1
|
Huang M, Li W, Sun Y, Dong J, Li C, Jia H, Jiao Y, Wang L, Zhang S, Wang F, Chen J. Janus piezoelectric adhesives regulate macrophage TRPV1/Ca 2+/cAMP axis to stimulate tendon-to-bone healing by multi-omics analysis. Bioact Mater 2025; 50:134-151. [PMID: 40242507 PMCID: PMC12002942 DOI: 10.1016/j.bioactmat.2025.03.029] [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: 02/07/2025] [Revised: 03/14/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
Piezoelectric stimulation has garnered substantial interest as a promising strategy for tissue regeneration. However, studies investigating its impact on tendon-to-bone healing characterized by fibrocartilage remain scarce. Moreover, there are considerable technical challenges in achieving minimally invasive application of piezoelectric stimulation on the irregular tendon-to-bone interface. Herein, we developed Janus asymmetric piezoelectric adhesives by assembling adhesive hydrogel (GAN) and non-adhesive hydrogel (GM) on each side of piezoelectric poly (L-lactic acid) nanofiber. Piezoelectric adhesives exhibited superior anti-inflammatory effects both in vitro and ex vivo. Notably, the transient receptor potential (TRP) ion channels, a class of versatile signaling molecules, are closely associated with the regulation of inflammation. This study demonstrated that piezoelectric stimulation promoted Ca2+ influx through the activation of transient receptor potential vanilloid 1 (TRPV1), further enhancing cAMP signaling pathway in macrophages by RNA sequencing. Additionally, in vivo proteomic analysis revealed Arachidonic acid metabolism and TNF-α signaling pathway downregulation and VEGF signaling pathway upregulation in a rat rotator cuff repair model. Piezoelectric adhesives ultimately achieved inflammation alleviation, angiogenesis enhancement, and fibrocartilage regeneration promotion, improving the biomechanical strength of the enthesis. This study elucidated the mechanism by which piezoelectric stimulation regulated tendon-to-bone healing through multi-omics analysis. The piezoelectric adhesives hold promise as a convenient and effective strategy for enhancing tendon-to-bone healing in clinical practice.
Collapse
Affiliation(s)
- Moran Huang
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Wan Li
- Key Laboratory of Textile Science & Technology, Ministry of Education, Donghua University, Shanghai, 201620, China
| | - Yaying Sun
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Jize Dong
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Chaojing Li
- Key Laboratory of Textile Science & Technology, Ministry of Education, Donghua University, Shanghai, 201620, China
| | - Henjie Jia
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yongjie Jiao
- Key Laboratory of Textile Science & Technology, Ministry of Education, Donghua University, Shanghai, 201620, China
| | - Lu Wang
- Key Laboratory of Textile Science & Technology, Ministry of Education, Donghua University, Shanghai, 201620, China
| | - Shanxing Zhang
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Fujun Wang
- Key Laboratory of Textile Science & Technology, Ministry of Education, Donghua University, Shanghai, 201620, China
- Shanghai Frontiers Science Center of Advanced Textiles, College of Textiles, Donghua University, Shanghai 201620, China
| | - Jiwu Chen
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| |
Collapse
|
2
|
Bhattacharyya A, Vasconcelos D, Spicarova D, Palecek J. 20:4-NAPE induced changes of mechanical sensitivity and DRG neurons excitability are concentration dependent and mediated via NAPE-PLD. Sci Rep 2025; 15:14131. [PMID: 40269193 DOI: 10.1038/s41598-025-98567-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 04/14/2025] [Indexed: 04/25/2025] Open
Abstract
Alterations in the excitability of dorsal root ganglion (DRG) neurons are critical in the pathogenesis of acute and chronic pain. Neurotransmitter release from the terminals of DRG neurons is regulated by cannabinoid receptor 1 (CB1) and transient receptor potential vanilloid 1 (TRPV1), both activated by anandamide (AEA). In our experiments, the AEA precursor N-arachidonoylphosphatidylethanolamine (20:4-NAPE) was used to study the modulation of nociceptive DRG neurons excitability using K+-evoked Ca2+ transients. Intrathecal administration was used to evaluate in vivo effects. Application of 20:4-NAPE at lower concentrations (10 nM - 1 µM) decreased the excitability of DRG neurons, whereas the higher (10 µM) increased it. Both effects of 20:4-NAPE were blocked by the N-acylphosphatidylethanolamine phospholipase D (NAPE-PLD) inhibitor LEI-401. Similarly, lower concentrations of externally applied AEA (1 nM - 10 nM) inhibited DRG neurons, whereas higher concentration (100 nM) did not change it. High AEA concentration (10 µM) evoked Ca2+ transients dependent on TRPV1 activation in separate experiments. Inhibition of the CB1 receptor by PF514273 (400 nM) prevented the 20:4-NAPE- and AEA-induced inhibition, whereas TRPV1 inhibition by SB366791 (1 µM) prevented the increased DRG neuron excitability. In behavioral tests, lower 20:4-NAPE concentration caused hyposensitivity, while higher evoked mechanical allodynia. Intrathecal LEI-401 prevented both in vivo effects of 20:4-NAPE. These results highlight anti- and pro-nociceptive effects of 20:4-NAPE mediated by CB1 and TRPV1 in concentration-dependent manner. Our study underscores the complexity of endocannabinoid signaling in pain transmission modulation and highlights 20:4-NAPE as a potential therapeutic target, offering new insights for developing analgesic strategies.
Collapse
Affiliation(s)
- Anirban Bhattacharyya
- Laboratory of Pain Research, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Daniel Vasconcelos
- Laboratory of Pain Research, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Diana Spicarova
- Laboratory of Pain Research, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jiri Palecek
- Laboratory of Pain Research, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
3
|
Broad LM, Suico JG, Turner PK, Nie S, Johnson KW, Sanger HE, Wegiel LA, Sperry DC, Remick D, Moran M, Malekiani S, Del Camino D, Wu X, Chong JA, Blair NT, Wilke AV. Preclinical and clinical evaluation of a novel TRPA1 antagonist LY3526318. Pain 2025:00006396-990000000-00878. [PMID: 40258136 DOI: 10.1097/j.pain.0000000000003570] [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: 07/22/2024] [Accepted: 12/11/2024] [Indexed: 04/23/2025]
Abstract
ABSTRACT The transient receptor potential cation channel member A1 (TRPA1) is heavily implicated in nociceptive signaling in both physiological and pathological pain states. However, it has been challenging to develop TRPA1 antagonists with appropriate properties to advance into clinical development. Herein, we describe the preclinical characterization and early clinical development of LY3526318, a potent, selective, and orally bioavailable TRPA1 antagonist. In vitro studies showed that LY3526318 reversibly inhibited recombinant TRPA1 channels with nanomolar potency that was conserved across species. LY3526318 also inhibited the function of native human and rat TRPA1 channels, including nociceptive dorsal root ganglion neuronal TRPA1 channels. In vivo studies showed that LY3526318 blocked formalin-evoked flinching behaviors and chronic Freund adjuvant-induced cold hypersensitivity in rats. Only male rats were used in these studies. Initial phase 1, single- and multiple-ascending dose studies evaluating pharmacokinetic and safety parameters of LY3526318 revealed a suboptimal pharmacokinetic profile leading to the development and study of a spray-dried dispersion (SDD) formulation of LY3526318. When dosed once daily at 250 mg, LY3526318-SDD showed a tmax of 4 hours and t1/2 of 12 hours, maintaining plasma exposures demonstrated to engage the TRPA1 target. Adverse events were transient and mild across all phase 1 studies. In summary, LY3526318 blocked TRPA1 in vitro and in vivo, inhibited behavioral signs of enhanced nociception in animal models, and was safe and well tolerated in phase 1 clinical studies, with LY3526318-SDD displaying an appropriate pharmacokinetic profile to advance to proof-of-concept studies in patients with chronic pain.
Collapse
Affiliation(s)
- Lisa M Broad
- Eli Lilly and Company, Bracknell, United Kingdom
| | | | | | - Si Nie
- Eli Lilly and Company, Indianapolis, IN, United States
| | | | | | | | | | - David Remick
- Eli Lilly and Company, Indianapolis, IN, United States
| | | | | | | | - Xinyuan Wu
- Hydra Biosciences, Cambridge, MA, United States
| | | | | | | |
Collapse
|
4
|
Ren C, Xu Q, Luo Q, Qiao X, Ding T, Wang W, Zeng X, Chen C, Xiao Y, Hong X. Benzothiazole amide analogues as antagonists of TRPC 6 channels: A therapeutic approach for kidney fibrosis. Eur J Med Chem 2025; 291:117628. [PMID: 40267878 DOI: 10.1016/j.ejmech.2025.117628] [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: 02/06/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 04/25/2025]
Abstract
Transient receptor potential canonical 6 (TRPC6) channels, which function as receptor-operated, non-selective cation channels, are widely expressed in the kidney, lungs, and brain. Within these organs, they play crucial roles in regulating diverse physiological processes and contribute to the pathogenesis of various disorders. The resolution of the cryo-electron microscopy structure of TRPC6 has significantly advanced our understanding of its molecular mechanisms, thereby providing a robust platform for structure-based drug design. Building upon compound 1S as a lead, we developed and synthesized a series of benzothiazole derivatives, ultimately identifying compound X26 as a potent TRPC6 antagonist with an IC50 of 0.97 μM. In vitro administration of X26 significantly suppressed TGF-β1-induced myofibroblast differentiation in HK-2 cells, as evidenced by a reduced expression of α-SMA, collagen I, and fibronectin. Furthermore, in a unilateral ureteral obstruction (UUO)-induced kidney fibrosis mouse model, treatment with X26 resulted in a substantial reduction in serum urea nitrogen, serum creatinine, and urinary protein levels, as well as a decrease in renal collagen deposition. These findings establish X26 as a promising lead for the development of TRPC6 antagonists and therapeutic interventions for kidney fibrosis.
Collapse
Affiliation(s)
- Chunlin Ren
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
| | - Qiding Xu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
| | - Qiusi Luo
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
| | - Xue Qiao
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
| | - Taotao Ding
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
| | - Wumei Wang
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China; Shenzhen Institute of Wuhan University, Shenzhen, 518057, China
| | - Xiaodong Zeng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China.
| | - Cheng Chen
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Yuling Xiao
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China.
| | - Xuechuan Hong
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China; Shenzhen Institute of Wuhan University, Shenzhen, 518057, China.
| |
Collapse
|
5
|
Zeng Z, Chen E, Xue J. Emerging roles of mechanically activated ion channels in autoimmune disease. Autoimmun Rev 2025; 24:103813. [PMID: 40194731 DOI: 10.1016/j.autrev.2025.103813] [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: 01/21/2025] [Revised: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 04/09/2025]
Abstract
Mechanically activated (MA) ion channels have rapidly gained prominence as vital conduits bridging aberrant mechanical cues in tissues with the dysregulated immune responses at the core of autoimmune diseases. Once regarded as peripheral players in inflammation, these channels, exemplified by PIEZO1, TRPV4, and specific K2P family members, now play a central role in modulating T-cell effector functions, B- cell activation and the activity of macrophages and dendritic cells. Their gating is intimately tied to physical distortions such as increased tissue stiffness, osmotic imbalances, or fluid shear, triggering a cascade of ionic fluxes that elevate proinflammatory signaling and drive tissue-destructive loops. Recognition of these channels as central mediators of mechanical stress-induced inflammation responses in autoimmune pathogenesis is rapidly expanding. In parallel, the emerging therapeutic strategies aim to restrain overactive mechanosensors or selectively harness them in affected tissues. Small molecules, peptide blockers, and gene-targeting approaches show preclinical promise, although off-target effects and the broader homeostatic roles of these channels warrant caution. This review explores how integrating mechanobiological concepts with established immunological paradigms enables a more detailed understanding of autoimmune pathogenesis. By elucidating how mechanical forces potentiate or dampen pathological immunity, we propose innovative strategies that exploit mechanosensitivity to recalibrate immune responses across a spectrum of autoimmune conditions.
Collapse
Affiliation(s)
- Zhiru Zeng
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Engeng Chen
- Department of Zhejiang Provincial Key Laboratory of Biotherapy, Sir Run Run Shaw Hospital of Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Jing Xue
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China.
| |
Collapse
|
6
|
Jin Z, Peng Y, Zhang H, He X, Zhang Y, Pan X, Li M, Yang Q. Inhibition of TRPM3 by Primidone Provides a Potential Therapeutic Method for Adenomyosis Management. Drug Des Devel Ther 2025; 19:2533-2549. [PMID: 40190806 PMCID: PMC11972583 DOI: 10.2147/dddt.s494981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 03/19/2025] [Indexed: 04/09/2025] Open
Abstract
Purpose To test the expression profile of transient receptor potential channels (TRPs) in adenomyosis patients and evaluate the effects of primidone on tamoxifen-induced adenomyosis mice. Patients and Methods This study included in vivo animal model and human tissue samples. Eutopic endometrium from adenomyosis patients (n=20) was collected and subjected to mRNA analysis of TRP channels. TRPA1, TRPV1 and TRPM3 in adenomyosis patients (n=50) and tamoxifen-induced adenomyosis mice (n=6) were examined by immunohistochemistry. From 10 weeks after birth, primidone (2 mg/kg/d) and atosiban (1 mg/kg/d) were given separately to adenomyotic mice by intraperitoneal injection for 3 weeks. The hotplate test was conducted once a week beginning at 10 weeks, and then uterine samples were harvested for HE staining and RNA-seq at 13 weeks. Results The mRNA expression of 15 TRPs was significantly increased in the proliferative phase of the adenomyotic endometrium. TRPV1, TRPM3 or TRPA1 staining levels were positively correlated with dysmenorrhea severity, menses amount and uterine size. In tamoxifen-induced adenomyosis mice, primidone had a significant effect on both the depth of myometrial infiltration and analgesia. Forty-seven DEGSSs were identifieSd after primidone treatment, and bioinformatics analysis predicted that they were enriched in the cell cycle and cell division. Conclusion The expression profile of TRP channels varies significantly in adenomyosis patients, and primidone may provide a potential therapeutic method for adenomyosis management.
Collapse
Affiliation(s)
- Zhixing Jin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215123, People’s Republic of China
| | - Yaoming Peng
- Shanghai Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, People’s Republic of China
| | - He Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215123, People’s Republic of China
| | - Xiaoping He
- Shanghai Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, People’s Republic of China
| | - Yi Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215123, People’s Republic of China
| | - Xin Pan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215123, People’s Republic of China
| | - Min Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215123, People’s Republic of China
| | - Qianqian Yang
- Department of Pathology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215123, People’s Republic of China
| |
Collapse
|
7
|
Zhang L, Iannotti FA, R. Saber F, K. Arafa R, Schiano Moriello A, A. Rasle R, Soria‐Lopez A, G. Abd EL‐Gawwad S, Rocchetti G, Otero P, Kulinowski Ł, Skalicka‐Woźniak K, Lucini L, Simal‐Gandara J. The Phenolic Signature of Psidium cattleianum Fruits and Leaves Modulates TRPV1 and TRPA1 Transient Receptor Potential Channels: A Metabolomics, In Vitro, and In Silico Study. Food Sci Nutr 2025; 13:e70075. [PMID: 40129993 PMCID: PMC11931593 DOI: 10.1002/fsn3.70075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/27/2025] [Accepted: 02/09/2025] [Indexed: 03/26/2025] Open
Abstract
Although Psidium cattleianum (strawberry guava, Myrtaceae) is known for its anti-inflammatory, antioxidant, antimicrobial, and antidiabetic properties, its phytochemical profile and associated bioactivities remain largely underexplored. This study employed UHPLC-QTOF-HRMS for untargeted phenolic profiling of leaf and fruit extracts from P. cattleianum, followed by semi-quantification of phenolic subclasses and multivariate data analysis. Four hundred sixty-nine metabolites, including various phenolic subclasses-predominantly flavonoids and phenolic acids were- identified and annotated. Using HEK-293 cells stably transfected with TRPA1 or TRPV1 cation channels, it was found that both leaf and fruit extracts activate and rapidly desensitize TRPA1 in a concentration-dependent manner (EC50 18 and 30 μg/mL; IC50 60 and 47 μg/mL, respectively). Additionally, molecular docking analysis provided deeper insights into the interactions between P. cattleianum phytochemicals and the TRPA1 cation channel, identifying theaflavin 3,3'-O-digallate as the phenolic compound with the highest affinity (S score of -9.27 Kcal/mol). Interestingly, except for theaflavin 3,3'-O-digallate, compounds enriched in the leaf extract exhibited weaker binding interactions and lower S scores (approximately -7 Kcal/mol) compared to those enriched in the fruit extract. Also, a 100 ns molecular dynamics study of theaflavin 3,3'-O-digallate with TRAP1 demonstrated high binding stability of the complex. Overall, this study offers valuable insights into the phytochemical characteristics of P. cattleianum extracts and reveals their mechanism of action through affinity for the TRPA1 cation channel-receptors.
Collapse
Affiliation(s)
- Leilei Zhang
- Department for Sustainable Food ProcessUniversità Cattolica del Sacro CuorePiacenzaItaly
| | - Fabio Arturo Iannotti
- Institute of Biomolecular Chemistry (ICB); National Research Council (CNR)PozzuoliItaly
| | - Fatema R. Saber
- Pharmacognosy Department, Faculty of PharmacyCairo UniversityCairoEgypt
| | - Reem K. Arafa
- Drug Design and Discovery LabHelmy Institute for Medical Sciences, Zewail City of Science and TechnologyGizaEgypt
- Biomedical Sciences ProgramUniversity of Science and Technology, Zewail City of Science and TechnologyGizaEgypt
| | | | - Rasha A. Rasle
- Drug Design and Discovery LabHelmy Institute for Medical Sciences, Zewail City of Science and TechnologyGizaEgypt
- Biomedical Sciences ProgramUniversity of Science and Technology, Zewail City of Science and TechnologyGizaEgypt
| | - Anton Soria‐Lopez
- Department of Physical Chemistry, Faculty of SciencesUniversidade de VigoOurenseSpain
| | - Sara G. Abd EL‐Gawwad
- Drug Design and Discovery LabHelmy Institute for Medical Sciences, Zewail City of Science and TechnologyGizaEgypt
- Biomedical Sciences ProgramUniversity of Science and Technology, Zewail City of Science and TechnologyGizaEgypt
| | - Gabriele Rocchetti
- Department of Animal Science, Food and NutritionUniversità Cattolica del Sacro CuorePiacenzaItaly
| | - Paz Otero
- Analytical Chemistry and Food Science Department, Faculty of ScienceNutrition and Bromatology GroupOurenseSpain
| | - Łukasz Kulinowski
- Department of Natural Products ChemistryMedical University of LublinLublinPoland
| | | | - Luigi Lucini
- Department for Sustainable Food ProcessUniversità Cattolica del Sacro CuorePiacenzaItaly
| | - Jesus Simal‐Gandara
- Analytical Chemistry and Food Science Department, Faculty of ScienceNutrition and Bromatology GroupOurenseSpain
- CISPACFontan Building, City of CultureSantiago de CompostelaSpain
| |
Collapse
|
8
|
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
|
9
|
Göntér K, László S, Tékus V, Dombi Á, Fábián K, Pál S, Pozsgai G, Botz L, Wagner Ö, Pintér E, Hajna Z. New generation capsaicin-diclofenac containing, silicon-based transdermal patch provides prolonged analgesic effect in acute and chronic pain models. Eur J Pharm Sci 2025; 207:107035. [PMID: 39922237 DOI: 10.1016/j.ejps.2025.107035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/30/2025] [Accepted: 02/06/2025] [Indexed: 02/10/2025]
Abstract
OBJECTIVE Pain is one of the major public health burdens worldwide, however, conventional analgesics are often ineffective. Capsaicin-the active compound of Capsicum species, being responsible for their pungency-has been part of traditional medicine long ago. Capsaicin is a natural agonist of the Transient Receptor Potential Vanilloid 1 receptor-localized on capsaicin-sensitive sensory neurons and strongly involved in pain transmission-, and has been in focus of analgesic drug research for many years. In this study, we aimed to develop a sustained release transdermal patch (transdermal therapeutic system, TTS) combining the advantages of low-concentration capsaicin and diclofenac embedded in an innovative structure, as well as to perform complex preclinical investigations of its analgesic effect. METHODS Drug delivery properties of the TTS were investigated with Franz cell and flow-through cell tests. Analgesic effect of the TTS was examined in in vivo models of acute postoperative and inflammatory, chronic neuropathic and osteoarthritic pain. RESULTS Modified silicone polymer matrix-based TTS containing low-concentration capsaicin and diclofenac has been developed, releasing both compounds according to zero-order kinetics. Moreover, capsaicin and diclofenac facilitated the liberation of each other. Combined TTS significantly reduced acute postoperative and inflammatory pain, as well as chronic neuropathic and osteoarthritic pain. Interestingly, in acute postoperative and chronic osteoarthritic pain, capsaicin prolonged and potentiated the pain-relieving effect of diclofenac. CONCLUSIONS New generation combined low-concentration capsaicin-diclofenac containing TTS can be an effective therapeutic tool in acute and chronic pain states involving neuropathic and inflammatory components.
Collapse
Affiliation(s)
- Kitti Göntér
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti str. 12, H-7624, Pécs, Hungary; HUN-REN, Chronic Pain Research Group, University of Pécs, Pécs, Hungary; National Laboratory for Drug Research and Development, Magyar Tudósok Krt. 2, Budapest, 1117, Hungary
| | - Szabolcs László
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti str. 12, H-7624, Pécs, Hungary; Department of Inorganic and Analytical Chemistry, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111, Budapest, Hungary; HUN-REN, Computation-Driven Chemistry Research Group, Műegyetem rkp. 3, H-1111, Budapest, Hungary
| | - Valéria Tékus
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti str. 12, H-7624, Pécs, Hungary
| | - Ágnes Dombi
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Rókus str. 2, H-7624, Pécs, Hungary
| | - Katalin Fábián
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Rókus str. 2, H-7624, Pécs, Hungary
| | - Szilárd Pál
- Institute of Pharmaceutical Technology and Biopharmacy, Faculty of Pharmacy, University of Pécs, Rókus str. 2, H-7624, Pécs, Hungary
| | - Gábor Pozsgai
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Rókus str. 2, H-7624, Pécs, Hungary
| | - Lajos Botz
- Institute of Clinical Pharmacy, Clinical Centre, University of Pécs, Honvéd str. 3, H-7624, Pécs, Hungary
| | - Ödön Wagner
- Department of Inorganic and Analytical Chemistry, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111, Budapest, Hungary
| | - Erika Pintér
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti str. 12, H-7624, Pécs, Hungary; HUN-REN, Chronic Pain Research Group, University of Pécs, Pécs, Hungary; National Laboratory for Drug Research and Development, Magyar Tudósok Krt. 2, Budapest, 1117, Hungary.
| | - Zsófia Hajna
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti str. 12, H-7624, Pécs, Hungary; HUN-REN, Chronic Pain Research Group, University of Pécs, Pécs, Hungary; National Laboratory for Drug Research and Development, Magyar Tudósok Krt. 2, Budapest, 1117, Hungary
| |
Collapse
|
10
|
Touhara KK, Rossen ND, Deng F, Castro J, Harrington AM, Chu T, Garcia-Caraballo S, Brizuela M, O'Donnell T, Xu J, Cil O, Brierley SM, Li Y, Julius D. Topological segregation of stress sensors along the gut crypt-villus axis. Nature 2025; 640:732-742. [PMID: 39939779 DOI: 10.1038/s41586-024-08581-9] [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: 04/16/2024] [Accepted: 12/27/2024] [Indexed: 02/14/2025]
Abstract
The crypt-villus structure of the small intestine serves as an essential protective barrier. The integrity of this barrier is monitored by the complex sensory system of the gut, in which serotonergic enterochromaffin (EC) cells play an important part1,2. These rare sensory epithelial cells surveil the mucosal environment for luminal stimuli and transmit signals both within and outside the gut3-6. However, whether EC cells in crypts and villi detect different stimuli or produce distinct physiological responses is unknown. Here we address these questions by developing a reporter mouse model to quantitatively measure the release and propagation of serotonin from EC cells in live intestines. Crypt EC cells exhibit a tonic low-level mode that activates epithelial serotonin 5-HT4 receptors to modulate basal ion secretion and a stimulus-induced high-level mode that activates 5-HT3 receptors on sensory nerve fibres. Both these modes can be initiated by the irritant receptor TRPA1, which is confined to crypt EC cells. The activation of TRPA1 by luminal irritants is enhanced when the protective mucus layer is compromised. Villus EC cells also signal damage through a distinct mechanism, whereby oxidative stress activates TRPM2 channels, which leads to the release of both serotonin and ATP and consequent excitation of sensory nerve fibres. This topological segregation of EC cell functionality along the mucosal architecture constitutes a mechanism for the surveillance, maintenance and protection of gut integrity under diverse physiological conditions.
Collapse
Affiliation(s)
- Kouki K Touhara
- Department of Physiology, University of California San Franscisco, San Francisco, CA, USA.
| | - Nathan D Rossen
- Department of Physiology, University of California San Franscisco, San Francisco, CA, USA
- Tetrad Graduate Program, University of California San Francisco, San Francisco, CA, USA
| | - Fei Deng
- State Key Laboratory of Membrane Biology, New Cornerstone Science Laboratory, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Joel Castro
- Visceral Pain Research Group, Hopwood Centre for Neurobiology, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Andrea M Harrington
- Visceral Pain Research Group, Hopwood Centre for Neurobiology, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Tifany Chu
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Sonia Garcia-Caraballo
- Visceral Pain Research Group, Hopwood Centre for Neurobiology, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Mariana Brizuela
- Visceral Pain Research Group, Hopwood Centre for Neurobiology, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Tracey O'Donnell
- Visceral Pain Research Group, Hopwood Centre for Neurobiology, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Jinhao Xu
- Department of Physiology, University of California San Franscisco, San Francisco, CA, USA
| | - Onur Cil
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Stuart M Brierley
- Visceral Pain Research Group, Hopwood Centre for Neurobiology, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia.
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia.
| | - Yulong Li
- State Key Laboratory of Membrane Biology, New Cornerstone Science Laboratory, School of Life Sciences, Peking University, Beijing, China.
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China.
| | - David Julius
- Department of Physiology, University of California San Franscisco, San Francisco, CA, USA.
| |
Collapse
|
11
|
Steib A, Rozmer K, Szőke É, Kun J, Farkas N, Feller D, Pongrácz J, Pohóczky K, Helyes Z. The TRPA1 cation channel is upregulated by cigarette smoke in mouse and human macrophages modulating lung inflammation. Sci Rep 2025; 15:10661. [PMID: 40148437 PMCID: PMC11950515 DOI: 10.1038/s41598-025-95662-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 03/24/2025] [Indexed: 03/29/2025] Open
Abstract
Cigarette smoke (CS) is a well-known source of several inflammatory, cytotoxic and genotoxic compounds that cause chronic lung diseases. The transient receptor potential ankyrin 1 (TRPA1), a smoking-responsive, non-selective cation channel, is expressed by both capsaicin-sensitive peptidergic sensory nerves and non-neuronal cells of the lung, but there are few and controversial data on its expression and function on macrophages. Here, we investigated TRPA1 mRNA and protein expression in mouse and human lung tissues and human 3D spheroids, with a particular focus on its expression and potential regulatory effects on pro- and anti-inflammatory macrophage functions in response to CS. TRPA1 was stably expressed in both human and mouse alveolar macrophages, being upregulated after CS exposure and its functional activity was demonstrated in mouse macrophage culture. Moreover, besides CS, the TRPA1 genotype itself affected the expression of M1- (Il-1β, Il-23) and M2-type (Il-10, Tgfβ) macrophage cytokines. Furthermore, CS extract increased TRPA1 mRNA in human lung spheroids showing more prominent expression in macrophage-containing 3D aggregates, while CS extract influenced an elevated TGFβ expression specifically in macrophage-containing spheroids. These results suggest the fine-tuning role of TRPA1 activation in CS-induced airway inflammation, particularly in macrophages, but further studies are needed to draw precise conclusions.
Collapse
Affiliation(s)
- Anita Steib
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- Hungarian Research Network, Chronic Pain Research Group (HUN-REN PTE), Pécs, Hungary
| | - Katalin Rozmer
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- Hungarian Research Network, Chronic Pain Research Group (HUN-REN PTE), Pécs, Hungary
- Institute of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
| | - Éva Szőke
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- Hungarian Research Network, Chronic Pain Research Group (HUN-REN PTE), Pécs, Hungary
- National Laboratory for Drug Research and Development, Budapest, Hungary
| | - József Kun
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- National Laboratory for Drug Research and Development, Budapest, Hungary
- Hungarian Centre for Genomics and Bioinformatics, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Nelli Farkas
- Institute of Bioanalysis, Medical School, University of Pécs, Pécs, Hungary
| | - Diána Feller
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
| | - Judit Pongrácz
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
| | - Krisztina Pohóczky
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary.
- National Laboratory for Drug Research and Development, Budapest, Hungary.
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Pécs, Hungary.
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- Hungarian Research Network, Chronic Pain Research Group (HUN-REN PTE), Pécs, Hungary
- National Laboratory for Drug Research and Development, Budapest, Hungary
- PharmInVivo Ltd., Pécs, Hungary
| |
Collapse
|
12
|
Irwin RS, Madison JM. Unexplained or Refractory Chronic Cough in Adults. N Engl J Med 2025; 392:1203-1214. [PMID: 40138554 DOI: 10.1056/nejmra2309906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Affiliation(s)
- Richard S Irwin
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Massachusetts Chan Medical School, Worcester
| | - J Mark Madison
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Massachusetts Chan Medical School, Worcester
| |
Collapse
|
13
|
Enders JD, Prodoehl EK, Penn SM, Sriram A, Stucky CL. Episodic pain in Fabry disease is mediated by a heat shock protein-TRPA1 axis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.20.639340. [PMID: 40060522 PMCID: PMC11888165 DOI: 10.1101/2025.02.20.639340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
Abstract
Two-thirds of patients with Fabry disease suffer debilitating pain attacks triggered by exercise, fever, and exposure to environmental heat. These patients face an even greater risk of heat-related episodic pain in the face of global climate change. Almost nothing is known about the biological mechanisms underlying heat-induced pain crises in Fabry disease, and there is no preclinical model available to study Fabry crises. Here, we established the first model of heat-induced pain attacks in Fabry disease by exposing transgenic Fabry rats to environmental heat. Heat exposure precipitated robust mechanical hypersensitivity, closely matching temporal features reported by patients with Fabry disease. At the cellular level, heat exposure sensitized Fabry dorsal root ganglia (DRG) neurons to agonists for transient receptor potential cation channel A1 (TRPA1), but not TRPV1. The heat shock response, which normally confers heat-resilience, was impaired in Fabry disease, and we demonstrated that heat shock proteins (HSP70 and HSP90) regulate TRPA1. Strikingly, pharmacologically inhibiting HSP90 completely prevented cellular and behavioral sensitization by environmental heat in Fabry disease. Together, this work establishes the first model of episodic pain in Fabry disease, implicates the heat shock response in heat-evoked pain episodes, and identifies a novel heat shock protein-TRPA1 regulatory axis.
Collapse
Affiliation(s)
- Jonathan D Enders
- Department of Cell Biology, Neurobiology, and Anatomy; Medical College of Wisconsin, Milwaukee, WI
| | - Eve K Prodoehl
- Department of Cell Biology, Neurobiology, and Anatomy; Medical College of Wisconsin, Milwaukee, WI
| | - Signe M Penn
- Department of Cell Biology, Neurobiology, and Anatomy; Medical College of Wisconsin, Milwaukee, WI
| | - Anvitha Sriram
- Department of Cell Biology, Neurobiology, and Anatomy; Medical College of Wisconsin, Milwaukee, WI
| | - Cheryl L Stucky
- Department of Cell Biology, Neurobiology, and Anatomy; Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
14
|
Gong X, Yan Q, Chen L. Transient receptor potential a1b regulates primordial germ cell numbers and sex differentiation in developing zebrafish. JOURNAL OF FISH BIOLOGY 2025; 106:921-931. [PMID: 39587668 DOI: 10.1111/jfb.16005] [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: 06/07/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 11/27/2024]
Abstract
Temperature is a leading environmental factor determining the sex ratio of some animal populations, such as fish, amphibians, and reptiles. However, the underlying mechanism by which temperature affects gender is still poorly understood. Transient receptor potential a1b (Trpa1b) belongs to the ion channel family of transient receptor potentials and exhibits dual thermosensitivity to heat and cold. In this study, we have unveiled a novel function of the trpa1b gene. Zebrafish generated through clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 with Trpa1b-null manifest a male-biased sex ratio. The quantity of primordial germ cells (PGCs) in zebrafish is closely linked to gender determination and gonadal development. Yet the role of the trpa1b gene in zebrafish reproductive development remains unexplored in the literature. Our investigation revealed a significant reduction in PGCs in Trpa1b mutant zebrafish compared to their wild-type counterparts 24-h postfertilization (hpf). Transcriptome sequencing of tissues near the reproductive crest of embryos at 1.25 days postfertilization (dpf) revealed differential changes in PGC-related marker genes and genes related to sperm cell development and differentiation. The relative expression of ddx4 and sycp3 genes was significantly downregulated, whereas amh was significantly upregulated at 20 dpf in trpa1b-/- zebrafish. The results of this study provide valuable insights and references for studying the molecular mechanism of sex determination in zebrafish. Undoubtedly, these results will further enhance our understanding of gender differentiation and gonadal development in fish and other vertebrates.
Collapse
Affiliation(s)
- Xiaoting Gong
- Key Laboratory of Aquacultural Resources and Utilization, Ministry of Education, College of Fisheries and Life Sciences, Shanghai Ocean University, Shanghai, China
| | - Qianqian Yan
- Key Laboratory of Aquacultural Resources and Utilization, Ministry of Education, College of Fisheries and Life Sciences, Shanghai Ocean University, Shanghai, China
| | - Liangbiao Chen
- Key Laboratory of Aquacultural Resources and Utilization, Ministry of Education, College of Fisheries and Life Sciences, Shanghai Ocean University, Shanghai, China
| |
Collapse
|
15
|
Ratnasingham M, Bradding P, Roach KM. The role of TRP channels in lung fibrosis: Mechanisms and therapeutic potential. Int J Biochem Cell Biol 2025; 180:106728. [PMID: 39672503 DOI: 10.1016/j.biocel.2024.106728] [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: 09/26/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 12/15/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a severe lung disease affecting around 5 million people globally, with a median survival of 3-4 years. Characterized by excessive scarring of lung tissue, IPF results from the accumulation of myofibroblasts that deposit extracellular matrix (ECM), causing fibrosis. Current treatments, pirfenidone and nintedanib, slow the disease but do not stop its progression. IPF pathogenesis involves repeated alveolar injury, leading to pro-fibrotic mediators like TGFβ1, which trigger fibroblast-to-myofibroblast transitions and ECM deposition. Recent research suggests that transient receptor potential (TRP) channels, such as TRPV4, TRPC6, and TRPA1, play a key role in regulating calcium signalling and mechanical stress, crucial in myofibroblast activation. Targeting TRP channels may disrupt fibrosis and offer new therapeutic strategies. Preclinical studies indicate that inhibiting TRP channels could reduce fibrosis, warranting further trials to explore their efficacy and safety in treating IPF and related fibrotic conditions.
Collapse
Affiliation(s)
- M Ratnasingham
- NIHR Respiratory BRC, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - P Bradding
- NIHR Respiratory BRC, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - K M Roach
- NIHR Respiratory BRC, Department of Respiratory Sciences, University of Leicester, Leicester, UK.
| |
Collapse
|
16
|
Marynissen H, Pinto S, Van Ranst N, Van Cutsem E, Voets T, de Hoon J. Increased TRPA1 functionality in the skin of rats and cancer patients following oxaliplatin treatment. THE JOURNAL OF PAIN 2025; 28:104794. [PMID: 39892485 DOI: 10.1016/j.jpain.2025.104794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 01/06/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
Chemotherapy-induced peripheral neuropathy is a debilitating pathology affecting a majority of patients who are being treated with specific cytostatic compounds including oxaliplatin. Various in vitro, ex vivo and in vivo preclinical experiments indicate that transient receptor potential ankyrin 1 (TRPA1) plays a crucial role in the symptomatology of chemotherapy-induced peripheral neuropathy. However, it is unclear whether oxaliplatin also modulates the TRPA1 functionality in the skin of rodents or patients. Here, we quantified the vasodilation after topical application of the TRPA1 agonist cinnamaldehyde in a rodent model of chemotherapy-induced peripheral neuropathy (male Sprague Dawley rats, aged 6 weeks) as well as on fingers of patients suffering from chronic chemotherapy-induced peripheral neuropathy after oxaliplatin treatment. Compared to vehicle-treated rats, a cumulative dose of oxaliplatin 32 mg/kg enhanced the vasodilation after cinnamaldehyde application on rat abdominal skin. Likewise, also in patients with chronic chemotherapy-induced peripheral neuropathy after oxaliplatin, the response to cinnamaldehyde was significantly higher compared to sex- and age-matched healthy controls. Thereby, this study is the first to translate the evidence of increased TRPA1 functionality in vitro or ex vivo in rodents to in vivo conditions in human. The increased TRPA1 functionality in patients with chronic chemotherapy-induced peripheral neuropathy does not only confirm the potential of TRPA1 as target to hit to provide efficacious analgesia, it also paves the way for additional patient stratification on a molecular level and possible treatment response prediction. PERSPECTIVE: The cinnamaldehyde-induced, TRPA1-mediated vasodilation was enhanced in patients with oxaliplatin-induced peripheral neuropathy versus healthy controls, confirming the potential of TRPA1 as target-to-hit for this indication.
Collapse
Affiliation(s)
- Heleen Marynissen
- Center for Clinical Pharmacology, University Hospitals Leuven/KU Leuven, Herestraat 49, 3000 Leuven, Belgium.
| | - Sílvia Pinto
- Laboratory of Ion Channel Research, VIB-KU Leuven Center for Brain and Disease Research, Herestraat 49, 3000 Leuven, Belgium; Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Nele Van Ranst
- Laboratory of Ion Channel Research, VIB-KU Leuven Center for Brain and Disease Research, Herestraat 49, 3000 Leuven, Belgium; Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Eric Van Cutsem
- Digestieve Oncologie, UZ Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Thomas Voets
- Laboratory of Ion Channel Research, VIB-KU Leuven Center for Brain and Disease Research, Herestraat 49, 3000 Leuven, Belgium; Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Jan de Hoon
- Center for Clinical Pharmacology, University Hospitals Leuven/KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| |
Collapse
|
17
|
Zhang F, Mehta H, Choudhary HH, Islam R, Hanafy KA. TRPV4 Channel in Neurological Disease: from Molecular Mechanisms to Therapeutic Potential. Mol Neurobiol 2025; 62:3877-3891. [PMID: 39333347 PMCID: PMC11790740 DOI: 10.1007/s12035-024-04518-5] [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: 05/30/2024] [Accepted: 09/20/2024] [Indexed: 09/29/2024]
Abstract
Transient Receptor Potential Vanilloid 4 (TRPV4) is a non-selective cation channel with pivotal roles in various physiological processes, including osmosensitivity, mechanosensation, neuronal development, vascular tone regulation, and bone homeostasis in human bodies. Recent studies have made significant progress in understanding the structure and functional role of TRPV4, shedding light on its involvement in pathological processes, particularly in the realm of neurological diseases. Here, we aim to provide a comprehensive exploration of the multifaceted contributions of TRPV4 to neurological diseases, spanning its intricate molecular mechanisms to its potential as a target for therapeutic interventions. We delve into the structural and functional attributes of TRPV4, scrutinize its expression profile, and elucidate the possible mechanisms through which it participates in the pathogenesis of neurological disorders. Furthermore, we discussed recent years' progress in therapeutic strategies aimed at harnessing TRPV4 for the treatment of these diseases. These insights will provide a basis for understanding and designing modality-specific pharmacological agents to treat TRPV4-associated disorders.
Collapse
Affiliation(s)
- Feng Zhang
- Cooper Medical School at Rowan University, Camden, NJ, USA
- Cooper University Health Care, Camden, NJ, USA
- Center for Neuroinflammation at Cooper Medical School at Rowan University, Camden, NJ, USA
| | - Hritik Mehta
- Cooper Medical School at Rowan University, Camden, NJ, USA
- Cooper University Health Care, Camden, NJ, USA
- Center for Neuroinflammation at Cooper Medical School at Rowan University, Camden, NJ, USA
| | - Hadi Hasan Choudhary
- Cooper Medical School at Rowan University, Camden, NJ, USA
- Cooper University Health Care, Camden, NJ, USA
- Center for Neuroinflammation at Cooper Medical School at Rowan University, Camden, NJ, USA
| | - Rezwanul Islam
- Cooper Medical School at Rowan University, Camden, NJ, USA
- Cooper University Health Care, Camden, NJ, USA
- Center for Neuroinflammation at Cooper Medical School at Rowan University, Camden, NJ, USA
| | - Khalid A Hanafy
- Cooper Medical School at Rowan University, Camden, NJ, USA.
- Cooper University Health Care, Camden, NJ, USA.
- Center for Neuroinflammation at Cooper Medical School at Rowan University, Camden, NJ, USA.
- Cooper Neurological Institute Center for Neuroinflammation, Cooper Medical School at Rowan University, Camden, NJ, USA.
| |
Collapse
|
18
|
Zhang Y, Yi Y, Shu Y, Ru X, He S. TRP channels and breast cancer: the role of TRPs in the pathophysiological development. Front Mol Biosci 2025; 12:1528663. [PMID: 40078961 PMCID: PMC11896876 DOI: 10.3389/fmolb.2025.1528663] [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: 11/15/2024] [Accepted: 01/27/2025] [Indexed: 03/14/2025] Open
Abstract
TRP channels play important roles in regulating various physiological and pathological processes, including the progression of cancer. Several TRP channels mediate tumour development. This review focuses on the role of TRP channels in the development of breast cancer, including their involvement in proliferation, apoptosis, autophagy, metastasis, and angiogenesis. TRP channels are associated with breast carcinogenesis and their role as potential therapeutic targets and prognostic biomarkers is under investigation. This review summarizes the reported effects of inhibiting or agonizing various TRP channel in breast cancer cells. Although there are relatively mature protocols for the treatment of breast cancer, its treatment is not currently a breakthrough, and therapies targeting TRP channels may be a developable strategy for it.
Collapse
Affiliation(s)
- Yu Zhang
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, School of Medicine, Huzhou University, Huzhou, China
| | - Yanfeng Yi
- Department of Life Sciences and Health, School of Science and Engineering, Huzhou College, Huzhou, China
| | - Yinghao Shu
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, School of Medicine, Huzhou University, Huzhou, China
| | - Xiaochen Ru
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, School of Medicine, Huzhou University, Huzhou, China
| | - Shuaibing He
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, School of Medicine, Huzhou University, Huzhou, China
| |
Collapse
|
19
|
Deng Z, Chen X, Zhang R, Kong L, Fang Y, Guo J, Shen B, Zhang L. Delta opioid peptide [D-ala2, D-leu5]-Enkephalin's ability to enhance mitophagy via TRPV4 to relieve ischemia/reperfusion injury in brain microvascular endothelial cells. Stroke Vasc Neurol 2025; 10:32-44. [PMID: 38697767 PMCID: PMC11877439 DOI: 10.1136/svn-2023-003080] [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] [Accepted: 04/20/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND Local brain tissue can suffer from ischaemia/reperfusion (I/R) injury, which lead to vascular endothelial damage. The peptide δ opioid receptor (δOR) agonist [D-ala2, D-leu5]-Enkephalin (DADLE) can reduce apoptosis caused by acute I/R injury in brain microvascular endothelial cells (BMECs). OBJECTIVE This study aims to explore the mechanism by which DADLE enhances the level of mitophagy in BMECs by upregulating the expression of transient receptor potential vanilloid subtype 4 (TRPV4). METHODS BMECs were extracted and made to undergo oxygen-glucose deprivation/reoxygenation (OGD/R) accompanied by DADLE. RNA-seq analysis revealed that DADLE induced increased TRPV4 expression. The CCK-8 method was used to assess the cellular viability; quantitative PCR (qPCR) was used to determine the mRNA expression of Drp1; western blot was used to determine the expression of TRPV4 and autophagy-related proteins; and calcium imaging was used to detect the calcium influx. Autophagosomes in in the cells' mitochondria were observed by using transmission electron microscopy. ELISA was used to measure ATP content, and a JC-1 fluorescent probe was used to detect mitochondrial membrane potential. RESULTS When compared with the OGD/R group, OGD/R+DADLE group showed significantly enhanced cellular viability; increased expression of TRPV4, Beclin-1, LC3-II/I, PINK1 and Parkin; decreased p62 expression; a marked rise in calcium influx; further increases in mitophagy, an increase in ATP synthesis and an elevation of mitochondrial membrane potential. These protective effects of DADLE can be blocked by a TRPV4 inhibitor HC067047 or RNAi of TRPV4. CONCLUSION DADLE can promote mitophagy in BMECs through TRPV4, improving mitochondrial function and relieving I/R injury.
Collapse
Affiliation(s)
- Zhongfang Deng
- Department of Physiology, Anhui Medical University, Hefei, Anhui, China
| | - Xiaoyu Chen
- Department of Physiology, Anhui Medical University, Hefei, Anhui, China
| | - Ran Zhang
- Department of Physiology, Anhui Medical University, Hefei, Anhui, China
| | - Lingchao Kong
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yang Fang
- Department of Physiology, Anhui Medical University, Hefei, Anhui, China
| | - Jizheng Guo
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui, China
| | - Bing Shen
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao
| | - Lesha Zhang
- Department of Physiology, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
20
|
Krivoshein G, Rivera-Mancilla E, MaassenVanDenBrink A, Giniatullin R, van den Maagdenberg AMJM. Sex difference in TRPM3 channel functioning in nociceptive and vascular systems: an emerging target for migraine therapy in females? J Headache Pain 2025; 26:40. [PMID: 39994546 PMCID: PMC11853570 DOI: 10.1186/s10194-025-01966-9] [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: 12/20/2024] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
Transient Receptor Potential Melastatin 3 (TRPM3) channels are Ca2+ permeable ion channels that act as polymodal sensors of mechanical, thermal, and various chemical stimuli. TRPM3 channels are highly expressed in the trigeminovascular system, including trigeminal neurons and the vasculature. Their presence in dural afferents suggests that they are potential triggers of migraine pain, which is originating from the meningeal area. This area is densely innervated by autonomous and trigeminal nerves that contain the major migraine mediator calcitonin gene-related peptide (CGRP) in peptidergic nerve fibers. Co-expression of TRPM3 channels and CGRP receptors in meningeal nerves suggests a potential interplay between both signalling systems. Compared to other members of the TRP family, TRPM3 channels have a high sensitivity to sex hormones and to the endogenous neurosteroid pregnenolone sulfate (PregS). The predominantly female sex hormones estrogen and progesterone, of which the levels drop during menses, act as natural inhibitors of TRPM3 channels, while PregS is a known endogenous agonist of these channels. A decrease in sex hormone levels has also been suggested as trigger for attacks of menstrually-related migraine. Notably, there is a remarkable sex difference in TRPM3-mediated effects in trigeminal nociceptive signalling and the vasculature. In line with this, the relaxation of human isolated meningeal arteries induced by the activation of TRPM3 channels is greater in females. Additionally, the sex-dependent vasodilatory responses to CGRP in meningeal arteries seem to be influenced by age-related hormonal changes, which could contribute to sex differences in migraine pathology. Consistent with these observations, activation of TRPM3 channels triggers nociceptive sensory firing much more prominently in female than male mouse meninges, suggesting that pain processing in female patients with migraine may differ. Overall, the combined TRPM3-related neuronal and vascular mechanisms could provide a possible explanation for the higher prevalence and even the more severe quality of migraine attacks in females. This narrative review summarizes recent data on the sex-dependent roles of TRPM3 channels in migraine pathophysiology, the potential interplay between TRPM3 and CGRP signalling, and highlights the prospects for translational therapies targeting TRPM3 channels, which may be of particular relevance for women with migraine.
Collapse
Affiliation(s)
- Georgii Krivoshein
- Departments of Human Genetics and Neurology, Leiden University Medical Center, PO Box 9600 2300 RC, Leiden, The Netherlands
| | - Eduardo Rivera-Mancilla
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Antoinette MaassenVanDenBrink
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Rashid Giniatullin
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Arn M J M van den Maagdenberg
- Departments of Human Genetics and Neurology, Leiden University Medical Center, PO Box 9600 2300 RC, Leiden, The Netherlands.
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
21
|
Zhou T, Wang Z, Lv X, Guo M, Zhang N, Liu L, Geng L, Shao J, Zhang K, Gao M, Mao A, Zhu Y, Yu F, Feng L, Wang X, Zhai Q, Chen W, Ma X. Targeting gut S. aureofaciens Tü117 serves as a new potential therapeutic intervention for the prevention and treatment of hypertension. Cell Metab 2025; 37:496-513.e11. [PMID: 39908987 DOI: 10.1016/j.cmet.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 09/13/2024] [Accepted: 01/08/2025] [Indexed: 02/07/2025]
Abstract
Currently, the regulation of specific gut microbial metabolism for the development and/or treatment of hypertension remains largely unexplored. Here, we show that α-lipomycin, produced by Streptomyces aureofaciens (S. aureofaciens) Tü117, is upregulated in the serum of high-salt diet (HSD) mice and patients with essential hypertension. α-lipomycin causes vasodilation impairment involving transient receptor potential vanilloid 4 (TRPV4)-mediated nitric oxide and endothelium-derived hyperpolarizing factor pathways in mice. We also find that Lactobacillus plantarum (L. plantarum) CCFM639 attenuates the increase in blood pressure (BP) potentially through inhibiting the proliferation of S. aureofaciens Tü117 in mice. An exploratory intervention trial indicates that L. plantarum CCFM639 supplementation reduces BPs in subjects newly diagnosed with pre-hypertension or stage 1 hypertension without antihypertensive medication. Our findings provide evidence for a role of S. aureofaciens Tü117-associated α-lipomycin elevation in the pathogenesis of HSD-induced hypertension, highlighting that targeting gut bacteria serves as a new therapeutic intervention for hypertension.
Collapse
Affiliation(s)
- Tingting Zhou
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; Food Nutrition and Human Health Interdisciplinary Center, School of Food Science and Technology, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; Affiliated Hospital of Jiangnan University, Wuxi 214122, China
| | - Zhiwei Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; Food Nutrition and Human Health Interdisciplinary Center, School of Food Science and Technology, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China
| | - Xiaowang Lv
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; Food Nutrition and Human Health Interdisciplinary Center, School of Food Science and Technology, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China
| | - Mengting Guo
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Ning Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Liangju Liu
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China
| | - Li Geng
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China
| | - Jing Shao
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Ka Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China
| | - Mengru Gao
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China
| | - Aiqin Mao
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China
| | - Yifei Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China
| | - Fan Yu
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Lei Feng
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Xiaoyan Wang
- Affiliated Hospital of Jiangnan University, Wuxi 214122, China
| | - Qixiao Zhai
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; Food Nutrition and Human Health Interdisciplinary Center, School of Food Science and Technology, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Wei Chen
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; Food Nutrition and Human Health Interdisciplinary Center, School of Food Science and Technology, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xin Ma
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; Food Nutrition and Human Health Interdisciplinary Center, School of Food Science and Technology, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; Affiliated Hospital of Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
22
|
Dragoni S, Moccia F, Bootman MD. The Roles of Transient Receptor Potential (TRP) Channels Underlying Aberrant Calcium Signaling in Blood-Retinal Barrier Dysfunction. Cold Spring Harb Perspect Biol 2025; 17:a041763. [PMID: 39586624 PMCID: PMC11864113 DOI: 10.1101/cshperspect.a041763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
The inner blood-retinal barrier (iBRB) protects the retinal vasculature from the peripheral circulation. Endothelial cells (ECs) are the core component of the iBRB; their close apposition and linkage via tight junctions limit the passage of fluids, proteins, and cells from the bloodstream to the parenchyma. Dysfunction of the iBRB is a hallmark of many retinal disorders. Vascular endothelial growth factor (VEGF) has been identified as the primary driver leading to a dysfunctional iBRB, thereby becoming the main target for therapy. However, a complete understanding of the molecular mechanisms underlying iBRB dysfunction is elusive and alternative therapeutic targets remain unexplored. Calcium (Ca2+) is a universal intracellular messenger whose homeostasis and dynamics are dysregulated in many pathological disorders. Among the extensive components of the cellular Ca2+-signaling toolkit, cation-selective transient receptor potential (TRP) channels are broadly involved in cell physiology and disease and, therefore, are widely studied as possible targets for therapy. Albeit that TRP channels have been discovered in the photoreceptors of Drosophila and have been studied in the neuroretina, their presence and function in the iBRB have only recently emerged. Within this article, we discuss the structure and functions of the iBRB with a particular focus on Ca2+ signaling in retinal ECs and highlight the potential of TRP channels as new targets for retinal diseases.
Collapse
Affiliation(s)
- Silvia Dragoni
- Institute of Ophthalmology, University College London, London EC1V 9EL, United Kingdom
| | - Francesco Moccia
- Department of Biology and Biotechnology "Lazzaro Spallanzani," University of Pavia, Pavia 27100, Italy
| | - Martin D Bootman
- School of Life, Health and Chemical Sciences, Faculty of Science, Technology, Engineering and Mathematics, The Open University, Milton Keynes MK7 6AA, United Kingdom
| |
Collapse
|
23
|
Fleischmann R, Strauß S, Reuter U. Treating episodic migraine with precision: the evolving landscape of targeted therapies driven by insights in disease biology. Expert Opin Biol Ther 2025:1-15. [PMID: 39831521 DOI: 10.1080/14712598.2025.2456464] [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: 12/13/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 01/22/2025]
Abstract
INTRODUCTION Migraine is a disabling neurological disorder with a complex neurobiology. It appears as a cyclic disorder of sensory processing, affecting multiple systems beyond nociception. Overlapping mechanisms, including dysfunctional processing of sensory input from brain structures are involved in the generation of attacks. AREAS COVERED This review provides a comprehensive synthesis on migraine neurobiology, which was additionally informed by search of research databases (PubMed, ClinicalTrials.gov). Findings from the most recent literature are integrated in a pathophysiological framework. By combining mechanistic insights and clinical trial data, this review highlights the trajectory of precision medicine in migraine treatment, offering a perspective on the near future of targeted and individualized therapeutic strategies. EXPERT OPINION Recent advances in migraine neurobiology offer potential solutions to longstanding challenges. While targeted CGRP therapies have shown promise by addressing specific mechanisms, the pathophysiology of migraine suggests that combination therapies targeting multiple pathways could be beneficial in migraine prevention. The growing diversity of treatment options presents challenges in therapy selection, underscoring the need for predictive biomarkers. These innovations can optimize treatment strategies and improve patient outcomes. As the field progresses, personalized, multimodal approaches are poised to become the standard of care, significantly advancing precision medicine in this area.
Collapse
Affiliation(s)
- Robert Fleischmann
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany
| | - Sebastian Strauß
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany
| | - Uwe Reuter
- University Medicine Greifswald, Greifswald, Germany
- Department of Neurology, Charité - University Medicine Berlin, Berlin, Germany
| |
Collapse
|
24
|
Yadav V, Pandey V, Gaglani P, Srivastava A, Soni, Subhashini. SIRT-2 inhibition by AK-7 orchestrates fibrotic cascades in airways through neuroimmune interaction via TRPA1, TRPM8 and TGF-β signalling. Biochem Pharmacol 2025; 232:116689. [PMID: 39617211 DOI: 10.1016/j.bcp.2024.116689] [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: 08/28/2024] [Revised: 11/04/2024] [Accepted: 11/28/2024] [Indexed: 12/10/2024]
Abstract
Chronic obstructive pulmonary diseases (COPD) is characterized by airflow limitation, chronic inflammation and airway remodeling (AR) in airways and lung parenchyma. AR, a lung response, involves mucus production, airflow issues, and structural changes. It is exacerbated by neurogenic inflammation from activated sensory nerves, highlighting the interplay between neuronal and immune regulation in COPD. Sirtuins play a crucial role in lung remodeling, with SIRT-2 being the least studied. Present study explores how SIRT-2 regulates neurogenic inflammation and fibrosis in experimental BALB/c mice with cigarette smoke-induced COPD. Mice from each group, except the control, were exposed to CS for 60 days and AK-7 (100ug/kg and 200ug/kg) was administered intranasally. The study evaluated lung injury and inflammation marked by increased Cortisol, ACTH, COX-2 and LDH in COPD group with its attenuation by SIRT-2 inhibition. Additionally, CS exposure exhibited neurogenic inflammation represented by activated TPRV1 and TRPM8, elevated neuromediators levels (dopamine, acetylcholine, substance P, serotonin) and their respective receptors which were mitigated by AK-7. CS exposure enhanced fibrosis by targeting the fibrotic cascade, enhancing MMP-9, total collagen, hydroxyproline, and upregulating αSMA, MUC5AC, TGF-β, PKA, GATA-3, FOXO3, and STAT-6. SIRT-2 inhibition effectively reversed all these factors suppressing fibrosis further supported by downregulated SIRT-2 expression and histopathological studies where collagen deposition and mucus production were also attenuated by AK-7. Molecular docking revealed strong binding affinity of certain protein such as COX-2, D5DR and 5HT with AK-7. Overall, targeting SIRT-2 to modulate neuro-immune interplay presents a promising therapeutic approach for addressing AR in COPD.
Collapse
Affiliation(s)
- Vandana Yadav
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi 221005, India
| | - Vinita Pandey
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi 221005, India
| | - Pratikkumar Gaglani
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi 221005, India
| | - Atul Srivastava
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Soni
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi 221005, India
| | - Subhashini
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
25
|
Zong P, Legere N, Feng J, Yue L. TRP Channels in Excitotoxicity. Neuroscientist 2025; 31:80-97. [PMID: 38682490 DOI: 10.1177/10738584241246530] [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: 05/01/2024]
Abstract
Glutamate excitotoxicity is a central mechanism contributing to cellular dysfunction and death in various neurological disorders and diseases, such as stroke, traumatic brain injury, epilepsy, schizophrenia, addiction, mood disorders, Huntington's disease, Alzheimer's disease, Parkinson's disease, multiple sclerosis, pathologic pain, and even normal aging-related changes. This detrimental effect emerges from glutamate binding to glutamate receptors, including α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors, N-methyl-d-aspartate receptors, kainate receptors, and GluD receptors. Thus, excitotoxicity could be prevented by targeting glutamate receptors and their downstream signaling pathways. However, almost all the glutamate receptor antagonists failed to attenuate excitotoxicity in human patients, mainly due to the limited understanding of the underlying mechanisms regulating excitotoxicity. Transient receptor potential (TRP) channels serve as ancient cellular sensors capable of detecting and responding to both external and internal stimuli. The study of human TRP channels has flourished in recent decades since the initial discovery of mammalian TRP in 1995. These channels have been found to play pivotal roles in numerous pathologic conditions, including excitotoxicity. In this review, our focus centers on exploring the intricate interactions between TRP channels and glutamate receptors in excitotoxicity.
Collapse
Affiliation(s)
- Pengyu Zong
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
- Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, CT, USA
| | - Nicholas Legere
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Jianlin Feng
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
| | - Lixia Yue
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
| |
Collapse
|
26
|
Wang X, Wang M, Zhu TT, Zheng ZJ, Li S, Sui ZY, Guo X, Wu S, Zhang NN, Yu ZY, Hu CP, Tang YB, Wang Q, Zhang Z. The TRPM7 chanzyme in smooth muscle cells drives abdominal aortic aneurysm in mice. NATURE CARDIOVASCULAR RESEARCH 2025; 4:216-234. [PMID: 39953275 DOI: 10.1038/s44161-025-00613-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 01/15/2025] [Indexed: 02/17/2025]
Abstract
Ionic signaling in smooth muscle cells (SMCs) is critical for vascular homeostasis. In this study, we untangled the role of the bifunctional TRPM7 channel kinase (chanzyme) in abdominal aortic aneurysm (AAA) pathogenesis. Comparing SMC-specific, macrophage-specific and endothelial cell-specific Trpm7 knockout, we revealed that SMC-specific Trpm7 deficiency protected mice from AAA in two distinct preclinical models of the disease. We showed that the TRPM7 channel activity increased the Ca2+ and Zn2+ influx and the Ca2+/calcineurin/CRTC2/CREB-dependent and Zn2+/MTF1-dependent Mmp2 transcription. Repurposing the clinical drug FTY720 to prevent and treat AAA resulted in improved aortic phenotypes through inhibition of TRPM7 channel activity. This study highlights the ionic mechanisms underlying AAA, identifies TRPM7 as a potential therapeutic target and suggests that blocking TRPM7 channels could be a viable strategy for treating AAA.
Collapse
MESH Headings
- Animals
- TRPM Cation Channels/metabolism
- TRPM Cation Channels/genetics
- TRPM Cation Channels/antagonists & inhibitors
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/metabolism
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Disease Models, Animal
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Matrix Metalloproteinase 2/metabolism
- Matrix Metalloproteinase 2/genetics
- Fingolimod Hydrochloride/pharmacology
- Fingolimod Hydrochloride/therapeutic use
- Aorta, Abdominal/pathology
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/drug effects
- Mice
- Zinc/metabolism
- Mice, Inbred C57BL
- Male
- Protein Serine-Threonine Kinases/metabolism
- Protein Serine-Threonine Kinases/genetics
- Calcium Signaling/drug effects
- Mice, Knockout
- Cells, Cultured
- Humans
- Phenotype
Collapse
Affiliation(s)
- Xuan Wang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Mi Wang
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Hunan Key Laboratory of Cardiometabolic Medicine, Central South University, Changsha, China
| | - Tian-Tian Zhu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Zi-Jie Zheng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Shuang Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Zhao-Yi Sui
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Xin Guo
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Sha Wu
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Nai-Ning Zhang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhi-Yi Yu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chang-Ping Hu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Central South University, Changsha, China
| | - Yong-Bo Tang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Qing Wang
- Department of Interventional Radiology & Vascular Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Zheng Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China.
- Hunan Provincial Key Laboratory of Cardiovascular Research, Central South University, Changsha, China.
| |
Collapse
|
27
|
Zhu Q, Yang J, Shi L, Zhang J, Zhang P, Li J, Song X. Exploring the role of ubiquitination modifications in migraine headaches. Front Immunol 2025; 16:1534389. [PMID: 39958329 PMCID: PMC11825825 DOI: 10.3389/fimmu.2025.1534389] [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: 11/25/2024] [Accepted: 01/09/2025] [Indexed: 02/18/2025] Open
Abstract
Migraine is a complex neurovascular disorder whose pathogenesis involves activation of the trigeminal vascular system, central and peripheral sensitization, and neuroinflammation. Calcitonin gene-related peptide (CGRP) plays a dominant role and activation of MAPK and NF-κB signaling pathways regulates neuropeptide release, glial cell activation, and amplification of nociceptive signals. Aberrant activation of these pathways drives migraine onset and chronicity. The ubiquitin-proteasome system (UPS) is involved in neurological and inflammatory disorders. ubiquitination in the UPS is achieved through a cascade of enzymes, including Ub-activating enzyme (E1), Ub-coupling enzyme (E2), and Ub-ligase (E3). The aim of this review is to systematically explore the role of ubiquitination in the regulation of MAPK and NF-κB signaling pathways, with a focus on the mechanisms of ubiquitinating enzymes in neuroinflammation and pain signal amplification, and to explore their potential as diagnostics, biomarkers, predictors of response to therapy, and monitoring of chronicity in migraine disease.
Collapse
Affiliation(s)
- Qian Zhu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jin Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Lei Shi
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jieying Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Peng Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Junlong Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xiaoli Song
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
28
|
de Deus JL, Maia JM, Soriano RN, Amorim MR, Branco LGS. Psychedelics in neuroinflammation: Mechanisms and therapeutic potential. Prog Neuropsychopharmacol Biol Psychiatry 2025; 137:111278. [PMID: 39892847 DOI: 10.1016/j.pnpbp.2025.111278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 01/03/2025] [Accepted: 01/27/2025] [Indexed: 02/04/2025]
Abstract
Neuroinflammation is a critical factor in the pathogenesis of various neurodegenerative and psychiatric disorders, including Alzheimer's disease, Parkinson's disease, and major depressive disorder. Psychedelics, such as psilocybin, lysergic acid diethylamide (LSD), and dimethyltryptamine (DMT), have demonstrated promising therapeutic effects on neuroinflammation, primarily through interactions with serotonin (5-HT) receptors, particularly the 5-HT2A receptor. Activation of these receptors by psychedelics modulates the production of pro-inflammatory cytokines, regulates microglial activity, and shifts the balance between neurotoxic and neuroprotective metabolites. Additionally, psychedelics affect critical signaling pathways, including the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), phosphatidylinositol-3-kinase/protein kinase B (PI3K/Akt), and mechanistic target of rapamycin (mTOR) pathways, promoting neuroplasticity and exerting anti-inflammatory effects. Beyond the serotonergic system, other neurotransmitter systems-including the glutamatergic, dopaminergic, noradrenergic, gamma-aminobutyric acid (GABAergic), and cholinergic systems-also play significant roles in mediating the effects of psychedelics. This review examines the intricate mechanisms by which psychedelics modulate neuroinflammation and underscores their potential as innovative therapeutic agents for treating neuroinflammatory and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Junia Lara de Deus
- Department of Anesthesiology and Critical Care Medicine, George Washington University, Washington, DC, USA; Department of Oral and Basic Biology Ribeirão Preto, Dental School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Juliana Marino Maia
- Department of Medicine, Federal University of Juiz de Fora, Governador Valadares,MG, Brazil
| | - Renato Nery Soriano
- Division of Physiology and Biophysics, Department of Basic Life Sciences, Federal University of Juiz de Fora, Governador Valadares, MG, Brazil
| | - Mateus R Amorim
- Department of Anesthesiology and Critical Care Medicine, George Washington University, Washington, DC, USA; Program of Physiology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Luiz G S Branco
- Department of Oral and Basic Biology Ribeirão Preto, Dental School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil; Program of Physiology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
29
|
Chen S, Luo Y, Yu P, Yue X, Yang W. [Advances in the development of transient receptor potential melastatin 2 channel inhibitors]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2025; 54:120-130. [PMID: 39909465 PMCID: PMC11956857 DOI: 10.3724/zdxbyxb-2024-0586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 12/20/2024] [Indexed: 02/07/2025]
Abstract
Studies on specific transient receptor potential melastatin 2 (TRPM2) channel inhibitors can deepen our understanding of the pathological mechanism of related diseases, and allow discovery of novel, effective targets and drugs for therapy. The development of TRPM2 channel inhibitors can be broadly classified into four categories with distinct characteristics: reutilization and structural modification of homologous ion channel modulators to produce a diverse array of TRPM2 channel inhibitors with strong inhibitory effects; TRPM2 channel inhibitors based on channel gating mechanism with high specificity; inhibitors identified through high-throughput screening with novel chemical structures; inhibitors developed from natural antioxidants with higher safety. In recent years, the application of computer-aided drug design has significantly accelerated the development of TRPM2 channel inhibitors. Several promising compounds such as ZA18, A1 and D9 have been discovered, and it is expected that more potent and selective TRPM2 channel inhibitor scaffolds will be discovered in the future. This article reviews the advances on the studies of TRPM2 channel inhibitors, aiming to provide insights for further research and clinical application of TRPM2 channel inhibitors.
Collapse
Affiliation(s)
- Shiyao Chen
- Department of Biophysics, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Yanping Luo
- Department of Biophysics, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Peilin Yu
- Department of Toxicology, School of Public Health, Zhejiang University School of Medicine , Hangzhou 310058, China
| | - Xiaomin Yue
- Department of Biophysics, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Wei Yang
- Department of Biophysics, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
30
|
Chandy M, Jimenez-Tellez N, Wu JC. The relationship between cannabis and cardiovascular disease: clearing the haze. Nat Rev Cardiol 2025:10.1038/s41569-025-01121-6. [PMID: 39849111 DOI: 10.1038/s41569-025-01121-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/08/2025] [Indexed: 01/25/2025]
Abstract
Cannabis has been consumed for centuries, but global regulatory changes over the past three decades have increased the availability and consumption of cannabis. Cannabinoids are touted to have therapeutic potential for many diseases and could be a replacement for opioids for analgesia and sedation. However, cannabinoids can cause substantial adverse cardiovascular events that would mitigate any potential benefit. The endocannabinoid system regulates mood, satiety and memory, and modulates the cardiovascular system. The link between cannabinoids and cardiovascular disease, which used to be limited to evidence from preclinical studies, case reports and case series, is now evident in epidemiological studies. Cannabinoids adversely affect the cardiovascular system, causing myocardial infarction, cerebrovascular accidents, arrhythmia and heart failure. The effects of novel cannabinoids are unknown, and synthetic cannabinoids have the potential to cause even more substantial harm than traditional cannabinoids. Therefore, with the increasing availability and use of cannabis, the acute and chronic effects of this drug are becoming apparent.
Collapse
Affiliation(s)
- Mark Chandy
- Stanford Cardiovascular Institute, Stanford, CA, USA.
- Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Medicine, Western University, London, Ontario, Canada.
| | - Nerea Jimenez-Tellez
- Stanford Cardiovascular Institute, Stanford, CA, USA
- Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford, CA, USA.
- Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
31
|
Ma L, Ma C, Wang Z, Wei Y, Li N, Wang J, Li M, Wu Z, Du Y. Unraveling the Synergistic Neuroprotective Mechanism of Natural Drug Candidates Targeting TRPV1 and TRPM8 on an Ischemic Stroke. Anal Chem 2025; 97:1199-1209. [PMID: 39789730 DOI: 10.1021/acs.analchem.4c04442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
The development of multitargeted drugs is urgent for ischemic stroke. TRPV1 and TRPM8 are important targets of ischemic stroke. Previous drug candidate screening has identified that muscone, l-borneol, and ferulic acid may target TRPV1 and TRPM8 for ischemic stroke. However, the mechanisms of these drug candidates on targets were ill-informed. Therefore, firstly, a tongue-tissue biosensor was constructed. It explored the activation or inhibition mechanisms of drug candidates targeting TRPV1 and TRPM8 in a near-physiological environment. It was found that muscone could specifically inhibit TRPM8 and selectively activate TRPV1, while l-borneol exhibited the opposite effect. It suggested a synergistic network between these two drug candidates. Furthermore, more selective protein biosensors were developed to delve deeper into the synergistic mechanisms. A strong synergistic effect of muscone and l-borneol was proved. Molecular docking revealed that the synergistic effect was caused by different action sites, respectively. Subsequently, the synergistic effect of muscone and l-borneol was further confirmed by hypoxic nerve injury models of Caenorhabditis elegans (C. elegans) and antithrombus and anti-ischemic models of zebrafish. Ultimately, through nontargeted metabolomics, it was found that muscone and l-borneol mainly regulated Ca2+ concentration and energy metabolism by pathways such as purine and amino acid metabolisms. In conclusion, this research identified critical targets and synergistic drug candidates for multitarget neuroprotection of ischemic stroke. In addition, it has systemically demonstrated the feasibility of the integration of tissue/protein biosensors and metabolomics for the research and development of multitarget drugs. Compared to other screening and validation methods for drugs and targets, the biosensors we developed not only achieved higher sensitivity and specificity in complex physiological environments, ensuring a wider detection range, but also greatly saved biological samples. Simultaneously, they could be extended to other complex systems, such as biomarker screening in clinical samples and exosomes isolated from stem cells.
Collapse
Affiliation(s)
- Lijuan Ma
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Chaofu Ma
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zijian Wang
- China Beijing Tongrentang Group Co., Ltd., Beijing 100062, China
| | - Yunan Wei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Nan Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jing Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Mingshuang Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zhisheng Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yang Du
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| |
Collapse
|
32
|
Ekundayo B, Arullampalam P, Gerber CE, Hämmerli AF, Guichard S, Boukenna M, Ross-Kaschitza D, Lochner M, Rougier JS, Stahlberg H, Abriel H, Ni D. Identification of a binding site for small molecule inhibitors targeting human TRPM4. Nat Commun 2025; 16:833. [PMID: 39828793 PMCID: PMC11743598 DOI: 10.1038/s41467-025-56131-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
Transient receptor potential (TRP) melastatin 4 (TRPM4) protein is a calcium-activated monovalent cation channel associated with various genetic and cardiovascular disorders. The anthranilic acid derivative NBA is a potent and specific TRPM4 inhibitor, but its binding site in TRPM4 has been unknown, although this information is crucial for drug development targeting TRPM4. We determine three cryo-EM structures of full-length human TRPM4 embedded in native lipid nanodiscs without inhibitor, bound to NBA, and an anthranilic acid derivative, IBA. We found that the small molecules NBA and IBA were bound in a pocket formed between the S3, S4, and TRP helices and the S4-S5 linker of TRPM4. Our structural data and results from patch clamp experiments enable validation of a binding site for small molecule inhibitors, paving the way for further drug development targeting TRPM4.
Collapse
Affiliation(s)
- Babatunde Ekundayo
- Laboratory of Biological Electron Microscopy, IPHYS, SB, EPFL, and Dept. Fundamental Microbiology, Faculty of Biology and Medicine, UNIL, Cubotron, Rt. de la Sorge, Lausanne, Switzerland
| | - Prakash Arullampalam
- Institute of Biochemistry and Molecular Medicine and Swiss National Centre of Competence in Research TransCure, University of Bern, Bern, Switzerland
| | - Christian E Gerber
- Institute of Biochemistry and Molecular Medicine and Swiss National Centre of Competence in Research TransCure, University of Bern, Bern, Switzerland
| | - Anne-Flore Hämmerli
- Institute of Biochemistry and Molecular Medicine and Swiss National Centre of Competence in Research TransCure, University of Bern, Bern, Switzerland
| | - Sabrina Guichard
- Institute of Biochemistry and Molecular Medicine and Swiss National Centre of Competence in Research TransCure, University of Bern, Bern, Switzerland
| | - Mey Boukenna
- Institute of Biochemistry and Molecular Medicine and Swiss National Centre of Competence in Research TransCure, University of Bern, Bern, Switzerland
| | - Daniela Ross-Kaschitza
- Institute of Biochemistry and Molecular Medicine and Swiss National Centre of Competence in Research TransCure, University of Bern, Bern, Switzerland
| | - Martin Lochner
- Institute of Biochemistry and Molecular Medicine and Swiss National Centre of Competence in Research TransCure, University of Bern, Bern, Switzerland
| | - Jean-Sebastien Rougier
- Institute of Biochemistry and Molecular Medicine and Swiss National Centre of Competence in Research TransCure, University of Bern, Bern, Switzerland
| | - Henning Stahlberg
- Laboratory of Biological Electron Microscopy, IPHYS, SB, EPFL, and Dept. Fundamental Microbiology, Faculty of Biology and Medicine, UNIL, Cubotron, Rt. de la Sorge, Lausanne, Switzerland.
| | - Hugues Abriel
- Institute of Biochemistry and Molecular Medicine and Swiss National Centre of Competence in Research TransCure, University of Bern, Bern, Switzerland.
| | - Dongchun Ni
- Laboratory of Biological Electron Microscopy, IPHYS, SB, EPFL, and Dept. Fundamental Microbiology, Faculty of Biology and Medicine, UNIL, Cubotron, Rt. de la Sorge, Lausanne, Switzerland
- International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China
| |
Collapse
|
33
|
Guadarrama E, Vanoye CG, DeCaen PG. Defining the Polycystin Pharmacophore Through HTS & Computational Biophysics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.13.632808. [PMID: 39868095 PMCID: PMC11761769 DOI: 10.1101/2025.01.13.632808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Background and Purpose Polycystins (PKD2, PKD2L1) are voltage-gated and Ca2+-modulated members of the transient receptor potential (TRP) family of ion channels. Loss of PKD2L1 expression results in seizure-susceptibility and autism-like features in mice, whereas variants in PKD2 cause autosomal dominant polycystic kidney disease. Despite decades of evidence clearly linking their dysfunction to human disease and demonstrating their physiological importance in the brain and kidneys, the polycystin pharmacophore remains undefined. Contributing to this knowledge gap is their resistance to drug screening campaigns, which are hindered by these channels' unique subcellular trafficking to organelles such as the primary cilium. PKD2L1 is the only member of the polycystin family to form constitutively active ion channels on the plasma membrane when overexpressed. Experimental Approach HEK293 cells stably expressing PKD2L1 F514A were pharmacologically screened via high-throughput electrophysiology to identify potent polycystin channel modulators. In-silico docking analysis and mutagenesis were used to define the receptor sites of screen hits. Inhibition by membrane-impermeable QX-314 was used to evaluate PKD2L1's binding site accessibility. Key Results Screen results identify potent PKD2L1 antagonists with divergent chemical core structures and highlight striking similarities between the molecular pharmacology of PKD2L1 and voltage-gated sodium channels. Docking analysis, channel mutagenesis, and physiological recordings identify an open-state accessible lateral fenestration receptor within the pore, and a mechanism of inhibition that stabilizes the PKD2L1 inactivated state. Conclusion and Implication Outcomes establish the suitability of our approach to expand our chemical knowledge of polycystins and delineates novel receptor moieties for the development of channel-specific antagonists in TRP channel research.
Collapse
Affiliation(s)
- Eduardo Guadarrama
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Carlos G. Vanoye
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Paul G. DeCaen
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois, USA
| |
Collapse
|
34
|
Li YS, Ren HC, Li H, Xing M, Cao JH. From oxidative stress to metabolic dysfunction: The role of TRPM2. Int J Biol Macromol 2025; 284:138081. [PMID: 39603285 DOI: 10.1016/j.ijbiomac.2024.138081] [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: 10/04/2024] [Revised: 11/14/2024] [Accepted: 11/24/2024] [Indexed: 11/29/2024]
Abstract
Metabolic syndromes including atherosclerosis, diabetes, obesity, and hypertension are increasingly prevalent worldwide. The disorders are the primary attributes of oxidative stress and inflammation. The transient receptor potential M2 (TRPM2) channel is a pivotal mediator linking oxidative stress to metabolic dysfunction. TRPM2, a non-selective cation channel activated by reactive oxygen species (ROS) and adenosine diphosphate ribose (ADPR), regulates calcium influx, inflammation, and cell death across various tissues. This review explores the structural and activation mechanisms of TRPM2, emphasizing its significance in metabolic diseases. Elevated levels of TRPM2 play a vital role in the disease progression by influencing physiological and cellular processes such as endothelial dysfunction, immune cell activation, and mitochondrial impairment. In conditions such as atherosclerosis, ischemic stroke, diabetes, obesity, and hypertension; TRPM2 exacerbates oxidative damage, amplifies inflammatory responses, and disrupts metabolic homeostasis. Recent research highlights the potential of TRPM2 as a therapeutic target, developing specified inhibitors. This review underscores the multifaceted role of TRPM2 in metabolic disorders and its promise as a target for therapeutic interventions.
Collapse
Affiliation(s)
- Ying-Shuang Li
- Intravenous Drug Administration Center, Department of Pharmacy, Qingdao Third People's Hospital affiliated with Qingdao University, Qingdao, Shandong 266041, PR China
| | - Hua-Cheng Ren
- Intravenous Drug Administration Center, Department of Pharmacy, Qingdao Third People's Hospital affiliated with Qingdao University, Qingdao, Shandong 266041, PR China
| | - Hui Li
- Intravenous Drug Administration Center, Department of Pharmacy, Qingdao Third People's Hospital affiliated with Qingdao University, Qingdao, Shandong 266041, PR China
| | - Man Xing
- Intravenous Drug Administration Center, Department of Pharmacy, Qingdao Third People's Hospital affiliated with Qingdao University, Qingdao, Shandong 266041, PR China
| | - Jian-Hua Cao
- Intravenous Drug Administration Center, Department of Pharmacy, Qingdao Third People's Hospital affiliated with Qingdao University, Qingdao, Shandong 266041, PR China.
| |
Collapse
|
35
|
Li J, Zhou Z, Wu Y, Zhao J, Duan H, Peng Y, Wang X, Fan Z, Yin L, Li M, Liu F, Yang Y, Du L, Li J, Zhong H, Hou W, Zhang F, Ma H, Zhang X. Heat acclimation defense against exertional heat stroke by improving the function of preoptic TRPV1 neurons. Theranostics 2025; 15:1376-1398. [PMID: 39816678 PMCID: PMC11729562 DOI: 10.7150/thno.101422] [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: 07/03/2024] [Accepted: 11/19/2024] [Indexed: 01/18/2025] Open
Abstract
Rationale: Record-breaking heatwaves caused by greenhouse effects lead to multiple hyperthermia disorders, the most serious of which is exertional heat stroke (EHS) with the mortality reaching 60 %. Repeat exercise with heat exposure, termed heat acclimation (HA), protects against EHS by fine-tuning feedback control of body temperature (Tb), the mechanism of which is opaque. This study aimed to explore the molecular and neural circuit mechanisms of the HA training against EHS. Methods: Male C57BL/6 mice (6-8 weeks) and male TRPV1-Cre mice (6-8 weeks) were used in our experiments. The EHS model with or without HA training were established for this study. RNA sequencing, qPCR, immunoblot, immunofluorescent assays, calcium imaging, optogenetic/ chemical genetic intervention, virus tracing, patch clamp, and other methods were employed to investigate the molecular mechanism and neural circuit by which HA training improves the function of the medial preoptic area (mPOA) neurons. Furthermore, a novel exosome-based strategy targeting the central nervous system to deliver irisin, a protective peptide generated by HA, was established to protect against EHS. Results: HA-related neurons in the mPOA expressing transient receptor potential vanilloid-1 (TRPV1) were identified as a population whose activation reduces Tb; inversely, dysfunction of these neurons contributes to hyperthermia and EHS. mPOATRPV1 neurons facilitate vasodilation and reduce adipose tissue thermogenesis, which is associated with their inhibitory projection to the raphe pallidus nucleus (RPa) and dorsal medial hypothalamus (DMH) neurons, respectively. Furthermore, HA improves the function of preoptic heat-sensitive neurons by enhancing TRPV1 expression, and Trpv1 ablation reverses the HA-induced heat tolerance. A central nervous system-targeted exosome strategy to deliver irisin, a protective peptide generated by HA, can promote preoptic TRPV1 expression and exert similar protective effects against EHS. Conclusions: Preoptic TRPV1 neurons could be enhanced by HA, actively contributing to heat defense through the mPOA"DMH/RPa circuit during EHS, which results in the suppression of adipose tissue thermogenesis and facilitation of vasodilatation. A delivery strategy of exosomes engineered with RVG-Lamp2b-Irisin significantly improves the function of mPOATRPV1 neurons, providing a promising preventive strategy for EHS in the future.
Collapse
Affiliation(s)
- Jing Li
- Department of Critical Care Medicine and Department of Anaesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Ziqing Zhou
- Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China, 100071
| | - You Wu
- Department of Critical Care Medicine and Department of Anaesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Jianshuai Zhao
- Department of Critical Care Medicine and Department of Anaesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Haokai Duan
- Department of Microbiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Yuliang Peng
- Department of Critical Care Medicine and Department of Anaesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Xiaoke Wang
- Department of Microbiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Zhongmin Fan
- Department of Critical Care Medicine and Department of Anaesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Lu Yin
- Department of Critical Care Medicine and Department of Anaesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Mengyun Li
- Department of Microbiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Fuhong Liu
- Department of Critical Care Medicine and Department of Anaesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Yongheng Yang
- Department of Critical Care Medicine and Department of Anaesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Lixia Du
- Department of Critical Care Medicine and Department of Anaesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Jin Li
- Department of Critical Care Medicine and Department of Anaesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Haixing Zhong
- Department of Critical Care Medicine and Department of Anaesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Wugang Hou
- Department of Critical Care Medicine and Department of Anaesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Fanglin Zhang
- Department of Microbiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Hongwei Ma
- Department of Critical Care Medicine and Department of Anaesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
- Department of Microbiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Xijing Zhang
- Department of Critical Care Medicine and Department of Anaesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| |
Collapse
|
36
|
Rubaiy HN. Transient Receptor Potential Canonical Channels in Cardiovascular Pathology and Their Modulators. J Cardiovasc Pharmacol 2025; 85:21-34. [PMID: 39405561 DOI: 10.1097/fjc.0000000000001643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 10/05/2024] [Indexed: 01/18/2025]
Abstract
ABSTRACT Ion channels play a crucial role in various aspects of cardiac function, such as regulating rhythm and contractility. As a result, they serve as key targets for therapeutic interventions in cardiovascular diseases. Cell function is substantially influenced by the concentration of free cytosolic calcium (Ca 2+ ) and the voltage across the plasma membrane. These characteristics are known to be regulated by Ca 2+ -permeable nonselective cationic channels, although our knowledge of these channels is still inadequate. The transient receptor potential (TRP) superfamily comprises of many nonselective cation channels with diverse Ca 2+ permeability. Canonical or classical TRP (TRPC) channels are a subgroup of the TRP superfamily that are expressed ubiquitously in mammalian cells. TRPC channels are multidimensional signaling protein complexes that play essential roles in a variety of physiological and pathological processes in humans, including cancer, neurological disorders, cardiovascular diseases, and others. The objective of this article was to focus on the role that TRPC channels play in the cardiovascular system. The role of TRPC channels will be deeply discussed in cardiovascular pathology. Together, a critical element in developing novel treatments that target TRPC channels is comprehending the molecular mechanisms and regulatory pathways of TRPC channels in related cardiovascular diseases and conditions.
Collapse
Affiliation(s)
- Hussein N Rubaiy
- Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
37
|
Lin C, Hu Y, Lin Z, Du L, Hu Y, Ouyang L, Xie X, Cheng P, Liao J, Lu L, Zeng R, Xia P, Hou Z, Liu G, Hu H. MMP-9 responsive hydrogel promotes diabetic wound healing by suppressing ferroptosis of endothelial cells. Bioact Mater 2025; 43:240-254. [PMID: 39386223 PMCID: PMC11461830 DOI: 10.1016/j.bioactmat.2024.09.006] [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: 06/27/2024] [Revised: 08/20/2024] [Accepted: 09/04/2024] [Indexed: 10/12/2024] Open
Abstract
Ferroptosis plays a crucial role in the progression of diabetic wounds, suggesting potential therapeutic strategies to target ferroptosis. Transient receptor potential ankyrin 1 (TRPA1) is a non-selective calcium channel that acts as a receptor for a variety of physical or chemical stimuli. Cinnamaldehyde (CA) is a specific TRPA1 agonist. In in vitro experiments, we observed that high glucose (HG) treatment induced endothelial cell ferroptosis, impairing cell function. CA successfully inhibited endothelial cell ferroptosis, improving migration, proliferation, and tube formation. Further mechanistic studies showed that CA-activated TRPA1-induced Ca2+ influx promoted the phosphorylation of calmodulin-dependent protein kinase II (CaMKII) and nuclear factor-E 2-related factor 2 (Nrf2) translocation, which contributed to the elevation of glutathione peroxidase 4 (GPX4), leading to the inhibition of endothelial cell ferroptosis. In addition, CA was incorporated into an MMP-9-responsive injectable duplex hybrid hydrogel (CA@HA-Gel), allowing its efficient sustained release into diabetic wounds in an inflammation-responsive manner. The results showed that CA@HA-Gel inhibited wound endothelial cell ferroptosis and significantly promoted diabetic wound healing. In summary, the results presented in this study emphasize the potential therapeutic application of CA@HA-Gel in the treatment of diseases associated with ferroptosis.
Collapse
Affiliation(s)
- Chuanlu Lin
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Yiqiang Hu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Ze Lin
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Longyu Du
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Yixin Hu
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Lizhi Ouyang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Xudong Xie
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Peng Cheng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Jiewen Liao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Li Lu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Ruiyin Zeng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Ping Xia
- Department of Orthopaedics, Wuhan Fourth Hospital (Puai Hospital), Wuhan, China
| | - Zhiyong Hou
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, China
- Key Laboratory of Orthopaedic Biomechanics of Hebei Province, Shijiazhuang, China
| | - Guohui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Hankun Hu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
- Hubei Micro-explore Innovative Pharmaceutical Research Co., Ltd, Wuhan, Hubei, 430074, China
- Suzhou Organ-on-a-Chip System Science and Technology Co., Ltd, Suzhou, Jiangsu, 215000, China
| |
Collapse
|
38
|
Hansen CE, Hollaus D, Kamermans A, de Vries HE. Tension at the gate: sensing mechanical forces at the blood-brain barrier in health and disease. J Neuroinflammation 2024; 21:325. [PMID: 39696463 PMCID: PMC11657007 DOI: 10.1186/s12974-024-03321-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/07/2024] [Indexed: 12/20/2024] Open
Abstract
Microvascular brain endothelial cells tightly limit the entry of blood components and peripheral cells into the brain by forming the blood-brain barrier (BBB). The BBB is regulated by a cascade of mechanical and chemical signals including shear stress and elasticity of the adjacent endothelial basement membrane (BM). During physiological aging, but especially in neurological diseases including multiple sclerosis (MS), stroke, small vessel disease, and Alzheimer's disease (AD), the BBB is exposed to inflammation, rigidity changes of the BM, and disturbed cerebral blood flow (CBF). These altered forces lead to increased vascular permeability, reduced endothelial reactivity to vasoactive mediators, and promote leukocyte transmigration. Whereas the molecular players involved in leukocyte infiltration have been described in detail, the importance of mechanical signalling throughout this process has only recently been recognized. Here, we review relevant features of mechanical forces acting on the BBB under healthy and pathological conditions, as well as the endothelial mechanosensory elements detecting and responding to altered forces. We demonstrate the underlying complexity by focussing on the family of transient receptor potential (TRP) ion channels. A better understanding of these processes will provide insights into the pathogenesis of several neurological disorders and new potential leads for treatment.
Collapse
Affiliation(s)
- Cathrin E Hansen
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands
| | - David Hollaus
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Alwin Kamermans
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands.
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
39
|
Stein C. Effects of pH on opioid receptor activation and implications for drug design. Biophys J 2024; 123:4158-4166. [PMID: 38970252 PMCID: PMC11700362 DOI: 10.1016/j.bpj.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/29/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024] Open
Abstract
G-protein-coupled receptors are integral membrane proteins that transduce chemical signals from the extracellular matrix into the cell. Traditional drug design has considered ligand-receptor interactions only under normal conditions. However, studies on opioids indicate that such interactions are very different in diseased tissues. In such microenvironments, protons play an important role in structural and functional alterations of both ligands and receptors. The pertinent literature strongly suggests that future drug design should take these aspects into account in order to reduce adverse side effects while preserving desired effects of novel compounds.
Collapse
Affiliation(s)
- Christoph Stein
- Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Experimental Anaesthesiology, Berlin, Germany.
| |
Collapse
|
40
|
Mellado Lagarde MM, Wilbraham D, Martins RF, Zhao HS, Jackson K, Johnson KW, Knopp KL, DiBenedetto D, Broad LM. Clinical proof-of-concept results with a novel TRPA1 antagonist (LY3526318) in 3 chronic pain states. Pain 2024:00006396-990000000-00793. [PMID: 39679712 DOI: 10.1097/j.pain.0000000000003487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/17/2024] [Indexed: 12/17/2024]
Abstract
ABSTRACT Transient receptor potential ankyrin 1 (TRPA1) is implicated in physiological and pathological nociceptive signaling, but the clinical benefit of TRPA1 antagonists in chronic pain is not clearly demonstrated. LY3526318 is an oral, potent, and selective novel TRPA1 antagonist. The Chronic Pain Master Protocol was used to evaluate the safety and efficacy of LY3526318 in 3 randomized, placebo-controlled, proof-of-concept studies in knee osteoarthritis pain (OA), chronic low back pain (CLBP), and diabetic peripheral neuropathic pain (DPNP). Participants were randomized (1:2, placebo:LY3526318, 250 mg daily) into an 8-week double-blinded period. At 4 weeks, participants treated with LY3526318 transitioned to a placebo. The primary endpoint was the self-reported daily pain intensity measured using a Numerical Rating Scale (NRS) at 4 weeks. All endpoints were collected for up to 8 weeks. Change from baseline in average weekly NRS was analyzed using Bayesian mixed model repeated measures in the OA (N = 160), CLBP (N = 159), and DPNP (N = 154) studies. Baseline characteristics were balanced between treatment arms. Mean NRS change from baseline to week 4 did not differ significantly between placebo and LY3526318; however, a numerical improvement was observed in the CLBP, not in the OA or DPNP populations. Safety analysis integrated across studies enhanced understanding of the safety profile of LY3526318. LY3526318 showed a potential drug-induced hepatotoxic effect posing a risk for clinical development. No other safety signals were identified. LY3526318 showed potential for different responses among chronic pain indications and patient subpopulations, highlighting challenges in developing TRPA1 antagonists but supporting their value as a target in managing chronic pain.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Kelly L Knopp
- Eli Lilly and Company, Indianapolis, IN, United States
| | | | - Lisa M Broad
- Eli Lilly and Company, Bracknell, United Kingdom
| |
Collapse
|
41
|
Li J, Wei Y, Wang Y, Zhang Y, Xu Y, Ma H, Ma L, Zeng Q. Metabolomics study of APETx2 post-conditioning on myocardial ischemia-reperfusion injury. Front Pharmacol 2024; 15:1470142. [PMID: 39712499 PMCID: PMC11658994 DOI: 10.3389/fphar.2024.1470142] [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: 07/25/2024] [Accepted: 11/18/2024] [Indexed: 12/24/2024] Open
Abstract
Background Acid-sensing ion channels are activated during myocardial ischemia and are implicated in the mechanism of myocardial ischemia-reperfusion injury (MIRI). Acid-sensing ion channel 3 (ASIC3), the most pH-sensitive member of the ASIC family, is highly expressed in myocardial tissues. However, the role of ASIC3 in MIRI and its precise effects on the myocardial metabolome remain unclear. These unknowns might be related to the cardioprotective effects observed with APETx2 post-conditioning. Method Rat hearts subjected to Langendorff perfusion were randomly assigned to the normal (Nor) group, ischemia/reperfusion (I/R) group, ASIC3 blockade (AP) group. Rat hearts in group AP were treated with the ASIC3-specific inhibitor APETx2 (630 nM). Molecular and morphological changes were observed to elucidate the role of ASIC3 in MIRI. Bioinformatics analyses identified differential metabolites and pathways associated with APETx2 post-conditioning. Results APETx2 post-conditioning stabilized hemodynamics in the isolated rat heart model of MIRI. It also reduced myocardial infarct size, mitigated mitochondrial damage at the ultrastructural level, and improved markers of myocardial injury and oxidative stress. Further more, we observed that phosphatidylcholine, phosphatidylethanolamine, citric acid, cyanidin 5-O-beta-D-glucoside, and L-aspartic acid decreased after MIRI. The levels of these metabolites were partially restored by APETx2 post-conditioning. These metabolites are primarily involved in autophagy and endogenous cannabinoid signaling pathways. Conclusion ASIC3 is potentially a key player in MIRI. APETx2 post-conditioning may improve MIRI through specific metabolic changes. This study provides valuable data for future research on the metabolic mechanisms underlying the effects of APETx2 post-conditioning in MIRI.
Collapse
Affiliation(s)
- Jing Li
- Department of Anesthesiology, The Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
- School of Anesthesiology, Guizhou Medical University, Guiyang, Guizhou Province, China
- School of Anesthesiology, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Yiyong Wei
- Department of Anesthesiology, Affiliated Shenzhen Women and Children’s Hospital (Longgang) of Shantou University Medical College (Longgang District Maternity & Child Healthcare Hospital of Shenzhen City), Shenzhen, Guangdong Province, China
| | - Yi Wang
- School of Anesthesiology, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Yue Zhang
- School of Anesthesiology, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Ying Xu
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Huanhuan Ma
- School of Anesthesiology, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Lulin Ma
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, China
| | - Qingfan Zeng
- Department of Anesthesiology, The Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
- School of Anesthesiology, Guizhou Medical University, Guiyang, Guizhou Province, China
| |
Collapse
|
42
|
Lamberti A, Serafini M, Aprile S, Bhela IP, Goutsiou G, Pessolano E, Fernandez-Ballester G, Ferrer-Montiel A, Di Martino RMC, Fernandez-Carvajal A, Pirali T. The multicomponent Passerini reaction as a means of accessing diversity in structure, activity and properties: Soft and hard vanilloid/cannabinoid modulators. Eur J Med Chem 2024; 279:116845. [PMID: 39265249 DOI: 10.1016/j.ejmech.2024.116845] [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: 05/06/2024] [Revised: 07/23/2024] [Accepted: 08/22/2024] [Indexed: 09/14/2024]
Abstract
A growing body of evidence points to the existence of a crosstalk between the endovanilloid (EV)- and the endocannabinoid (EC) systems, leading to the concept of a single system based on a shared set of endogenous ligands and regulation mechanisms. The EV/EC system encompasses the ion channel TRPV1, the G protein coupled receptors CB1 and CB2, their endogenous ligands and the enzymes for biosynthesis and inactivation. Disorders in which the EV/EC interaction is involved are inflammation, pain, neurodegenerative diseases and disorders of bones and skin. In the present paper, with the aim of targeting the EV/EC system, the Passerini reaction is used in a diversity-oriented approach to generate a series of α-acyloxycarboxamides bearing different substructures that resemble endogenous ligands. Compounds have been screened for activity on TRPV1, CB1 and CB2 and metabolic stability in skin cells, liver subcellular fractions and plasma. This protocol allowed to generate agents characterized by a diverse activity on TRPV1, CB1 and CB2, as well as heterogeneous metabolic stability that could allow different routes of administration, from soft drugs for topical treatment of skin diseases to hard drugs for systemic use in inflammation and pain. Compared to natural mediators, these compounds have a better drug-likeness. Among them, 41 stands out as an agonist endowed with a well-balanced activity on both TRPV1 and CB2, high selectivity over TRPM8, TRPA1 and CB1, metabolic stability and synthetic accessibility.
Collapse
Affiliation(s)
- Angela Lamberti
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Elche, Spain
| | - Marta Serafini
- Department of Pharmaceutical Sciences, Università Degli Studi Del Piemonte Orientale, Largo Donegani 2, 28100, Novara, Italy
| | - Silvio Aprile
- Department of Pharmaceutical Sciences, Università Degli Studi Del Piemonte Orientale, Largo Donegani 2, 28100, Novara, Italy
| | - Irene Preet Bhela
- Department of Pharmaceutical Sciences, Università Degli Studi Del Piemonte Orientale, Largo Donegani 2, 28100, Novara, Italy
| | - Georgia Goutsiou
- Department of Pharmaceutical Sciences, Università Degli Studi Del Piemonte Orientale, Largo Donegani 2, 28100, Novara, Italy
| | - Emanuela Pessolano
- Department of Pharmaceutical Sciences, Università Degli Studi Del Piemonte Orientale, Largo Donegani 2, 28100, Novara, Italy
| | - Gregorio Fernandez-Ballester
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Elche, Spain
| | - Antonio Ferrer-Montiel
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Elche, Spain
| | - Rita Maria Concetta Di Martino
- Department of Pharmaceutical Sciences, Università Degli Studi Del Piemonte Orientale, Largo Donegani 2, 28100, Novara, Italy.
| | - Asia Fernandez-Carvajal
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Elche, Spain.
| | - Tracey Pirali
- Department of Pharmaceutical Sciences, Università Degli Studi Del Piemonte Orientale, Largo Donegani 2, 28100, Novara, Italy
| |
Collapse
|
43
|
Liu Q, Hu M, Li S, Zhang X, Zhang R, Lyu H, Xiao S, Guo D, Chen XZ, Tang J, Zhou C. TRPM channels in human cancers: regulatory mechanism and therapeutic prospects. Biomark Res 2024; 12:152. [PMID: 39633507 PMCID: PMC11616203 DOI: 10.1186/s40364-024-00699-2] [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: 09/04/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024] Open
Abstract
The transient receptor potential melastatin (TRPM) channel family has been previously implicated in various diseases, including those related to temperature sensing, cardiovascular health, and neurodegeneration. Nowadays, increasing evidence indicates that TRPM family members also play significant roles in various types of cancers, exhibiting both pro- and anti-tumorigenic functions. They are involved in tumor cell proliferation, survival, invasion, and metastasis, serving as potential diagnostic and prognostic biomarkers for cancer. This paper begins by describing the structure and physiological functions of the TRPM family members. It then outlines their roles in several common malignancies, including pancreatic, prostate, colorectal, breast, brain cancer, and melanoma. Subsequently, we focused on investigating the specific mechanisms by which TRPM family members are involved in tumorigenesis and development from both the tumor microenvironment (TME) and intracellular signaling. TRPM channels not only transmit signals from the TME to regulate tumor cell functions, but also mediate extracellular matrix remodeling, which is conducive to the malignant transformation of tumor cells. Importantly, TRPM channels depend on the regulation of the inflow of various ions in cells, and participate in key signaling pathways involved in tumor progression, such as Wnt/β-catenin, MAPK, PI3K/AKT, p53, and autophagy. Finally, we summarize the current strategies and challenges of targeting TRPM channels in tumor treatment, and discuss the feasibility of combining targeted TRPM channel drugs with cancer immunotherapy.
Collapse
Affiliation(s)
- Qinfeng Liu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, 430074, China
| | - Mengyu Hu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, 430074, China
| | - Shi Li
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, 430074, China
| | - Xin Zhang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, 430074, China
| | - Rui Zhang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, 430074, China
| | - Hao Lyu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, 430074, China
| | - Shuai Xiao
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, 430074, China
| | - Dong Guo
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, 430074, China
| | - Xing-Zhen Chen
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Jingfeng Tang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, 430074, China
| | - Cefan Zhou
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, 430074, China.
| |
Collapse
|
44
|
Zhang H, Wu A, Nan X, Yang L, Zhang D, Zhang Z, Liu H. The Application and Pharmaceutical Development of Etomidate: Challenges and Strategies. Mol Pharm 2024; 21:5989-6006. [PMID: 39495089 DOI: 10.1021/acs.molpharmaceut.4c00325] [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: 11/05/2024]
Abstract
Etomidate is a synthetic imidazole anesthetic that exerts hypnotic effects by potentiating the action of the inhibitory neurotransmitter γ-aminobutyric acid (GABA) or directly activating the anionic GABA (GABAA) receptor. It stands out among many anesthetics because of its multiple advantages, such as good hemodynamic stability and minimal inhibition of spontaneous respiration. However, its low water solubility and side effects, such as adrenal cortex inhibition and myoclonus, have limited the clinical application of this drug. To address these issues, extensive research has been conducted on the drug delivery of etomidate in recent decades, which has led to the emergence of different etomidate preparations. Despite so many etomidate preparations, so far some of the toxic side effects have not yet been effectively addressed. Herein we discuss the pharmaceutical design of etomidate that may resolve the above problem. We also propose targeted strategies for future research on etomidate preparations and discuss the feasibility of different administration routes and dosage forms to expand the application of this drug. Through this review, we hope to draw more attention to the potential of etomidate and its application challenges and provide valuable insights into the development of new etomidate preparations.
Collapse
Affiliation(s)
- Hao Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
- Department of Pharmacy, Zigong First People's Hospital, Zigong, Sichuan 643000, People's Republic of China
| | - Ailing Wu
- Department of Anesthesiology, Second People's Hospital of Neijiang, Southwest Medical University, Neijiang, Sichuan 641000, People's Republic of China
- Department of Anesthesiology, First People's Hospital of Neijiang, Neijiang, Sichuan 641099, People's Republic of China
| | - Xichen Nan
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Luhan Yang
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Dan Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Zhuo Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Hao Liu
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| |
Collapse
|
45
|
Zhu M, Fang Y, Sun Y, Li S, Yu J, Xiong B, Wen C, Zhou B, Huang B, Yin H, Xu H. Sonogenetics in the Treatment of Chronic Diseases: A New Method for Cell Regulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407373. [PMID: 39488795 DOI: 10.1002/advs.202407373] [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: 08/28/2024] [Revised: 10/21/2024] [Indexed: 11/04/2024]
Abstract
Sonogenetics is an innovative technology that integrates ultrasound with genetic editing to precisely modulate cellular activities in a non-invasive manner. This method entails introducing and activating mechanosensitive channels on the cell membrane of specific cells using gene delivery vectors. When exposed to ultrasound, these channels can be manipulated to open or close, thereby impacting cellular functions. Sonogenetics is currently being used extensively in the treatment of various chronic diseases, including Parkinson's disease, vision restoration, and cancer therapy. This paper provides a comprehensive review of key components of sonogenetics and focuses on evaluating its prospects and potential challenges in the treatment of chronic disease.
Collapse
Affiliation(s)
- Mingrui Zhu
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Yan Fang
- Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Yikang Sun
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Shaoyue Li
- Department of Medical Ultrasound, Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Jifeng Yu
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Bing Xiong
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Congjian Wen
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Boyang Zhou
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Bin Huang
- Zhejiang Hospital, Hangzhou, 310013, P. R. China
| | - Haohao Yin
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Huixiong Xu
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| |
Collapse
|
46
|
Liu G, Dong BB, Devanarayana S, Chen RC, Liu Q. Emerging roles of mechanosensitive ion channels in ventilator induced lung injury: a systematic review. Front Immunol 2024; 15:1479230. [PMID: 39664395 PMCID: PMC11631737 DOI: 10.3389/fimmu.2024.1479230] [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: 08/11/2024] [Accepted: 11/08/2024] [Indexed: 12/13/2024] Open
Abstract
Background The pathogenetic mechanisms of ventilator-induced lung injury (VILI) still need to be elucidated. The mechanical forces during mechanical ventilation are continually sensed and transmitted by mechanosensitive ion channels (MSICs) in pulmonary endothelial, epithelial, and immune cells. In recent years, MSICs have been shown to be involved in VILI. Methods A systematic search across PubMed, the Cochrane Library, Web of Science, and ScienceDirect was performed from inception to March 2024, and the review was conducted in accordance with PRISMA guidelines. The potential eligible studies were evaluated by two authors independently. Study characteristics, quality assessment, and potential mechanisms were analyzed. Results We included 23 eligible studies, most of which were performed with murine animals in vivo. At the in vitro level, 52% and 48% of the experiments were conducted with human or animal cells, respectively. No clinical studies were found. The most reported MSICs include Piezo channels, transient receptor potential channels, potassium channels, and stretch-activated sodium channels. Piezo1 has been the most concerned channel in the recent five years. This study found that signal pathways, such as RhoA/ROCK1, could be enhanced by cyclic stretch-activated MSICs, which contribute to VILI through dysregulated inflammation and immune responses mediated by ion transport. The review indicates the emerging role of MSICs in the pathogenesis of VILI, especially as a signal-transmitting link between mechanical stretch and pathogenesis such as inflammation, disruption of cell junctions, and edema formation. Conclusions Mechanical stretch stimulates MSICs to increase transcellular ion exchange and subsequently generates VILI through inflammation and other pathogeneses mediated by MSICs signal-transmitting pathways. These findings make it possible to identify potential therapeutic targets for the prevention of lung injury through further exploration and more studies. Systematic review registration https://inplasy.com/inplasy-2024-10-0115/, identifier INPLASY2024100115.
Collapse
Affiliation(s)
- Gang Liu
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Bin-bin Dong
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shalika Devanarayana
- School of International Education, Zhengzhou University, Zhengzhou, Henan, China
| | - Rong-Chang Chen
- Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Shenzhen Institute of Respiratory Diseases, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Qi Liu
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
47
|
Liu Y, He Y, Tong J, Guo S, Zhang X, Luo Z, Sun L, Chang C, Zhuang B, Liu X. Solvent-mediated analgesia via the suppression of water permeation through TRPV1 ion channels. Nat Biomed Eng 2024:10.1038/s41551-024-01288-2. [PMID: 39572786 DOI: 10.1038/s41551-024-01288-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 10/01/2024] [Indexed: 12/15/2024]
Abstract
Activation of the ion channel transient receptor potential vanilloid 1 (TRPV1), which is integral to pain perception, leads to an expansion of channel width, facilitating the passage of cations and large organic molecules. However, the permeability of TRPV1 channels to water remains uncertain, owing to a lack of suitable tools to study water dynamics. Here, using upconversion nanophosphors to discriminate between H2O and D2O, by monitoring water permeability across activated TRPV1 at the single-cell and single-molecule levels, and by combining single-channel current measurements with molecular dynamics simulations, we show that water molecules flow through TRPV1 and reveal a direct connection between water migration, cation flow and TRPV1 functionality. We also show in mouse models of acute or chronic inflammatory pain that the administration of deuterated water suppresses TRPV1 activity, interrupts the transmission of pain signals and mitigates pain without impacting other neurological responses. Solvent-mediated analgesia may inspire alternative options for pain management.
Collapse
Affiliation(s)
- Yuxia Liu
- Department of Chemistry, National University of Singapore, Singapore, Singapore
- The N.1 Institute for Health, National University of Singapore, Singapore, Singapore
| | - Yuanyuan He
- School of Physics, Peking University, Beijing, China
- Innovation Laboratory of Terahertz Biophysics, National Innovation Institute of Defense Technology, Beijing, China
- School of Safety Engineering, North China Institute of Science and Technology, Hebei, China
| | - Jiahuan Tong
- Yale-NUS College, National University of Singapore, Singapore, Singapore
| | - Shengyang Guo
- Department of Neurobiology, School of Basic Medicine, Peking University, Beijing, China
- Key Laboratory for Neuroscience of Ministry of Education and National Health Commission of China, Beijing, China
| | - Xinyu Zhang
- Department of Neurobiology, School of Basic Medicine, Peking University, Beijing, China
- Key Laboratory for Neuroscience of Ministry of Education and National Health Commission of China, Beijing, China
| | - Zichao Luo
- Department of Chemistry, National University of Singapore, Singapore, Singapore
- The N.1 Institute for Health, National University of Singapore, Singapore, Singapore
| | - Linlin Sun
- Department of Neurobiology, School of Basic Medicine, Peking University, Beijing, China
- Key Laboratory for Neuroscience of Ministry of Education and National Health Commission of China, Beijing, China
| | - Chao Chang
- School of Physics, Peking University, Beijing, China.
- Innovation Laboratory of Terahertz Biophysics, National Innovation Institute of Defense Technology, Beijing, China.
| | - Bilin Zhuang
- Yale-NUS College, National University of Singapore, Singapore, Singapore.
| | - Xiaogang Liu
- Department of Chemistry, National University of Singapore, Singapore, Singapore.
- The N.1 Institute for Health, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
48
|
Zhang Y, Ding Y, Zeng Z, Zhu R, Zheng P, Fan S, Cao Q, Chen H, Ren W, Wu M, Wang L, Du J. Intra-channel bi-epitopic crosslinking unleashes ultrapotent antibodies targeting Na V1.7 for pain alleviation. Cell Rep Med 2024; 5:101800. [PMID: 39461335 PMCID: PMC11604545 DOI: 10.1016/j.xcrm.2024.101800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 09/01/2024] [Accepted: 09/30/2024] [Indexed: 10/29/2024]
Abstract
Crucial for cell activities, ion channels are key drug discovery targets. Although small-molecule and peptide modulators dominate ion channel drug discovery, antibodies are emerging as an alternative modality. However, challenges persist in generating potent antibodies, especially for channels with limited extracellular epitopes. We herein present a bi-epitopic crosslinking strategy to overcome these challenges, focusing on NaV1.7, a potential analgesic target. Aiming to crosslink two non-overlapping epitopes on voltage-sensing domains II and IV, we construct bispecific antibodies and ligand-antibody conjugates. Enhanced affinity and potency are observed in comparison to the monospecific controls. Among them, a ligand-antibody conjugate (1080-PEG7-ACDTB) displays a two-orders-of-magnitude improvement in potency (IC50 of 0.06 ± 0.01 nM) and over 1,000-fold selectivity for NaV1.7. Additionally, this conjugate demonstrates robust analgesic effects in mouse pain models. Our study introduces an approach to developing effective antibodies against NaV1.7, thereby initiating a promising direction for the advancement of pain therapeutics.
Collapse
Affiliation(s)
- Yaning Zhang
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China; Peking University-Tsinghua University-National Institute Biological Sciences (PTN) Joint Graduate Program, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yanchao Ding
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Ziyan Zeng
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Rui Zhu
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Peiyuan Zheng
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Shilong Fan
- The Technology Center for Protein Sciences, Tsinghua University, Beijing 100084, China
| | - Qingjuan Cao
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Hang Chen
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Weishuai Ren
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Mengling Wu
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Luyao Wang
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Juanjuan Du
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
49
|
Sukumar VK, Tai YK, Chan CW, Iversen JN, Wu KY, Fong CHH, Lim JSJ, Franco-Obregón A. Brief Magnetic Field Exposure Stimulates Doxorubicin Uptake into Breast Cancer Cells in Association with TRPC1 Expression: A Precision Oncology Methodology to Enhance Chemotherapeutic Outcome. Cancers (Basel) 2024; 16:3860. [PMID: 39594815 PMCID: PMC11592624 DOI: 10.3390/cancers16223860] [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: 10/21/2024] [Revised: 11/04/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Background/Objectives: Doxorubicin (DOX) is commonly used as a chemotherapeutic agent for the treatment of breast cancer. Nonetheless, its systemic delivery via intravenous injection and toxicity towards healthy tissues commonly result in a broad range of detrimental side effects. Breast cancer severity was previously shown to be correlated with TRPC1 channel expression that conferred upon it enhanced vulnerability to pulsed electromagnetic field (PEMF) therapy. PEMF therapy was also previously shown to enhance breast cancer cell vulnerability to DOX in vitro and in vivo that correlated with TRPC1 expression and mitochondrial respiratory rates. Methods: DOX uptake was assessed by measuring its innate autofluorescence within murine 4T1 or human MCF7 breast cancer cells following magnetic exposure. Cellular vulnerability to doxorubicin uptake was assessed by monitoring mitochondrial activity and cellular DNA content. Results: Here, we demonstrate that 10 min of PEMF exposure could augment DOX uptake into 4T1 and MCF7 breast cancer cells. DOX uptake could be increased by TRPC1 overexpression, whereas inhibiting the activity of TRPC1 channels with SKF-96356 or genetic knockdown, precluded DOX uptake. PEMF exposure enhances DOX-mediated killing of breast cancer cells, reducing the IC50 value of DOX by half, whereas muscle cells, representative of collateral tissues, were less sensitive to PEMF-enhanced DOX-mediated cytotoxicity. Vesicular loading of DOX correlated with TRPC1 expression. Conclusions: This study presents a novel TRPC1-mediated mechanism through which PEMF therapy may enhance DOX cytotoxicity in breast cancer cells, paving the way for the development of localized non-invasive PEMF platforms to improve cancer outcomes with lower systemic levels of DOX.
Collapse
Affiliation(s)
- Viresh Krishnan Sukumar
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; (V.K.S.); (J.S.J.L.)
- BICEPS Lab (Biolonic Currents Electromagnetic Pulsing Systems), National University of Singapore, Singapore 117599, Singapore; (J.N.I.); (K.Y.W.); (C.H.H.F.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
| | - Yee Kit Tai
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; (V.K.S.); (J.S.J.L.)
- BICEPS Lab (Biolonic Currents Electromagnetic Pulsing Systems), National University of Singapore, Singapore 117599, Singapore; (J.N.I.); (K.Y.W.); (C.H.H.F.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
| | - Ching Wan Chan
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
| | - Jan Nikolas Iversen
- BICEPS Lab (Biolonic Currents Electromagnetic Pulsing Systems), National University of Singapore, Singapore 117599, Singapore; (J.N.I.); (K.Y.W.); (C.H.H.F.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
| | - Kwan Yu Wu
- BICEPS Lab (Biolonic Currents Electromagnetic Pulsing Systems), National University of Singapore, Singapore 117599, Singapore; (J.N.I.); (K.Y.W.); (C.H.H.F.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
| | - Charlene Hui Hua Fong
- BICEPS Lab (Biolonic Currents Electromagnetic Pulsing Systems), National University of Singapore, Singapore 117599, Singapore; (J.N.I.); (K.Y.W.); (C.H.H.F.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
| | - Joline Si Jing Lim
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; (V.K.S.); (J.S.J.L.)
- Experimental Therapeutics Programme, Cancer Science Institute, Singapore 117599, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University Singapore, Singapore 119228, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, National University Hospital, Singapore 119074, Singapore
| | - Alfredo Franco-Obregón
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; (V.K.S.); (J.S.J.L.)
- BICEPS Lab (Biolonic Currents Electromagnetic Pulsing Systems), National University of Singapore, Singapore 117599, Singapore; (J.N.I.); (K.Y.W.); (C.H.H.F.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| |
Collapse
|
50
|
Zhang N, Zhang H, Li S, Wu W, Luo P, Liu Z, Chen Y, Xia Z, Huang C, Cheng Q. Uncovering the predictive and immunomodulatory potential of transient receptor potential melastatin family-related CCNE1 in pan-cancer. Mol Cancer 2024; 23:258. [PMID: 39551726 PMCID: PMC11572178 DOI: 10.1186/s12943-024-02169-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 11/05/2024] [Indexed: 11/19/2024] Open
Abstract
Millions of new cases of cancer are diagnosed worldwide each year, making it a serious public health concern. Developments in customized therapy and early detection have significantly enhanced treatment for and results from cancer. Therefore, it is important to investigate new molecular biomarkers. In this study, we created an efficient transient receptor potential melastatin (TRPM) family members-related TRPM-Score for 17 solid tumors. CCNE1, produced from TRPM-Score, was found to be an exceptional biomarker through several sophisticated machine learning and deep learning computational techniques. TRPM-Score and CCNE1 immunotherapeutic prediction, immunological characteristics, and predictive value were thoroughly assessed. In most cancer types, CCNE1 was a substantially dangerous marker. Additional in vitro tests validated CCNE1's immunomodulatory properties, demonstrating that silencing impeded macrophage movement and decreased PD-L1 expression. Additionally, CCNE1 may accurately predict responses to cancer immunotherapy. These findings indicate that the TRPM family-particularly CCNE1, which is associated with TRPM-is a significant player in the pan-cancer domain and can be utilized as a therapeutic target and prognostic biomarkers, especially in immuno-oncology. The thorough characterization of the TRPM family and the discovery of CCNE1 as a crucial downstream effector mark important developments in our comprehension of pan-cancer biology.
Collapse
Affiliation(s)
- Nan Zhang
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Shuyu Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wantao Wu
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Chen
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhiwei Xia
- Department of Neurology, Hunan Aerospace Hospital, Hunan Normal University, Changsha, China.
| | - Chenshen Huang
- Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- Xiangya Hospital, National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.
| |
Collapse
|