the latter effect being prevented by the overexpression of a dominant negative (d. n.) form of FoxO (d.n.FoxO). The sustained overexpression of PKAcat induced fiber-type transition toward a smaller, slower, and more oxidative phenotype and improved muscle resistance to fatigue. Taken together, our data provide the first evidence that endogenous PKA activity is required to restrain the basal activity of FoxO and physiologically important to maintain skeletal muscle mass.
Collapse
Affiliation(s)
- Wilian A Silveira
- Departments of Physiology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil.,Institute of Biological and Natural Science, Federal University of Triângulo Mineiro (UFTM), Uberaba, Brazil
| | - Dawit A Gonçalves
- Departments of Physiology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil.,Departments of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil.,Department of Physical Education, School of Physical Education, Physiotherapy and Occupational Therapy, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil.,Department of Biomedical Sciences, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Juliano Machado
- Departments of Physiology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil.,Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Neuherberg, Germany
| | - Natalia Lautherbach
- Departments of Physiology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Danilo Lustrino
- Departments of Physiology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Silvia Paula-Gomes
- Departments of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Marcelo G Pereira
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Elen H Miyabara
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Marco Sandri
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy.,Myology Center, University of Padova, Padova, Italy
| | - Isis C Kettelhut
- Departments of Physiology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil.,Departments of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Luiz C Navegantes
- Departments of Physiology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
20
|
Keyes J, Ganesan A, Molinar-Inglis O, Hamidzadeh A, Zhang J, Ling M, Trejo J, Levchenko A, Zhang J. Signaling diversity enabled by Rap1-regulated plasma membrane ERK with distinct temporal dynamics. eLife 2020; 9:57410. [PMID: 32452765 PMCID: PMC7289600 DOI: 10.7554/elife.57410] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/22/2020] [Indexed: 12/23/2022] Open
Abstract
A variety of different signals induce specific responses through a common, extracellular-signal regulated kinase (ERK)-dependent cascade. It has been suggested that signaling specificity can be achieved through precise temporal regulation of ERK activity. Given the wide distrubtion of ERK susbtrates across different subcellular compartments, it is important to understand how ERK activity is temporally regulated at specific subcellular locations. To address this question, we have expanded the toolbox of Förster Resonance Energy Transfer (FRET)-based ERK biosensors by creating a series of improved biosensors targeted to various subcellular regions via sequence specific motifs to measure spatiotemporal changes in ERK activity. Using these sensors, we showed that EGF induces sustained ERK activity near the plasma membrane in sharp contrast to the transient activity observed in the cytoplasm and nucleus. Furthermore, EGF-induced plasma membrane ERK activity involves Rap1, a noncanonical activator, and controls cell morphology and EGF-induced membrane protrusion dynamics. Our work strongly supports that spatial and temporal regulation of ERK activity is integrated to control signaling specificity from a single extracellular signal to multiple cellular processes.
Collapse
Affiliation(s)
- Jeremiah Keyes
- Department of Pharmacology, University of California San Diego, La Jolla, United States
| | - Ambhighainath Ganesan
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, United States
| | - Olivia Molinar-Inglis
- Department of Pharmacology, University of California San Diego, La Jolla, United States
| | - Archer Hamidzadeh
- Department of Biomedical Engineering and Yale Systems Biology Institute, Yale University, New Haven, United States
| | - Jinfan Zhang
- Department of Pharmacology, University of California San Diego, La Jolla, United States
| | - Megan Ling
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, United States
| | - JoAnn Trejo
- Department of Pharmacology, University of California San Diego, La Jolla, United States
| | - Andre Levchenko
- Department of Biomedical Engineering and Yale Systems Biology Institute, Yale University, New Haven, United States
| | - Jin Zhang
- Department of Pharmacology, University of California San Diego, La Jolla, United States.,Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, United States.,Department of Bioengineering, University of California San Diego, La Jolla, United States
| |
Collapse
|
21
|
Integration of Rap1 and Calcium Signaling. Int J Mol Sci 2020; 21:ijms21051616. [PMID: 32120817 PMCID: PMC7084553 DOI: 10.3390/ijms21051616] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 02/07/2023] Open
Abstract
Ca2+ is a universal intracellular signal. The modulation of cytoplasmic Ca2+ concentration regulates a plethora of cellular processes, such as: synaptic plasticity, neuronal survival, chemotaxis of immune cells, platelet aggregation, vasodilation, and cardiac excitation–contraction coupling. Rap1 GTPases are ubiquitously expressed binary switches that alternate between active and inactive states and are regulated by diverse families of guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs). Active Rap1 couples extracellular stimulation with intracellular signaling through secondary messengers—cyclic adenosine monophosphate (cAMP), Ca2+, and diacylglycerol (DAG). Much evidence indicates that Rap1 signaling intersects with Ca2+ signaling pathways to control the important cellular functions of platelet activation or neuronal plasticity. Rap1 acts as an effector of Ca2+ signaling when activated by mechanisms involving Ca2+ and DAG-activated (CalDAG-) GEFs. Conversely, activated by other GEFs, such as cAMP-dependent GEF Epac, Rap1 controls cytoplasmic Ca2+ levels. It does so by regulating the activity of Ca2+ signaling proteins such as sarcoendoplasmic reticulum Ca2+-ATPase (SERCA). In this review, we focus on the physiological significance of the links between Rap1 and Ca2+ signaling and emphasize the molecular interactions that may offer new targets for the therapy of Alzheimer’s disease, hypertension, and atherosclerosis, among other diseases.
Collapse
|
22
|
Potential of epigenetic events in human thyroid cancer. Cancer Genet 2019; 239:13-21. [PMID: 31472323 DOI: 10.1016/j.cancergen.2019.08.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 07/27/2019] [Accepted: 08/22/2019] [Indexed: 12/26/2022]
Abstract
Thyroid cancer remains the highest prevailing endocrine malignancy, and its incidence rate has progressively increased in the previous years. Above 95% of thyroid tumor are follicular cells types of carcinoma in which are considered invasive type of tumor. The pathogenesis and molecular mechanism of thyroid tumors are yet remains elucidated, in spite of activating RET, RAS and BRAF carcinogenesis have been well introduced. Nemours molecular alterations have been defined and have revealed promise for their diagnostic, prognostic and therapeutic capacity but still need further confirmation. Among different types of mechanisms, the current article reviews the importance of epigenetic modifications in thyroid cancer. Increasing data from previous reports demonstrate that acquired epigenetic abnormalities together with genetic changes plays an important role in alteration of gene expression patterns. Aberrant DNA methylation has been well known in the CpG regions and profile of microRNAs (mi-RNAs) expression also involved in cancer development. In addition, the gene expression through epigenetic control contribution to thyroid cancer is analyzed and it is semi considered in the clinic. However the epigenetic of the thyroid cancer is yet remains in its early stages, and it carries encouraging potential thyroid cancer detections in its early stages, assessment of prognosis and targeted cancer treatment.
Collapse
|
23
|
Epac2 Elevation Reverses Inhibition by Chondroitin Sulfate Proteoglycans In Vitro and Transforms Postlesion Inhibitory Environment to Promote Axonal Outgrowth in an Ex Vivo Model of Spinal Cord Injury. J Neurosci 2019; 39:8330-8346. [PMID: 31409666 DOI: 10.1523/jneurosci.0374-19.2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 08/05/2019] [Accepted: 08/08/2019] [Indexed: 02/07/2023] Open
Abstract
Millions of patients suffer from debilitating spinal cord injury (SCI) without effective treatments. Elevating cAMP promotes CNS neuron growth in the presence of growth-inhibiting molecules. cAMP's effects on neuron growth are partly mediated by Epac, comprising Epac1 and Epac2; the latter predominantly expresses in postnatal neural tissue. Here, we hypothesized that Epac2 activation would enhance axonal outgrowth after SCI. Using in vitro assays, we demonstrated, for the first time, that Epac2 activation using a specific soluble agonist (S-220) significantly enhanced neurite outgrowth of postnatal rat cortical neurons and markedly overcame the inhibition by chondroitin sulfate proteoglycans and mature astrocytes on neuron growth. We further investigated the novel potential of Epac2 activation in promoting axonal outgrowth by an ex vivo rat model of SCI mimicking post-SCI environment in vivo and by delivering S-220 via a self-assembling Fmoc-based hydrogel that has suitable properties for SCI repair. We demonstrated that S-220 significantly enhanced axonal outgrowth across the lesion gaps in the organotypic spinal cord slices, compared with controls. Furthermore, we elucidated, for the first time, that Epac2 activation profoundly modulated the lesion environment by reducing astrocyte/microglial activation and transforming astrocytes into elongated morphology that guided outgrowing axons. Finally, we showed that S-220, when delivered by the gel at 3 weeks after contusion SCI in male adult rats, resulted in significantly better locomotor performance for up to 4 weeks after treatment. Our data demonstrate a promising therapeutic potential of S-220 in SCI, via beneficial effects on neurons and glia after injury to facilitate axonal outgrowth.SIGNIFICANCE STATEMENT During development, neuronal cAMP levels decrease significantly compared with the embryonic stage when the nervous system is established. This has important consequences following spinal cord injury, as neurons fail to regrow. Elevating cAMP levels encourages injured CNS neurons to sprout and extend neurites. We have demonstrated that activating its downstream effector, Epac2, enhances neurite outgrowth in vitro, even in the presence of an inhibitory environment. Using a novel biomaterial-based drug delivery system in the form of a hydrogel to achieve local delivery of an Epac2 agonist, we further demonstrated that specific activation of Epac2 enhances axonal outgrowth and minimizes glial activation in an ex vivo model of spinal cord injury, suggesting a new strategy for spinal cord repair.
Collapse
|
24
|
Sunilkumar S, Ford SM. Elevated glucose concentration in culture media decreases membrane trafficking of SGLT2 in LLC-PK 1 cells via a cAMP/PKA-dependent pathway. Am J Physiol Cell Physiol 2019; 316:C913-C924. [PMID: 30943059 DOI: 10.1152/ajpcell.00433.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Na+-dependent glucose reabsorption in the renal proximal tubule is dynamically regulated by changes in blood glucose levels. There is, however, a disparity in reports studying the relationship between hyperglycemia and Na+-glucose-linked transporter (SGLT) function and expression. Similarly, manipulation of the glucose content in growth media of cultured renal cells has been shown to influence SGLT activity. In this investigation, SGLT activity was significantly lower in proximal tubule LLC-PK1 cells cultured in medium containing 17.5 than 5 mM glucose. α-Methyl d-glucopyranoside (AMG) transport kinetics showed reduced apparent Vmax and Km in cells grown in 17.5 mM glucose. SGLT2 was identified as the isoform responsible for glucose transport, and protein expression analyses showed decreased apical membrane localization of SGLT2 in cells grown in 17.5 mM glucose, explaining the reduced activity. Multiple signaling pathways have been implicated in regulation of SGLT activity and trafficking. Elevated media glucose decreased intracellular cAMP and PKA activation, leading to decreased SGLT2 trafficking into the plasma membrane, which was reversed after treatment with 1 µM forskolin. The effects of media glucose on SGLT activity were found to be dependent on p38 MAPK activation due to PKA-mediated signaling. Glucose-modulated AMG uptake is reversible and was associated with altered SGLT2 membrane trafficking and cAMP alterations. In summary, elevated glucose concentrations in culture medium decrease SGLT activity in LLC-PK1 cells by reducing membrane trafficking of SGLT2 via decreasing intracellular cAMP, resulting in a lowered PKA-dependent phosphorylation of p38 MAPK.
Collapse
Affiliation(s)
| | - Sue M Ford
- Department of Pharmaceutical Sciences, St. John's University , Queens, New York
| |
Collapse
|
25
|
Li WP, Ma K, Jiang XY, Yang R, Lu PH, Nie BM, Lu Y. Molecular mechanism of panaxydol on promoting axonal growth in PC12 cells. Neural Regen Res 2018; 13:1927-1936. [PMID: 30233066 PMCID: PMC6183029 DOI: 10.4103/1673-5374.239439] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2018] [Indexed: 12/04/2022] Open
Abstract
Nerve growth factor (NGF) promotes axonal growth in PC12 cells primarily by regulating the RTK-RAS-MEK-ERK pathway. Panaxydol, a polyacetylene isolated from Panax notoginseng, can mimic the effects of NGF. Panaxydol promotes neurite outgrowth in PC12 cells, but its molecular mechanism remains unclear. Indeed, although alkynol compounds such as panaxydol can increase intracellular cyclic adenosine 3',5'-monophosphate (cAMP) levels and the ERK inhibitor U0126 inhibits alkynol-induced axonal growth, how pathways downstream of cAMP activate ERK have not been investigated. This study observed the molecular mechanism of panaxydol-, NGF- and forskolin-induced PC12 cell axon growth using specific signaling pathway inhibitors. The results demonstrated that although the RTK inhibitor SU5416 obviously inhibited the growth-promoting effect of NGF, it could not inhibit the promoting effect of panaxydol on axonal growth of PC12 cells. The adenylate cyclase inhibitor SQ22536 and cAMP-dependent protein kinase inhibitor RpcAMPS could suppress the promoting effect of forskolin and panaxydol on axonal growth. The ERK inhibitor U0126 inhibited axonal growth induced by all three factors. However, the PKA inhibitor H89 inhibited the promoting effect of forskolin on axonal growth but could not suppress the promoting effect of panaxydol. A western blot assay was used to determine the effects of stimulating factors and inhibitors on ERK phosphorylation levels. The results revealed that NGF activates the ERK pathway through tyrosine receptors to induce axonal growth of PC12 cells. In contrast, panaxydol and forskolin increased cellular cAMP levels and were inhibited by adenylyl cyclase inhibitors. The protein kinase A inhibitor H89 completely inhibited forskolin-induced axonal outgrowth and ERK phosphorylation, but could not inhibit panaxydol-induced axonal growth and ERK phosphorylation. These results indicated that panaxydol promoted axonal growth of PC12 cells through different pathways downstream of cAMP. Considering that exchange protein directly activated by cAMP 1 (Epac1) plays an important role in mediating cAMP signaling pathways, RNA interference experiments targeting the Epac1 gene were employed. The results verified that Epac1 could mediate the axonal growth signaling pathway induced by panaxydol. These findings suggest that compared with NGF and forskolin, panaxydol elicits axonal growth through the cAMP-Epac1-Rap1-MEK-ERK-CREB pathway, which is independent of PKA.
Collapse
Affiliation(s)
- Wei-Peng Li
- Department of Nuclear Medicine, the First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui Province, China
| | - Ke Ma
- Department of Pharmacy, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Yan Jiang
- Key Laboratory of Arrhythmias of Ministry of Education of China, Tongji University School of Medicine, Shanghai, China
| | - Rui Yang
- Department of Pharmacy, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pei-Hua Lu
- Department of Pharmacy, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bao-Ming Nie
- Department of Pharmacy, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang Lu
- Department of Pharmacy, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
26
|
Dumbacher M, Van Dooren T, Princen K, De Witte K, Farinelli M, Lievens S, Tavernier J, Dehaen W, Wera S, Winderickx J, Allasia S, Kilonda A, Spieser S, Marchand A, Chaltin P, Hoogenraad CC, Griffioen G. Modifying Rap1-signalling by targeting Pde6δ is neuroprotective in models of Alzheimer's disease. Mol Neurodegener 2018; 13:50. [PMID: 30257685 PMCID: PMC6158915 DOI: 10.1186/s13024-018-0283-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 09/13/2018] [Indexed: 01/06/2023] Open
Abstract
Background Neuronal Ca2+ dyshomeostasis and hyperactivity play a central role in Alzheimer’s disease pathology and progression. Amyloid-beta together with non-genetic risk-factors of Alzheimer’s disease contributes to increased Ca2+ influx and aberrant neuronal activity, which accelerates neurodegeneration in a feed-forward fashion. As such, identifying new targets and drugs to modulate excessive Ca2+ signalling and neuronal hyperactivity, without overly suppressing them, has promising therapeutic potential. Methods Here we show, using biochemical, electrophysiological, imaging, and behavioural tools, that pharmacological modulation of Rap1 signalling by inhibiting its interaction with Pde6δ normalises disease associated Ca2+ aberrations and neuronal activity, conferring neuroprotection in models of Alzheimer’s disease. Results The newly identified inhibitors of the Rap1-Pde6δ interaction counteract AD phenotypes, by reconfiguring Rap1 signalling underlying synaptic efficacy, Ca2+ influx, and neuronal repolarisation, without adverse effects in-cellulo or in-vivo. Thus, modulation of Rap1 by Pde6δ accommodates key mechanisms underlying neuronal activity, and therefore represents a promising new drug target for early or late intervention in neurodegenerative disorders. Conclusion Targeting the Pde6δ-Rap1 interaction has promising therapeutic potential for disorders characterised by neuronal hyperactivity, such as Alzheimer’s disease. Electronic supplementary material The online version of this article (10.1186/s13024-018-0283-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Michael Dumbacher
- reMYND NV, Gaston Geenslaan 1, Leuven-Heverlee, 3001, Belgium.,Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584CH, Utrecht, The Netherlands
| | - Tom Van Dooren
- reMYND NV, Gaston Geenslaan 1, Leuven-Heverlee, 3001, Belgium
| | - Katrien Princen
- reMYND NV, Gaston Geenslaan 1, Leuven-Heverlee, 3001, Belgium
| | - Koen De Witte
- reMYND NV, Gaston Geenslaan 1, Leuven-Heverlee, 3001, Belgium
| | - Mélissa Farinelli
- E-Phy-Science, IPMC, 660 route des Lucioles, 06560, Sophia Antipolis, France
| | - Sam Lievens
- Orionis Biosciences, Technologiepark 12B, Zwijnaarde-Ghent, 9052, Belgium.,Cytokine Receptor Lab, Flanders Institute of Biotechnology, Medical Biotechnology Center, Faculty of Medicine and Health Sciences, Ghent University, Albert Baertsoenkaai 3, 9000, Ghent, Belgium
| | - Jan Tavernier
- Cytokine Receptor Lab, Flanders Institute of Biotechnology, Medical Biotechnology Center, Faculty of Medicine and Health Sciences, Ghent University, Albert Baertsoenkaai 3, 9000, Ghent, Belgium
| | - Wim Dehaen
- Department of Chemistry, KU Leuven, Celestijnenlaan 200f - box 2404, Leuven-Heverlee, 3001, Belgium
| | - Stefaan Wera
- ViroVet NV, Ambachtenlaan 1, Leuven-Heverlee, 3001, Belgium
| | - Joris Winderickx
- Department of Biology, Functional Biology, KU Leuven, Kasteelpark Arenberg 31 box 2433, Leuven-Heverlee, 3001, Belgium
| | - Sara Allasia
- Cistim Leuven vzw, Gaston Geenslaan 2, Leuven-Heverlee, 3001, Belgium
| | - Amuri Kilonda
- Cistim Leuven vzw, Gaston Geenslaan 2, Leuven-Heverlee, 3001, Belgium
| | - Stéphane Spieser
- Cistim Leuven vzw, Gaston Geenslaan 2, Leuven-Heverlee, 3001, Belgium
| | - Arnaud Marchand
- Cistim Leuven vzw, Gaston Geenslaan 2, Leuven-Heverlee, 3001, Belgium
| | - Patrick Chaltin
- Cistim Leuven vzw, Gaston Geenslaan 2, Leuven-Heverlee, 3001, Belgium.,Center for Drug Design and Development (CD3), KU Leuven, Waaistraat 6, 3000, Leuven, Belgium
| | - Casper C Hoogenraad
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584CH, Utrecht, The Netherlands
| | | |
Collapse
|
27
|
Huk DJ, Ashtekar A, Magner A, La Perle K, Kirschner LS. Deletion of Rap1b, but not Rap1a or Epac1, Reduces Protein Kinase A-Mediated Thyroid Cancer. Thyroid 2018; 28:1153-1161. [PMID: 29882482 PMCID: PMC6154455 DOI: 10.1089/thy.2017.0528] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
BACKGROUND Thyroid cancer is an emerging health problem in the United States and worldwide. With incidence rates of thyroid cancer rapidly rising, the need to develop new treatment options is becoming a priority, and understanding the molecular mechanisms of this disease is crucial to furthering these efforts. Thyroid growth is driven by the TSH/cAMP/PKA signaling pathway, and it has previously been shown that activation of PKA through genetic ablation of the regulatory subunit Prkar1a (Prkar1a KO) is sufficient to cause follicular thyroid cancer in mouse models. cAMP also activates the Epac proteins and their downstream effectors, Rap1a and Rap1b. METHODS Previously, the authors' laboratory generated a mouse model of follicular thyroid cancer by conferring thyroid-specific deletion of Prkar1a (R1a-TpoKO). To probe the roles of other components of the PKA signaling system in the development of thyroid cancer, this study deleted Rap1 and Epac1 in the setting of the Prkar1a knockout. RESULTS Deletion of Rap1 significantly decreases thyroid size and cancer incidence in Prkar1a KO thyroids. Further, isoform-specific ablation of Rap1a and Rap1b implicates Rap1b as the downstream effector of PKA during thyroid carcinogenesis. In vivo modeling provides definitive evidence that Epac1 plays little role in thyroid proliferation and is dispensable for thyroid carcinogenesis arising from the deletion of Prkar1a. CONCLUSIONS This study demonstrate that PKA signaling to Rap1b is a key signaling node for follicular thyroid carcinogenesis, while Epac1 activity is not required for tumor development. This work sheds new light on the pathways involved in FTC development and identifies a possible target for the development of new therapies in the treatment of FTC.
Collapse
Affiliation(s)
- Danielle J. Huk
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio
| | - Amruta Ashtekar
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio
| | - Alexa Magner
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio
| | - Krista La Perle
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio
| | - Lawrence S. Kirschner
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio
- Division of Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
- Address correspondence to:Lawrence S. Kirschner, MD, PhDThe Ohio State University460 W 12th Ave, Rm 510Columbus, OH 43210
| |
Collapse
|
28
|
Jain R, Watson U, Vasudevan L, Saini DK. ERK Activation Pathways Downstream of GPCRs. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 338:79-109. [PMID: 29699693 DOI: 10.1016/bs.ircmb.2018.02.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
GPCRs, the 7-TM receptors, represent a class of cell surface receptors which modulate a variety of physiological responses. The serpentine structure in addition to contributing the diversity of stimuli these receptors can sense also provides flexibility to the extracellular and intracellular regions where other proteins can interact with and can form functionally active multimeric entities. The range in signaling and physiological responses generated by these receptors can be attributed to a large repertoire of the receptor subtypes as well as their differential coupling to various classes of G-protein subunits and other proteins which facilitate multistate activation. A multistate GPCR can engage diverse signaling molecules, thereby modulating not only the canonical cellular responses but also noncanonical responses typically associated with activation of other cascades such as RTK and MAPK/ERK signaling. Given the crucial involvement of MAP kinase/ERK signaling in cell fate determination specially with respect to regulating cell proliferation, cellular apoptosis, and survival, GPCR-mediated cross-activation of MAPK has been explored in various systems and shown to involve functional integration of multiple pathways. This review describes the present knowledge of the different mechanisms of ERK activation downstream of GPCRs and our present understanding of receptor-dependent and -independent MAPK activation cascades.
Collapse
Affiliation(s)
- Ruchi Jain
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| | - Uchenna Watson
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India; Department of Studies in Zoology, University of Mysore, Manasagangothri, Mysore, India
| | - Lakshmi Vasudevan
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India; L-GEST-Laboratory of Toxicology, Faculty of Pharmaceutical Sciences, Ghent, Belgium
| | - Deepak K Saini
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India; Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India.
| |
Collapse
|
29
|
Shah S, Brock EJ, Ji K, Mattingly RR. Ras and Rap1: A tale of two GTPases. Semin Cancer Biol 2018; 54:29-39. [PMID: 29621614 DOI: 10.1016/j.semcancer.2018.03.005] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 03/16/2018] [Accepted: 03/29/2018] [Indexed: 02/07/2023]
Abstract
Ras oncoproteins play pivotal roles in both the development and maintenance of many tumor types. Unfortunately, these proteins are difficult to directly target using traditional pharmacological strategies, in part due to their lack of obvious binding pockets or allosteric sites. This obstacle has driven a considerable amount of research into pursuing alternative ways to effectively inhibit Ras, examples of which include inducing mislocalization to prevent Ras maturation and inactivating downstream proteins in Ras-driven signaling pathways. Ras proteins are archetypes of a superfamily of small GTPases that play specific roles in the regulation of many cellular processes, including vesicle trafficking, nuclear transport, cytoskeletal rearrangement, and cell cycle progression. Several other superfamily members have also been linked to the control of normal and cancer cell growth and survival. For example, Rap1 has high sequence similarity to Ras, has overlapping binding partners, and has been demonstrated to both oppose and mimic Ras-driven cancer phenotypes. Rap1 plays an important role in cell adhesion and integrin function in a variety of cell types. Mechanistically, Ras and Rap1 cooperate to initiate and sustain ERK signaling, which is activated in many malignancies and is the target of successful therapeutics. Here we review the role activated Rap1 in ERK signaling and other downstream pathways to promote invasion and cell migration and metastasis in various cancer types.
Collapse
Affiliation(s)
- Seema Shah
- Program in Cancer Biology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Ethan J Brock
- Program in Cancer Biology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Kyungmin Ji
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Raymond R Mattingly
- Program in Cancer Biology, Wayne State University School of Medicine, Detroit, MI 48201, USA; Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
30
|
Synthesis and degradation of cAMP in Giardia lamblia: possible role and characterization of a nucleotidyl cyclase with a single cyclase homology domain. Biochem J 2017; 474:4001-4017. [PMID: 29054977 DOI: 10.1042/bcj20170590] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 10/17/2017] [Accepted: 10/19/2017] [Indexed: 01/25/2023]
Abstract
Despite its importance in the regulation of growth and differentiation processes of a variety of organisms, the mechanism of synthesis and degradation of cAMP (cyclic AMP) has not yet been described in Giardia lamblia In this work, we measured significant quantities of cAMP in trophozoites of G. lamblia incubated in vitro and later detected how it increases during the first hours of encystation, and how it then returns to basal levels at 24 h. Through an analysis of the genome of G. lamblia, we found sequences of three putative enzymes - one phosphodiesterase (gPDE) and two nucleotidyl cyclases (gNC1 and gNC2) - that should be responsible for the regulation of cAMP in G. lamblia Later, an RT-PCR assay confirmed that these three genes are expressed in trophozoites. The bioinformatic analysis indicated that gPDE is a transmembrane protein of 154 kDa, with a single catalytic domain in the C-terminal end; gNC1 is predicted to be a transmembrane protein of 74 kDa, with only one class III cyclase homology domain (CHD) at the C-terminal end; and gNC2 should be a transmembrane protein of 246 kDa, with two class III CHDs. Finally, we cloned and enriched the catalytic domain of gNC1 (gNC1cd) from bacteria. After that, we confirmed that gNC1cd has adenylyl cyclase (AC) activity. This enzymatic activity depends on the presence of Mn2+ and Ca2+, but no significant activity was displayed in the presence of Mg2+ Additionally, the AC activity of gNC1cd is competitively inhibited with GTP, so it is highly possible that gNC1 has guanylyl cyclase activity as well.
Collapse
|
31
|
Phosphorylation of the C-Raf N Region Promotes Raf Dimerization. Mol Cell Biol 2017; 37:MCB.00132-17. [PMID: 28694330 DOI: 10.1128/mcb.00132-17] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 06/28/2017] [Indexed: 12/11/2022] Open
Abstract
The activation of Raf kinases by the small GTPase Ras requires two major sets of phosphorylations. One set lies within the activation loop, and the other lies within the N-terminal acidic region (N region). In the most abundant isoform of Raf, C-Raf, N-region phosphorylations occur on serine 338 (S338) and tyrosine 341 (Y341) and are thought to provide allosteric activation of the Raf dimer. We show that the phosphorylations of these N-region sites does not require C-Raf dimerization, but rather, they precede dimerization. One of these phosphorylations (phospho-Y341) is required for C-Raf dimerization, and this action can be replicated by phosphomimetic mutants both in vivo and in vitro The role of the phosphorylation of Y341 in promoting Raf dimerization is distinct from its well-known function in facilitating S338 phosphorylation. In Ras mutant pancreatic cancer cell lines, the phosphorylation and dimerization of C-Raf are basally elevated. Dimerization is thought to contribute to their elevated growth rate through their activation of the mitogen-activated protein (MAP) kinase (extracellular signal-regulated kinase [ERK]) signaling cascade. Blocking the tyrosine phosphorylation of C-Raf with Src family inhibitors blocks growth, basal dimerization, and ERK activation in these cells. We suggest that the kinases mediating C-Raf Y341 phosphorylation are potential candidate drug targets in selected Ras-dependent cancers.
Collapse
|
32
|
Jȩdrzejewska-Szmek J, Luczak V, Abel T, Blackwell KT. β-adrenergic signaling broadly contributes to LTP induction. PLoS Comput Biol 2017; 13:e1005657. [PMID: 28742159 PMCID: PMC5546712 DOI: 10.1371/journal.pcbi.1005657] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 08/07/2017] [Accepted: 06/27/2017] [Indexed: 12/18/2022] Open
Abstract
Long-lasting forms of long-term potentiation (LTP) represent one of the major cellular mechanisms underlying learning and memory. One of the fundamental questions in the field of LTP is why different molecules are critical for long-lasting forms of LTP induced by diverse experimental protocols. Further complexity stems from spatial aspects of signaling networks, such that some molecules function in the dendrite and some are critical in the spine. We investigated whether the diverse experimental evidence can be unified by creating a spatial, mechanistic model of multiple signaling pathways in hippocampal CA1 neurons. Our results show that the combination of activity of several key kinases can predict the occurrence of long-lasting forms of LTP for multiple experimental protocols. Specifically Ca2+/calmodulin activated kinase II, protein kinase A and exchange protein activated by cAMP (Epac) together predict the occurrence of LTP in response to strong stimulation (multiple trains of 100 Hz) or weak stimulation augmented by isoproterenol. Furthermore, our analysis suggests that activation of the β-adrenergic receptor either via canonical (Gs-coupled) or non-canonical (Gi-coupled) pathways underpins most forms of long-lasting LTP. Simulations make the experimentally testable prediction that a complete antagonist of the β-adrenergic receptor will likely block long-lasting LTP in response to strong stimulation. Collectively these results suggest that converging molecular mechanisms allow CA1 neurons to flexibly utilize signaling mechanisms best tuned to temporal pattern of synaptic input to achieve long-lasting LTP and memory storage. Long-term potentiation of the strength of synaptic connections is a mechanism of learning and memory storage. One of the most confusing aspects of hippocampal synaptic potentiation is that numerous experiments have revealed the requirement for a plethora of signaling molecules. Furthermore the degree to which molecules activated by the stress response modify hippocampal synaptic potentiation and memory is still unclear. We used a computational model to demonstrate that this molecular diversity can be explained by considering a combination of several key molecules. We also show that activation of β-adrenergic receptors by the stress response appears to be involved in most forms of synaptic potentiation, though in some cases unconventional mechanisms are utilized. This suggests that novel treatments for stress-related disorders may have more success if they target unconventional mechanisms activated by β-adrenergic receptors.
Collapse
Affiliation(s)
- Joanna Jȩdrzejewska-Szmek
- The Krasnow Institute for Advanced Studies, George Mason University, Fairfax, Virginia, United States of America
| | - Vincent Luczak
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ted Abel
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Kim T Blackwell
- The Krasnow Institute for Advanced Studies, George Mason University, Fairfax, Virginia, United States of America
- * E-mail:
| |
Collapse
|
33
|
Emery AC, Xu W, Eiden MV, Eiden LE. Guanine nucleotide exchange factor Epac2-dependent activation of the GTP-binding protein Rap2A mediates cAMP-dependent growth arrest in neuroendocrine cells. J Biol Chem 2017; 292:12220-12231. [PMID: 28546426 DOI: 10.1074/jbc.m117.790329] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 05/23/2017] [Indexed: 11/06/2022] Open
Abstract
First messenger-dependent activation of MAP kinases in neuronal and endocrine cells is critical for cell differentiation and function and requires guanine nucleotide exchange factor (GEF)-mediated activation of downstream Ras family small GTPases, which ultimately lead to ERK, JNK, and p38 phosphorylation. Because there are numerous GEFs and also a host of Ras family small GTPases, it is important to know which specific GEF-small GTPase dyad functions in a given cellular process. Here we investigated the upstream activators and downstream effectors of signaling via the GEF Epac2 in the neuroendocrine NS-1 cell line. Three cAMP sensors, Epac2, PKA, and neuritogenic cAMP sensor-Rapgef2, mediate distinct cellular outputs: p38-dependent growth arrest, cAMP response element-binding protein-dependent cell survival, and ERK-dependent neuritogenesis, respectively, in these cells. Previously, we found that cAMP-induced growth arrest of PC12 and NS-1 cells requires Epac2-dependent activation of p38 MAP kinase, which posed the important question of how Epac2 engages p38 without simultaneously activating other MAP kinases in neuronal and endocrine cells. We now show that the small GTP-binding protein Rap2A is the obligate effector for, and GEF substrate of, Epac2 in mediating growth arrest through p38 activation in NS-1 cells. This new pathway is distinctly parcellated from the G protein-coupled receptor → Gs → adenylate cyclase → cAMP → PKA → cAMP response element-binding protein pathway mediating cell survival and the G protein-coupled receptor → Gs → adenylate cyclase → cAMP → neuritogenic cAMP sensor-Rapgef2 → B-Raf → MEK → ERK pathway mediating neuritogenesis in NS-1 cells.
Collapse
Affiliation(s)
- Andrew C Emery
- Section on Molecular Neuroscience, National Institute of Mental Health Intramural Research Program, Bethesda, Maryland 20892
| | - Wenqin Xu
- Section on Molecular Neuroscience, National Institute of Mental Health Intramural Research Program, Bethesda, Maryland 20892
| | - Maribeth V Eiden
- Office of the Scientific Director, National Institute of Mental Health Intramural Research Program, Bethesda, Maryland 20892
| | - Lee E Eiden
- Section on Molecular Neuroscience, National Institute of Mental Health Intramural Research Program, Bethesda, Maryland 20892.
| |
Collapse
|
34
|
Multiplexing PKA and ERK1&2 kinases FRET biosensors in living cells using single excitation wavelength dual colour FLIM. Sci Rep 2017; 7:41026. [PMID: 28106114 PMCID: PMC5247693 DOI: 10.1038/srep41026] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 12/14/2016] [Indexed: 02/04/2023] Open
Abstract
Monitoring of different signalling enzymes in a single assay using multiplex biosensing provides a multidimensional workspace to elucidate biological processes, signalling pathway crosstalk, and determine precise sequence of events at the single living cell level. In this study, we interrogate the complexity in cAMP/PKA-MAPK/ERK1&2 crosstalk by using multi-parameter biosensing experiments to correlate biochemical activities simultaneously in time and space. Using a single excitation wavelength dual colour FLIM method we are able to detect fluorescence lifetime images of two donors to simultaneously measure PKA and ERK1&2 kinase activities in the same cellular localization by using FRET biosensors. To this end, we excite two FRET donors mTFP1 and LSSmOrange with a 440 nm wavelength and we alleviate spectral bleed-through associated limitations with the very dim-fluorescent acceptor ShadowG for mTFP1 and the red-shifted mKate2 for LSSmOrange. The simultaneous recording of PKA and ERK1&2 kinase activities reveals concomitant EGF-mediated activations of both kinases in HeLa cells. Under these conditions the subsequent Forskolin-induced cAMP release reverses the transient increase of EGF-mediated ERK1&2 kinase activity while reinforcing PKA activation. Here we propose a validated methodology for multiparametric kinase biosensing in living cells using FRET-FLIM.
Collapse
|
35
|
Takahashi M, Li Y, Dillon TJ, Stork PJS. Phosphorylation of Rap1 by cAMP-dependent Protein Kinase (PKA) Creates a Binding Site for KSR to Sustain ERK Activation by cAMP. J Biol Chem 2016; 292:1449-1461. [PMID: 28003362 DOI: 10.1074/jbc.m116.768986] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 12/16/2016] [Indexed: 12/31/2022] Open
Abstract
Cyclic adenosine monophosphate (cAMP) is an important mediator of hormonal stimulation of cell growth and differentiation through its activation of the extracellular signal-regulated kinase (ERK) cascade. Two small G proteins, Ras and Rap1 have been proposed to mediate this activation. Using HEK293 cells as a model system, we have recently shown that both Ras and Rap1 are required for cAMP signaling to ERKs. However, cAMP-dependent Ras signaling to ERKs is transient and rapidly terminated by PKA phosphorylation of the Raf isoforms C-Raf and B-Raf. In contrast, cAMP-dependent Rap1 signaling to ERKs and Rap1 is potentiated by PKA. We show that this is due to sustained binding of B-Raf to Rap1. One of the targets of PKA is Rap1 itself, directly phosphorylating Rap1a on serine 180 and Rap1b on serine 179. We show that these phosphorylations create potential binding sites for the adaptor protein 14-3-3 that links Rap1 to the scaffold protein KSR. These results suggest that Rap1 activation of ERKs requires PKA phosphorylation and KSR binding. Because KSR and B-Raf exist as heterodimers within the cell, this binding also brings B-Raf to Rap1, allowing Rap1 to couple to ERKs through B-Raf binding to Rap1 independently of its Ras-binding domain.
Collapse
Affiliation(s)
- Maho Takahashi
- From the Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239-3098
| | - Yanping Li
- From the Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239-3098
| | - Tara J Dillon
- From the Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239-3098
| | - Philip J S Stork
- From the Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239-3098
| |
Collapse
|
36
|
Miklos W, Heffeter P, Pirker C, Hager S, Kowol CR, van Schoonhoven S, Stojanovic M, Keppler BK, Berger W. Loss of phosphodiesterase 4D mediates acquired triapine resistance via Epac-Rap1-Integrin signaling. Oncotarget 2016; 7:84556-84574. [PMID: 27602951 PMCID: PMC5356681 DOI: 10.18632/oncotarget.11821] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 08/24/2016] [Indexed: 12/27/2022] Open
Abstract
Triapine, an anticancer thiosemicarbazone, is currently under clinical investigation. Whereas promising results were obtained in hematological diseases, trials in solid tumors widely failed. To understand mechanisms causing triapine insensitivity, we have analysed genomic alterations in a triapine-resistant SW480 subline (SW480/tria). Only one distinct genomic loss was observed specifically in SW480/tria cells affecting the phosphodiesterase 4D (PDE4D) gene locus. Accordingly, pharmacological inhibition of PDE4D resulted in significant triapine resistance in SW480 cells. Hence, we concluded that enhanced cyclic AMP levels might confer protection against triapine. Indeed, hyperactivation of both major downstream pathways, namely the protein kinase A (PKA)-cAMP response element-binding protein (Creb) and the exchange protein activated by cAMP (Epac)-Ras-related protein 1 (Rap1) signaling axes, was observed in SW480/tria cells. Unexpectedly, inhibition of PKA did not re-sensitize SW480/tria cells against triapine. In contrast, Epac activation resulted in distinct triapine resistance in SW480 cells. Conversely, knock-down of Epac expression and pharmacological inhibition of Rap1 re-sensitized SW480/tria cells against triapine. Rap1 is a well-known regulator of integrins. Accordingly, SW480/tria cells displayed enhanced plasma membrane expression of several integrin subunits, enhanced adhesion especially to RGD-containing matrix components, and bolstered activation/expression of the integrin downstream effectors Src and RhoA/Rac. Accordingly, integrin and Src inhibition resulted in potent triapine re-sensitization especially of SW480/tria cells. In summary, we describe for the first time integrin activation based on cAMP-Epac-Rap1 signaling as acquired drug resistance mechanism. combinations of triapine with inhibitors of several steps in this resistance cascade might be feasible strategies to overcome triapine insensitivity of solid tumors.
Collapse
Affiliation(s)
- Walter Miklos
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
| | - Petra Heffeter
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
- Research Platform “Translational Cancer Therapy Research”, University Vienna and Medical University Vienna, Vienna, Austria
| | - Christine Pirker
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
| | - Sonja Hager
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
| | - Christian R. Kowol
- Institute of Inorganic Chemistry, University of Vienna, A-1090 Vienna, Austria
- Research Platform “Translational Cancer Therapy Research”, University Vienna and Medical University Vienna, Vienna, Austria
| | - Sushilla van Schoonhoven
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
| | - Mirjana Stojanovic
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
| | - Bernhard K. Keppler
- Institute of Inorganic Chemistry, University of Vienna, A-1090 Vienna, Austria
- Research Platform “Translational Cancer Therapy Research”, University Vienna and Medical University Vienna, Vienna, Austria
| | - Walter Berger
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
- Research Platform “Translational Cancer Therapy Research”, University Vienna and Medical University Vienna, Vienna, Austria
| |
Collapse
|
37
|
Baameur F, Singhmar P, Zhou Y, Hancock JF, Cheng X, Heijnen CJ, Kavelaars A. Epac1 interacts with importin β1 and controls neurite outgrowth independently of cAMP and Rap1. Sci Rep 2016; 6:36370. [PMID: 27808165 PMCID: PMC5093460 DOI: 10.1038/srep36370] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 10/14/2016] [Indexed: 01/23/2023] Open
Abstract
Exchange protein directly activated by cAMP-1 (Epac1) is a cAMP sensor that regulates multiple cellular functions including cellular migration, proliferation and differentiation. Classically, Epac1 is thought to exert its effects through binding of cAMP leading to a conformational change in Epac1 and its accumulation at the plasma membrane (PM) where it activates Rap1. In search for regulators of Epac1 activity, we show here that importin β1 (impβ1) is an Epac1 binding partner that prevents PM accumulation of Epac1. We demonstrate that in the absence of impβ1, endogenous as well as overexpressed Epac1 accumulate at the PM. Moreover, agonist-induced PM translocation of Epac1 leads to dissociation of Epac1 from impβ1. Localization of Epac1 at the PM in the absence of impβ1, requires residue R82 in its DEP domain. Notably, the PM accumulation of Epac1 in the absence of impβ1 does not require binding of cAMP to Epac1 and does not result in Rap1 activation. Functionally, PM accumulation of Epac1, an Epac1 mutant deficient in cAMP binding, or an Epac1 mutant tethered to the PM, is sufficient to inhibit neurite outgrowth. In conclusion, we uncover a cAMP-independent function of Epac1 at the PM and demonstrate that impβ1 controls subcellular localization of Epac1.
Collapse
Affiliation(s)
- Faiza Baameur
- Laboratory of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Pooja Singhmar
- Laboratory of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yong Zhou
- Department of Integrative Biology and Pharmacology and Texas Therapeutics Institute, The University of Texas Health Science Center, Houston, TX 77030, USA
| | - John F Hancock
- Department of Integrative Biology and Pharmacology and Texas Therapeutics Institute, The University of Texas Health Science Center, Houston, TX 77030, USA
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology and Texas Therapeutics Institute, The University of Texas Health Science Center, Houston, TX 77030, USA
| | - Cobi J Heijnen
- Laboratory of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Annemieke Kavelaars
- Laboratory of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
38
|
Host Epac1 is required for cAMP-mediated invasion by Trypanosoma cruzi. Mol Biochem Parasitol 2016; 211:67-70. [PMID: 27984073 DOI: 10.1016/j.molbiopara.2016.10.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 10/25/2016] [Accepted: 10/26/2016] [Indexed: 02/03/2023]
Abstract
Mechanistic details of the modulation by cAMP of Trypanosoma cruzi host cell invasion remain ill-defined. Here we report that activation of host's Epac1 stimulated invasion, whereas specific pharmacological inhibition or maneuvers that alter Epac1 subcellular localization significantly reduced invasion. Furthermore, while specific activation of host cell PKA showed no effect, its inhibition resulted in an increased invasion, revealing a crosstalk between the PKA and Epac signaling pathways during the process of invasion. Therefore, our data suggests that subcellular localization of Epac might be playing an important role during invasion and that specific activation of the host cell cAMP/Epac1 pathway is required for cAMP-mediated invasion.
Collapse
|
39
|
Zhou L, Ma SL, Yeung PKK, Wong YH, Tsim KWK, So KF, Lam LCW, Chung SK. Anxiety and depression with neurogenesis defects in exchange protein directly activated by cAMP 2-deficient mice are ameliorated by a selective serotonin reuptake inhibitor, Prozac. Transl Psychiatry 2016; 6:e881. [PMID: 27598965 PMCID: PMC5048194 DOI: 10.1038/tp.2016.129] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 05/03/2016] [Accepted: 05/05/2016] [Indexed: 01/29/2023] Open
Abstract
Intracellular cAMP and serotonin are important modulators of anxiety and depression. Fluoxetine, a selective serotonin reuptake inhibitor (SSRI) also known as Prozac, is widely used against depression, potentially by activating cAMP response element-binding protein (CREB) and increasing brain-derived neurotrophic factor (BDNF) through protein kinase A (PKA). However, the role of Epac1 and Epac2 (Rap guanine nucleotide exchange factors, RAPGEF3 and RAPGEF4, respectively) as potential downstream targets of SSRI/cAMP in mood regulations is not yet clear. Here, we investigated the phenotypes of Epac1 (Epac1(-/-)) or Epac2 (Epac2(-/-)) knockout mice by comparing them with their wild-type counterparts. Surprisingly, Epac2(-/-) mice exhibited a wide range of mood disorders, including anxiety and depression with learning and memory deficits in contextual and cued fear-conditioning tests without affecting Epac1 expression or PKA activity. Interestingly, rs17746510, one of the three single-nucleotide polymorphisms (SNPs) in RAPGEF4 associated with cognitive decline in Chinese Alzheimer's disease (AD) patients, was significantly correlated with apathy and mood disturbance, whereas no significant association was observed between RAPGEF3 SNPs and the risk of AD or neuropsychiatric inventory scores. To further determine the detailed role of Epac2 in SSRI/serotonin/cAMP-involved mood disorders, we treated Epac2(-/-) mice with a SSRI, Prozac. The alteration in open field behavior and impaired hippocampal cell proliferation in Epac2(-/-) mice were alleviated by Prozac. Taken together, Epac2 gene polymorphism is a putative risk factor for mood disorders in AD patients in part by affecting the hippocampal neurogenesis.
Collapse
Affiliation(s)
- L Zhou
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - S L Ma
- Department of Psychiatry, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - P K K Yeung
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China,State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Y H Wong
- Division of Life Science and the Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong SAR, China,State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong SAR, China
| | - K W K Tsim
- State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong SAR, China,Division of Life Science and Center for Chinese Medicine, Hong Kong University of Science and Technology, Clear Water Bay, Clear Water Bay, Hong Kong SAR, China
| | - K F So
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China,Research Center of Heart, Brain, Hormone and Healthy Aging, The University of Hong Kong, Pokfulam, Hong Kong SAR, China,State Key Laboratory of Brain and Cognitive Science, The University of Hong Kong, Pokfulam, Hong Kong SAR, China,Department of Ophthalmology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - L C W Lam
- Department of Psychiatry, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - S K Chung
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China,State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China,Research Center of Heart, Brain, Hormone and Healthy Aging, The University of Hong Kong, Pokfulam, Hong Kong SAR, China,School of Biomedical Sciences, The University of Hong Kong, 1/F, Laboratory Block, Faculty of Medicine Building, 21 Sassoon Road, Pokfulam, Hong Kong SAR, China. E-mail:
| |
Collapse
|
40
|
Li Y, Dillon TJ, Takahashi M, Earley KT, Stork PJS. Protein Kinase A-independent Ras Protein Activation Cooperates with Rap1 Protein to Mediate Activation of the Extracellular Signal-regulated Kinases (ERK) by cAMP. J Biol Chem 2016; 291:21584-21595. [PMID: 27531745 DOI: 10.1074/jbc.m116.730978] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 08/03/2016] [Indexed: 11/06/2022] Open
Abstract
Cyclic adenosine monophosphate (cAMP) is an important mediator of hormonal stimulation of cell growth and differentiation through its activation of the extracellular signal-regulated kinase (ERK) cascade. Two small G proteins, Ras and Rap1, have been proposed to mediate this activation, with either Ras or Rap1 acting in distinct cell types. Using Hek293 cells, we show that both Ras and Rap1 are required for cAMP signaling to ERKs. The roles of Ras and Rap1 were distinguished by their mechanism of activation, dependence on the cAMP-dependent protein kinase (PKA), and the magnitude and kinetics of their effects on ERKs. Ras was required for the early portion of ERK activation by cAMP and was activated independently of PKA. Ras activation required the Ras/Rap guanine nucleotide exchange factor (GEF) PDZ-GEF1. Importantly, this action of PDZ-GEF1 was disrupted by mutation within its putative cyclic nucleotide-binding domain within PDZ-GEF1. Compared with Ras, Rap1 activation of ERKs was of longer duration. Rap1 activation was dependent on PKA and required Src family kinases and the Rap1 exchanger C3G. This is the first report of a mechanism for the cooperative actions of Ras and Rap1 in cAMP activation of ERKs. One physiological role for the sustained activation of ERKs is the transcription and stabilization of a range of transcription factors, including c-FOS. We show that the induction of c-FOS by cAMP required both the early and sustained phases of ERK activation, requiring Ras and Rap1, as well as for each of the Raf isoforms, B-Raf and C-Raf.
Collapse
Affiliation(s)
- Yanping Li
- From the Vollum Institute, Oregon Health and Science University, Portland, Oregon 97239-3098
| | - Tara J Dillon
- From the Vollum Institute, Oregon Health and Science University, Portland, Oregon 97239-3098
| | - Maho Takahashi
- From the Vollum Institute, Oregon Health and Science University, Portland, Oregon 97239-3098
| | - Keith T Earley
- From the Vollum Institute, Oregon Health and Science University, Portland, Oregon 97239-3098
| | - Philip J S Stork
- From the Vollum Institute, Oregon Health and Science University, Portland, Oregon 97239-3098
| |
Collapse
|
41
|
Alonso N, Diaz Nebreda A, Monczor F, Gutkind JS, Davio C, Fernandez N, Shayo C. PI3K pathway is involved in ERK signaling cascade activation by histamine H2R agonist in HEK293T cells. Biochim Biophys Acta Gen Subj 2016; 1860:1998-2007. [PMID: 27316323 DOI: 10.1016/j.bbagen.2016.06.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 06/12/2016] [Accepted: 06/13/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND Histamine, through histamine H2 receptor (H2R), modulates different biological processes, involving the modulation of PI3K/AKT/mTOR and RAS/RAF/MEK/ERK pathways. Many evidences have demonstrated the existence and importance of the crossregulation between these two signaling pathways. The aim of the present work was to determine the molecular mechanisms leading to PI3K and ERK pathways modulation induced by the H2R agonist amthamine and to evaluate the possible interplay between them. METHODS Phosphorylation levels of ERK and Akt were examined by Western blot in HEK293T cells expressing the human H2R, in the presence of H2R agonist and dominant negative mutants or pharmacological inhibitors of different proteins/pathways. Transcriptional activity assays were assessed to determine SRE activity. Amthamine-mediated cellular proliferation was investigated in MA-10A cells in the presence of PI3K inhibitor. RESULTS H2R agonist inhibits PI3K/Akt/mTOR and stimulates Ras/MEK/ERK pathways. Moreover, PI3K/Akt/mTOR signaling inhibition is necessary to achieve H2R mediated ERK activation. In the presence of a constitutive active mutant of Akt, amthamine is not able to mediate ERK activation. This crosstalk affects classical ERK downstream targets such as Elk1 phosphorylation and the transcriptional activity of the SRE, classically associated to proliferation. We further demonstrate that amthamine-induced proliferation in Leydig MA-10 tumor cells, is enhanced by LY294002, a PI3K inhibitor. CONCLUSIONS These results describe a crosstalk between PI3K/AKT/mTOR and Ras/MEK/ERK pathways induced by H2R stimulation with implications in cell proliferation. GENERAL SIGNIFICANCE This work indicates that the modulation of PI3K/AKT/mTOR pathway by H2R in turn regulates Ras/MEK/ERK activation conditioning the proliferative capacity of the cells.
Collapse
Affiliation(s)
- Natalia Alonso
- IBYME, Instituto de Biología y Medicina Experimental, Laboratorio de Patología y Farmacología Molecular, CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Antonela Diaz Nebreda
- IBYME, Instituto de Biología y Medicina Experimental, Laboratorio de Patología y Farmacología Molecular, CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Federico Monczor
- Cátedra de Química Medicinal, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina; ININFA, Instituto de Investigaciones Farmacológicas, UBA-CONICET, Facultad de Farmacia y Bioquímica, Junin 956, Ciudad Autónoma de Buenos Aires, Argentina
| | - J Silvio Gutkind
- Oral and Pharyngeal Cancer Branch, National Institute of Dental Research, NIH, Bethesda, MD, USA
| | - Carlos Davio
- Cátedra de Química Medicinal, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina; ININFA, Instituto de Investigaciones Farmacológicas, UBA-CONICET, Facultad de Farmacia y Bioquímica, Junin 956, Ciudad Autónoma de Buenos Aires, Argentina
| | - Natalia Fernandez
- Cátedra de Química Medicinal, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina; ININFA, Instituto de Investigaciones Farmacológicas, UBA-CONICET, Facultad de Farmacia y Bioquímica, Junin 956, Ciudad Autónoma de Buenos Aires, Argentina
| | - Carina Shayo
- IBYME, Instituto de Biología y Medicina Experimental, Laboratorio de Patología y Farmacología Molecular, CONICET, Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
42
|
Activation of MEK/ERK Signaling by PACAP in Guinea Pig Cardiac Neurons. J Mol Neurosci 2016; 59:309-16. [PMID: 27194157 DOI: 10.1007/s12031-016-0766-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 04/29/2016] [Indexed: 10/21/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) signaling can increase guinea pig cardiac neuron excitability in part through extracellular signal-regulated kinase (ERK) activation. The present study examined the PACAP receptors and signaling cascades that stimulate guinea pig cardiac neuron ERK signaling using confocal microscopy to quantify PACAP-induced neuronal phosphorylated ERK (pERK) immunoreactivity. PACAP and maxadilan, but not vasoactive intestinal polypeptide (VIP), increased cardiac neuron pERK, implicating primary roles for PACAP-selective PAC1 receptor (Adcyap1r1) signaling rather than VPAC receptors (Vipr1 and Vipr2) in the generation of cardiac neuron pERK. The adenylyl cyclase (AC) activator forskolin, but not the protein kinase C (PKC) activator phorbol myristate acetate (PMA), increased pERK. Also, Bim1 did not blunt PACAP activation of pERK. Together, the results suggest PAC1 receptor signal transduction via Gs/adenylyl cyclase (AC)/cAMP rather than Gq/phospholipase C (PLC) generated neuronal pERK. Activator and inhibitor studies suggested that the PACAP-mediated pERK activation was PKA-dependent rather than an exchange protein directly activated by a cAMP (EPAC), PKA-independent mechanism. The PACAP-induced pERK was inhibited by the clathrin inhibitor Pitstop2 to block receptor internalization and endosomal signaling. We propose that the PACAP-mediated MEK/ERK activation in cardiac neurons involves both AC/cAMP/PKA signaling and PAC1 receptor internalization/activation of signaling endosomes.
Collapse
|
43
|
Zou M, Baitei EY, Al-Rijjal RA, Parhar RS, Al-Mohanna FA, Kimura S, Pritchard C, BinEssa H, Alanazi AA, Alzahrani AS, Akhtar M, Assiri AM, Meyer BF, Shi Y. KRAS(G12D)-mediated oncogenic transformation of thyroid follicular cells requires long-term TSH stimulation and is regulated by SPRY1. J Transl Med 2015; 95:1269-77. [PMID: 26146959 PMCID: PMC6289253 DOI: 10.1038/labinvest.2015.90] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 05/05/2015] [Indexed: 11/09/2022] Open
Abstract
KRAS(G12D) can cause lung cancer rapidly, but is not sufficient to induce thyroid cancer. It is not clear whether long-term serum thyroid stimulating hormone (TSH) stimulation can promote KRAS(G12D)-mediated thyroid follicular cell transformation. In the present study, we investigated the effect of long-term TSH stimulation in KRAS(G12D) knock-in mice and the role of Sprouty1 (SPRY1) in KRAS(G12D)-mediated signaling. We used TPO-KRAS(G12D) mice for thyroid-specific expression of KRAS(G12D) under the endogenous KRAS promoter. Twenty TPO-KRAS(G12D) mice were given anti-thyroid drug propylthiouracil (PTU, 0.1% w/v) in drinking water to induce serum TSH and 20 mice were without PTU treatment. Equal number of wild-type littermates (TPO-KRAS(WT)) was given the same treatment. The expression of SPRY1, a negative regulator of receptor tyrosine kinase (RTK) signaling, was analyzed in both KRAS(G12D)-and BRAF(V600E)-induced thyroid cancers. Without PTU treatment, only mild thyroid enlargement and hyperplasia were observed in TPO-KRAS(G12D) mice. With PTU treatment, significant thyroid enlargement and hyperplasia occurred in both TPO-KRAS(G12D) and TPO-KRAS(WT) littermates. Thyroids from TPO-KRAS(G12D) mice were six times larger than TPO-KRAS(WT) littermates. Distinct thyroid histology was found between TPO-KRAS(G12D) and TPO-KRAS(WT) mice: thyroid from TPO-KRAS(G12D) mice showed hyperplasia with well-maintained follicular architecture whereas in TPO-KRAS(WT) mice this structure was replaced by papillary hyperplasia. Among 10 TPO-KRAS(G12D) mice monitored for 14 months, two developed follicular thyroid cancer (FTC), one with pulmonary metastasis. Differential SPRY1 expression was demonstrated: increased in FTC and reduced in papillary thyroid cancer (PTC). The increased SPRY1 expression in FTC promoted TSH-RAS signaling through PI3K/AKT pathway whereas downregulation of SPRY1 by BRAF(V600E) in PTC resulted in both MAPK and PI3K/AKT activation. We conclude that chronic TSH stimulation can enhance KRAS(G12D)-mediated oncogenesis, leading to FTC. SPRY1 may function as a molecular switch to control MAPK signaling and its downregulation by BRAF(V600E) favors PTC development.
Collapse
Affiliation(s)
- Minjing Zou
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Essa Y Baitei
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Roua A Al-Rijjal
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Ranjit S Parhar
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Futwan A Al-Mohanna
- Department of Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Shioko Kimura
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Catrin Pritchard
- Department of Biochemistry, University of Leicester, Leicester, UK
| | - Huda BinEssa
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Azizah A Alanazi
- Department of Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Ali S Alzahrani
- Department of Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Mohammed Akhtar
- Department of Pathology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Abdullah M Assiri
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Brian F Meyer
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Yufei Shi
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
44
|
O'Leary AP, Fox JM, Pullar CE. Beta-Adrenoceptor Activation Reduces Both Dermal Microvascular Endothelial Cell Migration via a cAMP-Dependent Mechanism and Wound Angiogenesis. J Cell Physiol 2015; 230:356-65. [PMID: 24986762 PMCID: PMC4263239 DOI: 10.1002/jcp.24716] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 06/26/2014] [Indexed: 12/26/2022]
Abstract
Angiogenesis is an essential process during tissue regeneration; however, the amount of angiogenesis directly correlates with the level of wound scarring. Angiogenesis is lower in scar-free foetal wounds while angiogenesis is raised and abnormal in pathophysiological scarring such as hypertrophic scars and keloids. Delineating the mechanisms that modulate angiogenesis and could reduce scarring would be clinically useful. Beta-adrenoceptors (β-AR) are G protein-coupled receptors (GPCRs) expressed on all skin cell-types. They play a role in wound repair but their specific role in angiogenesis is unknown. In this study, a range of in vitro assays (single cell migration, scratch wound healing, ELISAs for angiogenic growth factors and tubule formation) were performed with human dermal microvascular endothelial cells (HDMEC) to investigate and dissect mechanisms underpinning β-AR-mediated modulation of angiogenesis in chick chorioallantoic membranes (CAM) and murine excisional skin wounds. β-AR activation reduced HDMEC migration via cyclic adenosine monophosphate (cAMP)-dependent and protein kinase A (PKA)-independent mechanisms as demonstrated through use of an EPAC agonist that auto-inhibited the cAMP-mediated β-AR transduced reduction in HDMEC motility; a PKA inhibitor was, conversely, ineffective. ELISA studies demonstrated that β-AR activation reduced pro-angiogenic growth factor secretion from HDMECs (fibroblast growth factor 2) and keratinocytes (vascular endothelial growth factor A) revealing possible β-AR-mediated autocrine and paracrine anti-angiogenic mechanisms. In more complex environments, β-AR activation delayed HDMEC tubule formation and decreased angiogenesis both in the CAM assay and in murine excisional skin wounds in vivo. β-AR activation reduced HDMEC function in vitro and angiogenesis in vivo; therefore, β-AR agonists could be promising anti-angiogenic modulators in skin. J. Cell. Physiol. 230: 356–365, 2015. © 2014 The Authors. Journal of Cellular Physiology Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Andrew P O'Leary
- Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, UK
| | - James M Fox
- Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, UK
| | - Christine E Pullar
- Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, UK
| |
Collapse
|
45
|
Scott J, Harris GJ, Pinder EM, Macfarlane JG, Hellyer TP, Rostron AJ, Conway Morris A, Thickett DR, Perkins GD, McAuley DF, Widdrington JD, Wiscombe S, Baudouin SV, Roy AI, Linnett VC, Wright SE, Ruchaud-Sparagano MH, Simpson AJ. Exchange protein directly activated by cyclic AMP (EPAC) activation reverses neutrophil dysfunction induced by β2-agonists, corticosteroids, and critical illness. J Allergy Clin Immunol 2015; 137:535-44. [PMID: 26388312 DOI: 10.1016/j.jaci.2015.07.036] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 06/20/2015] [Accepted: 07/14/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND Neutrophils play a role in the pathogenesis of asthma, chronic obstructive pulmonary disease, and pulmonary infection. Impaired neutrophil phagocytosis predicts hospital-acquired infection. Despite this, remarkably few neutrophil-specific treatments exist. OBJECTIVES We sought to identify novel pathways for the restoration of effective neutrophil phagocytosis and to activate such pathways effectively in neutrophils from patients with impaired neutrophil phagocytosis. METHODS Blood neutrophils were isolated from healthy volunteers and patients with impaired neutrophil function. In healthy neutrophils phagocytic impairment was induced experimentally by using β2-agonists. Inhibitors and activators of cyclic AMP (cAMP)-dependent pathways were used to assess the influence on neutrophil phagocytosis in vitro. RESULTS β2-Agonists and corticosteroids inhibited neutrophil phagocytosis. Impairment of neutrophil phagocytosis by β2-agonists was associated with significantly reduced RhoA activity. Inhibition of protein kinase A (PKA) restored phagocytosis and RhoA activity, suggesting that cAMP signals through PKA to drive phagocytic impairment. However, cAMP can signal through effectors other than PKA, such as exchange protein directly activated by cyclic AMP (EPAC). An EPAC-activating analog of cAMP (8CPT-2Me-cAMP) reversed neutrophil dysfunction induced by β2-agonists or corticosteroids but did not increase RhoA activity. 8CPT-2Me-cAMP reversed phagocytic impairment induced by Rho kinase inhibition but was ineffective in the presence of Rap-1 GTPase inhibitors. 8CPT-2Me-cAMP restored function to neutrophils from patients with known acquired impairment of neutrophil phagocytosis. CONCLUSIONS EPAC activation consistently reverses clinical and experimental impairment of neutrophil phagocytosis. EPAC signals through Rap-1 and bypasses RhoA. EPAC activation represents a novel potential means by which to reverse impaired neutrophil phagocytosis.
Collapse
Affiliation(s)
- Jonathan Scott
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Graham J Harris
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Emma M Pinder
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - James G Macfarlane
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Thomas P Hellyer
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Anthony J Rostron
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Andrew Conway Morris
- Division of Anaesthesia, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - David R Thickett
- Centre for Translational Inflammation Research, University of Birmingham, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Gavin D Perkins
- Warwick Medical School Clinical Trials Unit and Heart of England Foundation Trust, University of Warwick, Coventry, United Kingdom
| | - Daniel F McAuley
- Centre for Infection and Immunity, Health Sciences Building, Queen's University Belfast, Belfast, United Kingdom; Regional Intensive Care Unit, Royal Victoria Hospital, Belfast, United Kingdom
| | - John D Widdrington
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Sarah Wiscombe
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Simon V Baudouin
- Department of Anaesthetics, Royal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
| | - Alistair I Roy
- Integrated Critical Care Unit, Sunderland Royal Hospital, Sunderland, United Kingdom
| | - Vanessa C Linnett
- Intensive Care Unit, Queen Elizabeth Hospital, Gateshead, United Kingdom
| | - Stephen E Wright
- Intensive Care Unit, Freeman Hospital, Newcastle upon Tyne, United Kingdom
| | | | - A John Simpson
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.
| |
Collapse
|
46
|
Banerjee U, Cheng X. Exchange protein directly activated by cAMP encoded by the mammalian rapgef3 gene: Structure, function and therapeutics. Gene 2015; 570:157-67. [PMID: 26119090 PMCID: PMC4556420 DOI: 10.1016/j.gene.2015.06.063] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Accepted: 06/23/2015] [Indexed: 01/08/2023]
Abstract
Mammalian exchange protein directly activated by cAMP isoform 1 (EPAC1), encoded by the RAPGEF3 gene, is one of the two-membered family of cAMP sensors that mediate the intracellular functions of cAMP by acting as guanine nucleotide exchange factors for the Ras-like Rap small GTPases. Extensive studies have revealed that EPAC1-mediated cAMP signaling is highly coordinated spatiotemporally through the formation of dynamic signalosomes by interacting with a diverse array of cellular partners. Recent functional analyses of genetically engineered mouse models further suggest that EPAC1 functions as an important stress response switch and is involved in pathophysiological conditions of cardiac stresses, chronic pain, cancer and infectious diseases. These findings, coupled with the development of EPAC specific small molecule modulators, validate EPAC1 as a promising target for therapeutic interventions. Human gene RAPGEF3 encodes for EPAC1 protein. Along with PKA, CNG & HCN, EPAC is an important cAMP sensor. Selective modulators of EPAC1 have been developed for use as pharmacological probes. Formation of EPAC1 signalosomes allows spatiotemporal control of cAMP signaling. EPAC1 is implicated in major pathophysiological conditions and is an attractive therapeutic target.
Collapse
Affiliation(s)
- Upasana Banerjee
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Health Science Center, Houston, TX 77030, United States
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Health Science Center, Houston, TX 77030, United States.
| |
Collapse
|
47
|
Parnell E, Smith BO, Yarwood SJ. The cAMP sensors, EPAC1 and EPAC2, display distinct subcellular distributions despite sharing a common nuclear pore localisation signal. Cell Signal 2015; 27:989-96. [PMID: 25683912 PMCID: PMC4372255 DOI: 10.1016/j.cellsig.2015.02.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 02/08/2015] [Indexed: 11/22/2022]
Abstract
We have identified a conserved nuclear pore localisation signal (NPLS; amino acids 764-838 of EPAC1) in the catalytic domains of the cAMP-sensors, EPAC1 and EPAC2A. Consequently, EPAC1 is mainly localised to the nuclear pore complex in HEK293T cells where it becomes activated following stimulation with cAMP. In contrast, structural models indicate that the cAMP-binding domain of EPAC2A (CNBD1) blocks access to the conserved NPLS in EPAC2A, reducing its ability to interact with nuclear binding sites. Consequently, a naturally occurring EPAC2 isoform, EPAC2B, which lacks CNBD1 is enriched in nuclear fractions, similar to EPAC1. Structural differences in EPAC isoforms may therefore determine their intracellular location and their response to elevations in intracellular cAMP.
Collapse
Affiliation(s)
- Euan Parnell
- Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, G12 8QQ, UK
| | - Brian O Smith
- Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, G12 8QQ, UK
| | - Stephen J Yarwood
- Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, G12 8QQ, UK.
| |
Collapse
|
48
|
Faam B, Ghaffari MA, Ghadiri A, Azizi F. Epigenetic modifications in human thyroid cancer. Biomed Rep 2014; 3:3-8. [PMID: 25469237 DOI: 10.3892/br.2014.375] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 09/09/2014] [Indexed: 12/24/2022] Open
Abstract
Thyroid carcinoma is the most common endocrine malignancy of the endocrine organs, and its incidence rate has steadily increased over the last decade. Over 95% of thyroid carcinoma is derived from follicular cells that have a spectrum of differentiation to the most invasive malignancy. The molecular pathogenesis of thyroid cancer remains to be clarified, although activating the RET, RAS and BRAF oncogenes have been well characterized. Increasing evidence from previous studies demonstrates that acquired epigenetic abnormalities participating with genetic alteration results in altered patterns of gene expression/function. Aberrant DNA methylation has been established in the CpG regions and microRNAs (miRNAs) expression profile recognized in cancer development. In the present review, a literature review was performed using MEDLINE and PubMed with the terms 'epigenetic patterns in thyroid cancer [or papillary thyroid carcinoma (PTC), follicular thyroid carcinoma (FTC), medullary thyroid cancer (MTC), anaplastic thyroid cancer (ATC)]', 'DNA methylation in thyroid cancer (or PTC, FTC, MTC, ATC)', 'miRNA expression in thyroid cancer (or PTC, FTC, MTC, ATC)', 'epigenetic patterns in cancer' and the current understanding of epigenetic patterns in thyroid cancer was discussed.
Collapse
Affiliation(s)
- Bita Faam
- Cellular and Molecular Research Center, Jundishapur University of Medical Sciences, Ahvaz, Tehran, Iran
| | - Mohammad Ali Ghaffari
- Cellular and Molecular Research Center, Department of Biochemistry, School of Medicine, Tehran, Iran
| | - Ata Ghadiri
- Cellular and Molecular Research Center, Jundishapur University of Medical Sciences, Ahvaz, Tehran, Iran ; Department of Immunology, Faculty of Medicine, Jundishapur University of Medical Sciences, Ahvaz, Tehran, Iran
| | - Fereidoun Azizi
- Endocrine Research Center, Research Institute for Endocrine Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
49
|
Blackwell KT, Jedrzejewska-Szmek J. Molecular mechanisms underlying neuronal synaptic plasticity: systems biology meets computational neuroscience in the wilds of synaptic plasticity. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2013; 5:717-31. [PMID: 24019266 PMCID: PMC3947422 DOI: 10.1002/wsbm.1240] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 07/25/2013] [Accepted: 07/29/2013] [Indexed: 12/29/2022]
Abstract
Interactions among signaling pathways that are activated by transmembrane receptors produce complex networks and emergent dynamical behaviors that are implicated in synaptic plasticity. Temporal dynamics and spatial aspects are critical determinants of cell responses such as synaptic plasticity, although the mapping between spatiotemporal activity pattern and direction of synaptic plasticity is not completely understood. Computational modeling of neuronal signaling pathways has significantly contributed to understanding signaling pathways underlying synaptic plasticity. Spatial models of signaling pathways in hippocampal neurons have revealed mechanisms underlying the spatial distribution of extracellular signal-related kinase (ERK) activation in hippocampal neurons. Other spatial models have demonstrated that the major role of anchoring proteins in striatal and hippocampal synaptic plasticity is to place molecules near their activators. Simulations of yet other models have revealed that the spatial distribution of synaptic plasticity may differ for potentiation versus depression. In general, the most significant advances have been made by interactive modeling and experiments; thus, an interdisciplinary approach should be applied to investigate critical issues in neuronal signaling pathways. These issues include identifying which transmembrane receptors are key for activating ERK in neurons, and the crucial targets of kinases that produce long-lasting synaptic plasticity. Although the number of computer programs for computationally efficient simulation of large reaction-diffusion networks is increasing, parameter estimation and sensitivity analysis in these spatial models remain more difficult than in single compartment models. Advances in live cell imaging coupled with further software development will continue to accelerate the development of spatial models of synaptic plasticity.
Collapse
Affiliation(s)
- KT Blackwell
- Molecular Neuroscience Department, The Krasnow Institute for Advanced Studies George Mason University, Fairfax, VA 22030-444, USA
| | - J Jedrzejewska-Szmek
- Molecular Neuroscience Department, The Krasnow Institute for Advanced Studies George Mason University, Fairfax, VA 22030-444, USA
| |
Collapse
|
50
|
Takahashi M, Dillon TJ, Liu C, Kariya Y, Wang Z, Stork PJS. Protein kinase A-dependent phosphorylation of Rap1 regulates its membrane localization and cell migration. J Biol Chem 2013; 288:27712-23. [PMID: 23946483 PMCID: PMC3784689 DOI: 10.1074/jbc.m113.466904] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 08/13/2013] [Indexed: 11/06/2022] Open
Abstract
The small G protein Rap1 can mediate "inside-out signaling" by recruiting effectors to the plasma membrane that signal to pathways involved in cell adhesion and cell migration. This action relies on the membrane association of Rap1, which is dictated by post-translational prenylation as well as by a stretch of basic residues within its carboxyl terminus. One feature of this stretch of acidic residues is that it lies adjacent to a functional phosphorylation site for the cAMP-dependent protein kinase PKA. This phosphorylation has two effects on Rap1 action. One, it decreases the level of Rap1 activity as measured by GTP loading and the coupling of Rap1 to RapL, a Rap1 effector that couples Rap1 GTP loading to integrin activation. Two, it destabilizes the membrane localization of Rap1, promoting its translocation into the cytoplasm. These two actions, decreased GTP loading and decreased membrane localization, are related, as the translocation of Rap1-GTP into the cytoplasm is associated with its increased GTP hydrolysis and inactivation. The consequences of this phosphorylation in Rap1-dependent cell adhesion and cell migration were also examined. Active Rap1 mutants that lack this phosphorylation site had a minimal effect on cell adhesion but strongly reduced cell migration, when compared with an active Rap1 mutant that retained the phosphorylation site. This suggests that optimal cell migration is associated with cycles of Rap1 activation, membrane egress, and inactivation, and requires the regulated phosphorylation of Rap1 by PKA.
Collapse
Affiliation(s)
| | | | | | - Yumi Kariya
- From the Vollum Institute, and
- the Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Zhiping Wang
- From the Vollum Institute, and
- Department of Surgery, Oregon Health and Science University, Portland, Oregon 97239 and
| | | |
Collapse
|