1
|
Zhang Y, Zeng W, Zhang W, Wang Y, Jin S, Liu T, Luo H, Lu H. UVA Irradiation Promotes Melanoma Cell Proliferation Mediated by OPN3 Independently of ROS Production. Pigment Cell Melanoma Res 2025; 38:e13206. [PMID: 39474694 DOI: 10.1111/pcmr.13206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 08/12/2024] [Accepted: 09/26/2024] [Indexed: 12/29/2024]
Abstract
UVA radiation, a primary risk factor in melanoma progression, partly acts through the mediation of reactive oxygen species (ROS). The role of ROS in driving cutaneous melanoma toward an invasive phenotype and whether it occurs through opsins (OPNs), which are photosensitive G protein-coupled receptors, is not fully understood. This study focuses on the impact of UVA radiation on melanoma cell proliferation, with a special emphasis on OPN3. Investigating the biphasic response to various UVA doses (0.75-9 J/cm2) in A375 and MV3 cell lines, and using EdU and CCK-8 assays, we observed dose-dependent changes in cell proliferation. Interestingly, UVA irradiation at these doses of 0.75, 1.5 and 3 J/cm2 did not significantly induce ROS production. Our study further delves into the role of OPN3, a photosensitive receptor, in melanoma progression. Following UVA exposure, an increase in OPN3 expression was observed in melanoma cells lines A375 and MV3, indicating its role as a UVA-sensitive sensor and its influence on cell proliferation. Additionally, UVA-induced calcium flux in two melanoma cells lines pointed to a calcium-dependent G protein-coupled pathway in melanoma proliferation, mediated by OPN3 and not reliant on ROS. This research sheds light on the mechanism of UVA-induced melanoma progression, underscoring OPN3 as a pivotal regulator and advancing our understanding of UVA's interaction with opsins in melanoma progression.
Collapse
Affiliation(s)
- Yulei Zhang
- Department of Dermatology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Wen Zeng
- Department of Dermatology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Wei Zhang
- Department of Dermatology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Yu Wang
- Department of Dermatology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Shuqi Jin
- Department of Dermatology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Ting Liu
- Department of Dermatology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Huanhuan Luo
- Department of Dermatology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Hongguang Lu
- Department of Dermatology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
2
|
Ahuja K, Raju S, Dahiya S, Motiani RK. ROS and calcium signaling are critical determinant of skin pigmentation. Cell Calcium 2024; 125:102987. [PMID: 39708588 DOI: 10.1016/j.ceca.2024.102987] [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: 10/21/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
Pigmentation is a protective phenomenon that shields skin cells from UV-induced DNA damage. Perturbations in pigmentation pathways predispose to skin cancers and lead to pigmentary disorders. These ailments impart psychological trauma and severely affect the patients' quality of life. Emerging literature suggests that reactive oxygen species (ROS) and calcium (Ca2+) signaling modules regulate physiological pigmentation. Further, pigmentary disorders are associated with dysregulated ROS homeostasis and changes in Ca2+ dynamics. Here, we systemically review the literature that demonstrates key role of ROS and Ca2+ signaling in pigmentation and pigmentary disorders. Further, we discuss recent studies, which have revealed that organelle-specific Ca2+ transport mechanisms are critical determinant of pigmentation. Importantly, we deliberate upon the possibility of clinical management of pigmentary disorders by therapeutically targeting ROS generation and cellular Ca2+ handling toolkit. Finally, we highlight the key outstanding questions in the field that demand critical and timely attention. Although an important role of ROS and Ca2+ signaling in regulating skin pigmentation has emerged, the underlying molecular mechanisms remain poorly understood. In future, it would be vital to investigate in detail the signaling cascades that connect perturbed ROS homeostasis and Ca2+ signaling to human pigmentary disorders.
Collapse
Affiliation(s)
- Kriti Ahuja
- Laboratory of Calciomics and Systemic Pathophysiology (LCSP), Regional Centre for Biotechnology (RCB), Faridabad-121001, Delhi-NCR, India
| | - Sharon Raju
- Laboratory of Calciomics and Systemic Pathophysiology (LCSP), Regional Centre for Biotechnology (RCB), Faridabad-121001, Delhi-NCR, India
| | - Sakshi Dahiya
- Laboratory of Calciomics and Systemic Pathophysiology (LCSP), Regional Centre for Biotechnology (RCB), Faridabad-121001, Delhi-NCR, India
| | - Rajender K Motiani
- Laboratory of Calciomics and Systemic Pathophysiology (LCSP), Regional Centre for Biotechnology (RCB), Faridabad-121001, Delhi-NCR, India.
| |
Collapse
|
3
|
Lapajne L, Lakk M, Rudzitis CN, Vemaraju S, Lang RA, Hawlina M, Križaj D. Neuropsin, TRPV4 and intracellular calcium mediate intrinsic photosensitivity in corneal epithelial cells. Ocul Surf 2024; 36:1-9. [PMID: 39681161 DOI: 10.1016/j.jtos.2024.12.002] [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: 10/27/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024]
Abstract
PURPOSE To investigate intrinsic phototransduction in the corneal epithelium and its role in intracellular and inflammatory signaling. METHODS Optical imaging in isolated corneal epithelial cells (CECs) and debrided epithelia was combined with molecular, biochemical, pharmacological assays and gene deletion studies to track UVB-induced calcium signaling and release of cytokines, chemokines and matrix remodeling enzymes. Results from wild type mouse CECs were compared to data obtained from Opn5-/- and Trpv4-/- cells. RESULTS UVB stimuli and TRPV4 activity induced epithelial release of IL-1β, IL-17, matrix metalloproteinases MMP-3/MMP-9, and thymic stromal lymphopoietin (TSLP). UVB stimuli evoked [Ca2+]i elevations in dissociated mouse CECs that were partially reduced by inhibition of TRPV4 channels, Trpv4 knockdown and replacement of control saline with Ca2+-free saline. UVB-induced Ca2+ responses were significantly suppressed by OPN5 deletion and by inhibition of phospholipase C signaling, and responses were abrogated in cells with depleted intracellular Ca2+ stores. CONCLUSIONS Mammalian CECs are intrinsically and constitutively photosensitive. UVB photons are transduced by neuropsin, phospholipase C and CICR signaling, with mouse but not human CE transduction exhibiting a UVB-sensitive TRPV4 component. TRPV4 activity and UVB transduction are linked to cell-autonomous release of proinflammatory, matrix remodeling and nociceptive interleukins and MMPS. TRPV4-induced cytokine release may contribute to the pain induced by mechanical injury of the cornea and CEC photosensing may alert and protect the visual system from ultraviolet B (UVB) radiation -induced snow blindness, injury, vision loss and cancer.
Collapse
Affiliation(s)
- Luka Lapajne
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT, USA; Department of Ophthalmology, University Medical Center, Ljubljana, Slovenia
| | - Monika Lakk
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Christopher N Rudzitis
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT, USA; Interdepartmental Program in Neuroscience, University of Utah, USA
| | - Shruti Vemaraju
- Department of Ophthalmology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Richard A Lang
- Department of Ophthalmology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Marko Hawlina
- Department of Ophthalmology, University Medical Center, Ljubljana, Slovenia
| | - David Križaj
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT, USA; Interdepartmental Program in Neuroscience, University of Utah, USA; Department of Bioengineering, University of Utah, Salt Lake City, UT, USA; Department of Neurobiology, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
4
|
Tanwar J, Ahuja K, Sharma A, Sehgal P, Ranjan G, Sultan F, Agrawal A, D’Angelo D, Priya A, Yenamandra VK, Singh A, Raffaello A, Madesh M, Rizzuto R, Sivasubbu S, Motiani RK. Mitochondrial calcium uptake orchestrates vertebrate pigmentation via transcriptional regulation of keratin filaments. PLoS Biol 2024; 22:e3002895. [PMID: 39527653 PMCID: PMC11581414 DOI: 10.1371/journal.pbio.3002895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 11/21/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024] Open
Abstract
Mitochondria regulate several physiological functions through mitochondrial Ca2+ dynamics. However, role of mitochondrial Ca2+ signaling in melanosome biology remains unknown. Here, we show that pigmentation requires mitochondrial Ca2+ uptake. In vitro gain and loss of function studies demonstrate that mitochondrial Ca2+ uniporter (MCU) is crucial for melanogenesis while MCU rheostat, MCUb negatively control melanogenesis. Zebrafish, MCU+/- and MCUb-/- mice models show that MCU complex drives pigmentation in vivo. Mechanistically, MCU silencing activates transcription factor NFAT2 to induce expression of keratin (5, 7, and 8) filaments. Interestingly, keratin5 in turn augments mitochondrial Ca2+ uptake and potentiates melanogenesis by regulating melanosome biogenesis and maturation. Hence this signaling module acts as a negative feedback loop that fine-tunes both mitochondrial Ca2+ signaling and pigmentation. Notably, mitoxantrone, an FDA approved drug that inhibits MCU, reduces pigmentation thereby highlighting therapeutic potential of targeting mitochondrial Ca2+ uptake for clinical management of pigmentary disorders. Taken together, we reveal an MCU-NFAT2-Keratin5 driven signaling axis that acts as a critical determinant of mitochondrial Ca2+ uptake and pigmentation. Given the vital role of mitochondrial Ca2+ signaling and keratin filaments in cellular physiology, this feedback loop could be operational in a variety of other patho-physiological processes.
Collapse
Affiliation(s)
- Jyoti Tanwar
- Laboratory of Calciomics and Systemic Pathophysiology (LCSP), Regional Centre for Biotechnology (RCB), Faridabad, Delhi-NCR, India
| | - Kriti Ahuja
- Laboratory of Calciomics and Systemic Pathophysiology (LCSP), Regional Centre for Biotechnology (RCB), Faridabad, Delhi-NCR, India
| | - Akshay Sharma
- Laboratory of Calciomics and Systemic Pathophysiology (LCSP), Regional Centre for Biotechnology (RCB), Faridabad, Delhi-NCR, India
| | - Paras Sehgal
- CSIR-Institute of Genomics and Integrative Biology (IGIB), New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Gyan Ranjan
- Laboratory of Calciomics and Systemic Pathophysiology (LCSP), Regional Centre for Biotechnology (RCB), Faridabad, Delhi-NCR, India
| | - Farina Sultan
- Laboratory of Calciomics and Systemic Pathophysiology (LCSP), Regional Centre for Biotechnology (RCB), Faridabad, Delhi-NCR, India
| | - Anushka Agrawal
- Laboratory of Calciomics and Systemic Pathophysiology (LCSP), Regional Centre for Biotechnology (RCB), Faridabad, Delhi-NCR, India
| | - Donato D’Angelo
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Anshu Priya
- CSIR-Institute of Genomics and Integrative Biology (IGIB), New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Vamsi K. Yenamandra
- CSIR-Institute of Genomics and Integrative Biology (IGIB), New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Archana Singh
- CSIR-Institute of Genomics and Integrative Biology (IGIB), New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Anna Raffaello
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Muniswamy Madesh
- Department of Medicine, Center for Mitochondrial Medicine, Cardiology Division, University of Texas Health San Antonio, San Antonio, Texas, United States of America
| | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- National Center on Gene Therapy and RNA-Based Drugs, Padua, Italy
| | - Sridhar Sivasubbu
- CSIR-Institute of Genomics and Integrative Biology (IGIB), New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Rajender K. Motiani
- Laboratory of Calciomics and Systemic Pathophysiology (LCSP), Regional Centre for Biotechnology (RCB), Faridabad, Delhi-NCR, India
| |
Collapse
|
5
|
Huo L, Zhang X, Pang Y, Qi Y, Ren S, Wu F, Shang Y, Xi J. Expression and Mutation of SLC45A2 Affects Iris Color in Quail. J Poult Sci 2024; 61:2024015. [PMID: 38818526 PMCID: PMC11130394 DOI: 10.2141/jpsa.2024015] [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: 01/22/2024] [Accepted: 05/09/2024] [Indexed: 06/01/2024] Open
Abstract
Iris color is a prominent phenotypic feature of quail. To understand the mechanism of melanin deposition related to quail iris color, iris tissues were selected from Beijing white and Chinese yellow quail for transcriptome analysis. Differentially expressed genes (DEGs) associated with pigmentation were identified using RNA sequencing and validated by quantitative real-time polymerase chain reaction (RT-qPCR). The identified single nucleotide polymorphisms were studied using bioinformatics and iris color correlation analyses. A total of 485 DEGs were obtained, with 223 upregulated and 262 downregulated. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses were performed. Thirty-two genes were annotated using the GO database. Three important pigment synthesis pathways (Notch signaling, melanogenesis, and tyrosine metabolism) were identified in quail iris tissue (P < 0.05). The expression levels of solute carrier family 45 member 2 (SLC45A2), tyrosinase-related protein 1, vitamin D receptor, opsin 5, and docking protein 5 were significantly different between Beijing white and Chinese yellow quail, as verified by RT-qPCR. The c.1061C>T mutation in SLC45A2, which caused a single amino acid change at position 354 (threonine to methionine), was significantly associated with iris color in Beijing white and Chinese yellow quail, and might be the main reason for the different iris colors between these two quail species.
Collapse
Affiliation(s)
- Linke Huo
- College of Animal Science, Henan University of Science and
Technology, Luoyang, 471003 He’nan, P.R. China
| | - Xiaohui Zhang
- College of Animal Science, Henan University of Science and
Technology, Luoyang, 471003 He’nan, P.R. China
- Luoyang Key Laboratory of Animal Genetics and Breeding,
Luoyang 471003, P.R. China
| | - Youzhi Pang
- College of Animal Science, Henan University of Science and
Technology, Luoyang, 471003 He’nan, P.R. China
- Luoyang Key Laboratory of Animal Genetics and Breeding,
Luoyang 471003, P.R. China
| | - Yanxia Qi
- College of Animal Science, Henan University of Science and
Technology, Luoyang, 471003 He’nan, P.R. China
- Luoyang Key Laboratory of Animal Genetics and Breeding,
Luoyang 471003, P.R. China
| | - Shiwei Ren
- College of Animal Science, Henan University of Science and
Technology, Luoyang, 471003 He’nan, P.R. China
| | - Fanghu Wu
- College of Animal Science, Henan University of Science and
Technology, Luoyang, 471003 He’nan, P.R. China
| | - Yuanyuan Shang
- College of Animal Science, Henan University of Science and
Technology, Luoyang, 471003 He’nan, P.R. China
| | - Jinquan Xi
- College of Animal Science, Henan University of Science and
Technology, Luoyang, 471003 He’nan, P.R. China
| |
Collapse
|
6
|
Slominski RM, Chen JY, Raman C, Slominski AT. Photo-neuro-immuno-endocrinology: How the ultraviolet radiation regulates the body, brain, and immune system. Proc Natl Acad Sci U S A 2024; 121:e2308374121. [PMID: 38489380 PMCID: PMC10998607 DOI: 10.1073/pnas.2308374121] [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: 03/17/2024] Open
Abstract
Ultraviolet radiation (UVR) is primarily recognized for its detrimental effects such as cancerogenesis, skin aging, eye damage, and autoimmune disorders. With exception of ultraviolet B (UVB) requirement in the production of vitamin D3, the positive role of UVR in modulation of homeostasis is underappreciated. Skin exposure to UVR triggers local responses secondary to the induction of chemical, hormonal, immune, and neural signals that are defined by the chromophores and extent of UVR penetration into skin compartments. These responses are not random and are coordinated by the cutaneous neuro-immuno-endocrine system, which counteracts the action of external stressors and accommodates local homeostasis to the changing environment. The UVR induces electrical, chemical, and biological signals to be sent to the brain, endocrine and immune systems, as well as other central organs, which in concert regulate body homeostasis. To achieve its central homeostatic goal, the UVR-induced signals are precisely computed locally with transmission through nerves or humoral signals release into the circulation to activate and/or modulate coordinating central centers or organs. Such modulatory effects will be dependent on UVA and UVB wavelengths. This leads to immunosuppression, the activation of brain and endocrine coordinating centers, and the modification of different organ functions. Therefore, it is imperative to understand the underlying mechanisms of UVR electromagnetic energy penetration deep into the body, with its impact on the brain and internal organs. Photo-neuro-immuno-endocrinology can offer novel therapeutic approaches in addiction and mood disorders; autoimmune, neurodegenerative, and chronic pain-generating disorders; or pathologies involving endocrine, cardiovascular, gastrointestinal, or reproductive systems.
Collapse
Affiliation(s)
- Radomir M. Slominski
- Departments of Genetics, the University of Alabama at Birmingham, Birmingham, AL35294
| | - Jake Y. Chen
- Department of Biomedical Informatics and Data Science, the University of Alabama at Birmingham, Birmingham, AL35294
- Comprehensive Cancer Center, Cancer Chemoprevention Program, University of Alabama at Birmingham, Birmingham, AL35294
| | - Chander Raman
- Department of Dermatology, the University of Alabama at Birmingham, Birmingham, AL35294
| | - Andrzej T. Slominski
- Comprehensive Cancer Center, Cancer Chemoprevention Program, University of Alabama at Birmingham, Birmingham, AL35294
- Department of Dermatology, the University of Alabama at Birmingham, Birmingham, AL35294
- Veteran Administration Medical Center, Birmingham, AL35294
| |
Collapse
|
7
|
Wu S, Liu G, Shao P, Lin X, Yu J, Chen H, Li H, Feng S. Transdermal Sustained Release Properties and Anti-Photoaging Efficacy of Liposome-Thermosensitive Hydrogel System. Adv Healthc Mater 2024; 13:e2301933. [PMID: 37607774 DOI: 10.1002/adhm.202301933] [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: 06/19/2023] [Revised: 08/13/2023] [Indexed: 08/24/2023]
Abstract
Drug delivery systems have become a research priority in the biomedical field. The incorporation of liposomes to hydrogels further forms more robust multifunctional systems for more effective and sustained topical drug delivery. In this study, carboxymethyl-modified chitosan/hyaluronic acid (CMC/HA, CMH) thermosensitive hydrogel is developed for sustained transdermal delivery of liposomes. Hydrogels are crosslinked by hydrogen bonding, hydrophobic interaction and electrostatic interaction. The gel properties can be regulated by substitution degree (DS), and when DS = 18.20 ± 0.67% (CMH2), the gel temperature is 37.8 °C, allowing rapid gelation at body temperature (315 s). Moreover, CMH2 hydrogel has suitable spreadability (17.7-57.2 cm2 ), viscosity (2133.4 mPa s) and porous structure, which facilitated its adhesion and application on the skin and liposomes delivery. The hydrogel can retard the liposomes release, and the release rate of ascorbyl glucoside (AA2G) is 33.92-49.35% in 24 h. Hydrogel avoids the rapid clearance of liposomes from the skin and improved the skin retention, achieving the long-term release of bioactive components. Liposome-hydrogel system more efficiently promotes the anti-photoaging effect of AA2G on skin, reducing epidermal thickness, melanin deposition and lipid oxidative damage and increasing collagen density. Therefore, liposome-hydrogel systems are proposed as multifunctional delivery systems for sustained transdermal delivery.
Collapse
Affiliation(s)
- Sijie Wu
- Department of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Gaodan Liu
- Department of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Ping Shao
- Department of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
- Key Laboratory of Food Macromolecular Resources Processing Technology Research (Zhejiang University of Technology), China National Light Industry, Hangzhou, Zhejiang, 310014, China
| | - Xingyu Lin
- College of Biosystems Engineering and Food Science, State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China
| | - Jiahao Yu
- Department of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
- Key Laboratory of Food Macromolecular Resources Processing Technology Research (Zhejiang University of Technology), China National Light Industry, Hangzhou, Zhejiang, 310014, China
| | - Hanchi Chen
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Huiliang Li
- Zhejiang Yige Beauty Group, Hangzhou, 310000, China
| | - Simin Feng
- Department of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
- Key Laboratory of Food Macromolecular Resources Processing Technology Research (Zhejiang University of Technology), China National Light Industry, Hangzhou, Zhejiang, 310014, China
| |
Collapse
|
8
|
Qu Y, Sun X, Wei N, Wang K. Inhibition of cutaneous heat-sensitive Ca 2+ -permeable transient receptor potential vanilloid 3 channels alleviates UVB-induced skin lesions in mice. FASEB J 2023; 37:e23309. [PMID: 37983944 DOI: 10.1096/fj.202301591rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/20/2023] [Accepted: 10/30/2023] [Indexed: 11/22/2023]
Abstract
Ultraviolet B (UVB) radiation causes skin injury by trigging excessive calcium influx and signaling cascades in the skin keratinocytes. The heat-sensitive Ca2+ -permeable transient receptor potential vanilloid 3 (TRPV3) channels robustly expressed in the keratinocytes play an important role in skin barrier formation and wound healing. Here, we report that inhibition of cutaneous TRPV3 alleviates UVB radiation-induced skin lesions. In mouse models of ear swelling and dorsal skin injury induced by a single exposure of weak UVB radiation, TRPV3 genes and proteins were upregulated in quantitative real-time PCR and Western blot assays. In accompany with TRPV3 upregulations, the expressions of proinflammatory cytokines tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) were also increased. Knockout of the TRPV3 gene alleviates UVB-induced ear swelling and dorsal skin inflammation. Furthermore, topical applications of two selective TRPV3 inhibitors, osthole and verbascoside, resulted in a dose-dependent attenuation of skin inflammation and lesions. Taken together, our findings demonstrate the causative role of overactive TRPV3 channel function in the development of UVB-induced skin injury. Therefore, topical inhibition of TRPV3 may hold potential therapy or prevention of UVB radiation-induced skin injury.
Collapse
Affiliation(s)
- Yaxuan Qu
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, China
| | - Xiaoying Sun
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, China
- Institute of Innovative Drugs, Qingdao University, Qingdao, China
| | - Ningning Wei
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, China
- Institute of Innovative Drugs, Qingdao University, Qingdao, China
| | - KeWei Wang
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, China
- Institute of Innovative Drugs, Qingdao University, Qingdao, China
| |
Collapse
|
9
|
Marini M, Titiz M, Souza Monteiro de Araújo D, Geppetti P, Nassini R, De Logu F. TRP Channels in Cancer: Signaling Mechanisms and Translational Approaches. Biomolecules 2023; 13:1557. [PMID: 37892239 PMCID: PMC10605459 DOI: 10.3390/biom13101557] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/16/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Ion channels play a crucial role in a wide range of biological processes, including cell cycle regulation and cancer progression. In particular, the transient receptor potential (TRP) family of channels has emerged as a promising therapeutic target due to its involvement in several stages of cancer development and dissemination. TRP channels are expressed in a large variety of cells and tissues, and by increasing cation intracellular concentration, they monitor mechanical, thermal, and chemical stimuli under physiological and pathological conditions. Some members of the TRP superfamily, namely vanilloid (TRPV), canonical (TRPC), melastatin (TRPM), and ankyrin (TRPA), have been investigated in different types of cancer, including breast, prostate, lung, and colorectal cancer. TRP channels are involved in processes such as cell proliferation, migration, invasion, angiogenesis, and drug resistance, all related to cancer progression. Some TRP channels have been mechanistically associated with the signaling of cancer pain. Understanding the cellular and molecular mechanisms by which TRP channels influence cancer provides new opportunities for the development of targeted therapeutic strategies. Selective inhibitors of TRP channels are under initial scrutiny in experimental animals as potential anti-cancer agents. In-depth knowledge of these channels and their regulatory mechanisms may lead to new therapeutic strategies for cancer treatment, providing new perspectives for the development of effective targeted therapies.
Collapse
Affiliation(s)
| | | | | | | | - Romina Nassini
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, 50139 Florence, Italy; (M.M.); (M.T.); (D.S.M.d.A.); (P.G.); (F.D.L.)
| | | |
Collapse
|
10
|
Tagore M, Hergenreder E, Perlee SC, Cruz NM, Menocal L, Suresh S, Chan E, Baron M, Melendez S, Dave A, Chatila WK, Nsengimana J, Koche RP, Hollmann TJ, Ideker T, Studer L, Schietinger A, White RM. GABA Regulates Electrical Activity and Tumor Initiation in Melanoma. Cancer Discov 2023; 13:2270-2291. [PMID: 37553760 PMCID: PMC10551668 DOI: 10.1158/2159-8290.cd-23-0389] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/27/2023] [Accepted: 08/02/2023] [Indexed: 08/10/2023]
Abstract
Oncogenes can initiate tumors only in certain cellular contexts, which is referred to as oncogenic competence. In melanoma, whether cells in the microenvironment can endow such competence remains unclear. Using a combination of zebrafish transgenesis coupled with human tissues, we demonstrate that GABAergic signaling between keratinocytes and melanocytes promotes melanoma initiation by BRAFV600E. GABA is synthesized in melanoma cells, which then acts on GABA-A receptors in keratinocytes. Electron microscopy demonstrates specialized cell-cell junctions between keratinocytes and melanoma cells, and multielectrode array analysis shows that GABA acts to inhibit electrical activity in melanoma/keratinocyte cocultures. Genetic and pharmacologic perturbation of GABA synthesis abrogates melanoma initiation in vivo. These data suggest that GABAergic signaling across the skin microenvironment regulates the ability of oncogenes to initiate melanoma. SIGNIFICANCE This study shows evidence of GABA-mediated regulation of electrical activity between melanoma cells and keratinocytes, providing a new mechanism by which the microenvironment promotes tumor initiation. This provides insights into the role of the skin microenvironment in early melanomas while identifying GABA as a potential therapeutic target in melanoma. See related commentary by Ceol, p. 2128. This article is featured in Selected Articles from This Issue, p. 2109.
Collapse
Affiliation(s)
- Mohita Tagore
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Emiliano Hergenreder
- The Center for Stem Cell Biology, Sloan Kettering Institute for Cancer Research, New York, New York
- Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, New York
- Weill Graduate School of Medical Sciences of Cornell University, New York, New York
| | - Sarah C. Perlee
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, New York
- Gerstner Sloan Kettering Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nelly M. Cruz
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Laura Menocal
- Weill Graduate School of Medical Sciences of Cornell University, New York, New York
| | - Shruthy Suresh
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eric Chan
- Molecular Cytology Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Maayan Baron
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, California
| | - Stephanie Melendez
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Asim Dave
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Walid K. Chatila
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jeremie Nsengimana
- Biostatistics Research Group, Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Richard P. Koche
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Travis J. Hollmann
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Trey Ideker
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, California
| | - Lorenz Studer
- The Center for Stem Cell Biology, Sloan Kettering Institute for Cancer Research, New York, New York
- Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, New York
| | - Andrea Schietinger
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Richard M. White
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Medical College, New York, New York
- Nuffield Department of Medicine, Ludwig Institute for Cancer Research, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
11
|
Sakamoto A, Terui Y, Igarashi K, Kashiwagi K. Transient Receptor Potential Ankyrin 1 (TRPA1) Channel Mediates Acrolein Cytotoxicity in Human Lung Cancer Cells. Int J Mol Sci 2023; 24:11847. [PMID: 37511605 PMCID: PMC10380761 DOI: 10.3390/ijms241411847] [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: 06/26/2023] [Revised: 07/22/2023] [Accepted: 07/23/2023] [Indexed: 07/30/2023] Open
Abstract
Transient receptor potential ankyrin 1 (TRPA1) is a nonselective ion channel implicated in thermosensation and inflammatory pain. It has been reported that expression of the TRPA1 channel is induced by cigarette smoke extract. Acrolein found in cigarette smoke is highly toxic and known as an agonist of the TRPA1 channel. However, the role of TRPA1 in the cytotoxicity of acrolein remains unclear. Here, we investigated whether the TRPA1 channel is involved in the cytotoxicity of acrolein in human lung cancer A549 cells. The IC50 of acrolein in A549 cells was 25 μM, and acrolein toxicity increased in a concentration- and time-dependent manner. When the effect of acrolein on TRPA1 expression was examined, the expression of TRPA1 in A549 cells was increased by treatment with 50 μM acrolein for 24 h or 500 μM acrolein for 30 min. AP-1, a transcription factor, was activated in the cells treated with 50 μM acrolein for 24 h, while induction of NF-κB and HIF-1α was observed in the cells treated with 500 μM acrolein for 30 min. These results suggest that acrolein induces TRPA1 expression by activating these transcription factors. Overexpression of TRPA1 in A549 cells increased acrolein sensitivity and the level of protein-conjugated acrolein (PC-Acro), while knockdown of TRPA1 in A549 cells or treatment with a TRPA1 antagonist caused tolerance to acrolein. These findings suggest that acrolein induces the TRPA1 channel and that an increase in TRPA1 expression promotes the cytotoxicity of acrolein.
Collapse
Affiliation(s)
- Akihiko Sakamoto
- Faculty of Pharmacy, Chiba Institute of Science, Choshi 288-0025, Japan
| | - Yusuke Terui
- Faculty of Pharmacy, Chiba Institute of Science, Choshi 288-0025, Japan
| | - Kazuei Igarashi
- Amine Pharma Research Institute, Innovation Plaza at Chiba University, Chiba 260-0856, Japan
| | - Keiko Kashiwagi
- Faculty of Pharmacy, Chiba Institute of Science, Choshi 288-0025, Japan
| |
Collapse
|
12
|
Tanwar J, Ahuja K, Sharma A, Sehgal P, Ranjan G, Sultan F, Priya A, Venkatesan M, Yenamandra VK, Singh A, Madesh M, Sivasubbu S, Motiani RK. Mitochondrial calcium signaling mediated transcriptional regulation of keratin filaments is a critical determinant of melanogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.26.542250. [PMID: 37292659 PMCID: PMC10245956 DOI: 10.1101/2023.05.26.542250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Mitochondria are versatile organelles that regulate several physiological functions. Many mitochondria-controlled processes are driven by mitochondrial Ca2+ signaling. However, role of mitochondrial Ca2+ signaling in melanosome biology remains unknown. Here, we show that pigmentation requires mitochondrial Ca2+ uptake. In vitro gain and loss of function studies demonstrated that Mitochondrial Ca2+ Uniporter (MCU) is crucial for melanogenesis while the MCU rheostats, MCUb and MICU1 negatively control melanogenesis. Zebrafish and mouse models showed that MCU plays a vital role in pigmentation in vivo. Mechanistically, MCU controls activation of transcription factor NFAT2 to induce expression of three keratins (keratin 5, 7 and 8), which we report as positive regulators of melanogenesis. Interestingly, keratin 5 in turn modulates mitochondrial Ca2+ uptake thereby this signaling module acts as a negative feedback loop that fine-tunes both mitochondrial Ca2+ signaling and melanogenesis. Mitoxantrone, an FDA approved drug that inhibits MCU, decreases physiological melanogenesis. Collectively, our data demonstrates a critical role for mitochondrial Ca2+ signaling in vertebrate pigmentation and reveal the therapeutic potential of targeting MCU for clinical management of pigmentary disorders. Given the centrality of mitochondrial Ca2+ signaling and keratin filaments in cellular physiology, this feedback loop may be functional in a variety of other pathophysiological conditions.
Collapse
Affiliation(s)
- Jyoti Tanwar
- Laboratory of Calciomics and Systemic Pathophysiology (LCSP), Regional Centre for Biotechnology (RCB), Faridabad-121001, Delhi-NCR, India
| | - Kriti Ahuja
- Laboratory of Calciomics and Systemic Pathophysiology (LCSP), Regional Centre for Biotechnology (RCB), Faridabad-121001, Delhi-NCR, India
| | - Akshay Sharma
- Laboratory of Calciomics and Systemic Pathophysiology (LCSP), Regional Centre for Biotechnology (RCB), Faridabad-121001, Delhi-NCR, India
| | - Paras Sehgal
- CSIR-Institute of Genomics and Integrative Biology (IGIB), New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002 India
| | - Gyan Ranjan
- CSIR-Institute of Genomics and Integrative Biology (IGIB), New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002 India
| | - Farina Sultan
- Laboratory of Calciomics and Systemic Pathophysiology (LCSP), Regional Centre for Biotechnology (RCB), Faridabad-121001, Delhi-NCR, India
| | - Anshu Priya
- CSIR-Institute of Genomics and Integrative Biology (IGIB), New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002 India
| | - Manigandan Venkatesan
- Department of Medicine, Center for Mitochondrial Medicine, Cardiology Division, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Vamsi K Yenamandra
- CSIR-Institute of Genomics and Integrative Biology (IGIB), New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002 India
| | - Archana Singh
- CSIR-Institute of Genomics and Integrative Biology (IGIB), New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002 India
| | - Muniswamy Madesh
- Department of Medicine, Center for Mitochondrial Medicine, Cardiology Division, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Sridhar Sivasubbu
- CSIR-Institute of Genomics and Integrative Biology (IGIB), New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002 India
| | - Rajender K Motiani
- Laboratory of Calciomics and Systemic Pathophysiology (LCSP), Regional Centre for Biotechnology (RCB), Faridabad-121001, Delhi-NCR, India
| |
Collapse
|
13
|
Lan Y, Zeng W, Wang Y, Dong X, Shen X, Gu Y, Zhang W, Lu H. Opsin 3 mediates UVA-induced keratinocyte supranuclear melanin cap formation. Commun Biol 2023; 6:238. [PMID: 36869204 PMCID: PMC9984416 DOI: 10.1038/s42003-023-04621-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 02/22/2023] [Indexed: 03/05/2023] Open
Abstract
Solar ultraviolet (UV) radiation-induced DNA damage is a major risk factor for skin cancer development. UV-induced redistribution of melanin near keratinocyte nuclei leads to the formation of a supranuclear cap, which acts as a natural sunscreen and protects DNA by absorbing and scattering UV radiation. However, the mechanism underlying the intracellular movement of melanin in nuclear capping is poorly understood. In this study, we found that OPN3 is an important photoreceptor in human epidermal keratinocytes and is critical for UVA-mediated supranuclear cap formation. OPN3 mediates supranuclear cap formation via the calcium-dependent G protein-coupled receptor signaling pathway and ultimately upregulates Dync1i1 and DCTN1 expression in human epidermal keratinocytes via activating calcium/CaMKII, CREB, and Akt signal transduction. Together, these results clarify the role of OPN3 in regulating melanin cap formation in human epidermal keratinocytes, greatly expanding our understanding of the phototransduction mechanisms involved in physiological function in skin keratinocytes.
Collapse
Affiliation(s)
- Yinghua Lan
- Department of Dermatology, Affiliated Hospital of Guizhou Medical University, 550001, Guiyang, Guizhou, P.R. China
| | - Wen Zeng
- Department of Dermatology, Affiliated Hospital of Guizhou Medical University, 550001, Guiyang, Guizhou, P.R. China
| | - Yu Wang
- Department of Dermatology, Affiliated Hospital of Guizhou Medical University, 550001, Guiyang, Guizhou, P.R. China
| | - Xian Dong
- Department of Dermatology, Affiliated Hospital of Guizhou Medical University, 550001, Guiyang, Guizhou, P.R. China
| | - Xiaoping Shen
- Department of Dermatology, Affiliated Hospital of Guizhou Medical University, 550001, Guiyang, Guizhou, P.R. China
| | - Yangguang Gu
- Department of Dermatology, Affiliated Hospital of Guizhou Medical University, 550001, Guiyang, Guizhou, P.R. China
| | - Wei Zhang
- Department of Dermatology, Affiliated Hospital of Guizhou Medical University, 550001, Guiyang, Guizhou, P.R. China
| | - Hongguang Lu
- Department of Dermatology, Affiliated Hospital of Guizhou Medical University, 550001, Guiyang, Guizhou, P.R. China.
| |
Collapse
|
14
|
Wu W, Wang Y, Liu Y, Guo H, Li Z, Zou W, Liu J, Song Z. TRPA1 promotes UVB-induced skin pigmentation by regulating melanosome luminal pH. Exp Dermatol 2023; 32:165-176. [PMID: 36302111 DOI: 10.1111/exd.14693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 09/25/2022] [Accepted: 10/24/2022] [Indexed: 11/28/2022]
Abstract
Melanocytes stimulated by ultraviolet radiation (UVR) produce melanin and melanosomes, which causes skin pigmentation and acts as an important physiological defence process for photoprotection. Neutral luminal pH of melanosomes is critical for providing optimal conditions for the rate-limiting, pH-sensitive melanin synthesizing enzyme tyrosinase (TYR). As a major component of extraocular phototransduction pathway, transient receptor potential ankyrin1 (TRPA1) can be activated by ultraviolet B (UVB) and reported to be expressed in melanocytes. However, whether TRPA1 is involved in the regulation of melanogenesis remains unclear. Melanogenic activity of TRPA1 was evaluated in primary normal human epidermal melanocytes (HEMs) and murine B16-F10 cell cultures, and the effects of topical applications of TRPA1 specific agonist and antagonist on UVB-induced skin pigmentation were confirmed on in vivo guinea pig models. Calcium (Ca2+ ) imaging and pH imaging were performed to analyse the effects of TRPA1 on intracellular Ca2+ concentration ([Ca2+ ]ic ) and melanosome luminal pH. TRPA1 regulated melanin synthesis, UVB-induced Ca2+ influx and melanosome luminal pH in HEMs and B16-F10 cells. Topical treatment of TRPA1 specific agonist JT010 increased UVB-induced skin pigmentation in guinea pigs, while topical using of TRPA1 selective antagonist HC-030031 mitigated such pigmentation. Our results indicated that TRPA1 activated by UVB enhanced the skin pigmentation, most likely by regulating the [Ca2+ ]ic and the melanosomal pH, consequently influencing the enzymatic activity of TYR. Therefore, the results suggest TRPA1 as a potential therapeutic target in the treatment of skin pigmented disorders that are at high risk under UVB irradiation.
Collapse
Affiliation(s)
- Wei Wu
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yupeng Wang
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ying Liu
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Handan Guo
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhou Li
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Wei Zou
- College of Life Science, Liaoning Normal University, Dalian, China
| | - Jing Liu
- Stem Cell Clinical Research Center, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhiqi Song
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
15
|
Uzunbajakava NE, Tobin DJ, Botchkareva NV, Dierickx C, Bjerring P, Town G. Highlighting nuances of blue light phototherapy: Mechanisms and safety considerations. JOURNAL OF BIOPHOTONICS 2023; 16:e202200257. [PMID: 36151769 DOI: 10.1002/jbio.202200257] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/17/2022] [Accepted: 09/21/2022] [Indexed: 06/16/2023]
Abstract
The efficacy of blue light therapy in dermatology relies on numerous clinical studies. The safety remains a topic of controversy, where potentially deleterious effects were derived from in vitro rather than in vivo experiments. The objectives of this work were (1) to highlight the nuances behind "colors" of blue light, light propagation in tissue and the plurality of modes of action; and (2) to rigorously analyze studies on humans reporting both clinical and histological data from skin biopsies with focus on DNA damage, proliferation, apoptosis, oxidative stress, impact on collagen, elastin, immune cells, and pigmentation. We conclude that blue light therapy is safe for human skin. It induces intriguing skin pigmentation, in part mediated by photoreceptor Opsin-3, which might have a photoprotective effect against ultraviolet irradiation. Future research needs to unravel photochemical reactions and the most effective and safe parameters of blue light in dermatology.
Collapse
Affiliation(s)
| | - Desmond J Tobin
- Charles Institute of Dermatology, University College Dublin, Dublin, Ireland
| | - Natalia V Botchkareva
- Department of Dermatology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Christine Dierickx
- Skinperium Laser and Cosmetic Dermatology Clinic, Skinperium, Luxembourg City, Luxembourg
| | - Peter Bjerring
- Dermatology Department, Aalborg University Hospital, Aalborg, Denmark
| | - Godfrey Town
- Dermatology Department, Aalborg University Hospital, Aalborg, Denmark
| |
Collapse
|
16
|
Mäki-Opas I, Hämäläinen M, Moilanen E, Scotece M. TRPA1 as a potential factor and drug target in scleroderma: dermal fibrosis and alternative macrophage activation are attenuated in TRPA1-deficient mice in bleomycin-induced experimental model of scleroderma. Arthritis Res Ther 2023; 25:12. [PMID: 36698198 PMCID: PMC9875496 DOI: 10.1186/s13075-023-02994-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 01/13/2023] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Systemic sclerosis is a rheumatoid disease best known for its fibrotic skin manifestations called scleroderma. Alternatively activated (M2-type) macrophages are normally involved in the resolution of inflammation and wound healing but also in fibrosing diseases such as scleroderma. TRPA1 is a non-selective cation channel, activation of which causes pain and neurogenic inflammation. In the present study, we investigated the role of TRPA1 in bleomycin-induced skin fibrosis mimicking scleroderma. METHODS Wild type and TRPA1-deficient mice were challenged with intradermal bleomycin injections to induce a scleroderma-mimicking disease. Macrophages were investigated in vitro to evaluate the underlying mechanisms. RESULTS Bleomycin induced dermal thickening and collagen accumulation in wild type mice and that was significantly attenuated in TRPA1-deficient animals. Accordingly, the expression of collagens 1A1, 1A2, and 3A1 as well as pro-fibrotic factors TGF-beta, CTGF, fibronectin-1 and YKL-40, and M2 macrophage markers Arg1 and MRC1 were lower in TRPA1-deficient than wild type mice. Furthermore, bleomycin was discovered to significantly enhance M2-marker expression particularly in the presence of IL-4 in wild type macrophages in vitro, but not in macrophages harvested from TRPA1-deficient mice. IL-4-induced PPARγ-expression in macrophages was increased by bleomycin, providing a possible mechanism behind the phenomenon. CONCLUSIONS In conclusion, the results indicate that interfering TRPA1 attenuates fibrotic and inflammatory responses in bleomycin-induced scleroderma. Therefore, TRPA1-blocking treatment could potentially alleviate M2 macrophage driven diseases like systemic sclerosis and scleroderma.
Collapse
Affiliation(s)
- Ilari Mäki-Opas
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, 33014, Tampere, Finland.
| | - Mari Hämäläinen
- grid.412330.70000 0004 0628 2985The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, 33014 Tampere, Finland
| | - Eeva Moilanen
- grid.412330.70000 0004 0628 2985The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, 33014 Tampere, Finland
| | - Morena Scotece
- grid.412330.70000 0004 0628 2985The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, 33014 Tampere, Finland ,grid.428472.f0000 0004 1794 2467Current affiliation: Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer (CIC), Instituto de Biología Molecular y Celular del Cáncer (IBMCC), CSIC-USAL, 37007 Salamanca, Spain
| |
Collapse
|
17
|
Alimohammadi S, Pénzes Z, Horváth D, Gyetvai Á, Bácsi A, Kis NG, Németh Á, Arany J, Oláh A, Lisztes E, Tóth BI, Bíró T, Szöllősi AG. TRPV4 Activation Increases the Expression of CD207 (Langerin) of Monocyte-Derived Langerhans Cells without Affecting their Maturation. J Invest Dermatol 2022; 143:801-811.e10. [PMID: 36502939 DOI: 10.1016/j.jid.2022.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 10/20/2022] [Accepted: 10/22/2022] [Indexed: 12/14/2022]
Abstract
Langerhans cells (LCs) are the sole professional antigen-presenting cell normally found in the human epidermal compartment. Research into their physiological role is hindered by the fact that they are invariably activated during isolation from the skin. To overcome this challenge, we turned to a monocyte-derived LC (moLC) model, which we characterized with RNA sequencing, and compared the transcriptome of moLCs with that of donor-matched immature dendritic cells. We found that moLCs express markers characteristic of LC2 cells as well as TRPV4. TRPV4 is especially important in the skin because it has been linked to the conservation of the skin barrier, immunological responses, as well as acute and chronic itch, but we know little about its function on LCs. Our results show that TRPV4 activation increased the expression of Langerin and led to increased intracellular calcium concentration in moLCs. Regarding the functionality of moLCs, we found that TRPV4 agonism had a mitigating effect on their inflammatory responses because it decreased their cytokine production and T-cell activating capability. Because TRPV4 has emerged as a potential therapeutic target in dermatological conditions, it is important to highlight LCs as, to our knowledge, a previously unreported target of these therapies.
Collapse
Affiliation(s)
- Shahrzad Alimohammadi
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Doctoral School of Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsófia Pénzes
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Doctoral School of Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Dorottya Horváth
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Doctoral School of Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Ágnes Gyetvai
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Bácsi
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Nikoletta Gréta Kis
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ákos Németh
- Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Doctoral School of Health Sciences, Faculty of Public Health, University of Debrecen, Debrecen, Hungary
| | - József Arany
- Doctoral School of Molecular Medicine, University of Debrecen, Debrecen, Hungary; Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Oláh
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Erika Lisztes
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Balázs István Tóth
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamás Bíró
- Monasterium Laboratory Skin & Hair Research Solutions GmbH, Münster, Germany
| | - Attila Gábor Szöllősi
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
18
|
Swindell WR, Bojanowski K, Chaudhuri RK. Isosorbide Fatty Acid Diesters Have Synergistic Anti-Inflammatory Effects in Cytokine-Induced Tissue Culture Models of Atopic Dermatitis. Int J Mol Sci 2022; 23:ijms232214307. [PMID: 36430783 PMCID: PMC9696169 DOI: 10.3390/ijms232214307] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/08/2022] [Accepted: 11/11/2022] [Indexed: 11/19/2022] Open
Abstract
Atopic dermatitis (AD) is a chronic disease in which epidermal barrier disruption triggers Th2-mediated eruption of eczematous lesions. Topical emollients are a cornerstone of chronic management. This study evaluated efficacy of two plant-derived oil derivatives, isosorbide di-(linoleate/oleate) (IDL) and isosorbide dicaprylate (IDC), using AD-like tissue culture models. Treatment of reconstituted human epidermis with cytokine cocktail (IL-4 + IL-13 + TNF-α + IL-31) compromised the epidermal barrier, but this was prevented by co-treatment with IDL and IDC. Cytokine stimulation also dysregulated expression of keratinocyte (KC) differentiation genes whereas treatment with IDC or IDL + IDC up-regulated genes associated with early (but not late) KC differentiation. Although neither IDL nor IDC inhibited Th2 cytokine responses, both compounds repressed TNF-α-induced genes and IDL + IDC led to synergistic down-regulation of inflammatory (IL1B, ITGA5) and neurogenic pruritus (TRPA1) mediators. Treatment of cytokine-stimulated skin explants with IDC decreased lactate dehydrogenase (LDH) secretion by more than 50% (more than observed with cyclosporine) and in vitro LDH activity was inhibited by IDL and IDC. These results demonstrate anti-inflammatory mechanisms of isosorbide fatty acid diesters in AD-like skin models. Our findings highlight the multifunctional potential of plant oil derivatives as topical ingredients and support studies of IDL and IDC as therapeutic candidates.
Collapse
Affiliation(s)
- William R. Swindell
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Correspondence:
| | | | | |
Collapse
|
19
|
Pyatigorskaya NV, Filippova OV, Nikolenko NS, Kravchenko AD. Transient receptor potential Ankyrin 1: structure, function and ligands. RESEARCH RESULTS IN PHARMACOLOGY 2022. [DOI: 10.3897/rrpharmacology.8.90214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Introduction: Transient receptor potential ankyrin 1 (TRPA1) is a protein expressed in many living organisms. During the study of TRPA1, its unique biological role as a universal and polymodal sensor of various altering agents was found. The aim of this study is to search and generalize information about structural features and molecular determinants, mechanisms of activation, action and modulation of TRPA1 as a universal pain and inflammation sensor, as well as the nature of activators and antagonists of this target and their therapeutic potential.
Materials and methods: This article presents an overview of the results of scientific research of TRPA1, its modulators, as well as an overview of their pharmacological potential over the period from the discovery of these channels to the present, with an emphasis on the last decade.
Results and discussion: The main collected data on expression, structural features and molecular determinants, mechanisms of activation and action of TRPA1 indicate its role as a universal and labile element of the primary response of the body to adverse exogenous and endogenous factors. Regardless of the nature of the stimulus, hyperstimulation of TRPA1 channels can lead to such phenomena as pain, inflammation, itching, edema and other manifestations of alteration, and therefore TRPA1 blockade can be used in the treatment of various diseases accompanied by these pathological conditions. Currently, TRPA1 antagonists are being actively searched for and studied, as evidenced by a high patent activity over the past 14 years; however, the molecular mechanisms of action and pharmacological properties of TRPA1 blockers remain understudied.
Conclusion: Acquire of new information about TRPA1 will help in the development of its modulators, which can become promising analgesics, anti-inflammatory drugs, bronchodilators, and agents for the treatment of cardiovascular diseases of new generations.
Collapse
|
20
|
Dai R, Yu T, Weng D, Li H, Cui Y, Wu Z, Guo Q, Zou H, Wu W, Gao X, Qi Z, Ren Y, Wang S, Li Y, Luo M. A neuropsin-based optogenetic tool for precise control of G q signaling. SCIENCE CHINA. LIFE SCIENCES 2022; 65:1271-1284. [PMID: 35579776 DOI: 10.1007/s11427-022-2122-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 04/29/2022] [Indexed: 06/15/2023]
Abstract
Gq-coupled receptors regulate numerous physiological processes by activating enzymes and inducing intracellular Ca2+ signals. There is a strong need for an optogenetic tool that enables powerful experimental control over Gq signaling. Here, we present chicken opsin 5 (cOpn5) as the long sought-after, single-component optogenetic tool that mediates ultra-sensitive optical control of intracellular Gq signaling with high temporal and spatial resolution. Expressing cOpn5 in HEK 293T cells and primary mouse astrocytes enables blue light-triggered, Gq-dependent Ca2+ release from intracellular stores and protein kinase C activation. Strong Ca2+ transients were evoked by brief light pulses of merely 10 ms duration and at 3 orders lower light intensity of that for common optogenetic tools. Photostimulation of cOpn5-expressing cells at the subcellular and single-cell levels generated fast intracellular Ca2+ transition, thus demonstrating the high spatial precision of cOpn5 optogenetics. The cOpn5-mediated optogenetics could also be applied to activate neurons and control animal behavior in a circuit-dependent manner. cOpn5 optogenetics may find broad applications in studying the mechanisms and functional relevance of Gq signaling in both non-excitable cells and excitable cells in all major organ systems.
Collapse
Affiliation(s)
- Ruicheng Dai
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China
- School of Life Sciences, Peking University, Beijing, 100871, China
- Peking University-Tsinghua University-NIBS Joint Graduate Program, NIBS, Beijing, 102206, China
| | - Tao Yu
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China
- Peking University-Tsinghua University-NIBS Joint Graduate Program, NIBS, Beijing, 102206, China
| | - Danwei Weng
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China
- Graduate School of Peking Union Medical College, Beijing, 100730, China
| | - Heng Li
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research (TIMBR), Beijing, 102206, China
| | - Yuting Cui
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China
| | - Zhaofa Wu
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, 100871, China
- PKU-McGovern Institute for Brain Research, Beijing, 100871, China
| | - Qingchun Guo
- Chinese Institute for Brain Research, Beijing, 102206, China
- Capital Medical University, Beijing, 102206, China
| | - Haiyue Zou
- Chinese Institute for Brain Research, Beijing, 102206, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Wenting Wu
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China
- Peking University-Tsinghua University-NIBS Joint Graduate Program, NIBS, Beijing, 102206, China
| | - Xinwei Gao
- Chinese Institute for Brain Research, Beijing, 102206, China
| | - Zhongyang Qi
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China
| | - Yuqi Ren
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China
| | - Shu Wang
- Chinese Institute for Brain Research, Beijing, 102206, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, 100871, China
- PKU-McGovern Institute for Brain Research, Beijing, 100871, China
| | - Minmin Luo
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China.
- Graduate School of Peking Union Medical College, Beijing, 100730, China.
- Chinese Institute for Brain Research, Beijing, 102206, China.
- Tsinghua Institute of Multidisciplinary Biomedical Research (TIMBR), Beijing, 102206, China.
| |
Collapse
|
21
|
Liu Y, Li Z, Wu W, Wang Y, Zhao G, Liu Y, Liu J, Song Z. UVR Promotes Keratinocyte Phagocytosis and Skin Pigmentation Through TRPA1 Channels. CLINICAL, COSMETIC AND INVESTIGATIONAL DERMATOLOGY 2022; 15:1183-1193. [PMID: 35784271 PMCID: PMC9249384 DOI: 10.2147/ccid.s365682] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/15/2022] [Indexed: 11/23/2022]
Affiliation(s)
- Ying Liu
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Zhou Li
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Wei Wu
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Yupeng Wang
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Guangming Zhao
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Yuejian Liu
- Central Laboratory, First Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Jing Liu
- Stem Cell Clinical Research Center, First Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Zhiqi Song
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
- Correspondence: Zhiqi Song, Department of Dermatology, First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Road, Dalian, 116011, People’s Republic of China, Email
| |
Collapse
|
22
|
Camponogara C, Oliveira SM. Are TRPA1 and TRPV1 channel-mediated signalling cascades involved in UVB radiation-induced sunburn? ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2022; 92:103836. [PMID: 35248760 DOI: 10.1016/j.etap.2022.103836] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/09/2022] [Accepted: 02/28/2022] [Indexed: 06/14/2023]
Abstract
Burn injuries are underappreciated injuries associated with substantial morbidity and mortality. Overexposure to ultraviolet (UV) radiation has dramatic clinical effects in humans and is a significant public health concern. Although the mechanisms underlying UVB exposure are not fully understood, many studies have made substantial progress in the pathophysiology of sunburn in terms of its molecular aspects in the last few years. It is well established that the transient receptor potential ankyrin 1 (TRPA1), and vanilloid 1 (TRPV1) channels modulate the inflammatory, oxidative, and proliferative processes underlying UVB radiation exposure. However, it is still unknown which mechanisms underlying TRPV1/A1 channel activation are elicited in sunburn induced by UVB radiation. Therefore, in this review, we give an overview of the TRPV1/A1 channel-mediated signalling cascades that may be involved in the pathophysiology of sunburn induced by UVB radiation. These data will undoubtedly help to explain the various features of sunburn and contribute to the development of novel therapeutic approaches to better treat it.
Collapse
Affiliation(s)
- Camila Camponogara
- Graduated Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Sara Marchesan Oliveira
- Graduated Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, RS, Brazil; Department of Biochemistry and Molecular Biology, Centre of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil.
| |
Collapse
|
23
|
TRPA1 promotes melanosome phagocytosis in keratinocytes via PAR-2/CYLD axis. J Dermatol Sci 2022; 106:181-188. [DOI: 10.1016/j.jdermsci.2022.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 05/06/2022] [Accepted: 05/15/2022] [Indexed: 11/23/2022]
|
24
|
Landini L, Souza Monteiro de Araujo D, Titiz M, Geppetti P, Nassini R, De Logu F. TRPA1 Role in Inflammatory Disorders: What Is Known So Far? Int J Mol Sci 2022; 23:ijms23094529. [PMID: 35562920 PMCID: PMC9101260 DOI: 10.3390/ijms23094529] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/11/2022] [Accepted: 04/18/2022] [Indexed: 02/01/2023] Open
Abstract
The transient receptor potential ankyrin 1 (TRPA1), a member of the TRP superfamily of channels, is primarily localized in a subpopulation of primary sensory neurons of the trigeminal, vagal, and dorsal root ganglia, where its activation mediates neurogenic inflammatory responses. TRPA1 expression in resident tissue cells, inflammatory, and immune cells, through the indirect modulation of a large series of intracellular pathways, orchestrates a range of cellular processes, such as cytokine production, cell differentiation, and cytotoxicity. Therefore, the TRPA1 pathway has been proposed as a protective mechanism to detect and respond to harmful agents in various pathological conditions, including several inflammatory diseases. Specific attention has been paid to TRPA1 contribution to the transition of inflammation and immune responses from an early defensive response to a chronic pathological condition. In this view, TRPA1 antagonists may be regarded as beneficial tools for the treatment of inflammatory conditions.
Collapse
|
25
|
Presence of TRPA1 Modifies CD4+/CD8+ T Lymphocyte Ratio and Activation. Pharmaceuticals (Basel) 2022; 15:ph15010057. [PMID: 35056114 PMCID: PMC8781558 DOI: 10.3390/ph15010057] [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: 11/24/2021] [Revised: 12/24/2021] [Accepted: 12/28/2021] [Indexed: 12/10/2022] Open
Abstract
Transient Receptor Potential Ankyrin 1 (TRPA1) has been reported to influence neuroinflammation and lymphocyte function. We analysed the immune phenotype and activation characteristics of TRPA1-deficient mice (knockout—KO) generated by targeted deletion of the pore-loop domain of the ion channel. We compared TRPA1 mRNA and protein expression in monocyte and lymphocyte subpopulations isolated from primary and secondary lymphatic organs of wild type (WT) and KO mice. qRT-PCR and flow cytometric studies indicated a higher level of TRPA1 in monocytes than in lymphocytes, but both were orders of magnitude lower than in sensory neurons. We found lower CD4+/CD8+ thymocyte ratios, diminished CD4/CD8 rates, and B cell numbers in the KO mice. Early activation marker CD69 was lower in CD4+ T cells of KO, while the level of CD8+/CD25+ cells was higher. In vitro TcR-mediated activation did not result in significant differences in CD69 level between WT and KO splenocytes, but lower cytokine (IL-1β, IL-6, TNF-α, IL-17A, IL-22, and RANTES) secretion was observed in KO splenocytes. Basal intracellular Ca2+ level and TcR-induced Ca2+ signal in T lymphocytes did not differ significantly, but interestingly, imiquimod-induced Ca2+ level in KO thymocytes was higher. Our results support the role of TRPA1 in the regulation of activation, cytokine production, and T and B lymphocytes composition in mice.
Collapse
|
26
|
The TRPA1 Channel Amplifies the Oxidative Stress Signal in Melanoma. Cells 2021; 10:cells10113131. [PMID: 34831352 PMCID: PMC8624842 DOI: 10.3390/cells10113131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/09/2021] [Accepted: 11/09/2021] [Indexed: 11/16/2022] Open
Abstract
Macrophages (MΦs) and reactive oxygen species (ROS) are implicated in carcinogenesis. The oxidative stress sensor, transient receptor potential ankyrin 1 (TRPA1), activated by ROS, appears to contribute to lung and breast cancer progression. Although TRPA1 expression has been reported in melanoma cell lines, and oxidative stress has been associated with melanocytic transformation, their role in melanoma remains poorly known. Here, we localized MΦs, the final end-product of oxidative stress, 4-hydroxynonenal (4-HNE), and TRPA1 in tissue samples of human common dermal melanocytic nevi, dysplastic nevi, and thin (pT1) and thick (pT4) cutaneous melanomas. The number (amount) of intratumoral and peritumoral M2 MΦs and 4-HNE staining progressively increased with tumor severity, while TRPA1 expression was similar in all samples. Hydrogen peroxide (H2O2) evoked a TRPA1-dependent calcium response in two distinct melanoma cell lines (SK-MEL-28 and WM266-4). Furthermore, H2O2 induced a TRPA1-dependent H2O2 release that was prevented by the TRPA1 antagonist, A967079, or Trpa1 gene silencing (siRNA). ROS release from infiltrating M2 MΦs may target TRPA1-expressing melanoma cells to amplify the oxidative stress signal that affects tumor cell survival and proliferation.
Collapse
|
27
|
Naert R, López-Requena A, Talavera K. TRPA1 Expression and Pathophysiology in Immune Cells. Int J Mol Sci 2021; 22:ijms222111460. [PMID: 34768891 PMCID: PMC8583806 DOI: 10.3390/ijms222111460] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/22/2021] [Accepted: 10/22/2021] [Indexed: 12/18/2022] Open
Abstract
The non-selective cation channel TRPA1 is best known as a broadly-tuned sensor expressed in nociceptive neurons, where it plays key functions in chemo-, thermo-, and mechano-sensing. However, in this review we illustrate how this channel is expressed also in cells of the immune system. TRPA1 has been detected, mainly with biochemical techniques, in eosinophils, mast cells, macrophages, dendritic cells, T cells, and B cells, but not in neutrophils. Functional measurements, in contrast, remain very scarce. No studies have been reported in basophils and NK cells. TRPA1 in immune cells has been linked to arthritis (neutrophils), anaphylaxis and atopic dermatitis (mast cells), atherosclerosis, renal injury, cardiac hypertrophy and inflammatory bowel disease (macrophages), and colitis (T cells). The contribution of TRPA1 to immunity is dual: as detector of cell stress, tissue injury, and exogenous noxious stimuli it leads to defensive responses, but in conditions of aberrant regulation it contributes to the exacerbation of inflammatory conditions. Future studies should aim at characterizing the functional properties of TRPA1 in immune cells, an essential step in understanding its roles in inflammation and its potential as therapeutic target.
Collapse
Affiliation(s)
- Robbe Naert
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; (R.N.); (A.L.-R.)
| | - Alejandro López-Requena
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; (R.N.); (A.L.-R.)
- Ablynx, Technologiepark 21, 9052 Zwijnaarde, Belgium
| | - Karel Talavera
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; (R.N.); (A.L.-R.)
- Correspondence: ; Tel.: +32-16-330469
| |
Collapse
|
28
|
de Assis LVM, Moraes MN, Mendes D, Silva MM, Menck CFM, Castrucci AMDL. Loss of Melanopsin (OPN4) Leads to a Faster Cell Cycle Progression and Growth in Murine Melanocytes. Curr Issues Mol Biol 2021; 43:1436-1450. [PMID: 34698095 PMCID: PMC8929055 DOI: 10.3390/cimb43030101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/14/2021] [Accepted: 09/26/2021] [Indexed: 12/13/2022] Open
Abstract
Skin melanocytes harbor a complex photosensitive system comprised of opsins, which were shown, in recent years, to display light- and thermo-independent functions. Based on this premise, we investigated whether melanopsin, OPN4, displays such a role in normal melanocytes. In this study, we found that murine Opn4KO melanocytes displayed a faster proliferation rate compared to Opn4WT melanocytes. Cell cycle population analysis demonstrated that OPN4KO melanocytes exhibited a faster cell cycle progression with reduced G0–G1, and highly increased S and slightly increased G2/M cell populations compared to the Opn4WT counterparts. Expression of specific cell cycle-related genes in Opn4KO melanocytes exhibited alterations that corroborate a faster cell cycle progression. We also found significant modification in gene and protein expression levels of important regulators of melanocyte physiology. PER1 protein level was higher while BMAL1 and REV-ERBα decreased in Opn4KO melanocytes compared to Opn4WT cells. Interestingly, the gene expression of microphthalmia-associated transcription factor (MITF) was upregulated in Opn4KO melanocytes, which is in line with a higher proliferative capability. Taken altogether, we demonstrated that OPN4 regulates cell proliferation, cell cycle, and affects the expression of several important factors of the melanocyte physiology; thus, arguing for a putative tumor suppression role in melanocytes.
Collapse
Affiliation(s)
- Leonardo Vinícius Monteiro de Assis
- Laboratory of Comparative Physiology of Pigmentation, Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo 05508-090, Brazil; (M.N.M.); (A.M.d.L.C.)
- Correspondence:
| | - Maria Nathália Moraes
- Laboratory of Comparative Physiology of Pigmentation, Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo 05508-090, Brazil; (M.N.M.); (A.M.d.L.C.)
| | - Davi Mendes
- DNA Repair Lab, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (D.M.); (M.M.S.); (C.F.M.M.)
| | - Matheus Molina Silva
- DNA Repair Lab, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (D.M.); (M.M.S.); (C.F.M.M.)
| | - Carlos Frederico Martins Menck
- DNA Repair Lab, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (D.M.); (M.M.S.); (C.F.M.M.)
| | - Ana Maria de Lauro Castrucci
- Laboratory of Comparative Physiology of Pigmentation, Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo 05508-090, Brazil; (M.N.M.); (A.M.d.L.C.)
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| |
Collapse
|
29
|
Hsu WL, Noda M, Yoshioka T, Ito E. A novel strategy for treating cancer: understanding the role of Ca2+ signaling from nociceptive TRP channels in regulating cancer progression. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:401-415. [PMID: 36045706 PMCID: PMC9400763 DOI: 10.37349/etat.2021.00053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/12/2021] [Indexed: 11/19/2022] Open
Abstract
Cancer is an aging-associated disease and caused by genomic instability that is driven by the accumulation of mutations and epimutations in the aging process. Although Ca2+ signaling, reactive oxygen species (ROS) accumulation, DNA damage response (DDR) and senescence inflammation response (SIR) are processed during genomic instability, the underlying mechanism for the cause of genomic instability and cancer development is still poorly understood and needs to be investigated. Nociceptive transient receptor potential (TRP) channels, which firstly respond to environmental stimuli, such as microbes, chemicals or physical injuries, potentiate regulation of the aging process by Ca2+ signaling. In this review, the authors provide an explanation of the dual role of nociceptive TRP channels in regulating cancer progression, initiating cancer progression by aging-induced genomic instability, and promoting malignancy by epigenetic regulation. Thus, therapeutically targeting nociceptive TRP channels seems to be a novel strategy for treating cancers.
Collapse
Affiliation(s)
- Wen-Li Hsu
- Department of Dermatology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80145, Taiwan; Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Mami Noda
- Laboratory of Pathophysiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Tohru Yoshioka
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Graduate Institute of Medicine, School of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Etsuro Ito
- Graduate Institute of Medicine, School of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Waseda Research Institute for Science and Engineering, Waseda University, Tokyo 162-8480, Japan; Department of Biology, Waseda University, Tokyo 162-8480, Japan
| |
Collapse
|
30
|
Jia Q, Tian W, Li B, Chen W, Zhang W, Xie Y, Cheng N, Chen Q, Xiao J, Zhang Y, Yang J, Wang S. TRPV1 and TRPA1 in melanocytes synergize UV-dependent and UV-independent melanogenesis. Br J Pharmacol 2021; 178:4646-4662. [PMID: 34363226 DOI: 10.1111/bph.15643] [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: 01/25/2021] [Revised: 07/18/2021] [Accepted: 07/21/2021] [Indexed: 10/20/2022] Open
Abstract
BACKGROUND AND PURPOSE Melanogenesis is essential for pigmentation, and deregulated melanogenesis causes pigmentary diseases. PUVA therapy (psoralen plus ultraviolet A, UVA) strongly stimulates pigmentation, but the underlying molecular mechanisms are elusive. EXPERIMENTAL APPROACH Melanin content of cultured human melanocytes was spectrophotometrically measured. Patch-clamp recordings were made in human melanocytes or HEK 293 cells transiently expressing wild type or mutant human TRPV1 and TRPA1 channels. Endogenous expression of TRPV1 and TRPA1 in melanocytes was analyzed by western blotting and was knocked down with siRNA. In vivo pigmentary responses were measured by a colorimeter in mouse ear skin. The expression of TRPV1 and TRPA1 in human pigmented lesions was examined by immunohistochemical staining. KEY RESULTS PUVA strongly stimulated melanogenesis, and PUVA-induced TRPV1 and TRPA1 channel activation in melanocytes and the resulting Ca2+ influx were required for the stimulated melanogenesis both in vitro and in vivo. Agonists-induced TRPV1 and TRPA1 activation alone did not stimulate melanogenesis, but it synergized UVA or intrinsic cAMP and NO signaling pathways to stimulate UV-dependent or UV-independent melanogenesis. Moreover, the expressions of TRPV1 and TRPA1 were increased in human melanocytic lesions, and inhibition of both channels decreased melanin content in melanoma cells. CONCLUSION AND IMPLICATIONS TRPV1 and TRPA1 are key molecular sensors and enhancers of extrinsic and intrinsic melanogenic signals in both physiological and pathological conditions, and activation of both channels in melanocytes contributes to PUVA therapy-induced pigmentation. Our work provides a common mechanism of melanogenic regulation and highlights TRPV1 and TRPA1 as potential therapeutic targets for pigmentary disorders.
Collapse
Affiliation(s)
- Qi Jia
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China.,Department of Orthopedic Oncology, Changzheng Hospital, Navy Medical University, Shanghai, China
| | - Weifeng Tian
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, and Ion Channel Research and Drug Development Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Binbin Li
- Department of Pathology, Changzheng Hospital, Navy Medical University, Shanghai, China
| | - Wen Chen
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Wenjie Zhang
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yang Xie
- Department of Dermatology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Na Cheng
- Department of Pathology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qi Chen
- Department of Biostatistics, Navy Medical University, Shanghai, China
| | - Jianru Xiao
- Department of Orthopedic Oncology, Changzheng Hospital, Navy Medical University, Shanghai, China
| | - Yiwang Zhang
- Department of Pathology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jian Yang
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Shu Wang
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
31
|
How does the skin sense sun light? An integrative view of light sensing molecules. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY C-PHOTOCHEMISTRY REVIEWS 2021. [DOI: 10.1016/j.jphotochemrev.2021.100403] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
32
|
Molecular and Biochemical Basis of Minocycline-Induced Hyperpigmentation-The Study on Normal Human Melanocytes Exposed to UVA and UVB Radiation. Int J Mol Sci 2021; 22:ijms22073755. [PMID: 33916535 PMCID: PMC8038496 DOI: 10.3390/ijms22073755] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/01/2021] [Accepted: 04/01/2021] [Indexed: 01/04/2023] Open
Abstract
Minocycline is a drug which induces skin hyperpigmentation. Its frequency reaches up to 50% of treated patients. The adverse effect diminishes the great therapeutic potential of minocycline, including antibacterial, neuroprotective, anti-inflammatory and anti-cancer actions. It is supposed that an elevated melanin level and drug accumulation in melanin-containing cells are related to skin hyperpigmentation. This study aimed to evaluate molecular and biochemical mechanism of minocycline-induced hyperpigmentation in human normal melanocytes, as well as the contribution of UV radiation to this side effect. The experiments involved the evaluation of cyto- and phototoxic potential of the drug using cell imaging with light and confocal microscopes as well as biochemical and molecular analysis of melanogenesis. We showed that minocycline induced melanin synthesis in epidermal melanocytes. The action was intensified by UV irradiation, especially with the UVB spectrum. Minocycline stimulated the expression of microphthalmia-associated transcription factor (MITF) and tyrosinase (TYR) gene. Higher levels of melanin and increased activity of tyrosinase were also observed in treated cells. Moreover, minocycline triggered the supranuclear accumulation of tyrosinase, similar to UV radiation. The decreased level of premelanosome protein PMEL17 observed in all minocycline-treated cultures suggests disorder of the formation, maturation or distribution of melanosomes. The study revealed that minocycline itself was able to enhance melanin synthesis. The action was intensified by irradiation, especially with the UVB spectrum. Demonstrated results confirmed the potential role of melanin and UV radiation minocycline-induced skin hyperpigmentation.
Collapse
|
33
|
Mechanisms of Broad-Band UVB Irradiation‒Induced Itch in Mice. J Invest Dermatol 2021; 141:2499-2508.e3. [PMID: 33812858 DOI: 10.1016/j.jid.2021.03.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 03/02/2021] [Accepted: 03/09/2021] [Indexed: 11/24/2022]
Abstract
Although sunburn can produce severe uncontrollable itching, the underlying mechanisms of UV irradiation‒induced itch are poorly understood because of a lack of experimental animal models of sunburn itch. In this study, we established a sunburn-related mouse model and found that broad-band UVB irradiation elicited scratching but not wiping behavior in mice. Using a combination of live-cell calcium ion imaging and quantitative RT-PCR on dorsal root ganglion neurons, H&E staining, immunofluorescence staining of skin preparations, and behavioral testing, in combination with genetic and pharmacological approaches, we showed that TRPV1-positive dorsal root ganglion neurons but not mast cells are involved in broad-band UVB irradiation‒induced itch. Moreover, both genetic and pharmacological inhibition of TRPV1 function significantly alleviated the broad-band UVB irradiation‒induced itch response. Collectively, our results suggest that broad-band UVB irradiation evokes itch sensation in mice by promoting TRPV1 channel function in dorsal root ganglion neurons and provide potential therapeutic targets for sunburn-related itch.
Collapse
|
34
|
Luostarinen S, Hämäläinen M, Moilanen E. Transient Receptor Potential Ankyrin 1 (TRPA1)-An Inflammation-Induced Factor in Human HaCaT Keratinocytes. Int J Mol Sci 2021; 22:ijms22073322. [PMID: 33805042 PMCID: PMC8037497 DOI: 10.3390/ijms22073322] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/10/2021] [Accepted: 03/17/2021] [Indexed: 12/13/2022] Open
Abstract
Transient receptor potential ankyrin 1 (TRPA1) is an ion channel mainly studied in sensory neurons where it mediates itch, pain and neurogenic inflammation. Recently, some nonneuronal cells have also been shown to express TRPA1 to support inflammatory responses. To address the role of TRPA1 in skin inflammation, we aimed to investigate TRPA1 expression in keratinocytes. HaCaT cells (a model of human keratinocytes) and skin biopses from wild-type and TRPA1 deficient mice were used in the studies. TRPA1 expression in nonstimulated keratinocytes was very low but significantly inducible by the proinflammatory cytokine tumor necrosis factor (TNF) in an nuclear factor kappa B (NF-κB), and mitogen-activated protein (MAP) kinase (p38 and c-Jun N-terminal kinase, JNK)-dependent manner. Interestingly, drugs widely used to treat skin inflammation, the calcineurin inhibitors tacrolimus and cyclosporine and the glucocorticoid dexamethasone, significantly decreased TRPA1 expression. Furthermore, pharmacological inhibition and genetic deletion of TRPA1 reduced the synthesis of TNF-induced monocyte chemoattractant protein 1 (MCP-1) in keratinocytes and mouse skin biopsies. In conclusion, these findings point to an inflammatory role for TRPA1 in keratinocytes and present TRPA1 as a potential drug target in inflammatory skin diseases.
Collapse
|
35
|
Gating of the capsaicin receptor TRPV1 by UVA-light and oxidants are mediated by distinct mechanisms. Cell Calcium 2021; 96:102391. [PMID: 33752082 DOI: 10.1016/j.ceca.2021.102391] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 02/22/2021] [Accepted: 03/09/2021] [Indexed: 02/07/2023]
Abstract
Redox-sensitivity is a common property of several transient receptor potential (TRP) ion channels. Oxidants and UVA-light activate TRPV2 by oxidizing methionine pore residues which are conserved in the capsaicin-receptor TRPV1. However, the redox-sensitivity of TRPV1 is regarded to depend on intracellular cysteine residues. In this study we examined if TRPV1 is gated by UVA-light, and if the conserved methionine residues are relevant for redox-sensitivity of TRPV1. Patch clamp recordings were performed to explore wildtype (WT) and mutants of human TRPV1 (hTRPV1). UVA-light induced hTRPV1-mediated membrane currents and potentiated both proton- and heat-evoked currents. The reducing agent dithiothreitol (DTT) prevented and partially reversed UVA-light induced sensitization of hTRPV1. UVA-light induced sensitization was reduced in the mutant hTRPV1-C158A/C387S/C767S (hTRPV1-3C). The remaining sensitivity to UVA-light of hTRRPV1-3C was not further reduced upon exchange of the methionine residues M568 and M645. While UVA-induced sensitization was reduced in the protein kinase C-insensitive mutant hTRPV1-S502A/S801A, the PKC-inhibitors chelerythrine chloride, staurosporine and Gö6976 did not reduce UVA-induced effects on hTRPV1-WT. While hTRPV1-3C was insensitive to the cysteine-selective oxidant diamide, it displayed a residual sensitivity to H2O2 and chloramine-T. However, the exchange of M568 and M645 in hTRPV1-3C did not further reduce these effects. Our data demonstrate that oxidants and UVA-light gate hTRPV1 by cysteine-dependent as well as cysteine-independent mechanisms. In contrast to TRPV2, the methionine residues 568 and 645 seem to be of limited relevance for redox-sensitivity of hTRPV1. Finally, UVA-light induced gating of hTRPV1 does not seem to require activation of protein kinase C.
Collapse
|
36
|
Maglie R, Souza Monteiro de Araujo D, Antiga E, Geppetti P, Nassini R, De Logu F. The Role of TRPA1 in Skin Physiology and Pathology. Int J Mol Sci 2021; 22:3065. [PMID: 33802836 PMCID: PMC8002674 DOI: 10.3390/ijms22063065] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/15/2021] [Accepted: 03/15/2021] [Indexed: 12/13/2022] Open
Abstract
The transient receptor potential ankyrin 1 (TRPA1), a member of the TRP superfamily of channels, acts as 'polymodal cellular sensor' on primary sensory neurons where it mediates the peripheral and central processing of pain, itch, and thermal sensation. However, the TRPA1 expression extends far beyond the sensory nerves. In recent years, much attention has been paid to its expression and function in non-neuronal cell types including skin cells, such as keratinocytes, melanocytes, mast cells, dendritic cells, and endothelial cells. TRPA1 seems critically involved in a series of physiological skin functions, including formation and maintenance of physico-chemical skin barriers, skin cells, and tissue growth and differentiation. TRPA1 appears to be implicated in mechanistic processes in various immunological inflammatory diseases and cancers of the skin, such as atopic and allergic contact dermatitis, psoriasis, bullous pemphigoid, cutaneous T-cell lymphoma, and melanoma. Here, we report recent findings on the implication of TRPA1 in skin physiology and pathophysiology. The potential use of TRPA1 antagonists in the treatment of inflammatory and immunological skin disorders will be also addressed.
Collapse
Affiliation(s)
- Roberto Maglie
- Department of Health Sciences, Section of Dermatology, University of Florence, 50139 Florence, Italy; (R.M.); (E.A.)
| | - Daniel Souza Monteiro de Araujo
- Department of Health Sciences, Clinical Pharmacology Unit, University of Florence, 50139 Florence, Italy; (D.S.M.d.A.); (P.G.); (F.D.L.)
| | - Emiliano Antiga
- Department of Health Sciences, Section of Dermatology, University of Florence, 50139 Florence, Italy; (R.M.); (E.A.)
| | - Pierangelo Geppetti
- Department of Health Sciences, Clinical Pharmacology Unit, University of Florence, 50139 Florence, Italy; (D.S.M.d.A.); (P.G.); (F.D.L.)
| | - Romina Nassini
- Department of Health Sciences, Clinical Pharmacology Unit, University of Florence, 50139 Florence, Italy; (D.S.M.d.A.); (P.G.); (F.D.L.)
| | - Francesco De Logu
- Department of Health Sciences, Clinical Pharmacology Unit, University of Florence, 50139 Florence, Italy; (D.S.M.d.A.); (P.G.); (F.D.L.)
| |
Collapse
|
37
|
Lan Y, Zeng W, Dong X, Lu H. Opsin 5 is a key regulator of ultraviolet radiation-induced melanogenesis in human epidermal melanocytes. Br J Dermatol 2021; 185:391-404. [PMID: 33400324 PMCID: PMC8453816 DOI: 10.1111/bjd.19797] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2021] [Indexed: 12/24/2022]
Abstract
Background Human skin, which is constantly exposed to solar ultraviolet radiation (UVR), has a unique ability to respond by increasing its pigmentation in a protective process driven by melanogenesis in human epidermal melanocytes (HEMs). However, the molecular mechanisms used by HEMs to detect and respond to UVR remain unclear. Objectives To investigate the function and potential mechanism of opsin 5 (OPN5), a photoreceptor responsive to UVR wavelengths, in melanogenesis in HEMs. Methods Melanin content in HEMs was determined using the NaOH method, and activity of tyrosinase (TYR) (a key enzyme in melanin synthesis) was determined by the l‐DOPA method. OPN5 expression in UVR‐treated vs. untreated HEMs and explant tissues was detected by reverse‐transcription quantitative polymerase chain reaction (RT‐qPCR), Western blotting and immunofluorescence. Short interfering RNA‐mediated OPN5 knockdown and a lentivirus OPN5 overexpression model were used to examine their respective effects on TYR, tyrosinase‐related protein 1 (TRP1), TRP2 and microphthalmia‐associated transcription factor (MITF) expression, under UVR. Changes in expression of TYR, TRP1 and TRP2 caused by changes in OPN5 expression level were detected by RT‐qPCR and Western blot. Furthermore, changes in signalling pathway proteins were assayed. Results We found that OPN5 is the key sensor in HEMs responsible for UVR‐induced melanogenesis. OPN5‐induced melanogenesis required Ca2+‐dependent G protein‐coupled receptor‐ and protein kinase C signal transduction, thus contributing to the UVR‐induced MITF response to mediate downstream cellular effects, and providing evidence of OPN5 function in mammalian phototransduction. Remarkably, OPN5 activation was necessary for UVR‐induced increase in cellular melanin and has an inherent function in melanocyte melanogenesis. Conclusions Our results provide insight into the molecular mechanisms of UVR sensing and phototransduction in melanocytes, and may reveal molecular targets for preventing pigmentation or pigment diseases.
What is already known about this topic?
Ultraviolet radiation (UVR) induces a protective response to DNA damage mediated by melanin synthesis in human epidermal melanocytes (HEMs). Tyrosinase (TYR), with tyrosinase‐related proteins (TRP1, TRP2), are the key enzymes for melanin synthesis. Microphthalmia‐associated transcription factor regulates key genes for melanocyte development and differentiation, and can stimulate melanogenesis by activating transcription of TYR and other pigmentation genes, including TRP1. Opsin 5 (OPN5) is known to function as a photoreceptor responsive to wavelengths in the near UV spectrum.
What does this study add?UVR induces melanogenesis in HEMs via OPN5. OPN5 regulates expression of TYR, TRP1 and TRP2 through the calcium‐dependent G protein‐coupled and protein kinase C signalling pathways. OPN5 has an inherent role in HEMs in mediating melanogenesis.
What is the translational message?OPN5 was discovered as a key sensor for UVR‐induced melanogenesis in human skin melanocytes. It could be a target for early treatment of pigmentation or pigment diseases, to provide a more personalized and economically feasible method.
Linked Comment: L.V.M. de Assis and A.M. de Lauro Castrucci. Br J Dermatol 2021; 185:249–250. Plain language summary available online
Collapse
Affiliation(s)
- Y Lan
- School of Public Health, Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - W Zeng
- Department of Immunology, Basic Medical School, Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - X Dong
- Department of Dermatology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550001, China
| | - H Lu
- School of Public Health, Guizhou Medical University, Guiyang, Guizhou, 550004, China.,Department of Immunology, Basic Medical School, Guizhou Medical University, Guiyang, Guizhou, 550004, China.,Department of Dermatology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550001, China
| |
Collapse
|
38
|
Woo JH, Nam DY, Kim HJ, Hong PTL, Kim WK, Nam JH. Nootkatol prevents ultraviolet radiation-induced photoaging via ORAI1 and TRPV1 inhibition in melanocytes and keratinocytes. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2021; 25:87-94. [PMID: 33361541 PMCID: PMC7756533 DOI: 10.4196/kjpp.2021.25.1.87] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/06/2020] [Accepted: 12/06/2020] [Indexed: 12/31/2022]
Abstract
Skin photoaging occurs due to chronic exposure to solar ultraviolet radiation (UV), the main factor contributing to extrinsic skin aging. Clinical signs of photoaging include the formation of deep, coarse skin wrinkles and hyperpigmentation. Although melanogenesis and skin wrinkling occur in different skin cells and have different underlying mechanisms, their initiation involves intracellular calcium signaling via calcium ion channels. The ORAI1 channel initiates melanogenesis in melanocytes, and the TRPV1 channel initiates MMP-1 production in keratinocytes in response to UV stimulation. We aimed to develop a drug that may simultaneously inhibit ORAI1 and TRPV1 activity to help prevent photoaging. We synthesized nootkatol, a chemical derivative of valencene. TRPV1 and ORAI1 activities were measured using the whole-cell patch-clamp technique. Intracellular calcium concentration [Ca2+]i was measured using calcium-sensitive fluorescent dye (Fura-2 AM). UV-induced melanin formation and MMP-1 production were quantified in B16F10 melanoma cells and HaCaT cells, respectively. Our results indicate that nootkatol (90 μM) reduced TRPV1 current by 94% ± 2% at –60 mV and ORAI1 current by 97% ± 1% at –120 mV. Intracellular calcium signaling was significantly inhibited by nootkatol in response to ORAI1 activation in human primary melanocytes (51.6% ± 0.98% at 100 μM). Additionally, UV-induced melanin synthesis was reduced by 76.38% ± 5.90% in B16F10 melanoma cells, and UV-induced MMP-1 production was reduced by 59.33% ± 1.49% in HaCaT cells. In conclusion, nootkatol inhibits both TRPV1 and ORAI1 to prevent photoaging, and targeting ion channels may be a promising strategy for preventing photoaging.
Collapse
Affiliation(s)
- Joo Han Woo
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Korea.,Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Korea
| | | | - Hyun Jong Kim
- Department of Internal Medicine, Graduate School of Medicine, Dongguk University, Goyang 10326, Korea
| | - Phan Thi Lam Hong
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Korea.,Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Korea
| | - Woo Kyung Kim
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Korea.,Department of Internal Medicine, Graduate School of Medicine, Dongguk University, Goyang 10326, Korea
| | - Joo Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Korea.,Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Korea
| |
Collapse
|
39
|
De Logu F, Ugolini F, Caporalini C, Palomba A, Simi S, Portelli F, Campanacci DA, Beltrami G, Massi D, Nassini R. TRPA1 Expression in Synovial Sarcoma May Support Neural Origin. Biomolecules 2020; 10:biom10101446. [PMID: 33076385 PMCID: PMC7602570 DOI: 10.3390/biom10101446] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 10/13/2020] [Indexed: 12/16/2022] Open
Abstract
Synovial sarcoma (SS) is a malignant mesenchymal soft tissue neoplasm. Despite its name, the cells of origin are not synovial cells, but rather neural, myogenic, or multipotent mesenchymal stem cells have been proposed as possible cells originators. Unlike other sarcomas, an unusual presentation of long-term pain at the tumor site has been documented, but the exact mechanisms have not been fully clarified yet. The transient receptor potential ankyrin 1 (TRPA1) is a nonselective cation channel mainly expressed in primary sensory neurons, where it functions as a pain sensor. TRPA1 have also been described in multiple non-excitable cells, including those derived from neural crest stem cells such as glial cells and, in particular, Schwann cell oligodendrocytes and astrocytes. We evaluated TRPA1 expression in SS. We selected a cohort of 41 SSs, and by immunohistochemistry, we studied TRPA1 expression. TRPA1 was found in 92.6% of cases. Triple TRPA1/pS100/SOX10 and TRPA1/SLUG/SNAIL staining strongly supports a neural origin of SS. TRPA1 positivity was also observed in a subset of cases negative with pS100, SOX10 and/or SLUG/SNAIL, and these divergent phenotypes may reflect a process of tumor plasticity and dedifferentiation of neural-derived SSs. Given the functional diversity of TRPA1 and its expression in neuronal and non-neuronal multipotent neural crest stem cells, it remains to be determined whether TRPA1 expression in SSs neoplastic cells plays a role in the molecular mechanism associated with premonitory pain symptoms and tumor progression.
Collapse
Affiliation(s)
- Francesco De Logu
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, 50139 Florence, Italy; (F.D.L.); (R.N.)
| | - Filippo Ugolini
- Section of Pathological Anatomy, Department of Health Sciences, University of Florence, 50139 Florence, Italy; (F.U.); (A.P.); (S.S.); (F.P.)
| | | | - Annarita Palomba
- Section of Pathological Anatomy, Department of Health Sciences, University of Florence, 50139 Florence, Italy; (F.U.); (A.P.); (S.S.); (F.P.)
| | - Sara Simi
- Section of Pathological Anatomy, Department of Health Sciences, University of Florence, 50139 Florence, Italy; (F.U.); (A.P.); (S.S.); (F.P.)
| | - Francesca Portelli
- Section of Pathological Anatomy, Department of Health Sciences, University of Florence, 50139 Florence, Italy; (F.U.); (A.P.); (S.S.); (F.P.)
| | - Domenico Andrea Campanacci
- Orthopedics and Traumatology Section, Department of Health Sciences, University of Florence, 50139 Florence, Italy; (D.A.C.); (G.B.)
| | - Giovanni Beltrami
- Orthopedics and Traumatology Section, Department of Health Sciences, University of Florence, 50139 Florence, Italy; (D.A.C.); (G.B.)
| | - Daniela Massi
- Section of Pathological Anatomy, Department of Health Sciences, University of Florence, 50139 Florence, Italy; (F.U.); (A.P.); (S.S.); (F.P.)
- Correspondence: ; Tel.: +39-055-794-9082
| | - Romina Nassini
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, 50139 Florence, Italy; (F.D.L.); (R.N.)
| |
Collapse
|
40
|
Souza Monteiro de Araujo D, Nassini R, Geppetti P, De Logu F. TRPA1 as a therapeutic target for nociceptive pain. Expert Opin Ther Targets 2020; 24:997-1008. [PMID: 32838583 PMCID: PMC7610834 DOI: 10.1080/14728222.2020.1815191] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction Chronic pain affects approximatively 30–50% of the population globally. Pathologies such as migraine, diabetic neuropathy, nerve injury and treatment with chemotherapeutic agents, can induce chronic pain. Members of the transient receptor potential (TRP) channels, including the TRP ankyrin 1 (TRPA1), have a major role in pain. Areas covered We focus on TRPA1 as a therapeutic target for pain relief. The structure, localization, and activation of the channel and its implication in different pathways to signal pain are described. This paper underlines the role of pharmacological interventions on TRPA1 to reduce pain in numerous pain conditions. We conducted a literature search in PubMed up to and including July 2020. Expert opinion Our understanding of the molecular mechanisms underlying the sensitization of central and peripheral nociceptive pathways is limited. Preclinical evidence indicates that, in murine models of pain diseases, numerous mechanisms converge on the pathway that encompasses oxidative stress and Schwann cell TRPA1 to sustain chronic pain. Programs to identify and develop treatments to attenuate TRPA1-mediated chronic pain have emerged from this knowledge. Antagonists explored as a novel class of analgesics have a new and promising target in the TRPA1 expressed by peripheral glial cells.
Collapse
Affiliation(s)
| | - Romina Nassini
- Department of Health Sciences, Clinical Pharmacology Unit, University of Florence , Florence, Italy
| | - Pierangelo Geppetti
- Department of Health Sciences, Clinical Pharmacology Unit, University of Florence , Florence, Italy
| | - Francesco De Logu
- Department of Health Sciences, Clinical Pharmacology Unit, University of Florence , Florence, Italy
| |
Collapse
|
41
|
Dumbuya H, Hafez SY, Oancea E. Cross talk between calcium and ROS regulate the UVA-induced melanin response in human melanocytes. FASEB J 2020; 34:11605-11623. [PMID: 32658369 DOI: 10.1096/fj.201903024r] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 06/01/2020] [Accepted: 06/15/2020] [Indexed: 12/31/2022]
Abstract
Exposure to high doses of solar long wavelength ultraviolet radiation (UVA) damages human skin via reactive oxygen species (ROS). Whether physiological UVA doses also generate ROS that has an effect on the skin remains unknown. We previously showed that in human epidermal melanocytes UVA activates a G-protein coupled signaling pathway that leads to calcium mobilization and increased melanin. Here, we report that ROS generated by the UVA phototransduction pathway are critical cellular messengers required to augment melanin. Using simultaneous UVA exposure and live-cell imaging of primary human melanocytes, we found that physiological doses of UVA generate two spatiotemporally distinct sources of ROS: one upstream of the G-protein activation that potentiates calcium responses, and another source downstream of calcium, in the mitochondria (ROSmito ). UVA-evoked signaling led to mitochondrial calcium uptake via mitochondrial calcium uniporter to promote ROSmito production leading to melanin synthesis. Our findings reveal a novel mechanism in which ROS function as signaling messengers necessary for melanin production, thus having a protective role in the UVA-induced skin response.
Collapse
Affiliation(s)
- Hawasatu Dumbuya
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, RI, USA
| | - Salwa Y Hafez
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, RI, USA.,College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
| | - Elena Oancea
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, RI, USA
| |
Collapse
|
42
|
Wu Q, Xia Y, Dai K, Bai P, Kwan KKL, Guo MSS, Dong TTX, Tsim KWK. Solar light induces the release of acetylcholine from skin keratinocytes affecting melanogenesis. FASEB J 2020; 34:8941-8958. [PMID: 32519787 DOI: 10.1096/fj.202000708r] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/15/2020] [Accepted: 05/19/2020] [Indexed: 12/26/2022]
Abstract
Cholinergic system conducts signal transmission in brain and muscle. Besides nervous system, the nonneuronal functions of cholinergic system have been proposed in various tissues. The expression of cholinergic proteins and release of acetylcholine in human skin have been reported, but its mechanism and influence on dermatological functions is not elucidated. Here, the expression profile of cholinergic markers was further investigated in skin and keratinocyte. The expression levels of choline acetyltransferase (ChAT), acetylcholinesterase (AChE), vesicular acetylcholine transporter (VAChT), and synaptophysin, were upregulated during differentiation of keratinocytes. In cultured keratinocytes, a transient exposure of solar light induced the release of acetylcholine, which was mediated by intracellular Ca2+ mobilization. The light-induced acetylcholine release was mediated by the present of opsin. The light-induced melanogenesis was inhibited by acetylcholine or AChE inhibitor in melanocyte in vitro and mouse skin ex vivo. These results indicated that the potential role of cholinergic system could be a negative regulator in skin pigmentation.
Collapse
Affiliation(s)
- Qiyun Wu
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, Shenzhen Research Institute, Shenzhen, China.,Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yiteng Xia
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Kun Dai
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, Shenzhen Research Institute, Shenzhen, China.,Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Panzhu Bai
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, Shenzhen Research Institute, Shenzhen, China.,Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Kenneth K L Kwan
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Maggie S S Guo
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, Shenzhen Research Institute, Shenzhen, China
| | - Tina T X Dong
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, Shenzhen Research Institute, Shenzhen, China.,Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Karl W K Tsim
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, Shenzhen Research Institute, Shenzhen, China.,Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China
| |
Collapse
|
43
|
Fialho MFP, Brum EDS, Pegoraro NS, Couto ACG, Trevisan G, Cruz L, Oliveira SM. Topical transient receptor potential ankyrin 1 antagonist treatment attenuates nociception and inflammation in an ultraviolet B radiation-induced burn model in mice. J Dermatol Sci 2020; 97:135-142. [PMID: 31982303 DOI: 10.1016/j.jdermsci.2020.01.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 01/07/2020] [Accepted: 01/14/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Ultraviolet B (UVB) radiation exposure promotes sunburn and thereby acute and chronic inflammatory processes, contributing to pain development and maintenance. New therapeutic alternatives are necessary because typical treatments can cause adverse effects. An attractive alternative would be to target the transient receptor potential ankyrin 1 (TRPA1), a calcium-permeable, non-selective cation channel, which is involved in a variety of inflammatory pain models. OBJECTIVE Evaluate the peripheral participation of TRPA1 using a topical treatment (HC030031 gel formulation; a selective TRPA1 antagonist) in nociception and inflammation caused by a UVB radiation-induced burn model in male mice (25-30 g). METHODS The mice were anaesthetised, and just the right hind paw was exposed to UVB radiation (0.75 J/cm2). Topical treatments were applied immediately after irradiation and once a day for 8 days. RESULTS HC030031 gel presented suitable pH and spreadability factor, ensuring its quality and the therapeutic effect. HC030031 0.05 % reversed UVB-induced mechanical and cold allodynia, with maximum inhibition (Imax) of 69 ± 13 % and 100 % (on day 4), respectively. HC030031 0.05 % also reduced the paw edema and MPO activity, with Imax of 77 ± 6 % (on day 5) and 69 ± 28 %, respectively. Likewise, UVB radiation increased the H2O2 levels (a TRPA1 agonist) and the Ca2+ influx in mice spinal cord synaptosomes. UVB radiation-induced Ca2+ influx was reduced by HC030031. CONCLUSION These findings confirm the activation of the TRPA1 channel by UVB radiation, suggesting that topical TRPA1 antagonists can be a new strategy for the adjuvant treatment of sunburn-associated pain and inflammation.
Collapse
Affiliation(s)
- Maria Fernanda Pessano Fialho
- Neurotoxicity and Psychopharmacology Laboratory, Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Evelyne da Silva Brum
- Neurotoxicity and Psychopharmacology Laboratory, Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Natháli Schopf Pegoraro
- Neurotoxicity and Psychopharmacology Laboratory, Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Ana Claudia Gontijo Couto
- Institute of Genetics and Biochemistry, Graduate Program in Genetics and Biochemistry, Federal University of Uberlandia, Uberlandia, MG, Brazil
| | - Gabriela Trevisan
- Graduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Letícia Cruz
- Graduate Program in Pharmaceutical Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Sara Marchesan Oliveira
- Neurotoxicity and Psychopharmacology Laboratory, Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, RS, Brazil.
| |
Collapse
|
44
|
Olinski LE, Lin EM, Oancea E. Illuminating insights into opsin 3 function in the skin. Adv Biol Regul 2020; 75:100668. [PMID: 31653550 PMCID: PMC7059126 DOI: 10.1016/j.jbior.2019.100668] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 09/24/2019] [Accepted: 09/30/2019] [Indexed: 12/21/2022]
Abstract
Because sunlight is essential for human survival, we have developed complex mechanisms for detecting and responding to light stimuli. The eyes and skin are major organs for sensing light and express several light-sensitive opsin receptors. These opsins mediate cellular responses to spectrally-distinct wavelengths of visible and ultraviolet light. How the eyes mediate visual phototransduction is well understood, but less is known about how the skin detects light. Both human and murine skin express a wide array of opsins, with one of the most highly expressed being the functionally elusive opsin 3 (OPN3). In this review we explore light reception, opsin expression and signaling in skin cells; we compile data elucidating potential functions for human OPN3 in skin, with emphasis on recent studies investigating OPN3 regulation of melanin within epidermal melanocytes.
Collapse
Affiliation(s)
- Lauren E Olinski
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence RI, 02912, USA.
| | - Erica M Lin
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence RI, 02912, USA
| | - Elena Oancea
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence RI, 02912, USA.
| |
Collapse
|
45
|
Oxidation of methionine residues activates the high-threshold heat-sensitive ion channel TRPV2. Proc Natl Acad Sci U S A 2019; 116:24359-24365. [PMID: 31719194 DOI: 10.1073/pnas.1904332116] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Thermosensitive transient receptor potential (TRP) ion channels detect changes in ambient temperature to regulate body temperature and temperature-dependent cellular activity. Rodent orthologs of TRP vanilloid 2 (TRPV2) are activated by nonphysiological heat exceeding 50 °C, and human TRPV2 is heat-insensitive. TRPV2 is required for phagocytic activity of macrophages which are rarely exposed to excessive heat, but what activates TRPV2 in vivo remains elusive. Here we describe the molecular mechanism of an oxidation-induced temperature-dependent gating of TRPV2. While high concentrations of H2O2 induce a modest sensitization of heat-induced inward currents, the oxidant chloramine-T (ChT), ultraviolet A light, and photosensitizing agents producing reactive oxygen species (ROS) activate and sensitize TRPV2. This oxidation-induced activation also occurs in excised inside-out membrane patches, indicating a direct effect on TRPV2. The reducing agent dithiothreitol (DTT) in combination with methionine sulfoxide reductase partially reverses ChT-induced sensitization, and the substitution of the methionine (M) residues M528 and M607 to isoleucine almost abolishes oxidation-induced gating of rat TRPV2. Mass spectrometry on purified rat TRPV2 protein confirms oxidation of these residues. Finally, macrophages generate TRPV2-like heat-induced inward currents upon oxidation and exhibit reduced phagocytosis when exposed to the TRP channel inhibitor ruthenium red (RR) or to DTT. In summary, our data reveal a methionine-dependent redox sensitivity of TRPV2 which may be an important endogenous mechanism for regulation of TRPV2 activity and account for its pivotal role for phagocytosis in macrophages.
Collapse
|
46
|
Talavera K, Startek JB, Alvarez-Collazo J, Boonen B, Alpizar YA, Sanchez A, Naert R, Nilius B. Mammalian Transient Receptor Potential TRPA1 Channels: From Structure to Disease. Physiol Rev 2019; 100:725-803. [PMID: 31670612 DOI: 10.1152/physrev.00005.2019] [Citation(s) in RCA: 218] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The transient receptor potential ankyrin (TRPA) channels are Ca2+-permeable nonselective cation channels remarkably conserved through the animal kingdom. Mammals have only one member, TRPA1, which is widely expressed in sensory neurons and in non-neuronal cells (such as epithelial cells and hair cells). TRPA1 owes its name to the presence of 14 ankyrin repeats located in the NH2 terminus of the channel, an unusual structural feature that may be relevant to its interactions with intracellular components. TRPA1 is primarily involved in the detection of an extremely wide variety of exogenous stimuli that may produce cellular damage. This includes a plethora of electrophilic compounds that interact with nucleophilic amino acid residues in the channel and many other chemically unrelated compounds whose only common feature seems to be their ability to partition in the plasma membrane. TRPA1 has been reported to be activated by cold, heat, and mechanical stimuli, and its function is modulated by multiple factors, including Ca2+, trace metals, pH, and reactive oxygen, nitrogen, and carbonyl species. TRPA1 is involved in acute and chronic pain as well as inflammation, plays key roles in the pathophysiology of nearly all organ systems, and is an attractive target for the treatment of related diseases. Here we review the current knowledge about the mammalian TRPA1 channel, linking its unique structure, widely tuned sensory properties, and complex regulation to its roles in multiple pathophysiological conditions.
Collapse
Affiliation(s)
- Karel Talavera
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven; VIB Center for Brain and Disease Research, Leuven, Belgium
| | - Justyna B Startek
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven; VIB Center for Brain and Disease Research, Leuven, Belgium
| | - Julio Alvarez-Collazo
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven; VIB Center for Brain and Disease Research, Leuven, Belgium
| | - Brett Boonen
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven; VIB Center for Brain and Disease Research, Leuven, Belgium
| | - Yeranddy A Alpizar
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven; VIB Center for Brain and Disease Research, Leuven, Belgium
| | - Alicia Sanchez
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven; VIB Center for Brain and Disease Research, Leuven, Belgium
| | - Robbe Naert
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven; VIB Center for Brain and Disease Research, Leuven, Belgium
| | - Bernd Nilius
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven; VIB Center for Brain and Disease Research, Leuven, Belgium
| |
Collapse
|
47
|
The phospholipase C inhibitor U73122 is a potent agonist of the polymodal transient receptor potential ankyrin type 1 (TRPA1) receptor channel. Naunyn Schmiedebergs Arch Pharmacol 2019; 393:177-189. [DOI: 10.1007/s00210-019-01722-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 08/26/2019] [Indexed: 01/07/2023]
|
48
|
Huang KF, Ma KH, Jhap TY, Liu PS, Chueh SH. Ultraviolet B irradiation induced Nrf2 degradation occurs via activation of TRPV1 channels in human dermal fibroblasts. Free Radic Biol Med 2019; 141:220-232. [PMID: 31220549 DOI: 10.1016/j.freeradbiomed.2019.06.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 06/10/2019] [Accepted: 06/15/2019] [Indexed: 12/21/2022]
Abstract
Ultraviolet (UV) irradiation causes cellular oxidative stress. Under redox imbalance, Keap1-dependent Nrf2 degradation is minimal. In this study, we examined the role of Ca2+ in Nrf2 homeostasis after UVB irradiation using human dermal fibroblasts. UVB irradiation stimulates 12-lipoxygenase and the product 12-hydroxyeicosatetraenoic acid then activates TRPV1 increasing the cell's cytosolic Ca2+ concentration. UVB irradiation induced reactive oxygen species generation and apoptosis are inhibited in the absence of Ca2+ or in the presence of either a 12-lipoxygenase inhibitor or a TRPV1 inhibitor during and after UVB irradiation. Thus, the Ca2+ increase via TRPV1 is a critical factor in UVB irradiation induced oxidative stress. UVB irradiation induces a Ca2+ dependent Nrf2 degradation and thus activation of TRPV1 with 12-hydroxyeicosatetraenoic acid also decreasing Nrf2 levels. UVB irradiation induced Nrf2 degradation is inhibited by co-treatment of cells with W-7, cyclosporin A, SB-216763 or MG-132, which are inhibitors of calmodulin, calcineurin, GSK3β and the proteasome, respectively. Furthermore, UVB irradiation in parallel induces GSK3β dephosphorylation in a Ca2+ dependent manner. Co-immunoprecipitation showed that UVB irradiation induces an increase in Nrf2 phosphorylation, an increase in the binding of β-TrCP and Nrf2, and an increase in Nrf2 ubiquitination; these effects are all Ca2+ dependent. These findings suggest that UVB irradiation induced GSK3β activation in a Ca2+ dependent manner, which then stimulates the phosphorylation and ubiquitination of Nrf2 via β-TrCP. Indeed, silencing of β-TrCP was found to inhibit UVB irradiation-induced oxidative stress, Nrf2 degradation and apoptosis, while it had no effect on the Ca2+ increase. Taken together, our results suggest that a Ca2+ influx via TRPV1 is responsible for UVB irradiation-induced Nrf2 degradation and that modulation of the Ca2+-calmodulin-calcineurin-GSK3β-Nrf2-β-TrCP-Cullin-1 pathway may explain Ca2+ dependent Nrf2 degradation.
Collapse
Affiliation(s)
- Kuo-Feng Huang
- Division of Plastic Surgery, Department of Surgery, Chi Mei Medical Center, Tainan, Taiwan, ROC
| | - Kuo-Hsing Ma
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Tian-You Jhap
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Pei-Shan Liu
- Department of Microbiology, Soochow University, Taipei, Taiwan, ROC
| | - Sheau-Huei Chueh
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, ROC.
| |
Collapse
|
49
|
Norões MM, Santos LG, Gavioli EC, de Paula Soares Rachetti V, Otuki MF, de Almeida Cabrini D, da Silveira Prudente A, Oliveira JRJM, de Carvalho Gonçalves M, Ferreira J, Preti D, De Logu F, Nassini R, André E. Role of TRPA1 receptors in skin inflammation induced by volatile chemical irritants in mice. Eur J Pharmacol 2019; 858:172460. [PMID: 31228448 DOI: 10.1016/j.ejphar.2019.172460] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 06/13/2019] [Accepted: 06/14/2019] [Indexed: 01/26/2023]
Abstract
Contact dermatitis is a very common inflammatory reaction in the skin, causing not only aesthetic problems but also loss functionality at work. The molecular mechanisms of contact dermatitis induced by chemical irritants are still unclear. Considering that transient receptor potential channels (TRP) may induce neurogenic inflammation and the exacerbation of inflammatory responses, here we investigated the role of transient receptor potential channel ankyrin type-1 (TRPA1) in skin inflammation evoked by chemical irritants. Ear oedema and nociceptive responses elicited by the topical application of xylene and toluene were measured in Swiss mice, wild type and TRPA1 knockout (Trpa1-/-) C57BL/6 mice. Histological analyses were performed in mice subjected to the ear oedema assay. Topical application of xylene and toluene in the mouse ear induced an edematogenic response (0.113 ± 0.008 mm and 0.067 ± 0.011 mm), compared to vehicle (0.008 ± 0.008 mm), assessed by ear thickness measurements and histological analyses. These responses were prevented by topical pretreatment with a selective TRPA1 antagonist, HC-030031 (% inhibition: xylene 36.8 ± 9.4% and toluene 50.7 ± 11.0%), and by the genetic deletion of TRPA1 ((% inhibition: xylene 66.6 ± 16.7% and toluene 75 ± 0%). In addition, the topical application of xylene and toluene to the mouse paw elicited nociceptive responses, which were significantly reduced by oral treatment with HC-030031 ((% of inhibition: 84.9 ± 1.3% and 27.1 ± 8.0%, respectively); nociceptive responses were almost completely abolished in Trpa1-/-mice. Our data suggest that the activation of TRPA1 could be involved in some of the symptoms of irritant-mediated contact dermatitis, such as oedema, pain and neurogenic inflammation.
Collapse
Affiliation(s)
- Maíra Macedo Norões
- Department of Pharmacology, Federal University of Santa Catarina, Campus Universitário, Córrego Grande, Florianópolis, SC, Brazil
| | - Larissa Gonzaga Santos
- Department of Pharmacology, Federal University of Paraná, Centro Politécnico - Jardim das Américas, Curitiba, PR, Brazil
| | - Elaine Cristina Gavioli
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Campus Universitário UFRN - Lagoa Nova, Natal, RN, Brazil
| | - Vanessa de Paula Soares Rachetti
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Campus Universitário UFRN - Lagoa Nova, Natal, RN, Brazil
| | - Michel Fleith Otuki
- Department of Pharmacology, Federal University of Paraná, Centro Politécnico - Jardim das Américas, Curitiba, PR, Brazil
| | - Daniela de Almeida Cabrini
- Department of Pharmacology, Federal University of Paraná, Centro Politécnico - Jardim das Américas, Curitiba, PR, Brazil
| | - Arthur da Silveira Prudente
- Department of Pharmacology, Federal University of Paraná, Centro Politécnico - Jardim das Américas, Curitiba, PR, Brazil; (f)Federal University of Latin American Integration, Avenida Silvio Américo Sasdelli, Bairro Itaipu A, Foz do Iguaçu, PR, Brazil
| | | | - Muryel de Carvalho Gonçalves
- Department of Pharmacology, Federal University of Santa Catarina, Campus Universitário, Córrego Grande, Florianópolis, SC, Brazil
| | - Juliano Ferreira
- Department of Pharmacology, Federal University of Santa Catarina, Campus Universitário, Córrego Grande, Florianópolis, SC, Brazil
| | - Delia Preti
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Ferrara, Italy
| | - Francesco De Logu
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Romina Nassini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Eunice André
- Department of Pharmacology, Federal University of Paraná, Centro Politécnico - Jardim das Américas, Curitiba, PR, Brazil.
| |
Collapse
|
50
|
Human nonvisual opsin 3 regulates pigmentation of epidermal melanocytes through functional interaction with melanocortin 1 receptor. Proc Natl Acad Sci U S A 2019; 116:11508-11517. [PMID: 31097585 DOI: 10.1073/pnas.1902825116] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Opsins form a family of light-activated, retinal-dependent, G protein-coupled receptors (GPCRs) that serve a multitude of visual and nonvisual functions. Opsin 3 (OPN3 or encephalopsin), initially identified in the brain, remains one of the few members of the mammalian opsin family with unknown function and ambiguous light absorption properties. We recently discovered that OPN3 is highly expressed in human epidermal melanocytes (HEMs)-the skin cells that produce melanin. The melanin pigment is a critical defense against ultraviolet radiation (UVR), and its production is mediated by the Gαs-coupled melanocortin 1 receptor (MC1R). The physiological function and light sensitivity of OPN3 in melanocytes are yet to be determined. Here, we show that in HEMs, OPN3 acts as a negative regulator of melanin production by modulating the signaling of MC1R. OPN3 negatively regulates the cyclic adenosine monophosphate (cAMP) response evoked by MC1R via activation of the Gαi subunit of G proteins, thus decreasing cellular melanin levels. In addition to their functional relationship, OPN3 and MC1R colocalize at both the plasma membrane and in intracellular structures, and can form a physical complex. Remarkably, OPN3 can bind retinal, but does not mediate light-induced signaling in melanocytes. Our results identify a function for OPN3 in the regulation of the melanogenic pathway in epidermal melanocytes; we have revealed a light-independent function for the poorly characterized OPN3 and a pathway that greatly expands our understanding of melanocyte and skin physiology.
Collapse
|