1
|
Kou F, Li XY, Feng Z, Hua J, Wu X, Gao H, Lin J, Kang D, Li A, Li J, Ding Y, Ban T, Zhang Q, Liu Z. GPR171 restrains intestinal inflammation by suppressing FABP5-mediated Th17 cell differentiation and lipid metabolism. Gut 2025:gutjnl-2024-334010. [PMID: 40074327 DOI: 10.1136/gutjnl-2024-334010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 03/02/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND GPR171 suppresses T cell immune responses involved in antitumour immunity, while its role in inflammatory bowel disease (IBD) pathogenesis remains unclear. OBJECTIVE We aimed to investigate the role of GPR171 in modulating CD4+ T cell effector functions in IBD and evaluate its therapeutic potential. DESIGN We analysed GPR171 expression in colon biopsies and peripheral blood samples from patients with IBD and assessed the impact of GPR171 on CD4+ T cell differentiation through administration of its endogenous ligand (BigLEN). We further determined the role of GPR171 in dextran sulfate sodium (DSS)-induced colitis and CD45RBhighCD4+ T-cell transfer colitis model and deciphered the underlying mechanisms using RNA sequencing (RNA-seq) and lipidomics. We developed a novel BigLEN-based Fc fusion protein (BigLEN-Fc) and evaluated its potential in preventing and treating colitis. RESULTS GPR171 was markedly increased in inflamed mucosa and CD4+ T cells of patients with IBD compared with controls. BigLEN-triggered GPR171 activation inhibited Th17 cell differentiation in vitro. GPR171 deficiency exacerbated DSS- and CD45RBhighCD4+ T cell-induced colitis in mice, characterised by increased Th17 cell responses in intestinal mucosa. Mechanistically, GPR171 deficiency promoted Th17 cell differentiation and altered lipidome profile in Th17 cells via the cAMP-pCREB-FABP5 axis. Blockage of FABP5 reduced Th17 cell differentiation in vitro and ameliorated DSS-induced colitis in Gpr171 -/- mice. Furthermore, BigLEN-mutFc administration potently mitigated colitis in mice. CONCLUSIONS GPR171 deficiency promotes Th17 cell differentiation and causes lipid metabolism perturbation, contributing to intestinal inflammation in a FABP5-dependent manner. Target therapy (eg, BigLEN-Fc) represents a novel therapeutic approach for IBD treatment.
Collapse
Affiliation(s)
- Fushun Kou
- Center for Inflammatory Bowel Disease Research and Department of Gastroenterology, Tongji University School of Medicine, Shanghai Tenth People's Hospital, Shanghai, China
| | - Xiao-Yu Li
- Center for Inflammatory Bowel Disease Research and Department of Gastroenterology, Tongji University School of Medicine, Shanghai Tenth People's Hospital, Shanghai, China
| | - Zhongsheng Feng
- Center for Inflammatory Bowel Disease Research and Department of Gastroenterology, Tongji University School of Medicine, Shanghai Tenth People's Hospital, Shanghai, China
| | - Jinghan Hua
- Center for Inflammatory Bowel Disease Research and Department of Gastroenterology, Tongji University School of Medicine, Shanghai Tenth People's Hospital, Shanghai, China
| | - Xiaohan Wu
- Center for Inflammatory Bowel Disease Research and Department of Gastroenterology, Tongji University School of Medicine, Shanghai Tenth People's Hospital, Shanghai, China
| | - Han Gao
- Center for Inflammatory Bowel Disease Research and Department of Gastroenterology, Tongji University School of Medicine, Shanghai Tenth People's Hospital, Shanghai, China
| | - Jian Lin
- Center for Inflammatory Bowel Disease Research and Department of Gastroenterology, Tongji University School of Medicine, Shanghai Tenth People's Hospital, Shanghai, China
| | - Dengfeng Kang
- Center for Inflammatory Bowel Disease Research and Department of Gastroenterology, Tongji University School of Medicine, Shanghai Tenth People's Hospital, Shanghai, China
| | - Ai Li
- Center for Inflammatory Bowel Disease Research and Department of Gastroenterology, Tongji University School of Medicine, Shanghai Tenth People's Hospital, Shanghai, China
| | - Junxiang Li
- Department of Gastroenterology, Beijing University of Chinese Medicine, Dongfang Hospital, Beijing, China
| | - Yao Ding
- Ailomics Therapeutics Co Ltd, Shanghai, China
| | - Ting Ban
- Ailomics Therapeutics Co Ltd, Shanghai, China
| | - Qing Zhang
- Ailomics Therapeutics Co Ltd, Shanghai, China
| | - Zhanju Liu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Shanghai, China
| |
Collapse
|
2
|
Xu C, Wang Y, Ni H, Yao M, Cheng L, Lin X. The role of orphan G protein-coupled receptors in pain. Heliyon 2024; 10:e28818. [PMID: 38590871 PMCID: PMC11000026 DOI: 10.1016/j.heliyon.2024.e28818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/10/2024] Open
Abstract
G protein-coupled receptors (GPCRs), which form the largest family of membrane protein receptors in humans, are highly complex signaling systems with intricate structures and dynamic conformations and locations. Among these receptors, a specific subset is referred to as orphan GPCRs (oGPCRs) and has garnered significant interest in pain research due to their role in both central and peripheral nervous system function. The diversity of GPCR functions is attributed to multiple factors, including allosteric modulators, signaling bias, oligomerization, constitutive signaling, and compartmentalized signaling. This review primarily focuses on the recent advances in oGPCR research on pain mechanisms, discussing the role of specific oGPCRs including GPR34, GPR37, GPR65, GPR83, GPR84, GPR85, GPR132, GPR151, GPR160, GPR171, GPR177, and GPR183. The orphan receptors among these receptors associated with central nervous system diseases are also briefly described. Understanding the functions of these oGPCRs can contribute not only to a deeper understanding of pain mechanisms but also offer a reference for discovering new targets for pain treatment.
Collapse
Affiliation(s)
- Chengfei Xu
- Department of Anesthesiology, The Third People's Hospital of Bengbu, Bengbu, 233000, PR China
| | - Yahui Wang
- Department of Anesthesiology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, PR China
| | - Huadong Ni
- Department of Anesthesiology and Pain Research Center, Affiliated Hospital of Jiaxing University, Jiaxing, 314000, PR China
| | - Ming Yao
- Department of Anesthesiology and Pain Research Center, Affiliated Hospital of Jiaxing University, Jiaxing, 314000, PR China
| | - Liang Cheng
- Department of Anesthesiology, The Third People's Hospital of Bengbu, Bengbu, 233000, PR China
| | - Xuewu Lin
- Department of Anesthesiology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, PR China
| |
Collapse
|
3
|
Yin G, Zheng S, Zhang W, Dong X, Qi L, Li Y. Classification of bladder cancer based on immune cell infiltration and construction of a risk prediction model for prognosis. Zhejiang Da Xue Xue Bao Yi Xue Ban 2023; 53:47-57. [PMID: 38229504 PMCID: PMC10945491 DOI: 10.3724/zdxbyxb-2023-0343] [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: 07/20/2023] [Accepted: 11/24/2023] [Indexed: 01/18/2024]
Abstract
OBJECTIVES To classify bladder cancer based on immune cell infiltration score and to construct a prognosis assessment model of patients with bladder cancer. METHODS The transcriptome data and clinical data of breast cancer patients were obtained from the The Cancer Genome Atlas (TCGA) database. Single sample gene set enrichment analysis was used to calculate the infiltration scores of 16 immune cells. The classification of breast cancer patients was achieved by unsupervised clustering, and the sensitivity of patients with different types to immunotherapy and chemotherapy was analyzed. The key modules significantly related to the infiltration of key immune cells were identified by weighted correlation network analysis (WGCNA), and the key genes in the modules were identified. A risk scoring model and a nomogram for prognosis assessment of bladder cancer patients were constructed and verified. RESULTS B cells, mast cells, neutrophils, T helper cells and tumor infiltrating lymphocytes were determined to be the key immune cells of bladder cancer. The patients were clustered into two groups (Cluster 1 ´ and Custer 2) based on immune cell infiltration scores. Compared with patients with Cluster 1 ´, patients with Cluster 2 were more likely to benefit from immunotherapy (P<0.05), and patients with Cluster 2 were more sensitive to Enbeaten, Docetaxel, Cyclopamine, and Akadixin (P<0.05). 35 genes related to key immune cells were screened out by WGCNA and 4 genes (GPR171, HOXB3, HOXB5 and HOXB6) related to the prognosis of bladder cancer were further screened by LASSO Cox regression. The areas under the ROC curve (AUC) of the bladder cancer prognosis risk scoring model based on these 4 genes to predict the 1-, 3- and 5-year survival of patients were 0.735, 0.765 and 0.799, respectively. The nomogram constructed by combining risk score and clinical parameters has high accuracy in predicting the 1-, 3-, and 5-year overall survival of bladder cancer patients. CONCLUSIONS According to the immune cell infiltration score, bladder cancer patients can be classified. Furthermore, bladder cancer prognosis risk scoring model and nomogram based on key immune cell-related genes have high accuracy in predicting the prognosis of bladder cancer patients.
Collapse
Affiliation(s)
- Guicao Yin
- Department of Urology, the Affiliated Hospital of Yangzhou University, Yangzhou 225000, Jiangsu Province, China.
| | - Shengqi Zheng
- Department of Urology, the Affiliated Hospital of Yangzhou University, Yangzhou 225000, Jiangsu Province, China
| | - Wei Zhang
- Department of Urology, the Affiliated Hospital of Yangzhou University, Yangzhou 225000, Jiangsu Province, China
| | - Xin Dong
- School of Nursing, School of Public Health, Yangzhou University, Yangzhou 225000, Jiangsu Province, China
| | - Lezhong Qi
- Department of Urology, the Affiliated Hospital of Yangzhou University, Yangzhou 225000, Jiangsu Province, China
| | - Yifan Li
- Department of Urology, the Affiliated Hospital of Yangzhou University, Yangzhou 225000, Jiangsu Province, China.
| |
Collapse
|
4
|
Kawasoe K, Watanabe T, Yoshida-Sakai N, Yamamoto Y, Kurahashi Y, Kidoguchi K, Ureshino H, Kamachi K, Fukuda-Kurahashi Y, Kimura S. A Combination of Alectinib and DNA-Demethylating Agents Synergistically Inhibits Anaplastic-Lymphoma-Kinase-Positive Anaplastic Large-Cell Lymphoma Cell Proliferation. Cancers (Basel) 2023; 15:5089. [PMID: 37894456 PMCID: PMC10605931 DOI: 10.3390/cancers15205089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 10/07/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
The recent evolution of molecular targeted therapy has improved clinical outcomes in several human malignancies. The translocation of anaplastic lymphoma kinase (ALK) was originally identified in anaplastic large-cell lymphoma (ALCL) and subsequently in non-small cell lung carcinoma (NSCLC). Since ALK fusion gene products act as a driver of carcinogenesis in both ALCL and NSCLC, several ALK tyrosine kinase inhibitors (TKIs) have been developed. Crizotinib and alectinib are first- and second-generation ALK TKIs, respectively, approved for the treatment of ALK-positive ALCL (ALK+ ALCL) and ALK+ NSCLC. Although most ALK+ NSCLC patients respond to crizotinib and alectinib, they generally relapse after several years of treatment. We previously found that DNA-demethylating agents enhanced the efficacy of ABL TKIs in chronic myeloid leukemia cells. Moreover, aberrant DNA methylation has also been observed in ALCL cells. Thus, to improve the clinical outcomes of ALK+ ALCL therapy, we investigated the synergistic efficacy of the combination of alectinib and the DNA-demethylating agent azacytidine, decitabine, or OR-2100 (an orally bioavailable decitabine derivative). As expected, the combination of alectinib and DNA-demethylating agents synergistically suppressed ALK+ ALCL cell proliferation, concomitant with DNA hypomethylation and a reduction in STAT3 (a downstream target of ALK fusion proteins) phosphorylation. The combination of alectinib and OR-2100 markedly altered gene expression in ALCL cells, including that of genes implicated in apoptotic signaling, which possibly contributed to the synergistic anti-ALCL effects of this drug combination. Therefore, alectinib and OR-2100 combination therapy has the potential to improve the outcomes of patients with ALK+ ALCL.
Collapse
Affiliation(s)
- Kazunori Kawasoe
- Department of Drug Discovery and Biomedical Sciences, Faculty of Medicine, Saga University, Saga 849-8501, Japan
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga 849-8501, Japan
| | - Tatsuro Watanabe
- Department of Drug Discovery and Biomedical Sciences, Faculty of Medicine, Saga University, Saga 849-8501, Japan
| | - Nao Yoshida-Sakai
- Department of Drug Discovery and Biomedical Sciences, Faculty of Medicine, Saga University, Saga 849-8501, Japan
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga 849-8501, Japan
| | - Yuta Yamamoto
- Department of Drug Discovery and Biomedical Sciences, Faculty of Medicine, Saga University, Saga 849-8501, Japan
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga 849-8501, Japan
| | - Yuki Kurahashi
- Department of Drug Discovery and Biomedical Sciences, Faculty of Medicine, Saga University, Saga 849-8501, Japan
- OHARA Pharmaceutical Co., Ltd., Koka 520-3403, Japan
| | - Keisuke Kidoguchi
- Department of Drug Discovery and Biomedical Sciences, Faculty of Medicine, Saga University, Saga 849-8501, Japan
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga 849-8501, Japan
| | - Hiroshi Ureshino
- Department of Drug Discovery and Biomedical Sciences, Faculty of Medicine, Saga University, Saga 849-8501, Japan
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga 849-8501, Japan
| | - Kazuharu Kamachi
- Department of Drug Discovery and Biomedical Sciences, Faculty of Medicine, Saga University, Saga 849-8501, Japan
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga 849-8501, Japan
| | - Yuki Fukuda-Kurahashi
- Department of Drug Discovery and Biomedical Sciences, Faculty of Medicine, Saga University, Saga 849-8501, Japan
- OHARA Pharmaceutical Co., Ltd., Koka 520-3403, Japan
| | - Shinya Kimura
- Department of Drug Discovery and Biomedical Sciences, Faculty of Medicine, Saga University, Saga 849-8501, Japan
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga 849-8501, Japan
| |
Collapse
|
5
|
Baalmann F, Brendler J, Butthof A, Popkova Y, Engel KM, Schiller J, Winter K, Lede V, Ricken A, Schöneberg T, Schulz A. Reduced urine volume and changed renal sphingolipid metabolism in P2ry14-deficient mice. Front Cell Dev Biol 2023; 11:1128456. [PMID: 37250906 PMCID: PMC10213973 DOI: 10.3389/fcell.2023.1128456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 04/28/2023] [Indexed: 05/31/2023] Open
Abstract
The UDP-glucose receptor P2RY14, a rhodopsin-like G protein-coupled receptor (GPCR), was previously described as receptor expressed in A-intercalated cells of the mouse kidney. Additionally, we found P2RY14 is abundantly expressed in mouse renal collecting duct principal cells of the papilla and epithelial cells lining the renal papilla. To better understand its physiological function in kidney, we took advantage of a P2ry14 reporter and gene-deficient (KO) mouse strain. Morphometric studies showed that the receptor function contributes to kidney morphology. KO mice had a broader cortex relative to the total kidney area than wild-type (WT) mice. In contrast, the area of the outer stripe of the outer medulla was larger in WT compared to KO mice. Transcriptome comparison of the papilla region of WT and KO mice revealed differences in the gene expression of extracellular matrix proteins (e.g., decorin, fibulin-1, fibulin-7) and proteins involved in sphingolipid metabolism (e.g., small subunit b of the serine palmitoyltransferase) and other related GPCRs (e.g., GPR171). Using mass spectrometry, changes in the sphingolipid composition (e.g., chain length) were detected in the renal papilla of KO mice. At the functional level, we found that KO mice had a reduced urine volume but an unchanged glomerular filtration rate under normal chow and salt diets. Our study revealed P2ry14 as a functionally important GPCR in collecting duct principal cells and cells lining the renal papilla and the possible involvement of P2ry14 in nephroprotection by regulation of decorin.
Collapse
Affiliation(s)
- Fabian Baalmann
- Rudolf Schönheimer Institute of Biochemistry, Faculty of Medicine, Leipzig University, Leipzig, Germany
| | - Jana Brendler
- Institute of Anatomy, Faculty of Medicine, Leipzig University, Leipzig, Germany
| | - Anne Butthof
- Rudolf Schönheimer Institute of Biochemistry, Faculty of Medicine, Leipzig University, Leipzig, Germany
| | - Yulia Popkova
- Institute of Medical Physics and Biophysics, Faculty of Medicine, Leipzig University, Leipzig, Germany
| | - Kathrin M. Engel
- Institute of Medical Physics and Biophysics, Faculty of Medicine, Leipzig University, Leipzig, Germany
| | - Jürgen Schiller
- Institute of Medical Physics and Biophysics, Faculty of Medicine, Leipzig University, Leipzig, Germany
| | - Karsten Winter
- Institute of Anatomy, Faculty of Medicine, Leipzig University, Leipzig, Germany
| | - Vera Lede
- Rudolf Schönheimer Institute of Biochemistry, Faculty of Medicine, Leipzig University, Leipzig, Germany
| | - Albert Ricken
- Institute of Anatomy, Faculty of Medicine, Leipzig University, Leipzig, Germany
| | - Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Faculty of Medicine, Leipzig University, Leipzig, Germany
| | - Angela Schulz
- Rudolf Schönheimer Institute of Biochemistry, Faculty of Medicine, Leipzig University, Leipzig, Germany
| |
Collapse
|
6
|
McDermott MV, Ram A, Mattoon MT, Haderlie EE, Raddatz MC, Thomason MK, Bobeck EN. A small molecule ligand for the novel pain target, GPR171, produces minimal reward in mice. Pharmacol Biochem Behav 2023; 224:173543. [PMID: 36933620 PMCID: PMC11472835 DOI: 10.1016/j.pbb.2023.173543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 03/10/2023] [Accepted: 03/12/2023] [Indexed: 03/19/2023]
Abstract
ProSAAS is one of the most abundant proteins in the brain and is processed into several smaller peptides. One of which, BigLEN, is an endogenous ligand for the G protein-coupled receptor, GPR171. Recent work in rodent models has shown that a small-molecule ligand for GPR171, MS15203, increases morphine antinociception and is effective in lessening chronic pain. While these studies provide evidence for GPR171 as a possible pain target, its abuse liability has not yet been assessed and was evaluated in the current study. We first mapped the distribution of GPR171 and ProSAAS throughout the reward circuit of the brain using immunohistochemistry and showed that GPR171 and ProSAAS are localized in the hippocampus, basolateral amygdala, nucleus accumbens, prefrontal cortex. In the major dopaminergic structure, the ventral tegmental area (VTA), GPR171 appeared to be primarily localized in dopamine neurons while ProSAAS is outside of dopamine neurons. Next, MS15203 was administered to mice with or without morphine, and VTA slices were stained for the immediate early gene c-Fos as a marker of neuronal activation. Quantification of c-Fos-positive cells revealed no statistical difference between MS15203 and saline, suggesting that MS15203 does not increase VTA activation and dopamine release. The results of a conditioned place preference experiment showed that treatment with MS15203 produced no place preference indicating a lack of reward-related behavior. Taken together this data provides evidence that the novel pain therapeutic, MS15203, has minimal reward liability. Therefore, GPR171 deserves further exploration as a pain target. SIGNIFICANCE STATEMENT: MS15203, a drug that activates the receptor GPR171, was previously shown to increase morphine analgesia. The authors use in vivo and histological techniques to show that it fails to activate the rodent reward circuitry, providing support for the continued exploration of MS15203 as a novel pain drug, and GPR171 a novel pain target.
Collapse
Affiliation(s)
- Max V McDermott
- Dept. of Biology, Utah State University, 5305 Old Main Hill BNR117, Logan, UT 84322-5305, United States of America; Interdisciplinary Neuroscience Program, Utah State University, 5305 Old Main Hill BNR117, Logan, UT 84322-5305, United States of America
| | - Akila Ram
- Dept. of Biology, Utah State University, 5305 Old Main Hill BNR117, Logan, UT 84322-5305, United States of America
| | - Matthew T Mattoon
- Dept. of Biology, Utah State University, 5305 Old Main Hill BNR117, Logan, UT 84322-5305, United States of America
| | - Emmaline E Haderlie
- Dept. of Biology, Utah State University, 5305 Old Main Hill BNR117, Logan, UT 84322-5305, United States of America
| | - Megan C Raddatz
- Dept. of Biology, Utah State University, 5305 Old Main Hill BNR117, Logan, UT 84322-5305, United States of America; Interdisciplinary Neuroscience Program, Utah State University, 5305 Old Main Hill BNR117, Logan, UT 84322-5305, United States of America
| | - Madi K Thomason
- Dept. of Biology, Utah State University, 5305 Old Main Hill BNR117, Logan, UT 84322-5305, United States of America
| | - Erin N Bobeck
- Dept. of Biology, Utah State University, 5305 Old Main Hill BNR117, Logan, UT 84322-5305, United States of America; Interdisciplinary Neuroscience Program, Utah State University, 5305 Old Main Hill BNR117, Logan, UT 84322-5305, United States of America.
| |
Collapse
|
7
|
Afrose L, McDermott MV, Bhuiyan AI, Pathak SK, Bobeck EN. GPR171 activation regulates morphine tolerance but not withdrawal in a test-dependent manner in mice. Behav Pharmacol 2022; 33:442-451. [PMID: 35942845 PMCID: PMC9477863 DOI: 10.1097/fbp.0000000000000692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/07/2022] [Indexed: 11/26/2022]
Abstract
A newly deorphanized G protein-coupled receptor, GPR171, is found to be highly expressed within the periaqueductal gray, a pain-modulating region in the brain. Our recent research has shown that a GPR171 agonist increases morphine antinociception in male mice and opioid signaling in vitro . The objective of this study was to evaluate the effects of combination treatment in females as well as whether chronic treatment can be used without exacerbating morphine-induced tolerance and withdrawal in female and male mice. Our results demonstrate that activation of GPR171 with an agonist attenuates morphine tolerance in both female and male mice on the tail-flick test, but not the hotplate test. Importantly, the GPR171 agonist in combination with morphine does not exacerbate morphine-induced tolerance and withdrawal during long-term morphine treatment. Taken together, these data suggest that the GPR171 agonist may be combined with morphine to maintain antinociception while reducing the dose of morphine and therefore reducing side effects and abuse liability. The outcome of this study is clearly an important step toward understanding the functional interactions between opioid receptors and GPR171 and developing safer therapeutics for long-term pain management.
Collapse
Affiliation(s)
| | - Max V. McDermott
- Department of Biology
- Interdisciplinary Neuroscience Program, Utah State University, Logan, Utah
| | - Ashif I. Bhuiyan
- Chemistry and Biochemistry Department, Queens College of The City University of New York, Flushing, New York
- Chemistry Doctoral Program
| | - Sanjai K. Pathak
- Chemistry and Biochemistry Department, Queens College of The City University of New York, Flushing, New York
- Chemistry Doctoral Program
- Biochemistry Doctoral Program, The Graduate Center of The City University of New York, New York
| | - Erin N. Bobeck
- Department of Biology
- Interdisciplinary Neuroscience Program, Utah State University, Logan, Utah
| |
Collapse
|
8
|
Li Y, Yu X, Ma Y, Su J, Li Y, Zhu S, Bai T, Wei Q, Becker B, Ding Z, Wang K, Tian Y, Wang J. Neural signatures of default mode network in major depression disorder after electroconvulsive therapy. Cereb Cortex 2022; 33:3840-3852. [PMID: 36089839 DOI: 10.1093/cercor/bhac311] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 06/17/2022] [Accepted: 07/08/2022] [Indexed: 11/12/2022] Open
Abstract
Functional abnormalities of default mode network (DMN) have been well documented in major depressive disorder (MDD). However, the association of DMN functional reorganization with antidepressant treatment and gene expression is unclear. Moreover, whether the functional interactions of DMN could predict treatment efficacy is also unknown. Here, we investigated the link of treatment response with functional alterations of DMN and gene expression with a comparably large sample including 46 individuals with MDD before and after electroconvulsive therapy (ECT) and 46 age- and sex-matched healthy controls. Static and dynamic functional connectivity (dFC) analyses showed increased intrinsic/static but decreased dynamic functional couplings of inter- and intra-subsystems and between nodes of DMN. The changes of static functional connections of DMN were spatially correlated with brain gene expression profiles. Moreover, static and dFC of the DMN before treatment as features could predict depressive symptom improvement following ECT. Taken together, these results shed light on the underlying neural and genetic basis of antidepressant effect of ECT and the intrinsic functional connectivity of DMN have the potential to serve as prognostic biomarkers to guide accurate personalized treatment.
Collapse
Affiliation(s)
- Yuanyuan Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China.,School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 625014, China
| | - Xiaohui Yu
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China
| | - Yingzi Ma
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China
| | - Jing Su
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China
| | - Yue Li
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 625014, China
| | - Shunli Zhu
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 625014, China
| | - Tongjian Bai
- Department of Neurology, The First Hospital of Anhui Medical University, Hefei 230022, China
| | - Qiang Wei
- Department of Neurology, The First Hospital of Anhui Medical University, Hefei 230022, China
| | - Benjamin Becker
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 625014, China
| | - Zhiyong Ding
- Medical Imaging Department, Maternal and Child Health-care Hospital of Qujing, Qujing 655000, China
| | - Kai Wang
- Department of Neurology, The First Hospital of Anhui Medical University, Hefei 230022, China.,Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei 230022, China.,Anhui Medical University, School of Mental Health and Psychological Sciences, Hefei 230022, China.,Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei 230022, China.,Anhui Province Clinical Research Center for Neurological Disease, Hefei 230022, China
| | - Yanghua Tian
- Department of Neurology, The First Hospital of Anhui Medical University, Hefei 230022, China.,Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China.,Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei 230022, China.,Anhui Medical University, School of Mental Health and Psychological Sciences, Hefei 230022, China.,Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei 230022, China.,Anhui Province Clinical Research Center for Neurological Disease, Hefei 230022, China.,Department of Psychology and Sleep Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Jiaojian Wang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China.,Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan 650500, China
| |
Collapse
|
9
|
Aryal DK, Rodriguiz RM, Nguyen NL, Pease MW, Morgan DJ, Pintar J, Fricker LD, Wetsel WC. Mice lacking proSAAS display alterations in emotion, consummatory behavior and circadian entrainment. GENES, BRAIN, AND BEHAVIOR 2022; 21:e12827. [PMID: 35878875 PMCID: PMC9444949 DOI: 10.1111/gbb.12827] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 07/07/2022] [Accepted: 07/07/2022] [Indexed: 11/30/2022]
Abstract
ProSAAS is a neuroendocrine protein that is cleaved by neuropeptide-processing enzymes into more than a dozen products including the bigLEN and PEN peptides, which bind and activate the receptors GPR171 and GPR83, respectively. Previous studies have suggested that proSAAS-derived peptides are involved in physiological functions that include body weight regulation, circadian rhythms and anxiety-like behavior. In the present study, we find that proSAAS knockout mice display robust anxiety-like behaviors in the open field, light-dark emergence and elevated zero maze tests. These mutant mice also show a reduction in cued fear and an impairment in fear-potentiated startle, indicating an important role for proSAAS-derived peptides in emotional behaviors. ProSAAS knockout mice exhibit reduced water consumption and urine production relative to wild-type controls. No differences in food consumption and overall energy expenditure were observed between the genotypes. However, the respiratory exchange ratio was elevated in the mutants during the light portion of the light-dark cycle, indicating decreased fat metabolism during this period. While proSAAS knockout mice show normal circadian patterns of activity, even upon long-term exposure to constant darkness, they were unable to shift their circadian clock upon exposure to a light pulse. Taken together, these results show that proSAAS-derived peptides modulate a wide range of behaviors including emotion, metabolism and the regulation of the circadian clock.
Collapse
Affiliation(s)
- Dipendra K. Aryal
- Department of Psychiatry and Behavioral SciencesDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Ramona M. Rodriguiz
- Department of Psychiatry and Behavioral SciencesDuke University Medical CenterDurhamNorth CarolinaUSA,Department of Psychiatry and Behavioral Sciences, Mouse Behavioral and Neuroendocrine Analysis Core FacilityDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Ngoc Lien Nguyen
- Department of Psychiatry and Behavioral SciencesDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Matthew W. Pease
- Department of Psychiatry and Behavioral SciencesDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Daniel J. Morgan
- Department of Anesthesiology and Perioperative Medicine, Pennsylvania StateUniversity College of MedicineHersheyPennsylvaniaUSA
| | - John Pintar
- Department of Neuroscience and Cell BiologyRutgers Robert Wood Johnson Medical SchoolPiscatawayNew JerseyUSA
| | - Lloyd D. Fricker
- Departments of Molecular Pharmacology and NeuroscienceAlbert Einstein College of MedicineBronxNew YorkUSA
| | - William C. Wetsel
- Department of Psychiatry and Behavioral Sciences, Mouse Behavioral and Neuroendocrine Analysis Core FacilityDuke University Medical CenterDurhamNorth CarolinaUSA,Department of Cell BiologyDuke University Medical CenterDurhamNorth CarolinaUSA,Department of NeurobiologyDuke University Medical CenterDurhamNCUSA
| |
Collapse
|
10
|
Hunger dampens a nucleus accumbens circuit to drive persistent food seeking. Curr Biol 2022; 32:1689-1702.e4. [DOI: 10.1016/j.cub.2022.02.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/11/2022] [Accepted: 02/09/2022] [Indexed: 02/07/2023]
|
11
|
Fujiwara Y, Torphy RJ, Sun Y, Miller EN, Ho F, Borcherding N, Wu T, Torres RM, Zhang W, Schulick RD, Zhu Y. The GPR171 pathway suppresses T cell activation and limits antitumor immunity. Nat Commun 2021; 12:5857. [PMID: 34615877 PMCID: PMC8494883 DOI: 10.1038/s41467-021-26135-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 09/15/2021] [Indexed: 12/17/2022] Open
Abstract
The recently identified G-protein-coupled receptor GPR171 and its ligand BigLEN are thought to regulate food uptake and anxiety. Though GPR171 is commonly used as a T cell signature gene in transcriptomic studies, its potential role in T cell immunity has not been explored. Here we show that GPR171 is transcribed in T cells and its protein expression is induced upon antigen stimulation. The neuropeptide ligand BigLEN interacts with GPR171 to suppress T cell receptor-mediated signalling pathways and to inhibit T cell proliferation. Loss of GPR171 in T cells leads to hyperactivity to antigen stimulation and GPR171 knockout mice exhibit enhanced antitumor immunity. Blockade of GPR171 signalling by an antagonist promotes antitumor T cell immunity and improves immune checkpoint blockade therapies. Together, our study identifies the GPR171/BigLEN axis as a T cell checkpoint pathway that can be modulated for cancer immunotherapy.
Collapse
Affiliation(s)
- Yuki Fujiwara
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Robert J Torphy
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Yi Sun
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Emily N Miller
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Felix Ho
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Nicholas Borcherding
- Department of Pathology and Immunology, Washington University, St. Louis, MO, 63110, USA
| | - Tuoqi Wu
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Raul M Torres
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Weizhou Zhang
- Department of Pathology, University of Florida, Gainesville, FL, 32610, USA
| | - Richard D Schulick
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Yuwen Zhu
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
12
|
Fakira AK, Lueptow LM, Trimbake NA, Devi LA. PEN Receptor GPR83 in Anxiety-Like Behaviors: Differential Regulation in Global vs Amygdalar Knockdown. Front Neurosci 2021; 15:675769. [PMID: 34512237 PMCID: PMC8427670 DOI: 10.3389/fnins.2021.675769] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 07/26/2021] [Indexed: 11/13/2022] Open
Abstract
Anxiety disorders are prevalent across the United States and result in a large personal and societal burden. Currently, numerous therapeutic and pharmaceutical treatment options exist. However, drugs to classical receptor targets have shown limited efficacy and often come with unpleasant side effects, highlighting the need to identify novel targets involved in the etiology and treatment of anxiety disorders. GPR83, a recently deorphanized receptor activated by the abundant neuropeptide PEN, has also been identified as a glucocorticoid regulated receptor (and named GIR) suggesting that this receptor may be involved in stress-responses that underlie anxiety. Consistent with this, GPR83 null mice have been found to be resistant to stress-induced anxiety. However, studies examining the role of GPR83 within specific brain regions or potential sex differences have been lacking. In this study, we investigate anxiety-related behaviors in male and female mice with global knockout and following local GPR83 knockdown in female mice. We find that a global knockdown of GPR83 has minimal impact on anxiety-like behaviors in female mice and a decrease in anxiety-related behaviors in male mice. In contrast, a local GPR83 knockdown in the basolateral amygdala leads to more anxiety-related behaviors in female mice. Local GPR83 knockdown in the central amygdala or nucleus accumbens (NAc) showed no significant effect on anxiety-related behaviors. Finally, dexamethasone administration leads to a significant decrease in receptor expression in the amygdala and NAc of female mice. Together, our studies uncover a significant, but divergent role for GPR83 in different brain regions in the regulation of anxiety-related behaviors, which is furthermore dependent on sex.
Collapse
Affiliation(s)
| | | | | | - Lakshmi A. Devi
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
13
|
Matthews DG, Caruso M, Alcazar Magana A, Wright KM, Maier CS, Stevens JF, Gray NE, Quinn JF, Soumyanath A. Caffeoylquinic Acids in Centella asiatica Reverse Cognitive Deficits in Male 5XFAD Alzheimer's Disease Model Mice. Nutrients 2020; 12:E3488. [PMID: 33202902 PMCID: PMC7698091 DOI: 10.3390/nu12113488] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/04/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023] Open
Abstract
Centella asiatica (CA) is an edible plant and a popular botanical dietary supplement. It is reputed, in Ayurveda, to mitigate age-related cognitive decline. There is a considerable body of preclinical literature supporting CA's ability to improve learning and memory. This study evaluated the contribution of CA's triterpenes (TT), widely considered its active compounds, and caffeoylquinic acids (CQA) to the cognitive effects of CA water extract (CAW) in 5XFAD mice, a model of Alzheimer's disease. 5XFAD mice were fed a control diet alone, or one containing 1% CAW or compound groups (TT, CQA, or TT + CQA) equivalent to their content in 1% CAW. Wild-type (WT) littermates received the control diet. Conditioned fear response (CFR) was evaluated after 4.5 weeks. Female 5XFAD controls showed no deficit in CFR compared to WT females, nor any effects from treatment. In males, CFR of 5XFAD controls was attenuated compared to WT littermates (p = 0.005). 5XFAD males receiving CQA or TT + CQA had significantly improved CFR (p < 0.05) compared to 5XFAD male controls. CFR did not differ between 5XFAD males receiving treatment diets and WT males. These data confirm a role for CQA in CAW's cognitive effects.
Collapse
Affiliation(s)
- Donald G. Matthews
- Department of Neurology, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA; (D.G.M.); (M.C.); (K.M.W.); (N.E.G.); (J.F.Q.)
| | - Maya Caruso
- Department of Neurology, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA; (D.G.M.); (M.C.); (K.M.W.); (N.E.G.); (J.F.Q.)
| | - Armando Alcazar Magana
- Department of Chemistry, Oregon State University, Corvallis, OR 97331, USA; (A.A.M.); (C.S.M.)
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA;
| | - Kirsten M. Wright
- Department of Neurology, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA; (D.G.M.); (M.C.); (K.M.W.); (N.E.G.); (J.F.Q.)
| | - Claudia S. Maier
- Department of Chemistry, Oregon State University, Corvallis, OR 97331, USA; (A.A.M.); (C.S.M.)
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA;
| | - Jan F. Stevens
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA;
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| | - Nora E. Gray
- Department of Neurology, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA; (D.G.M.); (M.C.); (K.M.W.); (N.E.G.); (J.F.Q.)
| | - Joseph F. Quinn
- Department of Neurology, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA; (D.G.M.); (M.C.); (K.M.W.); (N.E.G.); (J.F.Q.)
- Parkinson’s Disease Research Education and Clinical Care Center, Veterans’ Administration Portland Health Care System, Portland, OR 97239, USA
| | - Amala Soumyanath
- Department of Neurology, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA; (D.G.M.); (M.C.); (K.M.W.); (N.E.G.); (J.F.Q.)
| |
Collapse
|
14
|
Liu R, Wei P, Keller C, Orefice NS, Shi Y, Li Z, Huang J, Cui Y, Frost DC, Han S, Cross TWL, Rey FE, Li L. Integrated Label-Free and 10-Plex DiLeu Isobaric Tag Quantitative Methods for Profiling Changes in the Mouse Hypothalamic Neuropeptidome and Proteome: Assessment of the Impact of the Gut Microbiome. Anal Chem 2020; 92:14021-14030. [PMID: 32926775 DOI: 10.1021/acs.analchem.0c02939] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Gut microbiota can regulate host physiological and pathological status through gut-brain communications or pathways. However, the impact of the gut microbiome on neuropeptides and proteins involved in regulating brain functions and behaviors is still not clearly understood. To address the problem, integrated label-free and 10-plex DiLeu isobaric tag-based quantitative methods were implemented to compare the profiling of neuropeptides and proteins in the hypothalamus of germ-free (GF)- vs conventionally raised (ConvR)-mice. A total of 2943 endogenous peptides from 63 neuropeptide precursors and 3971 proteins in the mouse hypothalamus were identified. Among these 368 significantly changed peptides (fold changes over 1.5 and a p-value of <0.05), 73.6% of the peptides showed higher levels in GF-mice than in ConvR-mice, and 26.4% of the peptides had higher levels in ConvR-mice than in GF-mice. These peptides were mainly from secretogranin-2, phosphatidylethanolamine-binding protein-1, ProSAAS, and proenkephalin-A. A quantitative proteomic analysis employing DiLeu isobaric tags revealed that 282 proteins were significantly up- or down-regulated (fold changes over 1.2 and a p-value of <0.05) among the 3277 quantified proteins. These neuropeptides and proteins were mainly involved in regulating behaviors, transmitter release, signaling pathways, and synapses. Interestingly, pathways including long-term potentiation, long-term depression, and circadian entrainment were involved. In the present study, a combined label-free and 10-plex DiLeu-based quantitative method enabled a comprehensive profiling of gut microbiome-induced dynamic changes of neuropeptides and proteins in the hypothalamus, suggesting that the gut microbiome might mediate a range of behavioral changes, brain development, and learning and memory through these neuropeptides and proteins.
Collapse
Affiliation(s)
- Rui Liu
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States.,School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China.,Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing 210023, P. R. China.,Jiangsu Key Laboratory of Research and Development in Marine Bio-resource Pharmaceutics, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Pingli Wei
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Caitlin Keller
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Nicola Salvatore Orefice
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States.,Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Yatao Shi
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Zihui Li
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Junfeng Huang
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Yusi Cui
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Dustin C Frost
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Shuying Han
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States.,School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China.,Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing 210023, P. R. China
| | - Tzu-Wen L Cross
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States.,Cardiovascular Research Center, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Federico E Rey
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Lingjun Li
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States.,Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| |
Collapse
|
15
|
Davenport AP, Scully CCG, de Graaf C, Brown AJH, Maguire JJ. Advances in therapeutic peptides targeting G protein-coupled receptors. Nat Rev Drug Discov 2020; 19:389-413. [PMID: 32494050 DOI: 10.1038/s41573-020-0062-z] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2020] [Indexed: 02/06/2023]
Abstract
Dysregulation of peptide-activated pathways causes a range of diseases, fostering the discovery and clinical development of peptide drugs. Many endogenous peptides activate G protein-coupled receptors (GPCRs) - nearly 50 GPCR peptide drugs have been approved to date, most of them for metabolic disease or oncology, and more than 10 potentially first-in-class peptide therapeutics are in the pipeline. The majority of existing peptide therapeutics are agonists, which reflects the currently dominant strategy of modifying the endogenous peptide sequence of ligands for peptide-binding GPCRs. Increasingly, novel strategies are being employed to develop both agonists and antagonists, to both introduce chemical novelty and improve drug-like properties. Pharmacodynamic improvements are evolving to allow biasing ligands to activate specific downstream signalling pathways, in order to optimize efficacy and reduce side effects. In pharmacokinetics, modifications that increase plasma half-life have been revolutionary. Here, we discuss the current status of the peptide drugs targeting GPCRs, with a focus on evolving strategies to improve pharmacokinetic and pharmacodynamic properties.
Collapse
Affiliation(s)
- Anthony P Davenport
- Experimental Medicine and Immunotherapeutics, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK.
| | | | | | | | - Janet J Maguire
- Experimental Medicine and Immunotherapeutics, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| |
Collapse
|
16
|
McDermott MV, Afrose L, Gomes I, Devi LA, Bobeck EN. Opioid-Induced Signaling and Antinociception Are Modulated by the Recently Deorphanized Receptor, GPR171. J Pharmacol Exp Ther 2019; 371:56-62. [PMID: 31308196 PMCID: PMC6750184 DOI: 10.1124/jpet.119.259242] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 07/10/2019] [Indexed: 12/13/2022] Open
Abstract
ProSAAS is one of the most widely expressed proteins throughout the brain and was recently found to be upregulated in chronic fibromyalgia patients. BigLEN is a neuropeptide that is derived from ProSAAS and was recently discovered to be the endogenous ligand for the orphan G protein-coupled receptor GPR171. Although BigLEN-GPR171 has been found to play a role in feeding and anxiety behaviors, it has not yet been explored in pain and opioid modulation. The purpose of this study was to evaluate this novel neuropeptide-receptor system in opioid-induced antinociception. We found that GPR171 is expressed in GABAergic neurons within the periaqueductal gray, which is a key brain area involved in pain modulation and opioid functions. We also found that, although the GPR171 agonist and antagonist do not have nociceptive effects on their own, they oppositely regulate morphine-induced antinociception with the agonist enhancing and antagonist reducing antinociception. Lastly, we showed that the GPR171 antagonist or receptor knockdown decreases signaling by the mu-opioid receptor, but not the delta-opioid receptor. Taken together, these results suggest that antagonism of the GPR171 receptor reduces mu opioid receptor signaling and morphine-induced antinociception, whereas the GPR171 agonist enhances morphine antinociception, suggesting that GPR171 may be a novel target toward the development of pain therapeutics. SIGNIFICANCE STATEMENT: GPR171 is a recently deorphanized receptor that is expressed within the periaqueductal gray and can regulate mu opioid receptor signaling and antinociception. This research may contribute to the development of new therapeutics to treat pain.
Collapse
Affiliation(s)
- Max V McDermott
- Department of Biology, Utah State University, Logan, Utah (M.V.M., L.A., E.N.B.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (I.G., L.A.D.)
| | - Leela Afrose
- Department of Biology, Utah State University, Logan, Utah (M.V.M., L.A., E.N.B.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (I.G., L.A.D.)
| | - Ivone Gomes
- Department of Biology, Utah State University, Logan, Utah (M.V.M., L.A., E.N.B.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (I.G., L.A.D.)
| | - Lakshmi A Devi
- Department of Biology, Utah State University, Logan, Utah (M.V.M., L.A., E.N.B.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (I.G., L.A.D.)
| | - Erin N Bobeck
- Department of Biology, Utah State University, Logan, Utah (M.V.M., L.A., E.N.B.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (I.G., L.A.D.)
| |
Collapse
|
17
|
HCN2 Channels in Cholinergic Interneurons of Nucleus Accumbens Shell Regulate Depressive Behaviors. Neuron 2019; 101:662-672.e5. [PMID: 30638901 DOI: 10.1016/j.neuron.2018.12.018] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 09/17/2018] [Accepted: 12/12/2018] [Indexed: 12/13/2022]
Abstract
Cholinergic interneurons (ChIs) in the nucleus accumbens (NAc) have been implicated in drug addiction, reward, and mood disorders. However, the physiological role of ChIs in depression has not been characterized. Here, we show that the tonic firing rate of ChIs in NAc shell is reduced in chronic stress mouse models and in a genetic mouse model of depression. Chemogenetic inhibition of NAc ChIs renders naive mice susceptible to stress, whereas enhancement of ChI activity reverses depressive phenotypes. As a component of the molecular mechanism, we found that the expression and function of the hyperpolarization-activated cyclic nucleotide-gated channel 2 (HCN2) are decreased in ChIs of NAc shell in depressed mice. Overexpression of HCN2 channels in ChIs enhances cell activity and is sufficient to rescue depressive phenotypes. These data suggest that enhancement of HCN2 channel activity in NAc ChIs is a feasible approach for the development of a new class of antidepressants.
Collapse
|
18
|
From Synapse to Function: A Perspective on the Role of Neuroproteomics in Elucidating Mechanisms of Drug Addiction. Proteomes 2018; 6:proteomes6040050. [PMID: 30544849 PMCID: PMC6315754 DOI: 10.3390/proteomes6040050] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 12/05/2018] [Accepted: 12/07/2018] [Indexed: 12/18/2022] Open
Abstract
Drug addiction is a complex disorder driven by dysregulation in molecular signaling across several different brain regions. Limited therapeutic options currently exist for treating drug addiction and related psychiatric disorders in clinical populations, largely due to our incomplete understanding of the molecular pathways that influence addiction pathology. Recent work provides strong evidence that addiction-related behaviors emerge from the convergence of many subtle changes in molecular signaling networks that include neuropeptides (neuropeptidome), protein-protein interactions (interactome) and post-translational modifications such as protein phosphorylation (phosphoproteome). Advancements in mass spectrometry methodology are well positioned to identify these novel molecular underpinnings of addiction and further translate these findings into druggable targets for therapeutic development. In this review, we provide a general perspective of the utility of novel mass spectrometry-based approaches for addressing critical questions in addiction neuroscience, highlighting recent innovative studies that exemplify how functional assessments of the neuroproteome can provide insight into the mechanisms of drug addiction.
Collapse
|
19
|
Abstract
Neuropeptides are the largest class of intercellular signaling molecules, contributing to a wide variety of physiological processes. Neuropeptide receptors are therapeutic targets for a broad range of drugs, including medications to treat pain, addiction, sleep disorders, and nausea. In addition to >100 peptides with known functions, many peptides have been identified in mammalian brain for which the cognate receptors have not been identified. Similarly, dozens of "orphan" G protein-coupled receptors have been identified in the mammalian genome. While it would seem straightforward to match the orphan peptides and receptors, this is not always easily accomplished. In this review we focus on peptides named PEN and big LEN, which are among the most abundant neuropeptides in mouse brain, and their recently identified receptors: GPR83 and GPR171. These receptors are co-expressed in some brain regions and are able to interact. Because PEN and big LEN are produced from the same precursor protein and co-secreted, the interaction of GPR83 and GPR171 is physiologically relevant. In addition to interactions of these two peptides/receptors, PEN and LEN are co-localized with neuropeptide Y and Agouti-related peptide in neurons that regulate feeding. In this review, using these peptide receptors as an example, we highlight the multiple modes of regulation of receptors and present the emerging view that neuropeptides function combinatorially to generate a network of signaling messages. The complexity of neuropeptides, receptors, and their signaling pathways is important to consider both in the initial deorphanization of peptides and receptors, and in the subsequent development of therapeutic applications.
Collapse
Affiliation(s)
- Lloyd D Fricker
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, United States
| | - Lakshmi A Devi
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
20
|
The G protein-coupled receptors deorphanization landscape. Biochem Pharmacol 2018; 153:62-74. [PMID: 29454621 DOI: 10.1016/j.bcp.2018.02.016] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 02/13/2018] [Indexed: 12/14/2022]
Abstract
G protein-coupled receptors (GPCRs) are usually highlighted as being both the largest family of membrane proteins and the most productive source of drug targets. However, most of the GPCRs are understudied and hence cannot be used immediately for innovative therapeutic strategies. Besides, there are still around 100 orphan receptors, with no described endogenous ligand and no clearly defined function. The race to discover new ligands for these elusive receptors seems to be less intense than before. Here, we present an update of the various strategies employed to assign a function to these receptors and to discover new ligands. We focus on the recent advances in the identification of endogenous ligands with a detailed description of newly deorphanized receptors. Replication being a key parameter in these endeavors, we also discuss the latest controversies about problematic ligand-receptor pairings. In this context, we propose several recommendations in order to strengthen the reporting of new ligand-receptor pairs.
Collapse
|
21
|
Fricker LD. Carboxypeptidase E and the Identification of Novel Neuropeptides as Potential Therapeutic Targets. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 82:85-102. [PMID: 29413529 DOI: 10.1016/bs.apha.2017.09.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Peptides and small molecules that bind to peptide receptors are important classes of drugs that are used for a wide variety of different applications. The search for novel neuropeptides traditionally involved a time-consuming approach to purify each peptide to homogeneity and determine its amino acid sequence. The discovery in the 1980s of enkephalin convertase/carboxypeptidase E (CPE), and the observation that this enzyme was involved in the production of nearly every known neuropeptide led to the idea for a one-step affinity purification of CPE substrates. This approach was successfully used to isolate hundreds of known neuropeptides in mouse brain, as well as over a dozen novel peptides. Some of the novel peptides found using this approach are among the most abundant peptides present in brain, but had not been previously identified by traditional approaches. Recently, receptors for two of the novel peptides have been identified, confirming their role as neuropeptides that function in cell-cell signaling. Small molecules that bind to one of these receptors have been developed and found to significantly reduce food intake and anxiety-like behavior in an animal model. This review describes the entire project, from discovery of CPE to the novel peptides and their receptors.
Collapse
Affiliation(s)
- Lloyd D Fricker
- Albert Einstein College of Medicine, Bronx, NY, United States.
| |
Collapse
|