1
|
Brouillette RL, Mona CE, Desgagné M, Hassanzedeh M, Breault É, Lussier F, Belleville K, Longpré JM, Grandbois M, Boudreault PL, Besserer-Offroy É, Sarret P. A lipidated peptide derived from the C-terminal tail of the vasopressin 2 receptor shows promise as a new β-arrestin inhibitor. Pharmacol Res 2025; 212:107597. [PMID: 39800176 DOI: 10.1016/j.phrs.2025.107597] [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: 08/23/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/15/2025]
Abstract
β-arrestins play pivotal roles in seven transmembrane receptor (7TMR) signalling and trafficking. To study their functional role in regulating specific receptor systems, current research relies mainly on genetic tools, as few pharmacological options are available. To address this issue, we designed and synthesised a novel lipidated phosphomimetic peptide inhibitor targeting β-arrestins, called ARIP, which was developed based on the C-terminal tail (A343-S371) of the vasopressin V2 receptor. As the V2R sequence has been shown to bind β-arrestins with high affinity, we added an N-terminal palmitate residue to allow membrane tethering and cell entry. Here, using BRET2-based biosensors, we demonstrated the ability of ARIP to inhibit agonist-induced β-arrestin recruitment on a series of 7TMRs that includes both stable and transient β-arrestin binders, with efficiencies that depend on receptor type. In addition, we showed that ARIP was unable to recruit β-arrestins to the cell membrane by itself, and that it did not interfere with G protein signalling. Molecular modelling studies also revealed that ARIP binds β-arrestins as does V2Rpp, the phosphorylated peptide derived from V2R, and that replacing the p-Ser and p-Thr residues of V2Rpp with Glu residues does not alter ARIP's inhibitory activity on β-arrestin recruitment. Importantly, ARIP exerted an opioid-sparing effect in vivo, as intrathecal injection of ARIP potentiated morphine's analgesic effect in the tail-flick test, consistent with previous findings of genetic inhibition of β-arrestins. ARIP therefore represents a promising pharmacological tool for investigating the fine-tuning roles of β-arrestins in 7TMR-driven pathophysiological processes.
Collapse
Affiliation(s)
- Rebecca L Brouillette
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Christine E Mona
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, UCLA Health, Los Angeles, CA, USA
| | - Michael Desgagné
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Malihe Hassanzedeh
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Émile Breault
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Frédérique Lussier
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Karine Belleville
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Jean-Michel Longpré
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Michel Grandbois
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Pierre-Luc Boudreault
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada; RECITAL International Partnership Lab, Université de Caen-Normandie, Caen, France & Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Élie Besserer-Offroy
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Université de Caen Normandie, INSERM U1086 - Anticipe, Normandie Université, Caen, France; Baclesse Comprehensive Cancer Center, UNICANCER, Caen, France; RECITAL International Partnership Lab, Université de Caen-Normandie, Caen, France & Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Philippe Sarret
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada; RECITAL International Partnership Lab, Université de Caen-Normandie, Caen, France & Université de Sherbrooke, Sherbrooke, QC, Canada.
| |
Collapse
|
2
|
Boileve A, Romito O, Hof T, Levallois A, Brard L, d'Hers S, Fouchet A, Simard C, Guinamard R, Brette F, Sallé L. EPAC1 and 2 inhibit K + currents via PLC/PKC and NOS/PKG pathways in rat ventricular cardiomyocytes. Am J Physiol Cell Physiol 2024; 327:C557-C570. [PMID: 38985989 DOI: 10.1152/ajpcell.00582.2023] [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: 10/31/2023] [Revised: 05/29/2024] [Accepted: 06/19/2024] [Indexed: 07/12/2024]
Abstract
The exchange protein directly activated by cAMP (EPAC) has been implicated in cardiac proarrhythmic signaling pathways including spontaneous diastolic Ca2+ leak from sarcoplasmic reticulum and increased action potential duration (APD) in isolated ventricular cardiomyocytes. The action potential (AP) lengthening following acute EPAC activation is mainly due to a decrease of repolarizing steady-state K+ current (IKSS) but the mechanisms involved remain unknown. This study aimed to assess the role of EPAC1 and EPAC2 in the decrease of IKSS and to investigate the underlying signaling pathways. AP and K+ currents were recorded with the whole cell configuration of the patch-clamp technique in freshly isolated rat ventricular myocytes. EPAC1 and EPAC2 were pharmacologically activated with 8-(4-chlorophenylthio)-2'-O-methyl-cAMP acetoxymethyl ester (8-CPTAM, 10 µmol/L) and inhibited with R-Ce3F4 and ESI-05, respectively. Inhibition of EPAC1 and EPAC2 significantly decreased the effect of 8-CPTAM on APD and IKSS showing that both EPAC isoforms are involved in these effects. Unexpectedly, calmodulin-dependent protein kinase II (CaMKII) inhibition by AIP or KN-93, and Ca2+ chelation by intracellular BAPTA, did not impact the response to 8-CPTAM. However, inhibition of PLC/PKC and nitric oxide synthase (NOS)/PKG pathways partially prevents the 8-CPTAM-dependent decrease of IKSS. Finally, the cumulative inhibition of PKC and PKG blocked the 8-CPTAM effect, suggesting that these two actors work along parallel pathways to regulate IKSS upon EPAC activation. On the basis of such findings, we propose that EPAC1 and EPAC2 are involved in APD lengthening by inhibiting a K+ current via both PLC/PKC and NOS/PKG pathways. This may have pathological implications since EPAC is upregulated in diseases such as cardiac hypertrophy.NEW & NOTEWORHTY Exchange protein directly activated by cAMP (EPAC) proteins modulate ventricular electrophysiology at the cellular level. Both EPAC1 and EPAC2 isoforms participate in this effect. Mechanistically, PLC/PKC and nitric oxide synthase (NO)/PKG pathways are involved in regulating K+ repolarizing current whereas the well-known downstream effector of EPAC, calmodulin-dependent protein kinase II (CaMKII), does not participate. This may have pathological implications since EPAC is upregulated in diseases such as cardiac hypertrophy. Thus, EPAC inhibition may be a new approach to prevent arrhythmias under pathological conditions.
Collapse
Affiliation(s)
- Arthur Boileve
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| | - Olivier Romito
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| | - Thomas Hof
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| | - Aurélia Levallois
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| | - Laura Brard
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| | - Sarah d'Hers
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| | - Alexandre Fouchet
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| | - Christophe Simard
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| | - Romain Guinamard
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| | - Fabien Brette
- PhyMedExp, INSERM U1046, CNRS 9412, Université de Montpellier, Montpellier, France
| | - Laurent Sallé
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| |
Collapse
|
3
|
Yang W, Mei FC, Lin W, White MA, Li L, Li Y, Pan S, Cheng X. Protein SUMOylation promotes cAMP-independent EPAC1 activation. Cell Mol Life Sci 2024; 81:283. [PMID: 38963422 PMCID: PMC11335207 DOI: 10.1007/s00018-024-05315-y] [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: 03/29/2024] [Revised: 05/20/2024] [Accepted: 06/12/2024] [Indexed: 07/05/2024]
Abstract
Protein SUMOylation is a prevalent stress-response posttranslational modification crucial for maintaining cellular homeostasis. Herein, we report that protein SUMOylation modulates cellular signaling mediated by cAMP, an ancient and universal stress-response second messenger. We identify K561 as a primary SUMOylation site in exchange protein directly activated by cAMP (EPAC1) via site-specific mapping of SUMOylation using mass spectrometry. Sequence and site-directed mutagenesis analyses reveal that a functional SUMO-interacting motif in EPAC1 is required for the binding of SUMO-conjugating enzyme UBC9, formation of EPAC1 nuclear condensate, and EPAC1 cellular SUMOylation. Heat shock-induced SUMO modification of EPAC1 promotes Rap1/2 activation in a cAMP-independent manner. Structural modeling and molecular dynamics simulation studies demonstrate that SUMO substituent on K561 of EPAC1 promotes Rap1 interaction by increasing the buried surface area between the SUMOylated receptor and its effector. Our studies identify a functional SUMOylation site in EPAC1 and unveil a novel mechanism in which SUMOylation of EPAC1 leads to its autonomous activation. The findings of SUMOylation-mediated activation of EPAC1 not only provide new insights into our understanding of cellular regulation of EPAC1 but also will open up a new field of experimentation concerning the cross-talk between cAMP/EPAC1 signaling and protein SUMOylation, two major cellular stress response pathways, during cellular homeostasis.
Collapse
Affiliation(s)
- Wenli Yang
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center, Houston, TX, USA
- Texas Therapeutics Institute, 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
- Texas Therapeutics Institute, 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
- Texas Therapeutics Institute, 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
| | - Mark A White
- Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Li Li
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Yue Li
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center, Houston, TX, USA
- Cell Therapy Manufacturing Center, 2130 W Holcombe Blvd, Houston, TX, 77030, USA
| | - Sheng Pan
- 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
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center, Houston, TX, USA.
- Texas Therapeutics Institute, 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
|
4
|
Li Z, Liu Q, Cai Y, Ye N, He Z, Yao Y, Ding Y, Wang P, Qi C, Zheng L, Wang L, Zhou J, Zhang QQ. EPAC inhibitor suppresses angiogenesis and tumor growth of triple-negative breast cancer. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167114. [PMID: 38447883 DOI: 10.1016/j.bbadis.2024.167114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/08/2024]
Abstract
AIMS Exchange protein directly activated by cAMP 1 (EPAC1), a major isoform of guanine nucleotide exchange factors, is highly expressed in vascular endothelia cells and regulates angiogenesis in the retina. High intratumor microvascular densities (MVD) resulting from angiogenesis is responsible for breast cancer development. Downregulation of EPAC1 in tumor cell reduces triple-negative breast cancer (TNBC)-induced angiogenesis. However, whether Epac1 expressed in vascular endothelial cells contributes to angiogenesis and tumor development of TNBC remains elusive. MAIN METHODS We employed NY0123, a previously identified potent EPAC inhibitor, to explore the anti-angiogenic biological role of EPAC1 in vitro and in vivo through vascular endothelial cells, rat aortic ring, Matrigel plug, and chick embryo chorioallantoic membrane (CAM) and yolk sac membrane (YSM) assays, as well as the in vivo xenograft tumor models of TNBC in both chick embryo and mice. KEY FINDINGS Inhibiting EPAC1 in vascular endothelial cells by NY0123 significantly suppresses angiogenesis and tumor growth of TNBC. In addition, NY0123 possesses a better inhibitory efficacy than ESI-09, a reported specific EPAC inhibitor tool compound. Importantly, inhibiting EPAC1 in vascular endothelia cells regulates the typical angiogenic signaling network, which is associated with not only vascular endothelial growth factor (VEGF)/vascular endothelial growth factor receptor-2 (VEGFR2) signaling, but also PI3K/AKT, MEK/ERK and Notch pathway. CONCLUSIONS Our findings support that EPAC1 may serve as an effective anti-angiogenic therapeutic target of TNBC, and EPAC inhibitor NY0123 has the therapeutic potential to be developed for the treatment of TNBC.
Collapse
Affiliation(s)
- Zishuo Li
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Qiao Liu
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yuhao Cai
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Na Ye
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Zinan He
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yuying Yao
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yi Ding
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Pingyuan Wang
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Cuiling Qi
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Lingyun Zheng
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Lijing Wang
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States.
| | - Qian-Qian Zhang
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
5
|
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
|
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
|
Wang F, Yu Z, Zhai S, Li Y, Xu Y, Ye Y, Wei X, Xu J, Xue B. CuO decorated vacancy-rich CeO 2 nanopencils for highly efficient catalytic NO reduction by CO at low temperature. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:31895-31904. [PMID: 36459322 DOI: 10.1007/s11356-022-24508-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 11/27/2022] [Indexed: 06/17/2023]
Abstract
With the rapid development of transportation and vehicles, the elimination of NOx and CO has highly attracted public attention. In this work, vacancy-rich CeO2 nanopencil supported CuO catalysts (CuO/CeO2-NPC) were successfully prepared for NO reduction by CO. Importantly, CeO2 with nanopencil-like shape (CeO2-NPC) have been synthesis by solvothermal method for the first time. The physicochemical properties of all samples were studied in detail by combining the means of X-ray diffraction (XRD), Raman spectroscopy, electron paramagnetic resonance (EPR), X-ray photoelectron spectroscopy (XPS), H2-temperature-programmed reduction (H2-TPR), transmission electron microscopy (TEM), scanning electron microscopy (SEM), N2 physisorption (Brunauer-Emmett-Teller), and NO and CO temperature-programmed desorption (NO-TPD and CO-TPD) techniques. Compared with CeO2 nanorods and nanoparticles supported CuO catalysts (CuO/CeO2-NR and CuO/CeO2-NP), the CuO/CeO2-NPC catalysts showed the highest catalytic activity, affording more than 90% NO conversion at 69 °C as well as excellent H2O tolerance at 150 °C, which is superior to catalysts previously reported. Characterization results indicated that the synergistic effect between the well-dispersed CuO and the CeO2 nanopencil support enables a favorable electron transfer between these components and enhances the density of surface oxygen vacancies and Cu+ species, which consequently accelerating the redox cycle. The results indicated that the morphology control of CeO2 support could be an efficient way to evidently enhance the catalytic performance for NO + CO reaction.
Collapse
Affiliation(s)
- Fei Wang
- Advanced Catalysis and Green Manufacturing Collaborative Innovation Center, School of Petrochemical and Engineering, Changzhou University, Changzhou, 213164, People's Republic of China.
| | - Zairan Yu
- Advanced Catalysis and Green Manufacturing Collaborative Innovation Center, School of Petrochemical and Engineering, Changzhou University, Changzhou, 213164, People's Republic of China
| | - Shuai Zhai
- Advanced Catalysis and Green Manufacturing Collaborative Innovation Center, School of Petrochemical and Engineering, Changzhou University, Changzhou, 213164, People's Republic of China
| | - Yuanyuan Li
- Advanced Catalysis and Green Manufacturing Collaborative Innovation Center, School of Petrochemical and Engineering, Changzhou University, Changzhou, 213164, People's Republic of China
| | - Yang Xu
- Advanced Catalysis and Green Manufacturing Collaborative Innovation Center, School of Petrochemical and Engineering, Changzhou University, Changzhou, 213164, People's Republic of China
| | - Yuyang Ye
- Advanced Catalysis and Green Manufacturing Collaborative Innovation Center, School of Petrochemical and Engineering, Changzhou University, Changzhou, 213164, People's Republic of China
| | - Xuejiao Wei
- School of Chemical Engineering and Materials, Changzhou Institute of Technology, Changzhou, 213032, People's Republic of China
| | - Jie Xu
- Advanced Catalysis and Green Manufacturing Collaborative Innovation Center, School of Petrochemical and Engineering, Changzhou University, Changzhou, 213164, People's Republic of China
| | - Bing Xue
- Advanced Catalysis and Green Manufacturing Collaborative Innovation Center, School of Petrochemical and Engineering, Changzhou University, Changzhou, 213164, People's Republic of China
| |
Collapse
|
8
|
Protein interaction, cytotoxic, transcriptomic and proteomic responses to structurally distinct EPAC1 activators in HUVECs. Sci Rep 2022; 12:16505. [PMID: 36198739 PMCID: PMC9534843 DOI: 10.1038/s41598-022-20607-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/15/2022] [Indexed: 11/15/2022] Open
Abstract
The N-acylsulfonamide derivative, I942, represents the first non-cyclic nucleotide partial agonist of EPAC1. This was soon followed by the identification of the I942 analogues, PW0381, PW0521 and PWO577 and a series of benzofuran oxoacetic acid EPAC1 activators, SY006, SY007 and SY009. Protein interaction, cytotoxicity and EPAC1 activation assays applied here identify PWO577 and SY007 as being effective EPAC1 binders that are well tolerated in HUVECs at concentrations greater than 100 μM and up to 48 h incubation and are effective activators of transfected EPAC1 in U2OS cells. Using RNAseq in HUVECs we show that PWO577 and SY007 regulate approximately 11,000 shared genes, with only few differential gene changes being “off-target”. The genes significantly regulated by both PWO577 and SY007 included a subset of genes normally associated with endothelial activation, including ICAM1, MMP1 and CCL2. Of these, only the expression of MMP1 was markedly increased at the protein level, as determined by LC–MS-based proteomics. Both PWO577 and SY007 suppressed IL-6-induced STAT3 activation and associated downstream gene expression, including inhibition of SOCS3, STAT3, IL6ST and JAK3 genes. Together these results demonstrate the utility of structurally distinct, specific and non-toxic EPAC1 activators. Future modifications will be aimed at eliminating the few noted off-target effects.
Collapse
|
9
|
Pan Y, Liu J, Ren J, Luo Y, Sun X. Epac: A Promising Therapeutic Target for Vascular Diseases: A Review. Front Pharmacol 2022; 13:929152. [PMID: 35910387 PMCID: PMC9330031 DOI: 10.3389/fphar.2022.929152] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Vascular diseases affect the circulatory system and comprise most human diseases. They cause severe symptoms and affect the quality of life of patients. Recently, since their identification, exchange proteins directly activated by cAMP (Epac) have attracted increasing scientific interest, because of their role in cyclic adenosine monophosphate (cAMP) signaling, a well-known signal transduction pathway. The role of Epac in cardiovascular disease and cancer is extensively studied, whereas their role in kidney disease has not been comprehensively explored yet. In this study, we aimed to review recent studies on the regulatory effects of Epac on various vascular diseases, such as cardiovascular disease, cerebrovascular disease, and cancer. Accumulating evidence has shown that both Epac1 and Epac2 play important roles in vascular diseases under both physiological and pathological conditions. Additionally, there has been an increasing focus on Epac pharmacological modulators. Therefore, we speculated that Epac could serve as a novel therapeutic target for the treatment of vascular diseases.
Collapse
Affiliation(s)
- Yunfeng Pan
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Jia Liu
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Jiahui Ren
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Yun Luo
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Xiaobo Sun
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
10
|
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: 63] [Impact Index Per Article: 21.0] [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
|
11
|
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
|
12
|
Martinez Pomier K, Akimoto M, Byun JA, Khamina M, Melacini G. Allosteric Regulation of Cyclic Nucleotide Dependent Protein Kinases. CAN J CHEM 2022. [DOI: 10.1139/cjc-2021-0359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Kinases include a wide variety of valuable drug targets, but full therapeutic exploitation requires a high degree of selectivity. A promising avenue to engineer the desired kinase selectivity relies on allosteric sites. Here we provide a focused minireview of recent progress in allosteric modulation of cyclic nucleotide-dependent kinases, including protein kinases A and G. We show how apparently diverse emerging concepts such as allosteric pluripotency, allosteric non-additive binding and uncompetitive allosteric inhibition are all manifestations of complex conformational ensembles. Such ensembles include not only the typical apo-inactive and effector-bound-active states, but also mixed intermediates that feature attributes of the former states within a single molecule. We also discuss how allosteric responses are amplified by aggregation processes, thus establishing a novel interface between the signaling and amyloid fields. Finally, we critically evaluate the challenges and opportunities for clinical translation opened by these emerging allosteric concepts.
Collapse
Affiliation(s)
| | | | - Jung Ah Byun
- McMaster University, 3710, Hamilton, Ontario, Canada
| | | | | |
Collapse
|
13
|
Tomilin VN, Pyrshev K, Stavniichuk A, Hassanzadeh Khayyat N, Ren G, Zaika O, Khedr S, Staruschenko A, Mei FC, Cheng X, Pochynyuk O. Epac1-/- and Epac2-/- mice exhibit deficient epithelial Na+ channel regulation and impaired urinary Na+ conservation. JCI Insight 2022; 7:e145653. [PMID: 34914636 PMCID: PMC8855822 DOI: 10.1172/jci.insight.145653] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/15/2021] [Indexed: 12/03/2022] Open
Abstract
Exchange proteins directly activated by cAMP (Epacs) are abundantly expressed in the renal tubules. We used genetic and pharmacological tools in combination with balance, electrophysiological, and biochemical approaches to examine the role of Epac1 and Epac2 in renal sodium handling. We demonstrate that Epac1-/- and Epac2-/- mice exhibit a delayed anti-natriuresis to dietary sodium restriction despite augmented aldosterone levels. This was associated with a significantly lower response to the epithelial Na+ channel (ENaC) blocker amiloride, reduced ENaC activity in split-opened collecting ducts, and defective posttranslational processing of α and γENaC subunits in the KO mice fed with a Na+-deficient diet. Concomitant deletion of both isoforms led to a marginally greater natriuresis but further increased aldosterone levels. Epac2 blocker ESI-05 and Epac1&2 blocker ESI-09 decreased ENaC activity in Epac WT mice kept on the Na+-deficient diet but not on the regular diet. ESI-09 injections led to natriuresis in Epac WT mice on the Na+-deficient diet, which was caused by ENaC inhibition. In summary, our results demonstrate similar but nonredundant actions of Epac1 and Epac2 in stimulation of ENaC activity during variations in dietary salt intake. We speculate that inhibition of Epac signaling could be instrumental in treatment of hypertensive states associated with ENaC overactivation.
Collapse
Affiliation(s)
- Viktor N. Tomilin
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Kyrylo Pyrshev
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Anna Stavniichuk
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Naghmeh Hassanzadeh Khayyat
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Guohui Ren
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Oleg Zaika
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Sherif Khedr
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Department of Physiology, Faculty of Medicine, Ain-Shams University, Cairo, Egypt
| | - Alexander Staruschenko
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, USA
| | - Fang C. Mei
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Oleh Pochynyuk
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
14
|
Musheshe N, Oun A, Sabogal-Guáqueta AM, Trombetta-Lima M, Mitchel SC, Adzemovic A, Speek O, Morra F, van der Veen CHJT, Lezoualc’h F, Cheng X, Schmidt M, Dolga AM. Pharmacological Inhibition of Epac1 Averts Ferroptosis Cell Death by Preserving Mitochondrial Integrity. Antioxidants (Basel) 2022; 11:antiox11020314. [PMID: 35204198 PMCID: PMC8868285 DOI: 10.3390/antiox11020314] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 01/27/2023] Open
Abstract
Exchange proteins directly activated by cAMP (Epac) proteins are implicated in a wide range of cellular functions including oxidative stress and cell survival. Mitochondrial-dependent oxidative stress has been associated with progressive neuronal death underlying the pathology of many neurodegenerative diseases. The role of Epac modulation in neuronal cells in relation to cell survival and death, as well as its potential effect on mitochondrial function, is not well established. In immortalized hippocampal (HT-22) neuronal cells, we examined mitochondria function in the presence of various Epac pharmacological modulators in response to oxidative stress due to ferroptosis. Our study revealed that selective pharmacological modulation of Epac1 or Epac2 isoforms, exerted differential effects in erastin-induced ferroptosis conditions in HT-22 cells. Epac1 inhibition prevented cell death and loss of mitochondrial integrity induced by ferroptosis, while Epac2 inhibition had limited effects. Our data suggest Epac1 as a plausible therapeutic target for preventing ferroptosis cell death associated with neurodegenerative diseases.
Collapse
Affiliation(s)
- Nshunge Musheshe
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (A.O.); (A.M.S.-G.); (M.T.-L.); (S.C.M.); (A.A.); (O.S.); (F.M.); (C.H.J.T.v.d.V.); (M.S.)
- Correspondence: (N.M.); (A.M.D.)
| | - Asmaa Oun
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (A.O.); (A.M.S.-G.); (M.T.-L.); (S.C.M.); (A.A.); (O.S.); (F.M.); (C.H.J.T.v.d.V.); (M.S.)
| | - Angélica María Sabogal-Guáqueta
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (A.O.); (A.M.S.-G.); (M.T.-L.); (S.C.M.); (A.A.); (O.S.); (F.M.); (C.H.J.T.v.d.V.); (M.S.)
| | - Marina Trombetta-Lima
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (A.O.); (A.M.S.-G.); (M.T.-L.); (S.C.M.); (A.A.); (O.S.); (F.M.); (C.H.J.T.v.d.V.); (M.S.)
| | - Sarah C. Mitchel
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (A.O.); (A.M.S.-G.); (M.T.-L.); (S.C.M.); (A.A.); (O.S.); (F.M.); (C.H.J.T.v.d.V.); (M.S.)
| | - Ahmed Adzemovic
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (A.O.); (A.M.S.-G.); (M.T.-L.); (S.C.M.); (A.A.); (O.S.); (F.M.); (C.H.J.T.v.d.V.); (M.S.)
| | - Oliver Speek
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (A.O.); (A.M.S.-G.); (M.T.-L.); (S.C.M.); (A.A.); (O.S.); (F.M.); (C.H.J.T.v.d.V.); (M.S.)
| | - Francesca Morra
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (A.O.); (A.M.S.-G.); (M.T.-L.); (S.C.M.); (A.A.); (O.S.); (F.M.); (C.H.J.T.v.d.V.); (M.S.)
| | - Christina H. J. T. van der Veen
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (A.O.); (A.M.S.-G.); (M.T.-L.); (S.C.M.); (A.A.); (O.S.); (F.M.); (C.H.J.T.v.d.V.); (M.S.)
| | - Frank Lezoualc’h
- Inserm UMR-1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université Toulouse Paul Sabatier, 31400 Toulouse, France;
| | - Xiaodong Cheng
- Department of Integrative Biology & Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center at Houston, Houston, TX 7000, USA;
| | - Martina Schmidt
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (A.O.); (A.M.S.-G.); (M.T.-L.); (S.C.M.); (A.A.); (O.S.); (F.M.); (C.H.J.T.v.d.V.); (M.S.)
- Groningen Research Institute of Asthma and COPD (GRIAC), Groningen Research Institute of Pharmacy (GRIP), University Medical Center Groningen (UMCG), University of Groningen, 9713 AV Groningen, The Netherlands
| | - Amalia M. Dolga
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (A.O.); (A.M.S.-G.); (M.T.-L.); (S.C.M.); (A.A.); (O.S.); (F.M.); (C.H.J.T.v.d.V.); (M.S.)
- Correspondence: (N.M.); (A.M.D.)
| |
Collapse
|
15
|
Chu W, Sun X, Zhu X, Zhao YC, Zhang J, Kong Q, Zhou L. Blockade of platelet glycoprotein receptor Ib ameliorates blood-brain barrier disruption following ischemic stroke via Epac pathway. Biomed Pharmacother 2021; 140:111698. [PMID: 34029954 DOI: 10.1016/j.biopha.2021.111698] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 04/04/2021] [Accepted: 04/21/2021] [Indexed: 10/21/2022] Open
Abstract
Glycoprotein (GP) Ib is a platelet membrane receptor complex exposed to vascular injury, proposed as an effective target for stroke therapy. Previously, we have observed that the GPIb antagonist anfibatide (ANF) could mitigate blood-brain barrier (BBB) disruption following cerebral ischemia/reperfusion (CI/R) injury. The current study was designed to investigate whether the amelioration of the BBB by ANF is mediated via the Epac signaling pathway. A murine model of CI/R injury was induced following 90 min of transient middle cerebral artery occlusion (MCAO). ANF (4 μg/kg) was intravenously injected 1 h after reperfusion. Herein, ANF ameliorated BBB disruption, increased the expression of tight junction proteins, suppressed F-actin cytoskeleton rearrangement, decreased the permeability of the ischemic brain tissue, and relieved brain edema. ANF-treated mice had smaller infarct volumes and less severe neurological deficits than the MCAO mice. Moreover, the effects of ANF and Epac1 agonists were very similar in the MCAO mice. Epac activation with a cAMP analog, 8-CPT-2'-O-Me-cAMP, mitigated the breakdown of BBB function and CI/R injury. The Epac specific antagonist, ESI-09, worsened barrier damage and cerebral impairment, antagonizing the protective effects afforded by ANF. In addition, ANF upregulated the expression of Epac1 protein in the ischemic cerebral cortex. Collectively, our results indicate that the protective effect of ANF on the BBB after CI/R could be attributed to the activation of the Epac pathway.
Collapse
Affiliation(s)
- Wei Chu
- Department of Pharmacology, School of Basic Medical Science, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei 230032, PR China; Department of Medical College, Shenzhen Polytechnic, Shenzhen 518055, PR China
| | - Xuemei Sun
- Department of Pharmacology, School of Basic Medical Science, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei 230032, PR China; Department of Medical College, Shenzhen Polytechnic, Shenzhen 518055, PR China
| | - Xiaoxiao Zhu
- Department of Pharmacology, School of Basic Medical Science, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei 230032, PR China; Department of Medical College, Shenzhen Polytechnic, Shenzhen 518055, PR China
| | - Yu Chen Zhao
- Department of Mathematics, University of California, Los Angeles, CA 90095, USA
| | - Jingcheng Zhang
- Department of Pharmacology, School of Basic Medical Science, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei 230032, PR China
| | - Qin Kong
- Department of Pharmacology, School of Basic Medical Science, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei 230032, PR China
| | - Lanlan Zhou
- Department of Pharmacology, School of Basic Medical Science, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei 230032, PR China; Department of Medical College, Shenzhen Polytechnic, Shenzhen 518055, PR China.
| |
Collapse
|
16
|
Guijarro-Belmar A, Domanski DM, Bo X, Shewan D, Huang W. The therapeutic potential of targeting exchange protein directly activated by cyclic adenosine 3',5'-monophosphate (Epac) for central nervous system trauma. Neural Regen Res 2021; 16:460-469. [PMID: 32985466 PMCID: PMC7996029 DOI: 10.4103/1673-5374.293256] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Millions of people worldwide are affected by traumatic spinal cord injury, which usually results in permanent sensorimotor disability. Damage to the spinal cord leads to a series of detrimental events including ischaemia, haemorrhage and neuroinflammation, which over time result in further neural tissue loss. Eventually, at chronic stages of traumatic spinal cord injury, the formation of a glial scar, cystic cavitation and the presence of numerous inhibitory molecules act as physical and chemical barriers to axonal regrowth. This is further hindered by a lack of intrinsic regrowth ability of adult neurons in the central nervous system. The intracellular signalling molecule, cyclic adenosine 3′,5′-monophosphate (cAMP), is known to play many important roles in the central nervous system, and elevating its levels as shown to improve axonal regeneration outcomes following traumatic spinal cord injury in animal models. However, therapies directly targeting cAMP have not found their way into the clinic, as cAMP is ubiquitously present in all cell types and its manipulation may have additional deleterious effects. A downstream effector of cAMP, exchange protein directly activated by cAMP 2 (Epac2), is mainly expressed in the adult central nervous system, and its activation has been shown to mediate the positive effects of cAMP on axonal guidance and regeneration. Recently, using ex vivo modelling of traumatic spinal cord injury, Epac2 activation was found to profoundly modulate the post-lesion environment, such as decreasing the activation of astrocytes and microglia. Pilot data with Epac2 activation also suggested functional improvement assessed by in vivo models of traumatic spinal cord injury. Therefore, targeting Epac2 in traumatic spinal cord injury could represent a novel strategy in traumatic spinal cord injury repair, and future work is needed to fully establish its therapeutic potential.
Collapse
Affiliation(s)
- Alba Guijarro-Belmar
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Aberdeen; Sainsbury Wellcome Centre, University College London, London, UK
| | - Dominik Mateusz Domanski
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Aberdeen, UK
| | - Xuenong Bo
- Center for Neuroscience, Surgery and Trauma, Queen Mary University of London, London, UK
| | - Derryck Shewan
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Aberdeen, UK
| | - Wenlong Huang
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
17
|
Maeda Y, Kikuchi R, Kawagoe J, Tsuji T, Koyama N, Yamaguchi K, Nakamura H, Aoshiba K. Anti-cancer strategy targeting the energy metabolism of tumor cells surviving a low-nutrient acidic microenvironment. Mol Metab 2020; 42:101093. [PMID: 33007425 PMCID: PMC7578269 DOI: 10.1016/j.molmet.2020.101093] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/17/2020] [Accepted: 09/24/2020] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE Tumor cells experience hypoxia, acidosis, and hypoglycemia. Metabolic adaptation to glucose shortage is essential to maintain tumor cells' survival because of their high glucose requirement. This study evaluated the hypothesis that acidosis might promote tumor survival during glucose shortage and if so, explored a novel drug targeting metabolic vulnerability to glucose shortage. METHODS Cell survival and bioenergetics metabolism were assessed in lung cancer cell lines. Our in-house small-molecule compounds were screened to identify those that kill cancer cells under low-glucose conditions. Cytotoxicity against non-cancerous cells was also assessed. Tumor growth was evaluated in vivo using a mouse engraft model. RESULTS Acidosis limited the cellular consumption of glucose and ATP, causing tumor cells to enter a metabolically dormant but energetically economic state, which promoted tumor cell survival during glucose deficiency. We identified ESI-09, a previously known exchange protein directly activated by cAMP (EAPC) inhibitor, as an anti-cancer compound that inhibited cancer cells under low-glucose conditions even when associated with acidosis. Bioenergetic studies showed that independent of EPAC inhibition, ESI-09 was a safer mitochondrial uncoupler than a classical uncoupler and created a futile cycle of mitochondrial respiration, leading to decreased ATP production, increased ATP dissipation, and fuel scavenging. Accordingly, ESI-09 exhibited more cytotoxic effects under low-glucose conditions than under normal glucose conditions. ESI-09 was also more effective than actively proliferating cells on quiescent glucose-restricted cells. Cisplatin showed opposite effects. ESI-09 inhibited tumor growth in lung cancer engraft mice. CONCLUSIONS This study highlights the acidosis-induced promotion of tumor survival during glucose shortage and demonstrates that ESI-09 is a novel potent anti-cancer mitochondrial uncoupler that targets a metabolic vulnerability to glucose shortage even when associated with acidosis. The higher cytotoxicity under lower-than-normal glucose conditions suggests that ESI-09 is safer than conventional chemotherapy, can target the metabolic vulnerability of tumor cells to low-glucose stress, and is applicable to many cancer cell types.
Collapse
Affiliation(s)
- Yuki Maeda
- Department of Respiratory Medicine, Tokyo Medical University Ibaraki Medical Center, 3-20-1 Chuou, Ami-machi, Inashiki-gun, Ibaraki, 300-0395, Japan
| | - Ryota Kikuchi
- Department of Respiratory Medicine, Tokyo Medical University Ibaraki Medical Center, 3-20-1 Chuou, Ami-machi, Inashiki-gun, Ibaraki, 300-0395, Japan; Department of Respiratory Medicine, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Junichiro Kawagoe
- Department of Respiratory Medicine, Tokyo Medical University Ibaraki Medical Center, 3-20-1 Chuou, Ami-machi, Inashiki-gun, Ibaraki, 300-0395, Japan; Department of Respiratory Medicine, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Takao Tsuji
- Department of Medicine, Otsuki Municipal Hospital, 1255 Hanasaki, Otsuki-chou, Otsuki-shi, Yamanashi, 401-0015, Japan
| | - Nobuyuki Koyama
- Department of Clinical Oncology, Tokyo Medical University Ibaraki Medical Center, 3-20-1 Chuou, Ami-machi, Inashiki-gun, Ibaraki, 300-0395, Japan
| | - Kazuhiro Yamaguchi
- Department of Respiratory Medicine, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Hiroyuki Nakamura
- Department of Respiratory Medicine, Tokyo Medical University Ibaraki Medical Center, 3-20-1 Chuou, Ami-machi, Inashiki-gun, Ibaraki, 300-0395, Japan
| | - Kazutetsu Aoshiba
- Department of Respiratory Medicine, Tokyo Medical University Ibaraki Medical Center, 3-20-1 Chuou, Ami-machi, Inashiki-gun, Ibaraki, 300-0395, Japan.
| |
Collapse
|
18
|
Liu W, Ha Y, Xia F, Zhu S, Li Y, Shi S, Mei FC, Merkley K, Vizzeri G, Motamedi M, Cheng X, Liu H, Zhang W. Neuronal Epac1 mediates retinal neurodegeneration in mouse models of ocular hypertension. J Exp Med 2020; 217:133574. [PMID: 31918438 PMCID: PMC7144517 DOI: 10.1084/jem.20190930] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 09/06/2019] [Accepted: 11/26/2019] [Indexed: 12/24/2022] Open
Abstract
Progressive loss of retinal ganglion cells (RGCs) leads to irreversible visual deficits in glaucoma. Here, we found that the level of cyclic AMP and the activity and expression of its mediator Epac1 were increased in retinas of two mouse models of ocular hypertension. Genetic depletion of Epac1 significantly attenuated ocular hypertension–induced detrimental effects in the retina, including vascular inflammation, neuronal apoptosis and necroptosis, thinning of ganglion cell complex layer, RGC loss, and retinal neuronal dysfunction. With bone marrow transplantation and various Epac1 conditional knockout mice, we further demonstrated that Epac1 in retinal neuronal cells (especially RGCs) was responsible for their death. Consistently, pharmacologic inhibition of Epac activity prevented RGC loss. Moreover, in vitro study on primary RGCs showed that Epac1 activation was sufficient to induce RGC death, which was mechanistically mediated by CaMKII activation. Taken together, these findings indicate that neuronal Epac1 plays a critical role in retinal neurodegeneration and suggest that Epac1 could be considered a target for neuroprotection in glaucoma.
Collapse
Affiliation(s)
- Wei Liu
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX.,Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yonju Ha
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX
| | - Fan Xia
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX
| | - Shuang Zhu
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX
| | - Yi Li
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX
| | - Shuizhen Shi
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX
| | - Fang C Mei
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center at Houston, Houston, TX
| | - Kevin Merkley
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX
| | - Gianmarco Vizzeri
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX
| | - Massoud Motamedi
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center at Houston, Houston, TX
| | - Hua Liu
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX
| | - Wenbo Zhang
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX.,Departments of Neuroscience, Cell Biology & Anatomy, University of Texas Medical Branch, Galveston, TX
| |
Collapse
|
19
|
Hoy JJ, Parra NS, Park J, Kuhn S, Iglesias-Bartolome R. Protein kinase A inhibitor proteins (PKIs) divert GPCR-Gαs-cAMP signaling toward EPAC and ERK activation and are involved in tumor growth. FASEB J 2020; 34:13900-13917. [PMID: 32830375 PMCID: PMC7722164 DOI: 10.1096/fj.202001515r] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/30/2020] [Accepted: 08/07/2020] [Indexed: 01/12/2023]
Abstract
The PKA-inhibitor (PKI) family members PKIα, PKIβ, and PKIγ bind with high affinity to PKA and block its kinase activity, modulating the extent, and duration of PKA-mediated signaling events. While PKA is a well-known regulator of physiological and oncogenic events, the role of PKI proteins in these pathways has remained elusive. Here, by measuring activation of the MAPK pathway downstream of GPCR-Gαs-cAMP signaling, we show that the expression levels of PKI proteins can alter the balance of activation of two major cAMP targets: PKA and EPAC. Our results indicate that PKA maintains repressive control over MAPK signaling as well as a negative feedback on cAMP concentration. Overexpression of PKI and its subsequent repression of PKA dysregulates these signaling pathways, resulting in increased intracellular cAMP, and enhanced activation of EPAC and MAPK. We also find that amplifications of PKIA are common in prostate cancer and are associated with reduced progression free survival. Depletion of PKIA in prostate cancer cells leads to reduced migration, increased sensitivity to anoikis and reduced tumor growth. By altering PKA activity PKI can act as a molecular switch, driving GPCR-Gαs-cAMP signaling toward activation of EPAC-RAP1 and MAPK, ultimately modulating tumor growth.
Collapse
Affiliation(s)
- James J. Hoy
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Natalia Salinas Parra
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jeannie Park
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Skyler Kuhn
- Collaborative Bioinformatics Resource, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ramiro Iglesias-Bartolome
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
20
|
Looi CK, Hii LW, Ngai SC, Leong CO, Mai CW. The Role of Ras-Associated Protein 1 (Rap1) in Cancer: Bad Actor or Good Player? Biomedicines 2020; 8:334. [PMID: 32906721 PMCID: PMC7555474 DOI: 10.3390/biomedicines8090334] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/02/2020] [Accepted: 09/05/2020] [Indexed: 02/05/2023] Open
Abstract
Metastasis is known as the most life-threatening event in cancer patients. In principle, the immune system can prevent tumor development. However, dysfunctional T cells may fail to eliminate the tumor cells effectively and provide additional survival advantages for tumor proliferation and metastasis. Constitutive activation of Ras-associated protein1 (Rap1) has not only led to T cell anergy, but also inhibited autophagy and supported cancer progression through various oncogenic events. Inhibition of Rap1 activity with its negative regulator, Rap1GAP, impairs tumor progression. However, active Rap1 reduces tumor invasion in some cancers, indicating that the pleiotropic effects of Rap1 signaling in cancers could be cancer-specific. All in all, targeting Rap1 signaling and its regulators could potentially control carcinogenesis, metastasis, chemoresistance and immune evasion. Rap1GAP could be a promising therapeutic target in combating cancer.
Collapse
Affiliation(s)
- Chin-King Looi
- School of Postgraduate Study, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia; (C.-K.L.); (L.-W.H.)
| | - Ling-Wei Hii
- School of Postgraduate Study, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia; (C.-K.L.); (L.-W.H.)
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia;
| | - Siew Ching Ngai
- School of Biosciences, Faculty of Science and Engineering, University of Nottingham Malaysia, Semenyih, Selangor 43500, Malaysia;
| | - Chee-Onn Leong
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia;
- Centre for Cancer and Stem Cells Research, Institute for Research, Development, and Innovation (IRDI), International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Chun-Wai Mai
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia;
- Centre for Cancer and Stem Cells Research, Institute for Research, Development, and Innovation (IRDI), International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| |
Collapse
|
21
|
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
|
22
|
Boulton S, Van K, VanSchouwen B, Augustine J, Akimoto M, Melacini G. Allosteric Mechanisms of Nonadditive Substituent Contributions to Protein-Ligand Binding. Biophys J 2020; 119:1135-1146. [PMID: 32882185 DOI: 10.1016/j.bpj.2020.07.038] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/30/2020] [Accepted: 07/06/2020] [Indexed: 02/07/2023] Open
Abstract
Quantifying chemical substituent contributions to ligand-binding free energies is challenging due to nonadditive effects. Protein allostery is a frequent cause of nonadditivity, but the underlying allosteric mechanisms often remain elusive. Here, we propose a general NMR-based approach to elucidate such mechanisms and we apply it to the HCN4 ion channel, whose cAMP-binding domain is an archetypal conformational switch. Using NMR, we show that nonadditivity arises not only from concerted conformational transitions, but also from conformer-specific effects, such as steric frustration. Our results explain how affinity-reducing functional groups may lead to affinity gains if combined. Surprisingly, our approach also reveals that nonadditivity depends markedly on the receptor conformation. It is negligible for the inhibited state but highly significant for the active state, opening new opportunities to tune potency and agonism of allosteric effectors.
Collapse
Affiliation(s)
- Stephen Boulton
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
| | - Katherine Van
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
| | - Bryan VanSchouwen
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Canada
| | - Jerry Augustine
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
| | - Madoka Akimoto
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Canada
| | - Giuseppe Melacini
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada; Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Canada.
| |
Collapse
|
23
|
Maity S, Chandanathil M, Millis RM, Connor SA. Norepinephrine stabilizes translation-dependent, homosynaptic long-term potentiation through mechanisms requiring the cAMP sensor Epac, mTOR and MAPK. Eur J Neurosci 2020; 52:3679-3688. [PMID: 32275785 DOI: 10.1111/ejn.14735] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 02/21/2020] [Accepted: 03/22/2020] [Indexed: 02/01/2023]
Abstract
Neuromodulators regulate higher-order cognitive processes including learning and memory through modulation of synaptic transmission and plasticity. Norepinephrine is a neuromodulator that is secreted throughout the brain in response to novelty or increased arousal, which alters neural circuits by increasing the modifiability of CNS synapses. Norepinephrine activates metabotropic receptors, initiating complex intracellular signalling cascades that can promote enduring changes in synaptic strength including long-term potentiation (LTP). In particular, activation of beta-adrenergic receptors (β-ARs) by norepinephrine enhances LTP through downstream engagement of signalling cascades which upregulate protein synthesis at synapses. Here, we sought to determine the select signalling pathways recruited by norepinephrine to promote homosynaptic LTP at hippocampal synapses in mice. Application of norepinephrine initiated a long-lasting form of homosynaptic LTP that requires protein synthesis. Norepinephrine-mediated enhancement of LTP was reduced by inhibition of mammalian target of rapamycin and exchange protein directly activated by cAMP (Epac) but not cAMP-dependent protein kinase A, suggesting that the endogenous β-AR ligand norepinephrine may preferentially recruit Epac signalling to promote enduring changes in synaptic strength. These findings advance our understanding of the mechanisms through which norepinephrine regulates synaptic plasticity associated with formation of new memories.
Collapse
Affiliation(s)
- Sabyasachi Maity
- Department of Physiology, Neuroscience and Behavioral Sciences, St. George's University School of Medicine, St. George, Grenada.,Department of Physiology, University of Alberta, Edmonton, AB, Canada
| | - Merin Chandanathil
- Department of Physiology, American University of Antigua College of Medicine, St. John's, Antigua
| | - Richard M Millis
- Department of Physiology, American University of Antigua College of Medicine, St. John's, Antigua
| | | |
Collapse
|
24
|
Itzhakov D, Nitzan Y, Breitbart H. Protein kinase A inhibition induces EPAC-dependent acrosomal exocytosis in human sperm. Asian J Androl 2020; 21:337-344. [PMID: 30632486 PMCID: PMC6628745 DOI: 10.4103/aja.aja_99_18] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
To interact with the egg, the spermatozoon must undergo several biochemical and motility modifications in the female reproductive tract, collectively called capacitation. Only capacitated sperm can undergo acrosomal exocytosis, near or on the egg, a process that allows the sperm to penetrate and fertilize the egg. In the present study, we investigated the involvement of cyclic adenosine monophosphate (cAMP)-dependent processes on acrosomal exocytosis. Inhibition of protein kinase A (PKA) at the end of capacitation induced acrosomal exocytosis. This process is cAMP-dependent; however, the addition of relatively high concentration of the membrane-permeable 8-bromo-cAMP (8Br-cAMP, 0.1 mmol l−1) analog induced significant inhibition of the acrosomal exocytosis. The induction of acrosomal exocytosis by PKA inhibition was significantly inhibited by an exchange protein directly activated by cAMP (EPAC) ESI09 inhibitor. The EPAC selective substrate activated AE at relatively low concentrations (0.02–0.1 μmol l−1), whereas higher concentrations (>5 μmol l−1) were inhibitory to the AE induced by PKA inhibition. Inhibition of PKA revealed about 50% increase in intracellular cAMP levels, conditions under which EPAC can be activated to induce the AE. Induction of AE by activating the actin severing-protein, gelsolin, which causes F-actin dispersion, was inhibited by the EPAC inhibitor. The AE induced by PKA inhibition was mediated by phospholipase C activity but not by the Ca2+-channel, CatSper. Thus, inhibition of PKA at the end of the capacitation process induced EPAC/phospholipase C-dependent acrosomal exocytosis. EPAC mediates F-actin depolymerization and/or activation of effectors downstream to F-actin breakdown that lead to acrosomal exocytosis.
Collapse
Affiliation(s)
- Diana Itzhakov
- The Mina and Everard Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Yeshayahu Nitzan
- Department of Clinical Laboratory Science, Zefat Academic College, Zefat 1320611, Israel
| | - Haim Breitbart
- The Mina and Everard Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
| |
Collapse
|
25
|
Wang P, Luchowska-Stańska U, van Basten B, Chen H, Liu Z, Wiejak J, Whelan P, Morgan D, Lochhead E, Barker G, Rehmann H, Yarwood SJ, Zhou J. Synthesis and Biochemical Evaluation of Noncyclic Nucleotide Exchange Proteins Directly Activated by cAMP 1 (EPAC1) Regulators. J Med Chem 2020; 63:5159-5184. [PMID: 32340447 DOI: 10.1021/acs.jmedchem.9b02094] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Exchange proteins directly activated by cAMP (EPAC) play a central role in various biological functions, and activation of the EPAC1 protein has shown potential benefits for the treatment of various human diseases. Herein, we report the synthesis and biochemical evaluation of a series of noncyclic nucleotide EPAC1 activators. Several potent EPAC1 binders were identified including 25g, 25q, 25n, 25u, 25e, and 25f, which promote EPAC1 guanine nucleotide exchange factor activity in vitro. These agonists can also activate EPAC1 protein in cells, where they exhibit excellent selectivity toward EPAC over protein kinase A and G protein-coupled receptors. Moreover, 25e, 25f, 25n, and 25u exhibited improved selectivity toward activation of EPAC1 over EPAC2 in cells. Of these, 25u was found to robustly inhibit IL-6-activated signal transducer and activator of transcription 3 (STAT3) and subsequent induction of the pro-inflammatory vascular cell adhesion molecule 1 (VCAM1) cell-adhesion protein. These novel EPAC1 activators may therefore act as useful pharmacological tools for elucidation of EPAC function and promising drug leads for the treatment of relevant human diseases.
Collapse
Affiliation(s)
- Pingyuan Wang
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Urszula Luchowska-Stańska
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Riccarton, Edinburgh EH14 4AS, U.K
| | - Boy van Basten
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Riccarton, Edinburgh EH14 4AS, U.K
| | - Haiying Chen
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Zhiqing Liu
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Jolanta Wiejak
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Riccarton, Edinburgh EH14 4AS, U.K
| | - Padraic Whelan
- Institute of Chemical Sciences, Heriot-Watt University, Riccarton, Edinburgh EH14 4AS, U.K
| | - David Morgan
- Institute of Chemical Sciences, Heriot-Watt University, Riccarton, Edinburgh EH14 4AS, U.K
| | - Emma Lochhead
- Institute of Chemical Sciences, Heriot-Watt University, Riccarton, Edinburgh EH14 4AS, U.K
| | - Graeme Barker
- Institute of Chemical Sciences, Heriot-Watt University, Riccarton, Edinburgh EH14 4AS, U.K
| | - Holger Rehmann
- Department of Molecular Cancer Research, Centre of Biomedical Genetics and Cancer Genomics Centre, University Medical Centre Utrecht, Utrecht 3584 CX, Netherlands
| | - Stephen J Yarwood
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Riccarton, Edinburgh EH14 4AS, U.K
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| |
Collapse
|
26
|
Shao H, Mohamed H, Boulton S, Huang J, Wang P, Chen H, Zhou J, Luchowska-Stańska U, Jentsch NG, Armstrong AL, Magolan J, Yarwood S, Melacini G. Mechanism of Action of an EPAC1-Selective Competitive Partial Agonist. J Med Chem 2020; 63:4762-4775. [PMID: 32297742 DOI: 10.1021/acs.jmedchem.9b02151] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The exchange protein activated by cAMP (EPAC) is a promising drug target for a wide disease range, from neurodegeneration and infections to cancer and cardiovascular conditions. A novel partial agonist of the EPAC isoform 1 (EPAC1), I942, was recently discovered, but its mechanism of action remains poorly understood. Here, we utilize NMR spectroscopy to map the I942-EPAC1 interactions at atomic resolution and propose a mechanism for I942 partial agonism. We found that I942 interacts with the phosphate binding cassette (PBC) and base binding region (BBR) of EPAC1, similar to cyclic adenosine monophosphate (cAMP). These results not only reveal the molecular basis for the I942 vs cAMP mimicry and competition, but also suggest that the partial agonism of I942 arises from its ability to stabilize an inhibition-incompetent activation intermediate distinct from both active and inactive EPAC1 states. The mechanism of action of I942 may facilitate drug design for EPAC-related diseases.
Collapse
Affiliation(s)
| | | | | | | | - Pingyuan Wang
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Haiying Chen
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Urszula Luchowska-Stańska
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh Campus, Edinburgh EH14 4AS, United Kingdom
| | | | | | | | - Stephen Yarwood
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh Campus, Edinburgh EH14 4AS, United Kingdom
| | | |
Collapse
|
27
|
Gray JL, von Delft F, Brennan PE. Targeting the Small GTPase Superfamily through Their Regulatory Proteins. Angew Chem Int Ed Engl 2020; 59:6342-6366. [PMID: 30869179 PMCID: PMC7204875 DOI: 10.1002/anie.201900585] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/11/2019] [Indexed: 12/11/2022]
Abstract
The Ras superfamily of small GTPases are guanine-nucleotide-dependent switches essential for numerous cellular processes. Mutations or dysregulation of these proteins are associated with many diseases, but unsuccessful attempts to target the small GTPases directly have resulted in them being classed as "undruggable". The GTP-dependent signaling of these proteins is controlled by their regulators; guanine nucleotide exchange factors (GEFs), GTPase activating proteins (GAPs), and in the Rho and Rab subfamilies, guanine nucleotide dissociation inhibitors (GDIs). This review covers the recent small molecule and biologics strategies to target the small GTPases through their regulators. It seeks to critically re-evaluate recent chemical biology practice, such as the presence of PAINs motifs and the cell-based readout using compounds that are weakly potent or of unknown specificity. It highlights the vast scope of potential approaches for targeting the small GTPases in the future through their regulatory proteins.
Collapse
Affiliation(s)
- Janine L. Gray
- Structural Genomics ConsortiumUniversity of Oxford, NDMRBOld Road CampusOxfordOX3 7DQUK
- Target Discovery InstituteNuffield Department of MedicineUniversity of OxfordOld Road CampusOxfordOX3 7FZUK
- Diamond Light SourceHarwell Science and Innovation CampusDidcotOX11 0QXUK
| | - Frank von Delft
- Structural Genomics ConsortiumUniversity of Oxford, NDMRBOld Road CampusOxfordOX3 7DQUK
- Diamond Light SourceHarwell Science and Innovation CampusDidcotOX11 0QXUK
- Department of BiochemistryUniversity of JohannesburgAuckland Park2006South Africa
| | - Paul E. Brennan
- Structural Genomics ConsortiumUniversity of Oxford, NDMRBOld Road CampusOxfordOX3 7DQUK
- Target Discovery InstituteNuffield Department of MedicineUniversity of OxfordOld Road CampusOxfordOX3 7FZUK
- Alzheimer's Research (UK) Oxford Drug Discovery InstituteNuffield Department of MedicineUniversity of OxfordOxfordOX3 7FZUK
| |
Collapse
|
28
|
Palano G, Jansson M, Backmark A, Martinsson S, Sabirsh A, Hultenby K, Åkerblad P, Granberg KL, Jennbacken K, Müllers E, Hansson EM. A high-content, in vitro cardiac fibrosis assay for high-throughput, phenotypic identification of compounds with anti-fibrotic activity. J Mol Cell Cardiol 2020; 142:105-117. [PMID: 32277974 DOI: 10.1016/j.yjmcc.2020.04.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 03/31/2020] [Accepted: 04/02/2020] [Indexed: 12/26/2022]
Abstract
A key feature in the pathogenesis of heart failure is cardiac fibrosis, but effective treatments that specifically target cardiac fibrosis are currently not available. A major impediment to progress has been the lack of reliable in vitro models with sufficient throughput to screen for activity against cardiac fibrosis. Here, we established cell culture conditions in micro-well format that support extracellular deposition of mature collagen from primary human cardiac fibroblasts - a hallmark of cardiac fibrosis. Based on robust biochemical characterization we developed a high-content phenotypic screening platform, that allows for high-throughput identification of compounds with activity against cardiac fibrosis. Our platform correctly identifies compounds acting on known cardiac fibrosis pathways. Moreover, it can detect anti-fibrotic activity for compounds acting on targets that have not previously been reported in in vitro cardiac fibrosis assays. Taken together, our experimental approach provides a powerful platform for high-throughput screening of anti-fibrotic compounds as well as discovery of novel targets to develop new therapeutic strategies for heart failure.
Collapse
Affiliation(s)
- G Palano
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine, Karolinska Institutet, Huddinge, Sweden
| | - M Jansson
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - A Backmark
- Discovery Biology, Discovery Sciences, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - S Martinsson
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - A Sabirsh
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - K Hultenby
- Clincal Research Center, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - P Åkerblad
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - K L Granberg
- Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - K Jennbacken
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - E Müllers
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden.
| | - E M Hansson
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine, Karolinska Institutet, Huddinge, Sweden.
| |
Collapse
|
29
|
Silva I, Magalhães-Cardoso MT, Ferreirinha F, Moreira S, Costa AF, Silva D, Vieira C, Silva-Ramos M, Correia-de-Sá P. β 3 Adrenoceptor-induced cholinergic inhibition in human and rat urinary bladders involves the exchange protein directly activated by cyclic AMP 1 favoring adenosine release. Br J Pharmacol 2020; 177:1589-1608. [PMID: 31721163 DOI: 10.1111/bph.14921] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 10/11/2019] [Accepted: 10/21/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE The mechanism by which β3 receptor agonists (e.g. mirabegron) control bladder overactivity may involve adenosine release from human and rat detrusor smooth muscle. Retrograde activation of adenosine A1 receptors reduces ACh release from cholinergic bladder nerves. β3 -Adrenoceptors usually couple to adenylyl cyclase. Here we investigated, which of the cAMP targets, protein kinase A or the exchange protein directly activated by cAMP (EPAC) could be involved in this cholinergic inhibition of the bladder. EXPERIMENTAL APPROACH [3 H]ACh and adenosine release from urothelium-denuded detrusor strips of cadaveric human organ donors and rats were measured by liquid scintillation spectrometry and HPLC, respectively. In vivo cystometry was also performed in urethane-anaesthetized rats. KEY RESULTS The exchange protein directly activated by cAMP (EPAC) inhibitor, ESI-09, prevented mirabegron- and isoprenaline-induced adenosine release from human and rat detrusor strips respectively. ESI-09, but not the PKA inhibitor, H-89, attenuated inhibition of [3 H]ACh release from stimulated (10 Hz) detrusor strips caused by activating β3 -adrenoceptors, AC (forskolin) and EPAC1 (8-CTP-2Me-cAMP). Isoprenaline-induced inhibition of [3 H]ACh release was also prevented by inhibitors of PKC (chelerythrine and Go6976) and of the equilibrative nucleoside transporter 1 (ENT1; dipyridamole and NBTI), but not by PLC inhibition with U73122. Pretreatment with ESI-09, but not with H-89, prevented the reduction of the voiding frequency caused by isoprenaline and forskolin in vivo. CONCLUSION AND IMPLICATIONS Data suggest that β3 -adrenoceptor-induced inhibition of cholinergic neurotransmission in human and rat urinary bladders involves activation of an EPAC1/PKC pathway downstream cAMP production resulting in adenosine outflow via ENT1.
Collapse
Affiliation(s)
- Isabel Silva
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - M Teresa Magalhães-Cardoso
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Fátima Ferreirinha
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Sílvia Moreira
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Ana Filipa Costa
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Diogo Silva
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Cátia Vieira
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Miguel Silva-Ramos
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Serviço de Urologia, Centro Hospitalar Universitário do Porto (CHUP), Porto, Portugal
| | - Paulo Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| |
Collapse
|
30
|
Gray JL, Delft F, Brennan PE. Targeting der kleinen GTPasen über ihre regulatorischen Proteine. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201900585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Janine L. Gray
- Structural Genomics ConsortiumUniversity of Oxford, NDMRB Old Road Campus Oxford OX3 7DQ Großbritannien
- Target Discovery InstituteNuffield Department of MedicineUniversity of Oxford Old Road Campus Oxford OX3 7FZ Großbritannien
- Diamond Light Source Harwell Science and Innovation Campus Didcot OX11 0QX Großbritannien
| | - Frank Delft
- Structural Genomics ConsortiumUniversity of Oxford, NDMRB Old Road Campus Oxford OX3 7DQ Großbritannien
- Diamond Light Source Harwell Science and Innovation Campus Didcot OX11 0QX Großbritannien
- Department of BiochemistryUniversity of Johannesburg Auckland Park 2006 Südafrika
| | - Paul E. Brennan
- Structural Genomics ConsortiumUniversity of Oxford, NDMRB Old Road Campus Oxford OX3 7DQ Großbritannien
- Target Discovery InstituteNuffield Department of MedicineUniversity of Oxford Old Road Campus Oxford OX3 7FZ Großbritannien
- Alzheimer's Research (UK) Oxford Drug Discovery InstituteNuffield Department of MedicineUniversity of Oxford Oxford OX3 7FZ Großbritannien
| |
Collapse
|
31
|
Liu H, Mei FC, Yang W, Wang H, Wong E, Cai J, Toth E, Luo P, Li YM, Zhang W, Cheng X. Epac1 inhibition ameliorates pathological angiogenesis through coordinated activation of Notch and suppression of VEGF signaling. SCIENCE ADVANCES 2020; 6:eaay3566. [PMID: 31911948 PMCID: PMC6938696 DOI: 10.1126/sciadv.aay3566] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 10/29/2019] [Indexed: 05/26/2023]
Abstract
In this study, we investigated the roles of Epac1 in pathological angiogenesis and its potential as a novel therapeutic target for the treatment of vasoproliferative diseases. Genetic deletion of Epac1 ameliorated pathological angiogenesis in mouse models of oxygen-induced retinopathy (OIR) and carotid artery ligation. Moreover, genetic deletion or pharmacological inhibition of Epac1 suppressed microvessel sprouting from ex vivo aortic ring explants. Mechanistic studies revealed that Epac1 acted as a previously unidentified inhibitor of the γ-secretase/Notch signaling pathway via interacting with γ-secretase and regulating its intracellular trafficking while enhancing vascular endothelial growth factor signaling to promote pathological angiogenesis. Pharmacological administration of an Epac-specific inhibitor suppressed OIR-induced neovascularization in wild-type mice, recapitulating the phenotype of genetic Epac1 knockout. Our results demonstrate that Epac1 signaling is critical for the progression of pathological angiogenesis but not for physiological angiogenesis and that the newly developed Epac-specific inhibitors are effective in combating proliferative retinopathy.
Collapse
Affiliation(s)
- Hua Liu
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Fang C. Mei
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, USA
- Texas Therapeutics Institute, University of Texas Health Science Center, Houston, TX, USA
| | - Wenli Yang
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, USA
- Texas Therapeutics Institute, University of Texas Health Science Center, Houston, TX, USA
| | - Hui Wang
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, USA
- Texas Therapeutics Institute, University of Texas Health Science Center, Houston, TX, USA
| | - Eitan Wong
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jingjing Cai
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Emma Toth
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, USA
- Texas Therapeutics Institute, University of Texas Health Science Center, Houston, TX, USA
| | - Pei Luo
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, USA
- Texas Therapeutics Institute, University of Texas Health Science Center, Houston, TX, USA
| | - Yue-Ming Li
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wenbo Zhang
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
- Department of Neuroscience, Cell Biology and Anatomy, University of Texas Medical Branch, Galveston, TX, USA
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, USA
- Texas Therapeutics Institute, University of Texas Health Science Center, Houston, TX, USA
| |
Collapse
|
32
|
The Epac1 Protein: Pharmacological Modulators, Cardiac Signalosome and Pathophysiology. Cells 2019; 8:cells8121543. [PMID: 31795450 PMCID: PMC6953115 DOI: 10.3390/cells8121543] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/22/2019] [Accepted: 11/24/2019] [Indexed: 12/11/2022] Open
Abstract
The second messenger 3′,5′-cyclic adenosine monophosphate (cAMP) is one of the most important signalling molecules in the heart as it regulates many physiological and pathophysiological processes. In addition to the classical protein kinase A (PKA) signalling route, the exchange proteins directly activated by cAMP (Epac) mediate the intracellular functions of cAMP and are now emerging as a new key cAMP effector in cardiac pathophysiology. In this review, we provide a perspective on recent advances in the discovery of new chemical entities targeting the Epac1 isoform and illustrate their use to study the Epac1 signalosome and functional characterisation in cardiac cells. We summarize the role of Epac1 in different subcompartments of the cardiomyocyte and discuss how cAMP–Epac1 specific signalling networks may contribute to the development of cardiac diseases. We also highlight ongoing work on the therapeutic potential of Epac1-selective small molecules for the treatment of cardiac disorders.
Collapse
|
33
|
Ahmed A, Boulton S, Shao H, Akimoto M, Natarajan A, Cheng X, Melacini G. Recent Advances in EPAC-Targeted Therapies: A Biophysical Perspective. Cells 2019; 8:E1462. [PMID: 31752286 PMCID: PMC6912387 DOI: 10.3390/cells8111462] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 02/07/2023] Open
Abstract
The universal second messenger cAMP regulates diverse intracellular processes by interacting with ubiquitously expressed proteins, such as Protein Kinase A (PKA) and the Exchange Protein directly Activated by cAMP (EPAC). EPAC is implicated in multiple pathologies, thus several EPAC-specific inhibitors have been identified in recent years. However, the mechanisms and molecular interactions underlying the EPAC inhibition elicited by such compounds are still poorly understood. Additionally, being hydrophobic low molecular weight species, EPAC-specific inhibitors are prone to forming colloidal aggregates, which result in non-specific aggregation-based inhibition (ABI) in aqueous systems. Here, we review from a biophysical perspective the molecular basis of the specific and non-specific interactions of two EPAC antagonists-CE3F4R, a non-competitive inhibitor, and ESI-09, a competitive inhibitor of EPAC. Additionally, we discuss the value of common ABI attenuators (e.g., TX and HSA) to reduce false positives at the expense of introducing false negatives when screening aggregation-prone compounds. We hope this review provides the EPAC community effective criteria to evaluate similar compounds, aiding in the optimization of existing drug leads, and informing the development of the next generation of EPAC-specific inhibitors.
Collapse
Affiliation(s)
- Alveena Ahmed
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada; (A.A.); (S.B.)
| | - Stephen Boulton
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada; (A.A.); (S.B.)
| | - Hongzhao Shao
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON L8S 4L8, Canada; (H.S.); (M.A.)
| | - Madoka Akimoto
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON L8S 4L8, Canada; (H.S.); (M.A.)
| | - Amarnath Natarajan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Xiaodong Cheng
- Department of Integrative Biology & Pharmacology, University of Texas Health Science Center at Houston, Houston, TX 77030, USA;
- Texas Therapeutics Institute, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Giuseppe Melacini
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada; (A.A.); (S.B.)
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON L8S 4L8, Canada; (H.S.); (M.A.)
| |
Collapse
|
34
|
Luchowska-Stańska U, Morgan D, Yarwood SJ, Barker G. Selective small-molecule EPAC activators. Biochem Soc Trans 2019; 47:1415-1427. [PMID: 31671184 PMCID: PMC6824682 DOI: 10.1042/bst20190254] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/22/2019] [Accepted: 08/27/2019] [Indexed: 02/06/2023]
Abstract
The cellular signalling enzymes, EPAC1 and EPAC2, have emerged as key intracellular sensors of the secondary messenger cyclic 3',5'-adenosine monophosphate (cyclic adenosine monophosphate) alongside protein kinase A. Interest has been galvanised in recent years thanks to the emergence of these species as potential targets for new cardiovascular disease therapies, including vascular inflammation and insulin resistance in vascular endothelial cells. We herein summarise the current state-of-the-art in small-molecule EPAC activity modulators, including cyclic nucleotides, sulphonylureas, and N-acylsulphonamides.
Collapse
Affiliation(s)
- Urszula Luchowska-Stańska
- Institute of Biological Chemistry, Biophysics, and Bioengineering, Heriot-Watt University, Edinburgh EH14 4AS, U.K
| | - David Morgan
- Institute of Chemical Sciences, Heriot-Watt University, Edinburgh EH14 4AS, U.K
| | - Stephen J. Yarwood
- Institute of Biological Chemistry, Biophysics, and Bioengineering, Heriot-Watt University, Edinburgh EH14 4AS, U.K
| | - Graeme Barker
- Institute of Chemical Sciences, Heriot-Watt University, Edinburgh EH14 4AS, U.K
| |
Collapse
|
35
|
Nash CA, Wei W, Irannejad R, Smrcka AV. Golgi localized β1-adrenergic receptors stimulate Golgi PI4P hydrolysis by PLCε to regulate cardiac hypertrophy. eLife 2019; 8:48167. [PMID: 31433293 PMCID: PMC6726460 DOI: 10.7554/elife.48167] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 08/21/2019] [Indexed: 12/25/2022] Open
Abstract
Increased adrenergic tone resulting from cardiovascular stress leads to development of heart failure, in part, through chronic stimulation of β1 adrenergic receptors (βARs) on cardiac myocytes. Blocking these receptors is part of the basis for β-blocker therapy for heart failure. Recent data demonstrate that G protein-coupled receptors (GPCRs), including βARs, are activated intracellularly, although the biological significance is unclear. Here we investigated the functional role of Golgi βARs in rat cardiac myocytes and found they activate Golgi localized, prohypertrophic, phosphoinositide hydrolysis, that is not accessed by cell surface βAR stimulation. This pathway is accessed by the physiological neurotransmitter norepinephrine (NE) via an Oct3 organic cation transporter. Blockade of Oct3 or specific blockade of Golgi resident β1ARs prevents NE dependent cardiac myocyte hypertrophy. This clearly defines a pathway activated by internal GPCRs in a biologically relevant cell type and has implications for development of more efficacious β-blocker therapies.
Collapse
Affiliation(s)
- Craig A Nash
- Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, United States
| | - Wenhui Wei
- Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, United States
| | - Roshanak Irannejad
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| | - Alan V Smrcka
- Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, United States
| |
Collapse
|
36
|
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
|
37
|
Boulton S, Selvaratnam R, Ahmed R, Van K, Cheng X, Melacini G. Mechanisms of Specific versus Nonspecific Interactions of Aggregation-Prone Inhibitors and Attenuators. J Med Chem 2019; 62:5063-5079. [PMID: 31074269 PMCID: PMC7255057 DOI: 10.1021/acs.jmedchem.9b00258] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A common source of false positives in drug discovery is ligand self-association into large colloidal assemblies that nonspecifically inhibit target proteins. However, the mechanisms of aggregation-based inhibition (ABI) and ABI-attenuation by additives, such as Triton X-100 (TX) and human serum albumin (HSA), are not fully understood. Here, we investigate the molecular basis of ABI and ABI-attenuation through the lens of NMR and coupled thermodynamic cycles. We unexpectedly discover a new class of aggregating ligands that exhibit negligible interactions with proteins but act as competitive sinks for the free inhibitor, resulting in bell-shaped dose-response curves. TX attenuates ABI by converting inhibitory, protein-binding aggregates into nonbinding coaggregates, whereas HSA minimizes nonspecific ligand interactions by functioning as a reservoir for free inhibitor and preventing self-association. Hence, both TX and HSA are useful tools to minimize false positives arising from nonspecific binding but at the cost of potentially introducing false negatives due to suppression of specific interactions.
Collapse
Affiliation(s)
- Stephen Boulton
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| | - Rajeevan Selvaratnam
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Department of Laboratory Medicine, University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5G 2C4, Canada
| | - Rashik Ahmed
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| | - Katherine Van
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology and Texas Therapeutics Institute, McGovern Medical School, University of Texas Health Science Center, Houston, Texas 77030, United States
| | - Giuseppe Melacini
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| |
Collapse
|
38
|
Guček A, Gandasi NR, Omar-Hmeadi M, Bakke M, Døskeland SO, Tengholm A, Barg S. Fusion pore regulation by cAMP/Epac2 controls cargo release during insulin exocytosis. eLife 2019; 8:41711. [PMID: 31099751 PMCID: PMC6557626 DOI: 10.7554/elife.41711] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 04/28/2019] [Indexed: 12/20/2022] Open
Abstract
Regulated exocytosis establishes a narrow fusion pore as initial aqueous connection to the extracellular space, through which small transmitter molecules such as ATP can exit. Co-release of polypeptides and hormones like insulin requires further expansion of the pore. There is evidence that pore expansion is regulated and can fail in diabetes and neurodegenerative disease. Here, we report that the cAMP-sensor Epac2 (Rap-GEF4) controls fusion pore behavior by acutely recruiting two pore-restricting proteins, amisyn and dynamin-1, to the exocytosis site in insulin-secreting beta-cells. cAMP elevation restricts and slows fusion pore expansion and peptide release, but not when Epac2 is inactivated pharmacologically or in Epac2-/- (Rapgef4-/-) mice. Consistently, overexpression of Epac2 impedes pore expansion. Widely used antidiabetic drugs (GLP-1 receptor agonists and sulfonylureas) activate this pathway and thereby paradoxically restrict hormone release. We conclude that Epac2/cAMP controls fusion pore expansion and thus the balance of hormone and transmitter release during insulin granule exocytosis. Insulin is the hormone that signals to the body to take up sugar from the blood. Specialized cells in the pancreas – known as β-cells – release insulin after a meal. Before that, insulin molecules are stored in tiny granules inside the β-cells; these granules must fuse with the cells’ surface membranes to release their contents. The first step in this process creates a narrow pore that allows small molecules, but not the larger insulin molecules, to seep out. The pore then widens to release the insulin. Since the small molecules are known to act locally in the pancreas, it is possible that this “molecular sieve” is biologically important. Yet it is not clear how the pore widens. One of the problems for people with type 2 diabetes is that they release less insulin into the bloodstream. Two kinds of drugs used to treat these patients work by stimulating β-cells to release their insulin. One way to achieve this is by raising the levels of a small molecule called cAMP, which is well known to help prepare insulin granules for release. The cAMP molecule also seems to slow the widening of the pore, and Gucek et al. have now investigated how this happens at a molecular level. By observing individual granules of human β-cells using a special microscope, Gucek et al. could watch how different drugs affect pore widening and content release. They also saw that cAMP activated a protein called Epac2, which then recruited two other proteins – amisyn and dynamin – to the small pores. These two proteins together then closed the pore, rather than expanding it to let insulin out. Type 2 diabetes patients sometimes have high levels of amisyn in their β-cells, which could explain why they do not release enough insulin. The microscopy experiments also revealed that two common anti-diabetic drugs activate Epac2 and prevent the pores from widening, thereby counteracting their positive effect on insulin release. The combined effect is likely a shift in the balance between insulin and the locally acting small molecules. These findings suggest that two common anti-diabetic drugs activate a common mechanism that may lead to unexpected outcomes, possibly even reducing how much insulin the β-cells can release. Future studies in mice and humans will have to investigate these effects in whole organisms.
Collapse
Affiliation(s)
- Alenka Guček
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Nikhil R Gandasi
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | | - Marit Bakke
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | - Anders Tengholm
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Sebastian Barg
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
39
|
Griggs RB, Santos DF, Laird DE, Doolen S, Donahue RR, Wessel CR, Fu W, Sinha GP, Wang P, Zhou J, Brings S, Fleming T, Nawroth PP, Susuki K, Taylor BK. Methylglyoxal and a spinal TRPA1-AC1-Epac cascade facilitate pain in the db/db mouse model of type 2 diabetes. Neurobiol Dis 2019; 127:76-86. [PMID: 30807826 DOI: 10.1016/j.nbd.2019.02.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 02/21/2019] [Indexed: 12/21/2022] Open
Abstract
Painful diabetic neuropathy (PDN) is a devastating neurological complication of diabetes. Methylglyoxal (MG) is a reactive metabolite whose elevation in the plasma corresponds to PDN in patients and pain-like behavior in rodent models of type 1 and type 2 diabetes. Here, we addressed the MG-related spinal mechanisms of PDN in type 2 diabetes using db/db mice, an established model of type 2 diabetes, and intrathecal injection of MG in conventional C57BL/6J mice. Administration of either a MG scavenger (GERP10) or a vector overexpressing glyoxalase 1, the catabolic enzyme for MG, attenuated heat hypersensitivity in db/db mice. In C57BL/6J mice, intrathecal administration of MG produced signs of both evoked (heat and mechanical hypersensitivity) and affective (conditioned place avoidance) pain. MG-induced Ca2+ mobilization in lamina II dorsal horn neurons of C57BL/6J mice was exacerbated in db/db, suggestive of MG-evoked central sensitization. Pharmacological and/or genetic inhibition of transient receptor potential ankyrin subtype 1 (TRPA1), adenylyl cyclase type 1 (AC1), protein kinase A (PKA), or exchange protein directly activated by cyclic adenosine monophosphate (Epac) blocked MG-evoked hypersensitivity in C57BL/6J mice. Similarly, intrathecal administration of GERP10, or inhibitors of TRPA1 (HC030031), AC1 (NB001), or Epac (HJC-0197) attenuated hypersensitivity in db/db mice. We conclude that MG and sensitization of a spinal TRPA1-AC1-Epac signaling cascade facilitate PDN in db/db mice. Our results warrant clinical investigation of MG scavengers, glyoxalase inducers, and spinally-directed pharmacological inhibitors of a MG-TRPA1-AC1-Epac pathway for the treatment of PDN in type 2 diabetes.
Collapse
Affiliation(s)
- Ryan B Griggs
- Department of Physiology and Center for Analgesia Research Excellence, College of Medicine, University of Kentucky Medical Center, Lexington, KY, United States of America; Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States of America.
| | - Diogo F Santos
- Department of Physiology and Center for Analgesia Research Excellence, College of Medicine, University of Kentucky Medical Center, Lexington, KY, United States of America
| | - Don E Laird
- Department of Physiology and Center for Analgesia Research Excellence, College of Medicine, University of Kentucky Medical Center, Lexington, KY, United States of America
| | - Suzanne Doolen
- Department of Physiology and Center for Analgesia Research Excellence, College of Medicine, University of Kentucky Medical Center, Lexington, KY, United States of America
| | - Renee R Donahue
- Department of Physiology and Center for Analgesia Research Excellence, College of Medicine, University of Kentucky Medical Center, Lexington, KY, United States of America
| | - Caitlin R Wessel
- Department of Physiology and Center for Analgesia Research Excellence, College of Medicine, University of Kentucky Medical Center, Lexington, KY, United States of America
| | - Weisi Fu
- Department of Physiology and Center for Analgesia Research Excellence, College of Medicine, University of Kentucky Medical Center, Lexington, KY, United States of America
| | - Ghanshyam P Sinha
- Department of Physiology and Center for Analgesia Research Excellence, College of Medicine, University of Kentucky Medical Center, Lexington, KY, United States of America
| | - Pingyuan Wang
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Jia Zhou
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Sebastian Brings
- Department of Nuclear Medicine, University Hospital of Heidelberg, INF 400 Heidelberg, Germany; Department of Medicine and Clinical Chemistry, University Hospital of Heidelberg, INF 410 Heidelberg, Germany
| | - Thomas Fleming
- Department of Medicine and Clinical Chemistry, University Hospital of Heidelberg, INF 410 Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Peter P Nawroth
- Department of Medicine and Clinical Chemistry, University Hospital of Heidelberg, INF 410 Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Helmholtz Zentrum München, Neuherberg, Germany
| | - Keiichiro Susuki
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States of America
| | - Bradley K Taylor
- Department of Physiology and Center for Analgesia Research Excellence, College of Medicine, University of Kentucky Medical Center, Lexington, KY, United States of America; Department of Anesthesiology, University of Pittsburgh, Pittsburgh, PA, United States of America.
| |
Collapse
|
40
|
Ramos-Alvarez I, Lee L, Jensen RT. Cyclic AMP-dependent protein kinase A and EPAC mediate VIP and secretin stimulation of PAK4 and activation of Na +,K +-ATPase in pancreatic acinar cells. Am J Physiol Gastrointest Liver Physiol 2019; 316:G263-G277. [PMID: 30520694 PMCID: PMC6397337 DOI: 10.1152/ajpgi.00275.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 11/20/2018] [Accepted: 11/20/2018] [Indexed: 01/31/2023]
Abstract
Rat pancreatic acinar cells possess only the p21-activated kinase (PAKs), PAK4 of the group II PAK, and it is activated by gastrointestinal hormones/neurotransmitters stimulating PLC and by a number of growth factors. However, little is known generally of cAMP agents causing PAK4 activation, and there are no studies with gastrointestinal hormones/neurotransmitters activating cAMP cascades. In the present study, we examined the ability of VIP and secretin, which stimulate cAMP generation in pancreatic acini, to stimulate PAK4 activation, the signaling cascades involved, and their possible role in activating sodium-potassium adenosine triphosphatase (Na+,K+-ATPase). PAK4 activation was compared with activation of the well-established cAMP target, cyclic AMP response element binding protein (CREB). Secretin-stimulated PAK4 activation was inhibited by KT-5720 and PKA Type II inhibitor (PKI), protein kinase A (PKA) inhibitors, whereas VIP activation was inhibited by ESI-09 and HJC0197, exchange protein directly activated by cAMP (EPAC) inhibitors. In contrast, both VIP/secretin-stimulated phosphorylation of CREB (pCREB) via EPAC activation; however, it was inhibited by the p44/42 inhibitor PD98059 and the p38 inhibitor SB202190. The specific EPAC agonist 8-CPT-2- O-Me-cAMP as well 8-Br-cAMP and forskolin stimulated PAK4 activation. Secretin/VIP activation of Na+,K+-ATPase, was inhibited by PAK4 inhibitors (PF-3758309, LCH-7749944). These results demonstrate PAK4 is activated in pancreatic acini by stimulation of both VIP-/secretin-preferring receptors, as is CREB. However, they differ in their signaling cascades. Furthermore, PAK4 activation is needed for Na+,K+ATPase activation, which mediates pancreatic fluid secretion. These results, coupled with recent studies reporting PAKs are involved in both pancreatitis/pancreatic cancer growth/enzyme secretion, show that PAK4, similar to PAK2, likely plays an important role in both pancreatic physiological/pathological responses. NEW & NOTEWORTHY Pancreatic acini possess only the group II p21-activated kinase, PAK4, which is activated by PLC-stimulating agents/growth factors and is important in enzyme-secretion/growth/pancreatitis. Little information exists on cAMP-activating agents stimulating group II PAKs. We studied ability/effect of cyclic AMP-stimulating agents [vasoactive intestinal polypeptide (VIP), secretin] on PAK4 activity in rat pancreatic-acini. Both VIP/secretin activated PAK4/CREB, but the cAMP signaling cascades differed for EPAC, MAPK, and PKA pathways. Both hormones require PAK4 activation to stimulate sodium-potassium adenosine triphosphatase activity. This study shows PAK4 plays an important role in VIP-/secretin-stimulated pancreatic fluid secretion and suggests it plays important roles in pancreatic acinar physiological/pathophysiological responses mediated by cAMP-activating agents.
Collapse
Affiliation(s)
- Irene Ramos-Alvarez
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health , Bethesda, Maryland
| | - Lingaku Lee
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health , Bethesda, Maryland
| | - R T Jensen
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health , Bethesda, Maryland
| |
Collapse
|
41
|
Cherezova A, Tomilin V, Buncha V, Zaika O, Ortiz PA, Mei F, Cheng X, Mamenko M, Pochynyuk O. Urinary concentrating defect in mice lacking Epac1 or Epac2. FASEB J 2019; 33:2156-2170. [PMID: 30252533 PMCID: PMC6338637 DOI: 10.1096/fj.201800435r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 08/27/2018] [Indexed: 11/11/2022]
Abstract
cAMP is a universal second messenger regulating a plethora of processes in the kidney. Two downstream effectors of cAMP are PKA and exchange protein directly activated by cAMP (Epac), which, unlike PKA, is often linked to elevation of [Ca2+]i. While both Epac isoforms (Epac1 and Epac2) are expressed along the nephron, their relevance in the kidney remains obscure. We combined ratiometric calcium imaging with quantitative immunoblotting, immunofluorescent confocal microscopy, and balance studies in mice lacking Epac1 or Epac2 to determine the role of Epac in renal water-solute handling. Epac1-/- and Epac2-/- mice developed polyuria despite elevated arginine vasopressin levels. We did not detect major deficiencies in arginine vasopressin [Ca2+]i signaling in split-opened collecting ducts or decreases in aquaporin water channel type 2 levels. Instead, sodium-hydrogen exchanger type 3 levels in the proximal tubule were dramatically reduced in Epac1-/- and Epac2-/- mice. Water deprivation revealed persisting polyuria, impaired urinary concentration ability, and augmented urinary excretion of Na+ and urea in both mutant mice. In summary, we report a nonredundant contribution of Epac isoforms to renal function. Deletion of Epac1 and Epac2 decreases sodium-hydrogen exchanger type 3 expression in the proximal tubule, leading to polyuria and osmotic diuresis.-Cherezova, A., Tomilin, V., Buncha, V., Zaika, O., Ortiz, P. A., Mei, F., Cheng, X., Mamenko, M., Pochynyuk, O. Urinary concentrating defect in mice lacking Epac1 or Epac2.
Collapse
Affiliation(s)
- Alena Cherezova
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Viktor Tomilin
- Department of Integrative Biology and Pharmacology The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Vadym Buncha
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Oleg Zaika
- Department of Integrative Biology and Pharmacology The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Pablo A. Ortiz
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA; and
| | - Fang Mei
- Department of Integrative Biology and Pharmacology The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology The University of Texas Health Science Center at Houston, Houston, Texas, USA
- Texas Therapeutics Institute, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Mykola Mamenko
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Oleh Pochynyuk
- Department of Integrative Biology and Pharmacology The University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
42
|
Brouwer S, Hoffmeister T, Gresch A, Schönhoff L, Düfer M. Resveratrol Influences Pancreatic Islets by Opposing Effects on Electrical Activity and Insulin Release. Mol Nutr Food Res 2019; 62. [PMID: 29341416 DOI: 10.1002/mnfr.201700902] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 12/19/2017] [Indexed: 01/27/2023]
Abstract
SCOPE Resveratrol is suggested to improve glycemic control by activation of sirtuin 1 (SIRT1) and has already been tested clinically. Our investigation characterizes the targets of resveratrol in pancreatic beta cells and their contribution to short- and long-term effects on insulin secretion. METHODS AND RESULTS Islets or beta cells are isolated from C57BL/6N mice. Electrophysiology is performed with microelectrode arrays and patch-clamp technique, insulin secretion and content are determined by radioimmunoassay, cAMP is measured by enzyme-linked immunosorbent assay, and cytosolic Ca2+ concentration by fluorescence methods. Resveratrol (25 μmol L-1 ) elevates [Ca2+ ]c and potentiates glucose-stimulated insulin secretion. These effects are associated with increased intracellular cAMP and are sensitive to the SIRT1 blocker Ex-527. Inhibition of EPAC1 by CE3F4 also abolishes the stimulatory effect of resveratrol. The underlying mechanism does not involve membrane depolarization as resveratrol even reduces electrical activity despite blocking KATP channels. Importantly, after prolonged exposure to resveratrol (14 days), the beneficial influence of the polyphenol on insulin release is lost. CONCLUSION Resveratrol addresses multiple targets in pancreatic islets. Potentiation of insulin secretion is mediated by SIRT1-dependent activation of cAMP/EPAC1. Considering resveratrol as therapeutic supplement for patients with type 2 diabetes mellitus, the inhibitory influence on electrical excitability attenuates positive effects.
Collapse
Affiliation(s)
- Simone Brouwer
- University of Münster, Pharmaceutical and Medicinal Chemistry, Dept. of Pharmacology, Münster, Germany
| | - Theresa Hoffmeister
- University of Münster, Pharmaceutical and Medicinal Chemistry, Dept. of Pharmacology, Münster, Germany
| | - Anne Gresch
- University of Münster, Pharmaceutical and Medicinal Chemistry, Dept. of Pharmacology, Münster, Germany
| | - Lisa Schönhoff
- University of Münster, Pharmaceutical and Medicinal Chemistry, Dept. of Pharmacology, Münster, Germany
| | - Martina Düfer
- University of Münster, Pharmaceutical and Medicinal Chemistry, Dept. of Pharmacology, Münster, Germany
| |
Collapse
|
43
|
Llavero F, Luque Montoro M, Arrazola Sastre A, Fernández-Moreno D, Lacerda HM, Parada LA, Lucia A, Zugaza JL. Epidermal growth factor receptor controls glycogen phosphorylase in T cells through small GTPases of the RAS family. J Biol Chem 2019; 294:4345-4358. [PMID: 30647127 DOI: 10.1074/jbc.ra118.005997] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/07/2019] [Indexed: 12/31/2022] Open
Abstract
We recently uncovered a regulatory pathway of the muscle isoform of glycogen phosphorylase (PYGM) that plays an important role in regulating immune function in T cells. Here, using various enzymatic, pulldown, and immunoprecipitation assays, we describe signaling cross-talk between the small GTPases RAS and RAP1A, member of RAS oncogene family (RAP1) in human Kit 225 lymphoid cells, which, in turn, is regulated by the epidermal growth factor receptor (EGFR). We found that this communication bridge is essential for glycogen phosphorylase (PYG) activation through the canonical pathway because this enzyme is inactive in the absence of adenylyl cyclase type 6 (ADCY6). PYG activation required stimulation of both exchange protein directly activated by cAMP 2 (EPAC2) and RAP1 via RAS and ADCY6 phosphorylation, with the latter being mediated by Raf-1 proto-oncogene, Ser/Thr kinase (RAF1). Consistent with this model, PYG activation was EGFR-dependent and may be initiated by the constitutively active form of RAS. Consequently, PYG activation in Kit 225 T cells could be blocked with specific inhibitors of RAS, EPAC, RAP1, RAF1, ADCY6, and cAMP-dependent protein kinase. Our results establish a new paradigm for the mechanism of PYG activation, which depends on the type of receptor involved.
Collapse
Affiliation(s)
- Francisco Llavero
- From the Achucarro Basque Center for Neuroscience, Science Park of the Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 48940 Leioa, Spain,
| | - Miriam Luque Montoro
- From the Achucarro Basque Center for Neuroscience, Science Park of the Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 48940 Leioa, Spain
| | - Alazne Arrazola Sastre
- From the Achucarro Basque Center for Neuroscience, Science Park of the Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 48940 Leioa, Spain.,the Department of Genetics, Physical Anthropology, and Animal Physiology, Faculty of Science and Technology, UPV/EHU, 48940 Leioa, Spain
| | - David Fernández-Moreno
- the Research Institute of the Hospital 12 de Octubre ("i+12"), 28041 Madrid, Spain.,the Faculty of Sports Science, Universidad Europea de Madrid, 28670 Madrid, Spain
| | | | - Luis A Parada
- the Instituto de Patología Experimental, Universidad Nacional de Salta, A4400 Salta, Argentina, and
| | - Alejandro Lucia
- the Research Institute of the Hospital 12 de Octubre ("i+12"), 28041 Madrid, Spain.,the Faculty of Sports Science, Universidad Europea de Madrid, 28670 Madrid, Spain
| | - José L Zugaza
- From the Achucarro Basque Center for Neuroscience, Science Park of the Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 48940 Leioa, Spain, .,the Department of Genetics, Physical Anthropology, and Animal Physiology, Faculty of Science and Technology, UPV/EHU, 48940 Leioa, Spain.,IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain
| |
Collapse
|
44
|
Curcumin pretreatment protects against hypoxia/reoxgenation injury via improvement of mitochondrial function, destabilization of HIF-1α and activation of Epac1-Akt pathway in rat bone marrow mesenchymal stem cells. Biomed Pharmacother 2018; 109:1268-1275. [PMID: 30551377 DOI: 10.1016/j.biopha.2018.11.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/01/2018] [Accepted: 11/01/2018] [Indexed: 11/22/2022] Open
Abstract
Bone marrow mesenchymal stem cells (BMSCs) possess promising therapeutic effects and have been considered as a highly desirable agent for tissue injury treatment. However, little survived cells after transplanting due to severe relocated conditions (characterized by prolonged hypoxia and oxidative stress) lead to hampered benefits of BMSCs-based cell therapy. Curcumin, a natural dietary product, has attracted increasing attention owing to its profound pharmacologic properties. Here, we report the protective effects of curcumin pretreatment in BMSCs against hypoxia and reoxygenation (H/R) triggered injury, which mimick ischemia/reperfusion in vivo. We found that curcumin pretreatment remarkably inhibited H/R-induced cell viability loss, cell nuclei condensation, LDH leakage, as well as caspase-3 activity increase in BMSCs. Furthermore, curcumin pretreatment prevented H/R-induced mitochondrial dysfunction through expediting adenosine triphosphate production and suppressing reactive oxygen species accumulation and mitochondrial membrane potential decline. In addition, curcumin pretreatment notably induced HIF-1α destabilization, Epac1 and Akt activation, and Erk1/2 and p38 deactivation. However, Epac1 inhibitor ESI-09 obviously restrained the increase of p-Akt induced by curcumin, but not p-Erk1/2 or p-p38, and abrogated the protective effect of curcumin on BMSCs' survival and arrested cell cycle in G0/G1 phase. Taken together, these results demonstrated that curcumin pretreatment conferred BMSCs the ability to survive from H/R injury, which might attribute to its protection on mitochondrial function, destabilization of HIF-1α and activation of Epac1-Akt signaling pathway. Thus, this study provides more pharmacologic aspects of curcumin, and suggests that pre-conditioning of BMSCs with curcumin could serve as an attractive approach for facilitating cell therapy in tissue repair treatment.
Collapse
|
45
|
Zhou D, Ota K, Nardin C, Feldman M, Widman A, Wind O, Simon A, Reilly M, Levin LR, Buck J, Wakamatsu K, Ito S, Zippin JH. Mammalian pigmentation is regulated by a distinct cAMP-dependent mechanism that controls melanosome pH. Sci Signal 2018; 11:11/555/eaau7987. [PMID: 30401788 DOI: 10.1126/scisignal.aau7987] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The production of melanin increases skin pigmentation and reduces the risk of skin cancer. Melanin production depends on the pH of melanosomes, which are more acidic in lighter-skinned than in darker-skinned people. We showed that inhibition of soluble adenylyl cyclase (sAC) controlled pigmentation by increasing the pH of melanosomes both in cells and in vivo. Distinct from the canonical melanocortin 1 receptor (MC1R)-dependent cAMP pathway that controls pigmentation by altering gene expression, we found that inhibition of sAC increased pigmentation by increasing the activity of tyrosinase, the rate-limiting enzyme in melanin synthesis, which is more active at basic pH. We demonstrated that the effect of sAC activity on pH and melanin production in human melanocytes depended on the skin color of the donor. Last, we identified sAC inhibitors as a new class of drugs that increase melanosome pH and pigmentation in vivo, suggesting that pharmacologic inhibition of this pathway may affect skin cancer risk or pigmentation conditions.
Collapse
Affiliation(s)
- Dalee Zhou
- Department of Dermatology, Weill Cornell Medical College, New York, NY 10021, USA
| | - Koji Ota
- Department of Dermatology, Weill Cornell Medical College, New York, NY 10021, USA
| | - Charlee Nardin
- Department of Dermatology, Weill Cornell Medical College, New York, NY 10021, USA.,Service de Dermatologie, Centre Hospitalier Universitaire, Besançon 25030, France
| | - Michelle Feldman
- Department of Dermatology, Weill Cornell Medical College, New York, NY 10021, USA
| | - Adam Widman
- Department of Dermatology, Weill Cornell Medical College, New York, NY 10021, USA
| | - Olivia Wind
- Department of Dermatology, Weill Cornell Medical College, New York, NY 10021, USA
| | - Amanda Simon
- Department of Dermatology, Weill Cornell Medical College, New York, NY 10021, USA
| | - Michael Reilly
- Department of Dermatology, Weill Cornell Medical College, New York, NY 10021, USA
| | - Lonny R Levin
- Department of Pharmacology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Jochen Buck
- Department of Pharmacology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Kazumasa Wakamatsu
- Department of Chemistry, Fujita Health University School of Health Sciences, Toyoake 470-1192, Japan
| | - Shosuke Ito
- Department of Chemistry, Fujita Health University School of Health Sciences, Toyoake 470-1192, Japan
| | - Jonathan H Zippin
- Department of Dermatology, Weill Cornell Medical College, New York, NY 10021, USA.
| |
Collapse
|
46
|
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
|
47
|
Wang XF, Song SD, Li YJ, Hu ZQ, Zhang ZW, Yan CG, Li ZG, Tang HF. Protective Effect of Quercetin in LPS-Induced Murine Acute Lung Injury Mediated by cAMP-Epac Pathway. Inflammation 2018; 41:1093-1103. [PMID: 29569077 DOI: 10.1007/s10753-018-0761-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Quercetin (Que) as an abundant flavonol element possesses potent antioxidative properties and has protective effect in lipopolysaccharide (LPS)-induced acute lung injury (ALI), but the specific mechanism is still unclear, so we investigated the effect of Que from in vivo and in vitro studies and the related mechanism of cAMP-PKA/Epac pathway. The results in mice suggested that Que can inhibit the release of inflammatory cytokine, block neutrophil recruitment, and decrease the albumin leakage in dose-dependent manners. At the same time, Que can increase the cAMP content of lung tissue, and Epac content, except PKA. The results in epithelial cell (MLE-12) suggested that Que also can inhibit the inflammatory mediators keratinocyte-derived chemokines release after LPS stimulation; Epac inhibitor ESI-09 functionally antagonizes the inhibitory effect of Que; meanwhile, PKA inhibitor H89 functionally enhances the inhibitory effect of Que. Overexpression of Epac1 in MLE-12 suggested that Epac1 enhance the effect of Que. All those results suggested that the protective effect of quercetin in ALI is involved in cAMP-Epac pathway.
Collapse
Affiliation(s)
- Xue-Feng Wang
- Second Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, 310005, China
| | - Shun-de Song
- Zhejiang Respiratory Drugs Research Laboratory, School of Basic Medical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ya-Jun Li
- Zhejiang Respiratory Drugs Research Laboratory, School of Basic Medical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Zheng Qiang Hu
- Zhejiang Respiratory Drugs Research Laboratory, School of Basic Medical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Zhe-Wen Zhang
- Zhejiang Respiratory Drugs Research Laboratory, School of Basic Medical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Chun-Guang Yan
- Department of Pathogenic Biology and Immunology, Southeast University School of Medicine, Nanjing, 210009, China
| | - Zi-Gang Li
- Department of Anesthesiology, Women's Hospital,School of Medicine, Zhejiang University , Hangzhou, 310006, China
| | - Hui-Fang Tang
- Zhejiang Respiratory Drugs Research Laboratory, School of Basic Medical Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
48
|
Boulton S, Selvaratnam R, Blondeau JP, Lezoualc'h F, Melacini G. Mechanism of Selective Enzyme Inhibition through Uncompetitive Regulation of an Allosteric Agonist. J Am Chem Soc 2018; 140:9624-9637. [PMID: 30016089 DOI: 10.1021/jacs.8b05044] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Classical uncompetitive inhibitors are potent pharmacological modulators of enzyme function. Since they selectively target enzyme-substrate complexes (E:S), their inhibitory potency is amplified by increasing substrate concentrations. Recently, an unconventional uncompetitive inhibitor, called CE3F4R, was discovered for the exchange protein activated by cAMP isoform 1 (EPAC1). Unlike conventional uncompetitive inhibitors, CE3F4R is uncompetitive with respect to an allosteric effector, cAMP, as opposed to the substrate (i.e., CE3F4R targets the E:cAMP rather than the E:S complex). However, the mechanism of CE3F4R as an uncompetitive inhibitor is currently unknown. Here, we elucidate the mechanism of CE3F4R's action using NMR spectroscopy. Due to limited solubility and line broadening, which pose major challenges for traditional structural determination approaches, we resorted to a combination of protein- and ligand-based NMR experiments to comparatively analyze EPAC mutations, inhibitor analogs, and cyclic nucleotide derivatives that trap EPAC at different stages of activation. We discovered that CE3F4R binds within the EPAC cAMP-binding domain (CBD) at a subdomain interface distinct from the cAMP binding site, acting as a wedge that stabilizes a cAMP-bound mixed-intermediate. The mixed-intermediate includes attributes of both the apo/inactive and cAMP-bound/active states. In particular, the intermediate targeted by CE3F4R traps a CBD's hinge helix in its inactive conformation, locking EPAC into a closed domain topology that restricts substrate access to the catalytic domain. The proposed mechanism of action also explains the isoform selectivity of CE3F4R in terms of a single EPAC1 versus EPAC2 amino acid difference that destabilizes the active conformation of the hinge helix.
Collapse
Affiliation(s)
| | | | - Jean-Paul Blondeau
- Université Paris-Sud , Faculté de Pharmacie , 92296 Cedex Châtenay-Malabry , France
| | - Frank Lezoualc'h
- Inserm, UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse III Paul Sabatier , 31432 Cedex 04 Toulouse , France
| | | |
Collapse
|
49
|
Compartmentalized cyclic nucleotides have opposing effects on regulation of hypertrophic phospholipase Cε signaling in cardiac myocytes. J Mol Cell Cardiol 2018; 121:51-59. [PMID: 29885334 DOI: 10.1016/j.yjmcc.2018.06.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 05/12/2018] [Accepted: 06/05/2018] [Indexed: 12/11/2022]
Abstract
In cardiac myocytes activation of an exchange factor activated by cAMP (Epac) leads to activation of phospholipase Cε (PLCε)-dependent hydrolysis of phosphatidylinositol 4-phosphate (PI4P) in the Golgi apparatus a process critical for development of cardiac hypertrophy. Here we show that β-adrenergic receptor (βAR) stimulation does not stimulate this pathway in the presence of the broad spectrum phosphodiesterase (PDE) inhibitor IBMX, but selective PDE3 inhibition revealed βAR-dependent PI4P depletion. On the other hand, selective inhibition of PDE2 or PDE9A blocked endothelin-1 (ET-1) and cAMP-dependent PI4P hydrolysis by PLCε. Direct activation of protein kinase A (PKA), protein kinase G (PKG), or the atrial natriuretic factor (ANF) receptor abolished PI4P hydrolysis in response to multiple upstream stimuli. These results reveal distinct pools of cyclic nucleotides that either inhibit PLCε at the Golgi through PKA/PKG, or activate PLCε at the Golgi through Epac. These data together reveal a new mechanism by which ANF and selective PDE inhibitors can protect against cardiac hypertrophy.
Collapse
|
50
|
Szanda G, Wisniewski É, Rajki A, Spät A. Mitochondrial cAMP exerts positive feedback on mitochondrial Ca 2+ uptake via the recruitment of Epac1. J Cell Sci 2018; 131:jcs.215178. [PMID: 29661848 DOI: 10.1242/jcs.215178] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 04/07/2018] [Indexed: 01/10/2023] Open
Abstract
We have previously demonstrated in H295R adrenocortical cells that the Ca2+-dependent production of mitochondrial cAMP (mt-cAMP) by the matrix soluble adenylyl cyclase (sAC; encoded by ADCY10) is associated with enhanced aldosterone production. Here, we examined whether mitochondrial sAC and mt-cAMP fine tune mitochondrial Ca2+ metabolism to support steroidogenesis. Reduction of mt-cAMP formation resulted in decelerated mitochondrial Ca2+ accumulation in intact cells during K+-induced Ca2+ signalling and also in permeabilized cells exposed to elevated perimitochondrial [Ca2+]. By contrast, treatment with the membrane-permeable cAMP analogue 8-Br-cAMP, inhibition of phosphodiesterase 2 and overexpression of sAC in the mitochondrial matrix all intensified Ca2+ uptake into the organelle. Identical mt-cAMP dependence of mitochondrial Ca2+ uptake was also observed in HeLa cells. Importantly, the enhancing effect of mt-cAMP on Ca2+ uptake was independent from both the mitochondrial membrane potential and Ca2+ efflux, but was reduced by Epac1 (also known as RAPGEF3) blockade both in intact and in permeabilized cells. Finally, overexpression of sAC in the mitochondrial matrix potentiated aldosterone production implying that the observed positive feedback mechanism of mt-cAMP on mitochondrial Ca2+ accumulation may have a role in the rapid initiation of steroidogenesis.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Gergő Szanda
- Department of Physiology, Semmelweis University Medical School, 1482 POB 2 Budapest, Hungary .,MTA-SE Laboratory of Molecular Physiology, Semmelweis University and Hungarian Academy of Sciences, 1482 POB 2 Budapest, Hungary
| | - Éva Wisniewski
- Department of Physiology, Semmelweis University Medical School, 1482 POB 2 Budapest, Hungary
| | - Anikó Rajki
- MTA-SE Laboratory of Molecular Physiology, Semmelweis University and Hungarian Academy of Sciences, 1482 POB 2 Budapest, Hungary
| | - András Spät
- Department of Physiology, Semmelweis University Medical School, 1482 POB 2 Budapest, Hungary .,MTA-SE Laboratory of Molecular Physiology, Semmelweis University and Hungarian Academy of Sciences, 1482 POB 2 Budapest, Hungary
| |
Collapse
|