1
|
Zheng Z, Zong Y, Ma Y, Tian Y, Pang Y, Zhang C, Gao J. Glucagon-like peptide-1 receptor: mechanisms and advances in therapy. Signal Transduct Target Ther 2024; 9:234. [PMID: 39289339 PMCID: PMC11408715 DOI: 10.1038/s41392-024-01931-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 06/17/2024] [Accepted: 07/16/2024] [Indexed: 09/19/2024] Open
Abstract
The glucagon-like peptide-1 (GLP-1) receptor, known as GLP-1R, is a vital component of the G protein-coupled receptor (GPCR) family and is found primarily on the surfaces of various cell types within the human body. This receptor specifically interacts with GLP-1, a key hormone that plays an integral role in regulating blood glucose levels, lipid metabolism, and several other crucial biological functions. In recent years, GLP-1 medications have become a focal point in the medical community due to their innovative treatment mechanisms, significant therapeutic efficacy, and broad development prospects. This article thoroughly traces the developmental milestones of GLP-1 drugs, from their initial discovery to their clinical application, detailing the evolution of diverse GLP-1 medications along with their distinct pharmacological properties. Additionally, this paper explores the potential applications of GLP-1 receptor agonists (GLP-1RAs) in fields such as neuroprotection, anti-infection measures, the reduction of various types of inflammation, and the enhancement of cardiovascular function. It provides an in-depth assessment of the effectiveness of GLP-1RAs across multiple body systems-including the nervous, cardiovascular, musculoskeletal, and digestive systems. This includes integrating the latest clinical trial data and delving into potential signaling pathways and pharmacological mechanisms. The primary goal of this article is to emphasize the extensive benefits of using GLP-1RAs in treating a broad spectrum of diseases, such as obesity, cardiovascular diseases, non-alcoholic fatty liver disease (NAFLD), neurodegenerative diseases, musculoskeletal inflammation, and various forms of cancer. The ongoing development of new indications for GLP-1 drugs offers promising prospects for further expanding therapeutic interventions, showcasing their significant potential in the medical field.
Collapse
Affiliation(s)
- Zhikai Zheng
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yao Zong
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, WA, 6009, Australia
| | - Yiyang Ma
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yucheng Tian
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yidan Pang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Changqing Zhang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
2
|
Yang S, Zhou D, Zhang C, Xiang J, Xi X. Function of m 5C RNA methyltransferase NOP2 in high-grade serous ovarian cancer. Cancer Biol Ther 2023; 24:2263921. [PMID: 37800580 PMCID: PMC10561575 DOI: 10.1080/15384047.2023.2263921] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 09/12/2023] [Indexed: 10/07/2023] Open
Abstract
RNA methyltransferase nucleolar protein p120 (NOP2), commonly referred to as NOP2/Sun RNA methyltransferase family member 1 (NSUN1), is involved in cell proliferation and is highly expressed in various cancers. However, its role in high-grade serous ovarian cancer (HGSOC) remains unclear. Our study investigated the expression of NOP2 in HGSOC tissues and normal fimbria tissues, and found that NOP2 was significantly upregulated in HGSOC tissues. Our experiments showed that NOP2 overexpression promoted cell proliferation in vivo and in vitro and increased the migration and invasion ability of HGSOC cells in vitro. Furthermore, we identified Rap guanine nucleotide exchange factor 4 (RAPGEF4) as a potential downstream target of NOP2 in HGSOC. Finally, our findings suggest that the regulation of NOP2 and RAPGEF4 may depend on m5C methylation levels.
Collapse
Affiliation(s)
- Shimin Yang
- Department of Gynecology and Obstetrics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongmei Zhou
- Department of Gynecology and Obstetrics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chunxiao Zhang
- Department of Gynecology and Obstetrics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiangdong Xiang
- Department of Gynecology and Obstetrics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaowei Xi
- Department of Gynecology and Obstetrics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Iannucci LF, D'Erchia AM, Picardi E, Bettio D, Conca F, Surdo NC, Di Benedetto G, Musso D, Arrigoni C, Lolicato M, Vismara M, Grisan F, Salviati L, Milanesi L, Pesole G, Lefkimmiatis K. Cyclic AMP induces reversible EPAC1 condensates that regulate histone transcription. Nat Commun 2023; 14:5521. [PMID: 37684224 PMCID: PMC10491619 DOI: 10.1038/s41467-023-41088-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
The second messenger cyclic AMP regulates many nuclear processes including transcription, pre-mRNA splicing and mitosis. While most functions are attributed to protein kinase A, accumulating evidence suggests that not all nuclear cyclic AMP-dependent effects are mediated by this kinase, implying that other effectors may be involved. Here we explore the nuclear roles of Exchange Protein Activated by cyclic AMP 1. We find that it enters the nucleus where forms reversible biomolecular condensates in response to cyclic AMP. This phenomenon depends on intrinsically disordered regions present at its amino-terminus and is independent of protein kinase A. Finally, we demonstrate that nuclear Exchange Protein Activated by cyclic AMP 1 condensates assemble at genomic loci on chromosome 6 in the proximity of Histone Locus Bodies and promote the transcription of a histone gene cluster. Collectively, our data reveal an unexpected mechanism through which cyclic AMP contributes to nuclear spatial compartmentalization and promotes the transcription of specific genes.
Collapse
Affiliation(s)
- Liliana Felicia Iannucci
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Veneto Institute of Molecular Medicine, 35129, Padova, Italy
| | - Anna Maria D'Erchia
- Department of Biosciences, Biotechnologies and Environment, University of Bari "Aldo Moro", Bari, Italy
| | - Ernesto Picardi
- Department of Biosciences, Biotechnologies and Environment, University of Bari "Aldo Moro", Bari, Italy
| | - Daniela Bettio
- Clinical Genetics Unit, Department of Women's and Children's Health, University of Padova, Padova, Italy
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Filippo Conca
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Veneto Institute of Molecular Medicine, 35129, Padova, Italy
| | - Nicoletta Concetta Surdo
- Veneto Institute of Molecular Medicine, 35129, Padova, Italy
- Institute of Neuroscience (IN-CNR), National Research Council of Italy, Padova, Italy
| | - Giulietta Di Benedetto
- Veneto Institute of Molecular Medicine, 35129, Padova, Italy
- Institute of Neuroscience (IN-CNR), National Research Council of Italy, Padova, Italy
| | - Deborah Musso
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | | | - Marco Lolicato
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Mauro Vismara
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Veneto Institute of Molecular Medicine, 35129, Padova, Italy
| | | | - Leonardo Salviati
- Clinical Genetics Unit, Department of Women's and Children's Health, University of Padova, Padova, Italy
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Luciano Milanesi
- Institute of Biomedical Technologies, National Research Council of Italy, Milan, Italy
| | - Graziano Pesole
- Department of Biosciences, Biotechnologies and Environment, University of Bari "Aldo Moro", Bari, Italy
| | - Konstantinos Lefkimmiatis
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.
- Veneto Institute of Molecular Medicine, 35129, Padova, Italy.
- Institute of Neuroscience (IN-CNR), National Research Council of Italy, Padova, Italy.
| |
Collapse
|
4
|
Mazevet M, Belhadef A, Ribeiro M, Dayde D, Llach A, Laudette M, Belleville T, Mateo P, Gressette M, Lefebvre F, Chen J, Bachelot-Loza C, Rucker-Martin C, Lezoualch F, Crozatier B, Benitah JP, Vozenin MC, Fischmeister R, Gomez AM, Lemaire C, Morel E. EPAC1 inhibition protects the heart from doxorubicin-induced toxicity. eLife 2023; 12:e83831. [PMID: 37551870 PMCID: PMC10484526 DOI: 10.7554/elife.83831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 08/03/2023] [Indexed: 08/09/2023] Open
Abstract
Anthracyclines, such as doxorubicin (Dox), are widely used chemotherapeutic agents for the treatment of solid tumors and hematologic malignancies. However, they frequently induce cardiotoxicity leading to dilated cardiomyopathy and heart failure. This study sought to investigate the role of the exchange protein directly activated by cAMP (EPAC) in Dox-induced cardiotoxicity and the potential cardioprotective effects of EPAC inhibition. We show that Dox induces DNA damage and cardiomyocyte cell death with apoptotic features. Dox also led to an increase in both cAMP concentration and EPAC1 activity. The pharmacological inhibition of EPAC1 (with CE3F4) but not EPAC2 alleviated the whole Dox-induced pattern of alterations. When administered in vivo, Dox-treated WT mice developed a dilated cardiomyopathy which was totally prevented in EPAC1 knock-out (KO) mice. Moreover, EPAC1 inhibition potentiated Dox-induced cell death in several human cancer cell lines. Thus, EPAC1 inhibition appears as a potential therapeutic strategy to limit Dox-induced cardiomyopathy without interfering with its antitumoral activity.
Collapse
Affiliation(s)
| | | | | | | | | | - Marion Laudette
- Institut des Maladies Metaboliques et Cardiovasculaires - I2MC, INSERM, Université de ToulouseToulouseFrance
| | - Tiphaine Belleville
- Innovations Thérapeutiques en Hémostase - UMR-S 1140, INSERM, Faculté de Pharmacie, Université Paris Descartes, Sorbonne Paris CitéParisFrance
| | | | | | | | - Ju Chen
- Basic Cardiac Research UCSD School of Medicine La JollaSan DiegoUnited States
| | - Christilla Bachelot-Loza
- Innovations Thérapeutiques en Hémostase - UMR-S 1140, INSERM, Faculté de Pharmacie, Université Paris Descartes, Sorbonne Paris CitéParisFrance
| | - Catherine Rucker-Martin
- Faculté de Médecine, Université Paris-SaclayLe Kremlin BicêtreFrance
- Inserm UMR_S 999, Hôpital Marie LannelongueLe Plessis RobinsonFrance
| | - Frank Lezoualch
- Institut des Maladies Metaboliques et Cardiovasculaires - I2MC, INSERM, Université de ToulouseToulouseFrance
| | | | | | | | | | | | - Christophe Lemaire
- Université Paris-SaclayOrsayFrance
- Université Paris-Saclay, UVSQ, InsermOrsayFrance
| | | |
Collapse
|
5
|
Yang W, Xia F, Mei F, Shi S, Robichaux WG, Lin W, Zhang W, Liu H, Cheng X. Upregulation of Epac1 Promotes Pericyte Loss by Inducing Mitochondrial Fission, Reactive Oxygen Species Production, and Apoptosis. Invest Ophthalmol Vis Sci 2023; 64:34. [PMID: 37651112 PMCID: PMC10476449 DOI: 10.1167/iovs.64.11.34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 08/09/2023] [Indexed: 09/01/2023] Open
Abstract
Purpose The pathogenic mechanisms behind the development of ischemic retinopathy are complex and poorly understood. This study investigates the involvement of exchange protein directly activated by cAMP (Epac)1 signaling in pericyte injury during ischemic retinopathy, including diabetic retinopathy, a disease that threatens vision. Methods Mouse models of retinal ischemia-reperfusion injury and type 1 diabetes induced by streptozotocin were used to investigate the pathogenesis of these diseases. The roles of Epac1 signaling in the pathogenesis of ischemic retinopathy were determined by an Epac1 knockout mouse model. The cellular and molecular mechanisms of Epac1-mediated pericyte dysfunction in response to high glucose were investigated by specific modulation of Epac1 activity in primary human retinal pericytes using Epac1-specific RNA interference and a pharmacological inhibitor. Results Ischemic injury or diabetes-induced retinal capillary degeneration were associated with an increased expression of Epac1 in the mouse retinal vasculature, including both endothelial cells and pericytes. Genetic deletion of Epac1 protected ischemic injury-induced pericyte loss and capillary degeneration in the mouse retina. Furthermore, high glucose-induced Epac1 expression in retinal pericytes was accompanied by increased Drp1 phosphorylation, mitochondrial fission, reactive oxygen species production, and caspase 3 activation. Inhibition of Epac1 via RNA interference or pharmacological approaches blocked high glucose-mediated mitochondrial dysfunction and caspase 3 activation. Conclusions Our study reveals an important role of Epac1 signaling in mitochondrial dynamics, reactive oxygen species production, and apoptosis in retinal pericytes and identifies Epac1 as a therapeutic target for treating ischemic retinopathy.
Collapse
Affiliation(s)
- Wenli Yang
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas, United States
- Texas Therapeutics Institute, University of Texas Health Science Center, Houston, Texas, United States
| | - Fan Xia
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
| | - Fang Mei
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas, United States
- Texas Therapeutics Institute, University of Texas Health Science Center, Houston, Texas, United States
| | - Shuizhen Shi
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
| | - William G. Robichaux
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas, United States
- Texas Therapeutics Institute, University of Texas Health Science Center, Houston, Texas, United States
| | - Wei Lin
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas, United States
- Texas Therapeutics Institute, University of Texas Health Science Center, Houston, Texas, United States
| | - Wenbo Zhang
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
- Department of Neurobiology, University of Texas Medical Branch, Galveston, Texas, United States
| | - Hua Liu
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas, United States
- Texas Therapeutics Institute, University of Texas Health Science Center, Houston, Texas, United States
| |
Collapse
|
6
|
Slika H, Mansour H, Nasser SA, Shaito A, Kobeissy F, Orekhov AN, Pintus G, Eid AH. Epac as a tractable therapeutic target. Eur J Pharmacol 2023; 945:175645. [PMID: 36894048 DOI: 10.1016/j.ejphar.2023.175645] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 02/26/2023] [Accepted: 03/06/2023] [Indexed: 03/09/2023]
Abstract
In 1957, cyclic adenosine monophosphate (cAMP) was identified as the first secondary messenger, and the first signaling cascade discovered was the cAMP-protein kinase A (PKA) pathway. Since then, cAMP has received increasing attention given its multitude of actions. Not long ago, a new cAMP effector named exchange protein directly activated by cAMP (Epac) emerged as a critical mediator of cAMP's actions. Epac mediates a plethora of pathophysiologic processes and contributes to the pathogenesis of several diseases such as cancer, cardiovascular disease, diabetes, lung fibrosis, neurological disorders, and others. These findings strongly underscore the potential of Epac as a tractable therapeutic target. In this context, Epac modulators seem to possess unique characteristics and advantages and hold the promise of providing more efficacious treatments for a wide array of diseases. This paper provides an in-depth dissection and analysis of Epac structure, distribution, subcellular compartmentalization, and signaling mechanisms. We elaborate on how these characteristics can be utilized to design specific, efficient, and safe Epac agonists and antagonists that can be incorporated into future pharmacotherapeutics. In addition, we provide a detailed portfolio for specific Epac modulators highlighting their discovery, advantages, potential concerns, and utilization in the context of clinical disease entities.
Collapse
Affiliation(s)
- Hasan Slika
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, P.O. Box 11-0236, Lebanon.
| | - Hadi Mansour
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, P.O. Box 11-0236, Lebanon.
| | | | - Abdullah Shaito
- Biomedical Research Center, Qatar University, Doha, P.O. Box: 2713, Qatar.
| | - Firas Kobeissy
- Department of Neurobiology and Neuroscience, Morehouse School of Medicine, Atlanta, Georgia, USA.
| | - Alexander N Orekhov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Institute of Human Morphology, 3 Tsyurupa Street, Moscow, 117418, Russia; Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, Moscow, 125315, Russia; Institute for Atherosclerosis Research, Skolkovo Innovative Center, Osennyaya Street 4-1-207, Moscow, 121609, Russia.
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, 07100, Sassari, Italy.
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, P.O. Box 2713, Qatar.
| |
Collapse
|
7
|
Ahmed MB, Alghamdi AAA, Islam SU, Lee JS, Lee YS. cAMP Signaling in Cancer: A PKA-CREB and EPAC-Centric Approach. Cells 2022; 11:cells11132020. [PMID: 35805104 PMCID: PMC9266045 DOI: 10.3390/cells11132020] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/17/2022] [Accepted: 06/23/2022] [Indexed: 02/01/2023] Open
Abstract
Cancer is one of the most common causes of death globally. Despite extensive research and considerable advances in cancer therapy, the fundamentals of the disease remain unclear. Understanding the key signaling mechanisms that cause cancer cell malignancy may help to uncover new pharmaco-targets. Cyclic adenosine monophosphate (cAMP) regulates various biological functions, including those in malignant cells. Understanding intracellular second messenger pathways is crucial for identifying downstream proteins involved in cancer growth and development. cAMP regulates cell signaling and a variety of physiological and pathological activities. There may be an impact on gene transcription from protein kinase A (PKA) as well as its downstream effectors, such as cAMP response element-binding protein (CREB). The position of CREB downstream of numerous growth signaling pathways implies its oncogenic potential in tumor cells. Tumor growth is associated with increased CREB expression and activation. PKA can be used as both an onco-drug target and a biomarker to find, identify, and stage tumors. Exploring cAMP effectors and their downstream pathways in cancer has become easier using exchange protein directly activated by cAMP (EPAC) modulators. This signaling system may inhibit or accelerate tumor growth depending on the tumor and its environment. As cAMP and its effectors are critical for cancer development, targeting them may be a useful cancer treatment strategy. Moreover, by reviewing the material from a distinct viewpoint, this review aims to give a knowledge of the impact of the cAMP signaling pathway and the related effectors on cancer incidence and development. These innovative insights seek to encourage the development of novel treatment techniques and new approaches.
Collapse
Affiliation(s)
- Muhammad Bilal Ahmed
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea; (M.B.A.); (J.-S.L.)
| | | | - Salman Ul Islam
- Department of Pharmacy, Cecos University, Peshawar, Street 1, Sector F 5 Phase 6 Hayatabad, Peshawar 25000, Pakistan;
| | - Joon-Seok Lee
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea; (M.B.A.); (J.-S.L.)
| | - Young-Sup Lee
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea; (M.B.A.); (J.-S.L.)
- Correspondence: ; Tel.: +82-53-950-6353; Fax: +82-53-943-2762
| |
Collapse
|
8
|
Yang W, Robichaux WG, Mei FC, Lin W, Li L, Pan S, White MA, Chen Y, Cheng X. Epac1 activation by cAMP regulates cellular SUMOylation and promotes the formation of biomolecular condensates. SCIENCE ADVANCES 2022; 8:eabm2960. [PMID: 35442725 PMCID: PMC9020664 DOI: 10.1126/sciadv.abm2960] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 03/04/2022] [Indexed: 06/14/2023]
Abstract
Protein SUMOylation plays an essential role in maintaining cellular homeostasis when cells are under stress. However, precisely how SUMOylation is regulated, and a molecular mechanism linking cellular stress to SUMOylation, remains elusive. Here, we report that cAMP, a major stress-response second messenger, acts through Epac1 as a regulator of cellular SUMOylation. The Epac1-associated proteome is highly enriched with components of the SUMOylation pathway. Activation of Epac1 by intracellular cAMP triggers phase separation and the formation of nuclear condensates containing Epac1 and general components of the SUMOylation machinery to promote cellular SUMOylation. Furthermore, genetic knockout of Epac1 obliterates oxidized low-density lipoprotein-induced cellular SUMOylation in macrophages, leading to suppression of foam cell formation. These results provide a direct nexus connecting two major cellular stress responses to define a molecular mechanism in which cAMP regulates the dynamics of cellular condensates to modulate protein SUMOylation.
Collapse
Affiliation(s)
- Wenli Yang
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center, Houston, TX, USA
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - William G. Robichaux
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center, Houston, TX, USA
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Fang C. Mei
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center, Houston, TX, USA
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Wei Lin
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center, Houston, TX, USA
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Li Li
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Sheng Pan
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Mark A. White
- Sealy Center for Structural Biology and Molecular Biophysics, The University of Texas Medical Branch, Galveston, TX, USA
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Yuan Chen
- Department of Surgery and Moores Cancer Center, UC San Diego Health, La Jolla, CA, USA
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center, Houston, TX, USA
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| |
Collapse
|
9
|
Ni Z, Cheng X. Origin and Isoform Specific Functions of Exchange Proteins Directly Activated by cAMP: A Phylogenetic Analysis. Cells 2021; 10:cells10102750. [PMID: 34685730 PMCID: PMC8534922 DOI: 10.3390/cells10102750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/09/2021] [Accepted: 10/09/2021] [Indexed: 12/21/2022] Open
Abstract
Exchange proteins directly activated by cAMP (EPAC1 and EPAC2) are one of the several families of cellular effectors of the prototypical second messenger cAMP. To understand the origin and molecular evolution of EPAC proteins, we performed a comprehensive phylogenetic analysis of EPAC1 and EPAC2. Our study demonstrates that unlike its cousin PKA, EPAC proteins are only present in multicellular Metazoa. Within the EPAC family, EPAC1 is only associated with chordates, while EPAC2 spans the entire animal kingdom. Despite a much more contemporary origin, EPAC1 proteins show much more sequence diversity among species, suggesting that EPAC1 has undergone more selection and evolved faster than EPAC2. Phylogenetic analyses of the individual cAMP binding domain (CBD) and guanine nucleotide exchange (GEF) domain of EPACs, two most conserved regions between the two isoforms, further reveal that EPAC1 and EPAC2 are closely clustered together within both the larger cyclic nucleotide receptor and RAPGEF families. These results support the notion that EPAC1 and EPAC2 share a common ancestor resulting from a fusion between the CBD of PKA and the GEF from RAPGEF1. On the other hand, the two terminal extremities and the RAS-association (RA) domains show the most sequence diversity between the two isoforms. Sequence diversities within these regions contribute significantly to the isoform-specific functions of EPACs. Importantly, unique isoform-specific sequence motifs within the RA domain have been identified.
Collapse
Affiliation(s)
- Zhuofu Ni
- Department of Integrative Biology & Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA;
| | - Xiaodong Cheng
- Department of Integrative Biology & Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA;
- Texas Therapeutics Institute, Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Correspondence: ; Tel.: +1-713-500-7487
| |
Collapse
|
10
|
Abstract
The well-known second messenger cyclic adenosine monophosphate (cAMP) regulates the morphology and physiology of neurons and thus higher cognitive brain functions. The discovery of exchange protein activated by cAMP (Epac) as a guanine nucleotide exchange factor for Rap GTPases has shed light on protein kinase A (PKA)-independent functions of cAMP signaling in neural tissues. Studies of cAMP-Epac-mediated signaling in neurons under normal and disease conditions also revealed its diverse contributions to neurodevelopment, synaptic remodeling, and neurotransmitter release, as well as learning, memory, and emotion. In this mini-review, the various roles of Epac isoforms, including Epac1 and Epac2, highly expressed in neural tissues are summarized, and controversies or issues are highlighted that need to be resolved to uncover the critical functions of Epac in neural tissues and the potential for a new therapeutic target of mental disorders.
Collapse
Affiliation(s)
- Kyungmin Lee
- Laboratory for Behavioral Neural Circuitry and Physiology, Department of Anatomy, Brain Science & Engineering Institute, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| |
Collapse
|
11
|
Wehbe N, Slika H, Mesmar J, Nasser SA, Pintus G, Baydoun S, Badran A, Kobeissy F, Eid AH, Baydoun E. The Role of Epac in Cancer Progression. Int J Mol Sci 2020; 21:ijms21186489. [PMID: 32899451 PMCID: PMC7555121 DOI: 10.3390/ijms21186489] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 12/14/2022] Open
Abstract
Cancer continues to be a prime contributor to global mortality. Despite tremendous research efforts and major advances in cancer therapy, much remains to be learned about the underlying molecular mechanisms of this debilitating disease. A better understanding of the key signaling events driving the malignant phenotype of cancer cells may help identify new pharmaco-targets. Cyclic adenosine 3',5'-monophosphate (cAMP) modulates a plethora of biological processes, including those that are characteristic of malignant cells. Over the years, most cAMP-mediated actions were attributed to the activity of its effector protein kinase A (PKA). However, studies have revealed an important role for the exchange protein activated by cAMP (Epac) as another effector mediating the actions of cAMP. In cancer, Epac appears to have a dual role in regulating cellular processes that are essential for carcinogenesis. In addition, the development of Epac modulators offered new routes to further explore the role of this cAMP effector and its downstream pathways in cancer. In this review, the potentials of Epac as an attractive target in the fight against cancer are depicted. Additionally, the role of Epac in cancer progression, namely its effect on cancer cell proliferation, migration/metastasis, and apoptosis, with the possible interaction of reactive oxygen species (ROS) in these phenomena, is discussed with emphasis on the underlying mechanisms and pathways.
Collapse
Affiliation(s)
- Nadine Wehbe
- Department of Biology, American University of Beirut, P.O. Box 11-0236 Beirut, Lebanon; (N.W.); (J.M.)
| | - Hasan Slika
- Department of Pharmacology and Therapeutics, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236 Beirut, Lebanon;
| | - Joelle Mesmar
- Department of Biology, American University of Beirut, P.O. Box 11-0236 Beirut, Lebanon; (N.W.); (J.M.)
| | - Suzanne A. Nasser
- Department of Pharmacology, Beirut Arab University, P.O. Box 11-5020 Beirut, Lebanon;
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sharjah, P.O. Box 27272 Sharjah, UAE;
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43, 07100 Sassari, Italy
| | - Serine Baydoun
- Department of Radiology, American University of Beirut, P.O. Box 11-0236 Beirut, Lebanon;
| | - Adnan Badran
- Department of Basic Sciences, University of Petra, P.O. Box 961343, Amman 11196, Jordan;
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Beirut, Lebanon;
| | - Ali H. Eid
- Department of Pharmacology and Therapeutics, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236 Beirut, Lebanon;
- Department of Pharmacology and Therapeutics, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Beirut, Lebanon
- Correspondence: (A.H.E.); (E.B.); Tel.: +961-1-350-000 (ext. 4891) (A.H.E. & E.B.)
| | - Elias Baydoun
- Department of Biology, American University of Beirut, P.O. Box 11-0236 Beirut, Lebanon; (N.W.); (J.M.)
- Correspondence: (A.H.E.); (E.B.); Tel.: +961-1-350-000 (ext. 4891) (A.H.E. & E.B.)
| |
Collapse
|
12
|
Sivertsen Åsrud K, Pedersen L, Aesoy R, Muwonge H, Aasebø E, Nitschke Pettersen IK, Herfindal L, Dobie R, Jenkins S, Berge RK, Henderson NC, Selheim F, Døskeland SO, Bakke M. Mice depleted for Exchange Proteins Directly Activated by cAMP (Epac) exhibit irregular liver regeneration in response to partial hepatectomy. Sci Rep 2019; 9:13789. [PMID: 31551444 PMCID: PMC6760117 DOI: 10.1038/s41598-019-50219-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 09/06/2019] [Indexed: 02/07/2023] Open
Abstract
The exchange proteins directly activated by cAMP 1 and 2 (Epac1 and Epac2) are expressed in a cell specific manner in the liver, but their biological functions in this tissue are poorly understood. The current study was undertaken to begin to determine the potential roles of Epac1 and Epac2 in liver physiology and disease. Male C57BL/6J mice in which expression of Epac1 and/or Epac2 are deleted, were subjected to partial hepatectomy and the regenerating liver was analyzed with regard to lipid accumulation, cell replication and protein expression. In response to partial hepatectomy, deletion of Epac1 and/or Epac2 led to increased hepatocyte proliferation 36 h post surgery, and the transient steatosis observed in wild type mice was virtually absent in mice lacking both Epac1 and Epac2. The expression of the protein cytochrome P4504a14, which is implicated in hepatic steatosis and fibrosis, was substantially reduced upon deletion of Epac1/2, while a number of factors involved in lipid metabolism were significantly decreased. Moreover, the number of Küpffer cells was affected, and Epac2 expression was increased in the liver of wild type mice in response to partial hepatectomy, further supporting a role for these proteins in liver function. This study establishes hepatic phenotypic abnormalities in mice deleted for Epac1/2 for the first time, and introduces Epac1/2 as regulators of hepatocyte proliferation and lipid accumulation in the regenerative process.
Collapse
Affiliation(s)
| | - Line Pedersen
- Department of Biomedicine, The University of Bergen, Bergen, Norway
| | - Reidun Aesoy
- Department of Clinical Science, The University of Bergen, Bergen, Norway
| | - Haruna Muwonge
- Department of Biomedicine, The University of Bergen, Bergen, Norway
| | - Elise Aasebø
- Department of Clinical Science, The University of Bergen, Bergen, Norway
- Department of Biomedicine, The Proteomic Unit at The University of Bergen (PROBE), University of Bergen, 5009, Bergen, Norway
| | | | - Lars Herfindal
- Department of Clinical Science, The University of Bergen, Bergen, Norway
| | - Ross Dobie
- Centre for Inflammation Research, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Stephen Jenkins
- Centre for Inflammation Research, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Rolf Kristian Berge
- Department of Clinical Science, The University of Bergen, Bergen, Norway
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Neil Cowan Henderson
- Centre for Inflammation Research, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Frode Selheim
- Department of Biomedicine, The University of Bergen, Bergen, Norway
- Department of Clinical Science, The University of Bergen, Bergen, Norway
| | | | - Marit Bakke
- Department of Biomedicine, The University of Bergen, Bergen, Norway
| |
Collapse
|
13
|
Orally active Epac inhibitor reverses mechanical allodynia and loss of intraepidermal nerve fibers in a mouse model of chemotherapy-induced peripheral neuropathy. Pain 2019; 159:884-893. [PMID: 29369966 DOI: 10.1097/j.pain.0000000000001160] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a major side effect of cancer treatment that significantly compromises quality of life of cancer patients and survivors. Identification of targets for pharmacological intervention to prevent or reverse CIPN is needed. We investigated exchange protein regulated by cAMP (Epac) as a potential target. Epacs are cAMP-binding proteins known to play a pivotal role in mechanical allodynia induced by nerve injury and inflammation. We demonstrate that global Epac1-knockout (Epac1-/-) male and female mice are protected against paclitaxel-induced mechanical allodynia. In addition, spinal cord astrocyte activation and intraepidermal nerve fiber (IENF) loss are significantly reduced in Epac1-/- mice as compared to wild-type mice. Moreover, Epac1-/- mice do not develop the paclitaxel-induced deficits in mitochondrial bioenergetics in the sciatic nerve that are a hallmark of CIPN. Notably, mice with cell-specific deletion of Epac1 in Nav1.8-positive neurons (N-Epac1-/-) also show reduced paclitaxel-induced mechanical allodynia, astrocyte activation, and IENF loss, indicating that CIPN develops downstream of Epac1 activation in nociceptors. The Epac-inhibitor ESI-09 reversed established paclitaxel-induced mechanical allodynia in wild-type mice even when dosing started 10 days after completion of paclitaxel treatment. In addition, oral administration of ESI-09 suppressed spinal cord astrocyte activation in the spinal cord and protected against IENF loss. Ex vivo, ESI-09 blocked paclitaxel-induced abnormal spontaneous discharges in dorsal root ganglion neurons. Collectively, these findings implicate Epac1 in nociceptors as a novel target for treatment of CIPN. This is clinically relevant because ESI-09 has the potential to reverse a debilitating and long-lasting side effect of cancer treatment.
Collapse
|
14
|
Santin JR, da Silva GF, Pastor MVD, Broering MF, Nunes R, Braga RC, de Sousa ITS, Stiz DS, da Silva KABS, Stoeberl LC, Corrêa R, Filho VC, Dos Santos CEM, Quintão NLM. Biological and Toxicological Evaluation of N-(4methyl-phenyl)-4-methylphthalimide on Bone Cancer in Mice. Anticancer Agents Med Chem 2019; 19:667-676. [PMID: 30734686 DOI: 10.2174/1871520619666190207130732] [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: 11/26/2018] [Revised: 01/19/2019] [Accepted: 01/26/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND It was recently demonstrated that the phthalimide N-(4-methyl-phenyl)-4- methylphthalimide (MPMPH-1) has important effects against acute and chronic pain in mice, with a mechanism of action correlated to adenylyl cyclase inhibition. Furthermore, it was also demonstrated that phthalimide derivatives presented antiproliferative and anti-tumor effects. Considering the literature data, the present study evaluated the effects of MPMPH-1 on breast cancer bone metastasis and correlated painful symptom, and provided additional toxicological information about the compound and its possible metabolites. METHODS In silico toxicological analysis was supported by in vitro and in vivo experiments to demonstrate the anti-tumor and anti-hypersensitivity effects of the compound. RESULTS The data obtained with the in silico toxicological analysis demonstrated that MPMPH-1 has mutagenic potential, with a low to moderate level of confidence. The mutagenicity potential was in vivo confirmed by micronucleus assay. MPMPH-1 treatments in the breast cancer bone metastasis model were able to prevent the osteoclastic resorption of bone matrix. Regarding cartilage, degradation was considerably reduced within the zoledronic acid group, while in MPMPH-1, chondrocyte multiplication was observed in random areas, suggesting bone regeneration. Additionally, the repeated treatment of mice with MPMPH-1 (10 mg/kg, i.p.), once a day for up to 36 days, significantly reduces the hypersensitivity in animals with breast cancer bone metastasis. CONCLUSION Together, the data herein obtained show that MPMPH-1 is relatively safe, and significantly control the cancer growth, allied to the reduction in bone reabsorption and stimulation of bone and cartilage regeneration. MPMPH-1 effects may be linked, at least in part, to the ability of the compound to interfere with adenylylcyclase pathway activation.
Collapse
Affiliation(s)
- José R Santin
- Postgraduate Program in Pharmaceutical Science, Universidade do Vale do Itajaí, Rua Uruguai, 458, Itajaí/SC, CEP 88302-901, Brazil
| | - Gislaine F da Silva
- Postgraduate Program in Pharmaceutical Science, Universidade do Vale do Itajaí, Rua Uruguai, 458, Itajaí/SC, CEP 88302-901, Brazil
| | - Maria V D Pastor
- Biomedicine Course, Universidade do Vale do Itajaí, Rua Uruguai, 458, Itajaí/SC, CEP 88302-901, Brazil
| | - Milena F Broering
- Postgraduate Program in Pharmaceutical Science, Universidade do Vale do Itajaí, Rua Uruguai, 458, Itajaí/SC, CEP 88302-901, Brazil
| | - Roberta Nunes
- Postgraduate Program in Pharmaceutical Science, Universidade do Vale do Itajaí, Rua Uruguai, 458, Itajaí/SC, CEP 88302-901, Brazil
| | | | | | - Dorimar S Stiz
- Postgraduate Program in Pharmaceutical Science, Universidade do Vale do Itajaí, Rua Uruguai, 458, Itajaí/SC, CEP 88302-901, Brazil
| | - Kathryn A B S da Silva
- Postgraduate Program in Pharmaceutical Science, Universidade do Vale do Itajaí, Rua Uruguai, 458, Itajaí/SC, CEP 88302-901, Brazil
| | - Luis C Stoeberl
- Postgraduate Program in Pharmaceutical Science, Universidade do Vale do Itajaí, Rua Uruguai, 458, Itajaí/SC, CEP 88302-901, Brazil
| | - Rogério Corrêa
- Postgraduate Program in Pharmaceutical Science, Universidade do Vale do Itajaí, Rua Uruguai, 458, Itajaí/SC, CEP 88302-901, Brazil
| | - Valdir C Filho
- Postgraduate Program in Pharmaceutical Science, Universidade do Vale do Itajaí, Rua Uruguai, 458, Itajaí/SC, CEP 88302-901, Brazil
| | | | - Nara L M Quintão
- Postgraduate Program in Pharmaceutical Science, Universidade do Vale do Itajaí, Rua Uruguai, 458, Itajaí/SC, CEP 88302-901, Brazil
| |
Collapse
|
15
|
Exchange Proteins Directly Activated by cAMP and Their Roles in Respiratory Syncytial Virus Infection. J Virol 2018; 92:JVI.01200-18. [PMID: 30185593 DOI: 10.1128/jvi.01200-18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 08/24/2018] [Indexed: 12/28/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of respiratory infection in young children and high-risk adults. However, a specific treatment for this viral infection is not currently available. In this study, we discovered that an exchange protein directly activated by cyclic AMP (EPAC) can serve as a potential therapeutic target for RSV. In both lower and upper epithelial cells, treatment with EPAC inhibitor (ESI-09), but not protein kinase A inhibitor (H89), significantly inhibits RSV replication and proinflammatory cytokine/chemokine induction. In addition, RSV-activated transcriptional factors belonging to the NF-κB and IRF families are also suppressed by ESI-09. Through isoform-specific gene knockdown, we found that EPAC2, but not EPAC1, plays a dominant role in controlling RSV replication and virus-induced host responses. Experiments using both EPAC2 knockout and EPAC2-specific inhibitor support such roles of EPAC2. Therefore, EPAC2 is a promising therapeutic target to regulate RSV replication and associated inflammation.IMPORTANCE RSV is a serious public health problem, as it is associated with bronchiolitis, pneumonia, and asthma exacerbations. Currently no effective treatment or vaccine is available, and many molecular mechanisms regarding RSV-induced lung disease are still significantly unknown. This project aims to elucidate an important and novel function of a protein, called EPAC2, in RSV replication and innate inflammatory responses. Our results should provide an important insight into the development of new pharmacologic strategies against RSV infection, thereby reducing RSV-associated morbidity and mortality.
Collapse
|
16
|
Illiano M, Sapio L, Salzillo A, Capasso L, Caiafa I, Chiosi E, Spina A, Naviglio S. Forskolin improves sensitivity to doxorubicin of triple negative breast cancer cells via Protein Kinase A-mediated ERK1/2 inhibition. Biochem Pharmacol 2018; 152:104-113. [PMID: 29574069 DOI: 10.1016/j.bcp.2018.03.023] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 03/20/2018] [Indexed: 01/07/2023]
Abstract
Triple negative breast cancer (TNBC) is an invasive, metastatic, highly aggressive tumor. Cytotoxic chemotherapy represents the current treatment for TNBC. However, relapse and chemo-resistance are very frequent. Therefore, new therapeutic approaches that are able to increase the sensitivity to cytotoxic drugs are needed. Forskolin, a natural cAMP elevating agent, has been used for several centuries in medicine and its safeness has also been demonstrated in modern studies. Recently, forskolin is emerging as a possible novel molecule for cancer therapy. Here, we investigate the effects of forskolin on the sensitivity of MDA-MB-231 and MDA-MB-468 TNBC cells to doxorubicin through MTT assay, flow cytometry-based assays (cell-cycle progression and cell death), cell number counting and immunoblotting experiments. We demonstrate that forskolin strongly enhances doxorubicin-induced antiproliferative effects by cell death induction. Similar effects are observed with IBMX and isoproterenol cAMP elevating agents and 8-Br-cAMP analog, but not by using 8-pCPT-2'-O-Me-cAMP Epac activator. It is important to note that the forskolin-induced potentiation of sensitivity to doxorubicin is accompanied by a strong inhibition of ERK1/2 phosphorylation, is mimicked by ERK inhibitor PD98059 and is prevented by pre-treatment with Protein Kinase A (PKA) and adenylate cyclase inhibitors. Altogether, our data indicate that forskolin sensitizes TNBC cells to doxorubicin via a mechanism depending on the cAMP/PKA-mediated ERK inhibition. Our findings sustain the evidence of anticancer activity mediated by forskolin and encourage the design of future in-vivo/clinical studies in order to explore forskolin as a doxorubicin sensitizer for possible use in TNBC patients.
Collapse
Affiliation(s)
- Michela Illiano
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "Luigi Vanvitelli", Medical School, Via L. De Crecchio 7, 80138 Naples, Italy.
| | - Luigi Sapio
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "Luigi Vanvitelli", Medical School, Via L. De Crecchio 7, 80138 Naples, Italy.
| | - Alessia Salzillo
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "Luigi Vanvitelli", Medical School, Via L. De Crecchio 7, 80138 Naples, Italy.
| | - Lucia Capasso
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "Luigi Vanvitelli", Medical School, Via L. De Crecchio 7, 80138 Naples, Italy.
| | - Ilaria Caiafa
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "Luigi Vanvitelli", Medical School, Via L. De Crecchio 7, 80138 Naples, Italy.
| | - Emilio Chiosi
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "Luigi Vanvitelli", Medical School, Via L. De Crecchio 7, 80138 Naples, Italy.
| | - Annamaria Spina
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "Luigi Vanvitelli", Medical School, Via L. De Crecchio 7, 80138 Naples, Italy.
| | - Silvio Naviglio
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "Luigi Vanvitelli", Medical School, Via L. De Crecchio 7, 80138 Naples, Italy.
| |
Collapse
|
17
|
Robichaux WG, Cheng X. Intracellular cAMP Sensor EPAC: Physiology, Pathophysiology, and Therapeutics Development. Physiol Rev 2018; 98:919-1053. [PMID: 29537337 PMCID: PMC6050347 DOI: 10.1152/physrev.00025.2017] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/05/2017] [Accepted: 09/06/2017] [Indexed: 12/13/2022] Open
Abstract
This review focuses on one family of the known cAMP receptors, the exchange proteins directly activated by cAMP (EPACs), also known as the cAMP-regulated guanine nucleotide exchange factors (cAMP-GEFs). Although EPAC proteins are fairly new additions to the growing list of cAMP effectors, and relatively "young" in the cAMP discovery timeline, the significance of an EPAC presence in different cell systems is extraordinary. The study of EPACs has considerably expanded the diversity and adaptive nature of cAMP signaling associated with numerous physiological and pathophysiological responses. This review comprehensively covers EPAC protein functions at the molecular, cellular, physiological, and pathophysiological levels; and in turn, the applications of employing EPAC-based biosensors as detection tools for dissecting cAMP signaling and the implications for targeting EPAC proteins for therapeutic development are also discussed.
Collapse
Affiliation(s)
- William G Robichaux
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center , Houston, Texas
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center , Houston, Texas
| |
Collapse
|
18
|
Insights into exchange factor directly activated by cAMP (EPAC) as potential target for cancer treatment. Mol Cell Biochem 2018; 447:77-92. [PMID: 29417338 DOI: 10.1007/s11010-018-3294-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Accepted: 01/19/2018] [Indexed: 01/02/2023]
Abstract
Cancer remains a global health problem and approximately 1.7 million new cancer cases are diagnosed every year worldwide. Although diverse molecules are currently being explored as targets for cancer therapy the tumor treatment and therapy is highly tricky. Secondary messengers are important for hormone-mediated signaling pathway. Cyclic AMP (cAMP), a secondary messenger responsible for various physiological processes regulates cell metabolism by activating Protein kinase A (PKA) and by targeting exchange protein directly activated by cAMP (EPAC). EPAC is present in two isoforms EPAC1 and EPAC2, which exhibit different tissue distribution and is involved in GDP/GTP exchange along with activating Rap1- and Rap2-mediated signaling pathways. EPAC is also known for its dual role in cancer as pro- and anti-proliferative in addition to metastasis. Results after perturbing EPAC activity suggests its involvement in cancer cell migration, proliferation, and cytoskeleton remodeling which makes it a potential therapeutic target for cancer treatments.
Collapse
|
19
|
Jansen SR, Poppinga WJ, de Jager W, Lezoualc'h F, Cheng X, Wieland T, Yarwood SJ, Gosens R, Schmidt M. Epac1 links prostaglandin E2 to β-catenin-dependent transcription during epithelial-to-mesenchymal transition. Oncotarget 2018; 7:46354-46370. [PMID: 27344171 PMCID: PMC5216803 DOI: 10.18632/oncotarget.10128] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 06/02/2016] [Indexed: 01/16/2023] Open
Abstract
In epithelial cells, β-catenin is localized at cell-cell junctions where it stabilizes adherens junctions. When these junctions are disrupted, β-catenin can translocate to the nucleus where it functions as a transcriptional cofactor. Recent research has indicated that PGE2 enhances the nuclear function of β-catenin through cyclic AMP. Here, we aim to study the role of the cyclic AMP effector Epac in β-catenin activation by PGE2 in non-small cell lung carcinoma cells. We show that PGE2 induces a down-regulation of E-cadherin, promotes cell migration and enhances β-catenin translocation to the nucleus. This results in β-catenin-dependent gene transcription. We also observed increased expression of Epac1. Inhibition of Epac1 activity using the CE3F4 compound or Epac1 siRNA abolished the effects of PGE2 on β-catenin. Further, we observed that Epac1 and β-catenin associate together. Expression of an Epac1 mutant with a deletion in the nuclear pore localization sequence prevents this association. Furthermore, the scaffold protein Ezrin was shown to be required to link Epac1 to β-catenin. This study indicates a novel role for Epac1 in PGE2-induced EMT and subsequent activation of β-catenin.
Collapse
Affiliation(s)
- Sepp R Jansen
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy (GRIP), University of Groningen, Groningen, The Netherlands.,Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Wilfred J Poppinga
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy (GRIP), University of Groningen, Groningen, The Netherlands
| | - Wim de Jager
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy (GRIP), University of Groningen, Groningen, The Netherlands
| | - Frank Lezoualc'h
- Inserm UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université Toulouse III, Toulouse, France
| | - Xiaodong Cheng
- Department of Integrative Biology & Pharmacology, Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas, Houston, TX, USA
| | - Thomas Wieland
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Stephen J Yarwood
- School of Life Sciences, Heriot-Watt University, Edinburgh, Scotland
| | - Reinoud Gosens
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy (GRIP), University of Groningen, Groningen, The Netherlands
| | - Martina Schmidt
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy (GRIP), University of Groningen, Groningen, The Netherlands
| |
Collapse
|
20
|
Overexpression of exchange protein directly activated by cAMP-1 (EPAC1) attenuates bladder cancer cell migration. Biochem Biophys Res Commun 2017; 495:64-70. [PMID: 29111327 DOI: 10.1016/j.bbrc.2017.10.142] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 10/26/2017] [Indexed: 12/11/2022]
Abstract
Exchange protein directly activated by cAMP (EPAC) is a mediator of a cAMP signaling pathway that is independent of protein kinase A. EPAC has two isoforms (EPAC1 and EPAC2) and is a cAMP-dependent guanine nucleotide exchange factor for the small GTPases, Rap1 and Rap2. Recent studies suggest that EPAC1 has both positive and negative influences on cancer and is involved in cell proliferation, apoptosis, migration and metastasis. We report that EPAC1 and EPAC2 expression levels were significantly lower in bladder cancer tissue than in normal bladder tissue. In addition, bladder cancer cell lines showed reduced EPAC1 mRNA expression. Furthermore, EPAC1 overexpression in bladder cancer cell lines induced morphologic changes and markedly suppressed cell migration without affecting cell viability. The overexpressed EPAC1 preferentially localized at cell-cell interfaces. In conclusion, reduced EPAC1 expression in bladder tumors and poor migration of EPAC1-overexpressing cells implicate EPAC1 as an inhibitor of bladder cancer cell migration.
Collapse
|
21
|
Liu Z, Zhu Y, Chen H, Wang P, Mei FC, Ye N, Cheng X, Zhou J. Structure-activity relationships of 2-substituted phenyl-N-phenyl-2-oxoacetohydrazonoyl cyanides as novel antagonists of exchange proteins directly activated by cAMP (EPACs). Bioorg Med Chem Lett 2017; 27:5163-5166. [PMID: 29100797 DOI: 10.1016/j.bmcl.2017.10.056] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 10/19/2017] [Accepted: 10/22/2017] [Indexed: 11/24/2022]
Abstract
Exchange proteins directly activated by cAMP (EPACs) are critical cAMP-dependent signaling pathway mediators that play important roles in cancer, diabetes, heart failure, inflammations, infections, neurological disorders and other human diseases. EPAC specific modulators are urgently needed to explore EPAC's physiological function, mechanism of action and therapeutic applications. On the basis of a previously identified EPAC specific inhibitor hit ESI-09, herein we have designed and synthesized a novel series of 2-substituted phenyl-N-phenyl-2-oxoacetohydrazonoyl cyanides as potent EPAC inhibitors. Compound 31 (ZL0524) has been discovered as the most potent EPAC inhibitor with IC50 values of 3.6 µM and 1.2 µM against EPAC1 and EPAC2, respectively. Molecular docking of 31 onto an active EPAC2 structure predicts that 31 occupies the hydrophobic pocket in cAMP binding domain (CBD) and also opens up new space leading to the solvent region. These findings provide inspirations for discovering next generation of EPAC inhibitors.
Collapse
Affiliation(s)
- Zhiqing Liu
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Yingmin Zhu
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The University of Texas Health Science Center, 7000 Fannin St #1200, Houston, TX 77030, United States
| | - Haiying Chen
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Pingyuan Wang
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Fang C Mei
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The University of Texas Health Science Center, 7000 Fannin St #1200, Houston, TX 77030, United States
| | - Na Ye
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The University of Texas Health Science Center, 7000 Fannin St #1200, Houston, TX 77030, United States.
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States.
| |
Collapse
|
22
|
Abstract
Malignant carcinomas are often characterized by metastasis, the movement of carcinoma cells from a primary site to colonize distant organs. For metastasis to occur, carcinoma cells first must adopt a pro-migratory phenotype and move through the surrounding stroma towards a blood or lymphatic vessel. Currently, there are very limited possibilities to target these processes therapeutically. The family of Rho GTPases is an ubiquitously expressed division of GTP-binding proteins involved in the regulation of cytoskeletal dynamics and intracellular signaling. The best characterized members of the Rho family GTPases are RhoA, Rac1 and Cdc42. Abnormalities in Rho GTPase function have major consequences for cancer progression. Rho GTPase activation is driven by cell surface receptors that activate GTP exchange factors (GEFs) and GTPase-activating proteins (GAPs). In this review, we summarize our current knowledge on Rho GTPase function in the regulation of metastasis. We will focus on key discoveries in the regulation of epithelial-mesenchymal-transition (EMT), cell-cell junctions, formation of membrane protrusions, plasticity of cell migration and adaptation to a hypoxic environment. In addition, we will emphasize on crosstalk between Rho GTPase family members and other important oncogenic pathways, such as cyclic AMP-mediated signaling, canonical Wnt/β-catenin, Yes-associated protein (YAP) and hypoxia inducible factor 1α (Hif1α) and provide an overview of the advancements and challenges in developing pharmacological tools to target Rho GTPase and the aforementioned crosstalk in the context of cancer therapeutics.
Collapse
|
23
|
Wang X, Luo C, Cheng X, Lu M. Lithium and an EPAC-specific inhibitor ESI-09 synergistically suppress pancreatic cancer cell proliferation and survival. Acta Biochim Biophys Sin (Shanghai) 2017; 49:573-580. [PMID: 28475672 DOI: 10.1093/abbs/gmx045] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Indexed: 01/03/2023] Open
Abstract
Our previous studies showed that while lithium suppresses proliferation and induces apoptosis in pancreatic cancer cells, the inhibition of exchange proteins directly activated by cyclic adenosine monophosphate (cAMP) (EPAC)1 blocks pancreatic cancer cell migration and invasion. In this study, we further investigated the combinatory effects of lithium and EPAC-specific inhibitor (ESI)-09, an EPAC-specific inhibitor, on pancreatic cancer cell proliferation and viability, and explored whether lithium synergistically cooperates with EPAC inhibition in suppressing pancreatic cancer cell tumorigenicity. The cell viability of pancreatic cancer cell lines PANC-1 and MiaPaCa-2 was measured after 48 h of incubation with different dose combination of lithium and ESI-09. Flow cytometric analysis was carried out to further verify the impact of lithium and ESI-09 upon PANC-1 cell proliferation and apoptosis. To investigate the mechanism that the effects generated by lithium and ESI-09 on PANC-1 cells, the intracellular cAMP level was measured by an ELISA-based cAMP immunoassay. Our data showed that lithium and ESI-09 synergistically inhibit pancreatic cancer cell growth and survival. Furthermore, our results revealed a novel mechanism in which the synergism between lithium and ESI-09 is not mediated by the inhibitory effect of lithium toward GSK3β, but by lithium's ability to suppress cAMP/protein kinase A signaling.
Collapse
Affiliation(s)
- Xinshuo Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Cheng Luo
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Meiling Lu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
24
|
Wang P, Liu Z, Chen H, Ye N, Cheng X, Zhou J. Exchange proteins directly activated by cAMP (EPACs): Emerging therapeutic targets. Bioorg Med Chem Lett 2017; 27:1633-1639. [PMID: 28283242 PMCID: PMC5397994 DOI: 10.1016/j.bmcl.2017.02.065] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/07/2017] [Accepted: 02/26/2017] [Indexed: 11/22/2022]
Abstract
Exchange proteins directly activated by cAMP (EPACs) are critical cAMP-dependent signaling pathway mediators. The discovery of EPAC proteins has significantly facilitated understanding on cAMP-dependent signaling pathway and efforts along this line open new avenues for developing novel therapeutics for cancer, diabetes, heart failure, inflammation, infections, neurological disorders and other human diseases. Over the past decade, important progress has been made in the identification of EPAC agonists, antagonists and their biological and pharmacological applications. In this review, we briefly summarize recently reported novel functions of EPACs and the discovery of their small molecule modulators. The challenges and future perspectives are also discussed.
Collapse
Affiliation(s)
- Pingyuan Wang
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Zhiqing Liu
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Haiying Chen
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Na Ye
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center, Houston, TX 77030, United States
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States.
| |
Collapse
|
25
|
Lewis AE, Aesoy R, Bakke M. Role of EPAC in cAMP-Mediated Actions in Adrenocortical Cells. Front Endocrinol (Lausanne) 2016; 7:63. [PMID: 27379015 PMCID: PMC4904129 DOI: 10.3389/fendo.2016.00063] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 05/30/2016] [Indexed: 12/31/2022] Open
Abstract
Adrenocorticotropic hormone regulates adrenal steroidogenesis mainly via the intracellular signaling molecule cAMP. The effects of cAMP are principally relayed by activating protein kinase A (PKA) and the more recently discovered exchange proteins directly activated by cAMP 1 and 2 (EPAC1 and EPAC2). While the intracellular roles of PKA have been extensively studied in steroidogenic tissues, those of EPACs are only emerging. EPAC1 and EPAC2 are encoded by the genes RAPGEF3 and RAPGEF4, respectively. Whereas EPAC1 is ubiquitously expressed, the expression of EPAC2 is more restricted, and typically found in endocrine tissues. Alternative promoter usage of RAPGEF4 gives rise to three different isoforms of EPAC2 that vary in their N-termini (EPAC2A, EPAC2B, and EPAC2C) and that exhibit distinct expression patterns. EPAC2A is expressed in the brain and pancreas, EPAC2B in steroidogenic cells of the adrenal gland and testis, and EPAC2C has until now only been found in the liver. In this review, we discuss current knowledge on EPAC expression and function with focus on the known roles of EPAC in adrenal gland physiology.
Collapse
Affiliation(s)
- Aurélia E. Lewis
- Department of Molecular Biology, University of Bergen, Bergen, Norway
- *Correspondence: Aurélia E. Lewis,
| | - Reidun Aesoy
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Marit Bakke
- Department of Biomedicine, University of Bergen, Bergen, Norway
| |
Collapse
|