1
|
Luo Y, Sun L, Peng Y. The structural basis of the G protein-coupled receptor and ion channel axis. Curr Res Struct Biol 2025; 9:100165. [PMID: 40083915 PMCID: PMC11904507 DOI: 10.1016/j.crstbi.2025.100165] [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: 09/26/2024] [Revised: 01/25/2025] [Accepted: 02/17/2025] [Indexed: 03/16/2025] Open
Abstract
Sensory neurons play an essential role in recognizing and responding to detrimental, irritating, and inflammatory stimuli from our surroundings, such as pain, itch, cough, and neurogenic inflammation. The transduction of these physiological signals is chiefly mediated by G protein-coupled receptors (GPCRs) and ion channels. The binding of ligands to GPCRs triggers a signaling cascade, recruiting G proteins or β-arrestins, which subsequently interact with ion channels (e.g., GIRK and TRP channels). This interaction leads to the sensitization and activation of these channels, initiating the neuron's protective mechanisms. This review delves into the complex interplay between GPCRs and ion channels that underpin these physiological processes, with a particular focus on the role of structural biology in enhancing our comprehension. Through unraveling the intricacies of the GPCR-ion channel axis, we aim to shed light on the sophisticated intermolecular dynamics within these pivotal membrane protein families, ultimately guiding the development of precise therapeutic interventions.
Collapse
Affiliation(s)
- Yulin Luo
- iHuman Institute, ShanghaiTech University, Ren Building, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
- School of Life Science and Technology, ShanghaiTech University, L Building, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
| | - Liping Sun
- iHuman Institute, ShanghaiTech University, Ren Building, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
| | - Yao Peng
- iHuman Institute, ShanghaiTech University, Ren Building, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
| |
Collapse
|
2
|
Xu Z, Hussain A, Jiang H, Zhang H, Raza Y, Mustafa G, Ye J, Yuan B, Yang B, Shi Q. AKAP14 is dispensable for mouse fertility. Cells Dev 2025:204032. [PMID: 40418986 DOI: 10.1016/j.cdev.2025.204032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/21/2025] [Accepted: 05/23/2025] [Indexed: 05/28/2025]
Abstract
A-kinase anchoring proteins (AKAPs) constitute a structurally diverse family of scaffold proteins with significant functional relevance. Various AKAP family members localize to distinct subcellular compartments, such as sperm flagella, via their specific domains. In this study, we investigated the physiological function of AKAP14, a potential functional gene for male fertility due to its high expression in human and mouse testicular tissues. We generated Akap14 knockout mice(Akap14ins/Y) using CRISPR/Cas9 technology to assess the role of AKAP14 in male fertility. Our results demonstrate that Akap14ins/Y mice exhibit normal fertility, with a comparable testes-to-body weight ratio, epididymal sperm count, sperm motility, and sperm morphology to wild-type male mice. Furthermore, examination of meiotic prophase I progression revealed a consistent distribution of each substage in Akap14ins/Y testes compared to wild-type testes, indicating that AKAP14 is dispensable for spermatogenesis in mice. In conclusion, despite its high expression level in the testes, AKAP14 depletion does not impact male fertility in mice, suggesting that further in-depth studies of its role in murine spermatogenesis may be unnecessary and could conserve valuable research resources. IN BRIEF: AKAP14 is highly expressed in the testes, but our study using Akap14 knockout mice reveals that it is dispensable for male fertility and spermatogenesis. Despite its expression in testicular tissues, AKAP14 depletion does not impair sperm function or meiotic progression, suggesting no critical role in murine reproductive health.
Collapse
Affiliation(s)
- Zishuo Xu
- Center for Reproduction and Genetics, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Ansar Hussain
- Center for Reproduction and Genetics, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Hanwei Jiang
- Center for Reproduction and Genetics, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Huan Zhang
- Center for Reproduction and Genetics, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Yousaf Raza
- Center for Reproduction and Genetics, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Ghulam Mustafa
- Center for Reproduction and Genetics, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Jingwei Ye
- Center for Reproduction and Genetics, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Botao Yuan
- Center for Reproduction and Genetics, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Bo Yang
- Center for Reproduction and Genetics, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, University of Science and Technology of China, Hefei, Anhui 230027, China..
| | - Qinghua Shi
- Center for Reproduction and Genetics, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, University of Science and Technology of China, Hefei, Anhui 230027, China..
| |
Collapse
|
3
|
Farhangdoost N, Liao C, Liu Y, Rochefort D, Aboasali F, Pietrantonio A, Alda M, Dion PA, Chaumette B, Khayachi A, Rouleau GA. Transcriptomic and epigenomic consequences of heterozygous loss-of-function mutations in AKAP11, a shared risk gene for bipolar disorder and schizophrenia. Mol Psychiatry 2025:10.1038/s41380-025-03040-x. [PMID: 40316678 DOI: 10.1038/s41380-025-03040-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/16/2025] [Accepted: 04/24/2025] [Indexed: 05/04/2025]
Abstract
The gene A-kinase anchoring protein 11 (AKAP11) recently emerged as a shared risk factor between bipolar disorder and schizophrenia, driven by large-effect loss-of-function (LoF) variants. Recent research has uncovered the neurophysiological characteristics and synapse proteomics profile of Akap11-mutant mouse models. Considering the role of AKAP11 in binding cAMP-dependent protein kinase A (PKA) and mediating phosphorylation of numerous substrates, such as transcription factors and epigenetic regulators, and given that chromatin alterations have been implicated in the brains of patients with bipolar disorder and schizophrenia, it is crucial to uncover the transcriptomic and chromatin dysregulations following the heterozygous knockout of AKAP11, particularly in human neurons. This study uses genome-wide approaches to investigate such aberrations in human induced pluripotent stem cell (iPSC)-derived neurons. We show the impact of heterozygous AKAP11 LoF mutations on the gene expression landscape and profile the DNA methylation and histone acetylation modifications. Altogether we highlight the involvement of aberrant activity of intergenic and intronic enhancers, which are enriched in PBX homeobox 2 (PBX2) and Nuclear Factor-1 (NF1) known binding motifs, respectively, in transcription dysregulations of genes mainly involved in DNA-binding transcription factor activity, actin binding and cytoskeleton regulation, and cytokine receptor binding. We also show significant downregulation of pathways related to ribosome structure and function, a pathway also altered in BD and SCZ post-mortem brain tissues and heterozygous Akap11-KO mice synapse proteomics. A better understanding of the dysregulations resulting from haploinsufficiency in AKAP11 improves our knowledge of the biological roots and pathophysiology of BD and SCZ, paving the way for better therapeutic approaches.
Collapse
Affiliation(s)
- Nargess Farhangdoost
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Montreal Neurological Institute-Hospital, Montreal, QC, Canada
| | - Calwing Liao
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Yumin Liu
- Montreal Neurological Institute-Hospital, Montreal, QC, Canada
- Department of Neurology & Neurosurgery, McGill University, Montreal, QC, Canada
| | - Daniel Rochefort
- Montreal Neurological Institute-Hospital, Montreal, QC, Canada
- Department of Neurology & Neurosurgery, McGill University, Montreal, QC, Canada
| | - Farah Aboasali
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Montreal Neurological Institute-Hospital, Montreal, QC, Canada
| | - Alessia Pietrantonio
- Montreal Neurological Institute-Hospital, Montreal, QC, Canada
- Department of Neurology & Neurosurgery, McGill University, Montreal, QC, Canada
| | - Martin Alda
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Patrick A Dion
- Montreal Neurological Institute-Hospital, Montreal, QC, Canada
- Department of Neurology & Neurosurgery, McGill University, Montreal, QC, Canada
| | - Boris Chaumette
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (INSERM U1266), Institut Pasteur (CNRS UMR3571), GHU Paris Psychiatrie et Neurosciences, Paris, France.
- Department of Psychiatry, McGill University, Montreal, QC, Canada.
| | - Anouar Khayachi
- Montreal Neurological Institute-Hospital, Montreal, QC, Canada.
- Department of Neurology & Neurosurgery, McGill University, Montreal, QC, Canada.
| | - Guy A Rouleau
- Department of Human Genetics, McGill University, Montreal, QC, Canada.
- Montreal Neurological Institute-Hospital, Montreal, QC, Canada.
- Department of Neurology & Neurosurgery, McGill University, Montreal, QC, Canada.
| |
Collapse
|
4
|
Kumar M, Mehan S, Kumar A, Sharma T, Khan Z, Tiwari A, Das Gupta G, Narula AS. Therapeutic efficacy of Genistein in activation of neuronal AC/cAMP/CREB/PKA and mitochondrial ETC-Complex pathways in experimental model of autism: Evidence from CSF, blood plasma and brain analysis. Brain Res 2024; 1846:149251. [PMID: 39384128 DOI: 10.1016/j.brainres.2024.149251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/10/2024] [Accepted: 09/22/2024] [Indexed: 10/11/2024]
Abstract
Autism is a complex neurodevelopmental condition characterized by repetitive behaviors, impaired social communication, and various associated conditions such as depression and anxiety. Its multifactorial etiology includes genetic, environmental, dietary, and gastrointestinal contributions. Pathologically, Autism is linked to mitochondrial dysfunction, oxidative stress, neuroinflammation, and neurotransmitter imbalances involving GABA, glutamate, dopamine, and oxytocin. Propionic acid (PRPA) is a short-chain fatty acid produced by gut bacteria, influencing central nervous system functions. Elevated PRPA levels can exacerbate Autism-related symptoms by disrupting metabolic processes and crossing the blood-brain barrier. Our research investigates the neuroprotective potential of Genistein (GNT), an isoflavone compound with known benefits in neuropsychiatric and neurodegenerative disorders, through modulation of the AC/cAMP/CREB/PKA signaling pathway and mitochondrial ETC complex (I-IV) function. In silico analyses revealed GNT's high affinity for these targets. Subsequent in vitro and in vivo experiments using a PRPA-induced rat model of autism demonstrated that GNT (40 and 80 mg/kg., orally) significantly improves locomotion, neuromuscular coordination, and cognitive functions in PRPA-treated rodents. Behavioral assessments showed reduced immobility in the forced swim test, enhanced Morris water maze performance, and restored regular locomotor activity. On a molecular level, GNT restored levels of key signaling molecules (AC, cAMP, CREB, PKA) and mitochondrial complexes (I-V), disrupted by PRPA exposure. Additionally, GNT reduced neuroinflammation and apoptosis, normalized neurotransmitter levels, and improved the complete blood count profile. Histopathological analyses confirmed that GNT ameliorated PRPA-induced brain injuries, restored normal brain morphology, reduced demyelination, and promoted neurogenesis. The study supports GNT's potential in autism treatment by modulating neural pathways, reducing inflammation, and restoring neurotransmitter balance.
Collapse
Affiliation(s)
- Manjeet Kumar
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India; Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India; Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India.
| | - Aakash Kumar
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India; Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Tarun Sharma
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India; Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India; Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Aarti Tiwari
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India; Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Ghanshyam Das Gupta
- Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India; Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC 27516, USA
| |
Collapse
|
5
|
Lyons AC, Mehta S, Zhang J. Fluorescent biosensors illuminate the spatial regulation of cell signaling across scales. Biochem J 2023; 480:1693-1717. [PMID: 37903110 PMCID: PMC10657186 DOI: 10.1042/bcj20220223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 11/01/2023]
Abstract
As cell signaling research has advanced, it has become clearer that signal transduction has complex spatiotemporal regulation that goes beyond foundational linear transduction models. Several technologies have enabled these discoveries, including fluorescent biosensors designed to report live biochemical signaling events. As genetically encoded and live-cell compatible tools, fluorescent biosensors are well suited to address diverse cell signaling questions across different spatial scales of regulation. In this review, methods of examining spatial signaling regulation and the design of fluorescent biosensors are introduced. Then, recent biosensor developments that illuminate the importance of spatial regulation in cell signaling are highlighted at several scales, including membranes and organelles, molecular assemblies, and cell/tissue heterogeneity. In closing, perspectives on how fluorescent biosensors will continue enhancing cell signaling research are discussed.
Collapse
Affiliation(s)
- Anne C. Lyons
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, U.S.A
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, U.S.A
| | - Sohum Mehta
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, U.S.A
| | - Jin Zhang
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, U.S.A
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, U.S.A
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, U.S.A
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, U.S.A
| |
Collapse
|
6
|
Bai X, Zhang K, Ou C, Nie B, Zhang J, Huang Y, Zhang Y, Huang J, Ouyang H, Cao M, Huang W. Selective activation of AKAP150/TRPV1 in ventrolateral periaqueductal gray GABAergic neurons facilitates conditioned place aversion in male mice. Commun Biol 2023; 6:742. [PMID: 37460788 PMCID: PMC10352381 DOI: 10.1038/s42003-023-05106-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 07/06/2023] [Indexed: 07/20/2023] Open
Abstract
Aversion refers to feelings of strong dislike or avoidance toward particular stimuli or situations. Aversion can be caused by pain stimuli and has a long-term negative impact on physical and mental health. Aversion can also be caused by drug abuse withdrawal, resulting in people with substance use disorder to relapse. However, the mechanisms underlying aversion remain unclear. The ventrolateral periaqueductal gray (vlPAG) is considered to play a key role in aversive behavior. Our study showed that inhibition of vlPAG GABAergic neurons significantly attenuated the conditioned place aversion (CPA) induced by hindpaw pain pinch or naloxone-precipitated morphine withdrawal. However, activating or inhibiting glutamatergic neurons, or activating GABAergic neurons cannot affect or alter CPA response. AKAP150 protein expression and phosphorylated TRPV1 (p-TRPV1) were significantly upregulated in these two CPA models. In AKAP150flox/flox mice and C57/B6J wild-type mice, cell-type-selective inhibition of AKAP150 in GABAergic neurons in the vlPAG attenuated aversion. However, downregulating AKAP150 in glutamatergic neurons did not attenuate aversion. Knockdown of AKAP150 in GABAergic neurons effectively reversed the p-TRPV1 upregulation in these two CPA models utilized in our study. Collectively, inhibition of the AKAP150/p-TRPV1 pathway in GABAergic neurons in the vlPAG may be considered a potential therapeutic target for the CPA response.
Collapse
Affiliation(s)
- Xiaohui Bai
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Anesthesiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation. Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kun Zhang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Chaopeng Ou
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Bilin Nie
- Department of Anesthesiology, Guangdong Women and Children Hospital, Guangzhou, China
| | - Jianxing Zhang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yongtian Huang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yingjun Zhang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jingxiu Huang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Handong Ouyang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| | - Minghui Cao
- Department of Anesthesiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation. Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Wan Huang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| |
Collapse
|
7
|
Long C, Li G, Meng Y, Huang X, Chen J, Liu J. Weighted gene co-expression network analysis identifies the prognosis-related models of left- and right-sided colon cancer. Medicine (Baltimore) 2023; 102:e33390. [PMID: 37144998 PMCID: PMC10158920 DOI: 10.1097/md.0000000000033390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 03/08/2023] [Indexed: 05/06/2023] Open
Abstract
Left-sided colon cancer (LC) and right-sided colon cancer (RC) are 2 essentially different diseases, and the potential mechanisms regulating them remain unidentified. In this study, we applied weighted gene co-expression network analysis (WGCNA) to confirm a yellow module, mainly enriched in metabolism-related signaling pathways related to LC and RC. Based on the RNA-seq data of colon cancer in The Cancer Genome Atlas (TCGA) and GSE41258 dataset with their corresponding clinical information, a training set (TCGA: LC: n = 171; RC: n = 260) and a validation set (GSE41258: LC: n = 94; RC: n = 77) were divided. Least absolute shrinkage and selection operator (LASSO) penalized COX regression analysis identified 20 prognosis-related genes (PRGs) and helped constructed 2 risk (LC-R and RC-R) models in LC and RC, respectively. The model-based risk scores accurately performed in risk stratification for colon cancer patients. The high-risk group of the LC-R model showed associations with ECM-receptor interaction, focal adhesion, and PI3K-AKT signaling pathway. Interestingly, the low-risk group of the LC-R model showed associations with immune-related signaling pathways like antigen processing and presentation. On the other hand, the high-risk group of the RC-R model showed enrichment for cell adhesion molecules and axon guidance signaling pathways. Furthermore, we identified 20 differentially expressed PRGs between LC and RC. Our findings provide new insights into the difference between LC and RC, and uncover the potential biomarkers for the treatment of LC and RC.
Collapse
Affiliation(s)
- Chenyan Long
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, The People’s Republic of China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, Guangxi Zhuang Autonomous Region, The People’s Republic of China
| | - Gang Li
- School of Public Health, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, The People’s Republic of China
| | - Yongsheng Meng
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, The People’s Republic of China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, Guangxi Zhuang Autonomous Region, The People’s Republic of China
| | - Xiaoliang Huang
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, The People’s Republic of China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, Guangxi Zhuang Autonomous Region, The People’s Republic of China
| | - Jianhong Chen
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, The People’s Republic of China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, Guangxi Zhuang Autonomous Region, The People’s Republic of China
| | - Jungang Liu
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, The People’s Republic of China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, Guangxi Zhuang Autonomous Region, The People’s Republic of China
| |
Collapse
|
8
|
Nuanced Interactions between AKAP79 and STIM1 with Orai1 Ca 2+ Channels at Endoplasmic Reticulum-Plasma Membrane Junctions Sustain NFAT Activation. Mol Cell Biol 2022; 42:e0017522. [PMID: 36317924 PMCID: PMC9670898 DOI: 10.1128/mcb.00175-22] [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] [Indexed: 11/06/2022] Open
Abstract
A-kinase anchoring protein 79 (AKAP79) is a human scaffolding protein that organizes Ca2+/calmodulin-dependent protein phosphatase calcineurin, calmodulin, cAMP-dependent protein kinase, protein kinase C, and the transcription factor nuclear factor of activated T cells (NFAT1) into a signalosome at the plasma membrane. Upon Ca2+ store depletion, AKAP79 interacts with the N-terminus of STIM1-gated Orai1 Ca2+ channels, enabling Ca2+ nanodomains to stimulate calcineurin. Calcineurin then dephosphorylates and activates NFAT1, which then translocates to the nucleus. A fundamental question is how signalosomes maintain long-term signaling when key effectors are released and therefore removed beyond the reach of the activating signal. Here, we show that the AKAP79-Orai1 interaction is considerably more transient than that of STIM1-Orai1. Free AKAP79, with calcineurin and NFAT1 in tow, is able to replace rapidly AKAP79 devoid of NFAT1 on Orai1, in the presence of continuous Ca2+ entry. We also show that Ca2+ nanodomains near Orai1 channels activate almost the entire cytosolic pool of NFAT1. Recycling of inactive NFAT1 from the cytoplasm to AKAP79 in the plasma membrane, coupled with the relatively weak interaction between AKAP79 and Orai1, maintain excitation-transcription coupling. By measuring rates for AKAP79-NFAT interaction, we formulate a mathematical model that simulates NFAT dynamics at the plasma membrane.
Collapse
|
9
|
Thomas R, Hernandez A, Benavides DR, Li W, Tan C, Umfress A, Plattner F, Chakraborti A, Pozzo-Miller L, Taylor SS, Bibb JA. Integrated regulation of PKA by fast and slow neurotransmission in the nucleus accumbens controls plasticity and stress responses. J Biol Chem 2022; 298:102245. [PMID: 35835216 PMCID: PMC9386499 DOI: 10.1016/j.jbc.2022.102245] [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: 04/19/2022] [Revised: 06/27/2022] [Accepted: 06/30/2022] [Indexed: 11/30/2022] Open
Abstract
Cortical glutamate and midbrain dopamine neurotransmission converge to mediate striatum-dependent behaviors, while maladaptations in striatal circuitry contribute to mental disorders. However, the crosstalk between glutamate and dopamine signaling has not been entirely elucidated. Here we uncover a molecular mechanism by which glutamatergic and dopaminergic signaling integrate to regulate cAMP-dependent protein kinase (PKA) via phosphorylation of the PKA regulatory subunit, RIIβ. Using a combination of biochemical, pharmacological, neurophysiological, and behavioral approaches, we find that glutamate-dependent reduction in cyclin-dependent kinase 5 (Cdk5)-dependent RIIβ phosphorylation alters the PKA holoenzyme autoinhibitory state to increase PKA signaling in response to dopamine. Furthermore, we show that disruption of RIIβ phosphorylation by Cdk5 enhances cortico-ventral striatal synaptic plasticity. In addition, we demonstrate that acute and chronic stress in rats inversely modulate RIIβ phosphorylation and ventral striatal infusion of a small interfering peptide that selectively targets RIIβ regulation by Cdk5 improves behavioral response to stress. We propose this new signaling mechanism integrating ventral striatal glutamate and dopamine neurotransmission is important to brain function, may contribute to neuropsychiatric conditions, and serves as a possible target for the development of novel therapeutics for stress-related disorders.
Collapse
Affiliation(s)
- Rachel Thomas
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104 USA; Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Adan Hernandez
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla 76230, Santiago de Querétaro, Querétaro, México; Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - David R Benavides
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Wei Li
- Department of Neurobiology, Civitan International Research Center, The University of Alabama Birmingham Medical Center, Birmingham, AL 35233, USA
| | - Chunfeng Tan
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030; Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Alan Umfress
- Department of Surgery, The University of Alabama Birmingham Medical Center, Birmingham, AL 35233, USA
| | - Florian Plattner
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ayanabha Chakraborti
- Department of Surgery, The University of Alabama Birmingham Medical Center, Birmingham, AL 35233, USA
| | - Lucas Pozzo-Miller
- Department of Neurobiology, Civitan International Research Center, The University of Alabama Birmingham Medical Center, Birmingham, AL 35233, USA
| | - Susan S Taylor
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA
| | - James A Bibb
- Department of Neurobiology, Civitan International Research Center, The University of Alabama Birmingham Medical Center, Birmingham, AL 35233, USA; Department of Surgery, The University of Alabama Birmingham Medical Center, Birmingham, AL 35233, USA; Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
10
|
Li H. Physiologic and pathophysiologic roles of AKAP12. Sci Prog 2022; 105:368504221109212. [PMID: 35775596 PMCID: PMC10450473 DOI: 10.1177/00368504221109212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A kinase anchoring protein (AKAP) 12 is a scaffolding protein that improves the specificity and efficiency of spatiotemporal signal through assembling intracellular signal proteins into a specific complex. AKAP12 is a negative mitogenic regulator that plays an important role in controlling cytoskeletal architecture, maintaining endothelial integrity, regulating glial function and forming blood-brain barrier (BBB) and blood retinal barrier (BRB). Moreover, elevated or reduced AKAP12 contributes to a variety of diseases. Complex connections between AKAP12 and various diseases including chronic liver diseases (CLDs), inflammatory diseases and a series of cancers will be tried to delineate in this paper. We first describe the expression, distribution and physiological function of AKAP12. Then we summarize the current knowledge of different connections between AKAP12 expression and various diseases. Some research groups have found paradoxical roles of AKAP12 in different diseases and further confirmation is needed. This paper aims to assess the role of AKAP12 in physiology and diseases to help lay the foundation for the design of small molecules for specific AKAP12 to correct the pathological signal defects.
Collapse
Affiliation(s)
- Hui Li
- Central Laboratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China
| |
Collapse
|
11
|
Qian L, Mehrabi Nasab E, Athari SM, Athari SS. Mitochondria signaling pathways in allergic asthma. J Investig Med 2022; 70:863-882. [PMID: 35168999 PMCID: PMC9016245 DOI: 10.1136/jim-2021-002098] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/05/2021] [Indexed: 12/23/2022]
Abstract
Mitochondria, as the powerhouse organelle of cells, are greatly involved in regulating cell signaling pathways, including those related to the innate and acquired immune systems, cellular differentiation, growth, death, apoptosis, and autophagy as well as hypoxic stress responses in various diseases. Asthma is a chronic complicated airway disease characterized by airway hyperresponsiveness, eosinophilic inflammation, mucus hypersecretion, and remodeling of airway. The asthma mortality and morbidity rates have increased worldwide, so understanding the molecular mechanisms underlying asthma progression is necessary for new anti-asthma drug development. The lung is an oxygen-rich organ, and mitochondria, by sensing and processing O2, contribute to the generation of ROS and activation of pro-inflammatory signaling pathways. Asthma pathophysiology has been tightly associated with mitochondrial dysfunction leading to reduced ATP synthase activity, increased oxidative stress, apoptosis induction, and abnormal calcium homeostasis. Defects of the mitochondrial play an essential role in the pro-remodeling mechanisms of lung fibrosis and airway cells' apoptosis. Identification of mitochondrial therapeutic targets can help repair mitochondrial biogenesis and dysfunction and reverse related pathological changes and lung structural remodeling in asthma. Therefore, we here overviewed the relationship between mitochondrial signaling pathways and asthma pathogenic mechanisms.
Collapse
Affiliation(s)
- Ling Qian
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Shanghai, China
| | - Entezar Mehrabi Nasab
- Department of Cardiology, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran (the Islamic Republic of)
| | | | - Seyyed Shamsadin Athari
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran (the Islamic Republic of)
| |
Collapse
|
12
|
Melick CH, Lama-Sherpa TD, Curukovic A, Jewell JL. G-Protein Coupled Receptor Signaling and Mammalian Target of Rapamycin Complex 1 Regulation. Mol Pharmacol 2022; 101:181-190. [PMID: 34965982 PMCID: PMC9092479 DOI: 10.1124/molpharm.121.000302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 11/29/2021] [Indexed: 11/30/2022] Open
Abstract
The mammalian target of rapamycin (mTOR) senses upstream stimuli to regulate numerous cellular functions such as metabolism, growth, and autophagy. Increased activation of mTOR complex 1 (mTORC1) is typically observed in human disease and continues to be an important therapeutic target. Understanding the upstream regulators of mTORC1 will provide a crucial link in targeting hyperactivated mTORC1 in human disease. In this mini-review, we will discuss the regulation of mTORC1 by upstream stimuli, with a specific focus on G-protein coupled receptor signaling to mTORC1. SIGNIFICANCE STATEMENT: mTORC1 is a master regulator of many cellular processes and is often hyperactivated in human disease. Therefore, understanding the molecular underpinnings of G-protein coupled receptor signaling to mTORC1 will undoubtedly be beneficial for human disease.
Collapse
Affiliation(s)
- Chase H Melick
- Department of Molecular Biology, Harold C. Simmons Comprehensive Cancer, and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Tshering D Lama-Sherpa
- Department of Molecular Biology, Harold C. Simmons Comprehensive Cancer, and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Adna Curukovic
- Department of Molecular Biology, Harold C. Simmons Comprehensive Cancer, and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jenna L Jewell
- Department of Molecular Biology, Harold C. Simmons Comprehensive Cancer, and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
13
|
Chinn AM, Salmerón C, Lee J, Sriram K, Raz E, Insel PA. PDE4B Is a Homeostatic Regulator of Cyclic AMP in Dendritic Cells. Front Pharmacol 2022; 13:833832. [PMID: 35387344 PMCID: PMC8977838 DOI: 10.3389/fphar.2022.833832] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 02/01/2022] [Indexed: 11/13/2022] Open
Abstract
Chronic decreases in the second messenger cyclic AMP (cAMP) occur in numerous settings, but how cells compensate for such decreases is unknown. We have used a unique system-murine dendritic cells (DCs) with a DC-selective depletion of the heterotrimeric GTP binding protein Gαs-to address this issue. These mice spontaneously develop Th2-allergic asthma and their DCs have persistently lower cAMP levels. We found that phosphodiesterase 4B (PDE4B) is the primary phosphodiesterase expressed in DCs and that its expression is preferentially decreased in Gαs-depleted DCs. PDE4B expression is dynamic, falling and rising in a protein kinase A-dependent manner with decreased and increased cAMP concentrations, respectively. Treatment of DCs that drive enhanced Th2 immunity with a PDE4B inhibitor ameliorated DC-induced helper T cell response. We conclude that PDE4B is a homeostatic regulator of cellular cAMP concentrations in DCs and may be a target for treating Th2-allergic asthma and other settings with low cellular cAMP concentrations.
Collapse
Affiliation(s)
- Amy M. Chinn
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, United States
| | - Cristina Salmerón
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, United States
| | - Jihyung Lee
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Krishna Sriram
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, United States
| | - Eyal Raz
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Paul A. Insel
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, United States
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
14
|
LaCroix R, Lin B, Kang TY, Levchenko A. Complex effects of kinase localization revealed by compartment-specific regulation of protein kinase A activity. eLife 2022; 11:e66869. [PMID: 35199643 PMCID: PMC8871369 DOI: 10.7554/elife.66869] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 02/10/2022] [Indexed: 12/12/2022] Open
Abstract
Kinase activity in signaling networks frequently depends on regulatory subunits that can both inhibit activity by interacting with the catalytic subunits and target the kinase to distinct molecular partners and subcellular compartments. Here, using a new synthetic molecular interaction system, we show that translocation of a regulatory subunit of the protein kinase A (PKA-R) to the plasma membrane has a paradoxical effect on the membrane kinase activity. It can both enhance it at lower translocation levels, even in the absence of signaling inputs, and inhibit it at higher translocation levels, suggesting its role as a linker that can both couple and decouple signaling processes in a concentration-dependent manner. We further demonstrate that superposition of gradients of PKA-R abundance across single cells can control the directionality of cell migration, reversing it at high enough input levels. Thus, complex in vivo patterns of PKA-R localization can drive complex phenotypes, including cell migration.
Collapse
Affiliation(s)
- Rebecca LaCroix
- Department of Biomedical Engineering, Yale UniversityNew HavenUnited States
- Yale Systems Biology Institute, Yale UniversityWest HavenUnited States
| | - Benjamin Lin
- Department of Biomedical Engineering, Yale UniversityNew HavenUnited States
- Yale Systems Biology Institute, Yale UniversityWest HavenUnited States
- Department of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone HealthNew YorkUnited States
| | - Tae-Yun Kang
- Department of Biomedical Engineering, Yale UniversityNew HavenUnited States
- Yale Systems Biology Institute, Yale UniversityWest HavenUnited States
| | - Andre Levchenko
- Department of Biomedical Engineering, Yale UniversityNew HavenUnited States
- Yale Systems Biology Institute, Yale UniversityWest HavenUnited States
| |
Collapse
|
15
|
Inferiority complex: why do sensory ion channels multimerize? Biochem Soc Trans 2022; 50:213-222. [PMID: 35166323 PMCID: PMC9022975 DOI: 10.1042/bst20211002] [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: 11/29/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 11/17/2022]
Abstract
Peripheral somatosensory nerves are equipped with versatile molecular sensors which respond to acute changes in the physical environment. Most of these sensors are ion channels that, when activated, depolarize the sensory nerve terminal causing it to generate action potentials, which is the first step in generation of most somatic sensations, including pain. The activation and inactivation of sensory ion channels is tightly regulated and modulated by a variety of mechanisms. Amongst such mechanisms is the regulation of sensory ion channel activity via direct molecular interactions with other proteins in multi-protein complexes at the plasma membrane of sensory nerve terminals. In this brief review, we will consider several examples of such complexes formed around a prototypic sensory receptor, transient receptor potential vanilloid type 1 (TRPV1). We will also discuss some inherent conceptual difficulties arising from the multitude of reported complexes.
Collapse
|
16
|
Peng GE, von Zastrow M. A Live-Cell Imaging Assay for Nuclear Entry of cAMP-Dependent Protein Kinase Catalytic Subunits Stimulated by Endogenous GPCR Activation. Methods Mol Biol 2022; 2483:339-349. [PMID: 35286686 DOI: 10.1007/978-1-0716-2245-2_21] [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: 06/14/2023]
Abstract
Nuclear entry of cAMP-dependent protein kinase catalytic subunits is typically inferred from changes in net protein amount or kinase activity in the nucleus. Previous methods to directly assess nuclear entry require kinase subunit overexpression and/or supraphysiological cAMP elevation. We describe a method to detect nuclear entry of catalytic subunits expressed at an endogenous level in living cells, stimulated by cAMP in a physiological range, and in real time.
Collapse
Affiliation(s)
- Grace E Peng
- Program in Cell Biology, University of California, San Francisco, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Mark von Zastrow
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA.
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA.
- Quantitative Biology Institute, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
17
|
Walker-Gray R, Pallien T, Miller DC, Oder A, Neuenschwander M, von Kries JP, Diecke S, Klussmann E. Disruptors of AKAP-Dependent Protein-Protein Interactions. Methods Mol Biol 2022; 2483:117-139. [PMID: 35286673 DOI: 10.1007/978-1-0716-2245-2_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
A-kinase anchoring proteins (AKAPs) are a family of multivalent scaffolding proteins. They engage in direct protein-protein interactions with protein kinases, kinase substrates and further signaling molecules. Each AKAP interacts with a specific set of protein interaction partners and such sets can vary between different cellular compartments and cells. Thus, AKAPs can coordinate signal transduction processes spatially and temporally in defined cellular environments. AKAP-dependent protein-protein interactions are involved in a plethora of physiological processes, including processes in the cardiovascular, nervous, and immune system. Dysregulation of AKAPs and their interactions is associated with or causes widespread diseases, for example, cardiac diseases such as heart failure. However, there are profound shortcomings in understanding functions of specific AKAP-dependent protein-protein interactions. In part, this is due to the lack of agents for specifically targeting defined protein-protein interactions. Peptidic and non-peptidic inhibitors are invaluable molecular tools for elucidating the functions of AKAP-dependent protein-protein interactions. In addition, such interaction disruptors may pave the way to new concepts for the treatment of diseases where AKAP-dependent protein-protein interactions constitute potential drug targets.Here we describe screening approaches for the identification of small molecule disruptors of AKAP-dependent protein-protein interactions. Examples include interactions of AKAP18 and protein kinase A (PKA) and of AKAP-Lbc and RhoA. We discuss a homogenous time-resolved fluorescence (HTRF) and an AlphaScreen® assay for small molecule library screening and human induced pluripotent stem cell-derived cardiac myocytes (hiPSC-CMs) as a cell system for the characterization of identified hits.
Collapse
Affiliation(s)
- Ryan Walker-Gray
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Tamara Pallien
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Duncan C Miller
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Andreas Oder
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | | | | | - Sebastian Diecke
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Enno Klussmann
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany.
| |
Collapse
|
18
|
Belleville-Rolland T, Leuci A, Mansour A, Decouture B, Martin F, Poirault-Chassac S, Rouaud M, Guerineau H, Dizier B, Pidard D, Gaussem P, Bachelot-Loza C. Role of Membrane Lipid Rafts in MRP4 (ABCC4) Dependent Regulation of the cAMP Pathway in Blood Platelets. Thromb Haemost 2021; 121:1628-1636. [PMID: 33851387 DOI: 10.1055/a-1481-2663] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND Platelet cytosolic cyclic adenosine monophosphate (cAMP) levels are balanced by synthesis, degradation, and efflux. Efflux can occur via multidrug resistant protein-4 (MRP4; ABCC4) present on dense granule and/or plasma membranes. As lipid rafts have been shown to interfere on cAMP homeostasis, we evaluated the relationships between the distribution and activity of MRP4 in lipid rafts and cAMP efflux. METHODS Platelet activation and cAMP homeostasis were analyzed in human and wild-type or MRP4-deleted mouse platelets in the presence of methyl-β-cyclodextrin (MßCD) to disrupt lipid rafts, and of activators of the cAMP signalling pathways. Human platelet MRP4 and effector proteins of the cAMP pathway were analyzed by immunoblots in lipid rafts isolated by differential centrifugation. RESULTS MßCD dose dependently inhibited human and mouse platelet aggregation without affecting per se cAMP levels. An additive inhibitory effect existed between the adenylate cyclase (AC) activator forskolin and MßCD that was accompanied by an overincrease of cAMP, and which was significantly enhanced upon MRP4 deletion. Finally, an efflux of cAMP out of resting platelets incubated with prostaglandin E1 (PGE1) was observed that was partly dependent on MRP4. Lipid rafts contained a small fraction (≈15%) of MRP4 and most of the inhibitory G-protein Gi, whereas Gs protein, AC3, and phosphodiesterases PDE2 and PDE3A were all present as only trace amounts. CONCLUSION Our results are in favour of part of MRP4 present at the platelet surface, including in lipid rafts. Lipid raft integrity is necessary for cAMP signalling regulation, although MRP4 and most players of cAMP homeostasis are essentially located outside rafts.
Collapse
Affiliation(s)
- Tiphaine Belleville-Rolland
- Service d'hématologie biologique, AH-HP, Hopital Européen Georges Pompidou, Paris, France
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | - Alexandre Leuci
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | - Alexandre Mansour
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | - Benoit Decouture
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | - Fanny Martin
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | | | - Margot Rouaud
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | - Hippolyte Guerineau
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | - Blandine Dizier
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | - Dominique Pidard
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | - Pascale Gaussem
- Service d'hématologie biologique, AH-HP, Hopital Européen Georges Pompidou, Paris, France
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | | |
Collapse
|
19
|
Martin TJ, Sims NA, Seeman E. Physiological and Pharmacological Roles of PTH and PTHrP in Bone Using Their Shared Receptor, PTH1R. Endocr Rev 2021; 42:383-406. [PMID: 33564837 DOI: 10.1210/endrev/bnab005] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Indexed: 12/13/2022]
Abstract
Parathyroid hormone (PTH) and the paracrine factor, PTH-related protein (PTHrP), have preserved in evolution sufficient identities in their amino-terminal domains to share equivalent actions upon a common G protein-coupled receptor, PTH1R, that predominantly uses the cyclic adenosine monophosphate-protein kinase A signaling pathway. Such a relationship between a hormone and local factor poses questions about how their common receptor mediates pharmacological and physiological actions of the two. Mouse genetic studies show that PTHrP is essential for endochondral bone lengthening in the fetus and is essential for bone remodeling. In contrast, the main postnatal function of PTH is hormonal control of calcium homeostasis, with no evidence that PTHrP contributes. Pharmacologically, amino-terminal PTH and PTHrP peptides (teriparatide and abaloparatide) promote bone formation when administered by intermittent (daily) injection. This anabolic effect is remodeling-based with a lesser contribution from modeling. The apparent lesser potency of PTHrP than PTH peptides as skeletal anabolic agents could be explained by lesser bioavailability to PTH1R. By contrast, prolongation of PTH1R stimulation by excessive dosing or infusion, converts the response to a predominantly resorptive one by stimulating osteoclast formation. Physiologically, locally generated PTHrP is better equipped than the circulating hormone to regulate bone remodeling, which occurs asynchronously at widely distributed sites throughout the skeleton where it is needed to replace old or damaged bone. While it remains possible that PTH, circulating within a narrow concentration range, could contribute in some way to remodeling and modeling, its main physiological role is in regulating calcium homeostasis.
Collapse
Affiliation(s)
- T John Martin
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia.,The University of Melbourne, Department of Medicine at St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Natalie A Sims
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia.,The University of Melbourne, Department of Medicine at St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Ego Seeman
- The University of Melbourne, Department of Medicine at Austin Health, Heidelberg, Victoria, Australia
| |
Collapse
|
20
|
Angelis I, Moussis V, Tsoukatos DC, Tsikaris V. Multidrug Resistance Protein 4 (MRP4/ABCC4): A Suspected Efflux Transporter for Human's Platelet Activation. Protein Pept Lett 2021; 28:983-995. [PMID: 33964863 DOI: 10.2174/0929866528666210505120659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/10/2021] [Accepted: 03/16/2021] [Indexed: 11/22/2022]
Abstract
The main role of platelets is to contribute to hemostasis. However, under pathophysiological conditions, platelet activation may lead to thrombotic events of cardiovascular diseases. Thus, anti-thrombotic treatment is important in patients with cardiovascular disease. This review focuses on a platelet receptor, a transmembrane protein, the Multidrug Resistance Protein 4, MRP4, which contributes to platelet activation by extruding endogenous molecules responsible for their activation and accumulation. The regulation of the intracellular concentration levels of these molecules by MRP4 turned to make the protein suspicious and, at the same time, an interesting regulatory factor of normal platelet function. Especially, the possible role of MRP4 in the excretion of xenobiotic and antiplatelet drugs such as aspirin is discussed, thus imparting platelet aspirin tolerance and correlating the protein with the ineffectiveness of aspirin antiplatelet therapy. Based on the above, this review finally underlines that the development of a highly selective and targeted strategy for platelet MRP4 inhibition will also lead to inhibition of platelet activation and accumulation.
Collapse
Affiliation(s)
- Ioannis Angelis
- Department of Chemistry, Organic Chemistry & Biochemistry, University of Ioannina, Ioannina. Greece
| | - Vassilios Moussis
- Department of Chemistry, Organic Chemistry & Biochemistry, University of Ioannina, Ioannina. Greece
| | - Demokritos C Tsoukatos
- Department of Chemistry, Organic Chemistry & Biochemistry, University of Ioannina, Ioannina. Greece
| | - Vassilios Tsikaris
- Department of Chemistry, Organic Chemistry & Biochemistry, University of Ioannina, Ioannina. Greece
| |
Collapse
|
21
|
Beavo JA, Golkowski M, Shimizu-Albergine M, Beltejar MC, Bornfeldt KE, Ong SE. Phosphoproteomic Analysis as an Approach for Understanding Molecular Mechanisms of cAMP-Dependent Actions. Mol Pharmacol 2021; 99:342-357. [PMID: 33574048 PMCID: PMC8058506 DOI: 10.1124/molpharm.120.000197] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/23/2020] [Indexed: 12/26/2022] Open
Abstract
In recent years, highly sensitive mass spectrometry-based phosphoproteomic analysis is beginning to be applied to identification of protein kinase substrates altered downstream of increased cAMP. Such studies identify a very large number of phosphorylation sites regulated in response to increased cAMP. Therefore, we now are tasked with the challenge of determining how many of these altered phosphorylation sites are relevant to regulation of function in the cell. This minireview describes the use of phosphoproteomic analysis to monitor the effects of cyclic nucleotide phosphodiesterase (PDE) inhibitors on cAMP-dependent phosphorylation events. More specifically, it describes two examples of this approach carried out in the authors' laboratories using the selective PDE inhibitor approach. After a short discussion of several likely conclusions suggested by these analyses of cAMP function in steroid hormone-producing cells and also in T-cells, it expands into a discussion about some newer and more speculative interpretations of the data. These include the idea that multiple phosphorylation sites and not a single rate-limiting step likely regulate these and, by analogy, many other cAMP-dependent pathways. In addition, the idea that meaningful regulation requires a high stoichiometry of phosphorylation to be important is discussed and suggested to be untrue in many instances. These new interpretations have important implications for drug design, especially for targeting pathway agonists. SIGNIFICANCE STATEMENT: Phosphoproteomic analyses identify thousands of altered phosphorylation sites upon drug treatment, providing many possible regulatory targets but also highlighting questions about which phosphosites are functionally important. These data imply that multistep processes are regulated by phosphorylation at not one but rather many sites. Most previous studies assumed a single step or very few rate-limiting steps were changed by phosphorylation. This concept should be changed. Previous interpretations also assumed substoichiometric phosphorylation was not of regulatory importance. This assumption also should be changed.
Collapse
Affiliation(s)
- Joseph A Beavo
- Departments of Pharmacology and Medicine (J.A.B., M.G., M.S.-A., M.-C.B., S.-E.O.), and Division of Metabolism, Endocrinology and Nutrition (K.E.B.), University of Washington, Seattle, Washington
| | - Martin Golkowski
- Departments of Pharmacology and Medicine (J.A.B., M.G., M.S.-A., M.-C.B., S.-E.O.), and Division of Metabolism, Endocrinology and Nutrition (K.E.B.), University of Washington, Seattle, Washington
| | - Masami Shimizu-Albergine
- Departments of Pharmacology and Medicine (J.A.B., M.G., M.S.-A., M.-C.B., S.-E.O.), and Division of Metabolism, Endocrinology and Nutrition (K.E.B.), University of Washington, Seattle, Washington
| | - Michael-Claude Beltejar
- Departments of Pharmacology and Medicine (J.A.B., M.G., M.S.-A., M.-C.B., S.-E.O.), and Division of Metabolism, Endocrinology and Nutrition (K.E.B.), University of Washington, Seattle, Washington
| | - Karin E Bornfeldt
- Departments of Pharmacology and Medicine (J.A.B., M.G., M.S.-A., M.-C.B., S.-E.O.), and Division of Metabolism, Endocrinology and Nutrition (K.E.B.), University of Washington, Seattle, Washington
| | - Shao-En Ong
- Departments of Pharmacology and Medicine (J.A.B., M.G., M.S.-A., M.-C.B., S.-E.O.), and Division of Metabolism, Endocrinology and Nutrition (K.E.B.), University of Washington, Seattle, Washington
| |
Collapse
|
22
|
Colombe AS, Pidoux G. Cardiac cAMP-PKA Signaling Compartmentalization in Myocardial Infarction. Cells 2021; 10:cells10040922. [PMID: 33923648 PMCID: PMC8073060 DOI: 10.3390/cells10040922] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/02/2021] [Accepted: 04/13/2021] [Indexed: 02/07/2023] Open
Abstract
Under physiological conditions, cAMP signaling plays a key role in the regulation of cardiac function. Activation of this intracellular signaling pathway mirrors cardiomyocyte adaptation to various extracellular stimuli. Extracellular ligand binding to seven-transmembrane receptors (also known as GPCRs) with G proteins and adenylyl cyclases (ACs) modulate the intracellular cAMP content. Subsequently, this second messenger triggers activation of specific intracellular downstream effectors that ensure a proper cellular response. Therefore, it is essential for the cell to keep the cAMP signaling highly regulated in space and time. The temporal regulation depends on the activity of ACs and phosphodiesterases. By scaffolding key components of the cAMP signaling machinery, A-kinase anchoring proteins (AKAPs) coordinate both the spatial and temporal regulation. Myocardial infarction is one of the major causes of death in industrialized countries and is characterized by a prolonged cardiac ischemia. This leads to irreversible cardiomyocyte death and impairs cardiac function. Regardless of its causes, a chronic activation of cardiac cAMP signaling is established to compensate this loss. While this adaptation is primarily beneficial for contractile function, it turns out, in the long run, to be deleterious. This review compiles current knowledge about cardiac cAMP compartmentalization under physiological conditions and post-myocardial infarction when it appears to be profoundly impaired.
Collapse
|
23
|
Manchanda Y, Bitsi S, Kang Y, Jones B, Tomas A. Spatiotemporal control of GLP-1 receptor activity. ACTA ACUST UNITED AC 2021. [DOI: 10.1016/j.coemr.2020.07.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
24
|
Bucko PJ, Garcia I, Manocha R, Bhat A, Wordeman L, Scott JD. Gravin-associated kinase signaling networks coordinate γ-tubulin organization at mitotic spindle poles. J Biol Chem 2020; 295:13784-13797. [PMID: 32732289 PMCID: PMC7535905 DOI: 10.1074/jbc.ra120.014791] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/21/2020] [Indexed: 01/16/2023] Open
Abstract
Mitogenic signals that regulate cell division often proceed through multienzyme assemblies within defined intracellular compartments. The anchoring protein Gravin restricts the action of mitotic kinases and cell-cycle effectors to defined mitotic structures. In this report we discover that genetic deletion of Gravin disrupts proper accumulation and asymmetric distribution of γ-tubulin during mitosis. We utilize a new precision pharmacology tool, Local Kinase Inhibition, to inhibit the Gravin binding partner polo-like kinase 1 at spindle poles. Using a combination of gene-editing approaches, quantitative imaging, and biochemical assays, we provide evidence that disruption of local polo-like kinase 1 signaling underlies the γ-tubulin distribution defects observed with Gravin loss. Our study uncovers a new role for Gravin in coordinating γ-tubulin recruitment during mitosis and illuminates the mechanism by which signaling enzymes regulate this process at a distinct subcellular location.
Collapse
Affiliation(s)
- Paula J Bucko
- Department of Pharmacology, University of Washington, Seattle, Washington, USA
| | - Irvin Garcia
- Department of Pharmacology, University of Washington, Seattle, Washington, USA
| | - Ridhima Manocha
- Department of Pharmacology, University of Washington, Seattle, Washington, USA
| | - Akansha Bhat
- Department of Pharmacology, University of Washington, Seattle, Washington, USA
| | - Linda Wordeman
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington, USA
| | - John D Scott
- Department of Pharmacology, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
25
|
Melick CH, Jewell JL. Regulation of mTORC1 by Upstream Stimuli. Genes (Basel) 2020; 11:genes11090989. [PMID: 32854217 PMCID: PMC7565831 DOI: 10.3390/genes11090989] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/22/2020] [Accepted: 08/23/2020] [Indexed: 01/08/2023] Open
Abstract
The mammalian target of rapamycin (mTOR) is an evolutionary conserved Ser/Thr protein kinase that senses multiple upstream stimuli to control cell growth, metabolism, and autophagy. mTOR is the catalytic subunit of mTOR complex 1 (mTORC1). A significant amount of research has uncovered the signaling pathways regulated by mTORC1, and the involvement of these signaling cascades in human diseases like cancer, diabetes, and ageing. Here, we review advances in mTORC1 regulation by upstream stimuli. We specifically focus on how growth factors, amino acids, G-protein coupled receptors (GPCRs), phosphorylation, and small GTPases regulate mTORC1 activity and signaling.
Collapse
Affiliation(s)
- Chase H. Melick
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jenna L. Jewell
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Correspondence:
| |
Collapse
|
26
|
Pyfrom SC, Quinn CC, Dorando HK, Luo H, Payton JE. BCALM (AC099524.1) Is a Human B Lymphocyte-Specific Long Noncoding RNA That Modulates B Cell Receptor-Mediated Calcium Signaling. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 205:595-607. [PMID: 32571842 PMCID: PMC7372127 DOI: 10.4049/jimmunol.2000088] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 05/27/2020] [Indexed: 12/12/2022]
Abstract
Of the thousands of long noncoding RNAs (lncRNA) identified in lymphocytes, very few have defined functions. In this study, we report the discovery and functional elucidation of a human B cell-specific lncRNA with high levels of expression in three types of B cell cancer and normal B cells. The AC099524.1 gene is upstream of the gene encoding the B cell-specific phospholipase C γ 2 (PLCG2), a B cell-specific enzyme that stimulates intracellular Ca2+ signaling in response to BCR activation. AC099524.1 (B cell-associated lncRNA modulator of BCR-mediated Ca+ signaling [BCALM]) transcripts are localized in the cytoplasm and, as expected, CRISPR/Cas9 knockout of AC099524.1 did not affect PLCG2 mRNA or protein expression. lncRNA interactome, RNA immunoprecipitation, and coimmunoprecipitation studies identified BCALM-interacting proteins in B cells, including phospholipase D 1 (PLD1), and kinase adaptor proteins AKAP9 (AKAP450) and AKAP13 (AKAP-Lbc). These two AKAP proteins form signaling complexes containing protein kinases A and C, which phosphorylate and activate PLD1 to produce phosphatidic acid (PA). BCR stimulation of BCALM-deficient B cells resulted in decreased PLD1 phosphorylation and increased intracellular Ca+ flux relative to wild-type cells. These results suggest that BCALM promotes negative feedback that downmodulates BCR-mediated Ca+ signaling by promoting phosphorylation of PLD1 by AKAP-associated kinases, enhancing production of PA. PA activates SHP-1, which negatively regulates BCR signaling. We propose the name BCALM for B-Cell Associated LncRNA Modulator of BCR-mediated Ca+ signaling. Our findings suggest a new, to our knowledge, paradigm for lncRNA-mediated modulation of lymphocyte activation and signaling, with implications for B cell immune response and BCR-dependent cancers.
Collapse
Affiliation(s)
- Sarah C Pyfrom
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Chaz C Quinn
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Hannah K Dorando
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Hong Luo
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Jacqueline E Payton
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
27
|
Bucko PJ, Scott JD. Drugs That Regulate Local Cell Signaling: AKAP Targeting as a Therapeutic Option. Annu Rev Pharmacol Toxicol 2020; 61:361-379. [PMID: 32628872 DOI: 10.1146/annurev-pharmtox-022420-112134] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cells respond to environmental cues by mobilizing signal transduction cascades that engage protein kinases and phosphoprotein phosphatases. Correct organization of these enzymes in space and time enables the efficient and precise transmission of chemical signals. The cyclic AMP-dependent protein kinase A is compartmentalized through its association with A-kinase anchoring proteins (AKAPs). AKAPs are a family of multivalent scaffolds that constrain signaling enzymes and effectors at subcellular locations to drive essential physiological events. More recently, it has been recognized that defective signaling in certain endocrine disorders and cancers proceeds through pathological AKAP complexes. Consequently, pharmacologically targeting these macromolecular complexes unlocks new therapeutic opportunities for a growing number of clinical indications. This review highlights recent findings on AKAP signaling in disease, particularly in certain cancers, and offers an overview of peptides and small molecules that locally regulate AKAP-binding partners.
Collapse
Affiliation(s)
- Paula J Bucko
- Department of Pharmacology, University of Washington, Seattle, Washington 98195, USA; ,
| | - John D Scott
- Department of Pharmacology, University of Washington, Seattle, Washington 98195, USA; ,
| |
Collapse
|
28
|
Kelly MP, Heckman PRA, Havekes R. Genetic manipulation of cyclic nucleotide signaling during hippocampal neuroplasticity and memory formation. Prog Neurobiol 2020; 190:101799. [PMID: 32360536 DOI: 10.1016/j.pneurobio.2020.101799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/14/2020] [Accepted: 03/26/2020] [Indexed: 12/12/2022]
Abstract
Decades of research have underscored the importance of cyclic nucleotide signaling in memory formation and synaptic plasticity. In recent years, several new genetic techniques have expanded the neuroscience toolbox, allowing researchers to measure and modulate cyclic nucleotide gradients with high spatiotemporal resolution. Here, we will provide an overview of studies using genetic approaches to interrogate the role cyclic nucleotide signaling plays in hippocampus-dependent memory processes and synaptic plasticity. Particular attention is given to genetic techniques that measure real-time changes in cyclic nucleotide levels as well as newly-developed genetic strategies to transiently manipulate cyclic nucleotide signaling in a subcellular compartment-specific manner with high temporal resolution.
Collapse
Affiliation(s)
- Michy P Kelly
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, 6439 Garners Ferry Rd, VA Bldg1, 3(rd) Fl, D-12, Columbia, 29209, SC, USA.
| | - Pim R A Heckman
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands.
| | - Robbert Havekes
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands.
| |
Collapse
|
29
|
Sussman CR, Wang X, Chebib FT, Torres VE. Modulation of polycystic kidney disease by G-protein coupled receptors and cyclic AMP signaling. Cell Signal 2020; 72:109649. [PMID: 32335259 DOI: 10.1016/j.cellsig.2020.109649] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/16/2020] [Accepted: 04/17/2020] [Indexed: 12/11/2022]
Abstract
Autosomal Dominant Polycystic Kidney Disease (ADPKD) is a systemic disorder associated with polycystic liver disease (PLD) and other extrarenal manifestations, the most common monogenic cause of end-stage kidney disease, and a major burden for public health. Many studies have shown that alterations in G-protein and cAMP signaling play a central role in its pathogenesis. As for many other diseases (35% of all approved drugs target G-protein coupled receptors (GPCRs) or proteins functioning upstream or downstream from GPCRs), treatments targeting GPCR have shown effectiveness in slowing the rate of progression of ADPKD. Tolvaptan, a vasopressin V2 receptor antagonist is the first drug approved by regulatory agencies to treat rapidly progressive ADPKD. Long-acting somatostatin analogs have also been effective in slowing the rates of growth of polycystic kidneys and liver. Although no treatment has so far been able to prevent the development or stop the progression of the disease, these encouraging advances point to G-protein and cAMP signaling as a promising avenue of investigation that may lead to more effective and safe treatments. This will require a better understanding of the relevant GPCRs, G-proteins, cAMP effectors, and of the enzymes and A-kinase anchoring proteins controlling the compartmentalization of cAMP signaling. The purpose of this review is to provide an overview of general GPCR signaling; the function of polycystin-1 (PC1) as a putative atypical adhesion GPCR (aGPCR); the roles of PC1, polycystin-2 (PC2) and the PC1-PC2 complex in the regulation of calcium and cAMP signaling; the cross-talk of calcium and cAMP signaling in PKD; and GPCRs, adenylyl cyclases, cyclic nucleotide phosphodiesterases, and protein kinase A as therapeutic targets in ADPKD.
Collapse
Affiliation(s)
- Caroline R Sussman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States of America
| | - Xiaofang Wang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States of America
| | - Fouad T Chebib
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States of America
| | - Vicente E Torres
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States of America.
| |
Collapse
|
30
|
Melick CH, Meng D, Jewell JL. A-kinase anchoring protein 8L interacts with mTORC1 and promotes cell growth. J Biol Chem 2020; 295:8096-8105. [PMID: 32312749 DOI: 10.1074/jbc.ac120.012595] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 04/14/2020] [Indexed: 01/28/2023] Open
Abstract
mTOR complex 1 (mTORC1) senses nutrients to mediate anabolic processes within the cell. Exactly how mTORC1 promotes cell growth remains unclear. Here, we identified a novel mTORC1-interacting protein called protein kinase A anchoring protein 8L (AKAP8L). Using biochemical assays, we found that the N-terminal region of AKAP8L binds to mTORC1 in the cytoplasm. Importantly, loss of AKAP8L decreased mTORC1-mediated processes such as translation, cell growth, and cell proliferation. AKAPs anchor protein kinase A (PKA) through PKA regulatory subunits, and we show that AKAP8L can anchor PKA through regulatory subunit Iα. Reintroducing full-length AKAP8L into cells restored mTORC1-regulated processes, whereas reintroduction of AKAP8L missing the N-terminal region that confers the interaction with mTORC1 did not. Our results suggest a multifaceted role for AKAPs in the cell. We conclude that mTORC1 appears to regulate cell growth, perhaps in part through AKAP8L.
Collapse
Affiliation(s)
- Chase H Melick
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390.,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Delong Meng
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390.,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Jenna L Jewell
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390 .,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| |
Collapse
|
31
|
Baarsma HA, Han B, Poppinga WJ, Driessen S, Elzinga CRS, Halayko AJ, Meurs H, Maarsingh H, Schmidt M. Disruption of AKAP-PKA Interaction Induces Hypercontractility With Concomitant Increase in Proliferation Markers in Human Airway Smooth Muscle. Front Cell Dev Biol 2020; 8:165. [PMID: 32328490 PMCID: PMC7160303 DOI: 10.3389/fcell.2020.00165] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 02/28/2020] [Indexed: 01/11/2023] Open
Abstract
With the ability to switch between proliferative and contractile phenotype, airway smooth muscle (ASM) cells can contribute to the progression of airway diseases such as asthma and chronic obstructive pulmonary disease (COPD), in which airway obstruction is associated with ASM hypertrophy and hypercontractility. A-kinase anchoring proteins (AKAPs) have emerged as important regulatory molecules in various tissues, including ASM cells. AKAPs can anchor the regulatory subunits of protein kinase A (PKA), and guide cellular localization via various targeting domains. Here we investigated whether disruption of the AKAP-PKA interaction, by the cell permeable peptide stearated (st)-Ht31, alters human ASM proliferation and contractility. Treatment of human ASM with st-Ht31 enhanced the expression of protein markers associated with cell proliferation in both cultured cells and intact tissue, although this was not accompanied by an increase in cell viability or cell-cycle progression, suggesting that disruption of AKAP-PKA interaction on its own is not sufficient to drive ASM cell proliferation. Strikingly, st-Ht31 enhanced contractile force generation in human ASM tissue with concomitant upregulation of the contractile protein α-sm-actin. This upregulation of α-sm-actin was independent of mRNA stability, transcription or translation, but was dependent on proteasome function, as the proteasome inhibitor MG-132 prevented the st-Ht31 effect. Collectively, the AKAP-PKA interaction appears to regulate markers of the multi-functional capabilities of ASM, and this alter the physiological function, such as contractility, suggesting potential to contribute to the pathophysiology of airway diseases.
Collapse
Affiliation(s)
- Hoeke A Baarsma
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands.,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Bing Han
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands.,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Wilfred J Poppinga
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands.,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Saskia Driessen
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
| | - Carolina R S Elzinga
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
| | - Andrew J Halayko
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB, Canada
| | - Herman Meurs
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands.,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Harm Maarsingh
- Department of Pharmaceutical Sciences, Lloyd L. Gregory School of Pharmacy, Palm Beach Atlantic University, West Palm Beach, FL, United States
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands.,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
32
|
Helton LG, Kennedy EJ. Targeting Plasmodium with constrained peptides and peptidomimetics. IUBMB Life 2020; 72:1103-1114. [PMID: 32037730 DOI: 10.1002/iub.2244] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 01/24/2020] [Indexed: 01/04/2023]
Abstract
Malaria remains a worldwide health concern with an estimated quarter of a billion people infected and nearly half a million deaths annually. Malaria is caused by a parasite infection from Plasmodium strains which are transmitted from mosquitoes into the human host. Although several small molecule inhibitors have been found to target the early stages of transmission and prevent parasite proliferation, multiple drug resistant parasite strains have emerged and drug resistance remains a major hurdle. As an alternative to small molecule inhibition, several peptide-based therapeutics have been explored for their potential as antimalarial compounds. Chemically constrained peptides or peptidomimetics were developed to target large binding interfaces of parasite-based proteins that have historically been difficult to selectively inhibit using small molecules. Here, we review ongoing research aimed at developing constrained peptides targeting protein-protein interactions pertinent to malaria pathogenesis. These targets include Falcipain-2, the J domain of CDPK1, myosin A tail domain interacting protein, the PKA signaling pathway, and an unclear signaling pathway involving angiotensin-derived peptides. Diverse synthetic methods were also used for each target. Merging parasite biology with synthetic strategies may provide new opportunities to develop alternative methods for uncovering novel antimalarials and may offer an alternate source for targeting drug-resistant parasite strains.
Collapse
Affiliation(s)
- Leah G Helton
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia
| | - Eileen J Kennedy
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia
| |
Collapse
|
33
|
The Role of Cyclic AMP Signaling in Cardiac Fibrosis. Cells 2019; 9:cells9010069. [PMID: 31888098 PMCID: PMC7016856 DOI: 10.3390/cells9010069] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/23/2019] [Accepted: 12/24/2019] [Indexed: 12/18/2022] Open
Abstract
Myocardial stress and injury invariably promote remodeling of the cardiac tissue, which is associated with cardiomyocyte death and development of fibrosis. The fibrotic process is initially triggered by the differentiation of resident cardiac fibroblasts into myofibroblasts. These activated fibroblasts display increased proliferative capacity and secrete large amounts of extracellular matrix. Uncontrolled myofibroblast activation can thus promote heart stiffness, cardiac dysfunction, arrhythmias, and progression to heart failure. Despite the well-established role of myofibroblasts in mediating cardiac disease, our current knowledge on how signaling pathways promoting fibrosis are regulated and coordinated in this cell type is largely incomplete. In this respect, cyclic adenosine monophosphate (cAMP) signaling acts as a major modulator of fibrotic responses activated in fibroblasts of injured or stressed hearts. In particular, accumulating evidence now suggests that upstream cAMP modulators including G protein-coupled receptors, adenylyl cyclases (ACs), and phosphodiesterases (PDEs); downstream cAMP effectors such as protein kinase A (PKA) and the guanine nucleotide exchange factor Epac; and cAMP signaling organizers such as A-kinase anchoring proteins (AKAPs) modulate a variety of fundamental cellular processes involved in myocardial fibrosis including myofibroblast differentiation, proliferation, collagen secretion, and invasiveness. The current review will discuss recent advances highlighting the role of cAMP and AKAP-mediated signaling in regulating pathophysiological responses controlling cardiac fibrosis.
Collapse
|
34
|
Bucko PJ, Lombard CK, Rathbun L, Garcia I, Bhat A, Wordeman L, Smith FD, Maly DJ, Hehnly H, Scott JD. Subcellular drug targeting illuminates local kinase action. eLife 2019; 8:e52220. [PMID: 31872801 PMCID: PMC6930117 DOI: 10.7554/elife.52220] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 11/30/2019] [Indexed: 01/02/2023] Open
Abstract
Deciphering how signaling enzymes operate within discrete microenvironments is fundamental to understanding biological processes. A-kinase anchoring proteins (AKAPs) restrict the range of action of protein kinases within intracellular compartments. We exploited the AKAP targeting concept to create genetically encoded platforms that restrain kinase inhibitor drugs at distinct subcellular locations. Local Kinase Inhibition (LoKI) allows us to ascribe organelle-specific functions to broad specificity kinases. Using chemical genetics, super resolution microscopy, and live-cell imaging we discover that centrosomal delivery of Polo-like kinase 1 (Plk1) and Aurora A (AurA) inhibitors attenuates kinase activity, produces spindle defects, and prolongs mitosis. Targeted inhibition of Plk1 in zebrafish embryos illustrates how centrosomal Plk1 underlies mitotic spindle assembly. Inhibition of kinetochore-associated pools of AurA blocks phosphorylation of microtubule-kinetochore components. This versatile precision pharmacology tool enhances investigation of local kinase biology.
Collapse
Affiliation(s)
- Paula J Bucko
- Department of PharmacologyUniversity of WashingtonSeattleUnited States
| | - Chloe K Lombard
- Department of ChemistryUniversity of WashingtonSeattleUnited States
| | - Lindsay Rathbun
- Department of BiologySyracuse UniversitySyracuseUnited States
| | - Irvin Garcia
- Department of PharmacologyUniversity of WashingtonSeattleUnited States
| | - Akansha Bhat
- Department of PharmacologyUniversity of WashingtonSeattleUnited States
| | - Linda Wordeman
- Department of Physiology and BiophysicsUniversity of WashingtonSeattleUnited States
| | - F Donelson Smith
- Department of PharmacologyUniversity of WashingtonSeattleUnited States
| | - Dustin J Maly
- Department of ChemistryUniversity of WashingtonSeattleUnited States
| | - Heidi Hehnly
- Department of BiologySyracuse UniversitySyracuseUnited States
| | - John D Scott
- Department of PharmacologyUniversity of WashingtonSeattleUnited States
| |
Collapse
|
35
|
Lobingier BT, von Zastrow M. When trafficking and signaling mix: How subcellular location shapes G protein-coupled receptor activation of heterotrimeric G proteins. Traffic 2019; 20:130-136. [PMID: 30578610 DOI: 10.1111/tra.12634] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 12/14/2022]
Abstract
G protein-coupled receptors (GPCRs) physically connect extracellular information with intracellular signal propagation. Membrane trafficking plays a supportive role by "bookending" signaling events: movement through the secretory pathway delivers GPCRs to the cell surface where receptors can sample the extracellular environment, while endocytosis and endolysosomal membrane trafficking provide a versatile system to titrate cellular signaling potential and maintain homeostatic control. Recent evidence suggests that, in addition to these important effects, GPCR trafficking actively shapes the cellular signaling response by altering the location and timing of specific receptor-mediated signaling reactions. Here, we review key experimental evidence underlying this expanding view, focused on GPCR signaling mediated through activation of heterotrimeric G proteins located in the cytoplasm. We then discuss lingering and emerging questions regarding the interface between GPCR signaling and trafficking.
Collapse
Affiliation(s)
- Braden T Lobingier
- Department of Psychiatry and Department of Cellular and Molecular Pharmacology, University of California, San Francisco School of Medicine, San Francisco, California
| | - Mark von Zastrow
- Department of Psychiatry and Department of Cellular and Molecular Pharmacology, University of California, San Francisco School of Medicine, San Francisco, California
| |
Collapse
|
36
|
Xiong J, He J, Xie WP, Hinojosa E, Ambati CSR, Putluri N, Kim HE, Zhu MX, Du G. Rapid affinity purification of intracellular organelles using a twin strep tag. J Cell Sci 2019; 132:jcs.235390. [PMID: 31780580 DOI: 10.1242/jcs.235390] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 11/15/2019] [Indexed: 12/19/2022] Open
Abstract
Cells are internally organized into compartmentalized organelles that execute specialized functions. To understand the functions of individual organelles and their regulations, it is critical to resolve the compositions of individual organelles, which relies on a rapid and efficient isolation method for specific organellar populations. Here, we introduce a robust affinity purification method for rapid isolation of intracellular organelles (e.g. lysosomes, mitochondria and peroxisomes) by taking advantage of the extraordinarily high affinity between the twin strep tag and streptavidin variants. With this method, we can isolate desired organelles with high purity and yield in 3 min from the post-nuclear supernatant of mammalian cells or less than 8 min for the whole purification process. Using lysosomes as an example, we show that the rapid procedure is especially useful for studying transient and fast cellular activities, such as organelle-initiated signaling and organellar contents of small-molecular metabolites. Therefore, our method offers a powerful tool to dissect spatiotemporal regulation and functions of intracellular organelles.
Collapse
Affiliation(s)
- Jian Xiong
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.,Biochemistry and Cell Biology Program, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Jingquan He
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Wendy P Xie
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Ezekiel Hinojosa
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Chandra Shekar R Ambati
- Dan L. Duncan Cancer Center, Advanced Technology Core, Alkek Center for Molecular Discovery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Nagireddy Putluri
- Dan L. Duncan Cancer Center, Advanced Technology Core, Alkek Center for Molecular Discovery, Baylor College of Medicine, Houston, TX 77030, USA.,Department of Molecular & Cell Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hyun-Eui Kim
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.,Biochemistry and Cell Biology Program, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA .,Biochemistry and Cell Biology Program, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Guangwei Du
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA .,Biochemistry and Cell Biology Program, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
37
|
Hu T, Wu X, Li K, Li Y, He P, Wu Z, Fan J, Liu W, Guan M. AKAP12 Endogenous Transcripts Suppress The Proliferation, Migration And Invasion Of Colorectal Cancer Cells By Directly Targeting oncomiR-183-5p. Onco Targets Ther 2019; 12:8301-8310. [PMID: 31632079 PMCID: PMC6790119 DOI: 10.2147/ott.s207600] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 09/15/2019] [Indexed: 12/18/2022] Open
Abstract
Purpose Restoring lost function to suppressor gene products has captured the interest of the research community in the field of gene therapy. AKAP12, also known as Gravin/AKAP250, is a tumor suppressor gene, and its deregulation may be responsible for cancer progression. The aim of this study was to investigate whether AKAP12 mRNA has an anti-cancer function by regulating onco-miRNA expression in colorectal cancer (CRC) cells. Methods miRNAs targeting AKAP12 were predicted by bioinformatics analysis and further confirmed by dual-luciferase reporter assays and RT-qPCR. The altered expression of microRNA was validated in early-stage CRC tumor tissues by miRseq. Cell proliferation was measured by Cell Counting Kit-8 (CCK-8) assay. Cell invasion and migration were detected by transwell and wound healing assays, respectively. In vivo experiments were conducted to confirm the in vitro findings. Results Among all miRNAs, reversed correlation between AKAP12 expression and miRNA-183-5p expression was most significant. Luciferase assays revealed that AKAP12 directly targeted miR-183-5p. The miRseq data showed that miR-183 was also dysregulated at the early stage of tumor development and upregulated in late sub-stage II CRC patients (P<0.01). Mechanistic analysis both in vitro and in vivo demonstrated that anti-miR-183-5p depressed cell proliferation, migration, and invasion in CRC cells while miR-183-5p overexpression resulted in opposite effects. Conclusion Our findings suggested that oncomiR-183-5p promoted the proliferation, migration, and invasion of CRC cells. AKAP12 miRNA-binding elements (MREs) suppressed miRNA-183-5p activities. Any change in expression of AKAP12 thus affected miRNA-183-5p. This may be another anti-tumor mechanism in addition to protein-mediation that regulates tumor suppressor genes.
Collapse
Affiliation(s)
- Tingting Hu
- Department of Clinical Laboratory, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Xuan Wu
- Central Laboratory and Department of Laboratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, People's Republic of China
| | - Ke Li
- Central Laboratory and Department of Laboratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, People's Republic of China
| | - Yuan Li
- Central Laboratory and Department of Laboratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, People's Republic of China
| | - Ping He
- Department of Nuclear Medicine, Nanjing Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Zhiyuan Wu
- Department of Clinical Laboratory, Huashan Hospital North, Fudan University, Shanghai, People's Republic of China
| | - Jie Fan
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Weiwei Liu
- Central Laboratory and Department of Laboratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, People's Republic of China.,Department of Laboratory Medicine, Shanghai Skin Disease Hospital, Tongji University, Shanghai, People's Republic of China
| | - Ming Guan
- Department of Clinical Laboratory, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.,Department of Clinical Laboratory, Huashan Hospital North, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
38
|
Ho PWM, Chan AS, Pavlos NJ, Sims NA, Martin TJ. Brief exposure to full length parathyroid hormone-related protein (PTHrP) causes persistent generation of cyclic AMP through an endocytosis-dependent mechanism. Biochem Pharmacol 2019; 169:113627. [PMID: 31476292 DOI: 10.1016/j.bcp.2019.113627] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 08/28/2019] [Indexed: 12/17/2022]
Abstract
Parathyroid hormone (PTH)-related protein (PTHrP) (gene name Pthlh) was discovered as the factor responsible for the humoral hypercalcemia of malignancy. It shares such sequence similarity with PTH in the amino-terminal region that the two are equally able to act through a single G protein-coupled receptor, PTH1R. A number of biological activities are ascribed to domains of PTHrP beyond the amino-terminal domain. PTH functions as a circulating hormone, but PTHrP is generated locally in many tissues including bone, where it acts as a paracrine factor on osteoblasts and osteocytes. The present study compares how PTH and PTHrP influence cyclic AMP (cAMP) formation through adenylyl cyclase, the first event in cell activation through PTH1R. Brief exposure to full length PTHrP(1-141) in several osteoblastic cell culture systems was followed by sustained adenylyl cyclase activity for more than an hour after ligand washout. This effect was dose-dependent and was not found with shorter PTHrP or PTH peptides even though they were fully able to activate adenylyl cyclase with acute treatment. The persistent activation response to PTHrP(1-141) was seen also with later events in the cAMP/PKA pathway, including persistent activation of CRE-luciferase and sustained regulation of several CREB-responsive mRNAs, up to 24 h after the initial exposure. Pharmacologic blockade of endocytosis prevented the persistent activation of cAMP and gene responses. We conclude that full length PTHrP, the likely local physiological effector in bone, differs in intracellular action to PTH by undergoing endosomal translocation to induce a prolonged adenylyl cyclase activation in its target cells.
Collapse
Affiliation(s)
- Patricia W M Ho
- Bone Biology and Disease Unit, St. Vincent's Institute of Medical Research, Melbourne, Victoria 3065, Australia
| | - Audrey S Chan
- Bone Biology and Disease Unit, St. Vincent's Institute of Medical Research, Melbourne, Victoria 3065, Australia; School of Biomedical Sciences, The University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Nathan J Pavlos
- School of Biomedical Sciences, The University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Natalie A Sims
- Bone Biology and Disease Unit, St. Vincent's Institute of Medical Research, Melbourne, Victoria 3065, Australia; Department of Medicine, The University of Melbourne, St. Vincent's Hospital, Melbourne, Victoria 3065, Australia
| | - T John Martin
- Bone Biology and Disease Unit, St. Vincent's Institute of Medical Research, Melbourne, Victoria 3065, Australia; Department of Medicine, The University of Melbourne, St. Vincent's Hospital, Melbourne, Victoria 3065, Australia.
| |
Collapse
|
39
|
Lu H, Deng S, Zheng M, Hu K. iTRAQ plasma proteomics analysis for candidate biomarkers of type 2 incipient diabetic nephropathy. Clin Proteomics 2019; 16:33. [PMID: 31384238 PMCID: PMC6668123 DOI: 10.1186/s12014-019-9253-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 07/22/2019] [Indexed: 12/11/2022] Open
Abstract
Background Diabetic nephropathy is the most frequent cause of end-stage renal disease worldwide. Identification of biomarkers for diabetic nephropathy for early diagnosis may be the key to avoiding damage from this condition. Methods Proteomic iTRAQ technology was first used to identify differentially expressed plasma proteins in type 2 incipient diabetic nephropathy (IDN) using a Q-Exactive mass spectrometer. Results Compared with controls, 57 proteins (32 upregulated and 25 downregulated proteins) were identified. Furthermore, the gelsolin, collectin-11, PTPRJ, and AKAP-7 proteins were confirmed by Western blots as candidate biomarkers for type 2 IDN through ROC analysis. Conclusions These findings offer a theoretical basis for the early treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- Hongmei Lu
- 1The Second Clinical Medical College, Guangdong Medical University, Dongguan, 523808 China
| | - Shaodong Deng
- 1The Second Clinical Medical College, Guangdong Medical University, Dongguan, 523808 China
| | - Minghui Zheng
- 2Department of Clinical Laboratory, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120 China
| | - Kunhua Hu
- 3Proteomics Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 China
| |
Collapse
|
40
|
Neuronal cAMP/PKA Signaling and Energy Homeostasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1090:31-48. [PMID: 30390284 DOI: 10.1007/978-981-13-1286-1_3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The brain plays a key role in the regulation of body weight and glucose metabolism. Peripheral signals including hormones, metabolites, and neural afferent signals are received and processed by the brain which in turn elicits proper behavioral and metabolic responses for maintaining energy and glucose homeostasis. The cAMP/protein kinase A (PKA) pathway acts downstream G-protein-coupled receptors (GPCR) to mediate the physiological effects of many hormones and neurotransmitters. Activated PKA phosphorylates various proteins including ion channels, enzymes, and transcription factors and regulates their activity. Recent studies have shown that neuronal cAMP/PKA activity in multiple brain regions are involved in the regulation of feeding, energy expenditure, and glucose homeostasis. In this chapter I summarize recent genetic and pharmacological studies concerning the regulation of body weight and glucose homeostasis by cAMP/PKA signaling in the brain.
Collapse
|
41
|
Zhou HY, He JG, Hu ZL, Xue SG, Xu JF, Cui QQ, Gao SQ, Zhou B, Wu PF, Long LH, Wang F, Chen JG. A-Kinase Anchoring Protein 150 and Protein Kinase A Complex in the Basolateral Amygdala Contributes to Depressive-like Behaviors Induced by Chronic Restraint Stress. Biol Psychiatry 2019; 86:131-142. [PMID: 31076080 DOI: 10.1016/j.biopsych.2019.03.967] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 02/11/2019] [Accepted: 03/05/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND The basolateral amygdala (BLA) has been widely implicated in the pathophysiology of major depressive disorder. A-kinase anchoring protein 150 (AKAP150) directs kinases and phosphatases to synaptic glutamate receptors, controlling synaptic transmission and plasticity. However, the role of the AKAP150 in the BLA in major depressive disorder remains poorly understood. METHODS Depressive-like behaviors in C57BL/6J mice were developed by chronic restraint stress (CRS). Mice received either intra-BLA injection of lentivirus-expressing Akap5 short hairpin RNA or Ht-31, a peptide to disrupt the interaction of AKAP150 and protein kinase A (PKA), followed by depressive-like behavioral tests. Alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid glutamate receptor (AMPAR)-mediated miniature excitatory postsynaptic currents were recorded by whole-cell patch-clamp techniques. RESULTS Chronic stress exposure induced depressive-like behaviors, which were accompanied by an increase in total and synaptic AKAP150 expression in the BLA. Accordingly, CRS facilitated the association of AKAP150 with PKA, but not of calcineurin in the BLA. Intra-BLA infusion of lentivirus-expressing Akap5 short hairpin RNA or Ht-31 prevented depressive-like behaviors and normalized phosphorylation of serine 845 and surface expression of AMPAR subunit 1 (GluA1) in the BLA of CRS mice. Finally, blockage of AKAP150-PKA complex signaling rescued the changes in AMPAR-mediated miniature excitatory postsynaptic currents in depressive-like mice. CONCLUSIONS These results suggest that AKAP150-PKA directly modulates BLA neuronal synaptic strength, and that AKAP150-PKA-GluA1 streamline signaling complex is responsible for CRS-induced disruption of synaptic AMPAR-mediated transmission and depressive-like behaviors in mice.
Collapse
Affiliation(s)
- Hai-Yun Zhou
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jin-Gang He
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Laboratory of Neuropsychiatric Diseases, Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Neurological Diseases, Huazhong University of Science and Technology, Ministry of Education of China, Wuhan, China
| | - Zhuang-Li Hu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Laboratory of Neuropsychiatric Diseases, Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Neurological Diseases, Huazhong University of Science and Technology, Ministry of Education of China, Wuhan, China
| | - Shi-Ge Xue
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun-Feng Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian-Qian Cui
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuang-Qi Gao
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bin Zhou
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng-Fei Wu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Laboratory of Neuropsychiatric Diseases, Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Neurological Diseases, Huazhong University of Science and Technology, Ministry of Education of China, Wuhan, China
| | - Li-Hong Long
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Laboratory of Neuropsychiatric Diseases, Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Neurological Diseases, Huazhong University of Science and Technology, Ministry of Education of China, Wuhan, China
| | - Fang Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Laboratory of Neuropsychiatric Diseases, Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Neurological Diseases, Huazhong University of Science and Technology, Ministry of Education of China, Wuhan, China.
| | - Jian-Guo Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Laboratory of Neuropsychiatric Diseases, Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Neurological Diseases, Huazhong University of Science and Technology, Ministry of Education of China, Wuhan, China.
| |
Collapse
|
42
|
Brautigan DL, Shenolikar S. Protein Serine/Threonine Phosphatases: Keys to Unlocking Regulators and Substrates. Annu Rev Biochem 2019; 87:921-964. [PMID: 29925267 DOI: 10.1146/annurev-biochem-062917-012332] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Protein serine/threonine phosphatases (PPPs) are ancient enzymes, with distinct types conserved across eukaryotic evolution. PPPs are segregated into types primarily on the basis of the unique interactions of PPP catalytic subunits with regulatory proteins. The resulting holoenzymes dock substrates distal to the active site to enhance specificity. This review focuses on the subunit and substrate interactions for PPP that depend on short linear motifs. Insights about these motifs from structures of holoenzymes open new opportunities for computational biology approaches to elucidate PPP networks. There is an expanding knowledge base of posttranslational modifications of PPP catalytic and regulatory subunits, as well as of their substrates, including phosphorylation, acetylation, and ubiquitination. Cross talk between these posttranslational modifications creates PPP-based signaling. Knowledge of PPP complexes, signaling clusters, as well as how PPPs communicate with each other in response to cellular signals should unlock the doors to PPP networks and signaling "clouds" that orchestrate and coordinate different aspects of cell physiology.
Collapse
Affiliation(s)
- David L Brautigan
- Center for Cell Signaling and Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA;
| | - Shirish Shenolikar
- Signature Research Programs in Cardiovascular and Metabolic Disorders and Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore 169857
| |
Collapse
|
43
|
Zhang Y, Wang WE, Zhang X, Li Y, Chen B, Liu C, Ai X, Zhang X, Tian Y, Zhang C, Tang M, Szeto C, Hua X, Xie M, Zeng C, Wu Y, Zhou L, Zhu W, Yu D, Houser SR, Chen X. Cardiomyocyte PKA Ablation Enhances Basal Contractility While Eliminates Cardiac β-Adrenergic Response Without Adverse Effects on the Heart. Circ Res 2019; 124:1760-1777. [PMID: 30982412 DOI: 10.1161/circresaha.118.313417] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
RATIONALE PKA (Protein Kinase A) is a major mediator of β-AR (β-adrenergic) regulation of cardiac function, but other mediators have also been suggested. Reduced PKA basal activity and activation are linked to cardiac diseases. However, how complete loss of PKA activity impacts on cardiac physiology and if it causes cardiac dysfunction have never been determined. OBJECTIVES We set to determine how the heart adapts to the loss of cardiomyocyte PKA activity and if it elicits cardiac abnormalities. METHODS AND RESULTS (1) Cardiac PKA activity was almost completely inhibited by expressing a PKA inhibitor peptide in cardiomyocytes (cPKAi) in mice; (2) cPKAi reduced basal phosphorylation of 2 myofilament proteins (TnI [troponin I] and cardiac myosin binding protein C), and one longitudinal SR (sarcoplasmic reticulum) protein (PLB [phospholamban]) but not of the sarcolemmal proteins (Cav1.2 α1c and PLM [phospholemman]), dyadic protein RyR2, and nuclear protein CREB (cAMP response element binding protein) at their PKA phosphorylation sites; (3) cPKAi increased the expression of CaMKII (Ca2+/calmodulin-dependent kinase II), the Cav1.2 β subunits and current, but decreased CaMKII phosphorylation and CaMKII-mediated phosphorylation of PLB and RyR2; (4) These changes resulted in significantly enhanced myofilament Ca2+ sensitivity, prolonged contraction, slowed relaxation but increased myocyte Ca2+ transient and contraction amplitudes; (5) Isoproterenol-induced PKA and CaMKII activation and their phosphorylation of proteins were prevented by cPKAi; (6) cPKAi abolished the increases of heart rate, and cardiac and myocyte contractility by a β-AR agonist (isoproterenol), showing an important role of PKA and a minimal role of PKA-independent β-AR signaling in acute cardiac regulation; (7) cPKAi mice have partial exercise capability probably by enhancing vascular constriction and ventricular filling during β-AR stimulation; and (8) cPKAi mice did not show any cardiac functional or structural abnormalities during the 1-year study period. CONCLUSIONS PKA activity suppression induces a unique Ca2+ handling phenotype, eliminates β-AR regulation of heart rates and cardiac contractility but does not cause cardiac abnormalities.
Collapse
Affiliation(s)
- Ying Zhang
- Cardiology, Daping Hospital, Third Military Medical University, Chongqing (Y.Z., W.E.W., C. Zeng, L.Z.)
- Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University (Y.Z.)
- Department of Physiology and Cardiovascular Research Center (Y.Z., W.E.W., X.Z., Y.L., C.L., X.A., X.Z., C.Z., M.T., C.S., S.R.H., X.C.), Temple University School of Medicine, Philadelphia, PA
| | - Wei Eric Wang
- Cardiology, Daping Hospital, Third Military Medical University, Chongqing (Y.Z., W.E.W., C. Zeng, L.Z.)
- Department of Physiology and Cardiovascular Research Center (Y.Z., W.E.W., X.Z., Y.L., C.L., X.A., X.Z., C.Z., M.T., C.S., S.R.H., X.C.), Temple University School of Medicine, Philadelphia, PA
| | - Xiaoying Zhang
- Department of Physiology and Cardiovascular Research Center (Y.Z., W.E.W., X.Z., Y.L., C.L., X.A., X.Z., C.Z., M.T., C.S., S.R.H., X.C.), Temple University School of Medicine, Philadelphia, PA
| | - Ying Li
- Department of Physiology and Cardiovascular Research Center (Y.Z., W.E.W., X.Z., Y.L., C.L., X.A., X.Z., C.Z., M.T., C.S., S.R.H., X.C.), Temple University School of Medicine, Philadelphia, PA
- The General Hospital of The PLA Rocket Force, Beijing, China (Y.L.)
| | - Biyi Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, François M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine (B.C.)
| | - Chong Liu
- Department of Physiology and Cardiovascular Research Center (Y.Z., W.E.W., X.Z., Y.L., C.L., X.A., X.Z., C.Z., M.T., C.S., S.R.H., X.C.), Temple University School of Medicine, Philadelphia, PA
- Pharmacology, Second Military Medical University, Shanghai (C.L.)
| | - Xiaojie Ai
- Department of Physiology and Cardiovascular Research Center (Y.Z., W.E.W., X.Z., Y.L., C.L., X.A., X.Z., C.Z., M.T., C.S., S.R.H., X.C.), Temple University School of Medicine, Philadelphia, PA
- School of Agriculture and Biology, Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai Jiao Tong University (X.A.)
| | - Xiaoxiao Zhang
- Department of Physiology and Cardiovascular Research Center (Y.Z., W.E.W., X.Z., Y.L., C.L., X.A., X.Z., C.Z., M.T., C.S., S.R.H., X.C.), Temple University School of Medicine, Philadelphia, PA
- Department of Ultrasound, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan (X.Z., M.X.)
| | - Ying Tian
- Department of Pharmacology, Center for Translational Medicine (Y.T., W.Z.), Temple University School of Medicine, Philadelphia, PA
| | - Chen Zhang
- Cardiology, Daping Hospital, Third Military Medical University, Chongqing (Y.Z., W.E.W., C. Zeng, L.Z.)
- Department of Physiology and Cardiovascular Research Center (Y.Z., W.E.W., X.Z., Y.L., C.L., X.A., X.Z., C.Z., M.T., C.S., S.R.H., X.C.), Temple University School of Medicine, Philadelphia, PA
| | - Mingxin Tang
- Department of Physiology and Cardiovascular Research Center (Y.Z., W.E.W., X.Z., Y.L., C.L., X.A., X.Z., C.Z., M.T., C.S., S.R.H., X.C.), Temple University School of Medicine, Philadelphia, PA
| | - Christopher Szeto
- Department of Physiology and Cardiovascular Research Center (Y.Z., W.E.W., X.Z., Y.L., C.L., X.A., X.Z., C.Z., M.T., C.S., S.R.H., X.C.), Temple University School of Medicine, Philadelphia, PA
| | - Xiang Hua
- Fox Chase Cancer Center, Philadelphia, PA (X.H.)
| | - Mingxin Xie
- Department of Ultrasound, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan (X.Z., M.X.)
| | | | - Yingjie Wu
- Institute of Genome Engineered Animal Models for Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Liaoning (Y.W.)
| | - Lin Zhou
- Cardiology, Daping Hospital, Third Military Medical University, Chongqing (Y.Z., W.E.W., C. Zeng, L.Z.)
| | - Weizhong Zhu
- Department of Pharmacology, Center for Translational Medicine (Y.T., W.Z.), Temple University School of Medicine, Philadelphia, PA
- Pharmacology, School of Pharmacy, Nantong University, Jiangsu (W.Z.)
| | - Daohai Yu
- Clinical Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (D.Y.)
| | - Steven R Houser
- Department of Physiology and Cardiovascular Research Center (Y.Z., W.E.W., X.Z., Y.L., C.L., X.A., X.Z., C.Z., M.T., C.S., S.R.H., X.C.), Temple University School of Medicine, Philadelphia, PA
| | - Xiongwen Chen
- Department of Physiology and Cardiovascular Research Center (Y.Z., W.E.W., X.Z., Y.L., C.L., X.A., X.Z., C.Z., M.T., C.S., S.R.H., X.C.), Temple University School of Medicine, Philadelphia, PA
| |
Collapse
|
44
|
Djillani A, Mazella J, Heurteaux C, Borsotto M. Role of TREK-1 in Health and Disease, Focus on the Central Nervous System. Front Pharmacol 2019; 10:379. [PMID: 31031627 PMCID: PMC6470294 DOI: 10.3389/fphar.2019.00379] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/26/2019] [Indexed: 01/22/2023] Open
Abstract
TREK-1 is the most studied background K2P channel. Its main role is to control cell excitability and maintain the membrane potential below the threshold of depolarization. TREK-1 is multi-regulated by a variety of physical and chemical stimuli which makes it a very promising and challenging target in the treatment of several pathologies. It is mainly expressed in the brain but also in heart, smooth muscle cells, endocrine pancreas, and prostate. In the nervous system, TREK-1 is involved in many physiological and pathological processes such as depression, neuroprotection, pain, and anesthesia. These properties explain why many laboratories and pharmaceutical companies have been focusing their research on screening and developing highly efficient modulators of TREK-1 channels. In this review, we summarize the different roles of TREK-1 that have been investigated so far in attempt to characterize pharmacological tools and new molecules to modulate cellular functions controlled by TREK-1.
Collapse
Affiliation(s)
- Alaeddine Djillani
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, Université Côte d'Azur, Valbonne, France
| | - Jean Mazella
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, Université Côte d'Azur, Valbonne, France
| | - Catherine Heurteaux
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, Université Côte d'Azur, Valbonne, France
| | - Marc Borsotto
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, Université Côte d'Azur, Valbonne, France
| |
Collapse
|
45
|
Personalized prediction of genes with tumor-causing somatic mutations based on multi-modal deep Boltzmann machine. Neurocomputing 2019. [DOI: 10.1016/j.neucom.2018.02.096] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
46
|
Single nucleotide polymorphisms alter kinase anchoring and the subcellular targeting of A-kinase anchoring proteins. Proc Natl Acad Sci U S A 2018; 115:E11465-E11474. [PMID: 30455320 DOI: 10.1073/pnas.1816614115] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
A-kinase anchoring proteins (AKAPs) shape second-messenger signaling responses by constraining protein kinase A (PKA) at precise intracellular locations. A defining feature of AKAPs is a helical region that binds to regulatory subunits (RII) of PKA. Mining patient-derived databases has identified 42 nonsynonymous SNPs in the PKA-anchoring helices of five AKAPs. Solid-phase RII binding assays confirmed that 21 of these amino acid substitutions disrupt PKA anchoring. The most deleterious side-chain modifications are situated toward C-termini of AKAP helices. More extensive analysis was conducted on a valine-to-methionine variant in the PKA-anchoring helix of AKAP18. Molecular modeling indicates that additional density provided by methionine at position 282 in the AKAP18γ isoform deflects the pitch of the helical anchoring surface outward by 6.6°. Fluorescence polarization measurements show that this subtle topological change reduces RII-binding affinity 8.8-fold and impairs cAMP responsive potentiation of L-type Ca2+ currents in situ. Live-cell imaging of AKAP18γ V282M-GFP adducts led to the unexpected discovery that loss of PKA anchoring promotes nuclear accumulation of this polymorphic variant. Targeting proceeds via a mechanism whereby association with the PKA holoenzyme masks a polybasic nuclear localization signal on the anchoring protein. This led to the discovery of AKAP18ε: an exclusively nuclear isoform that lacks a PKA-anchoring helix. Enzyme-mediated proximity-proteomics reveal that compartment-selective variants of AKAP18 associate with distinct binding partners. Thus, naturally occurring PKA-anchoring-defective AKAP variants not only perturb dissemination of local second-messenger responses, but also may influence the intracellular distribution of certain AKAP18 isoforms.
Collapse
|
47
|
Fyn Regulates Binding Partners of Cyclic-AMP Dependent Protein Kinase A. Proteomes 2018; 6:proteomes6040037. [PMID: 30274258 PMCID: PMC6313912 DOI: 10.3390/proteomes6040037] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/26/2018] [Accepted: 09/27/2018] [Indexed: 12/03/2022] Open
Abstract
The cAMP-dependent protein kinase A (PKA) is a serine/threonine kinase involved in many fundamental cellular processes, including migration and proliferation. Recently, we found that the Src family kinase Fyn phosphorylates the catalytic subunit of PKA (PKA-C) at Y69, thereby increasing PKA kinase activity. We also showed that Fyn induced the phosphorylation of cellular proteins within the PKA preferred target motif. This led to the hypothesis that Fyn could affect proteins in complex with PKA. To test this, we employed a quantitative mass spectrometry approach to identify Fyn-dependent binding partners in complex with PKA-C. We found Fyn enhanced the binding of PKA-C to several cytoskeletal regulators that localize to the centrosome and Golgi apparatus. Three of these Fyn-induced PKA interactors, AKAP9, PDE4DIP, and CDK5RAP2, were validated biochemically and were shown to exist in complex with Fyn and PKA in a glioblastoma cell line. Intriguingly, the complexes formed between PKA-C and these known AKAPs were dependent upon Fyn catalytic activity and expression levels. In addition, we identified Fyn-regulated phosphorylation sites on proteins in complex with PKA-C. We also identified and biochemically validated a novel PKA-C interactor, LARP4, which complexed with PKA in the absence of Fyn. These results demonstrate the ability of Fyn to influence the docking of PKA to specific cellular scaffolds and suggest that Fyn may affect the downstream substrates targeted by PKA.
Collapse
|
48
|
Abstract
Abstract Primary sensory neurons are responsible for transmitting sensory information from the peripheral to the central nervous system. Their responses to incoming stimulation become greatly enhanced and prolonged following inflammation, giving rise to exaggerated nociceptive responses and chronic pain. The inflammatory mediator, prostaglandin E2 (PGE2), released from the inflamed tissue surrounding the terminals of sensory neurons contributes to the abnormal pain responses. PGE2 acts on G protein-coupled EP receptors to activate adenylyl cyclase, which catalyzes the conversion of adenosine triphosphate to cyclic adenosine 3′,5′-monophosphate (cAMP). Under normal conditions, cAMP activates primarily protein kinase A. After inflammation, cAMP also activates the exchange proteins activated by cAMP (Epacs) to produce exaggerated PGE2-mediated hyperalgesia. The role of cAMP-Epac signaling in the generation of hypersensitivity is the topic of this review.
Collapse
Affiliation(s)
| | - Yanping Gu
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch Galveston, TX 77555-1069, USA
| |
Collapse
|
49
|
Shepard RD, Gouty S, Kassis H, Berenji A, Zhu W, Cox BM, Nugent FS. Targeting histone deacetylation for recovery of maternal deprivation-induced changes in BDNF and AKAP150 expression in the VTA. Exp Neurol 2018; 309:160-168. [PMID: 30102916 DOI: 10.1016/j.expneurol.2018.08.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/17/2018] [Accepted: 08/09/2018] [Indexed: 12/21/2022]
Abstract
Severe early life stressors increase the probability of developing psychiatric disorders later in life through modifications in neuronal circuits controlling brain monoaminergic signaling. Our previous work demonstrated that 24 h maternal deprivation (MD) in male Sprague Dawley rats modifies dopamine (DA) signaling from the ventral tegmental area (VTA) through changes at GABAergic synapses that were reversible by in vitro histone deacetylase (HDAC) inhibition which led to restoration of the scaffold A-kinase anchoring protein (AKAP150) signaling and subsequently recovered GABAergic plasticity (Authement et al., 2015). Using a combination of in situ hybridization, Western blots and immunohistochemistry, we confirmed that MD-induced epigenetic modifications at the level of histone acetylation were associated with an upregulation of HDAC2. MD also increased Akap5 mRNA levels in the VTA. Western blot analysis of AKAP150 protein expression showed an increase in synaptic levels of AKAP150 protein in the VTA with an accompanying decrease in synaptic levels of protein kinase A (PKA). Moreover, the abundance of mature brain-derived neurotrophic factor (BDNF) protein of VTA tissues from MD rats was significantly lower than in control groups. In vivo systemic injection with a selective class I HDAC inhibitor (CI-994) was sufficient to reverse MD-induced histone hypoacetylation in the VTA for 24 h after the injection. Furthermore, HDAC inhibition normalized the levels of mBDNF and AKAP150 proteins at 24 h. Our data suggest that HDAC-mediated targeting of BDNF and AKAP-dependent local signaling within VTA could provide novel therapeutics for prevention of later-life psychopathology.
Collapse
Affiliation(s)
- Ryan D Shepard
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - Shawn Gouty
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - Haifa Kassis
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - Aylar Berenji
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - William Zhu
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - Brian M Cox
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - Fereshteh S Nugent
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA.
| |
Collapse
|
50
|
The origin of GSKIP, a multifaceted regulatory factor in the mammalian Wnt pathway. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:1046-1059. [DOI: 10.1016/j.bbamcr.2018.04.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 04/13/2018] [Accepted: 04/19/2018] [Indexed: 11/17/2022]
|