1
|
Varlamova EG. Roles of selenium-containing glutathione peroxidases and thioredoxin reductases in the regulation of processes associated with glioblastoma progression. Arch Biochem Biophys 2025; 766:110344. [PMID: 39956249 DOI: 10.1016/j.abb.2025.110344] [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: 10/30/2024] [Revised: 02/07/2025] [Accepted: 02/12/2025] [Indexed: 02/18/2025]
Abstract
Glioblastoma remains the most common and aggressive primary tumor of the central nervous system in adults. Current treatment options include standard surgical resection combined with radiation/chemotherapy, but such protocol most likely only delays the inevitable. Therefore, the problem of finding therapeutic targets to prevent the occurrence and development of this severe oncological disease is currently acute. It is known that the functions of selenoproteins in the regulation of carcinogenesis processes are not unambiguous. Either they exhibit cytotoxic activity on cancer cells, or cytoprotective. A special place in the progression of oncological diseases of various etiologies is occupied by proteins of the thioredoxin and glutathione systems. These are two cellular antioxidant systems that regulate redox homeostasis, counteracting the increased production of reactive oxygen species in cells. The review reflects the latest data on the role of key enzymes of these redox systems in the regulation of processes associated with the progression of glioblastoma. A thorough consideration of these issues will expand fundamental knowledge about the functions of selenium-containing thioredoxin reductases and glutathione peroxidases in the therapy of glioblastomas and provide an understanding of the prospects for the treatment of this aggressive oncological disease.
Collapse
Affiliation(s)
- Elena G Varlamova
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", St. Institutskaya 3, Pushchino, 142290, Russia.
| |
Collapse
|
2
|
Liu X, Deng C, Deng Y, Luo X, Zhang W. Molecule-rich solutions for achieving novel non-opioid analgesics. Drug Discov Today 2025; 30:104329. [PMID: 40081520 DOI: 10.1016/j.drudis.2025.104329] [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: 05/09/2024] [Revised: 02/28/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025]
Abstract
Despite their efficacy, opioids have long been associated with risks of addiction, tolerance, and dependence, leaving an unmet clinical need for pain treatment. Efforts have been devoted to developing novel classes of pain-relieving medication that outperform current options in terms of pain relief, side-effect profiles, and potential for abuse, but with limited success. Recent advances in the neurobiology of pain have shed light on the potential of targeting non-opioid receptors involved in pain processing. In this review, we identify avenues, ranging from molecular-based approaches to molecule-rich solutions, for effectively identifying non-opioid analgesics free from the side effects associated with opioids.
Collapse
Affiliation(s)
- Xingxing Liu
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Chaoyi Deng
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research, Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yu Deng
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research, Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xudong Luo
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Pharmacy, West China Tianfu Hospital, Sichuan University, Chengdu 610213, China
| | - Wensheng Zhang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research, Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
3
|
Yu E, Oh SW, Park SH, Kwon K, Han SB, Kang SH, Lee JH, Ha H, Yoon D, Jung E, Song M, Cho JY, Lee J. The Pigmentation of Blue Light Is Mediated by Both Melanogenesis Activation and Autophagy Inhibition through OPN3-TRPV1. J Invest Dermatol 2025; 145:908-918.e6. [PMID: 39241981 DOI: 10.1016/j.jid.2024.07.034] [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: 09/21/2023] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 09/09/2024]
Abstract
Blue light, a high-energy radiation in the visible light spectrum, was recently reported to induce skin pigmentation. In this study, we investigated the involvement of TRPV1-mediated signaling along with OPN3 in blue light-induced melanogenesis as well as its signaling pathway. Operating downstream target of OPN3 in blue light-induced melanogenesis, blue light activated TRPV1 and upregulated its expression, resulting in calcium influx. Calcium ion induced the activation of calcium/calmodulin-dependent protein kinase II and MAPK. It also downregulated clusterin expression, leading to the nuclear translocation of PAX3, ultimately affecting melanin synthesis. In addition, blue light interfered with autophagy-mediated regulation of melanosomes by decreasing not only the interaction between clusterin and LC3B but the expression of activating transcription factor family. These findings demonstrate that the pigmenting effects of blue light are mediated by calcium/calmodulin-dependent protein kinase II- and MAPK-mediated signaling as well as clusterin-dependent inhibition of autophagy through OPN3-TRPV1-calcium influx, suggesting, to our knowledge, a previously unreported signaling pathway through which blue light regulates melanocyte biology. Furthermore, these results suggest that TRPV1 and clusterin could be potential therapeutic targets for blue light-induced pigmentation due to prolonged exposure to blue light.
Collapse
Affiliation(s)
- Eunbi Yu
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Korea
| | - Sae Woong Oh
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Korea
| | - See-Hyoung Park
- Department of Bio and Chemical Engineering, Hongik University, Sejong City, Korea
| | - Kitae Kwon
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Korea
| | - Su Bin Han
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Korea
| | - Su Hyun Kang
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Korea
| | - Jung Hyun Lee
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Korea
| | - Heejun Ha
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Korea
| | - Donghoon Yoon
- Myeloma Center, Department of Internal Medicine, The University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Eunsun Jung
- Biospectrum Life Science Institute, Seongnam, Korea
| | - Minkyung Song
- Integrative Research of T cells Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Korea; Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon, Korea
| | - Jae Youl Cho
- Molecular Immunology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Korea.
| | - Jongsung Lee
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Korea.
| |
Collapse
|
4
|
Liu L, Deng Y, Li Q, Cai Y, Zhang C, Zhang T, Xu G, Han M. Sympathetic nerve promotes renal fibrosis by activating M2 macrophages through β2-AR-Gsa. Clin Immunol 2025; 270:110397. [PMID: 39580043 DOI: 10.1016/j.clim.2024.110397] [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: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 11/25/2024]
Abstract
Sympathetic nervous system overactivation is directly related to renal fibrosis. This study focused on the role of and mechanism by which sympathetic signaling regulates macrophage activation, as well as the contribution to renal fibrosis. Renal denervation alleviated tubular necrosis, tubulointerstitial fibrosis, and macrophage accumulation induced by unilateral ureteral obstruction and ischemia-reperfusion injury. In vitro, norepinephrine (NE) promoted macrophage alternative (M2) polarization by activating β2-adrenergic receptor (β2-AR) and heterotrimeric G stimulatory protein α-subunit (Gsa). The effects of NE-induced macrophage M2 polarization were blocked by a β2-AR selective antagonist and Gsa siRNA. Importantly, ablation of Gsa in macrophages alleviated tubulointerstitial fibrosis, macrophage accumulation, and M2 polarization in the renal ischemia-reperfusion injury model. Sympathetic nervous system overactivation regulates M2 polarization in macrophages as an important neuroimmune mechanism of renal fibrosis. The β2-AR-Gsa signaling pathway was responsible for NE-induced macrophage M2 polarization, which may be a therapeutic target for renal fibrosis.
Collapse
Affiliation(s)
- Lele Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuanjun Deng
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Li
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Cai
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunjiang Zhang
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianjing Zhang
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Xu
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Min Han
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
5
|
Camici M, Del Duca G, Brita AC, Antinori A. Connecting dots of long COVID-19 pathogenesis: a vagus nerve- hypothalamic-pituitary- adrenal-mitochondrial axis dysfunction. Front Cell Infect Microbiol 2024; 14:1501949. [PMID: 39735263 PMCID: PMC11671747 DOI: 10.3389/fcimb.2024.1501949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 11/25/2024] [Indexed: 12/31/2024] Open
Abstract
The pathogenesis of long COVID (LC) still presents many areas of uncertainty. This leads to difficulties in finding an effective specific therapy. We hypothesize that the key to LC pathogenesis lies in the presence of chronic functional damage to the main anti-inflammatory mechanisms of our body: the three reflexes mediated by the vagus nerve, the hypothalamic-pituitary-adrenal (HPA) hormonal axis, and the mitochondrial redox status. We will illustrate that this neuro-endocrine-metabolic axis is closely interconnected and how the SARS-CoV-2 can damage it at all stages through direct, immune-inflammatory, epigenetic damage mechanisms, as well as through the reactivation of neurotropic viruses. According to our theory, the direct mitochondrial damage carried out by the virus, which replicates within these organelles, and the cellular oxidative imbalance, cannot be countered in patients who develop LC. This is because their anti-inflammatory mechanisms are inconsistent due to reduced vagal tone and direct damage to the endocrine glands of the HPA axis. We will illustrate how acetylcholine (ACh) and cortisol, with its cytoplasmatic and cellular receptors respectively, are fundamental players in the LC process. Both Ach and cortisol play multifaceted and synergistic roles in reducing inflammation. They achieve this by modulating the activity of innate and cell-mediated immunity, attenuating endothelial and platelet activation, and modulating mitochondrial function, which is crucial for cellular energy production and anti-inflammatory mechanisms. In our opinion, it is essential to study the sensitivity of the glucocorticoids receptor in people who develop LC and whether SARS-CoV-2 can cause long-term epigenetic variations in its expression and function.
Collapse
Affiliation(s)
- Marta Camici
- Clinical and Research Infectious Diseases Department, National Institute for Infectious Diseases Lazzaro Spallanzani Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Giulia Del Duca
- Clinical and Research Infectious Diseases Department, National Institute for Infectious Diseases Lazzaro Spallanzani Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Anna Clelia Brita
- Department of Clinical Psychology, National Institute for Infectious Diseases Lazzaro Spallanzani Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Andrea Antinori
- Clinical and Research Infectious Diseases Department, National Institute for Infectious Diseases Lazzaro Spallanzani Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| |
Collapse
|
6
|
Kuroyanagi G, Hioki T, Matsushima-Nishiwaki R, Omura T, Kozawa O, Tokuda H. Gallein increases prostaglandin F2α‑induced osteoprotegerin and IL‑6 secretion in osteoblasts. Biomed Rep 2024; 21:147. [PMID: 39640194 PMCID: PMC11618980 DOI: 10.3892/br.2024.1835] [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: 02/10/2024] [Accepted: 07/08/2024] [Indexed: 12/07/2024] Open
Abstract
Gallein is a known Gβγ subunit inhibitor, but its function in bone metabolism, especially in osteoblasts, and its molecular mechanism remains to be elucidated. Osteoprotegerin (OPG), which is secreted from osteoblasts, binds to nuclear factor kB receptor activator (RANK) ligand (RANKL) as a decoy receptor, prevents RANKL-RANK binding, and inhibits bone resorption. IL-6 is not only a bone resorption factor but also as a bone metabolism regulator. Prostaglandin F2α (PGF2α) promotes p44/p42 MAPK, p38 MAPK and stress-activated protein kinase/JNK phosphorylation in osteoblast-like MC3T3-E1 cells. In MC3T3-E1 cells, activated p44/p42 and p38 MAPKs promote IL-6 secretion and activated p44/p42 and p38 MAPKs and JNK promote OPG secretion. The present study aimed to investigate the effect and mechanism of gallein on PGF2α-induced OPG and IL-6 secretion using an osteoblastic MC3T3-E1 cell line. It was found that gallein significantly increased PGF2α-induced OPG and IL-6 secretion in the MC3T3-E1 cell. By contrast, fluorescein, which is a gallein-like compound that does not bind to Gβγ, did not affect PGF2α-induced OPG and IL-6 secretion. Gallein significantly improved the PGF2α-induced OPG and IL-6 mRNA expression levels. Gallein did not affect the PGF2α-activated phosphorylation of p44/p42 and p38 MAPKs and JNK. Gallein increased PGF2α-induced OPG and IL-6 secretion in osteoblasts, indicating that gallein may regulate bone remodeling via OPG/IL-6 in bone metabolism.
Collapse
Affiliation(s)
- Gen Kuroyanagi
- Department of Orthopedic Surgery, Nagoya City University, Nagoya 467-8601, Japan
- Department of Rehabilitation Medicine, Nagoya City University, Nagoya 467-8601, Japan
- Department of Pharmacology, Gifu University, Gifu 501-1194, Japan
- Department of Metabolic Research, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan
| | - Tomoyuki Hioki
- Department of Pharmacology, Gifu University, Gifu 501-1194, Japan
- Department of Metabolic Research, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan
- Department of Dermatology, Kizawa Memorial Hospital, Minokamo, Gifu 505-0034, Japan
| | - Rie Matsushima-Nishiwaki
- Department of Pharmacology, Gifu University, Gifu 501-1194, Japan
- Department of Metabolic Research, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan
| | - Takuya Omura
- Department of Metabolic Research, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan
| | - Osamu Kozawa
- Department of Pharmacology, Gifu University, Gifu 501-1194, Japan
- Department of Metabolic Research, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan
| | - Haruhiko Tokuda
- Department of Pharmacology, Gifu University, Gifu 501-1194, Japan
- Department of Metabolic Research, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan
- Department of Clinical Laboratory, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan
| |
Collapse
|
7
|
Mandal S, Flood BE, Lunzer M, Kumar D, Bailey JV. Fluoride and gallein regulate polyphosphate accumulation in dental caries-associated Lacticaseibacillus. MICROBIOLOGY (READING, ENGLAND) 2024; 170:001519. [PMID: 39607745 PMCID: PMC11604172 DOI: 10.1099/mic.0.001519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024]
Abstract
Inorganic polyphosphates (polyPs) are energy-storing biopolymers synthesized by all three domains of life. PolyP accumulation has been well studied with respect to its role in stress response, but its role in dental disease has received less attention. Dental decay can be promoted by changes in pH as well as the chemical activity of ions such as phosphate in oral fluids at the enamel interface. Previous work has shown that the drawdown of phosphate from biofilm fluids can alter the saturation state of oral fluids to thermodynamically favour mineral dissolution. The members of the Lactobacillaceae are known to accumulate polyP and play a role in early-stage and late-stage dental caries. In this study, we examined the effects of potential metabolic inhibitors on polyP accumulation in Lacticaseibacillus rhamnosus. We observed that two inhibitors of the enzyme responsible for polyP synthesis, gallein and fluoride, inhibited polyP accumulation in a balanced medium. However, fluoride and gallein treatments amended with either glucose or lactate were found to enhance polyP accumulation. These results illustrate the potential complexity of polyP metabolism in the oral environment.
Collapse
Affiliation(s)
- Subhrangshu Mandal
- Department of Earth & Environmental Sciences, University of Minnesota – Twin Cities, Minneapolis, MN 55455, USA
- Department of Botany, Visva Bharati University, Bolpur, West Bengal, India
| | - Beverly E. Flood
- Department of Earth & Environmental Sciences, University of Minnesota – Twin Cities, Minneapolis, MN 55455, USA
| | - Mark Lunzer
- Department of Diagnostic and Biological Sciences, University of Minnesota – Twin Cities, Minneapolis, MN 55455, USA
| | - Dhiraj Kumar
- Department of Diagnostic and Biological Sciences, University of Minnesota – Twin Cities, Minneapolis, MN 55455, USA
| | - Jake V. Bailey
- Department of Earth & Environmental Sciences, University of Minnesota – Twin Cities, Minneapolis, MN 55455, USA
| |
Collapse
|
8
|
Xiang Z, Wu F, He Z, Tan F, Hu H, Zou C, Yi P, Liu W, Yang M. D1-like dopamine receptors promote B-cell differentiation in systemic lupus erythematosus. Cell Commun Signal 2024; 22:502. [PMID: 39420360 PMCID: PMC11484144 DOI: 10.1186/s12964-024-01885-3] [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: 08/02/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is an autoimmune disease that currently cannot be completely cured with a great health burden. Since the production of autoantibodies plays a key role in the pathogenesis of SLE, discovering the underlying immunoregulation mechanism of B cells will be helpful for developing promising immunotherapy for SLE. In recent studies, dopamine receptors (DRDs), G protein-coupled receptors that include D1-like and D2-like subtypes, are expressed on B cells and participate in various physiological processes, involving immune responses. However, the regulatory effect of DRDs on B cells has not been determined. METHODS This study explored the expression of DRDs on B-cell subsets from SLE patients and healthy individuals. The effects of D1-like receptor on B-cell activation and differentiation were further explored using D1-like receptor agonists or antagonists. RNA-seq and bioinformatics analyses were used to identify specific molecular mechanisms involved. RESULTS The D1-like DRDs on B cells of SLE patients were highly expressed compared with those of healthy controls (HCs). D1-like receptor agonist treatment exacerbated lupus-like symptoms in pristane-induced lupus-like mice, while D1-like receptor antagonists alleviated the lupus-like phenotypes. Inhibition of D1-like receptor signals impeded B-cell differentiation, while activation of D1-like receptor signals could promote B cell differentiation. Further RNA-seq confirmed that PTGS2, a gene related to B-cell differentiation, was up-regulated once the D1-like receptor signals were activated, while BMP6 and IL-24 were up-regulated once the D1-like receptor signals were inhibited. CONCLUSION D1-like receptors probably promote B-cell differentiation through the PTGS2/PRDM1 pathway.
Collapse
Affiliation(s)
- Zhongyuan Xiang
- Department of Laboratory Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fengxi Wu
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Xiangya, Central South University, Hunan Cancer Hospital, Changsha, Hunan, China
| | - Zhenghao He
- Department of Plastic Surgery, Zhongshan City People's Hospital, Zhongshan, Guangdong, China
| | - Fen Tan
- Department of Dermatology, Second Xiangya Hospital, Hunan Key Laboratory of Medical Epigenomics, Central South University, Changsha, Hunan, China
| | - Haoran Hu
- Department of Dermatology, Second Xiangya Hospital, Hunan Key Laboratory of Medical Epigenomics, Central South University, Changsha, Hunan, China
| | - Chun Zou
- Department of Dermatology, Second Xiangya Hospital, Hunan Key Laboratory of Medical Epigenomics, Central South University, Changsha, Hunan, China
| | - Ping Yi
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wenen Liu
- Department of Laboratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Ming Yang
- Department of Dermatology, Second Xiangya Hospital, Hunan Key Laboratory of Medical Epigenomics, Central South University, Changsha, Hunan, China.
| |
Collapse
|
9
|
He MY, Tong KI, Liu T, Whittaker Hawkins R, Shelton V, Zeng Y, Bakhtiari M, Xiao Y, Zheng G, Sakhdari A, Yang L, Xu W, Brooks DG, Laister RC, He HH, Kridel R. GNAS knockout potentiates HDAC3 inhibition through viral mimicry-related interferon responses in lymphoma. Leukemia 2024; 38:2210-2224. [PMID: 39117798 PMCID: PMC11436380 DOI: 10.1038/s41375-024-02325-4] [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: 02/12/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 08/10/2024]
Abstract
Despite selective HDAC3 inhibition showing promise in a subset of lymphomas with CREBBP mutations, wild-type tumors generally exhibit resistance. Here, using unbiased genome-wide CRISPR screening, we identify GNAS knockout (KO) as a sensitizer of resistant lymphoma cells to HDAC3 inhibition. Mechanistically, GNAS KO-induced sensitization is independent of the canonical G-protein activities but unexpectedly mediated by viral mimicry-related interferon (IFN) responses, characterized by TBK1 and IRF3 activation, double-stranded RNA formation, and transposable element (TE) expression. GNAS KO additionally synergizes with HDAC3 inhibition to enhance CD8+ T cell-induced cytotoxicity. Moreover, we observe in human lymphoma patients that low GNAS expression is associated with high baseline TE expression and upregulated IFN signaling and shares common disrupted biological activities with GNAS KO in histone modification, mRNA processing, and transcriptional regulation. Collectively, our findings establish an unprecedented link between HDAC3 inhibition and viral mimicry in lymphoma. We suggest low GNAS expression as a potential biomarker that reflects viral mimicry priming for enhanced response to HDAC3 inhibition in the clinical treatment of lymphoma, especially the CREBBP wild-type cases.
Collapse
Affiliation(s)
- Michael Y He
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Kit I Tong
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Ting Liu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Ryder Whittaker Hawkins
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Cell Biology Program, Hospital for Sick Children, Toronto, ON, Canada
| | - Victoria Shelton
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Yong Zeng
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Mehran Bakhtiari
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Yufeng Xiao
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Guangrong Zheng
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Ali Sakhdari
- Laboratory Medicine and Pathobiology, University Health Network, Toronto, ON, Canada
| | - Lin Yang
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Wenxi Xu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - David G Brooks
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Rob C Laister
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Housheng Hansen He
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Robert Kridel
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.
- Department of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
10
|
Ji X, Zhou Y, He S, Chen H, Zhang X, Chen Z, Cai J. Bioinformatics analysis of G protein subunit gamma transduction protein 2-autophagy axis in CD11b+ dendritic cells as a potential regulator to skew airway neutrophilic inflammation in asthma endotypes. Immun Inflamm Dis 2024; 12:e70038. [PMID: 39417697 PMCID: PMC11484477 DOI: 10.1002/iid3.70038] [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: 12/13/2023] [Revised: 09/14/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Asthma is a heterogeneous inflammatory disease with two main clinical endotypes: type 2 (T2) high and low asthma. The plasticity and autophagy in dendritic cells (DCs) influence T helper (Th)2 or Th17 differentiation to regulate asthma endotypes. Enhanced autophagy in DCs fosters Th2 differentiation in allergic environments, while reduced autophagy favors Th17 cell differentiation in sensitized and infected environments. Autophagy regulation in DCs involves interaction with various pathways like G protein-coupled receptor (GPCR), mammalian target of rapamycin (mTOR), or phosphoinositide 3-kinase (PI3K) pathway. However, specific molecules within DCs influencing asthma endotypes remain unclear. METHODS Gene expression data series (GSE) 64896, 6858, 2276, and 55247 were obtained from gene expression omnibus (GEO) database. Differentially expressed genes (DEGs) between CD103+ and CD11b+ DCs after induction by ovalbumin (OVA) and lipopolysaccharide (LPS) were analyzed using GEO2R. DEGs were examined through Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and protein-protein interaction (PPI) analyses. The hub gene network was construct with STRING database and Cytoscape. Autophagy differences in DCs and the selected hub gene in GSE6858, GSE2276, and GSE55247 were evaluated using student t tests. RESULTS Our analysis identified 635 upregulated and 360 downregulated genes in CD11b+ DCs, compared to CD103+ DCs. These DEGs were associated with "PI3K-AKT signaling pathway," "Ras signaling pathway," and so forth. Thirty-five hub genes were identified, in which G protein subunit gamma transduction protein 2 (Gngt2) in CD11b+ DCs exhibited a relatively specific increase in expression associated with autophagy defects under the induction environment similar to T2 low asthma model. No significant difference was found in lung Gngt2 expression between T2 high asthma model and control group. CONCLUSION Our analysis suggested Gngt2 acted as an adapter molecule that inhibited autophagy, promoting Th17-mediated airway inflammation via the GPCR pathway in a T2 low asthma mice model. Targeting this pathway provides new asthma treatment strategies in preclinical research.
Collapse
Affiliation(s)
- Xiaoying Ji
- Department of Respiratory and Critical Care MedicineAffiliated Hospital of Guizhou Medical UniversityGuiyang CityGuizhou ProvinceChina
| | - Yaoliang Zhou
- Emergency and Disaster Medical CenterThe Seventh Affiliated Hospital, Sun Yat‐sen UniversityShenzhen CityGuangdong ProvinceChina
| | - Shendong He
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Guangxi University of Chinese Medicine, Xianhu DistrictNanningGuangxiChina
| | - Hongda Chen
- Department of Traditional Chinese MedicineThe Seventh Affiliated Hospital, Sun Yat‐sen UniversityShenzhen CityGuangdong ProvinceChina
| | - Xianming Zhang
- Department of Respiratory and Critical Care MedicineAffiliated Hospital of Guizhou Medical UniversityGuiyang CityGuizhou ProvinceChina
| | - Zhifeng Chen
- Department of Respiratory and Critical Care MedicineThe Second Xiangya Hospital, Central South UniversityChangsha CityHunan ProvinceChina
| | - Jinwen Cai
- Department of Respiratory and Critical Care MedicineThe Third Xiangya Hospital of Central South UniversityChangsha CityHunan ProvinceChina
| |
Collapse
|
11
|
Hao W, Luo D, Jiang Y, Wan S, Li X. An overview of sphingosine-1-phosphate receptor 2: Structure, biological function, and small-molecule modulators. Med Res Rev 2024; 44:2331-2362. [PMID: 38665010 DOI: 10.1002/med.22044] [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: 01/03/2024] [Revised: 04/02/2024] [Accepted: 04/14/2024] [Indexed: 08/09/2024]
Abstract
Over the past decade, there has been a notable increase in research on sphingosine-1-phosphate receptor 2 (S1PR2), which is a type of G-protein-coupled receptor. Upon activation by S1P or other ligands, S1PR2 initiates downstream signaling pathways such as phosphoinositide 3-kinase (PI3K), Mitogen-activated protein kinase (MAPK), Rho/Rho-associated coiled-coil containing kinases (ROCK), and others, contributing to the diverse biological functions of S1PR2 and playing a pivotal role in various physiological processes and disease progressions, such as multiple sclerosis, fibrosis, inflammation, and tumors. Due to the extensive biological functions of S1PR2, many S1PR2 modulators, including agonists and antagonists, have been developed and discovered by pharmaceutical companies (e.g., Novartis and Galapagos NV) and academic medicinal chemists for disease diagnosis and treatment. However, few reviews have been published that comprehensively overview the functions and regulators of S1PR2. Herein, we provide an in-depth review of the advances in the function of S1PR2 and its modulators. We first summarize the structure and biological function of S1PR2 and its pathological role in human diseases. We then focus on the discovery approach, design strategy, development process, and biomedical application of S1PR2 modulators. Additionally, we outline the major challenges and future directions in this field. Our comprehensive review will aid in the discovery and development of more effective and clinically applicable S1PR2 modulators.
Collapse
Affiliation(s)
- Wanting Hao
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Dongdong Luo
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Yuqi Jiang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Shengbiao Wan
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Xiaoyang Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Marine Biomedical Research, Institute of Qingdao, Qingdao, China
| |
Collapse
|
12
|
Bonifer C, Hanke W, Mühle J, Löhr F, Becker-Baldus J, Nagel J, Schertler GFX, Müller CE, König GM, Hilger D, Glaubitz C. Structural response of G protein binding to the cyclodepsipeptide inhibitor FR900359 probed by NMR spectroscopy. Chem Sci 2024; 15:12939-12956. [PMID: 39148790 PMCID: PMC11323312 DOI: 10.1039/d4sc01950d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 06/27/2024] [Indexed: 08/17/2024] Open
Abstract
The cyclodepsipeptide FR900359 (FR) and its analogs are able to selectively inhibit the class of Gq proteins by blocking GDP/GTP exchange. The inhibitor binding site of Gq has been characterized by X-ray crystallography, and various binding and functional studies have determined binding kinetics and mode of inhibition. Here we investigate isotope-labeled FR bound to the membrane-anchored G protein heterotrimer by solid-state nuclear magnetic resonance (ssNMR) and in solution by liquid-state NMR. The resulting data allowed us to identify regions of the inhibitor which show especially pronounced effects upon binding and revealed a generally rigid binding mode in the cis conformation under native-like conditions. The inclusion of the membrane environment allowed us to show a deep penetration of FR into the lipid bilayer illustrating a possible access mode of FR into the cell. Dynamic nuclear polarization (DNP)-enhanced ssNMR was used to observe the structural response of specific segments of the Gα subunit to inhibitor binding. This revealed rigidification of the switch I binding site and an allosteric response in the α5 helix as well as suppression of structural changes induced by nucleotide exchange due to inhibition by FR. Our NMR studies of the FR-G protein complex conducted directly within a native membrane environment provide important insights into the inhibitors access via the lipid membrane, binding mode, and structural allosteric effects.
Collapse
Affiliation(s)
- Christian Bonifer
- Institute of Biophysical Chemistry, Centre of Biomolecular Magnetic Resonance, Goethe University Frankfurt Max-von-Laue Str. 9 60438 Frankfurt Germany
| | - Wiebke Hanke
- Institute for Pharmaceutical Biology, University of Bonn Nussallee 6 53115 Bonn Germany
| | - Jonas Mühle
- Division of Biology and Chemistry, Laboratory of Biomolecular Research, Paul Scherrer Institute Forschungsstr. 111, 5232 Villigen PSI Switzerland
| | - Frank Löhr
- Institute of Biophysical Chemistry, Centre of Biomolecular Magnetic Resonance, Goethe University Frankfurt Max-von-Laue Str. 9 60438 Frankfurt Germany
| | - Johanna Becker-Baldus
- Institute of Biophysical Chemistry, Centre of Biomolecular Magnetic Resonance, Goethe University Frankfurt Max-von-Laue Str. 9 60438 Frankfurt Germany
| | - Jessica Nagel
- Department of Pharmaceutical & Medicinal Chemistry, Pharmaceutical Institute, University of Bonn An der Immenburg 4 53121 Bonn Germany
| | - Gebhard F X Schertler
- Division of Biology and Chemistry, Laboratory of Biomolecular Research, Paul Scherrer Institute Forschungsstr. 111, 5232 Villigen PSI Switzerland
| | - Christa E Müller
- Department of Pharmaceutical & Medicinal Chemistry, Pharmaceutical Institute, University of Bonn An der Immenburg 4 53121 Bonn Germany
| | - Gabriele M König
- Institute for Pharmaceutical Biology, University of Bonn Nussallee 6 53115 Bonn Germany
| | - Daniel Hilger
- Department of Pharmaceutical Chemistry, University of Marburg 35037 Marburg Germany
| | - Clemens Glaubitz
- Institute of Biophysical Chemistry, Centre of Biomolecular Magnetic Resonance, Goethe University Frankfurt Max-von-Laue Str. 9 60438 Frankfurt Germany
| |
Collapse
|
13
|
Kuroyanagi G, Hioki T, Matsushima-Nishiwaki R, Kozawa O, Tokuda H. Gallein increases the fibroblast growth factor 2-elicited osteoprotegerin synthesis in osteoblasts. Biochim Biophys Acta Gen Subj 2024; 1868:130635. [PMID: 38788984 DOI: 10.1016/j.bbagen.2024.130635] [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: 12/03/2023] [Revised: 04/28/2024] [Accepted: 05/20/2024] [Indexed: 05/26/2024]
Abstract
Gallein is known as an inhibitor of Gβγ subunits, but roles of gallein in bone metabolism have not been reported. Fibroblast growth factor 2 (FGF-2) increases angiogenesis and promotes bone regeneration during the early stages of fracture healing. Osteoprotegerin (OPG) secreted by osteoblasts, binds to the receptor activator of nuclear factor-κB (RANK) ligand (RANKL) as a decoy receptor and prevents RANKL from binding to RANK, resulting in the suppression of bone resorption. Our previous report demonstrated that FGF-2 activates the phosphorylation of p38 mitogen-activated protein kinase (MAPK), stress-activated protein kinase/c-Jun N-terminal kinase (JNK), and p44/p42 MAPK in osteoblast-like MC3T3-E1 cells. Additionally, FGF-2-activated phosphorylation of p38 MAPK and JNK but not p44/p42 MAPK is positively involved in OPG synthesis in these cells. This work aimed to investigate the effects of gallein on the FGF-2-elicited OPG synthesis in osteoblast-like MC3T3-E1 cells and the mechanism. Our findings demonstrated that gallein significantly increased the FGF-2-elicited OPG synthesis in MC3T3-E1 cells. By contrast, fluorescein, gallein-like compound that does not bind Gβγ, did not affect the FGF-2-elicited OPG synthesis. Gallein significantly enhanced the FGF-2-induced OPG mRNA expression levels. Gallein did not affect the FGF-2-activated phosphorylation of p38 MAPK and p44/p42 MAPK, but significantly increased the FGF-2-activated phosphorylation of JNK, while fluorescein did not affect JNK phosphorylation. SP600125, a specific JNK inhibitor, strongly inhibited gallein-induced enhancement of FGF-2-induced OPG synthesis and mRNA expression levels. Our results indicated that gallein increases the FGF-2-induced OPG synthesis due to the JNK activation in the osteoblast.
Collapse
Affiliation(s)
- Gen Kuroyanagi
- Department of Orthopaedic Surgery, Nagoya City University, Nagoya 467-8601, Japan; Department of Rehabilitation Medicine, Nagoya City University, Nagoya 467-8601, Japan; Department of Pharmacology, Gifu University, Gifu 501-1194, Japan; Department of Metabolic Research, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan.
| | - Tomoyuki Hioki
- Department of Pharmacology, Gifu University, Gifu 501-1194, Japan; Department of Metabolic Research, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan; Department of Dermatology, Kizawa Memorial Hospital, Minokamo, Gifu 505-0034, Japan
| | - Rie Matsushima-Nishiwaki
- Department of Pharmacology, Gifu University, Gifu 501-1194, Japan; Department of Metabolic Research, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan
| | - Osamu Kozawa
- Department of Pharmacology, Gifu University, Gifu 501-1194, Japan; Department of Metabolic Research, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan
| | - Haruhiko Tokuda
- Department of Pharmacology, Gifu University, Gifu 501-1194, Japan; Department of Metabolic Research, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan; Department of Clinical Laboratory, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan
| |
Collapse
|
14
|
Huang Y, Lei Y, Gong Z, Wang Y, Li M, Zhao D, Song L. Feeding Eucommia ulmoides extract enhances protection against high-temperature stress in chicks. Poult Sci 2024; 103:103827. [PMID: 38801811 PMCID: PMC11150962 DOI: 10.1016/j.psj.2024.103827] [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: 02/06/2024] [Revised: 04/18/2024] [Accepted: 05/02/2024] [Indexed: 05/29/2024] Open
Abstract
Chick's susceptibility to heat stress often leads to growth retardation, immune function impairment, disease, and mortality. This thesis explores the potential ameliorative effect of 0.8% Eucommia ulmoides extract (EUE) into the diet of heat-stressed chicks in a 15-d feeding trial. The investigation reveals that feeding EUE significantly enhances the BW, ADG, AFI, and F/G of chicks experiencing heat stress. Additionally, the EUE groups exhibited higher levels of T-AOC (at 7 and 15d), SOD (at 15 d), GSH-Px (at 15 d), as well as lower MDA concentrations (at 7 and 15d) in chick serum. Pathological changes and H&E staining revealed that EUE effectively improved tissue damage in the duodenum, heart, and stomach induced by heat stress in the chicks. The EUE groups also showed higher levels of IgA (at 7 d), IgG and IgM (at 7 and 15 d). RNA-seq and WGCNA analysis revealed that EUE mitigates cellular damage and losses in heat-stressed chicks primarily through pathways involving signal transduction, protein synthesis and degradation, as well as cell cycle regulation, particularly the latter. This investigation serves as a fundamental and cognitive framework for the development and application of Eucommia ulmoides feed additives aimed at safeguarding the well-being of chicks in adverse environmental conditions.
Collapse
Affiliation(s)
- Youwen Huang
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-Bioengineering, Guizhou University, Guiyang, Guizhou Province 550025, China; Guizhou Key Lab of Agro-Bioengineering, Guiyang, Guizhou Province 550025, China; National-Local Joint Engineering Research Center of Karst Region Plant Resources Utilization & Breeding (Guizhou), Guiyang, Guizhou Province 550025, China
| | - Yue Lei
- Guizhou Institute of Subtropical Crops, Xingyi, Guizhou Province 562400, China
| | - Zouxian Gong
- Clinical Medical College of Guizhou Medical University, Guiyang, Guizhou Province 550004, China
| | - Yifan Wang
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-Bioengineering, Guizhou University, Guiyang, Guizhou Province 550025, China; Guizhou Key Lab of Agro-Bioengineering, Guiyang, Guizhou Province 550025, China; National-Local Joint Engineering Research Center of Karst Region Plant Resources Utilization & Breeding (Guizhou), Guiyang, Guizhou Province 550025, China
| | - Minxue Li
- Agricultural and Rural Bureau, Shuicheng District, Liupanshui City, Guizhou Province 553040, China
| | - Degang Zhao
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-Bioengineering, Guizhou University, Guiyang, Guizhou Province 550025, China; National-Local Joint Engineering Research Center of Karst Region Plant Resources Utilization & Breeding (Guizhou), Guiyang, Guizhou Province 550025, China; Guizhou Academy of Agricultural Science, Guiyang, Guizhou Province 550006, China
| | - Li Song
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-Bioengineering, Guizhou University, Guiyang, Guizhou Province 550025, China; Guizhou Key Lab of Agro-Bioengineering, Guiyang, Guizhou Province 550025, China; National-Local Joint Engineering Research Center of Karst Region Plant Resources Utilization & Breeding (Guizhou), Guiyang, Guizhou Province 550025, China.
| |
Collapse
|
15
|
Peng X, Tao H, Xia F, Zhu M, Yang M, Liu K, Hou B, Li X, Li S, He Y, Huan W, Gao F. Molecular design, construction and analgesic mechanism insights into the novel transdermal fusion peptide ANTP-BgNPB. Bioorg Chem 2024; 148:107482. [PMID: 38795582 DOI: 10.1016/j.bioorg.2024.107482] [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: 02/27/2024] [Revised: 05/14/2024] [Accepted: 05/19/2024] [Indexed: 05/28/2024]
Abstract
Toad venom, a traditional Chinese medicine, exhibits remarkable medicinal properties of significant therapeutic value. The peptides present within toad venom possess a wide range of biological functions, yet the neuropeptide B (NPB) and it modification requires further exploration to comprehensively understand its mechanisms of action and potential applications. In this study, a fusion peptide, ANTP-BgNPB, was designed to possess better analgesic properties through the transdermal modification of BgNPB. After optimizing the conditions, the expression of ANTP-BgNPB was successfully induced. The molecular dynamics simulations suggested that the modified protein exhibited improved stability and receptor binding affinity compared to its unmodified form. The analysis of the active site of ANTP-BgNPB and the verification of mutants revealed that GLN3, SER38, and ARG42 were crucial for the protein's recognition and binding with G protein-coupled receptor 7 (GPR7). Moreover, experiments conducted on mice using the hot plate and acetic acid twist body models demonstrated that ANTP-BgNPB was effective in transdermal analgesia. These findings represent significant progress in the development of transdermal delivery medications and could have a significant impact on pain management.
Collapse
Affiliation(s)
- Xinmeng Peng
- Zhejiang Provincial Key Laboratory of Resources Protection and Innovation of Traditional Chinese Medicine, Zhejiang A&F University, Hangzhou 311300, China
| | - Han Tao
- Zhejiang Provincial Key Laboratory of Resources Protection and Innovation of Traditional Chinese Medicine, Zhejiang A&F University, Hangzhou 311300, China
| | - Fengyan Xia
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 313000, China
| | - Mingwei Zhu
- Zhejiang Provincial Key Laboratory of Resources Protection and Innovation of Traditional Chinese Medicine, Zhejiang A&F University, Hangzhou 311300, China
| | - Meiyun Yang
- Zhejiang Provincial Key Laboratory of Resources Protection and Innovation of Traditional Chinese Medicine, Zhejiang A&F University, Hangzhou 311300, China
| | - Kexin Liu
- Zhejiang Provincial Key Laboratory of Resources Protection and Innovation of Traditional Chinese Medicine, Zhejiang A&F University, Hangzhou 311300, China
| | - Bowen Hou
- Zhejiang Provincial Key Laboratory of Resources Protection and Innovation of Traditional Chinese Medicine, Zhejiang A&F University, Hangzhou 311300, China
| | - Xintong Li
- Zhejiang Provincial Key Laboratory of Resources Protection and Innovation of Traditional Chinese Medicine, Zhejiang A&F University, Hangzhou 311300, China
| | - Suwan Li
- Zhejiang Provincial Key Laboratory of Resources Protection and Innovation of Traditional Chinese Medicine, Zhejiang A&F University, Hangzhou 311300, China
| | - Yanling He
- Zhejiang Provincial Key Laboratory of Resources Protection and Innovation of Traditional Chinese Medicine, Zhejiang A&F University, Hangzhou 311300, China
| | - Weiwei Huan
- Zhejiang Provincial Key Laboratory of Chemical Utilization of Forestry Biomass, College of Chemistry and Materials Engineering, Zhejiang A & F University, Hangzhou, Zhejiang 311300, China.
| | - Fei Gao
- Zhejiang Provincial Key Laboratory of Resources Protection and Innovation of Traditional Chinese Medicine, Zhejiang A&F University, Hangzhou 311300, China.
| |
Collapse
|
16
|
Yang C, Wu L, Jin X, Liu A, Jing Z, Feng C, Guo Z, Zhang Y, Ma Y, Li F, Wen Z, Yan L, Yang Y, Ji X, Zhang Y. Decrease in GPSM2 mediated by the natural product luteolin contributes to colon adenocarcinoma treatment and increases the sensitivity to fluorouracil. Biomed Pharmacother 2024; 176:116847. [PMID: 38823277 DOI: 10.1016/j.biopha.2024.116847] [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: 02/10/2024] [Revised: 05/22/2024] [Accepted: 05/27/2024] [Indexed: 06/03/2024] Open
Abstract
Luteolin, a monomeric substance, is a natural product of the Brucea javanica (BJ) plant. Brucea javanica oil emulsion injection (BJOEI) is a proprietary Chinese medicine purified from BJ that is widely used clinically as an anti-tumor treatment. Although a growing body of research suggests that luteolin and BJOEI have anti-tumor effects, the molecular mechanism of action has not been fully elucidated. In this study, through molecular docking technology, we found that luteolin can interact directly with GPSM2 and regulate the FoxO signaling pathway through GPSM2. In addition, the inhibitory effect of luteolin on colon adenocarcinoma (COAD) cells was found to be offset by knockdown of GPSM2. In contrast, the anti-proliferative effects of luteolin could be notably reversed by overexpression of GPSM2. The results reveal that GPSM2 is crucial in luteolin-mediated anti-proliferative effects. The mediation of anti-proliferative effects by GPSM2 has also been indirectly demonstrated in RKO and SW480 xenograft mice models. In addition, we verified that BJOEI inhibits the progression of COAD by mediating GPSM2 and regulating the FoxO signaling pathway. We also found that BJOEI achieved a better anti-tumor effect when combined with fluorouracil injection. Collectively, our data show that the anti-tumor effects of BJOEI and luteolin on COAD are GPSM2-dependent and downregulating the expression of GPSM2 to regulate the FoxO signaling pathway may be an effective way to treat COAD.
Collapse
Affiliation(s)
- Chunjiao Yang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, China; Department of Oncology, The Fifth Affiliated Hospital of Guangxi Medical University & The First People's Hospital of Nanning, Nanning, Guangxi, China
| | - Lina Wu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, No. 36, San Hao Street, Shenyang, Liaoning 110004, China; Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, China
| | - Xin Jin
- Department of Respiratory Medicine, The Fifth Affiliated Hospital of Guangxi Medical University & The First People's Hospital of Nanning, Nanning, Guangxi, China
| | - Aoran Liu
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, China
| | - Zhitao Jing
- The First Hospital of China Medical University, Shenyang, China
| | - Chuhan Feng
- Liaoning University Of Traditional Chinese Medicine, Shenyang, China
| | - Zhengting Guo
- The First Clinical College, China Medical University, Shenyang, China
| | - Yuzhe Zhang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, China
| | - Yanju Ma
- Department of Medical Oncology, Cancer Hospital of China Medical University, China
| | - Fang Li
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, China
| | - Zhenpeng Wen
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, China; Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Lirong Yan
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, China
| | - Yi Yang
- Department of Laboratory Animal Science, China Medical University, Shenyang, China
| | - Xu Ji
- The First Hospital of China Medical University, Shenyang, China.
| | - Ye Zhang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
17
|
Fu X, Tasker JG. Neuromodulation of inhibitory synaptic transmission in the basolateral amygdala during fear and anxiety. Front Cell Neurosci 2024; 18:1421617. [PMID: 38994327 PMCID: PMC11236696 DOI: 10.3389/fncel.2024.1421617] [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: 04/22/2024] [Accepted: 06/18/2024] [Indexed: 07/13/2024] Open
Abstract
The basolateral amygdala plays pivotal roles in the regulation of fear and anxiety and these processes are profoundly modulated by different neuromodulatory systems that are recruited during emotional arousal. Recent studies suggest activities of BLA interneurons and inhibitory synaptic transmission in BLA principal cells are regulated by neuromodulators to influence the output and oscillatory network states of the BLA, and ultimately the behavioral expression of fear and anxiety. In this review, we first summarize a cellular mechanism of stress-induced anxiogenesis mediated by the interaction of glucocorticoid and endocannabinoid signaling at inhibitory synapses in the BLA. Then we discuss cell type-specific activity patterns induced by neuromodulators converging on the Gq signaling pathway in BLA perisomatic parvalbumin-expressing (PV) and cholecystokinin-expressing (CCK) basket cells and their effects on BLA network oscillations and fear learning.
Collapse
Affiliation(s)
- Xin Fu
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Jeffrey G. Tasker
- Tulane Brain Institute, Tulane University, New Orleans, LA, United States
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States
| |
Collapse
|
18
|
Sanchez GA, Smrcka AV, Jutkiewicz EM. Biasing G βγ Downstream Signaling with Gallein Inhibits Development of Morphine Tolerance and Potentiates Morphine-Induced Nociception in a Tolerant State. Mol Pharmacol 2024; 106:47-55. [PMID: 38769020 PMCID: PMC11187686 DOI: 10.1124/molpharm.124.000875] [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: 01/19/2024] [Revised: 05/03/2024] [Accepted: 05/09/2024] [Indexed: 05/22/2024] Open
Abstract
Opioid analgesics are widely used as a treatment option for pain management and relief. However, the misuse of opioid analgesics has contributed to the current opioid epidemic in the United States. Prescribed opioids such as morphine, codeine, oxycodone, and fentanyl are mu-opioid receptor (MOR) agonists primarily used in the clinic to treat pain or during medical procedures, but development of tolerance limits their utility for treatment of chronic pain. Here we explored the effects of biasing Gβγ signaling on tolerance development after chronic morphine treatment in vivo. We hypothesized that biasing Gβγ signaling with gallein could prevent activation of regulatory signaling pathways that result in tolerance to antinociceptive effects of MOR agonists. Gallein has been shown to bind to Gβγ and inhibit interactions of Gβγ with phospholipase-Cβ3 (PLCβ3) or G-protein-coupled receptor kinase 2 (GRK2) but not G-protein inwardly rectifying potassium (GIRK) channels. In mice, morphine-induced antinociception was evaluated in the 55°C warm water tail withdrawal assay. We used two paradigms for gallein treatment: administration during and after three times-daily morphine administration. Our results show that gallein cotreatment during repeated administration of morphine decreased opioid tolerance development and that gallein treatment in an opioid-tolerant state enhanced the potency of morphine. Mechanistically, our data suggest that PLCβ3 is necessary for potentiating effects of gallein in an opioid-tolerant state but not in preventing the development of tolerance. These studies demonstrate that small molecules that target Gβγ signaling could reduce the need for large doses of opioid analgesics to treat pain by producing an opioid-sparing effect. SIGNIFICANCE STATEMENT: Biasing Gβγ signaling prevents tolerance to repeated morphine administration in vivo and potentiates the antinociceptive effects of morphine in an opioid-tolerant state. Mechanistically, phospholipase-Cβ is necessary for potentiating effects of gallein in an opioid-tolerant state but not in preventing the development of tolerance. This study identifies a novel treatment strategy to decrease the development of tolerance to the analgesic effects of mu-opioid receptor agonists, which are necessary to improve pain treatment and decrease the incidence of opioid use disorder.
Collapse
Affiliation(s)
- Gissell A Sanchez
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan
| | - Alan V Smrcka
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan
| | | |
Collapse
|
19
|
Shen S, Wu C, Lin G, Yang X, Zhou Y, Zhao C, Miao Z, Tian X, Wang K, Yang Z, Liu Z, Guo N, Li Y, Xia A, Zhou P, Liu J, Yan W, Ke B, Yang S, Shao Z. Structure-based identification of a G protein-biased allosteric modulator of cannabinoid receptor CB1. Proc Natl Acad Sci U S A 2024; 121:e2321532121. [PMID: 38830102 PMCID: PMC11181136 DOI: 10.1073/pnas.2321532121] [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: 12/07/2023] [Accepted: 04/01/2024] [Indexed: 06/05/2024] Open
Abstract
Cannabis sativa is known for its therapeutic benefit in various diseases including pain relief by targeting cannabinoid receptors. The primary component of cannabis, Δ9-tetrahydrocannabinol (THC), and other agonists engage the orthosteric site of CB1, activating both Gi and β-arrestin signaling pathways. The activation of diverse pathways could result in on-target side effects and cannabis addiction, which may hinder therapeutic potential. A significant challenge in pharmacology is the design of a ligand that can modulate specific signaling of CB1. By leveraging insights from the structure-function selectivity relationship (SFSR), we have identified Gi signaling-biased agonist-allosteric modulators (ago-BAMs). Further, two cryoelectron microscopy (cryo-EM) structures reveal the binding mode of ago-BAM at the extrahelical allosteric site of CB1. Combining mutagenesis and pharmacological studies, we elucidated the detailed mechanism of ago-BAM-mediated biased signaling. Notably, ago-BAM CB-05 demonstrated analgesic efficacy with fewer side effects, minimal drug toxicity and no cannabis addiction in mouse pain models. In summary, our finding not only suggests that ago-BAMs of CB1 provide a potential nonopioid strategy for pain management but also sheds light on BAM identification for GPCRs.
Collapse
Affiliation(s)
- Siyuan Shen
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu610041, Sichuan, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu610212, Sichuan, China
| | - Chao Wu
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu610041, Sichuan, China
| | - Guifeng Lin
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu610041, Sichuan, China
| | - Xin Yang
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu610041, Sichuan, China
| | - Yangli Zhou
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu610041, Sichuan, China
| | - Chang Zhao
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu610041, Sichuan, China
| | - Zhuang Miao
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu610041, Sichuan, China
| | - Xiaowen Tian
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu610041, Sichuan, China
| | - Kexin Wang
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu610041, Sichuan, China
| | - Zhiqian Yang
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu610041, Sichuan, China
| | - Zhiyu Liu
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu610041, Sichuan, China
| | - Nihong Guo
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu610041, Sichuan, China
| | - Yueshan Li
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu610041, Sichuan, China
| | - Anjie Xia
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu610041, Sichuan, China
| | - Pei Zhou
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu610041, Sichuan, China
| | - Jingming Liu
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu610041, Sichuan, China
| | - Wei Yan
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu610041, Sichuan, China
| | - Bowen Ke
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu610041, Sichuan, China
| | - Shengyong Yang
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu610041, Sichuan, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu610212, Sichuan, China
| | - Zhenhua Shao
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu610041, Sichuan, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu610212, Sichuan, China
| |
Collapse
|
20
|
Li YH, Yang YS, Xue YB, Lei H, Zhang SS, Qian J, Yao Y, Zhou R, Huang L. G protein subunit G γ13-mediated signaling pathway is critical to the inflammation resolution and functional recovery of severely injured lungs. eLife 2024; 12:RP92956. [PMID: 38836551 DOI: 10.7554/elife.92956] [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] [Indexed: 06/06/2024] Open
Abstract
Tuft cells are a group of rare epithelial cells that can detect pathogenic microbes and parasites. Many of these cells express signaling proteins initially found in taste buds. It is, however, not well understood how these taste signaling proteins contribute to the response to the invading pathogens or to the recovery of injured tissues. In this study, we conditionally nullified the signaling G protein subunit Gγ13 and found that the number of ectopic tuft cells in the injured lung was reduced following the infection of the influenza virus H1N1. Furthermore, the infected mutant mice exhibited significantly larger areas of lung injury, increased macrophage infiltration, severer pulmonary epithelial leakage, augmented pyroptosis and cell death, greater bodyweight loss, slower recovery, worsened fibrosis and increased fatality. Our data demonstrate that the Gγ13-mediated signal transduction pathway is critical to tuft cells-mediated inflammation resolution and functional repair of the damaged lungs.To our best knowledge, it is the first report indicating subtype-specific contributions of tuft cells to the resolution and recovery.
Collapse
Affiliation(s)
- Yi-Hong Li
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Yi-Sen Yang
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Yan-Bo Xue
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Hao Lei
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Sai-Sai Zhang
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Junbin Qian
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Yushi Yao
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruhong Zhou
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Zhejiang University Shanghai Institute for Advanced Study, Shanghai, Shanghai, China
| | - Liquan Huang
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Zhejiang University Shanghai Institute for Advanced Study, Shanghai, Shanghai, China
- Monell Chemical Senses Center, Philadelphia, United States
| |
Collapse
|
21
|
Hioki T, Kuroyanagi G, Matsushima-Nishiwaki R, Omura T, Kozawa O, Tokuda H. Gallein but not fluorescein enhances the PGD 2-stimulated synthesis of osteoprotegerin and interleukin-6 in osteoblasts: Amplification of osteoprotegerin/interleukin-6 by gallein. Prostaglandins Leukot Essent Fatty Acids 2024; 203:102639. [PMID: 39270488 DOI: 10.1016/j.plefa.2024.102639] [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: 07/04/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024]
Abstract
Gallein, a small molecule related to fluorescein, is established as an inhibitor of Gβγ subunits to inhibit G protein (Gs) signaling. This agent is providing a potential therapeutic strategy to ameliorate organ dysfunctions especially involved in inflammation, however; the effects on bone metabolism have not yet been clarified. Prostaglandins (PGs) play important roles as autacoids including osteoblasts, and d-type prostanoid (DP) receptor, a member of G protein-coupled receptor specific to PGD2, is expressed on osteoblasts. We previously reported that prostaglandin D2 (PGD2) induces the syntheses of osteoprotegerin (OPG) and interleukin-6 (IL-6), essential factors in bone remodelling process, and p38 mitogen-activated protein kinase (MAPK), c-Jun N-terminal kinase (JNK), and p44/p42 MAPK are involved in the signal transduction of PGD2 in osteoblast-like MC3T3-E1 cells. Thus, we investigated in this study that the effect and the underlying mechanism of gallein, an inhibitor Gβɤ subunits, on the syntheses of OPG and IL-6 induced by PGD2 in these cells. The cultured cells were treated with gallein or fluorescein, a structurally related compound inactive to Gβɤ subunits, and subsequently stimulated with PGD2. Not fluorescein but gallein amplified the PGD2-stimulated releases of OPG and IL-6. Gallein enhanced the PGD2-upregulated mRNA expression levels of OPG and IL-6. Regarding the signaling mechanism, gallein did not affect the PGD2-induced phosphorylation of p38 MAPK, JNK, or p42 MAPK. In conclusion, gallein upregulates the PGD2-stimulated syntheses of OPG and IL-6 by the specific effect to inhibit Gβγ subunits in osteoblasts, but the effect is not exerted at the upstream of p38 MAPK, JNK, or p44/p42 MAPK activation.
Collapse
Affiliation(s)
- Tomoyuki Hioki
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of Dermatology, Central Japan International Medical Center, Minokamo 505-8510, Japan; Department of Metabolic Research, Research Institute, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan
| | - Gen Kuroyanagi
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of Rehabilitation Medicine, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | - Rie Matsushima-Nishiwaki
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of Metabolic Research, Research Institute, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan
| | - Takuya Omura
- Department of Metabolic Research, Research Institute, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan
| | - Osamu Kozawa
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of Metabolic Research, Research Institute, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan
| | - Haruhiko Tokuda
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of Metabolic Research, Research Institute, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan; Department of Clinical Laboratory, Hospital, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan.
| |
Collapse
|
22
|
Asulin M, Gorodetzer N, Fridman R, Shelly Ben-Shushan R, Cohen Z, Beyer AM, Chuyun D, Gutterman DD, Szuchman-Sapir A. 5,6-diHETE lactone (EPA-L) mediates hypertensive microvascular dilation by activating the endothelial GPR-PLC-IP 3 signaling pathway. Biochem Biophys Res Commun 2024; 700:149585. [PMID: 38290177 DOI: 10.1016/j.bbrc.2024.149585] [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: 12/20/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/01/2024]
Abstract
Endothelial microvascular dysfunction affects multi-organ pathologic processes that contribute to increased vascular tone and is at the base of impaired metabolic and cardiovascular diseases. The vascular dilation impaired by nitric oxide (NO) deficiency in such dysfunctional endothelium is often balanced by endothelial-derived hyperpolarizing factors (EDHFs), which play a critical role in managing vascular tone. Our latest research has uncovered a new group of lactone oxylipins produced in the polyunsaturated fatty acids (PUFAs) CYP450 epoxygenase pathway, significantly affecting vascular dilation. The lactone oxylipin, derived from arachidonic acid (5,6-diHET lactone, AA-L), has been previously shown to facilitate vasodilation dependent on the endothelium in isolated human microvessels. The administration of the lactone oxylipin derived from eicosapentaenoic acid (5,6-diHETE lactone, EPA-L) to hypertensive rats demonstrated a significant decrease in blood pressure and improvement in the relaxation of microvessels. However, the molecular signaling processes that underlie these observations were not fully understood. The current study delineates the molecular pathways through which EPA-L promotes endothelium-dependent vascular dilation. In microvessels from hypertensive individuals, it was found that EPA-L mediates endothelium-dependent vasodilation while the signaling pathway was not dependent on NO. In vitro studies on human endothelial cells showed that the hyperpolarization mediated by EPA-L relies on G-protein-coupled receptor (GPR)-phospholipase C (PLC)-IP3 signaling that further activates calcium-dependent potassium flux. The pathway was confirmed using a range of inhibitors and cells overexpressing GPR40, where a specific antagonist reduced the calcium levels and outward currents induced by EPA-L. The downstream AKT and endothelial NO synthase (eNOS) phosphorylations were non-significant. These findings show that the GPR-PLC-IP3 pathway is a key mediator in the EPA-L-triggered vasodilation of arterioles. Therefore, EPA-L is identified as a significant lactone-based PUFA metabolite that contributes to endothelial and vascular health.
Collapse
Affiliation(s)
- Meitar Asulin
- Laboratory of Vascular Signaling, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel; Tel-Hai College, Upper Galilee, Israel
| | - Nadav Gorodetzer
- Laboratory of Vascular Signaling, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel; Tel-Hai College, Upper Galilee, Israel
| | - Rotem Fridman
- Laboratory of Vascular Signaling, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel
| | | | - Zohar Cohen
- Laboratory of Vascular Signaling, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel; Tel-Hai College, Upper Galilee, Israel
| | - Andreas M Beyer
- Department of Medicine and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - David D Gutterman
- Department of Medicine and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Andrea Szuchman-Sapir
- Laboratory of Vascular Signaling, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel; Tel-Hai College, Upper Galilee, Israel.
| |
Collapse
|
23
|
Elbatsh AMO, Amin-Mansour A, Haberkorn A, Textor C, Ebel N, Renard E, Koch LM, Groenveld FC, Piquet M, Naumann U, Ruddy DA, Romanet V, Martínez Gómez JM, Shirley MD, Wipfli P, Schnell C, Wartmann M, Rausch M, Jager MJ, Levesque MP, Maira SM, Manchado E. INPP5A phosphatase is a synthetic lethal target in GNAQ and GNA11-mutant melanomas. NATURE CANCER 2024; 5:481-499. [PMID: 38233483 PMCID: PMC10965444 DOI: 10.1038/s43018-023-00710-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 12/14/2023] [Indexed: 01/19/2024]
Abstract
Activating mutations in GNAQ/GNA11 occur in over 90% of uveal melanomas (UMs), the most lethal melanoma subtype; however, targeting these oncogenes has proven challenging and inhibiting their downstream effectors show limited clinical efficacy. Here, we performed genome-scale CRISPR screens along with computational analyses of cancer dependency and gene expression datasets to identify the inositol-metabolizing phosphatase INPP5A as a selective dependency in GNAQ/11-mutant UM cells in vitro and in vivo. Mutant cells intrinsically produce high levels of the second messenger inositol 1,4,5 trisphosphate (IP3) that accumulate upon suppression of INPP5A, resulting in hyperactivation of IP3-receptor signaling, increased cytosolic calcium and p53-dependent apoptosis. Finally, we show that GNAQ/11-mutant UM cells and patients' tumors exhibit elevated levels of IP4, a biomarker of enhanced IP3 production; these high levels are abolished by GNAQ/11 inhibition and correlate with sensitivity to INPP5A depletion. Our findings uncover INPP5A as a synthetic lethal vulnerability and a potential therapeutic target for GNAQ/11-mutant-driven cancers.
Collapse
Affiliation(s)
- Ahmed M O Elbatsh
- Oncology, Novartis Institute for Biomedical Research, Basel, Switzerland
| | - Ali Amin-Mansour
- Oncology, Novartis Institute for Biomedical Research, Cambridge, MA, USA
| | - Anne Haberkorn
- Oncology, Novartis Institute for Biomedical Research, Basel, Switzerland
| | - Claudia Textor
- PK Sciences, Novartis Institute for Biomedical Research, Basel, Switzerland
| | - Nicolas Ebel
- Oncology, Novartis Institute for Biomedical Research, Basel, Switzerland
| | - Emilie Renard
- Oncology, Novartis Institute for Biomedical Research, Basel, Switzerland
| | - Lisa M Koch
- Oncology, Novartis Institute for Biomedical Research, Basel, Switzerland
| | - Femke C Groenveld
- Oncology, Novartis Institute for Biomedical Research, Basel, Switzerland
| | - Michelle Piquet
- Oncology, Novartis Institute for Biomedical Research, Cambridge, MA, USA
| | - Ulrike Naumann
- Chemical Biology and Therapeutics, Novartis Institute for Biomedical Research, Basel, Switzerland
| | - David A Ruddy
- Oncology, Novartis Institute for Biomedical Research, Cambridge, MA, USA
| | - Vincent Romanet
- Oncology, Novartis Institute for Biomedical Research, Basel, Switzerland
| | - Julia M Martínez Gómez
- Dermatology Department, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Matthew D Shirley
- Oncology, Novartis Institute for Biomedical Research, Cambridge, MA, USA
| | - Peter Wipfli
- PK Sciences, Novartis Institute for Biomedical Research, Basel, Switzerland
| | - Christian Schnell
- Oncology, Novartis Institute for Biomedical Research, Basel, Switzerland
| | - Markus Wartmann
- Oncology, Novartis Institute for Biomedical Research, Basel, Switzerland
| | - Martin Rausch
- Chemical Biology and Therapeutics, Novartis Institute for Biomedical Research, Basel, Switzerland
| | - Martine J Jager
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mitchell P Levesque
- Dermatology Department, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | | | - Eusebio Manchado
- Oncology, Novartis Institute for Biomedical Research, Basel, Switzerland.
| |
Collapse
|
24
|
Odnoshivkina JG, Averin AS, Khakimov IR, Trusov NA, Trusova DA, Petrov AM. The mechanism of 25-hydroxycholesterol-mediated suppression of atrial β1-adrenergic responses. Pflugers Arch 2024; 476:407-421. [PMID: 38253680 DOI: 10.1007/s00424-024-02913-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 12/27/2023] [Accepted: 01/14/2024] [Indexed: 01/24/2024]
Abstract
25-Hydroxycholesterol (25HC) is a biologically active oxysterol, whose production greatly increases during inflammation by macrophages and dendritic cells. The inflammatory reactions are frequently accompanied by changes in heart regulation, such as blunting of the cardiac β-adrenergic receptor (AR) signaling. Here, the mechanism of 25HC-dependent modulation of responses to β-AR activation was studied in the atria of mice. 25HC at the submicromolar levels decreased the β-AR-mediated positive inotropic effect and enhancement of the Ca2+ transient amplitude, without changing NO production. Positive inotropic responses to β1-AR (but not β2-AR) activation were markedly attenuated by 25HC. The depressant action of 25HC on the β1-AR-mediated responses was prevented by selective β3-AR antagonists as well as inhibitors of Gi protein, Gβγ, G protein-coupled receptor kinase 2/3, or β-arrestin. Simultaneously, blockers of protein kinase D and C as well as a phosphodiesterase inhibitor did not preclude the negative action of 25HC on the inotropic response to β-AR activation. Thus, 25HC can suppress the β1-AR-dependent effects via engaging β3-AR, Gi protein, Gβγ, G protein-coupled receptor kinase, and β-arrestin. This 25HC-dependent mechanism can contribute to the inflammatory-related alterations in the atrial β-adrenergic signaling.
Collapse
Affiliation(s)
- Julia G Odnoshivkina
- Kazan State Medical University, 49 Butlerova St, Kazan, RT, Russia, 420012
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center of RAS, 2/31 Lobachevsky St, Kazan, RT, Russia, 420111
| | - Alexey S Averin
- Institute of Cell Biophysics, Federal Research Center "Pushchino Scientific Center of Biological Research", Pushchino Branch, Russian Academy of Sciences, Pushchino, 142290, Russia
| | - Ildar R Khakimov
- Kazan State Medical University, 49 Butlerova St, Kazan, RT, Russia, 420012
| | - Nazar A Trusov
- Kazan State Medical University, 49 Butlerova St, Kazan, RT, Russia, 420012
| | - Diliara A Trusova
- Kazan State Medical University, 49 Butlerova St, Kazan, RT, Russia, 420012
| | - Alexey M Petrov
- Kazan State Medical University, 49 Butlerova St, Kazan, RT, Russia, 420012.
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center of RAS, 2/31 Lobachevsky St, Kazan, RT, Russia, 420111.
- Kazan Federal University, 18 Kremlyovskaya Street, Kazan, Russia, 420008.
| |
Collapse
|
25
|
Wang J, Ilyas N, Ren Y, Ji Y, Li S, Li C, Liu F, Gu D, Ang KW. Technology and Integration Roadmap for Optoelectronic Memristor. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307393. [PMID: 37739413 DOI: 10.1002/adma.202307393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/10/2023] [Indexed: 09/24/2023]
Abstract
Optoelectronic memristors (OMs) have emerged as a promising optoelectronic Neuromorphic computing paradigm, opening up new opportunities for neurosynaptic devices and optoelectronic systems. These OMs possess a range of desirable features including minimal crosstalk, high bandwidth, low power consumption, zero latency, and the ability to replicate crucial neurological functions such as vision and optical memory. By incorporating large-scale parallel synaptic structures, OMs are anticipated to greatly enhance high-performance and low-power in-memory computing, effectively overcoming the limitations of the von Neumann bottleneck. However, progress in this field necessitates a comprehensive understanding of suitable structures and techniques for integrating low-dimensional materials into optoelectronic integrated circuit platforms. This review aims to offer a comprehensive overview of the fundamental performance, mechanisms, design of structures, applications, and integration roadmap of optoelectronic synaptic memristors. By establishing connections between materials, multilayer optoelectronic memristor units, and monolithic optoelectronic integrated circuits, this review seeks to provide insights into emerging technologies and future prospects that are expected to drive innovation and widespread adoption in the near future.
Collapse
Affiliation(s)
- Jinyong Wang
- School of Optoelectronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 611731, P. R. China
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Nasir Ilyas
- School of Optoelectronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 611731, P. R. China
| | - Yujing Ren
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore, 117585, Singapore
| | - Yun Ji
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Sifan Li
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Changcun Li
- School of Optoelectronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 611731, P. R. China
| | - Fucai Liu
- School of Optoelectronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 611731, P. R. China
| | - Deen Gu
- School of Optoelectronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 611731, P. R. China
- State Key Laboratory of Electronic Thin Films and Integrated Devices, University of Electronic Science and Technology of China, Chengdu, 611731, P. R. China
| | - Kah-Wee Ang
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore, 117576, Singapore
- Institute of Materials Research and Engineering, A*STAR, Singapore, 138634, Singapore
| |
Collapse
|
26
|
Zhang D, Chen H, Wang J, Ji J, Imam M, Zhang Z, Yan S. Current progress and prospects for G protein-coupled estrogen receptor in triple-negative breast cancer. Front Cell Dev Biol 2024; 12:1338448. [PMID: 38476263 PMCID: PMC10928007 DOI: 10.3389/fcell.2024.1338448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 02/08/2024] [Indexed: 03/14/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a biologically and clinically heterogeneous disease. The G protein-coupled estrogen receptor (GPER) plays a crucial role in mediating the effect of estrogen and estrogen-like compounds in TNBC cells. Compared with other subtypes, GPER has a higher expression in TNBC. The GPER mechanisms have been thoroughly characterized and analyzed in estrogen receptor α (ERα) positive breast cancer, but not in TNBC. Our previous work revealed that a higher expression of GPER mRNA indicates a better prognosis for ERα-positive breast cancer; however, its effects in TNBC differ. Whether GPER could serve as a predictive prognostic marker or therapeutic target for TNBC remains unclear. In this review, we provide a detailed introduction to the subcellular localization of GPER, the different effects of various ligands, and the interactions between GPER and closely associated factors in TNBC. We focused on the internal molecular mechanisms specific to TNBC and thoroughly explored the role of GPER in promoting tumor development. We also discussed the interaction of GPER with specific cytokines and chemokines, and the relationship between GPER and immune evasion. Additionally, we discussed the feasibility of using GPER as a therapeutic target in the context of existing studies. This comprehensive review highlights the effects of GPER on TNBC, providing a framework and directions for future research.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shunchao Yan
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
27
|
Zi Z, Rao Y. Discoveries of GPR39 as an evolutionarily conserved receptor for bile acids and of its involvement in biliary acute pancreatitis. SCIENCE ADVANCES 2024; 10:eadj0146. [PMID: 38306436 PMCID: PMC10836733 DOI: 10.1126/sciadv.adj0146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 01/04/2024] [Indexed: 02/04/2024]
Abstract
Acute pancreatitis (AP) is one of the most common gastrointestinal diseases. Bile acids (BAs) were proposed to be a cause of AP nearly 170 years ago, though the underlying mechanisms remain unclear. Here, we report that two G protein-coupled receptors, GPR39 and GHSR, mediated cellular responses to BAs. Our results revealed GPR39 as an evolutionarily conserved receptor for BAs, particularly 3-O-sulfated lithocholic acids. In cultured cell lines, GPR39 is sufficient for BA-induced Ca2+ elevation. In pancreatic acinar cells, GPR39 mediated BA-induced Ca2+ elevation and necrosis. Furthermore, AP induced by BAs was significantly reduced in GPR39 knockout mice. Our findings provide in vitro and in vivo evidence demonstrating that GPR39 is necessary and sufficient to mediate BA signaling, highlighting its involvement in biliary AP pathogenesis, and suggesting it as a promising therapeutic target for biliary AP.
Collapse
Affiliation(s)
- Zhentao Zi
- Chinese Institutes for Medical Research, Beijing (CIMR, Beijing) and the State Key Laboratory of Digestive Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, School of Life Sciences, School of Pharmaceutical Sciences, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Yi Rao
- Chinese Institutes for Medical Research, Beijing (CIMR, Beijing) and the State Key Laboratory of Digestive Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, School of Life Sciences, School of Pharmaceutical Sciences, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Changping Laboratory, Chinese Institute of Brain Research Beijing and Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing 102206, China
| |
Collapse
|
28
|
Wang W, Li J, Cui S, Li J, Ye X, Wang Z, Zhang T, Jiang X, Kong Y, Chen X, Chen YQ, Zhu S. Microglial Ffar4 deficiency promotes cognitive impairment in the context of metabolic syndrome. SCIENCE ADVANCES 2024; 10:eadj7813. [PMID: 38306420 PMCID: PMC10836723 DOI: 10.1126/sciadv.adj7813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 01/03/2024] [Indexed: 02/04/2024]
Abstract
Metabolic syndrome (MetS) is closely associated with an increased risk of dementia and cognitive impairment, and a complex interaction of genetic and environmental dietary factors may be implicated. Free fatty acid receptor 4 (Ffar4) may bridge the genetic and dietary aspects of MetS development. However, the role of Ffar4 in MetS-related cognitive dysfunction is unclear. In this study, we found that Ffar4 expression is down-regulated in MetS mice and MetS patients with cognitive impairment. Conventional and microglial conditional knockout of Ffar4 exacerbated high-fat diet (HFD)-induced cognitive dysfunction and anxiety, whereas microglial Ffar4 overexpression improved HFD-induced cognitive dysfunction and anxiety. Mechanistically, we found that microglial Ffar4 regulated microglial activation through type I interferon signaling. Microglial depletion and NF-κB inhibition partially reversed cognitive dysfunction and anxiety in microglia-specific Ffar4 knockout MetS mice. Together, these findings uncover a previously unappreciated role of Ffar4 in negatively regulating the NF-κB-IFN-β signaling and provide an attractive therapeutic target for delaying MetS-associated cognitive decline.
Collapse
Affiliation(s)
- Wei Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Jinyou Li
- Affiliated Hospital of Jiangnan University, Wuxi 214122, China
| | - Siyuan Cui
- Jiangnan University Medical Center, Wuxi 214002, China
| | - Jiayu Li
- Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Xianlong Ye
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China
| | - Zhe Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Tingting Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Xuan Jiang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Yulin Kong
- Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Xin Chen
- Jiangnan University Medical Center, Wuxi 214002, China
| | - Yong Q. Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Shenglong Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
- Jiangnan University Medical Center, Wuxi 214002, China
| |
Collapse
|
29
|
Wang J, Guo Y, He Y, Qin Y, Li X, Yang L, Liu K, Xiao L. Hepatic regulator of G protein signaling 14 ameliorates NAFLD through activating cAMP-AMPK signaling by targeting Giα1/3. Mol Metab 2024; 80:101882. [PMID: 38237897 PMCID: PMC10844864 DOI: 10.1016/j.molmet.2024.101882] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 01/28/2024] Open
Abstract
OBJECTIVE Nonalcoholic fatty liver disease (NAFLD) is an emerging public health threat as the most common chronic liver disease worldwide. However, there remains no effective medication to improve NAFLD. G protein-coupled receptors (GPCRs) are the most frequently investigated drug targets family. The Regulator of G protein signaling 14 (RGS14), as an essential negative modulator of GPCR signaling, plays important regulatory roles in liver damage and inflammatory responses. However, the role of RGS14 in NAFLD remains largely unclear. METHODS AND RESULTS In this study, we found that RGS14 was decreased in hepatocytes in NAFLD individuals in a public database. We employed genetic engineering technique to explore the function of RGS14 in NAFLD. We demonstrated that RGS14 overexpression ameliorated lipid accumulation, inflammatory response and liver fibrosis in hepatocytes in vivo and in vitro. Whereas, hepatocyte specific Rgs14-knockout (Rgs14-HKO) exacerbated high fat high cholesterol diet (HFHC) induced NASH. Further molecular experiments demonstrated that RGS14 depended on GDI activity to attenuate HFHC-feeding NASH. More importantly, RGS14 interacted with Guanine nucleotide-binding protein (Gi) alpha 1 and 3 (Giα1/3, gene named GNAI1/3), promoting the generation of cAMP and then activating the subsequent AMPK pathways. GNAI1/3 knockdown abolished the protective role of RGS14, indicating that RGS14 binding to Giα1/3 was required for prevention against hepatic steatosis. CONCLUSIONS RGS14 plays a protective role in the progression of NAFLD. RGS14-Giα1/3 interaction accelerated the production of cAMP and then activated cAMP-AMPK signaling. Targeting RGS14 or modulating the RGS14-Giα1/3 interaction may be a potential strategy for the treatment of NAFLD in the future.
Collapse
Affiliation(s)
- Junyong Wang
- Center for Basic Medical Research, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yaping Guo
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yunduan He
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Yifan Qin
- Center for Basic Medical Research, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xiuling Li
- Department of Gastroenterology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan 450004, China
| | - Ling Yang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Kangdong Liu
- Center for Basic Medical Research, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Li Xiao
- Department of Gastroenterology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan 450004, China.
| |
Collapse
|
30
|
Gupta PK, Singh A, Rana S. Conformational variants of the ternary complex of C5a, C5aR1, and G-protein. J Biomol Struct Dyn 2024:1-16. [PMID: 38247266 DOI: 10.1080/07391102.2024.2305698] [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: 09/09/2023] [Accepted: 01/09/2024] [Indexed: 01/23/2024]
Abstract
The complement component fragment 5a (C5a) binds and activates two complement receptors like C5aR1 and C5aR2, which play a significant role in orchestrating the proinflammatory function of C5a in tissues through the recruitment of heterotrimeric G-proteins and β-arrestins. Dysregulation of the complement induces excessive production of C5a, which triggers aberrant activation of the C5a-C5aR1-G-protein and C5a-C5aR2-β-arrestin signalling axes in tissues, contributing to the pathology of numerous immune-inflammatory diseases. Thus, understanding the interaction of C5a with C5aR1 and C5aR2, as well as the interaction of G-protein and β-arrestins, respectively, with C5a-C5aR1 and C5a-C5aR2, holds tremendous therapeutic value. In the absence of structural data, we have previously elaborated the binary complexes of C5a-C5aR1 and C5a-C5aR2, as well as the ternary complex of C5a-C5aR2-β-arrestin1, in highly refined model structures. While our ternary model complex of C5a-C5aR1-G-protein was in progress, two cryo-electron microscopy-based ternary structural complexes of C5aR1 were made available by others. However, it is observed that the interaction of the crucial NT-peptide of C5aR1 with C5a, including the portion of the G⍺i-subunit that harbors the switch-I region, is not fully resolved in both complexes. The current study addresses the issues and provides two highly refined alternative model ternary complexes of C5a-C5aR1-G-protein. The study highlights the conformational heterogeneity in C5aR1 by comparing the two conformational variants of the model ternary complex in the context of C5a-C5aR2-β-arrestin1 for further devising methods and molecules targeting both surface and intracellular C5aR1/C5aR2 for effectively mitigating the proinflammatory role of C5a in various disease settings.
Collapse
Affiliation(s)
- Pulkit Kr Gupta
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha, India
| | - Aditi Singh
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha, India
| | - Soumendra Rana
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha, India
| |
Collapse
|
31
|
Kumar V, Chunchagatta Lakshman PK, Prasad TK, Manjunath K, Bairy S, Vasu AS, Ganavi B, Jasti S, Kamariah N. Target-based drug discovery: Applications of fluorescence techniques in high throughput and fragment-based screening. Heliyon 2024; 10:e23864. [PMID: 38226204 PMCID: PMC10788520 DOI: 10.1016/j.heliyon.2023.e23864] [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: 05/09/2023] [Revised: 12/14/2023] [Accepted: 12/14/2023] [Indexed: 01/17/2024] Open
Abstract
Target-based discovery of first-in-class therapeutics demands an in-depth understanding of the molecular mechanisms underlying human diseases. Precise measurements of cellular and biochemical activities are critical to gain mechanistic knowledge of biomolecules and their altered function in disease conditions. Such measurements enable the development of intervention strategies for preventing or treating diseases by modulation of desired molecular processes. Fluorescence-based techniques are routinely employed for accurate and robust measurements of in-vitro activity of molecular targets and for discovering novel chemical molecules that modulate the activity of molecular targets. In the current review, the authors focus on the applications of fluorescence-based high throughput screening (HTS) and fragment-based ligand discovery (FBLD) techniques such as fluorescence polarization (FP), Förster resonance energy transfer (FRET), fluorescence thermal shift assay (FTSA) and microscale thermophoresis (MST) for the discovery of chemical probe to exploring target's role in disease biology and ultimately, serve as a foundation for drug discovery. Some recent advancements in these techniques for compound library screening against important classes of drug targets, such as G-protein-coupled receptors (GPCRs) and GTPases, as well as phosphorylation- and acetylation-mediated protein-protein interactions, are discussed. Overall, this review presents a landscape of how these techniques paved the way for the discovery of small-molecule modulators and biologics against these targets for therapeutic benefits.
Collapse
Affiliation(s)
| | | | - Thazhe Kootteri Prasad
- Centre for Chemical Biology & Therapeutics, inStem & NCBS, Bellary Road, Bangalore, 560065, India
| | - Kavyashree Manjunath
- Centre for Chemical Biology & Therapeutics, inStem & NCBS, Bellary Road, Bangalore, 560065, India
| | - Sneha Bairy
- Centre for Chemical Biology & Therapeutics, inStem & NCBS, Bellary Road, Bangalore, 560065, India
| | - Akshaya S. Vasu
- Centre for Chemical Biology & Therapeutics, inStem & NCBS, Bellary Road, Bangalore, 560065, India
| | - B. Ganavi
- Centre for Chemical Biology & Therapeutics, inStem & NCBS, Bellary Road, Bangalore, 560065, India
| | - Subbarao Jasti
- Centre for Chemical Biology & Therapeutics, inStem & NCBS, Bellary Road, Bangalore, 560065, India
| | - Neelagandan Kamariah
- Centre for Chemical Biology & Therapeutics, inStem & NCBS, Bellary Road, Bangalore, 560065, India
| |
Collapse
|
32
|
Rodriguez FD, Covenas R. Association of Neurokinin-1 Receptor Signaling Pathways with Cancer. Curr Med Chem 2024; 31:6460-6486. [PMID: 37594106 DOI: 10.2174/0929867331666230818110812] [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: 02/03/2023] [Revised: 06/14/2023] [Accepted: 07/01/2023] [Indexed: 08/19/2023]
Abstract
BACKGROUND Numerous biochemical reactions leading to altered cell proliferation cause tumorigenesis and cancer treatment resistance. The mechanisms implicated include genetic and epigenetic changes, modified intracellular signaling, and failure of control mechanisms caused by intrinsic and extrinsic factors alone or combined. No unique biochemical events are responsible; entangled molecular reactions conduct the resident cells in a tissue to display uncontrolled growth and abnormal migration. Copious experimental research supports the etiological responsibility of NK-1R (neurokinin-1 receptor) activation, alone or cooperating with other mechanisms, in cancer appearance in different tissues. Consequently, a profound study of this receptor system in the context of malignant processes is essential to design new treatments targeting NK-1R-deviated activity. METHODS This study reviews and discusses recent literature that analyzes the main signaling pathways influenced by the activation of neurokinin 1 full and truncated receptor variants. Also, the involvement of NK-1R in cancer development is discussed. CONCLUSION NK-1R can signal through numerous pathways and cross-talk with other receptor systems. The participation of override or malfunctioning NK-1R in malignant processes needs a more precise definition in different types of cancers to apply satisfactory and effective treatments. A long way has already been traveled: the current disposal of selective and effective NK-1R antagonists and the capacity to develop new drugs with biased agonistic properties based on the receptor's structural states with functional significance opens immediate research action and clinical application.
Collapse
Affiliation(s)
- Francisco David Rodriguez
- Department of Biochemistry and Molecular Biology, Faculty of Chemical Sciences, University of Salamanca, 37007 Salamanca, Spain
- Group GIR USAL: BMD (Bases Moleculares del Desarrollo), University of Salamanca, Salamanca, Spain
| | - Rafael Covenas
- Group GIR USAL: BMD (Bases Moleculares del Desarrollo), University of Salamanca, Salamanca, Spain
- Laboratory of Neuroanatomy of the Peptidergic Systems, Institute of Neurosciences of Castilla y León (INCYL), University of Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
33
|
Su X, Yang Y, Gao Y, Wang J, Hao Y, Zhang Y, Yang R. Gut microbiota CLA and IL-35 induction in macrophages through Gαq/11-mediated STAT1/4 pathway: an animal-based study. Gut Microbes 2024; 16:2437253. [PMID: 39636005 PMCID: PMC11622586 DOI: 10.1080/19490976.2024.2437253] [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: 05/07/2024] [Revised: 11/05/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
Gut microbiota/metabolites not only participate in the food and energy metabolism but also contribute to the host immune response and homeostasis. The alternation of gut microbiota/metabolites has been widely related to intestinal and extra-intestinal disorders such as intestinal bowel diseases (IBDs). Bactericidal substances from gut epithelial cells can regulate the composition of gut microbiota. However, the effects of regenerating protein 4 (REG4) (human)/(Reg4) (mice), a potentially bactericidal substance from gut epithelial cells, on the gut immune homeostasis maintain elusive. Here, we found that REG4/Reg4 is essential in maintaining gut immune homeostasis through REG4/Reg4 associated gut microbiota. Reg4 knockout (KO) mice were highly sensitive to DSS-mediated colitis, whereas human REG4 intestine epithelial cell transgenic (huREG4IECtg) mice exhibited more resistance to DSS-mediated colitis. Mechanistically, sequencing of gut microbiota and liquid chromatography-mass spectrometry showed that REG4/Reg4 could affect the composition of gut microbiota. REG4/Reg4 associated gut microbiota such as Lactobacillus could metabolize linoleic acid (LA) into conjugated linoleic acid (CLA). Immunoprecipitation and immunoblot showed that CLA could effectively promote the expression of IL-35 in macrophages through Gαq/11 mediated activation STAT1/4. Thus, our results demonstrate that REG4/Reg4 plays a critical role in maintaining gut immune homeostasis through CLA-mediated IL-35+ macrophages.
Collapse
Affiliation(s)
- Xiaomin Su
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Yazheng Yang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Yunhuan Gao
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Juanjuan Wang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Yang Hao
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Yuan Zhang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Rongcun Yang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
- Translational Medicine Institute, Tianjin Union Medical Center of Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| |
Collapse
|
34
|
Lyu C, Bhimani AK, Draus WT, Weigel R, Chen S. Active Gα i/o Mutants Accelerate Breast Tumor Metastasis via the c-Src Pathway. Mol Cell Biol 2023; 43:650-663. [PMID: 38099640 PMCID: PMC10761066 DOI: 10.1080/10985549.2023.2285833] [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: 09/22/2023] [Revised: 11/14/2023] [Accepted: 11/14/2023] [Indexed: 12/21/2023] Open
Abstract
Constitutively active mutations in the Gαi2 and GαoA subunits of heterotrimeric G proteins have been found in various human cancers, including breast cancer, but their precise roles in tumor formation, progression, and metastasis remain poorly understood. This study focused on GαoAR243H and Gαi2R179C mutants in breast cancer. These mutants alone were insufficient to initiate mammary tumor formation in mice. However, when introduced into transgenic mouse models of breast cancer induced by Neu expression or PTEN loss, the Gαi2R179C mutant notably enhanced spontaneous lung metastasis, without affecting primary tumor initiation and growth. Ectopic expression of the GαoAR243H and Gαi2R179C mutants in tumor cells promoted cell migration in vitro and dissemination into multiple organs in vivo by activating the c-Src signaling pathway. These mutants activate c-Src through direct interaction, involving specific residues in the switch domains II of Gαi subunits, which only partially overlap with those involved in inhibiting adenylyl cyclases. This study uncovers a critical role of Gαi/o signaling in accelerating breast cancer metastasis through the c-Src pathway. These findings hold clinical significance as they may pave the way for personalized therapies targeting c-Src to inhibit breast cancer metastasis in patients with active Gαi/o mutations or elevated Gαi/o signaling.
Collapse
Affiliation(s)
- Cancan Lyu
- The Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Aarzoo K. Bhimani
- The Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - William T. Draus
- The Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Ronald Weigel
- The Department of Surgery, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Songhai Chen
- The Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- The Holden Comprehensive Cancer Center, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
35
|
Lin J, Scullion L, Garland CJ, Dora K. Gβγ subunit signalling underlies neuropeptide Y-stimulated vasoconstriction in rat mesenteric and coronary arteries. Br J Pharmacol 2023; 180:3045-3058. [PMID: 37460913 PMCID: PMC10953346 DOI: 10.1111/bph.16192] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 06/27/2023] [Accepted: 07/09/2023] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND AND PURPOSE Raised serum concentrations of the sympathetic co-transmitter neuropeptide Y (NPY) are linked to cardiovascular diseases. However, the signalling mechanism for vascular smooth muscle (VSM) constriction to NPY is poorly understood. Therefore, the present study investigated the mechanisms of NPY-induced vasoconstriction in rat small mesenteric (RMA) and coronary (RCA) arteries. EXPERIMENTAL APPROACH Third-order mesenteric or intra-septal arteries from male Wistar rats were assessed in wire myographs for isometric tension, VSM membrane potential and VSM intracellular Ca2+ events. KEY RESULTS NPY stimulated concentration-dependent vasoconstriction in both RMA and RCA, which was augmented by blocking NO synthase or endothelial denudation in RMA. NPY-mediated vasoconstriction was blocked by the selective Y1 receptor antagonist BIBO 3304 and Y1 receptor protein expression was detected in both the VSM and endothelial cells in RMA and RCA. The selective Gβγ subunit inhibitor gallein and the PLC inhibitor U-73122 attenuated NPY-induced vasoconstriction. Signalling via the Gβγ-PLC pathway stimulated VSM Ca2+ waves and whole-field synchronised Ca2+ flashes in RMA and increased the frequency of Ca2+ flashes in myogenically active RCA. Furthermore, in RMA, the Gβγ pathway linked NPY to VSM depolarization and generation of action potential-like spikes associated with intense vasoconstriction. This depolarization activated L-type voltage-gated Ca2+ channels, as nifedipine abolished NPY-mediated vasoconstriction. CONCLUSIONS AND IMPLICATIONS These data suggest that the Gβγ subunit, which dissociates upon Y1 receptor activation, initiates VSM membrane depolarization and Ca2+ mobilisation to cause vasoconstriction. This model may help explain the development of microvascular vasospasm during raised sympathetic nerve activity.
Collapse
Affiliation(s)
- JinHeng Lin
- Department of PharmacologyUniversity of OxfordOxfordUK
| | | | | | - Kim Dora
- Department of PharmacologyUniversity of OxfordOxfordUK
| |
Collapse
|
36
|
Kostenis E, Gomeza J, Miess-Tanneberg E, Blum NK, Benkel T, Chevigné A, Hoffmann C, Kolb P, Nikolaev V, Waldhoer M, Szpakowska M, Inoue A, Schulz S. Reply to: How carvedilol does not activate β 2-adrenoceptors. Nat Commun 2023; 14:7867. [PMID: 38036502 PMCID: PMC10689814 DOI: 10.1038/s41467-023-42849-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 10/18/2023] [Indexed: 12/02/2023] Open
Affiliation(s)
- Evi Kostenis
- Molecular, Cellular, and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, 53115, Bonn, Germany.
| | - Jesus Gomeza
- Molecular, Cellular, and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, 53115, Bonn, Germany
| | - Elke Miess-Tanneberg
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University of Jena, 07747, Jena, Germany
| | - Nina Kathleen Blum
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University of Jena, 07747, Jena, Germany
| | - Tobias Benkel
- Molecular, Cellular, and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, 53115, Bonn, Germany
- ISAR Bioscience, Semmelweisstraße 5, 82152, Planegg, Germany
| | - Andy Chevigné
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health (LIH), L-4354, Esch-sur-Alzette, Luxembourg
| | - Carsten Hoffmann
- Institute for Molecular Cell Biology, CMB-Center for Molecular Biomedicine, Jena University Hospital, Friedrich Schiller University of Jena, 07745, Jena, Germany
| | - Peter Kolb
- Department of Pharmaceutical Chemistry, Philipps-University of Marburg, 35032, Marburg, Germany
| | - Viacheslav Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Maria Waldhoer
- InterAx Biotech AG, 5234, Villigen, Switzerland
- Ikherma Consulting Ltd, Hitchin, SG4 0TY, UK
| | - Martyna Szpakowska
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health (LIH), L-4354, Esch-sur-Alzette, Luxembourg
| | - Asuka Inoue
- Graduate School of Pharmaceutical Science, Tohoku University, Sendai, 980-8578, Japan
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University of Jena, 07747, Jena, Germany
| |
Collapse
|
37
|
Zhang K, Zhang MX, Meng XX, Zhu J, Wang JJ, He YF, Li YH, Zhao SC, Shi ZM, Zheng LN, Han T, Hong W. Targeting GPR65 alleviates hepatic inflammation and fibrosis by suppressing the JNK and NF-κB pathways. Mil Med Res 2023; 10:56. [PMID: 38001521 PMCID: PMC10675918 DOI: 10.1186/s40779-023-00494-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND G-protein coupled receptors (GPCRs) are recognized as attractive targets for drug therapy. However, it remains poorly understood how GPCRs, except for a few chemokine receptors, regulate the progression of liver fibrosis. Here, we aimed to reveal the role of GPR65, a proton-sensing receptor, in liver fibrosis and to elucidate the underlying mechanism. METHODS The expression level of GPR65 was evaluated in both human and mouse fibrotic livers. Furthermore, Gpr65-deficient mice were treated with either bile duct ligation (BDL) for 21 d or carbon tetrachloride (CCl4) for 8 weeks to investigate the role of GPR65 in liver fibrosis. A combination of experimental approaches, including Western blotting, quantitative real-time reverse transcription‑polymerase chain reaction (qRT-PCR), and enzyme-linked immunosorbent assay (ELISA), confocal microscopy and rescue studies, were used to explore the underlying mechanisms of GPR65's action in liver fibrosis. Additionally, the therapeutic potential of GPR65 inhibitor in the development of liver fibrosis was investigated. RESULTS We found that hepatic macrophages (HMs)-enriched GPR65 was upregulated in both human and mouse fibrotic livers. Moreover, knockout of Gpr65 significantly alleviated BDL- and CCl4-induced liver inflammation, injury and fibrosis in vivo, and mouse bone marrow transplantation (BMT) experiments further demonstrated that the protective effect of Gpr65 knockout is primarily mediated by bone marrow-derived macrophages (BMMs). Additionally, in vitro data demonstrated that Gpr65 silencing and GPR65 antagonist inhibited, while GPR65 overexpression and application of GPR65 endogenous and exogenous agonists enhanced the expression and release of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and transforming growth factor-β (TGF-β), all of which subsequently promoted the activation of hepatic stellate cells (HSCs) and the damage of hepatocytes (HCs). Mechanistically, GPR65 overexpression, the acidic pH and GPR65 exogenous agonist induced up-regulation of TNF-α and IL-6 via the Gαq-Ca2+-JNK/NF-κB pathways, while promoted the expression of TGF-β through the Gαq-Ca2+-MLK3-MKK7-JNK pathway. Notably, pharmacological GPR65 inhibition retarded the development of inflammation, HCs injury and fibrosis in vivo. CONCLUSIONS GPR65 is a major regulator that modulates the progression of liver fibrosis. Thus, targeting GPR65 could be an effective therapeutic strategy for the prevention of liver fibrosis.
Collapse
Affiliation(s)
- Kun Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Meng-Xia Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Xiao-Xiang Meng
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Jing Zhu
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Jia-Jun Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Yi-Fan He
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Ye-Hua Li
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Si-Cong Zhao
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Zhe-Min Shi
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Li-Na Zheng
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Tao Han
- Department of Hepatology and Gastroenterology, Tianjin Union Medical Center, Tianjin Medical University, Tianjin Union Medical Center affiliated to Nankai University, Tianjin, 300000, China.
| | - Wei Hong
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
38
|
Alabdali R, Franchini L, Orlandi C. G α Protein Signaling Bias at Serotonin 1A Receptor. Mol Pharmacol 2023; 104:230-238. [PMID: 37567783 PMCID: PMC10586511 DOI: 10.1124/molpharm.123.000722] [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: 05/18/2023] [Revised: 07/11/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Serotonin 1A receptor (5-HT1AR) is a clinically relevant target because of its involvement in several central and peripheral functions, including sleep, temperature homeostasis, processing of emotions, and response to stress. As a G protein coupled receptor (GPCR) activating numerous Gα i/o/z family members, 5-HT1AR can potentially modulate multiple intracellular signaling pathways in response to different therapeutics. Here, we applied a cell-based bioluminescence resonance energy transfer assay to quantify how ten structurally diverse 5-HT1AR agonists exert biased signaling by differentially stimulating Gα i/o/z family members. Our concentration-response analysis of the activation of each Gα i/o/z protein revealed unique potency and efficacy profiles of selected agonists when compared with the reference 5-hydroxytryptamine, serotonin. Overall, our analysis of signaling bias identified groups of ligands sharing comparable G protein activation selectivity and also drugs with unique selectivity profiles. We observed, for example, a strong bias of F-15599 toward the activation of Gα i3 that was unique among the agonists tested: we found a biased factor of +2.19 when comparing the activation of Gα i3 versus Gα i2 by F-15599, while it was -0.29 for 8-hydroxy-2-(di-n-propylamino) tetralin. Similarly, vortioxetine showed a biased factor of +1.06 for Gα z versus Gα oA, while it was -1.38 for vilazodone. Considering that alternative signaling pathways are regulated downstream of each Gα protein, our data suggest that the unique pharmacological properties of the tested agonists could result in multiple unrelated cellular outcomes. Further investigation is needed to reveal how this type of ligand bias could affect cellular responses and to illuminate the molecular mechanisms underlying therapeutic profile and side effects of each drug. SIGNIFICANCE STATEMENT: Serotonin 1a receptor (5-HT1AR) activates several members of the Gi/o/z protein family. Here, we examined ten structurally diverse and clinically relevant agonists acting on 5-HT1AR and identified distinctive bias patterns among G proteins. Considering the diversity of their intracellular effectors and signaling properties, this data reveal novel mechanisms underlying both therapeutic and undesirable effects.
Collapse
Affiliation(s)
- Rana Alabdali
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY
| | - Luca Franchini
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY
| | - Cesare Orlandi
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY
| |
Collapse
|
39
|
Cui X, Qin B, Xia C, Li H, Li Z, Li Z, Nasir A, Bai Q. Transcriptome-wide analysis of trigeminal ganglion and subnucleus caudalis in a mouse model of chronic constriction injury-induced trigeminal neuralgia. Front Pharmacol 2023; 14:1230633. [PMID: 37841912 PMCID: PMC10568182 DOI: 10.3389/fphar.2023.1230633] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 09/05/2023] [Indexed: 10/17/2023] Open
Abstract
Trigeminal neuropathic pain (TNP) induces mechanical allodynia and hyperalgesia, which are known to alter gene expression in injured dorsal root ganglia primary sensory neurons. Non-coding RNAs (ncRNAs) have been linked to TNP. However, the functional mechanism underlying TNP and the expression profile of ncRNAs in the trigeminal ganglion (TG) and trigeminal subnucleus caudalis (Sp5C) are still unknown. We used RNA sequencing and bioinformatics analysis to examine the TG and Sp5C transcriptomes after infraorbital nerve chronic constrictive injury (IoN-CCI). The robust changes in the gene expression of lncRNAs, circRNAs, and mRNAs were observed within the TG and Sp5C from mice that underwent IoN-CCI and the sham-operated mice (day 7). In total, 111,003 lncRNAs were found in TG and 107,157 in Sp5C; 369 lncRNAs were differentially expressed in TG, and 279 lncRNAs were differentially expressed in Sp5C. In addition, 13,216 circRNAs in TG and 21,658 circRNAs in Sp5C were identified, with 1,155 circRNAs and 2,097 circRNAs differentially expressed in TG and Sp5C, respectively. Furthermore, 5,205 DE mRNAs in TG and 3,934 DE mRNAs in Sp5C were differentially expressed between IoN-CCI and sham groups. The study revealed a high correlation of pain-related differentially expressed genes in the TG and Sp5C to anxiety, depression, inflammation, neuroinflammation, and apoptosis. Gene Ontology analysis revealed that binding-related molecular functions and membrane-related cell components were significantly enriched. Kyoto Encyclopedia of Genes and Genomes analysis shows the most significant enrichments in neurogenesis, nervous system development, neuron differentiation, adrenergic signaling, cAMP signaling, MAPK signaling, and PI3K-Akt signaling pathways. Furthermore, protein-protein interaction analysis showed that hub genes were implicated in neuropeptide signaling pathways. Functional analysis of DE ncRNA-targeting genes was mostly enriched with nociception-related signaling pathways underpinning TNP. Our findings suggest that ncRNAs are involved in TNP development and open new avenues for research and treatment.
Collapse
Affiliation(s)
- Xiaona Cui
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Anesthesiology, International Peace Maternity & Child Health Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Bo Qin
- Translational Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chaoyun Xia
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hong Li
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhiye Li
- Department of Pharmacy, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhisong Li
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Abdul Nasir
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qian Bai
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
40
|
Pepanian A, Binbay FA, Roy S, Nubbemeyer B, Koley A, Rhodes CA, Ammer H, Pei D, Ghosh P, Imhof D. Bicyclic Peptide Library Screening for the Identification of Gαi Protein Modulators. J Med Chem 2023; 66:12396-12406. [PMID: 37587416 PMCID: PMC11000586 DOI: 10.1021/acs.jmedchem.3c00873] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
Noncanonical G protein activation and inactivation, particularly for the Gαi/s protein subfamilies, have long been a focus of chemical research. Combinatorial libraries were already effectively applied to identify modulators of the guanine-nucleotide exchange, as can be exemplified with peptides such as KB-752 and GPM-1c/d, the so-called guanine-nucleotide exchange modulators. In this study, we identified novel bicyclic peptides from a combinatorial library screening that show prominent properties as molecular switch-on/off modulators of Gαi signaling. Among the series of hits, the exceptional paradigm of GPM-3, a protein and state-specific bicyclic peptide, is the first chemically identified GAP (GTPase-activating protein) modulator with a high binding affinity for Gαi protein. Computational analyses identified and assessed the structure of the bicyclic peptides, novel ligand-protein interaction sites, and their subsequent impact on the nucleotide binding site. This approach can therefore lead the way for the development of efficient chemical biological probes targeting Gαi protein modulation within a cellular context.
Collapse
Affiliation(s)
- Anna Pepanian
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenbeurg 4, Bonn 53121, Germany
| | - Furkan Ayberk Binbay
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenbeurg 4, Bonn 53121, Germany
| | - Suchismita Roy
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California 92093, United States
| | - Britta Nubbemeyer
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenbeurg 4, Bonn 53121, Germany
| | - Amritendu Koley
- Department of Chemistry and Biochemistry, The Ohio State University, 578 Biological Sciences Building, 484 W 12th Avenue, Columbus, Ohio 43210, United States
| | - Curran A Rhodes
- Department of Chemistry and Biochemistry, The Ohio State University, 578 Biological Sciences Building, 484 W 12th Avenue, Columbus, Ohio 43210, United States
| | - Hermann Ammer
- Institute of Pharmacology Toxicology and Pharmacy, Veterinary Faculty, Ludwig Maximilian University of Munich, Königinstr. 16, Munich 80539, Germany
| | - Dehua Pei
- Department of Chemistry and Biochemistry, The Ohio State University, 578 Biological Sciences Building, 484 W 12th Avenue, Columbus, Ohio 43210, United States
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California 92093, United States
- Department of Medicine, University of California San Diego, La Jolla, California 92093, United States
| | - Diana Imhof
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenbeurg 4, Bonn 53121, Germany
| |
Collapse
|
41
|
Qi Z, Zhong W, Jiao B, Chen K, Yang X, Wang L, Zeng W, Huang J, Xie J. Activation of G-protein-coupled receptor 183 initiates inflammatory pain via macrophage CCL22 secretion. Eur J Pharmacol 2023; 954:175872. [PMID: 37353188 DOI: 10.1016/j.ejphar.2023.175872] [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: 02/23/2023] [Revised: 06/12/2023] [Accepted: 06/20/2023] [Indexed: 06/25/2023]
Abstract
Chronic pain is a major public health problem with limited effective therapeutic options. G-protein-coupled receptors play a significant role in pain modulation; however, whether and how G-protein-coupled receptor 183 participates in pain regulation remain unclear. In the present study, we found that G-protein-coupled receptor 183 expression was specifically upregulated in the hind paws of mice in various inflammatory pain models. Activation of G-protein-coupled receptor 183 induced acute pain, whereas inhibition or silencing of this receptor alleviated mechanical allodynia and thermal hyperalgesia in complete Freund's adjuvant (CFA) model. Mechanistically, activating G-protein-coupled receptor 183 triggers pain responses via the upregulation of C-C motif chemokine 22(CCL22) in macrophages while blocking the CCL22 receptor C-C motif chemokine receptor 4 (CCR4) attenuates pain hypersensitivity. Taken together, our findings indicate that the G-protein-coupled receptor 183-CCL22 axis has a critical role in the development and maintenance of inflammatory pain.
Collapse
Affiliation(s)
- Zhenhua Qi
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Weiqiang Zhong
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Boyu Jiao
- Department of Acupuncture, The First Affiliated Hospital, SunYat-sen University, Guangzhou, Guangdong, 510080, China
| | - Kang Chen
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xiaohua Yang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Linjie Wang
- Department of Human Anatomy and Physiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Weian Zeng
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Junting Huang
- Department of Human Anatomy and Physiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China.
| | - Jingdun Xie
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
42
|
Gebrie A. The melanocortin receptor signaling system and its role in neuroprotection against neurodegeneration: Therapeutic insights. Ann N Y Acad Sci 2023; 1527:30-41. [PMID: 37526975 DOI: 10.1111/nyas.15048] [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] [Indexed: 08/02/2023]
Abstract
The melanocortin signaling system consists of the melanocortin peptides, their distinctive receptors, accessory proteins, and endogenous antagonists. Melanocortin peptides are small peptide hormones that have been studied in a variety of physiological and pathological conditions. There are five types of melanocortin receptors, and they are distributed within the central nervous system and in several tissues of the periphery. The G protein-coupled melanocortin receptors typically signal through adenylyl cyclase and other downstream signaling pathways. Depending on the ligand, surface expression of melanocortin receptor, receptor occupancy period, related proteins, the type of cell, and other parameters, the signaling pathways are complicated and pleiotropic. While it is known that all five melanocortin receptors are coupled to Gs, they can also occasionally couple to Gq or Gi. Both direct and indirect neuroprotection are induced by the melanocortin signaling system. Targeting several of the components of the melanocortin signaling system (ligands, receptors, accessory proteins, signaling effectors, and regulators) may provide therapeutic opportunities. Activation of the melanocortin system improves different functional traits in neurodegenerative diseases. There is a potential for additional melanocortin system interventions by interfering with dimerization or dissociation. This review aims to discuss the melanocortin receptor signaling system and its role in neuroprotection, as well as its therapeutic potential.
Collapse
Affiliation(s)
- Alemu Gebrie
- Department of Biomedical Sciences, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
43
|
Zhu Y, Liu S, Zigmond J, Kaltenbronn KM, Blumer KJ, Moeller KD. A Building Block Approach for the Total Synthesis of YM-385781. European J Org Chem 2023; 26:e202300365. [PMID: 38188369 PMCID: PMC10766104 DOI: 10.1002/ejoc.202300365] [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: 04/19/2023] [Indexed: 01/09/2024]
Abstract
YM-254890 and FR900359 are potent and selective inhibitors of the Gq/11-signaling pathway. As such, they have been attractive targets for both synthesis and biological studies. Yet in spite of this effort, a versatile synthetic approach to the molecules that allows for the rapid construction of a variety of non-natural and labelled analogs and an increase in the amount of those analogs available remains elusive. We report here a convergent building block approach to the molecules that can solve this challenge.
Collapse
Affiliation(s)
- Yu Zhu
- Department of Chemistry, Washington University, St. Louis, MO 63130
| | - Siyue Liu
- Department of Chemistry, Washington University, St. Louis, MO 63130
| | - Johnny Zigmond
- Department of Chemistry, Washington University, St. Louis, MO 63130
| | - Kevin M Kaltenbronn
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO
| | - Kendall J Blumer
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO
| | - Kevin D Moeller
- Department of Chemistry, Washington University, St. Louis, MO 63130
| |
Collapse
|
44
|
Zhao J, DiGiacomo V, Ferreras-Gutierrez M, Dastjerdi S, Ibáñez de Opakua A, Park JC, Luebbers A, Chen Q, Beeler A, Blanco FJ, Garcia-Marcos M. Small-molecule targeting of GPCR-independent noncanonical G-protein signaling in cancer. Proc Natl Acad Sci U S A 2023; 120:e2213140120. [PMID: 37098067 PMCID: PMC10160980 DOI: 10.1073/pnas.2213140120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 03/06/2023] [Indexed: 04/26/2023] Open
Abstract
Activation of heterotrimeric G-proteins (Gαβγ) by G-protein-coupled receptors (GPCRs) is a quintessential mechanism of cell signaling widely targeted by clinically approved drugs. However, it has become evident that heterotrimeric G-proteins can also be activated via GPCR-independent mechanisms that remain untapped as pharmacological targets. GIV/Girdin has emerged as a prototypical non-GPCR activator of G proteins that promotes cancer metastasis. Here, we introduce IGGi-11, a first-in-class small-molecule inhibitor of noncanonical activation of heterotrimeric G-protein signaling. IGGi-11 binding to G-protein α-subunits (Gαi) specifically disrupted their engagement with GIV/Girdin, thereby blocking noncanonical G-protein signaling in tumor cells and inhibiting proinvasive traits of metastatic cancer cells. In contrast, IGGi-11 did not interfere with canonical G-protein signaling mechanisms triggered by GPCRs. By revealing that small molecules can selectively disable noncanonical mechanisms of G-protein activation dysregulated in disease, these findings warrant the exploration of therapeutic modalities in G-protein signaling that go beyond targeting GPCRs.
Collapse
Affiliation(s)
- Jingyi Zhao
- Department of Biochemistry & Cell Biology, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA02118
| | - Vincent DiGiacomo
- Department of Biochemistry & Cell Biology, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA02118
| | | | - Shiva Dastjerdi
- Department of Chemistry, Boston University, College of Arts & Sciences, Boston, MA02115
| | | | - Jong-Chan Park
- Department of Biochemistry & Cell Biology, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA02118
| | - Alex Luebbers
- Department of Biochemistry & Cell Biology, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA02118
| | - Qingyan Chen
- Department of Biochemistry & Cell Biology, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA02118
| | - Aaron Beeler
- Department of Chemistry, Boston University, College of Arts & Sciences, Boston, MA02115
| | - Francisco J. Blanco
- Centro de Investigaciones Biológicas-Centro Superior de Investigaciones Cientificas, Madrid, Spain
| | - Mikel Garcia-Marcos
- Department of Biochemistry & Cell Biology, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA02118
- Department of Biology, College of Arts & Sciences, Boston University, Boston, MA02115
| |
Collapse
|
45
|
Cao Y, Li Y, Liu R, Zhou J, Wang K. Preclinical and Basic Research Strategies for Overcoming Resistance to Targeted Therapies in HER2-Positive Breast Cancer. Cancers (Basel) 2023; 15:cancers15092568. [PMID: 37174034 PMCID: PMC10177527 DOI: 10.3390/cancers15092568] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/16/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
The amplification of epidermal growth factor receptor 2 (HER2) is associated with a poor prognosis and HER2 gene is overexpressed in approximately 15-30% of breast cancers. In HER2-positive breast cancer patients, HER2-targeted therapies improved clinical outcomes and survival rates. However, drug resistance to anti-HER2 drugs is almost unavoidable, leaving some patients with an unmet need for better prognoses. Therefore, exploring strategies to delay or revert drug resistance is urgent. In recent years, new targets and regimens have emerged continuously. This review discusses the fundamental mechanisms of drug resistance in the targeted therapies of HER2-positive breast cancer and summarizes recent research progress in this field, including preclinical and basic research studies.
Collapse
Affiliation(s)
- Yi Cao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Pathology, School of Basic Medical science, Central South University, Changsha 410008, China
| | - Yunjin Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Pathology, School of Basic Medical science, Central South University, Changsha 410008, China
| | - Ruijie Liu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jianhua Zhou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Pathology, School of Basic Medical science, Central South University, Changsha 410008, China
| | - Kuansong Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Pathology, School of Basic Medical science, Central South University, Changsha 410008, China
| |
Collapse
|
46
|
Wagner W, Sobierajska K, Pułaski Ł, Stasiak A, Ciszewski WM. Whole grain metabolite 3,5-dihydroxybenzoic acid is a beneficial nutritional molecule with the feature of a double-edged sword in human health: a critical review and dietary considerations. Crit Rev Food Sci Nutr 2023; 64:8786-8804. [PMID: 37096487 DOI: 10.1080/10408398.2023.2203762] [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] [Indexed: 04/26/2023]
Abstract
Nonprocessed foodstuffs of plant origin, especially whole-grain cereals, are considered to be health-promoting components of the human diet. While most of their well-studied effects derive from their high fiber content and low glycemic index, the presence of underrated phenolic phytonutrients has recently been brought to the attention of nutritionists. In this review, we report and discuss findings on the sources and bioactivities of 3,5-dihydroxybenzoic acid (3,5-DHBA), which is both a direct dietary component (found, e.g., in apples) and, more importantly, a crucial metabolite of whole-grain cereal-derived alkylresorcinols (ARs). 3,5-DHBA is a recently described exogenous agonist of the HCAR1/GPR81 receptor. We concentrate on the HCAR1-mediated effects of 3,5-DHBA in the nervous system, on the maintenance of cell stemness, regulation of carcinogenesis, and response to anticancer therapy. Unexpectedly, malignant tumors take advantage of HCAR1 expression to sense 3,5-DHBA to support their growth. Thus, there is an urgent need to fully identify the role of whole-grain-derived 3,5-DHBA during anticancer therapy and its contribution in the regulation of vital organs of the body via its specific HCAR1 receptor. We discuss here in detail the possible consequences of the modulatory capabilities of 3,5-DHBA in physiological and pathological conditions in humans.
Collapse
Affiliation(s)
- Waldemar Wagner
- Laboratory of Cellular Immunology, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
| | | | - Łukasz Pułaski
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
- Laboratory of Transcriptional Regulation, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
| | - Anna Stasiak
- Department of Hormone Biochemistry, Medical University of Lodz, Lodz, Poland
| | - Wojciech M Ciszewski
- Department of Molecular Cell Mechanisms, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
47
|
Brackx W, de Cássia Collaço R, Theys M, Cruyssen JV, Bosmans F. Understanding the physiological role of Na V1.9: Challenges and opportunities for pain modulation. Pharmacol Ther 2023; 245:108416. [PMID: 37061202 DOI: 10.1016/j.pharmthera.2023.108416] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/31/2023] [Accepted: 04/12/2023] [Indexed: 04/17/2023]
Abstract
Voltage-activated Na+ (NaV) channels are crucial contributors to rapid electrical signaling in the human body. As such, they are among the most targeted membrane proteins by clinical therapeutics and natural toxins. Several of the nine mammalian NaV channel subtypes play a documented role in pain or other sensory processes such as itch, touch, and smell. While causal relationships between these subtypes and biological function have been extensively described, the physiological role of NaV1.9 is less understood. Yet, mutations in NaV1.9 can cause striking disease phenotypes related to sensory perception such as loss or gain of pain and chronic itch. Here, we explore our current knowledge of the mechanisms by which NaV1.9 may contribute to pain and elaborate on the challenges associated with establishing links between experimental conditions and human disease. This review also discusses the lack of comprehensive insights into NaV1.9-specific pharmacology, an unfortunate situation since modulatory compounds may have tremendous potential in the clinic to treat pain or as precision tools to examine the extent of NaV1.9 participation in sensory perception processes.
Collapse
Affiliation(s)
- Wayra Brackx
- Molecular Physiology and Neurophysics Group, Department of Basic and Applied Medical Sciences, University of Ghent, Ghent, Belgium
| | - Rita de Cássia Collaço
- Molecular Physiology and Neurophysics Group, Department of Basic and Applied Medical Sciences, University of Ghent, Ghent, Belgium
| | - Margaux Theys
- Molecular Physiology and Neurophysics Group, Department of Basic and Applied Medical Sciences, University of Ghent, Ghent, Belgium
| | - Jolien Vander Cruyssen
- Molecular Physiology and Neurophysics Group, Department of Basic and Applied Medical Sciences, University of Ghent, Ghent, Belgium
| | - Frank Bosmans
- Molecular Physiology and Neurophysics Group, Department of Basic and Applied Medical Sciences, University of Ghent, Ghent, Belgium.
| |
Collapse
|
48
|
Two-step structural changes in M3 muscarinic receptor activation rely on the coupled G q protein cycle. Nat Commun 2023; 14:1276. [PMID: 36882424 PMCID: PMC9992711 DOI: 10.1038/s41467-023-36911-4] [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: 07/30/2022] [Accepted: 02/23/2023] [Indexed: 03/09/2023] Open
Abstract
G protein-coupled receptors (GPCRs) regulate diverse intracellular signaling pathways through the activation of heterotrimeric G proteins. However, the effects of the sequential activation-deactivation cycle of G protein on the conformational changes of GPCRs remains unknown. By developing a Förster resonance energy transfer (FRET) tool for human M3 muscarinic receptor (hM3R), we find that a single-receptor FRET probe can display the consecutive structural conversion of a receptor by G protein cycle. Our results reveal that the G protein activation evokes a two-step change in the hM3R structure, including the fast step mediated by Gq protein binding and the subsequent slower step mediated by the physical separation of the Gαq and Gβγ subunits. We also find that the separated Gαq-GTP forms a stable complex with the ligand-activated hM3R and phospholipase Cβ. In sum, the present study uncovers the real-time conformational dynamics of innate hM3R during the downstream Gq protein cycle.
Collapse
|
49
|
Hua Y, Liu Y, Li L, Liu G. Activation of hypermethylated P2RY1 mitigates gastric cancer by promoting apoptosis and inhibiting proliferation. Open Life Sci 2023; 18:20220078. [PMID: 36879646 PMCID: PMC9985447 DOI: 10.1515/biol-2022-0078] [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: 02/23/2022] [Revised: 04/12/2022] [Accepted: 04/20/2022] [Indexed: 03/06/2023] Open
Abstract
The P2RY1 receptor is known to cause cancer by activating the ERK signal pathway, and its DNA methylation status and corresponding regulatory mechanism remain unknown. This study used the DNA methylation chip to profile the genome-wide DNA methylation level in gastric cancer tissues. The proliferation and apoptosis of the SGC7901 gastric cancer cell line were determined after treatment with a selective P2RY1 receptor agonist, MRS2365. The promoter region of P2RY1 was found to be highly methylated with four hypermethylated sites (|Δβ value| > 0.2) in diffuse gastric cancer and was validated by bioinformatics analysis in the TCGA database. Also, immunohistochemical staining data obtained from the HPA database demonstrated the downregulated expression of proteins encoded by P2RY1 in stomach cancer tissue. The analysis of MRS2365-treated cells by annexin V/propidium iodide staining and caspase-3 activity assays indicated the induction of apoptosis in SGC7901 cells. The P2RY1 receptor activation in human SGC7901 gastric cancer cells via the MRS2365 agonist induced apoptosis and reduced cell growth. High DNA methylation in the promoter region of P2RY1 might have contributed to the reduced expression of P2RY1's mRNA, which was likely responsible for the "aggressive" nature of the diffuse gastric cancer.
Collapse
Affiliation(s)
- Yinggang Hua
- Department of Basic Medicine, Medical College of Xiamen University, Xiamen, Fujian, China
| | - Yanling Liu
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Long Li
- Department of Basic Medicine, Medical College of Xiamen University, Xiamen, Fujian, China
| | - Guoyan Liu
- Department of Gastrointestinal Surgery, Zhongshan Hospital Xiamen University, Xiamen, China
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
- Department of Basic Medicine, Medical College of Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
50
|
Zhao J, DiGiacomo V, Ferreras-Gutierrez M, Dastjerdi S, de Opakua AI, Park JC, Luebbers A, Chen Q, Beeler A, Blanco FJ, Garcia-Marcos M. Small-molecule targeting of GPCR-independent non-canonical G protein signaling inhibits cancer progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.18.529092. [PMID: 36824907 PMCID: PMC9949157 DOI: 10.1101/2023.02.18.529092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
Activation of heterotrimeric G-proteins (Gαβγ) by G-protein-coupled receptors (GPCRs) is a quintessential mechanism of cell signaling widely targeted by clinically-approved drugs. However, it has become evident that heterotrimeric G-proteins can also be activated via GPCR-independent mechanisms that remain untapped as pharmacological targets. GIV/Girdin has emerged as a prototypical non-GPCR activator of G proteins that promotes cancer metastasis. Here, we introduce IGGi-11, a first-in-class smallmolecule inhibitor of non-canonical activation of heterotrimeric G-protein signaling. IGGi-11 binding to G-protein α-subunits (Gαi) specifically disrupted their engagement with GIV/Girdin, thereby blocking non-canonical G-protein signaling in tumor cells, and inhibiting pro-invasive traits of metastatic cancer cells in vitro and in mice. In contrast, IGGi-11 did not interfere with canonical G-protein signaling mechanisms triggered by GPCRs. By revealing that small molecules can selectively disable non-canonical mechanisms of G-protein activation dysregulated in disease, these findings warrant the exploration of therapeutic modalities in G-protein signaling that go beyond targeting GPCRs.
Collapse
|