1
|
Elfawal MA, Goetz E, Kim Y, Chen P, Savinov SN, Barasa L, Thompson PR, Aroian RV. High-Throughput Screening of More Than 30,000 Compounds for Anthelmintics against Gastrointestinal Nematode Parasites. ACS Infect Dis 2025; 11:104-120. [PMID: 39653369 PMCID: PMC11731298 DOI: 10.1021/acsinfecdis.4c00327] [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: 04/25/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 12/18/2024]
Abstract
Gastrointestinal nematodes (GINs) are among the most common parasites of humans, livestock, and companion animals. GIN parasites infect 1-2 billion people worldwide, significantly impacting hundreds of millions of children, pregnant women, and adult workers, thereby perpetuating poverty. Two benzimidazoles with suboptimal efficacy are currently used to treat GINs in humans as part of mass drug administrations, with many instances of lower-than-expected or poor efficacy and possible resistance. Thus, new anthelmintics are urgently needed. However, screening methods for new anthelmintics using human GINs typically have low throughput. Here, using our novel screening pipeline that starts with human hookworms, we screened 30,238 unique small molecules from a wide range of compound libraries, including ones with generic diversity, repurposed drugs, natural derivatives, known mechanisms of action, as well as multiple target-focused libraries (e.g., targeting kinases, GPCRs, and neuronal proteins). We identified 55 compounds with broad-spectrum activity against adult stages of two evolutionary divergent GINs, hookworms (Ancylostoma ceylanicum) and whipworms (Trichuris muris). Based on known databases, the targets of these 55 compounds were predicted in nematode parasites. One novel scaffold from the diversity set library, F0317-0202, showed good activity (high motility inhibition) against both GINs. To better understand this novel scaffold's structure-activity relationships (SAR), we screened 28 analogs and created SAR models highlighting chemical and functional groups required for broad-spectrum activity. These studies validate our new and efficient screening pipeline at the level of tens of thousands of compounds and provide an important set of new GIN-active compounds for developing novel and broadly active anthelmintics.
Collapse
Affiliation(s)
- Mostafa A. Elfawal
- Program
in Molecular Medicine, University of Massachusetts
Chan Medical School, Worcester, Massachusetts 01605, United States
| | - Emily Goetz
- Program
in Molecular Medicine, University of Massachusetts
Chan Medical School, Worcester, Massachusetts 01605, United States
| | - Youmie Kim
- Program
in Molecular Medicine, University of Massachusetts
Chan Medical School, Worcester, Massachusetts 01605, United States
| | - Paulina Chen
- Program
in Molecular Medicine, University of Massachusetts
Chan Medical School, Worcester, Massachusetts 01605, United States
| | - Sergey N. Savinov
- Department
of Science, Rivier University, Nashua, New Hampshire 03060, United States
| | - Leonard Barasa
- Department
of Chemical Biology, University of Massachusetts
Chan Medical School, Worcester, Massachusetts 01605, United States
| | - Paul R. Thompson
- Department
of Chemical Biology, University of Massachusetts
Chan Medical School, Worcester, Massachusetts 01605, United States
| | - Raffi V. Aroian
- Program
in Molecular Medicine, University of Massachusetts
Chan Medical School, Worcester, Massachusetts 01605, United States
| |
Collapse
|
2
|
Elfawal MA, Goetz E, Kim YM, Chen P, Savinov SN, Barasa L, Thompson PR, Aroian RV. High-throughput screening of more than 30,000 compounds for anthelmintics against gastrointestinal nematode parasites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.16.594481. [PMID: 39554023 PMCID: PMC11565780 DOI: 10.1101/2024.05.16.594481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Gastrointestinal nematodes (GINs) are amongst the most common parasites of humans, livestock, and companion animals. GIN parasites infect 1-2 billion people worldwide, significantly impacting hundreds of millions of children, pregnant women, and adult workers, thereby perpetuating poverty. Two benzimidazoles with suboptimal efficacy are currently used to treat GINs in humans as part of mass drug administrations, with many instances of lower-than-expected or poor efficacy and possible resistance. Thus, new anthelmintics are urgently needed. However, screening methods for new anthelmintics using human GINs typically have low throughput. Here, using our novel screening pipeline that starts with human hookworms, we screened 30,238 unique small molecules from a wide range of compound libraries, including ones with generic diversity, repurposed drugs, natural derivatives, known mechanisms of action, as well as multiple target-focused libraries (e.g., targeting kinases, GPCRs, and neuronal proteins). We identified 55 compounds with broad-spectrum activity against adult stages of two evolutionary divergent GINs, hookworms ( Ancylostoma ceylanicum ) and whipworms ( Trichuris muris ). Based on known databases, the targets of these 55 compounds were predicted in nematode parasites. One novel scaffold from the diversity set library, F0317-0202, showed good activity (high motility inhibition) against both GINs. To better understand this novel scaffold's structure-activity relationships (SAR), we screened 28 analogs and created SAR models highlighting chemical and functional groups required for broad-spectrum activity. These studies validate our new and efficient screening pipeline at the level of tens of thousands of compounds and provide an important set of new GIN-active compounds for developing novel and broadly-active anthelmintics.
Collapse
|
3
|
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
|
4
|
Dong X, Chen Y, Lu J, Huang S, Pei G. β-arrestin 2 and Epac2 cooperatively mediate DRD1-stimulated proliferation of human neural stem cells and growth of human cerebral organoids. Stem Cells 2022; 40:857-869. [PMID: 35772103 DOI: 10.1093/stmcls/sxac046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 06/15/2022] [Indexed: 11/12/2022]
Abstract
G protein coupled receptors (GPCRs) reportedly relay specific signals, such as dopamine and serotonin, to regulate neurogenic processes though the underlying signaling pathways are not fully elucidated. Based on our previous work which demonstrated Dopamine receptor D1 (DRD1) effectively induces the proliferation of human neural stem cells, here we continued to show the knockout of β-arrestin 2 by CRISPR/Cas9 technology significantly weakened the DRD1-induced proliferation and neurosphere growth. Furthermore, inhibition of the downstream p38 MAPK by its specific inhibitors or small hairpin RNA mimicked the weakening effect of β-arrestin 2 knockout. In addition, blocking of Epac2, a PKA independent signal pathway, by its specific inhibitors or small hairpin RNA also significantly reduced DRD1-induced effects. Simultaneous inhibition of β-arrestin 2/p38 MAPK and Epac2 pathways nearly abolished the DRD1-stimulated neurogenesis, indicating the cooperative contribution of both pathways. Consistently, the expansion and folding of human cerebral organoids as stimulated by DRD1 were also mediated cooperatively by both β-arrestin 2/p38 MAPK and Epac2 pathways. Taken together, our results reveal that GPCRs apply at least two different signal pathways to regulate neurogenic processes in a delicate and balanced manners.
Collapse
Affiliation(s)
- Xiaoxu Dong
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China.,State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yujie Chen
- Uli Schwarz Quantitative Biology Core Facility, Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Juan Lu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Shichao Huang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Gang Pei
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China.,State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.,Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
5
|
Khan A, Ni W, Baltazar T, Lopez-Giraldez F, Pober JS, Pierce RW. ArhGEF12 activates Rap1A and not RhoA in human dermal microvascular endothelial cells to reduce tumor necrosis factor-induced leak. FASEB J 2022; 36:e22254. [PMID: 35294066 PMCID: PMC9103844 DOI: 10.1096/fj.202101873rr] [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: 12/06/2021] [Revised: 03/01/2022] [Accepted: 03/03/2022] [Indexed: 12/31/2022]
Abstract
Overwhelming inflammation in the setting of acute critical illness induces capillary leak resulting in hypovolemia, edema, tissue dysoxia, organ failure and even death. The tight junction (TJ)-dependent capillary barrier is regulated by small GTPases, but the specific regulatory molecules most active in this vascular segment under such circumstances are not well described. We set out to identify GTPase regulatory molecules specific to endothelial cells (EC) that form TJs. Transcriptional profiling of confluent monolayers of TJ-forming human dermal microvascular ECs (HDMECs) and adherens junction only forming-human umbilical vein EC (HUVECs) demonstrate ARHGEF12 is basally expressed at higher levels and is only downregulated in HDMECs by junction-disrupting tumor necrosis factor (TNF). HDMECs depleted of ArhGEF12 by siRNA demonstrate a significantly exacerbated TNF-induced decrease in trans-endothelial electrical resistance and disruption of TJ continuous staining. ArhGEF12 is established as a RhoA-GEF in HUVECs and its knock down would be expected to reduce RhoA activity and barrier disruption. Pulldown of active GEFs from HDMECs depleted of ArhGEF12 and treated with TNF show decreased GTP-bound Rap1A after four hours but increased GTP-bound RhoA after 12 h. In cell-free assays, ArhGEF12 immunoprecipitated from HDMECs is able to activate both Rap1A and RhoA, but not act on Rap2A-C, RhoB-C, or even Rap1B which shares 95% sequence identity with Rap1A. We conclude that in TJ-forming HDMECs, ArhGEF12 selectively activates Rap1A to limit capillary barrier disruption in a mechanism independent of cAMP-mediated Epac1 activation.
Collapse
Affiliation(s)
- Alamzeb Khan
- Department of Pediatrics, Yale School of Medicine, Yale University
| | - Weiming Ni
- Department of Pediatrics, Yale School of Medicine, Yale University
| | - Tania Baltazar
- Department of Immunobiology, Yale School of Medicine, Yale University
| | | | - Jordan S. Pober
- Department of Immunobiology, Yale School of Medicine, Yale University
| | | |
Collapse
|
6
|
Zhang X, Feng Y, Tuo Y, Zheng QZ. Metal-free sulfonylation of arenes with N-fluorobenzenesulfonimide via cleavage of S-N bonds: expeditious synthesis of diarylsulfones. Org Biomol Chem 2022; 20:768-772. [PMID: 34989387 DOI: 10.1039/d1ob02209a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A novel metal-free sulfonylation of arenes with N-fluorobenzenesulfonimide (NFSI) toward the synthesis of diarylsulfones has been developed. The reaction represents a rare example of sulfonylation reaction using NFSI as an efficient sulfonyl donor and the first example of acid-mediated sulfonylation of unactivated arenes with NFSI via selective cleavage of S-N bonds. This protocol provides a concise approach for the construction of pharmaceutically and biologically important diarylsulfones. Applications in the functionalization of natural products (e.g., β-estradiol) and in the synthesis of a key intermediate to an inhibitor of farnesyl-protein transferase, as well as in the gram-scale synthesis of the EPAC2 antagonist, are demonstrated.
Collapse
Affiliation(s)
- Xiaohui Zhang
- State Key Laboratory of High-efficiency Utilization of Coal and Green Chemical Engineering, National Demonstration Center for Experimental Chemistry Education, School of Chemistry and Chemical Engineering, Ningxia University, Yinchuan 750021, China.
| | - Yueji Feng
- State Key Laboratory of High-efficiency Utilization of Coal and Green Chemical Engineering, National Demonstration Center for Experimental Chemistry Education, School of Chemistry and Chemical Engineering, Ningxia University, Yinchuan 750021, China.
| | - Yanyan Tuo
- State Key Laboratory of High-efficiency Utilization of Coal and Green Chemical Engineering, National Demonstration Center for Experimental Chemistry Education, School of Chemistry and Chemical Engineering, Ningxia University, Yinchuan 750021, China.
| | - Qing-Zhong Zheng
- State Key Laboratory of High-efficiency Utilization of Coal and Green Chemical Engineering, National Demonstration Center for Experimental Chemistry Education, School of Chemistry and Chemical Engineering, Ningxia University, Yinchuan 750021, China. .,State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Xue Yuan Rd. 38, Beijing 100191, China
| |
Collapse
|
7
|
Song X, Wen H, Zuo L, Geng Z, Nian J, Wang L, Jiang Y, Tao J, Zhu Z, Wu X, Wang Z, Zhang X, Yu L, Zhao H, Xiang P, Li J, Shen L, Hu J. Epac-2 ameliorates spontaneous colitis in Il-10 -/- mice by protecting the intestinal barrier and suppressing NF-κB/MAPK signalling. J Cell Mol Med 2022; 26:216-227. [PMID: 34862717 PMCID: PMC8742196 DOI: 10.1111/jcmm.17077] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 07/25/2021] [Accepted: 11/17/2021] [Indexed: 02/06/2023] Open
Abstract
Intestinal barrier dysfunction and intestinal inflammation interact in the progression of Crohn's disease (CD). A recent study indicated that Epac-2 protected the intestinal barrier and had anti-inflammatory effects. The present study examined the function of Epac-2 in CD-like colitis. Interleukin-10 gene knockout (Il-10-/- ) mice exhibit significant spontaneous enteritis and were used as the CD model. These mice were treated with Epac-2 agonists (Me-cAMP) or Epac-2 antagonists (HJC-0350) or were fed normally (control), and colitis and intestinal barrier structure and function were compared. A Caco-2 and RAW 264.7 cell co-culture system were used to analyse the effects of Epac-2 on the cross-talk between intestinal epithelial cells and inflammatory cells. Epac-2 activation significantly ameliorated colitis in mice, which was indicated by reductions in the colitis inflammation score, the expression of inflammatory factors and intestinal permeability. Epac-2 activation also decreased Caco-2 cell permeability in an LPS-induced cell co-culture system. Epac-2 activation significantly suppressed nuclear factor (NF)-κB/mitogen-activated protein kinase (MAPK) signalling in vivo and in vitro. Epac-2 may be a therapeutic target for CD based on its anti-inflammatory functions and protective effects on the intestinal barrier.
Collapse
Affiliation(s)
- Xue Song
- Department of Central LaboratoryFirst Affiliated Hospital of Bengbu Medical CollegeBengbuChina
- Anhui Key Laboratory of Tissue TransplantationBengbu Medical CollegeBengbuChina
| | - Hexin Wen
- Department of Gastrointestinal SurgeryFirst Affiliated Hospital of Bengbu Medical CollegeBengbuChina
| | - Lugen Zuo
- Anhui Key Laboratory of Tissue TransplantationBengbu Medical CollegeBengbuChina
- Department of Gastrointestinal SurgeryFirst Affiliated Hospital of Bengbu Medical CollegeBengbuChina
| | - Zhijun Geng
- Department of Central LaboratoryFirst Affiliated Hospital of Bengbu Medical CollegeBengbuChina
- Anhui Key Laboratory of Tissue TransplantationBengbu Medical CollegeBengbuChina
| | - Jing Nian
- Department of ImagingSecond Affiliated Hospital of Bengbu Medical CollegeBengbuChina
| | - Luyao Wang
- Anhui Key Laboratory of Tissue TransplantationBengbu Medical CollegeBengbuChina
- Department of Clinical MedicineBengbu Medical CollegeBengbuChina
| | - Yifan Jiang
- Anhui Key Laboratory of Tissue TransplantationBengbu Medical CollegeBengbuChina
- Department of Clinical MedicineBengbu Medical CollegeBengbuChina
| | - Jing Tao
- Anhui Key Laboratory of Tissue TransplantationBengbu Medical CollegeBengbuChina
- Department of Clinical MedicineBengbu Medical CollegeBengbuChina
| | - Zihan Zhu
- Anhui Key Laboratory of Tissue TransplantationBengbu Medical CollegeBengbuChina
- Department of Clinical MedicineBengbu Medical CollegeBengbuChina
| | - Xiaopei Wu
- Anhui Key Laboratory of Tissue TransplantationBengbu Medical CollegeBengbuChina
- Department of Clinical MedicineBengbu Medical CollegeBengbuChina
| | - Zhikun Wang
- Anhui Key Laboratory of Tissue TransplantationBengbu Medical CollegeBengbuChina
- Department of Clinical MedicineBengbu Medical CollegeBengbuChina
| | - Xiaofeng Zhang
- Department of Central LaboratoryFirst Affiliated Hospital of Bengbu Medical CollegeBengbuChina
| | - Liang Yu
- Department of Central LaboratoryFirst Affiliated Hospital of Bengbu Medical CollegeBengbuChina
- Anhui Key Laboratory of Tissue TransplantationBengbu Medical CollegeBengbuChina
| | - Hao Zhao
- Department of Central LaboratoryFirst Affiliated Hospital of Bengbu Medical CollegeBengbuChina
| | - Ping Xiang
- Department of Central LaboratoryFirst Affiliated Hospital of Bengbu Medical CollegeBengbuChina
| | - Jing Li
- Anhui Key Laboratory of Tissue TransplantationBengbu Medical CollegeBengbuChina
- Department of Clinical LaboratoryFirst Affiliated Hospital of Bengbu Medical CollegeBengbuChina
| | - Lin Shen
- Anhui Key Laboratory of Tissue TransplantationBengbu Medical CollegeBengbuChina
| | - Jianguo Hu
- Anhui Key Laboratory of Tissue TransplantationBengbu Medical CollegeBengbuChina
- Department of Clinical LaboratoryFirst Affiliated Hospital of Bengbu Medical CollegeBengbuChina
| |
Collapse
|
8
|
Glycine Release Is Potentiated by cAMP via EPAC2 and Ca 2+ Stores in a Retinal Interneuron. J Neurosci 2021; 41:9503-9520. [PMID: 34620721 PMCID: PMC8612479 DOI: 10.1523/jneurosci.0670-21.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 11/21/2022] Open
Abstract
Neuromodulation via the intracellular second messenger cAMP is ubiquitous at presynaptic nerve terminals. This modulation of synaptic transmission allows exocytosis to adapt to stimulus levels and reliably encode information. The AII amacrine cell (AII-AC) is a central hub for signal processing in the mammalian retina. The main apical dendrite of the AII-AC is connected to several lobular appendages that release glycine onto OFF cone bipolar cells and ganglion cells. However, the influence of cAMP on glycine release is not well understood. Using membrane capacitance measurements from mouse AII-ACs to directly measure exocytosis, we observe that intracellular dialysis of 1 mm cAMP enhances exocytosis without affecting the L-type Ca2+ current. Responses to depolarizing pulses of various durations show that the size of the readily releasable pool of vesicles nearly doubles with cAMP, while paired-pulse depression experiments suggest that release probability does not change. Specific agonists and antagonists for exchange protein activated by cAMP 2 (EPAC2) revealed that the cAMP-induced enhancement of exocytosis requires EPAC2 activation. Furthermore, intact Ca2+ stores were also necessary for the cAMP potentiation of exocytosis. Postsynaptic recordings from OFF cone bipolar cells showed that increasing cAMP with forskolin potentiated the frequency of glycinergic spontaneous IPSCs. We propose that cAMP elevations in the AII-AC lead to a robust enhancement of glycine release through an EPAC2 and Ca2+ store signaling pathway. Our results thus contribute to a better understanding of how AII-AC crossover inhibitory circuits adapt to changes in ambient luminance.SIGNIFICANCE STATEMENT The mammalian retina operates over a wide dynamic range of light intensities and contrast levels. To optimize the signal-to-noise ratio of processed visual information, both excitatory and inhibitory synapses within the retina must modulate their gain in synaptic transmission to adapt to different levels of ambient light. Here we show that increases of cAMP concentration within AII amacrine cells produce enhanced exocytosis from these glycinergic interneurons. Therefore, we propose that light-sensitive neuromodulators may change the output of glycine release from AII amacrine cells. This novel mechanism may fine-tune the amount of tonic and phasic synaptic inhibition received by bipolar cell terminals and, consequently, the spiking patterns that ganglion cells send to the upstream visual areas of the brain.
Collapse
|
9
|
Chen W, McRoberts JA, Ennes HS, Marvizon JC. cAMP signaling through protein kinase A and Epac2 induces substance P release in the rat spinal cord. Neuropharmacology 2021; 189:108533. [PMID: 33744339 DOI: 10.1016/j.neuropharm.2021.108533] [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: 08/12/2020] [Revised: 03/01/2021] [Accepted: 03/11/2021] [Indexed: 11/18/2022]
Abstract
Using neurokinin 1 receptor (NK1R) internalization to measure of substance P release in rat spinal cord slices, we found that it was induced by the adenylyl cyclase (AC) activator forskolin, by the protein kinase A (PKA) activators 6-Bnz-cAMP and 8-Br-cAMP, and by the activator of exchange protein activated by cAMP (Epac) 8-pCPT-2-O-Me-cAMP (CPTOMe-cAMP). Conversely, AC and PKA inhibitors decreased substance P release induced by electrical stimulation of the dorsal root. Therefore, the cAMP signaling pathway mediates substance P release in the dorsal horn. The effects of forskolin and 6-Bnz-cAMP were not additive with NMDA-induced substance P release and were decreased by the NMDA receptor blocker MK-801. In cultured dorsal horn neurons, forskolin increased NMDA-induced Ca2+ entry and the phosphorylation of the NR1 and NR2B subunits of the NMDA receptor. Therefore, cAMP-induced substance P release is mediated by the activating phosphorylation by PKA of NMDA receptors. Voltage-gated Ca2+ channels, but not by TRPV1 or TRPA1, also contributed to cAMP-induced substance P release. Activation of PKA was required for the effects of forskolin and the three cAMP analogs. Epac2 contributed to the effects of forskolin and CPTOMe-cAMP, signaling through a Raf - mitogen-activated protein kinase pathway to activate Ca2+ channels. Epac1 inhibitors induced NK1R internalization independently of substance P release. In rats with latent sensitization to pain, the effect of 6-Bnz-cAMP was unchanged, whereas the effect of forskolin was decreased due to the loss of the stimulatory effect of Epac2. Hence, substance P release induced by cAMP decreases during pain hypersensitivity.
Collapse
Affiliation(s)
- Wenling Chen
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, 90095, USA; Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA
| | - James A McRoberts
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Helena S Ennes
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Juan Carlos Marvizon
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, 90095, USA; Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA.
| |
Collapse
|
10
|
Pd/NHC-catalyzed arylsulfonylation of boronic acids: A general and direct protocol to access diarylsulfones. Tetrahedron Lett 2021. [DOI: 10.1016/j.tetlet.2020.152708] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
11
|
Lopez ER, Carbajal AG, Tian JB, Bavencoffe A, Zhu MX, Dessauer CW, Walters ET. Serotonin enhances depolarizing spontaneous fluctuations, excitability, and ongoing activity in isolated rat DRG neurons via 5-HT 4 receptors and cAMP-dependent mechanisms. Neuropharmacology 2020; 184:108408. [PMID: 33220305 DOI: 10.1016/j.neuropharm.2020.108408] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/10/2020] [Accepted: 11/16/2020] [Indexed: 12/18/2022]
Abstract
Ongoing activity in nociceptors, a driver of spontaneous pain, can be generated in dorsal root ganglion neurons in the absence of sensory generator potentials if one or more of three neurophysiological alterations occur - prolonged depolarization of resting membrane potential (RMP), hyperpolarization of action potential (AP) threshold, and/or increased amplitude of depolarizing spontaneous fluctuations of membrane potential (DSFs) to bridge the gap between RMP and AP threshold. Previous work showed that acute, sustained exposure to serotonin (5-HT) hyperpolarized AP threshold and potentiated DSFs, leading to ongoing activity if a separate source of maintained depolarization was present. Cellular signaling pathways that increase DSF amplitude and promote ongoing activity acutely in nociceptors are not known for any neuromodulator. Here, isolated DRG neurons from male rats were used to define the pathway by which low concentrations of 5-HT enhance DSFs, hyperpolarize AP threshold, and promote ongoing activity. A selective 5-HT4 receptor antagonist blocked these 5-HT-induced hyperexcitable effects, while a selective 5-HT4 agonist mimicked the effects of 5-HT. Inhibition of cAMP effectors, protein kinase A (PKA) and exchange protein activated by cAMP (EPAC), attenuated 5-HT's hyperexcitable effects, but a blocker of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels had no significant effect. 5-HT4-dependent PKA activation was specific to DRG neurons that bind isolectin B4 (a nonpeptidergic nociceptor marker). 5-HT's effects on AP threshold, DSFs, and ongoing activity were mimicked by a cAMP analog. Sustained exposure to 5-HT promotes ongoing activity in nonpeptidergic nociceptors through the Gs-coupled 5-HT4 receptor and downstream cAMP signaling involving both PKA and EPAC.
Collapse
Affiliation(s)
- Elia R Lopez
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UT Health, Houston, TX, 77030, USA.
| | - Anibal Garza Carbajal
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UT Health, Houston, TX, 77030, USA.
| | - Jin Bin Tian
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UT Health, Houston, TX, 77030, USA.
| | - Alexis Bavencoffe
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UT Health, Houston, TX, 77030, USA.
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UT Health, Houston, TX, 77030, USA.
| | - Carmen W Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UT Health, Houston, TX, 77030, USA.
| | - Edgar T Walters
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UT Health, Houston, TX, 77030, USA.
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Richard SA. EPAC2: A new and promising protein for glioma pathogenesis and therapy. Oncol Rev 2020; 14:446. [PMID: 32395202 PMCID: PMC7204831 DOI: 10.4081/oncol.2020.446] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 04/16/2020] [Indexed: 01/02/2023] Open
Abstract
Gliomas are prime brain cancers which are initiated by malignant modification of neural stem cells, progenitor cells and differentiated glial cells such as astrocyte, oligodendrocyte as well as ependymal cells. Exchange proteins directly activated by cAMP (EPACs) are crucial cyclic adenosine 3’,5’-monophosphate (cAMP)-determined signaling pathways. Cyclic AMP-intermediated signaling events were utilized to transduce protein kinase A (PKA) leading to the detection of EPACs or cAMP-guanine exchange factors (cAMP-GEFs). EPACs have been detected as crucial proteins associated with the pathogenesis of neurological disorders as well as numerous human diseases. EPAC proteins have two isoforms. These isoforms are EPAC1 and EPAC2. EPAC2 also known as Rap guanine nucleotide exchange factor 4 (RAPGEF4) is generally expression in all neurites. Higher EAPC2 levels was detected in the cortex, hippocampus as well as striatum of adult mouse brain. Activation as well as over-secretion of EPAC2 triggers apoptosis in neurons and EPAC-triggered apoptosis was intermediated via the modulation of Bcl-2 interacting member protein (BIM). EPAC2 secretory levels has proven to be more in low-grade clinical glioma than high-grade clinical glioma. This review therefore explores the effects of EPAC2/RAPGEF4 on the pathogenesis of glioma instead of EPAC1 because EPAC2 and not EPAC1 is predominately expressed in the brain. Therefore, EPAC2 is most likely to modulate glioma pathogenesis rather than EPAC1.
Collapse
Affiliation(s)
- Seidu A Richard
- Department of Medicine, Princefield University, Ho, Ghana, West Africa
| |
Collapse
|
15
|
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
|
16
|
EPAC1 and EPAC2 promote nociceptor hyperactivity associated with chronic pain after spinal cord injury. NEUROBIOLOGY OF PAIN 2019; 7:100040. [PMID: 31890991 PMCID: PMC6926371 DOI: 10.1016/j.ynpai.2019.100040] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/15/2019] [Accepted: 11/20/2019] [Indexed: 12/19/2022]
Abstract
Chronic pain following spinal cord injury (SCI) is associated with electrical hyperactivity (spontaneous and evoked) in primary nociceptors. Cyclic adenosine monophosphate (cAMP) signaling is an important contributor to nociceptor excitability, and knockdown of the cAMP effector, exchange protein activated by cAMP (EPAC), has been shown to relieve pain-like responses in several chronic pain models. To examine potentially distinct roles of each EPAC isoform (EPAC1 and 2) in maintaining chronic pain, we used rat and mouse models of contusive spinal cord injury (SCI). Pharmacological inhibition of EPAC1 or 2 in a rat SCI model was sufficient to reverse SCI-induced nociceptor hyperactivity, indicating that EPAC1 and 2 signaling activity are complementary, with both required to maintain hyperactivity. However, EPAC activation was not sufficient to induce similar hyperactivity in nociceptors from naïve rats, and we observed no change in EPAC protein expression after SCI. In the mouse SCI model, inhibition of both EPAC isoforms through a combination of pharmacological inhibition and genetic deletion was required to reverse SCI-induced nociceptor hyperactivity. This was consistent with our finding that neither EPAC1-/- nor EPAC2-/- mice were protected against SCI-induced chronic pain as assessed with an operant mechanical conflict test. Thus, EPAC1 and 2 activity may play a redundant role in mouse nociceptors, although no corresponding change in EPAC protein expression levels was detected after SCI. Despite some differences between these species, our data demonstrate a fundamental role for both EPAC1 and EPAC2 in mechanisms maintaining nociceptor hyperactivity and chronic pain after SCI.
Collapse
|
17
|
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
|
18
|
M. Beck E, Parnell E, Cowley A, Porter A, Gillespie J, Robinson J, Robinson L, Pannifer AD, Hamon V, Jones P, Morrison A, McElroy S, Timmerman M, Rutjes H, Mahajan P, Wiejak J, Luchowska-Stańska U, Morgan D, Barker G, Rehmann H, Yarwood SJ. Identification of A Novel Class of Benzofuran Oxoacetic Acid-Derived Ligands that Selectively Activate Cellular EPAC1. Cells 2019; 8:cells8111425. [PMID: 31726720 PMCID: PMC6912754 DOI: 10.3390/cells8111425] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 12/20/2022] Open
Abstract
Cyclic AMP promotes EPAC1 and EPAC2 activation through direct binding to a specific cyclic nucleotide-binding domain (CNBD) within each protein, leading to activation of Rap GTPases, which control multiple cell responses, including cell proliferation, adhesion, morphology, exocytosis, and gene expression. As a result, it has become apparent that directed activation of EPAC1 and EPAC2 with synthetic agonists may also be useful for the future treatment of diabetes and cardiovascular diseases. To identify new EPAC agonists we have developed a fluorescent-based, ultra-high-throughput screening (uHTS) assay that measures the displacement of binding of the fluorescent cAMP analogue, 8-NBD-cAMP to the EPAC1 CNBD. Triage of the output of an approximately 350,000 compound screens using this assay identified a benzofuran oxaloacetic acid EPAC1 binder (SY000) that displayed moderate potency using orthogonal assays (competition binding and microscale thermophoresis). We next generated a limited library of 91 analogues of SY000 and identified SY009, with modifications to the benzofuran ring associated with a 10-fold increase in potency towards EPAC1 over SY000 in binding assays. In vitro EPAC1 activity assays confirmed the agonist potential of these molecules in comparison with the known EPAC1 non-cyclic nucleotide (NCN) partial agonist, I942. Rap1 GTPase activation assays further demonstrated that SY009 selectively activates EPAC1 over EPAC2 in cells. SY009 therefore represents a novel class of NCN EPAC1 activators that selectively activate EPAC1 in cellulae.
Collapse
Affiliation(s)
- Elizabeth M. Beck
- European Screening Centre Newhouse, University of Dundee, Biocity Scotland, Bo’Ness Road, Newhouse, Lanarkshire ML1 5UH, UK; (E.M.B.); (A.C.); (A.P.); (J.G.); (J.R.); (L.R.); (A.D.P.); (V.H.); (P.J.); (A.M.); (S.M.)
| | - Euan Parnell
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Angela Cowley
- European Screening Centre Newhouse, University of Dundee, Biocity Scotland, Bo’Ness Road, Newhouse, Lanarkshire ML1 5UH, UK; (E.M.B.); (A.C.); (A.P.); (J.G.); (J.R.); (L.R.); (A.D.P.); (V.H.); (P.J.); (A.M.); (S.M.)
| | - Alison Porter
- European Screening Centre Newhouse, University of Dundee, Biocity Scotland, Bo’Ness Road, Newhouse, Lanarkshire ML1 5UH, UK; (E.M.B.); (A.C.); (A.P.); (J.G.); (J.R.); (L.R.); (A.D.P.); (V.H.); (P.J.); (A.M.); (S.M.)
| | - Jonathan Gillespie
- European Screening Centre Newhouse, University of Dundee, Biocity Scotland, Bo’Ness Road, Newhouse, Lanarkshire ML1 5UH, UK; (E.M.B.); (A.C.); (A.P.); (J.G.); (J.R.); (L.R.); (A.D.P.); (V.H.); (P.J.); (A.M.); (S.M.)
| | - John Robinson
- European Screening Centre Newhouse, University of Dundee, Biocity Scotland, Bo’Ness Road, Newhouse, Lanarkshire ML1 5UH, UK; (E.M.B.); (A.C.); (A.P.); (J.G.); (J.R.); (L.R.); (A.D.P.); (V.H.); (P.J.); (A.M.); (S.M.)
| | - Lindsay Robinson
- European Screening Centre Newhouse, University of Dundee, Biocity Scotland, Bo’Ness Road, Newhouse, Lanarkshire ML1 5UH, UK; (E.M.B.); (A.C.); (A.P.); (J.G.); (J.R.); (L.R.); (A.D.P.); (V.H.); (P.J.); (A.M.); (S.M.)
| | - Andrew D. Pannifer
- European Screening Centre Newhouse, University of Dundee, Biocity Scotland, Bo’Ness Road, Newhouse, Lanarkshire ML1 5UH, UK; (E.M.B.); (A.C.); (A.P.); (J.G.); (J.R.); (L.R.); (A.D.P.); (V.H.); (P.J.); (A.M.); (S.M.)
| | - Veronique Hamon
- European Screening Centre Newhouse, University of Dundee, Biocity Scotland, Bo’Ness Road, Newhouse, Lanarkshire ML1 5UH, UK; (E.M.B.); (A.C.); (A.P.); (J.G.); (J.R.); (L.R.); (A.D.P.); (V.H.); (P.J.); (A.M.); (S.M.)
| | - Philip Jones
- European Screening Centre Newhouse, University of Dundee, Biocity Scotland, Bo’Ness Road, Newhouse, Lanarkshire ML1 5UH, UK; (E.M.B.); (A.C.); (A.P.); (J.G.); (J.R.); (L.R.); (A.D.P.); (V.H.); (P.J.); (A.M.); (S.M.)
| | - Angus Morrison
- European Screening Centre Newhouse, University of Dundee, Biocity Scotland, Bo’Ness Road, Newhouse, Lanarkshire ML1 5UH, UK; (E.M.B.); (A.C.); (A.P.); (J.G.); (J.R.); (L.R.); (A.D.P.); (V.H.); (P.J.); (A.M.); (S.M.)
| | - Stuart McElroy
- European Screening Centre Newhouse, University of Dundee, Biocity Scotland, Bo’Ness Road, Newhouse, Lanarkshire ML1 5UH, UK; (E.M.B.); (A.C.); (A.P.); (J.G.); (J.R.); (L.R.); (A.D.P.); (V.H.); (P.J.); (A.M.); (S.M.)
| | - Martin Timmerman
- Pivot Park Screening Centre, Kloosterstraat 9, 5349 AB Oss, The Netherlands; (M.T.); (H.R.)
| | - Helma Rutjes
- Pivot Park Screening Centre, Kloosterstraat 9, 5349 AB Oss, The Netherlands; (M.T.); (H.R.)
| | - Pravin Mahajan
- Structural Genomics Consortium, University of Oxford, Oxford OX3 7DQ, UK;
| | - Jolanta Wiejak
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh Campus, Edinburgh EH14 4AS, UK; (J.W.); (U.L.-S.)
| | - Urszula Luchowska-Stańska
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh Campus, Edinburgh EH14 4AS, UK; (J.W.); (U.L.-S.)
| | - David Morgan
- Institute of Chemical Sciences, Heriot-Watt University, Edinburgh EH14 4AS, UK; (D.M.); (G.B.)
| | - Graeme Barker
- Institute of Chemical Sciences, Heriot-Watt University, Edinburgh EH14 4AS, UK; (D.M.); (G.B.)
| | - Holger Rehmann
- Department of Molecular Cancer Research, Centre of Biomedical Genetics and Cancer Genomics Centre, University Medical Centre Utrecht, 3508 TC Utrecht, The Netherlands;
| | - Stephen J. Yarwood
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh Campus, Edinburgh EH14 4AS, UK; (J.W.); (U.L.-S.)
- Correspondence:
| |
Collapse
|
19
|
Fu W, Nelson TS, Santos DF, Doolen S, Gutierrez JJ, Ye N, Zhou J, Taylor B. An NPY Y1 receptor antagonist unmasks latent sensitization and reveals the contribution of protein kinase A and Epac to chronic inflammatory pain. Pain 2019; 160:1754-1765. [PMID: 31335645 PMCID: PMC6903783 DOI: 10.1097/j.pain.0000000000001557] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Peripheral inflammation produces a long-lasting latent sensitization of spinal nociceptive neurons, that is, masked by tonic inhibitory controls. We explored mechanisms of latent sensitization with an established four-step approach: (1) induction of inflammation; (2) allow pain hypersensitivity to resolve; (3) interrogate latent sensitization with a channel blocker, mutant mouse, or receptor antagonist; and (4) disrupt compensatory inhibition with a receptor antagonist so as to reinstate pain hypersensitivity. We found that the neuropeptide Y Y1 receptor antagonist BIBO3304 reinstated pain hypersensitivity, indicative of an unmasking of latent sensitization. BIBO3304-evoked reinstatement was not observed in AC1 knockout mice and was prevented with intrathecal co-administration of a pharmacological blocker to the N-methyl-D-aspartate receptor (NMDAR), adenylyl cyclase type 1 (AC1), protein kinase A (PKA), transient receptor potential cation channel A1 (TRPA1), channel V1 (TRPV1), or exchange protein activated by cAMP (Epac1 or Epac2). A PKA activator evoked both pain reinstatement and touch-evoked pERK expression in dorsal horn; the former was prevented with intrathecal co-administration of a TRPA1 or TRPV1 blocker. An Epac activator also evoked pain reinstatement and pERK expression. We conclude that PKA and Epac are sufficient to maintain long-lasting latent sensitization of dorsal horn neurons that is kept in remission by the NPY-Y1 receptor system. Furthermore, we have identified and characterized 2 novel molecular signaling pathways in the dorsal horn that drive latent sensitization in the setting of chronic inflammatory pain: NMDAR→AC1→PKA→TRPA1/V1 and NMDAR→AC1→Epac1/2. New treatments for chronic inflammatory pain might either increase endogenous NPY analgesia or inhibit AC1, PKA, or Epac.
Collapse
Affiliation(s)
- Weisi Fu
- Department of Physiology, University of Kentucky Medical Center, Lexington KY, USA
| | - Tyler S. Nelson
- Department of Anesthesiology, Pittsburgh Center for Pain Research, and the Opiate Research Center at the University of Pittsburgh, University of Pittsburgh, Pittsburgh, PA USA
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA USA
- Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA USA
| | - Diogo F. Santos
- Department of Anesthesiology, Pittsburgh Center for Pain Research, and the Opiate Research Center at the University of Pittsburgh, University of Pittsburgh, Pittsburgh, PA USA
| | - Suzanne Doolen
- Department of Anesthesiology, Pittsburgh Center for Pain Research, and the Opiate Research Center at the University of Pittsburgh, University of Pittsburgh, Pittsburgh, PA USA
| | - Javier J.P. Gutierrez
- Department of Anesthesiology, Pittsburgh Center for Pain Research, and the Opiate Research Center at the University of Pittsburgh, University of Pittsburgh, Pittsburgh, PA USA
| | - Na Ye
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jia Zhou
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Bradley Taylor
- Department of Physiology, University of Kentucky Medical Center, Lexington KY, USA
- Department of Anesthesiology, Pittsburgh Center for Pain Research, and the Opiate Research Center at the University of Pittsburgh, University of Pittsburgh, Pittsburgh, PA USA
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA USA
| |
Collapse
|
20
|
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
|
21
|
Wang W, Zhang L, Morlock L, Williams NS, Shay JW, De Brabander JK. Design and Synthesis of TASIN Analogues Specifically Targeting Colorectal Cancer Cell Lines with Mutant Adenomatous Polyposis Coli (APC). J Med Chem 2019; 62:5217-5241. [PMID: 31070915 DOI: 10.1021/acs.jmedchem.9b00532] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite advances in targeted anticancer therapies, there are still no small-molecule-based therapies available that specifically target colorectal cancer (CRC) development and progression, the second leading cause of cancer deaths. We previously disclosed the discovery of truncating adenomatous polyposis coli (APC)-selective inhibitor 1 (TASIN-1), a small molecule that specifically targets colorectal cancer cells lines with truncating mutations in the adenomatous polyposis coli (APC) tumor suppressor gene through inhibition of cholesterol biosynthesis. Here, we report a medicinal chemistry evaluation of a collection of TASIN analogues and activity against colon cancer cell lines and an isogenic cell line pair reporting on the status of APC-dependent selectivity. A number of potent and selective analogues were identified, including compounds with good metabolic stability and pharmacokinetic properties. The compounds reported herein represent a first-in-class genotype-selective series that specifically target apc mutations present in the majority of CRC patients and serve as a translational platform toward a targeted therapy for colon cancer.
Collapse
|
22
|
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
|
23
|
Inflammation induces Epac-protein kinase C alpha and epsilon signaling in TRPV1-mediated hyperalgesia. Pain 2019; 159:2383-2393. [PMID: 30015706 DOI: 10.1097/j.pain.0000000000001346] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The exchange proteins activated by cAMP (Epacs) have been shown to play important roles in producing inflammation-induced nociception. Transient receptor potential vanilloid type 1 (TRPV1) is a major receptor processing thermal and chemosensitive nociceptive information. The role of Epacs in modulating the activity of TRPV1 has yet to be determined. Studying the effect of complete Freund adjuvant (CFA)-induced inflammation on capsaicin-activated TRPV1 nociceptive responses in dorsal root ganglia (DRG), we found that CFA produced a large increase in capsaicin-induced responses. The increase was inhibited by Epac1 and Epac2 antagonists. Thus, activation of Epacs is critical in producing enhancement in TRPV1-mediated responses under inflammatory conditions. In addition, the inflammation-induced enhancement of TRPV1 responses was blocked by PKCα and PKCε inhibitors, suggesting the essential roles of these PKCs in enhancing TRPV1 responses. To determine the mechanism underlying the Epac actions on TRPV1, we studied the effects of the Epac activator, 8-(4-chlorophenylthio)-2-O-methyl-cAMP (CPT), on capsaicin-induced nociceptive behavioral responses, capsaicin-activated currents, expression and membrane trafficking of PKC and TRPV1 in DRG. CPT was found to enhance capsaicin-induced nociception and ionic currents. The enhancement was inhibited by PKCα and PKCε inhibitors. In addition, CPT increased the expression of phosphorylated PKCα (pPKCα) and membrane TRPV1 expression in DRG. Studying the colocalization of TRPV1 and pPKCα or pPKCε in DRG slices prepared from CFA-treated rats, we found that pPKCα or pPKCε expressed with TRPV1 in different-sized neurons to exert differential influences on TRPV1 activity. Thus, Epac-PKC signaling is critically important in producing inflammation-induced potentiation of TRPV1 functions.
Collapse
|
24
|
Zhu H, Shen Y, Wen D, Le ZG, Tu T. Selective Synthesis of ortho-Substituted Diarylsulfones by Using NHC-Au Catalysts under Mild Conditions. Org Lett 2019; 21:974-979. [DOI: 10.1021/acs.orglett.8b03957] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Haibo Zhu
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Department of Chemistry, Fudan University, 2205 Songhu Road, Shanghai 200438, China
- School of Chemistry, Biology and Material Science, East China University of Technology, Nanchang 330013, China
| | - Yajing Shen
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Department of Chemistry, Fudan University, 2205 Songhu Road, Shanghai 200438, China
| | - Daheng Wen
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Department of Chemistry, Fudan University, 2205 Songhu Road, Shanghai 200438, China
| | - Zhang-Gao Le
- School of Chemistry, Biology and Material Science, East China University of Technology, Nanchang 330013, China
| | - Tao Tu
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Department of Chemistry, Fudan University, 2205 Songhu Road, Shanghai 200438, China
- State Key Laboratory of Organometallic Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| |
Collapse
|
25
|
Wiejak J, van Basten B, Luchowska-Stańska U, Hamilton G, Yarwood SJ. The novel exchange protein activated by cyclic AMP 1 (EPAC1) agonist, I942, regulates inflammatory gene expression in human umbilical vascular endothelial cells (HUVECs). BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1866:264-276. [PMID: 30414891 PMCID: PMC6325792 DOI: 10.1016/j.bbamcr.2018.11.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/31/2018] [Accepted: 11/06/2018] [Indexed: 12/14/2022]
Abstract
Exchange protein activated by cyclic AMP (EPAC1) suppresses multiple inflammatory actions in vascular endothelial cells (VECs), partly due to its ability to induce expression of the suppressor of cytokine signalling 3 (SOCS3) gene, the protein product of which inhibits interleukin 6 (IL6) signalling through the JAK/STAT3 pathway. Here, for the first time, we use the non-cyclic nucleotide EPAC1 agonist, I942, to determine its actions on cellular EPAC1 activity and cyclic AMP-regulated gene expression in VECs. We demonstrate that I942 promotes EPAC1 and Rap1 activation in HEK293T cells and induces SOCS3 expression and suppresses IL6-stimulated JAK/STAT3 signalling in HUVECs. SOCS3 induction by I942 in HUVECs was blocked by the EPAC1 antagonist, ESI-09, and EPAC1 siRNA, but not by the broad-spectrum protein kinase A (PKA) inhibitor, H89, indicating that I942 regulates SOCS3 gene expression through EPAC1. RNA sequencing was carried out to further identify I942-regulated genes in HUVECs. This identified 425 I942-regulated genes that were also regulated by the EPAC1-selective cyclic AMP analogue, 007, and the cyclic AMP-elevating agents, forskolin and rolipram (F/R). The majority of genes identified were suppressed by I942, 007 and F/R treatment and many were involved in the control of key vascular functions, including the gene for the cell adhesion molecule, VCAM1. I942 and 007 also inhibited IL6-induced expression of VCAM1 at the protein level and blocked VCAM1-dependent monocyte adhesion to HUVECs. Overall, I942 represents the first non-cyclic nucleotide EPAC1 agonist in cells with the ability to suppress IL6 signalling and inflammatory gene expression in VECs. The novel EPAC1 ligand I942 activates cellular EPAC1 and Rap1 GTPase. I942 induces SOCS3 gene expression in vascular endothelial cells (VECs). I942 suppresses JAK/STAT3 signalling from the IL6 receptor in VECs. I942 regulates 425 novel gene targets in VECs. I942 suppresses VCAM1 expression and monocyte adhesion in VECs.
Collapse
Affiliation(s)
- Jolanta Wiejak
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh Campus, Edinburgh EH14 4AS, UK
| | - Boy van Basten
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh Campus, Edinburgh EH14 4AS, UK
| | - Urszula Luchowska-Stańska
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh Campus, Edinburgh EH14 4AS, UK
| | - Graham Hamilton
- Glasgow Polyomics, Wolfson Wohl Cancer Research Centre, Garscube Campus, University of Glasgow, Bearsden G61 1QH, UK
| | - Stephen J Yarwood
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh Campus, Edinburgh EH14 4AS, UK.
| |
Collapse
|
26
|
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
|
27
|
Schmidpeter PAM, Gao X, Uphadyay V, Rheinberger J, Nimigean CM. Ligand binding and activation properties of the purified bacterial cyclic nucleotide-gated channel SthK. J Gen Physiol 2018; 150:821-834. [PMID: 29752414 PMCID: PMC5987880 DOI: 10.1085/jgp.201812023] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 04/09/2018] [Indexed: 11/20/2022] Open
Abstract
SthK is a bacterial cyclic nucleotide–gated ion channel from Spirochaeta thermophila. By optimizing the expression and purification of SthK, Schmidpeter et al. show that cAMP and cGMP bind to the channel with similar affinity but activate it with different efficacy. Cyclic nucleotide–modulated ion channels play several essential physiological roles. They are involved in signal transduction in photoreceptors and olfactory sensory neurons as well as pacemaking activity in the heart and brain. Investigations of the molecular mechanism of their actions, including structural and electrophysiological characterization, are restricted by the availability of stable, purified protein obtained from accessible systems. Here, we establish that SthK, a cyclic nucleotide–gated (CNG) channel from Spirochaeta thermophila, is an excellent model for investigating the gating of eukaryotic CNG channels at the molecular level. The channel has high sequence similarity with its eukaryotic counterparts and was previously reported to be activated by cyclic nucleotides in patch-clamp experiments with Xenopus laevis oocytes. We optimized protein expression and purification to obtain large quantities of pure, homogeneous, and active recombinant SthK protein from Escherichia coli. A negative-stain electron microscopy (EM) single-particle analysis indicated that this channel is a promising candidate for structural studies with cryo-EM. Using radioactivity and fluorescence flux assays, as well as single-channel recordings in lipid bilayers, we show that the protein is partially activated by micromolar concentrations of cyclic adenosine monophosphate (cAMP) and that channel activity is increased by depolarization. Unlike previous studies, we find that cyclic guanosine monophosphate (cGMP) is also able to activate SthK, but with much lower efficiency than cAMP. The distinct sensitivities to different ligands resemble eukaryotic CNG and hyperpolarization-activated and cyclic nucleotide–modulated channels. Using a fluorescence binding assay, we show that cGMP and cAMP bind to SthK with similar apparent affinities, suggesting that the large difference in channel activation by cAMP or cGMP is caused by the efficacy with which each ligand promotes the conformational changes toward the open state. We conclude that the functional characteristics of SthK reported here will permit future studies to analyze ligand gating and discrimination in CNG channels.
Collapse
Affiliation(s)
| | - Xiaolong Gao
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY
| | - Vikrant Uphadyay
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY
| | - Jan Rheinberger
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY
| | - Crina M Nimigean
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY .,Department of Biochemistry, Weill Cornell Medicine, New York, NY.,Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY
| |
Collapse
|
28
|
Gao ZG, Inoue A, Jacobson KA. On the G protein-coupling selectivity of the native A 2B adenosine receptor. Biochem Pharmacol 2018; 151:201-213. [PMID: 29225130 PMCID: PMC5899946 DOI: 10.1016/j.bcp.2017.12.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 12/05/2017] [Indexed: 12/14/2022]
Abstract
A2B adenosine receptor (A2BAR) activation induces Gs-dependent cyclic AMP accumulation. However, A2BAR G protein-coupling to other signaling events, e.g. ERK1/2 and calcium, is not well documented. We explored Gi, Gq/11 and Gs coupling in 1321 N1 astrocytoma, HEK293, and T24 bladder cancer cells endogenously expressing human A2BAR, using NECA or nonnucleoside BAY60-6583 as agonist, selective Gi, Gs and Gq/11 blockers, and CRISPR/Cas9-based Gq- and Gs-null HEK293 cells. In HEK293 cells, A2BAR-mediated ERK1/2 activity occurred via both Gi and Gs, but not Gq/11. However, HEK293 cell calcium mobilization was completely blocked by Gq/11 inhibitor UBO-QIC and by Gq/11 knockout. In T24 cells, Gi was solely responsible for A2BAR-mediated ERK1/2 stimulation, and Gs suppressed ERK1/2 activity. A2BAR-mediated intracellular calcium mobilization in T24 cells was mainly via Gi, although Gs may also play a role, but Gq/11 is not involved. In 1321 N1 astrocytoma cells A2BAR activation suppressed rather than stimulated ERK1/2 activity. The ERK1/2 activity decrease was reversed by Gs downregulation using cholera toxin, but potentiated by Gi inhibitor pertussis toxin, and UBO-QIC had no effect. EPACs played an important role in A2BAR-mediated ERK1/2 signaling in all three cells. Thus, A2BAR may: couple to the same downstream pathway via different G proteins in different cell types; activate different downstream events via different G proteins in the same cell type; activate Gi and Gs, which have opposing or synergistic roles in different cell types/signaling pathways. The findings, relevant to drug discovery, address some reported controversial roles of A2BAR and could apply to signaling mechanisms in other GPCRs.
Collapse
Affiliation(s)
- Zhan-Guo Gao
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA.
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA.
| |
Collapse
|
29
|
Robichaux WG, Cheng X. Intracellular cAMP Sensor EPAC: Physiology, Pathophysiology, and Therapeutics Development. Physiol Rev 2018; 98:919-1053. [PMID: 29537337 PMCID: PMC6050347 DOI: 10.1152/physrev.00025.2017] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/05/2017] [Accepted: 09/06/2017] [Indexed: 12/13/2022] Open
Abstract
This review focuses on one family of the known cAMP receptors, the exchange proteins directly activated by cAMP (EPACs), also known as the cAMP-regulated guanine nucleotide exchange factors (cAMP-GEFs). Although EPAC proteins are fairly new additions to the growing list of cAMP effectors, and relatively "young" in the cAMP discovery timeline, the significance of an EPAC presence in different cell systems is extraordinary. The study of EPACs has considerably expanded the diversity and adaptive nature of cAMP signaling associated with numerous physiological and pathophysiological responses. This review comprehensively covers EPAC protein functions at the molecular, cellular, physiological, and pathophysiological levels; and in turn, the applications of employing EPAC-based biosensors as detection tools for dissecting cAMP signaling and the implications for targeting EPAC proteins for therapeutic development are also discussed.
Collapse
Affiliation(s)
- William G Robichaux
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center , Houston, Texas
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center , Houston, Texas
| |
Collapse
|
30
|
Kim DH, Lee J, Lee A. Visible-Light-Driven Silver-Catalyzed One-Pot Approach: A Selective Synthesis of Diaryl Sulfoxides and Diaryl Sulfones. Org Lett 2018; 20:764-767. [DOI: 10.1021/acs.orglett.7b03901] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Dong Hyuk Kim
- Department of Chemistry and ‡Department of Energy Science and Technology, Myongji University, Yongin 17058, Republic of Korea
| | - Juyoung Lee
- Department of Chemistry and ‡Department of Energy Science and Technology, Myongji University, Yongin 17058, Republic of Korea
| | - Anna Lee
- Department of Chemistry and ‡Department of Energy Science and Technology, Myongji University, Yongin 17058, Republic of Korea
| |
Collapse
|
31
|
Barker G, Parnell E, van Basten B, Buist H, Adams DR, Yarwood SJ. The Potential of a Novel Class of EPAC-Selective Agonists to Combat Cardiovascular Inflammation. J Cardiovasc Dev Dis 2017; 4:jcdd4040022. [PMID: 29367551 PMCID: PMC5753123 DOI: 10.3390/jcdd4040022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 11/23/2017] [Accepted: 11/30/2017] [Indexed: 02/07/2023] Open
Abstract
The cyclic 3′,5′-adenosine monophosphate (cAMP) sensor enzyme, EPAC1, is a candidate drug target in vascular endothelial cells (VECs) due to its ability to attenuate proinflammatory cytokine signalling normally associated with cardiovascular diseases (CVDs), including atherosclerosis. This is through the EPAC1-dependent induction of the suppressor of cytokine signalling gene, SOCS3, which targets inflammatory signalling proteins for ubiquitinylation and destruction by the proteosome. Given this important role for the EPAC1/SOCS3 signalling axis, we have used high throughput screening (HTS) to identify small molecule EPAC1 regulators and have recently isolated the first known non-cyclic nucleotide (NCN) EPAC1 agonist, I942. I942 therefore represents the first in class, isoform selective EPAC1 activator, with the potential to suppress pro-inflammatory cytokine signalling with a reduced risk of side effects associated with general cAMP-elevating agents that activate multiple response pathways. The development of augmented I942 analogues may therefore provide improved research tools to validate EPAC1 as a potential therapeutic target for the treatment of chronic inflammation associated with deadly CVDs.
Collapse
Affiliation(s)
- Graeme Barker
- Institute of Chemical Sciences, Heriot-Watt University, Edinburgh EH14 4AS, UK.
| | - Euan Parnell
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Boy van Basten
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Edinburgh EH14 4AS, UK.
| | - Hanna Buist
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Edinburgh EH14 4AS, UK.
| | - David R Adams
- Institute of Chemical Sciences, Heriot-Watt University, Edinburgh EH14 4AS, UK.
| | - Stephen J Yarwood
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Edinburgh EH14 4AS, UK.
| |
Collapse
|
32
|
Liu Z, Zhu Y, Chen H, Wang P, Mei FC, Ye N, Cheng X, Zhou J. Structure-activity relationships of 2-substituted phenyl-N-phenyl-2-oxoacetohydrazonoyl cyanides as novel antagonists of exchange proteins directly activated by cAMP (EPACs). Bioorg Med Chem Lett 2017; 27:5163-5166. [PMID: 29100797 DOI: 10.1016/j.bmcl.2017.10.056] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 10/19/2017] [Accepted: 10/22/2017] [Indexed: 11/24/2022]
Abstract
Exchange proteins directly activated by cAMP (EPACs) are critical cAMP-dependent signaling pathway mediators that play important roles in cancer, diabetes, heart failure, inflammations, infections, neurological disorders and other human diseases. EPAC specific modulators are urgently needed to explore EPAC's physiological function, mechanism of action and therapeutic applications. On the basis of a previously identified EPAC specific inhibitor hit ESI-09, herein we have designed and synthesized a novel series of 2-substituted phenyl-N-phenyl-2-oxoacetohydrazonoyl cyanides as potent EPAC inhibitors. Compound 31 (ZL0524) has been discovered as the most potent EPAC inhibitor with IC50 values of 3.6 µM and 1.2 µM against EPAC1 and EPAC2, respectively. Molecular docking of 31 onto an active EPAC2 structure predicts that 31 occupies the hydrophobic pocket in cAMP binding domain (CBD) and also opens up new space leading to the solvent region. These findings provide inspirations for discovering next generation of EPAC inhibitors.
Collapse
Affiliation(s)
- Zhiqing Liu
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Yingmin Zhu
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The University of Texas Health Science Center, 7000 Fannin St #1200, Houston, TX 77030, United States
| | - Haiying Chen
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Pingyuan Wang
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Fang C Mei
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The University of Texas Health Science Center, 7000 Fannin St #1200, Houston, TX 77030, United States
| | - Na Ye
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The University of Texas Health Science Center, 7000 Fannin St #1200, Houston, TX 77030, United States.
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States.
| |
Collapse
|
33
|
Zhu Y, Mei F, Luo P, Cheng X. A cell-based, quantitative and isoform-specific assay for exchange proteins directly activated by cAMP. Sci Rep 2017; 7:6200. [PMID: 28740152 PMCID: PMC5524698 DOI: 10.1038/s41598-017-06432-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 06/13/2017] [Indexed: 12/17/2022] Open
Abstract
Extensive functional studies of the exchange protein directly activated by cAMP (EPAC) family of signaling molecules have demonstrated that EPAC proteins play a fundamental role in several physiological and pathophysiological responses, therefore are attractive drug targets. In this report, the development of a cell-based, medium to high throughput screening assay that is capable of monitoring EPAC-mediated activation of cellular Rap1 in an isoform-specific manner is described. This assay adapts a conventional ELISA format with immobilized RalGDS-RBD as a bait to selectively capture GTP-bound active Rap1. As a result, it fills an urgent need for a cell-based EPAC assay that can be conveniently performed using microtiter plates for the discovery and/or validation of isoform-specific EPAC agonists and antagonists.
Collapse
Affiliation(s)
- Yingmin Zhu
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Health Science Center, Houston, Texas, USA
| | - Fang Mei
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Health Science Center, Houston, Texas, USA
| | - Pei Luo
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Health Science Center, Houston, Texas, USA
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Health Science Center, Houston, Texas, USA.
| |
Collapse
|
34
|
Hsu WS, Tsai MH, Barve IJ, Yellol GS, Sun CM. Synthesis of Aminofuran-Linked Benzimidazoles and Cyanopyrrole-Fused Benzimidazoles by Condition-Based Skeletal Divergence. ACS COMBINATORIAL SCIENCE 2017; 19:492-499. [PMID: 28445030 DOI: 10.1021/acscombsci.7b00052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A condition-based skeletal divergent synthesis was explored to achieve skeletal diversity in two component condensation reaction. Cyanomethyl benzimidazole was reacted with α-bromoketone under thermal conditions to furnish 2-aminofuranyl-benzimidazoles, while the same reaction afforded 3-cyano-benzopyrrolo-imidazoles under microwave irradiation. Two nonequivalent nucleophilic centers on benzimidazole moiety were manipulated elegantly by different reaction conditions to achieve the skeletal diversity.
Collapse
Affiliation(s)
- Wei-Shun Hsu
- Department
of Applied Chemistry, 1001 Ta-Hseuh Road, National Chiao-Tung University, Hsinchu 300-10, Taiwan
| | - Min-Huan Tsai
- Department
of Applied Chemistry, 1001 Ta-Hseuh Road, National Chiao-Tung University, Hsinchu 300-10, Taiwan
| | - Indrajeet J. Barve
- Department
of Applied Chemistry, 1001 Ta-Hseuh Road, National Chiao-Tung University, Hsinchu 300-10, Taiwan
| | - Gorakh S. Yellol
- Department
of Applied Chemistry, 1001 Ta-Hseuh Road, National Chiao-Tung University, Hsinchu 300-10, Taiwan
| | - Chung-Ming Sun
- Department
of Applied Chemistry, 1001 Ta-Hseuh Road, National Chiao-Tung University, Hsinchu 300-10, Taiwan
- Department
of Medicinal and Applied Chemistry, Kaohsiung Medical University, 100,
Shih-Chuan first Road, Kaohsiung 807-08, Taiwan
| |
Collapse
|
35
|
Hu B, Cilz NI, Lei S. Somatostatin depresses the excitability of subicular bursting cells: Roles of inward rectifier K + channels, KCNQ channels and Epac. Hippocampus 2017; 27:971-984. [PMID: 28558129 DOI: 10.1002/hipo.22744] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 05/08/2017] [Accepted: 05/18/2017] [Indexed: 12/17/2022]
Abstract
The hippocampus is a crucial component for cognitive and emotional processing. The subiculum provides much of the output for this structure but the modulation and function of this region is surprisingly under-studied. The neuromodulator somatostatin (SST) interacts with five subtypes of SST receptors (sst1 to sst5 ) and each of these SST receptor subtypes is coupled to Gi proteins resulting in inhibition of adenylyl cyclase (AC) and decreased level of intracellular cAMP. SST modulates many physiological functions including cognition, emotion, autonomic responses and locomotion. Whereas SST has been shown to depress neuronal excitability in the subiculum, the underlying cellular and molecular mechanisms have not yet been determined. Here, we show that SST hyperpolarized two classes of subicular neurons with a calculated EC50 of 0.1 μM. Application of SST (1 μM) induced outward holding currents by primarily activating K+ channels including the G-protein-activated inwardly-rectifying potassium channels (GIRK) and KCNQ (M) channels, although inhibition of cation channels in some cells may also be implicated. SST-elicited hyperpolarization was mediated by activation of sst2 receptors and required the function of G proteins. The SST-induced hyperpolarization resulted from decreased activity of AC and reduced levels of cAMP but did not require the activity of either PKA or PKC. Inhibition of Epac2, a guanine nucleotide exchange factor, partially blocked SST-mediated hyperpolarization of subicular neurons. Furthermore, application of SST resulted in a robust depression of subicular action potential firing and the SST-induced hyperpolarization was responsible for its inhibitory action on LTP at the CA1-subicilum synapses. Our results provide a novel cellular and molecular mechanism that may explain the roles of SST in modulation of subicular function and be relevant to SST-related physiological functions.
Collapse
Affiliation(s)
- Binqi Hu
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota 58203
| | - Nicholas I Cilz
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota 58203
| | - Saobo Lei
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota 58203
| |
Collapse
|
36
|
Richlitzki A, Latour P, Schwärzel M. Null EPAC mutants reveal a sequential order of versatile cAMP effects during Drosophila aversive odor learning. ACTA ACUST UNITED AC 2017; 24:210-215. [PMID: 28416632 PMCID: PMC5397686 DOI: 10.1101/lm.043646.116] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 03/10/2017] [Indexed: 11/25/2022]
Abstract
Here, we define a role of the cAMP intermediate EPAC in Drosophila aversive odor learning by means of null epac mutants. Complementation analysis revealed that EPAC acts downstream from the rutabaga adenylyl cyclase and in parallel to protein kinase A. By means of targeted knockdown and genetic rescue we identified mushroom body Kenyon cells (KCs) as a necessary and sufficient site of EPAC action. We provide mechanistic insights by analyzing acquisition dynamics and using the "performance increment" as a means to access the trial-based sequential organization of odor learning. Thereby we show that versatile cAMP-dependent mechanisms are engaged within a sequential order that correlate to individual trials of the training session.
Collapse
Affiliation(s)
- Antje Richlitzki
- Freie Universität Berlin, Biology/Neurobiology, D-14195 Berlin, Germany
| | - Philipp Latour
- Freie Universität Berlin, Biology/Neurobiology, D-14195 Berlin, Germany
| | - Martin Schwärzel
- Freie Universität Berlin, Biology/Neurobiology, D-14195 Berlin, Germany
| |
Collapse
|
37
|
Wang P, Liu Z, Chen H, Ye N, Cheng X, Zhou J. Exchange proteins directly activated by cAMP (EPACs): Emerging therapeutic targets. Bioorg Med Chem Lett 2017; 27:1633-1639. [PMID: 28283242 PMCID: PMC5397994 DOI: 10.1016/j.bmcl.2017.02.065] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/07/2017] [Accepted: 02/26/2017] [Indexed: 11/22/2022]
Abstract
Exchange proteins directly activated by cAMP (EPACs) are critical cAMP-dependent signaling pathway mediators. The discovery of EPAC proteins has significantly facilitated understanding on cAMP-dependent signaling pathway and efforts along this line open new avenues for developing novel therapeutics for cancer, diabetes, heart failure, inflammation, infections, neurological disorders and other human diseases. Over the past decade, important progress has been made in the identification of EPAC agonists, antagonists and their biological and pharmacological applications. In this review, we briefly summarize recently reported novel functions of EPACs and the discovery of their small molecule modulators. The challenges and future perspectives are also discussed.
Collapse
Affiliation(s)
- Pingyuan Wang
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Zhiqing Liu
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Haiying Chen
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Na Ye
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center, Houston, TX 77030, United States
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States.
| |
Collapse
|
38
|
Ye N, Zhu Y, Liu Z, Mei FC, Chen H, Wang P, Cheng X, Zhou J. Identification of novel 2-(benzo[d]isoxazol-3-yl)-2-oxo-N-phenylacetohydrazonoyl cyanide analoguesas potent EPAC antagonists. Eur J Med Chem 2017; 134:62-71. [PMID: 28399451 DOI: 10.1016/j.ejmech.2017.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/30/2017] [Accepted: 04/01/2017] [Indexed: 12/28/2022]
Abstract
Two series of novel EPAC antagonists are designed, synthesized and evaluated in an effort to develop diversified analogues based on the scaffold of the previously identified high-throughput (HTS) hit 1 (ESI-09). Further SAR studies reveal that the isoxazole ring A of 1 can tolerate chemical modifications with either introduction of flexible electron-donating substitutions or structurally restrictedly fusing with a phenyl ring, leading to identification of several more potent and diversified EPAC antagonists (e.g., 10 (NY0617), 14 (NY0460), 26 (NY0725), 32 (NY0561), and 33 (NY0562)) with low micromolar inhibitory activities. Molecular docking studies on compounds 10 and 33 indicate that these two series of compounds bind at a similar site with substantially different interactions with the EPAC proteins. The findings may serve as good starting points for the development of more potent EPAC antagonists as valuable pharmacological probes or potential drug candidates.
Collapse
Affiliation(s)
- Na Ye
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States; Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yingmin Zhu
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The University of Texas Health Science Center, Houston, TX 77030, United States
| | - Zhiqing Liu
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Fang C Mei
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The University of Texas Health Science Center, Houston, TX 77030, United States
| | - Haiying Chen
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Pingyuan Wang
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The University of Texas Health Science Center, Houston, TX 77030, United States.
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States.
| |
Collapse
|
39
|
Parnell E, McElroy SP, Wiejak J, Baillie GL, Porter A, Adams DR, Rehmann H, Smith BO, Yarwood SJ. Identification of a Novel, Small Molecule Partial Agonist for the Cyclic AMP Sensor, EPAC1. Sci Rep 2017; 7:294. [PMID: 28331191 PMCID: PMC5428521 DOI: 10.1038/s41598-017-00455-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 02/27/2017] [Indexed: 11/26/2022] Open
Abstract
Screening of a carefully selected library of 5,195 small molecules identified 34 hit compounds that interact with the regulatory cyclic nucleotide-binding domain (CNB) of the cAMP sensor, EPAC1. Two of these hits (I942 and I178) were selected for their robust and reproducible inhibitory effects within the primary screening assay. Follow-up characterisation by ligand observed nuclear magnetic resonance (NMR) revealed direct interaction of I942 and I178 with EPAC1 and EPAC2-CNBs in vitro. Moreover, in vitro guanine nucleotide exchange factor (GEF) assays revealed that I942 and, to a lesser extent, I178 had partial agonist properties towards EPAC1, leading to activation of EPAC1, in the absence of cAMP, and inhibition of GEF activity in the presence of cAMP. In contrast, there was very little agonist action of I942 towards EPAC2 or protein kinase A (PKA). To our knowledge, this is the first observation of non-cyclic-nucleotide small molecules with agonist properties towards EPAC1. Furthermore, the isoform selective agonist nature of these compounds highlights the potential for the development of small molecule tools that selectively up-regulate EPAC1 activity.
Collapse
Affiliation(s)
- Euan Parnell
- Institute of Molecular, Cellular and Systems Biology, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Stuart P McElroy
- European Screening Centre, University of Dundee, Biocity Scotland, Newhouse, ML1 5UH, UK
| | - Jolanta Wiejak
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Edinburgh Campus, Edinburgh, EH14 4AS, UK
| | - Gemma L Baillie
- European Screening Centre, University of Dundee, Biocity Scotland, Newhouse, ML1 5UH, UK
| | - Alison Porter
- European Screening Centre, University of Dundee, Biocity Scotland, Newhouse, ML1 5UH, UK
| | - David R Adams
- Institute of Chemical Sciences, Heriot-Watt University, Edinburgh Campus, Edinburgh, EH14 4AS, UK
| | - Holger Rehmann
- Department of Molecular Cancer Research, Centre of Biomedical Genetics and Cancer Genomics Centre, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Brian O Smith
- Institute of Molecular, Cellular and Systems Biology, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Stephen J Yarwood
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Edinburgh Campus, Edinburgh, EH14 4AS, UK.
| |
Collapse
|
40
|
Abstract
Protein kinase C alpha plays a major role in mediating Epac-dependent enhancement of purinergic P2X3R activity in dorsal root ganglion neurons after inflammation. Sensitization of purinergic P2X3 receptors (P2X3Rs) is a major mechanism contributing to injury-induced exaggerated pain responses. We showed in a previous study that cyclic adenosine monophosphate (cAMP)–dependent guanine nucleotide exchange factor 1 (Epac1) in rat sensory dorsal root ganglia (DRGs) is upregulated after inflammatory injury, and it plays a critical role in P2X3R sensitization by activating protein kinase C epsilon (PKCε) inside the cells. protein kinase C epsilon has been established as the major PKC isoform mediating injury-induced hyperalgesic responses. On the other hand, the role of PKCα in receptor sensitization was seldom considered. Here, we studied the participation of PKCα in Epac signaling in P2X3R-mediated hyperalgesia. The expression of both Epac1 and Epac2 and the level of cAMP in DRGs are greatly enhanced after complete Freund adjuvant (CFA)–induced inflammation. The expression of phosphorylated PKCα is also upregulated. Complete Freund adjuvant (CFA)–induced P2X3R-mediated hyperalgesia is not only blocked by Epac antagonists but also by the classical PKC isoform inhibitors, Go6976, and PKCα-siRNA. These CFA effects are mimicked by the application of the Epac agonist, 8-(4-chlorophenylthio)-2 -O-methyl-cAMP (CPT), in control rats, further confirming the involvement of Epacs. Because the application of Go6976 prior to CPT still reduces CPT-induced hyperalgesia, PKCα is downstream of Epacs to mediate the enhancement of P2X3R responses in DRGs. The pattern of translocation of PKCα inside DRG neurons in response to CPT or CFA stimulation is distinct from that of PKCε. Thus, in contrast to prevalent view, PKCα also plays an essential role in producing complex inflammation-induced receptor-mediated hyperalgesia.
Collapse
|
41
|
Quinault A, Gausseres B, Bailbe D, Chebbah N, Portha B, Movassat J, Tourrel-Cuzin C. Disrupted dynamics of F-actin and insulin granule fusion in INS-1 832/13 beta-cells exposed to glucotoxicity: partial restoration by glucagon-like peptide 1. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1401-11. [DOI: 10.1016/j.bbadis.2016.04.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 03/22/2016] [Accepted: 04/11/2016] [Indexed: 01/11/2023]
|
42
|
Abstract
Antiviral therapeutics with profiles of high potency, low resistance, panserotype, and low toxicity remain challenging, and obtaining such agents continues to be an active area of therapeutic development. Due to their unique three-dimensional structural features, spirooxindoles have been identified as privileged chemotypes for antiviral drug development. Among them, spiro-pyrazolopyridone oxindoles have been recently reported as potent inhibitors of dengue virus NS4B, leading to the discovery of an orally bioavailable preclinical candidate (R)-44 with excellent in vivo efficacy in a dengue viremia mouse model. This review highlights recent advances in the development of biologically active spirooxindoles for their antiviral potential, primarily focusing on the structure-activity relationships (SARs) and modes of action, as well as future directions to achieve more potent analogues toward a viable antiviral therapy.
Collapse
Affiliation(s)
- Na Ye
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Boulevard, Galveston, Texas 77555, United States
| | - Haiying Chen
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Boulevard, Galveston, Texas 77555, United States
| | - Eric A. Wold
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Boulevard, Galveston, Texas 77555, United States
| | - Pei-Yong Shi
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Boulevard, Galveston, Texas 77555, United States
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, 301 University Boulevard, Galveston, Texas 77555, United States
- Sealy Center for Structural Biology & Molecular Biophysics, University of Texas Medical Branch, 301 University Boulevard, Galveston, Texas 77555, United States
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Boulevard, Galveston, Texas 77555, United States
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, 301 University Boulevard, Galveston, Texas 77555, United States
| |
Collapse
|
43
|
Wild CT, Zhu Y, Na Y, Mei F, Ynalvez MA, Chen H, Cheng X, Zhou J. Functionalized N,N-Diphenylamines as Potent and Selective EPAC2 Inhibitors. ACS Med Chem Lett 2016; 7:460-4. [PMID: 27190593 DOI: 10.1021/acsmedchemlett.5b00477] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 03/28/2016] [Indexed: 02/08/2023] Open
Abstract
N,N-Diphenylamines were discovered as potent and selective EPAC2 inhibitors. A study was conducted to determine the structure-activity relationships in a series of inhibitors of which several compounds displayed submicromolar potencies. Selectivity over the related EPAC1 protein was also demonstrated. Computational modeling reveals an allosteric site that is distinct from the cAMP binding domain shared by both EPAC isoforms, providing a theory with regards to subtype selectivity.
Collapse
Affiliation(s)
- Christopher T. Wild
- Chemical
Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Yingmin Zhu
- Department
of Integrative Biology and Pharmacology and Texas Therapeutics Institute, University of Texas Health Science Center, Houston, Texas 77030, United States
| | - Ye Na
- Chemical
Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Fang Mei
- Department
of Integrative Biology and Pharmacology and Texas Therapeutics Institute, University of Texas Health Science Center, Houston, Texas 77030, United States
| | - Marcus A. Ynalvez
- 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
| | - Xiaodong Cheng
- Department
of Integrative Biology and Pharmacology and Texas Therapeutics Institute, University of Texas Health Science Center, Houston, Texas 77030, United States
| | - Jia Zhou
- Chemical
Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| |
Collapse
|
44
|
Bobin P, Varin A, Lefebvre F, Fischmeister R, Vandecasteele G, Leroy J. Calmodulin kinase II inhibition limits the pro-arrhythmic Ca2+ waves induced by cAMP-phosphodiesterase inhibitors. Cardiovasc Res 2016; 110:151-61. [PMID: 26851245 DOI: 10.1093/cvr/cvw027] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 01/22/2016] [Indexed: 01/29/2023] Open
Abstract
AIMS A major concern of using phosphodiesterase (PDE) inhibitors in heart failure is their potential to increase mortality by inducing arrhythmias. By diminishing cyclic adenosine monophosphate (cAMP) hydrolysis, they promote protein kinase A (PKA) activity under β-adrenergic receptor (β-AR) stimulation, hence enhancing Ca(2+) cycling and contraction. Yet, cAMP also activates CaMKII via PKA or the exchange protein Epac, but it remains unknown whether these pathways are involved in the pro-arrhythmic effect of PDE inhibitors. METHODS AND RESULTS Excitation-contraction coupling was investigated in isolated adult rat ventricular myocytes loaded with Fura-2 and paced at 1 Hz allowing coincident measurement of intracellular Ca(2+) and sarcomere shortening. The PDE4 inhibitor Ro 20-1724 (Ro) promoted the inotropic effects of the non-selective β-AR agonist isoprenaline (Iso) and also spontaneous diastolic Ca(2+) waves (SCWs). PDE4 inhibition potentiated RyR2 and PLB phosphorylation at specific PKA and CaMKII sites increasing sarcoplasmic reticulum (SR) Ca(2+) load and SR Ca(2+) leak measured in a 0Na(+)/0Ca(2+) solution ± tetracaine. PKA inhibition suppressed all the effects of Iso ± Ro, whereas CaMKII inhibition prevented SR Ca(2+) leak and diminished SCW incidence without affecting the inotropic effects of Ro. Inhibition of Epac2 but not Epac1 diminished the occurrence of SCWs. PDE3 inhibition with cilostamide induced an SR Ca(2+) leak, which was also blocked by CaMKII inhibition. CONCLUSION Our results show that PDE inhibitors exert inotropic effects via PKA but lead to SCWs via both PKA and CaMKII activation partly via Epac2, suggesting the potential use of CaMKII inhibitors as adjuncts to PDE inhibition to limit their pro-arrhythmic effects.
Collapse
Affiliation(s)
- Pierre Bobin
- Inserm, UMR-S 1180, Univ. Paris-Sud, Université Paris-Saclay, F-92296, Châtenay-Malabry, France
| | - Audrey Varin
- Inserm, UMR-S 1180, Univ. Paris-Sud, Université Paris-Saclay, F-92296, Châtenay-Malabry, France
| | - Florence Lefebvre
- Inserm, UMR-S 1180, Univ. Paris-Sud, Université Paris-Saclay, F-92296, Châtenay-Malabry, France
| | - Rodolphe Fischmeister
- Inserm, UMR-S 1180, Univ. Paris-Sud, Université Paris-Saclay, F-92296, Châtenay-Malabry, France
| | - Grégoire Vandecasteele
- Inserm, UMR-S 1180, Univ. Paris-Sud, Université Paris-Saclay, F-92296, Châtenay-Malabry, France
| | - Jérôme Leroy
- Inserm, UMR-S 1180, Univ. Paris-Sud, Université Paris-Saclay, F-92296, Châtenay-Malabry, France
| |
Collapse
|
45
|
Ye N, Zhu Y, Chen H, Liu Z, Mei FC, Wild C, Chen H, Cheng X, Zhou J. Structure-Activity Relationship Studies of Substituted 2-(Isoxazol-3-yl)-2-oxo-N'-phenyl-acetohydrazonoyl Cyanide Analogues: Identification of Potent Exchange Proteins Directly Activated by cAMP (EPAC) Antagonists. J Med Chem 2015; 58:6033-47. [PMID: 26151319 DOI: 10.1021/acs.jmedchem.5b00635] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Exchange proteins directly activated by cAMP (EPAC) as guanine nucleotide exchange factors mediate the effects of the pivotal second messenger cAMP, thereby regulating a wide variety of intracellular physiological and pathophysiological processes. A series of novel 2-(isoxazol-3-yl)-2-oxo-N'-phenyl-acetohydrazonoyl cyanide EPAC antagonists was synthesized and evaluated in an effort to optimize properties of the previously identified high-throughput (HTS) hit 1 (ESI-09). Structure-activity relationship (SAR) analysis led to the discovery of several more active EPAC antagonists (e.g., 22 (HJC0726), 35 (NY0123), and 47 (NY0173)) with low micromolar inhibitory activity. These inhibitors may serve as valuable pharmacological probes to facilitate our efforts in elucidating the biological functions of EPAC and developing potential novel therapeutics against human diseases. Our SAR results have also revealed that further modification at the 3-, 4-, and 5-positions of the phenyl ring as well as the 5-position of the isoxazole moiety may allow for the development of more potent EPAC antagonists.
Collapse
Affiliation(s)
- Na Ye
- †Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Yingmin Zhu
- ‡Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030, United States
| | - Haijun 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
| | - Fang C Mei
- ‡Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030, United States
| | - Christopher Wild
- †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
| | - Xiaodong Cheng
- ‡Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030, United States
| | - Jia Zhou
- †Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| |
Collapse
|
46
|
Banerjee U, Cheng X. Exchange protein directly activated by cAMP encoded by the mammalian rapgef3 gene: Structure, function and therapeutics. Gene 2015; 570:157-67. [PMID: 26119090 PMCID: PMC4556420 DOI: 10.1016/j.gene.2015.06.063] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Accepted: 06/23/2015] [Indexed: 01/08/2023]
Abstract
Mammalian exchange protein directly activated by cAMP isoform 1 (EPAC1), encoded by the RAPGEF3 gene, is one of the two-membered family of cAMP sensors that mediate the intracellular functions of cAMP by acting as guanine nucleotide exchange factors for the Ras-like Rap small GTPases. Extensive studies have revealed that EPAC1-mediated cAMP signaling is highly coordinated spatiotemporally through the formation of dynamic signalosomes by interacting with a diverse array of cellular partners. Recent functional analyses of genetically engineered mouse models further suggest that EPAC1 functions as an important stress response switch and is involved in pathophysiological conditions of cardiac stresses, chronic pain, cancer and infectious diseases. These findings, coupled with the development of EPAC specific small molecule modulators, validate EPAC1 as a promising target for therapeutic interventions. Human gene RAPGEF3 encodes for EPAC1 protein. Along with PKA, CNG & HCN, EPAC is an important cAMP sensor. Selective modulators of EPAC1 have been developed for use as pharmacological probes. Formation of EPAC1 signalosomes allows spatiotemporal control of cAMP signaling. EPAC1 is implicated in major pathophysiological conditions and is an attractive therapeutic target.
Collapse
Affiliation(s)
- Upasana Banerjee
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Health Science Center, Houston, TX 77030, United States
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Health Science Center, Houston, TX 77030, United States.
| |
Collapse
|
47
|
Schwede F, Chepurny OG, Kaufholz M, Bertinetti D, Leech CA, Cabrera O, Zhu Y, Mei F, Cheng X, Manning Fox JE, MacDonald PE, Genieser HG, Herberg FW, Holz GG. Rp-cAMPS Prodrugs Reveal the cAMP Dependence of First-Phase Glucose-Stimulated Insulin Secretion. Mol Endocrinol 2015; 29:988-1005. [PMID: 26061564 DOI: 10.1210/me.2014-1330] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
cAMP-elevating agents such as the incretin hormone glucagon-like peptide-1 potentiate glucose-stimulated insulin secretion (GSIS) from pancreatic β-cells. However, a debate has existed since the 1970s concerning whether or not cAMP signaling is essential for glucose alone to stimulate insulin secretion. Here, we report that the first-phase kinetic component of GSIS is cAMP-dependent, as revealed through the use of a novel highly membrane permeable para-acetoxybenzyl (pAB) ester prodrug that is a bioactivatable derivative of the cAMP antagonist adenosine-3',5'-cyclic monophosphorothioate, Rp-isomer (Rp-cAMPS). In dynamic perifusion assays of human or rat islets, a step-wise increase of glucose concentration leads to biphasic insulin secretion, and under these conditions, 8-bromoadenosine-3',5'-cyclic monophosphorothioate, Rp-isomer, 4-acetoxybenzyl ester (Rp-8-Br-cAMPS-pAB) inhibits first-phase GSIS by up to 80%. Surprisingly, second-phase GSIS is inhibited to a much smaller extent (≤20%). Using luciferase, fluorescence resonance energy transfer, and bioluminescence resonance energy transfer assays performed in living cells, we validate that Rp-8-Br-cAMPS-pAB does in fact block cAMP-dependent protein kinase activation. Novel effects of Rp-8-Br-cAMPS-pAB to block the activation of cAMP-regulated guanine nucleotide exchange factors (Epac1, Epac2) are also validated using genetically encoded Epac biosensors, and are independently confirmed in an in vitro Rap1 activation assay using Rp-cAMPS and Rp-8-Br-cAMPS. Thus, in addition to revealing the cAMP dependence of first-phase GSIS from human and rat islets, these findings establish a pAB-based chemistry for the synthesis of highly membrane permeable prodrug derivatives of Rp-cAMPS that act with micromolar or even nanomolar potency to inhibit cAMP signaling in living cells.
Collapse
Affiliation(s)
- Frank Schwede
- BIOLOG Life Science Institute (F.S., H.-G.G.), 28199 Bremen, Germany; Departments of Medicine (O.G.C., C.A.L., G.G.H.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Department of Biochemistry (M.K., D.B., F.W.H.), University of Kassel, 34132 Kassel, Germany; Eli Lilly and Company (O.C.), Indianapolis, Indiana 46225; Department of Integrative Biology and Pharmacology (Y.Z., F.M., X.C.), Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030; Department of Pharmacology and the Alberta Diabetes Institute (J.E.M.F., P.E.M.), University of Alberta, Edmonton, Canada AB T6G 2E1
| | - Oleg G Chepurny
- BIOLOG Life Science Institute (F.S., H.-G.G.), 28199 Bremen, Germany; Departments of Medicine (O.G.C., C.A.L., G.G.H.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Department of Biochemistry (M.K., D.B., F.W.H.), University of Kassel, 34132 Kassel, Germany; Eli Lilly and Company (O.C.), Indianapolis, Indiana 46225; Department of Integrative Biology and Pharmacology (Y.Z., F.M., X.C.), Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030; Department of Pharmacology and the Alberta Diabetes Institute (J.E.M.F., P.E.M.), University of Alberta, Edmonton, Canada AB T6G 2E1
| | - Melanie Kaufholz
- BIOLOG Life Science Institute (F.S., H.-G.G.), 28199 Bremen, Germany; Departments of Medicine (O.G.C., C.A.L., G.G.H.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Department of Biochemistry (M.K., D.B., F.W.H.), University of Kassel, 34132 Kassel, Germany; Eli Lilly and Company (O.C.), Indianapolis, Indiana 46225; Department of Integrative Biology and Pharmacology (Y.Z., F.M., X.C.), Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030; Department of Pharmacology and the Alberta Diabetes Institute (J.E.M.F., P.E.M.), University of Alberta, Edmonton, Canada AB T6G 2E1
| | - Daniela Bertinetti
- BIOLOG Life Science Institute (F.S., H.-G.G.), 28199 Bremen, Germany; Departments of Medicine (O.G.C., C.A.L., G.G.H.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Department of Biochemistry (M.K., D.B., F.W.H.), University of Kassel, 34132 Kassel, Germany; Eli Lilly and Company (O.C.), Indianapolis, Indiana 46225; Department of Integrative Biology and Pharmacology (Y.Z., F.M., X.C.), Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030; Department of Pharmacology and the Alberta Diabetes Institute (J.E.M.F., P.E.M.), University of Alberta, Edmonton, Canada AB T6G 2E1
| | - Colin A Leech
- BIOLOG Life Science Institute (F.S., H.-G.G.), 28199 Bremen, Germany; Departments of Medicine (O.G.C., C.A.L., G.G.H.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Department of Biochemistry (M.K., D.B., F.W.H.), University of Kassel, 34132 Kassel, Germany; Eli Lilly and Company (O.C.), Indianapolis, Indiana 46225; Department of Integrative Biology and Pharmacology (Y.Z., F.M., X.C.), Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030; Department of Pharmacology and the Alberta Diabetes Institute (J.E.M.F., P.E.M.), University of Alberta, Edmonton, Canada AB T6G 2E1
| | - Over Cabrera
- BIOLOG Life Science Institute (F.S., H.-G.G.), 28199 Bremen, Germany; Departments of Medicine (O.G.C., C.A.L., G.G.H.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Department of Biochemistry (M.K., D.B., F.W.H.), University of Kassel, 34132 Kassel, Germany; Eli Lilly and Company (O.C.), Indianapolis, Indiana 46225; Department of Integrative Biology and Pharmacology (Y.Z., F.M., X.C.), Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030; Department of Pharmacology and the Alberta Diabetes Institute (J.E.M.F., P.E.M.), University of Alberta, Edmonton, Canada AB T6G 2E1
| | - Yingmin Zhu
- BIOLOG Life Science Institute (F.S., H.-G.G.), 28199 Bremen, Germany; Departments of Medicine (O.G.C., C.A.L., G.G.H.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Department of Biochemistry (M.K., D.B., F.W.H.), University of Kassel, 34132 Kassel, Germany; Eli Lilly and Company (O.C.), Indianapolis, Indiana 46225; Department of Integrative Biology and Pharmacology (Y.Z., F.M., X.C.), Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030; Department of Pharmacology and the Alberta Diabetes Institute (J.E.M.F., P.E.M.), University of Alberta, Edmonton, Canada AB T6G 2E1
| | - Fang Mei
- BIOLOG Life Science Institute (F.S., H.-G.G.), 28199 Bremen, Germany; Departments of Medicine (O.G.C., C.A.L., G.G.H.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Department of Biochemistry (M.K., D.B., F.W.H.), University of Kassel, 34132 Kassel, Germany; Eli Lilly and Company (O.C.), Indianapolis, Indiana 46225; Department of Integrative Biology and Pharmacology (Y.Z., F.M., X.C.), Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030; Department of Pharmacology and the Alberta Diabetes Institute (J.E.M.F., P.E.M.), University of Alberta, Edmonton, Canada AB T6G 2E1
| | - Xiaodong Cheng
- BIOLOG Life Science Institute (F.S., H.-G.G.), 28199 Bremen, Germany; Departments of Medicine (O.G.C., C.A.L., G.G.H.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Department of Biochemistry (M.K., D.B., F.W.H.), University of Kassel, 34132 Kassel, Germany; Eli Lilly and Company (O.C.), Indianapolis, Indiana 46225; Department of Integrative Biology and Pharmacology (Y.Z., F.M., X.C.), Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030; Department of Pharmacology and the Alberta Diabetes Institute (J.E.M.F., P.E.M.), University of Alberta, Edmonton, Canada AB T6G 2E1
| | - Jocelyn E Manning Fox
- BIOLOG Life Science Institute (F.S., H.-G.G.), 28199 Bremen, Germany; Departments of Medicine (O.G.C., C.A.L., G.G.H.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Department of Biochemistry (M.K., D.B., F.W.H.), University of Kassel, 34132 Kassel, Germany; Eli Lilly and Company (O.C.), Indianapolis, Indiana 46225; Department of Integrative Biology and Pharmacology (Y.Z., F.M., X.C.), Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030; Department of Pharmacology and the Alberta Diabetes Institute (J.E.M.F., P.E.M.), University of Alberta, Edmonton, Canada AB T6G 2E1
| | - Patrick E MacDonald
- BIOLOG Life Science Institute (F.S., H.-G.G.), 28199 Bremen, Germany; Departments of Medicine (O.G.C., C.A.L., G.G.H.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Department of Biochemistry (M.K., D.B., F.W.H.), University of Kassel, 34132 Kassel, Germany; Eli Lilly and Company (O.C.), Indianapolis, Indiana 46225; Department of Integrative Biology and Pharmacology (Y.Z., F.M., X.C.), Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030; Department of Pharmacology and the Alberta Diabetes Institute (J.E.M.F., P.E.M.), University of Alberta, Edmonton, Canada AB T6G 2E1
| | - Hans-G Genieser
- BIOLOG Life Science Institute (F.S., H.-G.G.), 28199 Bremen, Germany; Departments of Medicine (O.G.C., C.A.L., G.G.H.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Department of Biochemistry (M.K., D.B., F.W.H.), University of Kassel, 34132 Kassel, Germany; Eli Lilly and Company (O.C.), Indianapolis, Indiana 46225; Department of Integrative Biology and Pharmacology (Y.Z., F.M., X.C.), Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030; Department of Pharmacology and the Alberta Diabetes Institute (J.E.M.F., P.E.M.), University of Alberta, Edmonton, Canada AB T6G 2E1
| | - Friedrich W Herberg
- BIOLOG Life Science Institute (F.S., H.-G.G.), 28199 Bremen, Germany; Departments of Medicine (O.G.C., C.A.L., G.G.H.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Department of Biochemistry (M.K., D.B., F.W.H.), University of Kassel, 34132 Kassel, Germany; Eli Lilly and Company (O.C.), Indianapolis, Indiana 46225; Department of Integrative Biology and Pharmacology (Y.Z., F.M., X.C.), Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030; Department of Pharmacology and the Alberta Diabetes Institute (J.E.M.F., P.E.M.), University of Alberta, Edmonton, Canada AB T6G 2E1
| | - George G Holz
- BIOLOG Life Science Institute (F.S., H.-G.G.), 28199 Bremen, Germany; Departments of Medicine (O.G.C., C.A.L., G.G.H.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Department of Biochemistry (M.K., D.B., F.W.H.), University of Kassel, 34132 Kassel, Germany; Eli Lilly and Company (O.C.), Indianapolis, Indiana 46225; Department of Integrative Biology and Pharmacology (Y.Z., F.M., X.C.), Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030; Department of Pharmacology and the Alberta Diabetes Institute (J.E.M.F., P.E.M.), University of Alberta, Edmonton, Canada AB T6G 2E1
| |
Collapse
|
48
|
Parnell E, Palmer TM, Yarwood SJ. The future of EPAC-targeted therapies: agonism versus antagonism. Trends Pharmacol Sci 2015; 36:203-14. [PMID: 25744542 PMCID: PMC4392396 DOI: 10.1016/j.tips.2015.02.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 02/03/2015] [Accepted: 02/05/2015] [Indexed: 02/06/2023]
Abstract
Although tractable to drug development, targeting of cAMP signalling has side effects. Selectively targeting EPAC1 and EPAC2 cAMP sensor enzymes may limit some of these off-target effects. EPAC agonists could be used to treat vascular inflammation (EPAC1) or type 2 diabetes (EPAC2). EPAC1 and EPAC2 antagonists could be used to treat heart disease.
Pharmaceutical manipulation of cAMP levels exerts beneficial effects through the regulation of the exchange protein activated by cAMP (EPAC) and protein kinase A (PKA) signalling routes. Recent attention has turned to the specific regulation of EPAC isoforms (EPAC1 and EPAC2) as a more targeted approach to cAMP-based therapies. For example, EPAC2-selective agonists could promote insulin secretion from pancreatic β cells, whereas EPAC1-selective agonists may be useful in the treatment of vascular inflammation. By contrast, EPAC1 and EPAC2 antagonists could both be useful in the treatment of heart failure. Here we discuss whether the best way forward is to design EPAC-selective agonists or antagonists and the current strategies being used to develop isoform-selective, small-molecule regulators of EPAC1 and EPAC2 activity.
Collapse
Affiliation(s)
- Euan Parnell
- Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Timothy M Palmer
- School of Pharmacy, University of Bradford, Bradford BD7 1DP, UK
| | - Stephen J Yarwood
- Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK.
| |
Collapse
|
49
|
Schwede F, Bertinetti D, Langerijs CN, Hadders MA, Wienk H, Ellenbroek JH, de Koning EJP, Bos JL, Herberg FW, Genieser HG, Janssen RAJ, Rehmann H. Structure-guided design of selective Epac1 and Epac2 agonists. PLoS Biol 2015; 13:e1002038. [PMID: 25603503 PMCID: PMC4300089 DOI: 10.1371/journal.pbio.1002038] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Accepted: 12/03/2014] [Indexed: 12/25/2022] Open
Abstract
The second messenger cAMP is known to augment glucose-induced insulin secretion. However, its downstream targets in pancreatic β-cells have not been unequivocally determined. Therefore, we designed cAMP analogues by a structure-guided approach that act as Epac2-selective agonists both in vitro and in vivo. These analogues activate Epac2 about two orders of magnitude more potently than cAMP. The high potency arises from increased affinity as well as increased maximal activation. Crystallographic studies demonstrate that this is due to unique interactions. At least one of the Epac2-specific agonists, Sp-8-BnT-cAMPS (S-220), enhances glucose-induced insulin secretion in human pancreatic cells. Selective targeting of Epac2 is thus proven possible and may be an option in diabetes treatment. cAMP is a small molecule produced by cells that activates proteins involved in a wide range of biological processes, including olfaction, pacemaker activity, regulation of gene expression, insulin secretion, and many others. In the case of insulin secretion, cAMP seems to impinge on different stages of the signalling cascade to regulate secretory activity in pancreatic β-cells. Here we have developed a chemically modified version of cAMP that specifically only activates Epac2, one of the cAMP-responsive proteins in this cascade. Furthermore, our cAMP analogue activates Epac2 more potently than cAMP itself does. We have determined several crystal structures of Epac2 in complex with cAMP analogues to help us explain the molecular basis of the observed selectivity and the strong activation potential. In addition, we were able to show that the analogue is able to potentiate glucose-induced secretion of insulin from human pancreatic islets. The principal challenge during this study was identifying and understanding small differences in the cAMP-binding domains of cAMP-regulated proteins and matching these differences with suitable modifications of the cAMP molecule. A newly developed analogue of cAMP that selectively activates Epac2 can potentiate glucose-induced insulin secretion from human pancreatic β-cells.
Collapse
Affiliation(s)
| | | | | | - Michael A. Hadders
- Department of Chemistry, Laboratory of Crystal and Structural Chemistry, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands
| | - Hans Wienk
- Department of Chemistry, NMR Spectroscopy, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands
| | | | - Eelco J. P. de Koning
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
- Hubrecht Institute/KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Johannes L. Bos
- Molecular Cancer Research and Cancer Genomics Netherlands, Center for Molecular Medicine, UMC Utrecht, Utrecht, The Netherlands
| | | | | | | | - Holger Rehmann
- Molecular Cancer Research and Cancer Genomics Netherlands, Center for Molecular Medicine, UMC Utrecht, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
50
|
Holz GG, Leech CA, Chepurny OG. New insights concerning the molecular basis for defective glucoregulation in soluble adenylyl cyclase knockout mice. Biochim Biophys Acta Mol Basis Dis 2014; 1842:2593-600. [PMID: 24980705 DOI: 10.1016/j.bbadis.2014.06.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 06/17/2014] [Accepted: 06/18/2014] [Indexed: 01/09/2023]
Abstract
Recently published findings indicate that a knockout (KO) of soluble adenylyl cyclase (sAC, also known as AC-10) gene expression in mice leads to defective glucoregulation that is characterized by reduced pancreatic insulin secretion and reduced intraperitoneal glucose tolerance. Summarized here are current concepts regarding the molecular basis for this phenotype, with special emphasis on the potential role of sAC as a determinant of glucose-stimulated insulin secretion. Highlighted is new evidence that in pancreatic beta cells, oxidative glucose metabolism stimulates mitochondrial CO₂production that in turn generates bicarbonate ion (HCO(3)(-)). Since HCO(3)(-) binds to and directly stimulates the activity of sAC, we propose that glucose-stimulated cAMP production in beta cells is mediated not simply by transmembrane adenylyl cyclases (TMACs), but also by sAC. Based on evidence that sAC is expressed in mitochondria, there exists the possibility that beta-cell glucose metabolism is linked to mitochondrial cAMP production with consequent facilitation of oxidative phosphorylation. Since sAC is also expressed in the cytoplasm, sAC catalyzed cAMP production may activate cAMP sensors such as PKA and Epac2 to control ion channel function, intracellular Ca²⁺ handling, and Ca²⁺-dependent exocytosis. Thus, we propose that the existence of sAC in beta cells provides a new and unexpected explanation for previously reported actions of glucose metabolism to stimulate cAMP production. It seems possible that alterations of sAC activity might be of importance when evaluating new strategies for the treatment of type 2 diabetes (T2DM), or when evaluating why glucose metabolism fails to stimulate insulin secretion in patients diagnosed with T2DM. This article is part of a Special Issue entitled: The role of soluble adenylyl cyclase in health and disease.
Collapse
Affiliation(s)
- George G Holz
- Department of Medicine, State University of New York (SUNY), Upstate Medical University, Syracuse, NY 13210, USA; Department of Pharmacology, State University of New York (SUNY), Upstate Medical University, Syracuse, NY 13210, USA.
| | - Colin A Leech
- Department of Medicine, State University of New York (SUNY), Upstate Medical University, Syracuse, NY 13210, USA
| | - Oleg G Chepurny
- Department of Medicine, State University of New York (SUNY), Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|