1
|
Lin W, Xu L, Li G, Tortorella MD. Molecular gene signature of circulating stromal/stem cells. J Hum Genet 2025; 70:275-280. [PMID: 40069498 DOI: 10.1038/s10038-025-01322-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/02/2025] [Accepted: 02/06/2025] [Indexed: 03/14/2025]
Abstract
The human skeleton is renewed and regenerated throughout life, by a cellular process known as bone remodeling. Stem cells are clono-genic cells that are capable of differentiation into multiple mature cell types (multipotency), and simultaneously replenishing stem cell pool (self-renewal), which allows them to sustain tissue development and maintenance. Circulating mesenchymal stromal/stem cells (MSCs), are mobile adult stem cells with specific gene expression profiling, as well as enhanced mitochondrial remodeling as a promising source for personalized cell and gene therapy. A global LGR5-associated genetic interaction network highlights the functional organization and molecular phenotype of circulating MSCs.
Collapse
Affiliation(s)
- Weiping Lin
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, SAR, China
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Liangliang Xu
- Key Laboratory of Orthopaedics & Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangdong, Guangzhou, China.
| | - Gang Li
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, SAR, China.
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| | - Micky Daniel Tortorella
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, SAR, China.
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.
| |
Collapse
|
2
|
Xue C, Liu Z. Unraveling the proton-sensing mechanisms of G protein-coupled receptors: Insights from cryogenic electron microscopy studies. Mol Cell 2025; 85:1479-1481. [PMID: 40250408 DOI: 10.1016/j.molcel.2025.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 03/24/2025] [Accepted: 03/24/2025] [Indexed: 04/20/2025]
Abstract
In this issue, Guo et al.1 and Chen et al.2 resolved the cryo-EM structures of G protein-coupled receptor (GPR) 4 and GPR68, unveiling that histidine protonation initiates conformational rearrangements and dictates G protein coupling bias, illuminating pH-sensing mechanisms.
Collapse
Affiliation(s)
- Chenyang Xue
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China; Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Zhongmin Liu
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China; Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| |
Collapse
|
3
|
Chen LN, Zhou H, Xi K, Cheng S, Liu Y, Fu Y, Ma X, Xu P, Ji SY, Wang WW, Shen DD, Zhang H, Shen Q, Chai R, Zhang M, Yang L, Han F, Mao C, Cai X, Zhang Y. Proton perception and activation of a proton-sensing GPCR. Mol Cell 2025; 85:1640-1657.e8. [PMID: 40215960 DOI: 10.1016/j.molcel.2025.02.030] [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: 10/21/2024] [Revised: 01/22/2025] [Accepted: 02/28/2025] [Indexed: 04/20/2025]
Abstract
Maintaining pH at cellular, tissular, and systemic levels is essential for human health. Proton-sensing GPCRs regulate physiological and pathological processes by sensing the extracellular acidity. However, the molecular mechanism of proton sensing and activation of these receptors remains elusive. Here, we present cryoelectron microscopy (cryo-EM) structures of human GPR4, a prototypical proton-sensing GPCR, in its inactive and active states. Our studies reveal that three extracellular histidine residues are crucial for proton sensing of human GPR4. The binding of protons induces substantial conformational changes in GPR4's ECLs, particularly in ECL2, which transforms from a helix-loop to a β-turn-β configuration. This transformation leads to the rearrangements of H-bond network and hydrophobic packing, relayed by non-canonical motifs to accommodate G proteins. Furthermore, the antagonist NE52-QQ57 hinders human GPR4 activation by preventing hydrophobic stacking rearrangement. Our findings provide a molecular framework for understanding the activation mechanism of a human proton-sensing GPCR, aiding future drug discovery.
Collapse
Affiliation(s)
- Li-Nan Chen
- Department of Pathology of Sir Run Run Shaw Hospital, Department of Pharmacology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Hui Zhou
- Department of Pathology of Sir Run Run Shaw Hospital, Department of Pharmacology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Kun Xi
- Department of Pathology of Sir Run Run Shaw Hospital, Department of Pharmacology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Shizhuo Cheng
- Department of Pathology of Sir Run Run Shaw Hospital, Department of Pharmacology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yongfeng Liu
- Department of Pathology of Sir Run Run Shaw Hospital, Department of Pharmacology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yifan Fu
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xiangyu Ma
- Department of Pathology of Sir Run Run Shaw Hospital, Department of Pharmacology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China; State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, School of Medicine, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing 210096, China
| | - Ping Xu
- Department of Pathology of Sir Run Run Shaw Hospital, Department of Pharmacology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China; Institute of Cytology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Su-Yu Ji
- Department of Pathology of Sir Run Run Shaw Hospital, Department of Pharmacology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Wei-Wei Wang
- Department of Pathology of Sir Run Run Shaw Hospital, Department of Pharmacology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Dan-Dan Shen
- Department of Pathology of Sir Run Run Shaw Hospital, Department of Pharmacology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Huibing Zhang
- Department of Pathology of Sir Run Run Shaw Hospital, Department of Pharmacology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Qingya Shen
- Department of Pathology of Sir Run Run Shaw Hospital, Department of Pharmacology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Renjie Chai
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, School of Medicine, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing 210096, China
| | - Min Zhang
- College of Computer Science and Technology, Zhejiang University, Hangzhou 310027, China
| | - Lin Yang
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Feng Han
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.
| | - Chunyou Mao
- Department of Pathology of Sir Run Run Shaw Hospital, Department of Pharmacology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China; Center for Structural Pharmacology and Therapeutics Development, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China.
| | - Xiujun Cai
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou 310016, China; Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Hangzhou 310016, China.
| | - Yan Zhang
- Department of Pathology of Sir Run Run Shaw Hospital, Department of Pharmacology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
4
|
Guo L, Zhu K, Zhong YN, Gao M, Liu J, Qi Z, Liu Z, Rong N, Zhang M, Li D, Zhang Q, Yang G, Zhang X, Zhang M, Ding N, Ping YQ, Yang Z, Xiao P, Xia M, Yu X, Gaole A, Sun JP, Yang F. Structural basis and biased signaling of proton sensation by GPCRs mediated by extracellular histidine rearrangement. Mol Cell 2025; 85:1658-1673.e7. [PMID: 40215959 DOI: 10.1016/j.molcel.2025.03.018] [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: 09/11/2024] [Revised: 01/27/2025] [Accepted: 03/21/2025] [Indexed: 04/20/2025]
Abstract
Proton sensing by G protein-coupled receptors (GPCRs) is crucial in many life activities. However, its underlying mechanism remains unclear. Here, we report 8 cryoelectron microscopy (cryo-EM) structures of human GPR4 and GPR68 at different pH values and in complex with Gs or Gq trimers or in apo state. Structural inspection, structure-based pKa calculations, and mutational and computational analyses revealed that protonation of two conserved extracellular histidines induced polar network formation and other conformational changes to tether 7-transmembrane (TM7) to second extracellular loop (ECL2), and these changes constitute the central mechanisms of proton-induced activation of GPR4 and GPR68. Unexpectedly, proton sensation by specific extracellular histidine determined biased G protein coupling of GPR4. Moreover, GPR68's additional pH-sensing H842.67 enhances its function in a more acidic optimal pH range. The propagation path connecting proton-sensing histidines to the toggle switch was characterized. Collectively, we provide structural insights into the proton sensing, activation, and downstream effector coupling mechanisms of proton-sensing GPCRs.
Collapse
MESH Headings
- Histidine/metabolism
- Histidine/chemistry
- Histidine/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/chemistry
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/ultrastructure
- Humans
- Protons
- Cryoelectron Microscopy
- Signal Transduction
- Hydrogen-Ion Concentration
- HEK293 Cells
- GTP-Binding Protein alpha Subunits, Gq-G11/metabolism
- GTP-Binding Protein alpha Subunits, Gq-G11/genetics
- GTP-Binding Protein alpha Subunits, Gq-G11/chemistry
- GTP-Binding Protein alpha Subunits, Gs/metabolism
- GTP-Binding Protein alpha Subunits, Gs/genetics
- GTP-Binding Protein alpha Subunits, Gs/chemistry
- Protein Conformation
- Protein Binding
Collapse
Affiliation(s)
- Lulu Guo
- Advanced Medical Research Institute, Cheeloo College of Medicine, The Second Hospital Shandong University, Jinan, China
| | - Kongkai Zhu
- Advanced Medical Research Institute, Cheeloo College of Medicine, The Second Hospital Shandong University, Jinan, China
| | - Ya-Ni Zhong
- Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, China
| | - Mingxin Gao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Junyan Liu
- Department of Orthopaedics, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Zhimin Qi
- School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Zili Liu
- Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, China
| | - Naikang Rong
- Advanced Medical Research Institute, Cheeloo College of Medicine, The Second Hospital Shandong University, Jinan, China
| | - Minghui Zhang
- Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, China
| | - Dongfang Li
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qiyue Zhang
- Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, China
| | - Gongming Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xinxin Zhang
- Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, China
| | - Mingyue Zhang
- Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, China
| | - Ning Ding
- Department of Anesthesiology, Shandong Provincial Key Medical and Health Laboratory of Intensive Care Rehabilitation, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yu-Qi Ping
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Zhao Yang
- Advanced Medical Research Institute, Cheeloo College of Medicine, The Second Hospital Shandong University, Jinan, China; Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Peng Xiao
- Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, China
| | - Ming Xia
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China.
| | - Xiao Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, China.
| | - Alatan Gaole
- School of Life Sciences, Inner Mongolia University, Hohhot, China.
| | - Jin-Peng Sun
- Advanced Medical Research Institute, Cheeloo College of Medicine, The Second Hospital Shandong University, Jinan, China; Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, China; Beijing Key Laboratory of Cardiovascular Receptors Research.
| | - Fan Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, China; State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine.
| |
Collapse
|
5
|
Yue X, Peng L, Liu S, Zhang B, Zhang X, Chang H, Pei Y, Li X, Liu J, Shui W, Wu L, Xu H, Liu ZJ, Hua T. Structural basis of stepwise proton sensing-mediated GPCR activation. Cell Res 2025:10.1038/s41422-025-01092-w. [PMID: 40211064 DOI: 10.1038/s41422-025-01092-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 02/23/2025] [Indexed: 04/12/2025] Open
Abstract
The regulation of pH homeostasis is crucial in many biological processes vital for survival, growth, and function of life. The pH-sensing G protein-coupled receptors (GPCRs), including GPR4, GPR65 and GPR68, play a pivotal role in detecting changes in extracellular proton concentrations, impacting both physiological and pathological states. However, comprehensive understanding of the proton sensing mechanism is still elusive. Here, we determined the cryo-electron microscopy structures of GPR4 and GPR65 in various activation states across different pH levels, coupled with Gs, Gq or G13 proteins, as well as a small molecule NE52-QQ57-bound inactive GPR4 structure. These structures reveal the dynamic nature of the extracellular loop 2 and its signature conformations in different receptor states, and disclose the proton sensing mechanism mediated by networks of extracellular histidine and carboxylic acid residues. Notably, we unexpectedly captured partially active intermediate states of both GPR4-Gs and GPR4-Gq complexes, and identified a unique allosteric binding site for NE52-QQ57 in GPR4. By integrating prior investigations with our structural analysis and mutagenesis data, we propose a detailed atomic model for stepwise proton sensation and GPCR activation. These insights may pave the way for the development of selective ligands and targeted therapeutic interventions for pH sensing-relevant diseases.
Collapse
Affiliation(s)
- Xiaolei Yue
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Li Peng
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Shenhui Liu
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Bingjie Zhang
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Xiaodan Zhang
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Hao Chang
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yuan Pei
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Xiaoting Li
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Junlin Liu
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Wenqing Shui
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Lijie Wu
- iHuman Institute, ShanghaiTech University, Shanghai, China.
| | - Huji Xu
- Department of Rheumatology and Immunology, Changzheng Hospital, Second Military Medical University, Shanghai, China.
- School of Clinical Medicine, Tsinghua University, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Tsinghua University, Beijing, China.
| | - Zhi-Jie Liu
- iHuman Institute, ShanghaiTech University, Shanghai, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| | - Tian Hua
- iHuman Institute, ShanghaiTech University, Shanghai, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
6
|
Luo W, Zhou J, Liang F, Chou X, Peng Z, Tan W, Yu Z, Wan H. The GPR4 antagonist NE 52-QQ57 prevents ox-LDL-induced cellular senescence by promoting the expression of SIRT1. Genes Genomics 2025:10.1007/s13258-024-01610-x. [PMID: 40208484 DOI: 10.1007/s13258-024-01610-x] [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: 05/14/2024] [Accepted: 09/12/2024] [Indexed: 04/11/2025]
Abstract
BACKGROUND Cell senescence-associated endothelia dysfunction is a vital point in the pathological progression of atherosclerosis (AS). G-protein coupled receptor 4 (GPR4) is a proton-sensing receptor involved in developing endothelial dysfunction. OBJECTIVE In this study, we investigated the protective role of NE 52-QQ57, a GPR4 inhibitor in endothelial cell senescence induced using an oxidized low-density lipoprotein (ox-LDL). We also unravel the underlying molecular mechanism of NE 52-QQ57 as a therapeutic agent. METHODS Endothelial cell senescence model was established using human aortic endothelial cells (HAECs) stimulated with ox-LDL. The expression levels of GPR4, p53, p16, and sirtuin1 (SIRT1) were evaluated using real-time PCR and western blot assays. ROS production was determined using dihydroethidium (DHE) staining. Further, interleukin-6 (IL-6) and monocyte chemotactic protein 1 (MCP-1) secretion and expression were determined using ELISA and real-time PCR analysis, respectively. Finally, β-galactosidase (SA-β-Gal) staining associated with cell senescence, telomerase activity, and cell cycle assay were used to determine the state of cell senescence. RESULTS Firstly, GPR4 was found to be upregulated in the ox-LDL-stimulated HAECs. We also identified elevated ROS, IL-6, and MCP-1 levels induced by ox-LDL and significantly abrogated by NE 52-QQ57 treatment. Second, a reversal in SA-β-Gal activity, telomerase activity, and G0/G1 proportion, with an upregulation in p53 and p16 expressions was observed on NE 52-QQ57 treatment in the ox-LDL induced model. Lastly, the decreased expression level of SIRT1 was extremely elevated by NE 52-QQ57. Notably, the inhibitory effect of NE 52-QQ57 against ox-LDL-induced cell senescence was abolished by the SIRT1 inhibitor EX-527. CONCLUSION The GPR4 antagonist NE 52-QQ57 might prevent cellular senescence by promoting the expression of SIRT1.
Collapse
Affiliation(s)
- Wei Luo
- Department of Cardiology, The First Affiliated Hospital of University of South China, No. 69, Chuanshan Road, Shigu District, Hengyang, 421001, Hunan, China
| | - Jiming Zhou
- Department of Cardiology, The First Affiliated Hospital of University of South China, No. 69, Chuanshan Road, Shigu District, Hengyang, 421001, Hunan, China
| | - Feng Liang
- Department of Anesthesiology, The First Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, China
| | - Xianghui Chou
- Department of Cardiology, The First Affiliated Hospital of University of South China, No. 69, Chuanshan Road, Shigu District, Hengyang, 421001, Hunan, China
| | - Zhengliang Peng
- Department of Emergency, The First Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, China
| | - Weihua Tan
- Department of Emergency, The First Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, China
| | - Ziying Yu
- Department of Emergency, The First Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, China
| | - Huan Wan
- Department of Cardiology, The First Affiliated Hospital of University of South China, No. 69, Chuanshan Road, Shigu District, Hengyang, 421001, Hunan, China.
| |
Collapse
|
7
|
Xu J, Shepard BD, Pluznick JL. Roles of sensory receptors in non-sensory organs: the kidney and beyond. Nat Rev Nephrol 2025; 21:253-263. [PMID: 39753689 PMCID: PMC11929601 DOI: 10.1038/s41581-024-00917-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2024] [Indexed: 02/02/2025]
Abstract
Olfactory receptors (ORs), taste receptors and opsins are well-known for their pivotal roles in mediating the senses of smell, taste and sight, respectively. However, in the past two decades, research has shown that these sensory receptors also regulate physiological processes in a variety of non-sensory tissues. Although ORs, taste receptors and opsins have all been shown to have physiological roles beyond their traditional locations, most work in the kidney has focused on ORs. To date, renal ORs have been shown to have roles in blood pressure regulation (OLFR78 and OLFR558) and glucose homeostasis (OLFR1393). However, sensory receptors remain drastically understudied outside of traditional sensory systems, in part because of inherent challenges in studying these receptors. Increased knowledge of the physiological and pathophysiological roles of sensory receptors has the potential to substantially improve understanding of the function of numerous organs and systems, including the kidney. In addition, most sensory receptors are G protein-coupled receptors, which are considered to be the most druggable class of proteins, and thus could potentially be exploited as future therapeutic targets.
Collapse
Affiliation(s)
- Jiaojiao Xu
- Department of Physiology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Blythe D Shepard
- Department of Human Science, Georgetown University, Washington, DC, USA
| | - Jennifer L Pluznick
- Department of Physiology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
8
|
Karki P, Ke Y, Zhang CO, Promnares K, Li Y, Williams CH, Hong CC, Birukov KG, Birukova AA. Inhibition of proton sensor GPR68 suppresses endothelial dysfunction and acute lung injury caused by Staphylococcus aureus bacterial particles. FASEB J 2025; 39:e70333. [PMID: 39907683 DOI: 10.1096/fj.202401947r] [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: 08/19/2024] [Revised: 12/21/2024] [Accepted: 01/08/2025] [Indexed: 02/06/2025]
Abstract
Lung bacterial infections, including hospital-acquired pneumonia, remain a serious problem for public health. Endothelial cell (EC) exposure to heat-killed Staphylococcus aureus (HKSA) represents a clinical scenario of high titers of killed bacterial particles present in the host after antibiotic therapy, which triggers inflammatory cascades, cytokine storms, and EC dysfunction leading to acute lung injury (ALI). GPR68 is a member of the proton-sensing G protein-coupled receptor family. Acting as a pH sensor, GPR68 becomes activated upon pH reduction and contributes to pathologic cell responses by activating ER stress and unfolded protein response. This study investigated the role of GPR68 in HKSA-induced EC dysfunction and HKSA-induced ALI. HKSA robustly increased GPR68 mRNA levels in human pulmonary EC and directly stimulated GPR68 activity. A selective GPR68 small molecule inhibitor, OGM-8345, attenuated HKSA-induced EC permeability and protected cell junction integrity. OGM-8345 inhibited HKSA-induced activation of inflammatory genes TNF-α, IL-6, IL-8, IL-1β, and CXCL5 and decreased cytokine secretion by HKSA-challenged EC. Co-treatment with the GPR68 activator Ogerin or medium acidification to pH 6.5 augmented HKSA-induced EC dysfunction, which was rescued by OGM-8345. Intratracheal HKSA injection increased vascular leak and lung inflammation in mice which were monitored by lung Evans blue extravasation, increased cell and protein count in bronchoalveolar lavage, and mRNA expression of inflammatory genes. ALI and barrier dysfunction was attenuated by OGM-8345. We show for the first time the role of GPR68 in mediating HKSA-induced lung injury and the strong potential for OGM-8345 as a therapeutic treatment of bacterial pathogen-induced ALI associated with tissue acidification.
Collapse
Affiliation(s)
- Pratap Karki
- Division of Pulmonary and Critical Care, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yunbo Ke
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Chen-Ou Zhang
- Division of Pulmonary and Critical Care, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Kamoltip Promnares
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yue Li
- Division of Pulmonary and Critical Care, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Charles H Williams
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Charles C Hong
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Konstantin G Birukov
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Anna A Birukova
- Division of Pulmonary and Critical Care, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
9
|
Foti F, Schuler C, Ruiz PA, Perren L, Malagola E, de Vallière C, Seuwen K, Hausmann M, Rogler G. The Simultaneous Deletion of pH-Sensing Receptors GPR4 and OGR1 (GPR68) Ameliorates Colitis with Additive Effects on Multiple Parameters of Inflammation. Int J Mol Sci 2025; 26:1552. [PMID: 40004018 PMCID: PMC11855581 DOI: 10.3390/ijms26041552] [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: 12/02/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 02/27/2025] Open
Abstract
G protein-coupled receptors (GPRs), including pro-inflammatory GPR4 and ovarian cancer GPR1 (OGR1/GPR68), are involved in the pH sensing of the extracellular space and have been implicated in inflammatory bowel disease (IBD). Previous data show that a loss of GPR4 or OGR1 independently is associated with reduced intestinal inflammation in mouse models of experimental colitis. In the present manuscript, we investigated the impact of the simultaneous loss of GPR4 and OGR1 in animal models of IBD. To study the effects of combined loss of Gpr4 Ogr1 in IBD we used the well-established acute dextran sodium sulfate (DSS) and spontaneous Il10-/- murine colitis models. Disease severity was assessed using multiple clinical scores (e.g., body weight loss, disease activity score, murine endoscopic index of colitis severity (MEICS) and histological analyses). Real-time quantitative polymerase chain reaction (qPCR), Western blot, and flow cytometry were used to investigate changes in pro-inflammatory cytokines expression and immune cells infiltration. We found that a combined loss of GPR4 and OGR1 significantly reduces colon inflammation in IBD relative to single deficiencies as evidenced by reduced body weight loss, disease score, CD4/CD8 ratio, and Il1β, Il6, and Tnf in the colon. Similarly, in the II10 deficiency model, the inflammation was significantly ameliorated upon the simultaneous deletion of GPR4 and OGR1, evidenced by a reduction in the MEICS score, colon length, Tnf and Il1β measurements, and a decrease in the number of macrophages in the colon, as compared to single deletions. Importantly, hydroxyproline levels were decreased close to baseline in Il10-/- × Gpr4-/- × Ogr1-/- mice. Our findings demonstrate that the simultaneous loss of GRP4 and OGR1 functions exerts an additive effect on multiple parameters associated with colonic inflammation. These results further reinforce the hypothesis that chronic inflammatory acidosis is a driver of fibrosis and is dependent on GPR4 and OGR1 signaling. The inhibition of both GPR4 and OGR1 by pH-sensing receptor modulators may constitute as a potential therapeutic option for IBD, as both pH-sensing receptors appear to sustain inflammation by acting on complementary pro-inflammatory pathways.
Collapse
|
10
|
Huang Y, Liang J, Wu H, Chen P, Xiao A, Guan BO. Microscale insight into the proton concentration during electrolytic reaction via an optical microfiber: potential for microcurrent monitoring by a dielectric probe. LIGHT, SCIENCE & APPLICATIONS 2025; 14:73. [PMID: 39915465 PMCID: PMC11802907 DOI: 10.1038/s41377-025-01770-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 01/25/2025] [Accepted: 01/27/2025] [Indexed: 02/09/2025]
Abstract
Local microcurrent monitoring is of great significance for biological and battery systems, yet it poses a formidable challenge. The current measurement techniques rely on electromagnetic materials which inevitably introduce interference to the system under examination. To address this issue, a promising approach based on a dielectric fiber-optic sensor is demonstrated. The microfiber is capable of detecting microcurrent through monitoring the localized proton concentration signal with a pH resolution of 0.0052 pH units. By sensing the refractive index variation surrounding the sensor induced by the interaction between local proton concentration changes and oxidizer-treated microfiber surface through the evanescent field, this sensing mechanism effectively avoids the interference of the electromagnetic material on the performance of the tested system. This sensor exhibits a limit of detection for microcurrent of 1 μA. The sensing region is a microfiber with a diameter of 8.8 μm. It can get invaluable information that cannot be obtained through conventional electrochemical methods. Examples include photocurrent attenuation in photogenerated carrier materials during illumination, electrical activation in nerve cells, and fluctuations in the efficiency of electrical energy generation during battery discharge. This approach provides a powerful complement to electrochemical methods for the elucidation of microscale reaction mechanisms. The information provided by the prepared dielectric fiber-optic sensor will shed more light on proton kinetics and electrochemical and electrobiological mechanisms, which may fill an important gap in the current bioelectricity and battery monitoring methods.
Collapse
Affiliation(s)
- Yunyun Huang
- Guangdong Provincial Key Laboratory of Optical Fiber Sensing and Communications, Institute of Photonics Technology, Jinan University, Guangzhou, 511143, China.
- College of Physics & Optoelectronic Engineering, Jinan University, Guangzhou, 510632, China.
| | - Jiaxuan Liang
- Guangdong Provincial Key Laboratory of Optical Fiber Sensing and Communications, Institute of Photonics Technology, Jinan University, Guangzhou, 511143, China
- College of Physics & Optoelectronic Engineering, Jinan University, Guangzhou, 510632, China
| | - Haotian Wu
- Guangdong Provincial Key Laboratory of Optical Fiber Sensing and Communications, Institute of Photonics Technology, Jinan University, Guangzhou, 511143, China
- College of Physics & Optoelectronic Engineering, Jinan University, Guangzhou, 510632, China
| | - Pengwei Chen
- Guangdong Provincial Key Laboratory of Optical Fiber Sensing and Communications, Institute of Photonics Technology, Jinan University, Guangzhou, 511143, China
- College of Physics & Optoelectronic Engineering, Jinan University, Guangzhou, 510632, China
| | - Aoxiang Xiao
- Guangdong Provincial Key Laboratory of Optical Fiber Sensing and Communications, Institute of Photonics Technology, Jinan University, Guangzhou, 511143, China
- College of Physics & Optoelectronic Engineering, Jinan University, Guangzhou, 510632, China
| | - Bai-Ou Guan
- Guangdong Provincial Key Laboratory of Optical Fiber Sensing and Communications, Institute of Photonics Technology, Jinan University, Guangzhou, 511143, China.
- College of Physics & Optoelectronic Engineering, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
11
|
Wen X, Shang P, Chen H, Guo L, Rong N, Jiang X, Li X, Liu J, Yang G, Zhang J, Zhu K, Meng Q, He X, Wang Z, Liu Z, Cheng H, Zheng Y, Zhang B, Pang J, Liu Z, Xiao P, Chen Y, Liu L, Luo F, Yu X, Yi F, Zhang P, Yang F, Deng C, Sun JP. Evolutionary study and structural basis of proton sensing by Mus GPR4 and Xenopus GPR4. Cell 2025; 188:653-670.e24. [PMID: 39753131 DOI: 10.1016/j.cell.2024.12.001] [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: 05/10/2024] [Revised: 11/20/2024] [Accepted: 12/02/2024] [Indexed: 02/09/2025]
Abstract
Animals have evolved pH-sensing membrane receptors, such as G-protein-coupled receptor 4 (GPR4), to monitor pH changes related to their physiology and generate adaptive reactions. However, the evolutionary trajectory and structural mechanism of proton sensing by GPR4 remain unresolved. Here, we observed a positive correlation between the optimal pH of GPR4 activity and the blood pH range across different species. By solving 7-cryoelectron microscopy (cryo-EM) structures of Xenopus tropicalis GPR4 (xtGPR4) and Mus musculus GPR4 (mmGPR4) under varying pH conditions, we identified that protonation of HECL2-45.47 and H7.36 enabled polar network establishment and tighter association between the extracellular loop 2 (ECL2) and 7 transmembrane (7TM) domain, as well as a conserved propagating path, which are common mechanisms underlying protonation-induced GPR4 activation across different species. Moreover, protonation of distinct extracellular HECL2-45.41 contributed to the more acidic optimal pH range of xtGPR4. Overall, our study revealed common and distinct mechanisms of proton sensing by GPR4, from a structural, functional, and evolutionary perspective.
Collapse
Affiliation(s)
- Xin Wen
- Key Laboratory Experimental Teratology of the Ministry of Education, New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China; NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Advanced Medical Research Institute, Shandong University, Jinan, China
| | - Pan Shang
- Key Laboratory Experimental Teratology of the Ministry of Education, New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Haidi Chen
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, Jiangsu, China
| | - Lulu Guo
- NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Advanced Medical Research Institute, Shandong University, Jinan, China
| | - Naikang Rong
- Key Laboratory Experimental Teratology of the Ministry of Education, New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Xiaoyu Jiang
- Key Laboratory Experimental Teratology of the Ministry of Education, New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Xuan Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Junyan Liu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Gongming Yang
- Key Laboratory Experimental Teratology of the Ministry of Education, New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Jiacheng Zhang
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Kongkai Zhu
- NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Advanced Medical Research Institute, Shandong University, Jinan, China
| | - Qingbiao Meng
- Key Laboratory Experimental Teratology of the Ministry of Education, New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Xuefei He
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhihai Wang
- Key Laboratory Experimental Teratology of the Ministry of Education, New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Zili Liu
- Key Laboratory Experimental Teratology of the Ministry of Education, New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Haoran Cheng
- Key Laboratory Experimental Teratology of the Ministry of Education, New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Yilin Zheng
- Key Laboratory Experimental Teratology of the Ministry of Education, New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Bifei Zhang
- Key Laboratory Experimental Teratology of the Ministry of Education, New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Jiaojiao Pang
- Emergency Department, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Zhaoqian Liu
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Peng Xiao
- Key Laboratory Experimental Teratology of the Ministry of Education, New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Yuguo Chen
- Emergency Department, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Lunxu Liu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Fengming Luo
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiao Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Fan Yi
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China.
| | - Pengju Zhang
- Key Laboratory Experimental Teratology of the Ministry of Education, New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China.
| | - Fan Yang
- Key Laboratory Experimental Teratology of the Ministry of Education, New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China; NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Advanced Medical Research Institute, Shandong University, Jinan, China; Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China; Department of Physiology and Pathophysiology, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
| | - Cheng Deng
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, Jiangsu, China.
| | - Jin-Peng Sun
- Key Laboratory Experimental Teratology of the Ministry of Education, New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China; NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Advanced Medical Research Institute, Shandong University, Jinan, China; Department of Physiology and Pathophysiology, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
| |
Collapse
|
12
|
Howard MK, Hoppe N, Huang XP, Mitrovic D, Billesbølle CB, Macdonald CB, Mehrotra E, Rockefeller Grimes P, Trinidad DD, Delemotte L, English JG, Coyote-Maestas W, Manglik A. Molecular basis of proton sensing by G protein-coupled receptors. Cell 2025; 188:671-687.e20. [PMID: 39753132 PMCID: PMC11849372 DOI: 10.1016/j.cell.2024.11.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 09/23/2024] [Accepted: 11/21/2024] [Indexed: 02/09/2025]
Abstract
Three proton-sensing G protein-coupled receptors (GPCRs)-GPR4, GPR65, and GPR68-respond to extracellular pH to regulate diverse physiology. How protons activate these receptors is poorly understood. We determined cryogenic-electron microscopy (cryo-EM) structures of each receptor to understand the spatial arrangement of proton-sensing residues. Using deep mutational scanning (DMS), we determined the functional importance of every residue in GPR68 activation by generating ∼9,500 mutants and measuring their effects on signaling and surface expression. Constant-pH molecular dynamics simulations provided insights into the conformational landscape and protonation patterns of key residues. This unbiased approach revealed that, unlike other proton-sensitive channels and receptors, no single site is critical for proton recognition. Instead, a network of titratable residues extends from the extracellular surface to the transmembrane region, converging on canonical motifs to activate proton-sensing GPCRs. Our approach integrating structure, simulations, and unbiased functional interrogation provides a framework for understanding GPCR signaling complexity.
Collapse
Affiliation(s)
- Matthew K Howard
- Tetrad graduate program, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Bioengineering and Therapeutic Science, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Nicholas Hoppe
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94143, USA; Biophysics graduate program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Xi-Ping Huang
- Department of Pharmacology and the National Institute of Mental Health Psychoactive Drug Screening Program (NIMH PDSP), The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Darko Mitrovic
- Science for Life Laboratory, Department of Applied Physics, KTH Royal Institute of Technology, 12121 Solna, Stockholm, Stockholm County 114 28, Sweden
| | - Christian B Billesbølle
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Christian B Macdonald
- Department of Bioengineering and Therapeutic Science, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Eshan Mehrotra
- Tetrad graduate program, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94143, USA; Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Patrick Rockefeller Grimes
- Department of Bioengineering and Therapeutic Science, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Donovan D Trinidad
- Department of Medicine, Division of Infectious Disease, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lucie Delemotte
- Science for Life Laboratory, Department of Applied Physics, KTH Royal Institute of Technology, 12121 Solna, Stockholm, Stockholm County 114 28, Sweden
| | - Justin G English
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Willow Coyote-Maestas
- Department of Bioengineering and Therapeutic Science, University of California, San Francisco, San Francisco, CA 94143, USA; Chan Zuckerberg Biohub, San Francisco, CA 94148, USA; Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Aashish Manglik
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94143, USA; Chan Zuckerberg Biohub, San Francisco, CA 94148, USA; Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA 94115, USA.
| |
Collapse
|
13
|
Neitzel LR, Fuller DT, Cornell J, Rea S, de Aguiar Ferreira C, Williams CH, Hong CC. Inhibition of GPR68 induces ferroptosis and radiosensitivity in diverse cancer cell types. Sci Rep 2025; 15:4074. [PMID: 39900965 PMCID: PMC11791087 DOI: 10.1038/s41598-025-88357-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 01/28/2025] [Indexed: 02/05/2025] Open
Abstract
Radioresistance is thought to be a major consequence of tumor milieu acidification resulting from the Warburg effect. Previously, using ogremorphin (OGM), a small molecule inhibitor of GPR68, an extracellular proton sensing receptor, we demonstrated that GPR68 is a key pro-survival pathway in glioblastoma cells. Here, we demonstrate that GPR68 inhibition also induces ferroptosis in lung cell carcinoma (A549) and pancreatic ductal adenocarcinoma (Panc02) cells. Moreover, OGM synergized with ionizing radiation to induce lipid peroxidation, a hallmark of ferroptosis, as well as reduce colony size in 2D and 3D cell culture. GPR68 inhibition is not acutely detrimental but increases intracellular free ferrous iron, which is known to trigger reactive oxygen species (ROS) generation. In summary, GPR68 inhibition induces lipid peroxidation in cancer cells and sensitizes them to ionizing radiation in part through the mobilization of intracellular free ferrous iron. Our results suggest that GPR68 is a key mediator of cancer cell radioresistance activated by acidic tumor microenvironment.
Collapse
Affiliation(s)
- Leif R Neitzel
- Department of Medicine, Michigan State University College of Human Medicine, East Lansing, MI, USA
- Henry Ford Health + Michigan State Health Sciences, Detroit, MI, USA
| | - Daniela T Fuller
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jessica Cornell
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Samantha Rea
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Carolina de Aguiar Ferreira
- The Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI, 48824, USA
- Department of Radiology, Michigan State University, East Lansing, MI, 48824, USA
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI, 48824, USA
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, 48824, USA
| | - Charles H Williams
- Department of Medicine, Michigan State University College of Human Medicine, East Lansing, MI, USA.
- Henry Ford Health + Michigan State Health Sciences, Detroit, MI, USA.
| | - Charles C Hong
- Department of Medicine, Michigan State University College of Human Medicine, East Lansing, MI, USA.
- Henry Ford Health + Michigan State Health Sciences, Detroit, MI, USA.
| |
Collapse
|
14
|
Li FB, Bao SQ, Sun XL, Ma JX, Ma XL. Extracellular acidification stimulates OGR1 to modify osteoclast differentiation and activity through the Ca2+‑calcineurin‑NFATc1 pathway. Exp Ther Med 2025; 29:28. [PMID: 39720672 PMCID: PMC11667423 DOI: 10.3892/etm.2024.12778] [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: 07/14/2024] [Accepted: 10/11/2024] [Indexed: 12/26/2024] Open
Abstract
The aim of the present study was to explore the role of ovarian cancer G protein-coupled receptor 1 (OGR1) in osteoclast differentiation and activity induced by extracellular acid. The impact of extracellular acidification on osteoclasts was investigated. Briefly, osteoclasts were generated from RAW 264.7 cells using 100 ng/ml receptor activator of nuclear factor-κB ligand in cell culture media at pH 6.8 or 7.4. Tartrate-resistant acid phosphatase (TRAP) staining and the bone resorption pit assay were used to detect the effects of extracellular acid on the number and absorptive capacity of osteoclasts. Intracellular Ca2+ levels were analyzed using laser scanning confocal microscopy. Reverse transcription-quantitative PCR was used to detect the expression levels of genes associated with osteoclast formation and bone erosion. The role of OGR1 in the acid-stimulated formation and bone resorption of osteoclasts was also investigated. The results showed that in the pH 6.8 medium group the number of osteoclasts was 511.2±54.72 and the area of bone absorption was 4,184.88±277.14 µm2; both were significantly higher than those in the pH 7.4 medium group (all P<0.01). Inhibition of OGR1 using copper ion (Cu2+) reduced the number of osteoclasts and the area of bone resorption in the pH 6.8 medium group (all P<0.05). Furthermore, extracellular acid (pH 6.8) was able to induce a transient increase of Ca2+ levels in osteoclasts; however, inhibition of OGR1 using Cu2+ effectively attenuated the acid-induced increase of Ca2+ in osteoclasts. In addition, the elevation in Ca2+ levels was inhibited when BAPTA, a cytoplasmic Ca2+ chelator with cellular permeability, was added to the cells; however, the extracellular Ca2+-chelating agent ethylene glycol tetraacetic acid did not inhibit the acid-stimulated increase in Ca2+. Treatment with the phospholipase C inhibitor U73122 also inhibited the acid-stimulated increase of Ca2+ in osteoclasts. Furthermore, the mRNA expression levels of TRAP, matrix metalloproteinase-9, osteoclast-related receptor, nuclear factor-activated T cell 1 (NFATc1), cathepsin K and integrin β3 were elevated in the pH 6.8 medium group compared with those in the pH 7.4 medium group (all P<0.05). By contrast, the inhibition of OGR1 using Cu2+ partially reduced the effects of the acidic environment on osteoclast differentiation and activity-related gene expression (all P<0.05). In addition, the mRNA and protein expression levels of calcineurin were increased in osteoclasts in the pH 6.8 group compared with those in the pH 7.4 group (P<0.05), whereas blocking OGR1 suppressed the expression of acid-induced calcineurin. The mRNA expression levels of NFATc1 in osteoclasts were also increased in the pH 6.8 medium group compared with those in the pH 7.4 medium group (P<0.05). By contrast, the specific calcineurin inhibitor cyclosporine A significantly inhibited the acid-induced expression of NFATc1 in osteoclasts. In conclusion, the present study revealed that extracellular acidification may increase osteoclast differentiation and bone resorption activity. Furthermore, OGR1-mediated Ca2+ elevation could have a crucial role in osteoclasts by regulating the Ca2+-calcineurin-NFATc1 signaling pathway and downstream signaling.
Collapse
Affiliation(s)
- Feng-Bo Li
- Department of Orthopedics, Tianjin Hospital, Tianjin 300211, P.R. China
| | - Su-Qing Bao
- Department of Endocrinology, Tianjin First Central Hospital, Tianjin 300192, P.R. China
| | - Xiao-Lei Sun
- Department of Orthopedics, Tianjin Hospital, Tianjin 300211, P.R. China
| | - Jian-Xiong Ma
- Institute of Orthopedics, Tianjin Hospital, Tianjin 300211, P.R. China
| | - Xin-Long Ma
- Department of Orthopedics, Tianjin Hospital, Tianjin 300211, P.R. China
- Institute of Orthopedics, Tianjin Hospital, Tianjin 300211, P.R. China
| |
Collapse
|
15
|
Kume H, Kazama K, Sato R, Sato Y. Possible Involvement of Lysophospholipids in Severe Asthma as Novel Lipid Mediators. Biomolecules 2025; 15:182. [PMID: 40001485 PMCID: PMC11852450 DOI: 10.3390/biom15020182] [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: 10/08/2024] [Revised: 12/25/2024] [Accepted: 01/02/2025] [Indexed: 02/27/2025] Open
Abstract
In severe asthma, symptoms are unstable despite intensive treatment based on high doses of inhaled corticosteroids and on-demand use of oral corticosteroids. Although, recently, various biological agents related to Th2 cytokines have been added to intensive controller medications for severe asthma, a significant progress has not been observed in the management for symptoms (dyspnea, wheezing and cough). Medical treatment focused on Type 2 inflammation is probably insufficient to maintain good long-term management for severe asthma. Airway eosinophilia and decreased reversibility in forced expiratory volume in 1 second (FEV1) are listed as major predictors for exacerbation-prone asthma. However, it is generally considered that asthma is complex and heterogeneous. It is necessary to establish precision medicine using treatable traits based on a multidimensional approach related to asthma. Since phospholipids generate lysophospholipids and arachidonic acid by phospholipases, lysophospholipids can be associated with the pathogenesis of this disease via action on smooth muscle, endothelium, and epithelium in the airways. Lysophosphatidic acid (LPA), lysophosphatidylcholine (LPC), and sphingosine 1-phosphate (S1P) are increased in bronchoalveolar fluid after allergen challenge. LPA, LPC, and S1P recruit eosinophils to the lungs and cause β2-adrenergic desensitization. LAP and S1P cause contraction and hyperresponsiveness in airway smooth muscle. Moreover, lysophosphatidylserine and S1P are associated with the allergic reaction related to IgE/FcεRI in mast cells. Lysophospholipid action is probably comprised of corticosteroid resistance and is independent of Type 2 inflammation, and may be corelated with oxidative stress. Lysophospholipids may be a novel molecular target in advancing the management and treatment of asthma. This review discusses the clinical relevance of lysophospholipids in asthma.
Collapse
Affiliation(s)
- Hiroaki Kume
- Department of Infectious Diseases and Respiratory Medicine, Fukushima Medical University Aizu Medical Center, 21-2 Maeda, Tanisawa, Kawahigashi, Aizuwakamatsu 969-3492, Japan; (K.K.); (R.S.); (Y.S.)
| | | | | | | |
Collapse
|
16
|
Ma Y, Wang Y, Tang M, Weng Y, Chen Y, Xu Y, An S, Wu Y, Zhao S, Xu H, Li D, Liu M, Lu W, Ru H, Song G. Cryo-EM structure of an activated GPR4-Gs signaling complex. Nat Commun 2025; 16:605. [PMID: 39799123 PMCID: PMC11724869 DOI: 10.1038/s41467-025-55901-2] [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: 06/05/2024] [Accepted: 01/03/2025] [Indexed: 01/15/2025] Open
Abstract
G protein-coupled receptor 4 (GPR4) belongs to the subfamily of proton-sensing GPCRs (psGPCRs), which detect pH changes in extracellular environment and regulate diverse physiological responses. GPR4 was found to be overactivated in acidic tumor microenvironment as well as inflammation sites, with a triad of acidic residues within the transmembrane domain identified as crucial for proton sensing. However, the 3D structure remains unknown, and the roles of other conserved residues within psGPCRs are not well understood. Here we report cryo-electron microscopy (cryo-EM) structures of active zebrafish GPR4 at both pH 6.5 and 8.5, each highlighting a distribution of histidine and acidic residues at the extracellular region. Cell-based assays show that these ionizable residues moderately influence the proton-sensing capacity of zebrafish GPR4, compared to the more significant effects of the triad residues. Furthermore, we reveal a cluster of aromatic residues within the orthosteric pocket that may propagate the signaling to the intercellular region via repacking the aromatic patch at the central region. This study provides a framework for future signaling and functional investigation of psGPCRs.
Collapse
Affiliation(s)
- Yitong Ma
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Yijie Wang
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Mengyuan Tang
- Life Sciences Institute, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yuan Weng
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Ying Chen
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Yueming Xu
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Shuxiao An
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Yiran Wu
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Suwen Zhao
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Huanhuan Xu
- College of Science, Yunnan Agricultural University, Kunming, China
| | - Dali Li
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Mingyao Liu
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Weiqiang Lu
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China.
| | - Heng Ru
- Life Sciences Institute, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Gaojie Song
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China.
| |
Collapse
|
17
|
Vögele M, Thomson NJ, Truong ST, McAvity J, Zachariae U, Dror RO. Systematic analysis of biomolecular conformational ensembles with PENSA. J Chem Phys 2025; 162:014101. [PMID: 39745157 PMCID: PMC11698571 DOI: 10.1063/5.0235544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/12/2024] [Indexed: 01/06/2025] Open
Abstract
Atomic-level simulations are widely used to study biomolecules and their dynamics. A common goal in such studies is to compare simulations of a molecular system under several conditions-for example, with various mutations or bound ligands-in order to identify differences between the molecular conformations adopted under these conditions. However, the large amount of data produced by simulations of ever larger and more complex systems often renders it difficult to identify the structural features that are relevant to a particular biochemical phenomenon. We present a flexible software package named Python ENSemble Analysis (PENSA) that enables a comprehensive and thorough investigation into biomolecular conformational ensembles. It provides featurization and feature transformations that allow for a complete representation of biomolecules such as proteins and nucleic acids, including water and ion binding sites, thus avoiding the bias that would come with manual feature selection. PENSA implements methods to systematically compare the distributions of molecular features across ensembles to find the significant differences between them and identify regions of interest. It also includes a novel approach to quantify the state-specific information between two regions of a biomolecule, which allows, for example, tracing information flow to identify allosteric pathways. PENSA also comes with convenient tools for loading data and visualizing results, making them quick to process and easy to interpret. PENSA is an open-source Python library maintained at https://github.com/drorlab/pensa along with an example workflow and a tutorial. We demonstrate its usefulness in real-world examples by showing how it helps us determine molecular mechanisms efficiently.
Collapse
Affiliation(s)
- Martin Vögele
- Authors to whom correspondence should be addressed: and
| | - Neil J. Thomson
- Department of Computational Biology, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Sang T. Truong
- Department of Computer Science, Stanford University, Stanford, California 94305, USA
| | - Jasper McAvity
- Department of Computer Science, Stanford University, Stanford, California 94305, USA
| | | | - Ron O. Dror
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
18
|
He X, Hawkins C, Lawley L, Phan TM, Park I, Joven N, Zhang J, Wunderlich M, Mizukawa B, Pei S, Patel A, VanOudenhove J, Halene S, Fang J. GPR68 supports AML cells through the calcium/calcineurin pro-survival pathway and confers chemoresistance by mediating glucose metabolic symbiosis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167565. [PMID: 39522891 DOI: 10.1016/j.bbadis.2024.167565] [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: 05/28/2024] [Revised: 10/21/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024]
Abstract
Accumulating evidence demonstrates that the "Warburg effect" that glycolysis is enhanced even in the presence of oxygen existed in hematopoietic malignancies, contributing to extracellular acidosis. G-protein coupled receptor 68 (GPR68), as a proton sensing GPCR responding to extracellular acidosis, is expected to play a critical role in hematopoietic malignancies. In the present study, we found that GPR68 was overexpressed in acute myeloid leukemia (AML) cells, and GPR68 deficiency impaired AML cell survival in vitro and cell engraftment in vivo. Mechanistic studies revealed that unlike GPR68 regulates Calpain1 in myelodysplastic syndromes (MDS) cells, GPR68 deficiency reduced cytosolic Ca2+ levels and calcineurin (CaN) activity in AML cells through an NFAT-independent mechanism. Moreover, the decreased Ca2+ levels disturbed cellular respiration (i.e., oxidative phosphorylation, OxPhos) by inhibiting isocitrate dehydrogenase (IDH) activity; this was more pronounced when BCL2 was inhibited simultaneously. Interestingly, GPR68 inhibition also decreased aerobic glycolysis in AML cells in a Ca2+-independent manner, suggesting that GPR68 mediated glucose metabolic symbiosis. As glucose metabolic symbiosis and the heterogeneous dependencies on aerobic glycolysis and cellular respiration tremendously impact chemosensitivity, the inhibition of GPR68 potentiated the tumoricidal effect of first-line chemotherapeutic agents, including BCL-2 inhibitors targeting OxPhos and cytarabine (Ara-C) targeting glycolysis. Consistent with these in vitro observations, higher levels of GPR68 were associated with inferior clinical outcomes in AML patients who received chemotherapies. In short, GPR68 drives the Ca2+/CaN pro-survival pathway and mediates glucose metabolic pathways in AML cells. Targeting GPR68 eradicates AML cells and alleviates chemoresistance, which could be exploited as a therapeutic target.
Collapse
MESH Headings
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/genetics
- Humans
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/drug therapy
- Calcineurin/metabolism
- Calcium/metabolism
- Glucose/metabolism
- Animals
- Drug Resistance, Neoplasm
- Mice
- Cell Survival/drug effects
- Cell Line, Tumor
- Glycolysis
Collapse
Affiliation(s)
- Xiaofei He
- First Affliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Zhejiang Province, China; Department of Drug Discovery and Biomedical Sciences, University of South Carolina College of Pharmacy, Columbia, SC 29208, USA
| | - Caleb Hawkins
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina College of Pharmacy, Columbia, SC 29208, USA
| | - Lauren Lawley
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina College of Pharmacy, Columbia, SC 29208, USA
| | - Tra Mi Phan
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina College of Pharmacy, Columbia, SC 29208, USA
| | - Isaac Park
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina College of Pharmacy, Columbia, SC 29208, USA
| | - Nicole Joven
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina College of Pharmacy, Columbia, SC 29208, USA
| | - Jiajia Zhang
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Mark Wunderlich
- Cancer and Blood Disease Institutes, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Benjamin Mizukawa
- Cancer and Blood Disease Institutes, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Shanshan Pei
- Division of Hematology, University of Colorado, Denver, CO 80045, USA
| | - Amisha Patel
- Section of Hematology, Yale Cancer Center and Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jennifer VanOudenhove
- Section of Hematology, Yale Cancer Center and Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Stephanie Halene
- Section of Hematology, Yale Cancer Center and Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Yale Stem Cell Center and Yale RNA Center, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jing Fang
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina College of Pharmacy, Columbia, SC 29208, USA.
| |
Collapse
|
19
|
Chen YJN, Shi RC, Xiang YC, Fan L, Tang H, He G, Zhou M, Feng XZ, Tan JD, Huang P, Ye X, Zhao K, Fu WY, Li LL, Bian XT, Chen H, Wang F, Wang T, Zhang CK, Zhou BH, Chen W, Liang TT, Lv JT, Kang X, Shi YX, Kim E, Qin YH, Hettinghouse A, Wang KD, Zhao XL, Yang MY, Tang YZ, Piao HL, Guo L, Liu CJ, Miao HM, Tang KL. Malate initiates a proton-sensing pathway essential for pH regulation of inflammation. Signal Transduct Target Ther 2024; 9:367. [PMID: 39737965 PMCID: PMC11683149 DOI: 10.1038/s41392-024-02076-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 01/01/2025] Open
Abstract
Metabolites can double as a signaling modality that initiates physiological adaptations. Metabolism, a chemical language encoding biological information, has been recognized as a powerful principle directing inflammatory responses. Cytosolic pH is a regulator of inflammatory response in macrophages. Here, we found that L-malate exerts anti-inflammatory effect via BiP-IRF2BP2 signaling, which is a sensor of cytosolic pH in macrophages. First, L-malate, a TCA intermediate upregulated in pro-inflammatory macrophages, was identified as a potent anti-inflammatory metabolite through initial screening. Subsequent screening with DARTS and MS led to the isolation of L-malate-BiP binding. Further screening through protein‒protein interaction microarrays identified a L-malate-restrained coupling of BiP with IRF2BP2, a known anti-inflammatory protein. Interestingly, pH reduction, which promotes carboxyl protonation of L-malate, facilitates L-malate and carboxylate analogues such as succinate to bind BiP, and disrupt BiP-IRF2BP2 interaction in a carboxyl-dependent manner. Both L-malate and acidification inhibit BiP-IRF2BP2 interaction, and protect IRF2BP2 from BiP-driven degradation in macrophages. Furthermore, both in vitro and in vivo, BiP-IRF2BP2 signal is required for effects of both L-malate and pH on inflammatory responses. These findings reveal a previously unrecognized, proton/carboxylate dual sensing pathway wherein pH and L-malate regulate inflammatory responses, indicating the role of certain carboxylate metabolites as adaptors in the proton biosensing by interactions between macromolecules.
Collapse
Affiliation(s)
- Yu-Jia-Nan Chen
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China.
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA.
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases & Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China.
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China.
| | - Rong-Chen Shi
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China
| | - Yuan-Cai Xiang
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Li Fan
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases & Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Hong Tang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Gang He
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Mei Zhou
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Xin-Zhe Feng
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
| | - Jin-Dong Tan
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Pan Huang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Xiao Ye
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Kun Zhao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China
| | - Wen-Yu Fu
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
- Department of Orthopedics and Rehabilitations, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - Liu-Li Li
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
| | - Xu-Ting Bian
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Huan Chen
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Feng Wang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Teng Wang
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China
| | - Chen-Ke Zhang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Bing-Hua Zhou
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Wan Chen
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Tao-Tao Liang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Jing-Tong Lv
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Xia Kang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China
| | - You-Xing Shi
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Ellen Kim
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
| | - Yin-Hua Qin
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Army Medical University, Chongqing, 400038, China
| | - Aubryanna Hettinghouse
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
| | - Kai-di Wang
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
- Department of Medical Experimental Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, 266000, China
| | - Xiang-Li Zhao
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
- Department of Orthopedics and Rehabilitations, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - Ming-Yu Yang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Yu-Zhen Tang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Hai-Long Piao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Lin Guo
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| | - Chuan-Ju Liu
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA.
- Department of Orthopedics and Rehabilitations, Yale University School of Medicine, New Haven, CT, 06519, USA.
| | - Hong-Ming Miao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China.
- Jinfeng Laboratory, Chongqing, 401329, China.
| | - Kang-Lai Tang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
20
|
Karki P, Ke Y, Zhang C, Promnares K, Li Y, Williams CH, Hong CC, Birukov KG, Birukova AA. GPR68 Mediates Lung Endothelial Dysfunction Caused by Bacterial Inflammation and Tissue Acidification. Cells 2024; 13:2125. [PMID: 39768215 PMCID: PMC11674861 DOI: 10.3390/cells13242125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 12/18/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Tissue acidification resulting from dysregulated cellular bioenergetics accompanies various inflammatory states. GPR68, along with other members of proton-sensing G protein-coupled receptors, responds to extracellular acidification and has been implicated in chronic inflammation-related diseases such as ischemia, cancer, and colitis. The present study examined the role of extracellular acidification on human pulmonary endothelial cell (EC) permeability and inflammatory status per se and investigated potential synergistic effects of acidosis on endothelial dysfunction caused by bacterial lipopolysaccharide (LPS, Klebsiella pneumoniae). Results showed that medium acidification to pH 6.5 caused a delayed increase in EC permeability illustrated by a decrease in transendothelial electrical resistance and loss of continuous VE-cadherin immunostaining at cell junctions. Likewise, acidic pH induced endothelial inflammation reflected by increased mRNA and protein expression of EC adhesion molecules VCAM-1 and ICAM-1, upregulated mRNA transcripts of tumor necrosis factor-α, IL-6, IL-8, IL-1β, and CXCL5, and increased secretion of ICAM-1, IL-6, and IL-8 in culture medium monitored by ELISA. Among the GPCRs tested, acidic pH selectively increased mRNA and protein expression of GPR68, and only the GPR68-specific small molecule inhibitor OGM-8345 rescued acidosis-induced endothelial permeability and inflammation. Furthermore, acidic pH exacerbated LPS-induced endothelial permeability and inflammatory response in cultured lung macrovascular as well as microvascular endothelial cells. These effects were suppressed by OGM-8345 in both EC types. Altogether, these results suggest that GPR68 is a critical mediator of acidic pH-induced dysfunction of human pulmonary vascular endothelial cells and mediates the augmenting effect of tissue acidification on LPS-induced endothelial cell injury.
Collapse
Affiliation(s)
- Pratap Karki
- Division of Pulmonary and Critical Care, Department of Medicine, UMSOM Lung Biology Program, University of Maryland School of Medicine, 20 Penn Street, HSF-2, Room S143, Baltimore, MD 21201, USA; (P.K.); (C.Z.); (Y.L.)
| | - Yunbo Ke
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (Y.K.); (K.P.); (K.G.B.)
| | - Chenou Zhang
- Division of Pulmonary and Critical Care, Department of Medicine, UMSOM Lung Biology Program, University of Maryland School of Medicine, 20 Penn Street, HSF-2, Room S143, Baltimore, MD 21201, USA; (P.K.); (C.Z.); (Y.L.)
| | - Kamoltip Promnares
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (Y.K.); (K.P.); (K.G.B.)
| | - Yue Li
- Division of Pulmonary and Critical Care, Department of Medicine, UMSOM Lung Biology Program, University of Maryland School of Medicine, 20 Penn Street, HSF-2, Room S143, Baltimore, MD 21201, USA; (P.K.); (C.Z.); (Y.L.)
| | - Charles H. Williams
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (C.H.W.); (C.C.H.)
| | - Charles C. Hong
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (C.H.W.); (C.C.H.)
| | - Konstantin G. Birukov
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (Y.K.); (K.P.); (K.G.B.)
| | - Anna A. Birukova
- Division of Pulmonary and Critical Care, Department of Medicine, UMSOM Lung Biology Program, University of Maryland School of Medicine, 20 Penn Street, HSF-2, Room S143, Baltimore, MD 21201, USA; (P.K.); (C.Z.); (Y.L.)
| |
Collapse
|
21
|
Hansen CE, Hollaus D, Kamermans A, de Vries HE. Tension at the gate: sensing mechanical forces at the blood-brain barrier in health and disease. J Neuroinflammation 2024; 21:325. [PMID: 39696463 PMCID: PMC11657007 DOI: 10.1186/s12974-024-03321-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/07/2024] [Indexed: 12/20/2024] Open
Abstract
Microvascular brain endothelial cells tightly limit the entry of blood components and peripheral cells into the brain by forming the blood-brain barrier (BBB). The BBB is regulated by a cascade of mechanical and chemical signals including shear stress and elasticity of the adjacent endothelial basement membrane (BM). During physiological aging, but especially in neurological diseases including multiple sclerosis (MS), stroke, small vessel disease, and Alzheimer's disease (AD), the BBB is exposed to inflammation, rigidity changes of the BM, and disturbed cerebral blood flow (CBF). These altered forces lead to increased vascular permeability, reduced endothelial reactivity to vasoactive mediators, and promote leukocyte transmigration. Whereas the molecular players involved in leukocyte infiltration have been described in detail, the importance of mechanical signalling throughout this process has only recently been recognized. Here, we review relevant features of mechanical forces acting on the BBB under healthy and pathological conditions, as well as the endothelial mechanosensory elements detecting and responding to altered forces. We demonstrate the underlying complexity by focussing on the family of transient receptor potential (TRP) ion channels. A better understanding of these processes will provide insights into the pathogenesis of several neurological disorders and new potential leads for treatment.
Collapse
Affiliation(s)
- Cathrin E Hansen
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands
| | - David Hollaus
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Alwin Kamermans
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands.
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
22
|
Stein C. Effects of pH on opioid receptor activation and implications for drug design. Biophys J 2024; 123:4158-4166. [PMID: 38970252 PMCID: PMC11700362 DOI: 10.1016/j.bpj.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/29/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024] Open
Abstract
G-protein-coupled receptors are integral membrane proteins that transduce chemical signals from the extracellular matrix into the cell. Traditional drug design has considered ligand-receptor interactions only under normal conditions. However, studies on opioids indicate that such interactions are very different in diseased tissues. In such microenvironments, protons play an important role in structural and functional alterations of both ligands and receptors. The pertinent literature strongly suggests that future drug design should take these aspects into account in order to reduce adverse side effects while preserving desired effects of novel compounds.
Collapse
Affiliation(s)
- Christoph Stein
- Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Experimental Anaesthesiology, Berlin, Germany.
| |
Collapse
|
23
|
Williams MD, Morgan JS, Bullock MT, Poovey CE, Wisniewski ME, Francisco JT, Barajas-Nunez JA, Hijazi AM, Theobald D, Sriramula S, Mansfield KD, Holland NA, Tulis DA. pH-sensing GPR68 inhibits vascular smooth muscle cell proliferation through Rap1A. Am J Physiol Heart Circ Physiol 2024; 327:H1210-H1229. [PMID: 39269448 PMCID: PMC11560072 DOI: 10.1152/ajpheart.00413.2024] [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: 06/21/2024] [Revised: 09/06/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024]
Abstract
Phenotypic transformation of vascular smooth muscle (VSM) from a contractile state to a synthetic, proliferative state is a hallmark of cardiovascular disease (CVD). In CVD, diseased tissue often becomes acidic from altered cellular metabolism secondary to compromised blood flow, yet the contribution of local acid/base imbalance to the disease process has been historically overlooked. In this study, we examined the regulatory impact of the pH-sensing G protein-coupled receptor GPR68 on vascular smooth muscle (VSM) proliferation in vivo and in vitro in wild-type (WT) and GPR68 knockout (KO) male and female mice. Arterial injury reduced GPR68 expression in WT vessels and exaggerated medial wall remodeling in GPR68 KO vessels. In vitro, KO VSM cells showed increased cell-cycle progression and proliferation compared with WT VSM cells, and GPR68-inducing acidic exposure reduced proliferation in WT cells. mRNA and protein expression analyses revealed increased Rap1A in KO cells compared with WT cells, and RNA silencing of Rap1A reduced KO VSM cell proliferation. In sum, these findings support a growth-inhibitory capacity of pH-sensing GPR68 and suggest a mechanistic role for the small GTPase Rap1A in GPR68-mediated VSM growth control. These results shed light on GPR68 and its effector Rap1A as potential targets to combat pathological phenotypic switching and proliferation in VSM.NEW & NOTEWORTHY Extracellular acidosis remains an understudied feature of many pathologies. We examined a potential regulatory role for pH-sensing GPR68 in vascular smooth muscle (VSM) growth in the context of CVD. With in vivo and in vitro growth models with GPR68-deficient mice and GPR68 induction strategies, novel findings revealed capacity of GPR68 to attenuate growth through the small GTPase Rap1A. These observations highlight GPR68 and its effector Rap1A as possible therapeutic targets to combat pathological VSM growth.
Collapse
MESH Headings
- Animals
- Female
- Male
- Mice
- Cell Proliferation
- Cells, Cultured
- Disease Models, Animal
- Hydrogen-Ion Concentration
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- rap1 GTP-Binding Proteins/metabolism
- rap1 GTP-Binding Proteins/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/genetics
- Signal Transduction
- Vascular Remodeling
Collapse
Affiliation(s)
- Madison D Williams
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States
| | - Joshua S Morgan
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States
| | - Michael T Bullock
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States
| | - Cere E Poovey
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States
| | - Michael E Wisniewski
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States
| | - Jake T Francisco
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States
| | - Jerry A Barajas-Nunez
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States
| | - Amira M Hijazi
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States
| | - Drew Theobald
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States
| | - Srinivas Sriramula
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States
| | - Kyle D Mansfield
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States
| | - Nathan A Holland
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States
| | - David A Tulis
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States
| |
Collapse
|
24
|
Hurtado-Lorenzo A, Swantek JL. The landscape of new therapeutic opportunities for IBD. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 101:1-83. [PMID: 39521596 DOI: 10.1016/bs.apha.2024.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
This chapter presents an overview of the emerging strategies to address the unmet needs in the management of inflammatory bowel diseases (IBD). IBD poses significant challenges, as over half of patients experience disease progression despite interventions, leading to irreversible complications, and a substantial proportion do not respond to existing therapies, such as biologics. To overcome these limitations, we describe a diverse array of novel therapeutic approaches. In the area of immune homeostasis restoration, the focus is on targeting cytokine networks, leukocyte trafficking, novel immune pathways, and cell therapies involving regulatory T cells and mesenchymal stem cells (MSC). Recognizing the critical role of impaired intestinal barrier integrity in IBD, we highlight therapies aimed at restoring barrier function and promoting mucosal healing, such as those targeting cell proliferation, tight junctions, and lipid mediators. Addressing the challenges posed by fibrosis and fistulas, we describe emerging targets for reversing fibrosis like kinase and cytokine inhibitors and nuclear receptor agonists, as well as the potential of MSC for fistulas. The restoration of a healthy gut microbiome, through strategies like fecal microbiota transplantation, rationally defined bacterial consortia, and targeted antimicrobials, is also highlighted. We also describe innovative approaches to gut-targeted drug delivery to enhance efficacy and minimize side effects. Reinforcing these advancements is the critical role of precision medicine, which emphasizes the use of multiomics analysis for the discovery of biomarkers to enable personalized IBD care. Overall, the emerging landscape of therapeutic opportunities for IBD holds great potential to surpass the therapeutic ceiling of current treatments.
Collapse
Affiliation(s)
- Andrés Hurtado-Lorenzo
- Translational Research & IBD Ventures, Research Department, Crohn's & Colitis Foundation, New York, NY, United States.
| | - Jennifer L Swantek
- Translational Research & IBD Ventures, Research Department, Crohn's & Colitis Foundation, New York, NY, United States
| |
Collapse
|
25
|
Cappellesso F, Mazzone M, Virga F. Acid affairs in anti-tumour immunity. Cancer Cell Int 2024; 24:354. [PMID: 39465367 PMCID: PMC11514911 DOI: 10.1186/s12935-024-03520-0] [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: 05/31/2024] [Accepted: 09/30/2024] [Indexed: 10/29/2024] Open
Abstract
Metabolic rewiring of cancer cells is one of the hallmarks of cancer. As a consequence, the metabolic landscape of the tumour microenvironment (TME) differs compared to correspondent healthy tissues. Indeed, due to the accumulation of acid metabolites, such as lactate, the pH of the TME is generally acidic with a pH drop that can be as low as 5.6. Disruptions in the acid-base balance and elevated lactate levels can drive malignant progression not only through cell-intrinsic mechanisms but also by impacting the immune response. Generally, acidity and lactate dampen the anti-tumour response of both innate and adaptive immune cells favouring tumour progression and reducing the response to immunotherapy. In this review, we summarize the current knowledge on the functional, metabolic and epigenetic effects of acidity and lactate on the cells of the immune system. In particular, we focus on the role of monocarboxylate transporters (MCTs) and other solute carrier transporters (SLCs) that, by mediating the exchange of lactate (among other metabolites) and bicarbonate, participate in pH regulation and lactate transport in the cancer context. Finally, we discuss advanced approaches to target pH or lactate in the TME to enhance the anti-tumour immune response.
Collapse
Affiliation(s)
- Federica Cappellesso
- Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium.
- Lab of Dendritic Cell Biology and Cancer Immunotherapy, Inflammation Research Center, VIB, Brussels, Belgium.
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Federico Virga
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, 28029, Spain.
| |
Collapse
|
26
|
Bouyer PG, Salameh AI, Zhou Y, Kolba TN, Boron WF. Effects of extracellular metabolic acidosis and out-of-equilibrium CO 2/HCO 3 - solutions on intracellular pH in cultured rat hippocampal neurons. Front Physiol 2024; 15:1434359. [PMID: 39444753 PMCID: PMC11496273 DOI: 10.3389/fphys.2024.1434359] [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: 05/17/2024] [Accepted: 08/28/2024] [Indexed: 10/25/2024] Open
Abstract
Metabolic acidosis (MAc)-an extracellular pH (pHo) decrease caused by a [HCO3 -]o decrease at constant [CO2]o-usually causes intracellular pH (pHi) to fall. Here we determine the extent to which the pHi decrease depends on the pHo decrease vs the concomitant [HCO3 -]o decrease. We use rapid-mixing to generate out-of-equilibrium CO2/HCO3 - solutions in which we stabilize [CO2]o and [HCO3 -]o while decreasing pHo (pure acidosis, pAc), or stabilize [CO2]o and pHo while decreasing [HCO3 -]o (pure metabolic/down, pMet↓). Using the fluorescent dye 2',7'-bis-2-carboxyethyl)-5(and-6)carboxyfluorescein (BCECF) to monitor pHi in rat hippocampal neurons in primary culture, we find that-in naïve neurons-the pHi decrease caused by MAc is virtually the sum of those caused by pAc (∼70%) + pMet↓ (∼30%). However, if we impose a first challenge (MAc1, pAc1, or pMet↓1), allow the neurons to recover, and then impose a second challenge (MAc2, pAc2, or pMet↓2), we find that pAc/pMet↓ additivity breaks down. In a twin-challenge protocol in which challenge #2 is MAc, the pHo and [HCO3 -]o decreases during challenge #1 must be coincident in order to mimic the effects of MAc1 on MAc2. Conversely, if challenge #1 is MAc, then the pHo and [HCO3 -]o decreases during challenge #2 must be coincident in order for MAc1 to produce its physiological effects during the challenge #2 period. We conclude that the history of challenge #1 (MAc1, pAc1, or pMet↓1)-presumably as detected by one or more acid-base sensors-has a major impact on the pHi response during challenge #2 (MAc2, pAc2, or pMet↓2).
Collapse
Affiliation(s)
- Patrice G. Bouyer
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, United States
- Department of Biology, Valparaiso University, Valparaiso, IN, United States
| | - Ahlam I. Salameh
- Preclinical Sciences Division, Kent State University College of Podiatric Medicine, Independence, OH, United States
- Department of Physiology & Biophysics Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Yuehan Zhou
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, United States
- Department of Physiology & Biophysics Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Tiffany N. Kolba
- Department of Mathematics & Statistics, Valparaiso University, Valparaiso, IN, United States
| | - Walter F. Boron
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, United States
- Department of Physiology & Biophysics Case Western Reserve University School of Medicine, Cleveland, OH, United States
| |
Collapse
|
27
|
Hamaguchi R, Isowa M, Narui R, Morikawa H, Okamoto T, Wada H. How Does Cancer Occur? How Should It Be Treated? Treatment from the Perspective of Alkalization Therapy Based on Science-Based Medicine. Biomedicines 2024; 12:2197. [PMID: 39457509 PMCID: PMC11504456 DOI: 10.3390/biomedicines12102197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 09/22/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
This review article investigates the relationship between mitochondrial dysfunction and cancer progression, emphasizing the metabolic shifts that promote tumor growth. Mitochondria are crucial for cellular energy production, but they also play a significant role in cancer progression by promoting glycolysis even under oxygen-rich conditions, a phenomenon known as the Warburg effect. This metabolic reprogramming enables cancer cells to maintain an alkaline internal pH and an acidic external environment, which are critical for their proliferation and survival in hypoxic conditions. The article also explores the acidic tumor microenvironment (TME), a consequence of intensive glycolytic activity and proton production by cancer cells. This acidic milieu enhances the invasiveness and metastatic potential of cancer cells and contributes to increased resistance to chemotherapy. Alkalization therapy, which involves neutralizing this acidity through dietary modifications and the administration of alkalizing agents such as sodium bicarbonate, is highlighted as an effective strategy to counteract these adverse conditions and impede cancer progression. Integrating insights from science-based medicine, the review evaluates the effectiveness of alkalization therapy across various cancer types through clinical assessments. Science-based medicine, which utilizes inductive reasoning from observed clinical outcomes, lends support to the hypothesis of metabolic reprogramming in cancer treatment. By addressing both metabolic and environmental disruptions, this review suggests that considering cancer as primarily a metabolic disorder could lead to more targeted and effective treatment strategies, potentially improving outcomes for patients with advanced-stage cancers.
Collapse
Affiliation(s)
- Reo Hamaguchi
- Japanese Society on Inflammation and Metabolism in Cancer, 119 Nishioshikouji-cho, Nakagyo-ku, Kyoto 604-0842, Japan; (R.H.); (M.I.); (R.N.); (H.M.)
| | - Masahide Isowa
- Japanese Society on Inflammation and Metabolism in Cancer, 119 Nishioshikouji-cho, Nakagyo-ku, Kyoto 604-0842, Japan; (R.H.); (M.I.); (R.N.); (H.M.)
| | - Ryoko Narui
- Japanese Society on Inflammation and Metabolism in Cancer, 119 Nishioshikouji-cho, Nakagyo-ku, Kyoto 604-0842, Japan; (R.H.); (M.I.); (R.N.); (H.M.)
| | - Hiromasa Morikawa
- Japanese Society on Inflammation and Metabolism in Cancer, 119 Nishioshikouji-cho, Nakagyo-ku, Kyoto 604-0842, Japan; (R.H.); (M.I.); (R.N.); (H.M.)
| | - Toshihiro Okamoto
- Department of Thoracic and Cardiovascular Surgery, Cleveland Clinic, Cleveland, OH 44195, USA;
| | - Hiromi Wada
- Japanese Society on Inflammation and Metabolism in Cancer, 119 Nishioshikouji-cho, Nakagyo-ku, Kyoto 604-0842, Japan; (R.H.); (M.I.); (R.N.); (H.M.)
| |
Collapse
|
28
|
Berdiaki A, Neagu M, Tzanakakis P, Spyridaki I, Pérez S, Nikitovic D. Extracellular Matrix Components and Mechanosensing Pathways in Health and Disease. Biomolecules 2024; 14:1186. [PMID: 39334952 PMCID: PMC11430160 DOI: 10.3390/biom14091186] [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: 08/07/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Glycosaminoglycans (GAGs) and proteoglycans (PGs) are essential components of the extracellular matrix (ECM) with pivotal roles in cellular mechanosensing pathways. GAGs, such as heparan sulfate (HS) and chondroitin sulfate (CS), interact with various cell surface receptors, including integrins and receptor tyrosine kinases, to modulate cellular responses to mechanical stimuli. PGs, comprising a core protein with covalently attached GAG chains, serve as dynamic regulators of tissue mechanics and cell behavior, thereby playing a crucial role in maintaining tissue homeostasis. Dysregulation of GAG/PG-mediated mechanosensing pathways is implicated in numerous pathological conditions, including cancer and inflammation. Understanding the intricate mechanisms by which GAGs and PGs modulate cellular responses to mechanical forces holds promise for developing novel therapeutic strategies targeting mechanotransduction pathways in disease. This comprehensive overview underscores the importance of GAGs and PGs as key mediators of mechanosensing in maintaining tissue homeostasis and their potential as therapeutic targets for mitigating mechano-driven pathologies, focusing on cancer and inflammation.
Collapse
Affiliation(s)
- Aikaterini Berdiaki
- Department of Histology-Embryology, Medical School, University of Crete, 712 03 Heraklion, Greece; (A.B.); (P.T.); (I.S.)
| | - Monica Neagu
- Immunology Department, “Victor Babes” National Institute of Pathology, 050096 Bucharest, Romania;
| | - Petros Tzanakakis
- Department of Histology-Embryology, Medical School, University of Crete, 712 03 Heraklion, Greece; (A.B.); (P.T.); (I.S.)
| | - Ioanna Spyridaki
- Department of Histology-Embryology, Medical School, University of Crete, 712 03 Heraklion, Greece; (A.B.); (P.T.); (I.S.)
| | - Serge Pérez
- Centre de Recherche sur les Macromolécules Végétales (CERMAV), Centre National de la Recherche Scientifique (CNRS), University Grenoble Alpes, 38000 Grenoble, France;
| | - Dragana Nikitovic
- Department of Histology-Embryology, Medical School, University of Crete, 712 03 Heraklion, Greece; (A.B.); (P.T.); (I.S.)
| |
Collapse
|
29
|
Gonye EC, Shi Y, Li K, Clements RT, Xu W, Bayliss DA. Intrinsic Molecular Proton Sensitivity Underlies GPR4 Effects on Retrotrapezoid Nucleus Neuronal Activation and CO 2-Stimulated Breathing. J Neurosci 2024; 44:e0799242024. [PMID: 39107057 PMCID: PMC11376338 DOI: 10.1523/jneurosci.0799-24.2024] [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: 04/29/2024] [Revised: 06/28/2024] [Accepted: 07/30/2024] [Indexed: 08/09/2024] Open
Abstract
An interoceptive homeostatic reflex monitors levels of CO2/H+ to maintain blood gas homeostasis and rapidly regulate tissue acid-base balance by driving lung ventilation and CO2 excretion-this CO2-evoked increase in respiration is the hypercapnic ventilatory reflex (HCVR). Retrotrapezoid nucleus (RTN) neurons provide crucial excitatory drive to downstream respiratory rhythm/pattern-generating circuits, and their activity is directly modulated by changes in CO2/H+ RTN neurons express GPR4 and TASK-2, global deletion of which abrogates CO2/H+ activation of RTN neurons and the HCVR. It has not been determined if the intrinsic pH sensitivity of these proton detectors is required for these effects. We used CRISPR/Cas9 genome editing to generate mice with mutations in either of two pH-sensing histidine residues in GPR4 to determine effects on RTN neuronal CO2/H+ sensitivity and the HCVR. In global GPR4(H81F) and GPR4(H167F) mice, CO2-stimulated breathing and CO2-induced RTN neuronal activation were strongly blunted, with no effect on hypoxia-stimulated breathing. In brainstem slices from GPR4(H81F) mice, peak firing of RTN neurons during bath acidification was significantly reduced compared with GPR4 wild-type mice, and a subpopulation of RTN neurons was rendered pH-insensitive, phenocopying previous results from GPR4-deleted mice. These effects were independent of changes in RTN number/distribution, neuronal excitability or transcript levels for GPR4 and TASK-2. CO2-stimulated breathing was reduced to a similar extent in GPR4(H81F) and TASK-2-deleted mice, with combined mutation yielding no additional deficit in the HCVR. Together, these data demonstrate that the intrinsic pH sensitivity of GPR4 is necessary for full elaboration of the HCVR.
Collapse
Affiliation(s)
- Elizabeth C Gonye
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22903
| | - Yingtang Shi
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22903
| | - Keyong Li
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22903
| | - Rachel T Clements
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22903
| | - Wenhao Xu
- Genetically Engineered Mouse Model Core, University of Virginia, Charlottesville, Virginia 22903
| | - Douglas A Bayliss
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22903
| |
Collapse
|
30
|
Justus CR, Marie MA, Sanderlin EJ, Yang LV. The Roles of Proton-Sensing G-Protein-Coupled Receptors in Inflammation and Cancer. Genes (Basel) 2024; 15:1151. [PMID: 39336742 PMCID: PMC11431078 DOI: 10.3390/genes15091151] [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: 08/01/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
The precise regulation of pH homeostasis is crucial for normal physiology. However, in tissue microenvironments, it can be impacted by pathological conditions such as inflammation and cancer. Due to the overproduction and accumulation of acids (protons), the extracellular pH is characteristically more acidic in inflamed tissues and tumors in comparison to normal tissues. A family of proton-sensing G-protein-coupled receptors (GPCRs) has been identified as molecular sensors for cells responding to acidic tissue microenvironments. Herein, we review the current research progress pertaining to these proton-sensing GPCRs, including GPR4, GPR65 (TDAG8), and GPR68 (OGR1), in inflammation and cancer. Growing evidence suggests that GPR4 and GPR68 are mainly pro-inflammatory, whereas GPR65 is primarily anti-inflammatory, in various inflammatory disorders. Both anti- and pro-tumorigenic effects have been reported for this family of receptors. Moreover, antagonists and agonists targeting proton-sensing GPCRs have been developed and evaluated in preclinical models. Further research is warranted to better understand the roles of these proton-sensing GPCRs in pathophysiology and is required in order to exploit them as potential therapeutic targets for disease treatment.
Collapse
Affiliation(s)
- Calvin R Justus
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Mona A Marie
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Edward J Sanderlin
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Li V Yang
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| |
Collapse
|
31
|
Tricou LP, Al-Hawat ML, Cherifi K, Manrique G, Freedman BR, Matoori S. Wound pH-Modulating Strategies for Diabetic Wound Healing. Adv Wound Care (New Rochelle) 2024; 13:446-462. [PMID: 38149883 PMCID: PMC11535470 DOI: 10.1089/wound.2023.0129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 12/22/2023] [Indexed: 12/28/2023] Open
Abstract
Significance: Chronic diabetic wounds on the lower extremities (diabetic foot ulcers, DFU) are one of the most prevalent and life-threatening complications of diabetes, responsible for significant loss of quality of life and cost to the health care system. Available pharmacologic treatments fail to achieve complete healing in many patients. Recent studies and investigational treatments have highlighted the potential of modulating wound pH in DFU. Recent Advances: Data from in vitro, preclinical, and clinical studies highlight the role of pH in the pathophysiology of DFU, and topical administration of pH-lowering agents have shown promise as a therapeutic strategy for diabetic wounds. In this critical review, we describe the role of pH in DFU pathophysiology and present selected low-molecular-weight and hydrogel-based pH-modulating systems for wound healing and infection control in diabetic wounds. Critical Issues: The molecular mechanisms leading to pH alterations in diabetic wounds are complex and may differ between in vitro models, animal models of diabetes, and the human pathophysiology. Wound pH-lowering bandages for DFU therapy must be tested in established animal models of diabetic wound healing and patients with diabetes to establish a comprehensive benefit-risk profile. Future Directions: As our understanding of the role of pH in the pathophysiology of diabetic wounds is deepening, new treatments for this therapeutic target are being developed and will be tested in preclinical and clinical studies. These therapeutic systems will establish a target product profile for pH-lowering treatments such as an optimal pH profile for each wound healing stage. Thus, controlling wound bed pH could become a powerful tool to accelerate chronic diabetic wound healing.
Collapse
Affiliation(s)
- Léo-Paul Tricou
- Faculté de Pharmacie, Université de Montréal, Montréal, Canada
- ISPB Faculté de Pharmacie, Université Claude Bernard Lyon 1, Lyon, France
- Chemical Engineering Department, Polytechnique Montreal, Montréal, Canada
| | | | - Katia Cherifi
- Faculté de Pharmacie, Université de Montréal, Montréal, Canada
| | | | - Benjamin R. Freedman
- Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Simon Matoori
- Faculté de Pharmacie, Université de Montréal, Montréal, Canada
| |
Collapse
|
32
|
Matsingos C, Howell LA, McCormick PJ, Fornili A. Elucidating the Activation Mechanism of the Proton-sensing GPR68 Receptor. J Mol Biol 2024; 436:168688. [PMID: 38936694 DOI: 10.1016/j.jmb.2024.168688] [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: 12/13/2023] [Revised: 06/08/2024] [Accepted: 06/20/2024] [Indexed: 06/29/2024]
Abstract
GPR68 is a proton-sensing G-protein Coupled Receptor (GPCR) involved in a variety of physiological processes and disorders including neoplastic pathologies. While GPR68 and few other GPCRs have been shown to be activated by a decrease in the extracellular pH, the molecular mechanism of their activation remains largely unknown. In this work, we used a combined computational and in vitro approach to provide new insight into the activation mechanism of the receptor. Molecular Dynamics simulations of GPR68 were used to model the changes in residue interactions and motions triggered by pH. Global and local rearrangements consistent with partial activation were observed upon protonation of the inactive state. Selected extracellular histidine and transmembrane acidic residues were found to have significantly upshifted pKa values during the simulations, consistently with their previously hypothesised role in activation through changes in protonation state. Moreover, a novel pairing between histidine and acidic residues in the extracellular region was highlighted by both sequence analyses and simulation data and tested through site-directed mutagenesis. At last, we identified a previously unknown hydrophobic lock in the extracellular region that might stabilise the inactive conformation and regulate the transition to the active state.
Collapse
Affiliation(s)
- Christos Matsingos
- Department of Chemistry, School of Physical and Chemical Sciences, Queen Mary University of London, London E1 4NS, United Kingdom.
| | - Lesley A Howell
- Department of Chemistry, School of Physical and Chemical Sciences, Queen Mary University of London, London E1 4NS, United Kingdom
| | - Peter J McCormick
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom; Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool L69 7BE, United Kingdom
| | - Arianna Fornili
- Department of Chemistry, School of Physical and Chemical Sciences, Queen Mary University of London, London E1 4NS, United Kingdom.
| |
Collapse
|
33
|
Yu S, Liu D, Yan C, Yuan C, Zhang C, Zheng S. A novel mutation in GPR68 causes hypomaturation amelogenesis imperfecta. Arch Oral Biol 2024; 164:105991. [PMID: 38761453 DOI: 10.1016/j.archoralbio.2024.105991] [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: 01/13/2024] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 05/20/2024]
Abstract
OBJECTIVES To identify the genetic cause of a Chinese family with hypomaturation amelogenesis imperfecta (AI) and to characterize the structure of GPR68 mutated enamel in order to develop a deeper understanding of the role of the GPR68 protein during the intricate process of amelogenesis. DESIGN One Chinese family with generalized hypomaturation AI was recruited. Two of the third molars from the proband were subjected to scanning electron microscopy (SEM) and energy dispersive X-ray spectroscopy (EDX). Whole exome sequencing (WES) was performed, and the identified mutation was confirmed by Sanger sequencing. Bioinformatics studies were further conducted to analyze the potential deleterious effects of the mutation. RESULTS The proband presented with a hypomaturation AI phenotype, characterized by fragile and discolored enamel surface. The AI enamel showed prismatic structure, which was sporadically obscured by areas of amorphous material and porous structure. EDX analysis showed the proband's enamel demonstrated a significant decrease in calcium and phosphorus content and a significant increase in oxygen compared with normal enamel. A novel homozygous mutation of G protein-coupled receptor 68 (GPR68) (c .149 T > A, p.Ile50Asn) was identified in the proband. Bioinformatics analysis indicated that the mutation site displayed a high level of evolutionary conservation among species, and the mutation might impact the stability and conformation of the protein. CONCLUSION The novel homozygous GPR68 mutation resulted in hypomaturation AI. We first described the effect of GPR68 mutation on enamel structure. Our results provide new genetic evidence that mutations involved in GPR68 contribute to hypomaturation AI.
Collapse
Affiliation(s)
- Shunlan Yu
- Department of Preventive Dentistry, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, PR China
| | - Dandan Liu
- Department of Preventive Dentistry, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, PR China
| | - Changqing Yan
- Department of Preventive Dentistry, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, PR China
| | - Chao Yuan
- Department of Preventive Dentistry, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, PR China
| | - Chenying Zhang
- Department of Preventive Dentistry, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, PR China.
| | - Shuguo Zheng
- Department of Preventive Dentistry, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, PR China.
| |
Collapse
|
34
|
Weng W, Fu J, Cheng F, Wang Y, Zhang J. Integrated Bulk and Single-Cell RNA-Sequencing Reveals the Effects of Circadian Rhythm Disruption on the Metabolic Reprogramming of CD4+ T Cells in Alzheimer's Disease. Mol Neurobiol 2024; 61:6013-6030. [PMID: 38265551 DOI: 10.1007/s12035-023-03907-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 12/22/2023] [Indexed: 01/25/2024]
Abstract
Although growing evidence suggests close correlations between Alzheimer's disease (AD) and circadian rhythm disruption (CRD), few studies have focused on the influence of circadian rhythm on levels of immune cells in AD. We aimed to delineate the mechanism underlying the effects of circadian related genes on T cell immune function in AD. A total of 112 brain samples were used to construct the CRD-related model by performing weighted gene co-expression network analysis and machine learning algorithms (LASSO, SVM-RFE, and RF). The ssGSEA method was used to calculate the CRDscore in order to quantify CRD status. Using single-cell transcriptome data of CSF cells, we investigated the CD4+ T cell metabolism and cell-cell communication in high- and low-risk CRD groups. Connectivity map (CMap) was applied to explore small molecule drugs targeting CRD, and the expression of the signature gene GPR4 was further validated in AD. The CRDscore algorithm, which is based on 23 circadian-related genes, can effectively classify the CRD status in AD datasets. The single-cell analysis revealed that the CD4+ T cells with high CRDscore were characterized by hypometabolism. Cell communication analysis revealed that CD4+ T cells might be involved in promoting CD8+ T cell adhesion under CRD, which may facilitate T cell infiltration into the brain parenchyma. Overall, this study indicates the potential connotation of circadian rhythm in AD, providing insights into understanding T cell metabolic reprogramming under CRD.
Collapse
Affiliation(s)
- Weipin Weng
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha, China
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jianhan Fu
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Fan Cheng
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha, China
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yixuan Wang
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha, China
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Jie Zhang
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha, China.
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China.
- Department of Neurology, Turpan City People's Hospital, Tulufan, China.
| |
Collapse
|
35
|
Neale I, Reddy C, Tan ZY, Li B, Nag PP, Park J, Park J, Carey KL, Graham DB, Xavier RJ. Small-molecule probe for IBD risk variant GPR65 I231L alters cytokine signaling networks through positive allosteric modulation. SCIENCE ADVANCES 2024; 10:eadn2339. [PMID: 39028811 PMCID: PMC11259170 DOI: 10.1126/sciadv.adn2339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 06/13/2024] [Indexed: 07/21/2024]
Abstract
The proton-sensing heterotrimeric guanine nucleotide-binding protein-coupled receptor GPR65 is expressed in immune cells and regulates tissue homeostasis in response to decreased extracellular pH, which occurs in the context of inflammation and tumorigenesis. Genome-wide association studies linked GPR65 to several autoimmune and inflammatory diseases such as multiple sclerosis and inflammatory bowel disease (IBD). The loss-of-function GPR65 I231L IBD risk variant alters cellular metabolism, impairs protective tissue functions, and increases proinflammatory cytokine production. Hypothesizing that a small molecule designed to potentiate GPR65 at subphysiological pH could decrease inflammatory responses, we found positive allosteric modulators of GPR65 that engage and activate both human and mouse orthologs of the receptor. We observed that the chemical probe BRD5075 alters cytokine and chemokine programs in dendritic cells, establishing that immune signaling can be modulated by targeting GPR65. Our investigation offers improved chemical probes to further interrogate the biology of human GPR65 and its clinically relevant genetic variants.
Collapse
Affiliation(s)
- Ilona Neale
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Clark Reddy
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Zher Yin Tan
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Bihua Li
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Partha P. Nag
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Joshua Park
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jihye Park
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Daniel B. Graham
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Ramnik J. Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
36
|
Hernandez-Olmos V, Heering J, Marinescu B, Schermeng T, Ivanov VV, Moroz YS, Nevermann S, Mathes M, Ehrler JHM, Alnouri MW, Wolf M, Haydo AS, Schmachtel T, Zaliani A, Höfner G, Kaiser A, Schubert-Zsilavecz M, Beck-Sickinger AG, Offermanns S, Gribbon P, Rieger MA, Merk D, Sisignano M, Steinhilber D, Proschak E. Development of a Potent and Selective G2A (GPR132) Agonist. J Med Chem 2024; 67:10567-10588. [PMID: 38917049 PMCID: PMC11249017 DOI: 10.1021/acs.jmedchem.3c02164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 04/08/2024] [Accepted: 04/17/2024] [Indexed: 06/27/2024]
Abstract
G protein-coupled receptor G2A was postulated to be a promising target for the development of new therapeutics in neuropathic pain, acute myeloid leukemia, and inflammation. However, there is still a lack of potent, selective, and drug-like G2A agonists to be used as a chemical tool or as the starting matter for the development of drugs. In this work, we present the discovery and structure-activity relationship elucidation of a new potent and selective G2A agonist scaffold. Systematic optimization resulted in (3-(pyridin-3-ylmethoxy)benzoyl)-d-phenylalanine (T-10418) exhibiting higher potency than the reference and natural ligand 9-HODE and high selectivity among G protein-coupled receptors. With its favorable activity, a clean selectivity profile, excellent solubility, and high metabolic stability, T-10418 qualifies as a pharmacological tool to investigate the effects of G2A activation.
Collapse
Affiliation(s)
- Victor Hernandez-Olmos
- Fraunhofer
Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer
Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Jan Heering
- Fraunhofer
Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer
Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Beatrice Marinescu
- Institute
of Pharmaceutical Chemistry, Goethe University
Frankfurt, Max-von-Laue-Street
9, 60438 Frankfurt
am Main, Germany
| | - Tina Schermeng
- Institute
of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstraße 34, 04103 Leipzig, Germany
| | | | - Yurii S. Moroz
- Taras Shevchenko
National University of Kyiv, 64 Volodymyrska Street, Kyiv 01601, Ukraine
- Chemspace
LLC, 85 Chervonotkatska
Street, Kyiv 02094, Ukraine
| | - Sheila Nevermann
- Fraunhofer
Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Marius Mathes
- Institute
of Pharmaceutical Chemistry, Goethe University
Frankfurt, Max-von-Laue-Street
9, 60438 Frankfurt
am Main, Germany
| | - Johanna H. M. Ehrler
- Institute
of Pharmaceutical Chemistry, Goethe University
Frankfurt, Max-von-Laue-Street
9, 60438 Frankfurt
am Main, Germany
| | - Mohamad Wessam Alnouri
- Department
of Pharmacology, Max Planck Institute for
Heart and Lung Research, Ludwigstr. 43, 61231Bad Nauheim, Germany
| | - Markus Wolf
- Fraunhofer
Institute for Translational Medicine and Pharmacology ITMP, Discovery
Research ScreeningPort, Schnackenburgallee 114, 22525 Hamburg, Germany
| | - Alicia S. Haydo
- Department
of Medicine, Hematology/Oncology, Goethe
University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Tessa Schmachtel
- Department
of Medicine, Hematology/Oncology, Goethe
University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Andrea Zaliani
- Fraunhofer
Institute for Translational Medicine and Pharmacology ITMP, Discovery
Research ScreeningPort, Schnackenburgallee 114, 22525 Hamburg, Germany
| | - Georg Höfner
- Department of Pharmacy, Ludwig-Maximilians-Universität
München, Butenandtstr. 5-13, 81377 Munich, Germany
| | - Astrid Kaiser
- Institute
of Pharmaceutical Chemistry, Goethe University
Frankfurt, Max-von-Laue-Street
9, 60438 Frankfurt
am Main, Germany
| | - Manfred Schubert-Zsilavecz
- Institute
of Pharmaceutical Chemistry, Goethe University
Frankfurt, Max-von-Laue-Street
9, 60438 Frankfurt
am Main, Germany
| | - Annette G. Beck-Sickinger
- Institute
of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstraße 34, 04103 Leipzig, Germany
| | - Stefan Offermanns
- Department
of Pharmacology, Max Planck Institute for
Heart and Lung Research, Ludwigstr. 43, 61231Bad Nauheim, Germany
- Center for Molecular Medicine, Goethe University
Frankfurt, Theodor-Stern-Kai
7, 60590 Frankfurt, Germany
| | - Philipp Gribbon
- Fraunhofer
Institute for Translational Medicine and Pharmacology ITMP, Discovery
Research ScreeningPort, Schnackenburgallee 114, 22525 Hamburg, Germany
| | - Michael A. Rieger
- Fraunhofer
Institute for Translational Medicine and Pharmacology ITMP, Discovery
Research ScreeningPort, Schnackenburgallee 114, 22525 Hamburg, Germany
- Frankfurt Cancer Institute, 60590 Frankfurt
am Main, Germany
- Cardio-Pulmonary Institute (CPI), 60590 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK) and German
Cancer Research Institute
(DKFZ), Im Neuenheimer
Feld 280, 69120 Heidelberg, Germany
| | - Daniel Merk
- Department of Pharmacy, Ludwig-Maximilians-Universität
München, Butenandtstr. 5-13, 81377 Munich, Germany
| | - Marco Sisignano
- Pharmazentrum
Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe University, Theodor-Stern-Kai
7, 60590 Frankfurt
am Main, Germany
| | - Dieter Steinhilber
- Fraunhofer
Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer
Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Institute
of Pharmaceutical Chemistry, Goethe University
Frankfurt, Max-von-Laue-Street
9, 60438 Frankfurt
am Main, Germany
| | - Ewgenij Proschak
- Fraunhofer
Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer
Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Institute
of Pharmaceutical Chemistry, Goethe University
Frankfurt, Max-von-Laue-Street
9, 60438 Frankfurt
am Main, Germany
| |
Collapse
|
37
|
Liang S, Zhou J, Cao C, Liu Y, Ming S, Liu X, Shang Y, Lao J, Peng Q, Yang J, Wu M. GITR exacerbates lysophosphatidylcholine-induced macrophage pyroptosis in sepsis via posttranslational regulation of NLRP3. Cell Mol Immunol 2024; 21:674-688. [PMID: 38740925 PMCID: PMC11214634 DOI: 10.1038/s41423-024-01170-w] [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: 11/03/2023] [Accepted: 04/16/2024] [Indexed: 05/16/2024] Open
Abstract
The NLRP3 inflammasome functions as an inflammatory driver, but its relationship with lipid metabolic changes in early sepsis remains unclear. Here, we found that GITR expression in monocytes/macrophages was induced by lysophosphatidylcholine (LPC) and was positively correlated with the severity of sepsis. GITR is a costimulatory molecule that is mainly expressed on T cells, but its function in macrophages is largely unknown. Our in vitro data showed that GITR enhanced LPC uptake by macrophages and specifically enhanced NLRP3 inflammasome-mediated macrophage pyroptosis. Furthermore, in vivo studies using either cecal ligation and puncture (CLP) or LPS-induced sepsis models demonstrated that LPC exacerbated sepsis severity/lethality, while conditional knockout of GITR in myeloid cells or NLRP3/caspase-1/IL-1β deficiency attenuated sepsis severity/lethality. Mechanistically, GITR specifically enhanced inflammasome activation by regulating the posttranslational modification (PTM) of NLRP3. GITR competes with NLRP3 for binding to the E3 ligase MARCH7 and recruits MARCH7 to induce deacetylase SIRT2 degradation, leading to decreasing ubiquitination but increasing acetylation of NLRP3. Overall, these findings revealed a novel role of macrophage-derived GITR in regulating the PTM of NLRP3 and systemic inflammatory injury, suggesting that GITR may be a potential therapeutic target for sepsis and other inflammatory diseases. GITR exacerbates LPC-induced macrophage pyroptosis in sepsis via posttranslational regulation of NLRP3. According to the model, LPC levels increase during the early stage of sepsis, inducing GITR expression on macrophages. GITR not only competes with NLRP3 for binding to the E3 ligase MARCH7 but also recruits MARCH7 to induce the degradation of the deacetylase SIRT2, leading to decreasing ubiquitination but increasing acetylation of NLRP3 and therefore exacerbating LPC-induced NLRP3 inflammasome activation, macrophage pyroptosis and systemic inflammatory injury.
Collapse
Affiliation(s)
- Siping Liang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Jinyu Zhou
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Can Cao
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Yiting Liu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Siqi Ming
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Xi Liu
- Department of Infectious Diseases, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Yuqi Shang
- Department of Infectious Diseases, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Juanfeng Lao
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Qin Peng
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Jiahui Yang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Minhao Wu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China.
| |
Collapse
|
38
|
Yoshida Y, Fukuoka K, Sakugawa M, Kurogi M, Hamamura K, Hamasaki K, Tsurusaki F, Sotono K, Nishi T, Fukuda T, Kumamoto T, Oyama K, Ogino T, Tsuruta A, Mayanagi K, Yamashita T, Fuchino H, Kawahara N, Yoshimatsu K, Kawakami H, Koyanagi S, Matsunaga N, Ohdo S. Inhibition of G protein-coupled receptor 68 using homoharringtonine attenuates chronic kidney disease-associated cardiac impairment. Transl Res 2024; 269:31-46. [PMID: 38401836 DOI: 10.1016/j.trsl.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/22/2023] [Accepted: 02/20/2024] [Indexed: 02/26/2024]
Abstract
Chronic kidney disease (CKD) induces cardiac inflammation and fibrosis and reduces survival. We previously demonstrated that G protein-coupled receptor 68 (GPR68) promotes cardiac inflammation and fibrosis in mice with 5/6 nephrectomy (5/6Nx) and patients with CKD. However, no method of GPR68 inhibition has been found that has potential for therapeutic application. Here, we report that Cephalotaxus harringtonia var. nana extract and homoharringtonine ameliorate cardiac inflammation and fibrosis under CKD by suppressing GPR68 function. Reagents that inhibit the function of GPR68 were explored by high-throughput screening using a medicinal plant extract library (8,008 species), and we identified an extract from Cephalotaxus harringtonia var. nana as a GPR68 inhibitor that suppresses inflammatory cytokine production in a GPR68 expression-dependent manner. Consumption of the extract inhibited inflammatory cytokine expression and cardiac fibrosis and improved the decreased survival attributable to 5/6Nx. Additionally, homoharringtonine, a cephalotaxane compound characteristic of C. harringtonia, inhibited inflammatory cytokine production. Homoharringtonine administration in drinking water alleviated cardiac fibrosis and improved heart failure and survival in 5/6Nx mice. A previously unknown effect of C. harringtonia extract and homoharringtonine was revealed in which GPR68-dependent inflammation and cardiac dysfunction were suppressed. Utilizing these compounds could represent a new strategy for treating GPR68-associated diseases, including CKD.
Collapse
Affiliation(s)
- Yuya Yoshida
- Department of Clinical Pharmacokinetics, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Kohei Fukuoka
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Miyu Sakugawa
- Department of Clinical Pharmacokinetics, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Masayuki Kurogi
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Kengo Hamamura
- Department of Clinical Pharmacokinetics, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Keika Hamasaki
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Fumiaki Tsurusaki
- Department of Clinical Pharmacokinetics, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Kurumi Sotono
- Department of Clinical Pharmacokinetics, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Takumi Nishi
- Department of Clinical Pharmacokinetics, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Taiki Fukuda
- Department of Clinical Pharmacokinetics, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Taisei Kumamoto
- Department of Clinical Pharmacokinetics, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Kosuke Oyama
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Takashi Ogino
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Akito Tsuruta
- Department of Glocal Healthcare Science, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Kouta Mayanagi
- Department of Drug Discovery Structural Biology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Tomohiro Yamashita
- Department of Drug Discovery Structural Biology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Hiroyuki Fuchino
- Tsukuba Division, Research Center for Medicinal Plant Resources, National Institutes of Biomedical Innovation, Health and Nutrition, 1-2 Hachimandai, Tsukuba, Ibaraki 305-0843, Japan
| | - Nobuo Kawahara
- Tsukuba Division, Research Center for Medicinal Plant Resources, National Institutes of Biomedical Innovation, Health and Nutrition, 1-2 Hachimandai, Tsukuba, Ibaraki 305-0843, Japan; The Kochi Prefectural Makino Botanical Garden, 4200-6, Godaisan, Kochi 781-8125, Japan
| | - Kayo Yoshimatsu
- Tsukuba Division, Research Center for Medicinal Plant Resources, National Institutes of Biomedical Innovation, Health and Nutrition, 1-2 Hachimandai, Tsukuba, Ibaraki 305-0843, Japan
| | - Hitomi Kawakami
- Tsukuba Division, Research Center for Medicinal Plant Resources, National Institutes of Biomedical Innovation, Health and Nutrition, 1-2 Hachimandai, Tsukuba, Ibaraki 305-0843, Japan
| | - Satoru Koyanagi
- Department of Glocal Healthcare Science, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Naoya Matsunaga
- Department of Clinical Pharmacokinetics, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan.
| | - Shigehiro Ohdo
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan.
| |
Collapse
|
39
|
Pissas KP, Gründer S, Tian Y. Functional expression of the proton sensors ASIC1a, TMEM206, and OGR1 together with BK Ca channels is associated with cell volume changes and cell death under strongly acidic conditions in DAOY medulloblastoma cells. Pflugers Arch 2024; 476:923-937. [PMID: 38627262 PMCID: PMC11139714 DOI: 10.1007/s00424-024-02964-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/25/2024] [Accepted: 04/05/2024] [Indexed: 06/01/2024]
Abstract
Fast growing solid tumors are frequently surrounded by an acidic microenvironment. Tumor cells employ a variety of mechanisms to survive and proliferate under these harsh conditions. In that regard, acid-sensitive membrane receptors constitute a particularly interesting target, since they can affect cellular functions through ion flow and second messenger cascades. Our knowledge of these processes remains sparse, however, especially regarding medulloblastoma, the most common pediatric CNS malignancy. In this study, using RT-qPCR, whole-cell patch clamp, and Ca2+-imaging, we uncovered several ion channels and a G protein-coupled receptor, which were regulated directly or indirectly by low extracellular pH in DAOY and UW228 medulloblastoma cells. Acidification directly activated acid-sensing ion channel 1a (ASIC1a), the proton-activated Cl- channel (PAC, ASOR, or TMEM206), and the proton-activated G protein-coupled receptor OGR1. The resulting Ca2+ signal secondarily activated the large conductance calcium-activated potassium channel (BKCa). Our analyses uncover a complex relationship of these transmembrane proteins in DAOY cells that resulted in cell volume changes and induced cell death under strongly acidic conditions. Collectively, our results suggest that these ion channels in concert with OGR1 may shape the growth and evolution of medulloblastoma cells in their acidic microenvironment.
Collapse
Affiliation(s)
| | - Stefan Gründer
- Institute of Physiology, RWTH Aachen University, Pauwelsstraße 30, 52074, Aachen, Germany.
| | - Yuemin Tian
- Institute of Physiology, RWTH Aachen University, Pauwelsstraße 30, 52074, Aachen, Germany
| |
Collapse
|
40
|
Li MS, Wang XH, Wang H. Immunomodulation of Proton-activated G Protein-coupled Receptors in Inflammation. Curr Med Sci 2024; 44:475-484. [PMID: 38748372 DOI: 10.1007/s11596-024-2872-4] [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: 12/13/2023] [Accepted: 03/22/2024] [Indexed: 06/29/2024]
Abstract
Proton-activated G protein-coupled receptors (GPCRs), initially discovered by Ludwig in 2003, are widely distributed in various tissues. These receptors have been found to modulate the immune system in several inflammatory diseases, including inflammatory bowel disease, atopic dermatitis, and asthma. Proton-activated GPCRs belong to the G protein-coupled receptor family and can detect alternations in extracellular pH. This detection triggers downstream signaling pathways within the cells, ultimately influencing the function of immune cells. In this review, we specifically focused on investigating the immune response of proton-activated GPCRs under inflammatory conditions.
Collapse
Affiliation(s)
- Min-Shan Li
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Clinical Research Center for Nasal Inflammatory Diseases, Wuhan, 430030, China
| | - Xiang-Hong Wang
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Clinical Research Center for Nasal Inflammatory Diseases, Wuhan, 430030, China
| | - Heng Wang
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Clinical Research Center for Nasal Inflammatory Diseases, Wuhan, 430030, China.
| |
Collapse
|
41
|
Howard MK, Hoppe N, Huang XP, Macdonald CB, Mehrota E, Grimes PR, Zahm A, Trinidad DD, English J, Coyote-Maestas W, Manglik A. Molecular basis of proton-sensing by G protein-coupled receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.17.590000. [PMID: 38659943 PMCID: PMC11042331 DOI: 10.1101/2024.04.17.590000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Three proton-sensing G protein-coupled receptors (GPCRs), GPR4, GPR65, and GPR68, respond to changes in extracellular pH to regulate diverse physiology and are implicated in a wide range of diseases. A central challenge in determining how protons activate these receptors is identifying the set of residues that bind protons. Here, we determine structures of each receptor to understand the spatial arrangement of putative proton sensing residues in the active state. With a newly developed deep mutational scanning approach, we determined the functional importance of every residue in proton activation for GPR68 by generating ~9,500 mutants and measuring effects on signaling and surface expression. This unbiased screen revealed that, unlike other proton-sensitive cell surface channels and receptors, no single site is critical for proton recognition in GPR68. Instead, a network of titratable residues extend from the extracellular surface to the transmembrane region and converge on canonical class A GPCR activation motifs to activate proton-sensing GPCRs. More broadly, our approach integrating structure and unbiased functional interrogation defines a new framework for understanding the rich complexity of GPCR signaling.
Collapse
Affiliation(s)
- Matthew K. Howard
- Tetrad graduate program, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Science, University of California, San Francisco, CA, USA
| | - Nicholas Hoppe
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Biophysics graduate program, University of California, San Francisco, CA, USA
| | - Xi-Ping Huang
- Department of Pharmacology and the National Institute of Mental Health Psychoactive Drug Screening Program (NIMH PDSP), The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Christian B. Macdonald
- Department of Bioengineering and Therapeutic Science, University of California, San Francisco, CA, USA
| | - Eshan Mehrota
- Tetrad graduate program, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Medical Scientist Training Program, University of California, San Francisco, CA, USA
| | | | - Adam Zahm
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Donovan D. Trinidad
- Department of Medicine, Division of Infectious Disease, University of California, San Francisco, United States
| | - Justin English
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Willow Coyote-Maestas
- Department of Bioengineering and Therapeutic Science, University of California, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Quantitative Biosciences Institute, University of California, San Francisco, USA
| | - Aashish Manglik
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Quantitative Biosciences Institute, University of California, San Francisco, USA
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA, USA
| |
Collapse
|
42
|
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
|
43
|
Otsugu M, Mine A, Uchida I, Miyake Y, Tachihara R, Fujiwara K, Ichimura A, Sato K, Tomura H. Low pH modulates lipopolysaccharide-induced tumor necrosis factor-alpha expression and macropinocytotic activity in RAW264.7 cells. J Recept Signal Transduct Res 2024; 44:63-71. [PMID: 39175331 DOI: 10.1080/10799893.2024.2395310] [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: 03/22/2024] [Revised: 08/12/2024] [Accepted: 08/17/2024] [Indexed: 08/24/2024]
Abstract
Inflammation triggers various types of diseases that need to be addressed. Macrophages play important roles in the inflammatory responses. As atherosclerosis progresses, macrophages transform into foam cells. Extracellular acidification is observed at and around bacterial infection and atherosclerotic sites. However, the effects of acidification on the inflammatory response of macrophages and the progression of atherosclerosis have not been fully understood. This study investigates the impact of extracellular acidification on lipopolysaccharide-induced tumor necrosis factor-alpha (TNF-α) expression and macropinocytotic activity in RAW264.7 cells. TNF-α expression is measured by real-time polymerase chain reaction (relative value to glyceraldehyde-3-phosphate dehydrogenase expression). Macropinocytotic activity is measured by neutral red uptake (absorbance at 540 nm). Results show that TNF-α expression increased with decreasing extracellular pH in both un-foamed and foamed cells. Macropinocytotic activity was upregulated at pH 6.8 in un-foamed cells, but downregulated in foamed cells stimulated at low pH. Proton-sensing G protein-coupled receptors (GPCRs) were involved in the expression of TNF-α and in the macropinocytotic activity of foamed cells. In conclusion, this study reveals that extracellular acidification differently affect various inflammatory responses such as LPS-induced TNF-α expression and macropinocytotic activity of RAW264.7 cells and different proton-sensing GPCRs are involved in the different inflammatory responses.
Collapse
Affiliation(s)
- Miku Otsugu
- Laboratory of Cell Signaling Regulation, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, Japan
| | - Ayumi Mine
- Laboratory of Cell Signaling Regulation, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, Japan
| | - Izumi Uchida
- Laboratory of Cell Signaling Regulation, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, Japan
| | - Yuta Miyake
- Laboratory of Cell Signaling Regulation, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, Japan
| | - Ryo Tachihara
- Laboratory of Cell Signaling Regulation, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, Japan
| | - Kurumi Fujiwara
- Laboratory of Cell Signaling Regulation, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, Japan
| | - Ayako Ichimura
- Laboratory of Cell Signaling Regulation, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, Japan
| | - Koichi Sato
- Laboratory of Signal Transduction, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Hideaki Tomura
- Laboratory of Cell Signaling Regulation, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, Japan
- Institute of Endocrinology, Meiji University, Kawasaki, Japan
| |
Collapse
|
44
|
Stock C. pH-regulated single cell migration. Pflugers Arch 2024; 476:639-658. [PMID: 38214759 PMCID: PMC11006768 DOI: 10.1007/s00424-024-02907-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/21/2023] [Accepted: 01/02/2024] [Indexed: 01/13/2024]
Abstract
Over the last two decades, extra- and intracellular pH have emerged as fundamental regulators of cell motility. Fundamental physiological and pathological processes relying on appropriate cell migration, such as embryonic development, wound healing, and a proper immune defense on the one hand, and autoimmune diseases, metastatic cancer, and the progression of certain parasitic diseases on the other, depend on surrounding pH. In addition, migrating single cells create their own localized pH nanodomains at their surface and in the cytosol. By this means, the migrating cells locally modulate their adhesion to, and the re-arrangement and digestion of, the extracellular matrix. At the same time, the cytosolic nanodomains tune cytoskeletal dynamics along the direction of movement resulting in concerted lamellipodia protrusion and rear end retraction. Extracellular pH gradients as found in wounds, inflamed tissues, or the periphery of tumors stimulate directed cell migration, and long-term exposure to acidic conditions can engender a more migratory and invasive phenotype persisting for hours up to several generations of cells after they have left the acidic milieu. In the present review, the different variants of pH-dependent single cell migration are described. The underlying pH-dependent molecular mechanisms such as conformational changes of adhesion molecules, matrix protease activity, actin (de-)polymerization, and signaling events are explained, and molecular pH sensors stimulated by H+ signaling are presented.
Collapse
Affiliation(s)
- Christian Stock
- Department of Gastroenterology, Hepatology, Infectiology & Endocrinology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
45
|
Becker HM, Seidler UE. Bicarbonate secretion and acid/base sensing by the intestine. Pflugers Arch 2024; 476:593-610. [PMID: 38374228 PMCID: PMC11006743 DOI: 10.1007/s00424-024-02914-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 02/21/2024]
Abstract
The transport of bicarbonate across the enterocyte cell membrane regulates the intracellular as well as the luminal pH and is an essential part of directional fluid movement in the gut. Since the first description of "active" transport of HCO3- ions against a concentration gradient in the 1970s, the fundamental role of HCO3- transport for multiple intestinal functions has been recognized. The ion transport proteins have been identified and molecularly characterized, and knockout mouse models have given insight into their individual role in a variety of functions. This review describes the progress made in the last decade regarding novel techniques and new findings in the molecular regulation of intestinal HCO3- transport in the different segments of the gut. We discuss human diseases with defects in intestinal HCO3- secretion and potential treatment strategies to increase luminal alkalinity. In the last part of the review, the cellular and organismal mechanisms for acid/base sensing in the intestinal tract are highlighted.
Collapse
Affiliation(s)
- Holger M Becker
- Department of Gastroenterology, Hannover Medical School, 30625, Hannover, Germany
| | - Ursula E Seidler
- Department of Gastroenterology, Hannover Medical School, 30625, Hannover, Germany.
| |
Collapse
|
46
|
Boedtkjer E, Ara T. Strengthening the basics: acids and bases influence vascular structure and function, tissue perfusion, blood pressure, and human cardiovascular disease. Pflugers Arch 2024; 476:623-637. [PMID: 38383822 DOI: 10.1007/s00424-024-02926-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 02/23/2024]
Abstract
Acids and their conjugate bases accumulate in or dissipate from the interstitial space when tissue perfusion does not match the metabolic demand. Extracellular acidosis dilates most arterial beds, but associated acid-base disturbances-e.g., intracellular acidification and decreases in HCO3- concentration-can also elicit pro-contractile influences that diminish vasodilation and even dominate in some vascular beds to cause vasoconstriction. The ensemble activities of the acid-base-sensitive reactions in vascular smooth muscle and endothelial cells optimize vascular resistance for blood pressure control and direct the perfusion towards active tissue. In this review, we describe the mechanisms of intracellular pH regulation in the vascular wall and discuss how vascular smooth muscle and endothelial cells sense acid-base disturbances. We further deliberate on the functional effects of local acid-base disturbances and their integrated cardiovascular consequences under physiological and pathophysiological conditions. Finally, we address how mutations and polymorphisms in the molecular machinery that regulates pH locally and senses acid-base disturbances in the vascular wall can result in cardiovascular disease. Based on the emerging molecular insight, we propose that targeting local pH-dependent effectors-rather than systemic acid-base disturbances-has therapeutic potential to interfere with the progression and reduce the severity of cardiovascular disease.
Collapse
Affiliation(s)
- Ebbe Boedtkjer
- Department of Biomedicine, Aarhus University, Hoegh-Guldbergs Gade 10, DK-8000, Aarhus, Denmark.
| | - Tarannum Ara
- Department of Biomedicine, Aarhus University, Hoegh-Guldbergs Gade 10, DK-8000, Aarhus, Denmark
| |
Collapse
|
47
|
Hausmann M, Seuwen K, de Vallière C, Busch M, Ruiz PA, Rogler G. Role of pH-sensing receptors in colitis. Pflugers Arch 2024; 476:611-622. [PMID: 38514581 PMCID: PMC11006753 DOI: 10.1007/s00424-024-02943-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/06/2024] [Accepted: 03/06/2024] [Indexed: 03/23/2024]
Abstract
Low pH in the gut is associated with severe inflammation, fibrosis, and colorectal cancer (CRC) and is a hallmark of active inflammatory bowel disease (IBD). Subsequently, pH-sensing mechanisms are of interest for the understanding of IBD pathophysiology. Tissue hypoxia and acidosis-two contributing factors to disease pathophysiology-are linked to IBD, and understanding their interplay is highly relevant for the development of new therapeutic options. One member of the proton-sensing G protein-coupled receptor (GPCR) family, GPR65 (T-cell death-associated gene 8, TDAG8), was identified as a susceptibility gene for IBD in a large genome-wide association study. In response to acidic extracellular pH, GPR65 induces an anti-inflammatory response, whereas the two other proton-sensing receptors, GPR4 and GPR68 (ovarian cancer G protein-coupled receptor 1, OGR1), mediate pro-inflammatory responses. Here, we review the current knowledge on the role of these proton-sensing receptors in IBD and IBD-associated fibrosis and cancer, as well as colitis-associated cancer (CAC). We also describe emerging small molecule modulators of these receptors as therapeutic opportunities for the treatment of IBD.
Collapse
Affiliation(s)
- Martin Hausmann
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland.
| | - Klaus Seuwen
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland
| | - Cheryl de Vallière
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland
| | - Moana Busch
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland
| | - Pedro A Ruiz
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland
| | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland
| |
Collapse
|
48
|
Caratis F, Opiełka M, Hausmann M, Velasco-Estevez M, Rojek B, de Vallière C, Seuwen K, Rogler G, Karaszewski B, Rutkowska A. The proton-sensing receptors TDAG8 and GPR4 are differentially expressed in human and mouse oligodendrocytes: Exploring their role in neuroinflammation and multiple sclerosis. PLoS One 2024; 19:e0283060. [PMID: 38527054 PMCID: PMC10962805 DOI: 10.1371/journal.pone.0283060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 02/13/2024] [Indexed: 03/27/2024] Open
Abstract
Acidosis is one of the hallmarks of demyelinating central nervous system (CNS) lesions in multiple sclerosis (MS). The response to acidic pH is primarily mediated by a family of G protein-coupled proton-sensing receptors: OGR1, GPR4 and TDAG8. These receptors are inactive at alkaline pH, reaching maximal activation at acidic pH. Genome-wide association studies have identified a locus within the TDAG8 gene associated with several autoimmune diseases, including MS. Accordingly, we here found that expression of TDAG8, as opposed to GPR4 or OGR1, is upregulated in MS plaques. This led us to investigate the expression of TDAG8 in oligodendrocytes using mouse and human in vitro and in vivo models. We observed significant upregulation of TDAG8 in human MO3.13 oligodendrocytes during maturation and in response to acidic conditions. However, its deficiency did not impact normal myelination in the mouse CNS, and its expression remained unaltered under demyelinating conditions in mouse organotypic cerebellar slices. Notably, our data revealed no expression of TDAG8 in primary mouse oligodendrocyte progenitor cells (OPCs), in contrast to its expression in primary human OPCs. Our investigations have revealed substantial species differences in the expression of proton-sensing receptors in oligodendrocytes, highlighting the limitations of the employed experimental models in fully elucidating the role of TDAG8 in myelination and oligodendrocyte biology. Consequently, the study does not furnish robust evidence for the role of TDAG8 in such processes. Nonetheless, our findings tentatively point towards a potential association between TDAG8 and myelination processes in humans, hinting at a potential link between TDAG8 and the pathophysiology of MS and warrants further research.
Collapse
Affiliation(s)
- Fionä Caratis
- Brain Diseases Centre, Medical University of Gdansk, Gdansk, Poland
- Department of Anatomy and Neurobiology, Medical University of Gdansk, Gdansk, Poland
| | - Mikołaj Opiełka
- Brain Diseases Centre, Medical University of Gdansk, Gdansk, Poland
| | - Martin Hausmann
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Maria Velasco-Estevez
- H12O-CNIO Hematological Malignancies Group, Clinical Research Unit, Centro Nacional de Investigaciones Oncologicas (CNIO), Madrid, Spain
| | - Bartłomiej Rojek
- Department of Adult Neurology, Medical University of Gdansk & University Clinical Centre, Gdansk, Poland
| | - Cheryl de Vallière
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Klaus Seuwen
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Bartosz Karaszewski
- Brain Diseases Centre, Medical University of Gdansk, Gdansk, Poland
- Department of Adult Neurology, Medical University of Gdansk & University Clinical Centre, Gdansk, Poland
| | - Aleksandra Rutkowska
- Brain Diseases Centre, Medical University of Gdansk, Gdansk, Poland
- Department of Anatomy and Neurobiology, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
49
|
Segeritz P, Kolesnik K, Scott DJ, Collins DJ. Quantitative mechanical stimulation of GPR68 using a novel 96 well flow plugin. LAB ON A CHIP 2024; 24:1616-1625. [PMID: 38288761 DOI: 10.1039/d3lc00767g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Mechanosensitive proteins play a crucial role in a range of physiological processes, including hearing, tactile sensation and regulating blood flow. While previous work has demonstrated the mechanosensitivity of several proteins, the ability to apply precisely defined mechanical forces to cells in a consistent, replicable manner remains a significant challenge. In this work we present a novel 96-well plate-compatible plugin device for generating highly-controlled flow-based mechanical simulation of cells, which enables quantitative assessment of mechanosensitive protein function. The device is used to mechanically stimulate HEK 293T cells expressing the mechanosensitive protein GPR68, a G protein-coupled receptor. By assaying intracellular calcium levels during flow-based cell stimulation, we determine that GPR68 signalling is a function of the applied shear-force. As this approach is compatible with conventional cell culture plates and allows for simultaneous readout in a conventional fluorescence plate reader, this represents a valuable new tool to investigate mechanotransduction.
Collapse
Affiliation(s)
- Philipp Segeritz
- Department of Biomedical Engineering, The University of Melbourne, Parkville, VIC 3010, Australia.
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, VIC 3052, Australia.
| | - Kirill Kolesnik
- Department of Biomedical Engineering, The University of Melbourne, Parkville, VIC 3010, Australia.
| | - Daniel J Scott
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, VIC 3052, Australia.
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - David J Collins
- Department of Biomedical Engineering, The University of Melbourne, Parkville, VIC 3010, Australia.
- The Graeme Clark Institute, The University of Melbourne, Parkville, VIC, 3010, Australia
| |
Collapse
|
50
|
Ji R, Chang L, An C, Zhang J. Proton-sensing ion channels, GPCRs and calcium signaling regulated by them: implications for cancer. Front Cell Dev Biol 2024; 12:1326231. [PMID: 38505262 PMCID: PMC10949864 DOI: 10.3389/fcell.2024.1326231] [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: 10/25/2023] [Accepted: 02/14/2024] [Indexed: 03/21/2024] Open
Abstract
Extracellular acidification of tumors is common. Through proton-sensing ion channels or proton-sensing G protein-coupled receptors (GPCRs), tumor cells sense extracellular acidification to stimulate a variety of intracellular signaling pathways including the calcium signaling, which consequently exerts global impacts on tumor cells. Proton-sensing ion channels, and proton-sensing GPCRs have natural advantages as drug targets of anticancer therapy. However, they and the calcium signaling regulated by them attracted limited attention as potential targets of anticancer drugs. In the present review, we discuss the progress in studies on proton-sensing ion channels, and proton-sensing GPCRs, especially emphasizing the effects of calcium signaling activated by them on the characteristics of tumors, including proliferation, migration, invasion, metastasis, drug resistance, angiogenesis. In addition, we review the drugs targeting proton-sensing channels or GPCRs that are currently in clinical trials, as well as the relevant potential drugs for cancer treatments, and discuss their future prospects. The present review aims to elucidate the important role of proton-sensing ion channels, GPCRs and calcium signaling regulated by them in cancer initiation and development. This review will promote the development of drugs targeting proton-sensing channels or GPCRs for cancer treatments, effectively taking their unique advantage as anti-cancer drug targets.
Collapse
Affiliation(s)
- Renhui Ji
- Foundational and Translational Medical Research Center, Department of Allergy and General Surgery, Hohhot First Hospital, Hohhot, China
- Department of Pathophysiology, Basic Medicine College of Inner Mongolia Medical University, Hohhot, China
| | - Li Chang
- Foundational and Translational Medical Research Center, Department of Allergy and General Surgery, Hohhot First Hospital, Hohhot, China
- Department of Pathophysiology, Basic Medicine College of Inner Mongolia Medical University, Hohhot, China
| | - Caiyan An
- Foundational and Translational Medical Research Center, Department of Allergy and General Surgery, Hohhot First Hospital, Hohhot, China
| | - Junjing Zhang
- Foundational and Translational Medical Research Center, Department of Allergy and General Surgery, Hohhot First Hospital, Hohhot, China
| |
Collapse
|