1
|
El Mir J, Nasrallah A, Thézé N, Cario M, Fayyad‐Kazan H, Thiébaud P, Rezvani H. Xenopus as a model system for studying pigmentation and pigmentary disorders. Pigment Cell Melanoma Res 2025; 38:e13178. [PMID: 38849973 PMCID: PMC11681847 DOI: 10.1111/pcmr.13178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/17/2024] [Accepted: 05/24/2024] [Indexed: 06/09/2024]
Abstract
Human pigmentary disorders encompass a broad spectrum of phenotypic changes arising from disruptions in various stages of melanocyte formation, the melanogenesis process, or the transfer of pigment from melanocytes to keratinocytes. A large number of pigmentation genes associated with pigmentary disorders have been identified, many of them awaiting in vivo confirmation. A more comprehensive understanding of the molecular basis of pigmentary disorders requires a vertebrate animal model where changes in pigmentation are easily observable in vivo and can be combined to genomic modifications and gain/loss-of-function tools. Here we present the amphibian Xenopus with its unique features that fulfill these requirements. Changes in pigmentation are particularly easy to score in Xenopus embryos, allowing whole-organism based phenotypic screening. The development and behavior of Xenopus melanocytes closely mimic those observed in mammals. Interestingly, both Xenopus and mammalian skins exhibit comparable reactions to ultraviolet radiation. This review highlights how Xenopus constitutes an alternative and complementary model to the more commonly used mouse and zebrafish, contributing to the advancement of knowledge in melanocyte cell biology and related diseases.
Collapse
Affiliation(s)
- Joudi El Mir
- University of Bordeaux, Inserm, BRIC, UMR 1312BordeauxFrance
| | - Ali Nasrallah
- University of Bordeaux, Inserm, BRIC, UMR 1312BordeauxFrance
| | - Nadine Thézé
- University of Bordeaux, Inserm, BRIC, UMR 1312BordeauxFrance
| | - Muriel Cario
- University of Bordeaux, Inserm, BRIC, UMR 1312BordeauxFrance
- Aquiderm, University of BordeauxBordeauxFrance
| | - Hussein Fayyad‐Kazan
- Laboratory of Cancer Biology and Molecular ImmunologyLebanese UniversityHadathLebanon
| | - Pierre Thiébaud
- University of Bordeaux, Inserm, BRIC, UMR 1312BordeauxFrance
| | - Hamid‐Reza Rezvani
- University of Bordeaux, Inserm, BRIC, UMR 1312BordeauxFrance
- Aquiderm, University of BordeauxBordeauxFrance
| |
Collapse
|
2
|
Sangtanoo P, Srimongkol P, Saisavoey T, Puthong S, Buakeaw A, Suttisuwan R, Jatupornpipat M, Pimtong W, Reamtong O, Karnchanatat A. Bee pollen peptides as potent tyrosinase inhibitors with anti-melanogenesis effects in murine b16f10 melanoma cells and zebrafish embryos. Sci Rep 2024; 14:30834. [PMID: 39730661 DOI: 10.1038/s41598-024-81495-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 11/27/2024] [Indexed: 12/29/2024] Open
Abstract
One important functional food ingredient today, valued for its health properties and ability to prevent disease, is bee pollen, which comprises a combination of nectar, pollen from plants, and the secretions of bees. In this research, the tyrosinase (TYR) inhibiting abilities of the peptides derived from bee pollen protein hydrolysates are investigated. Various proteases were utilized to generate these peptides, followed by testing at different concentrations. Tyrosinase inhibition activity was detected in all cases, while the hydrolysate drawn from 5.0% w/v neutrase exhibited the best IC50 value and was thus investigated further via ultrafiltration to separate the active fractions. The highest potential for tyrosinase inhibition was recorded for the fractions below 0.65 kDa. Subsequent purification steps via SEC and RP-HPLC led to the identification of the VDGYPAAGY (named VY-9) peptide via LC-Q-TOF-MS/MS in fraction F1-2, known for its non-toxic and hydrophobic characteristics albeit poor water solubility. The synthesized VY-9 peptide demonstrated competitive inhibition, with IC50 values of 0.55 ± 0.03 µM for mono-phenolase and 2.54 ± 0.06 µM for di-phenolase activities, as confirmed by molecular docking analysis revealing dominant hydrogen bond interactions with TYR. Effective concentrations of 0.2-1.6 µM of VY-9 showed negligible cytotoxicity in B16F10 cells. Melanin synthesis suppression was examined via qRT-PCR, and western blot in MITF, TYR, TRP-1, and TRP-2. Cell death in zebrafish embryos was evaluated in vivo using a toxicity assay which revealed no significant influence from VY-9, while anti-melanogenic effects were observed when the concentration was 4 µM, suggesting bee pollen-derived peptides' potential in cosmetic and pharmaceutical depigmentation applications.
Collapse
Affiliation(s)
- Papassara Sangtanoo
- Center of Excellence in Bioconversion and Bioseparation for Platform Chemical Production, Institute of Biotechnology and Genetic Engineering, Chulalongkorn University, 254 Phayathai Road, Pathumwan, Bangkok, 10330, Thailand
| | - Piroonporn Srimongkol
- Center of Excellence in Bioconversion and Bioseparation for Platform Chemical Production, Institute of Biotechnology and Genetic Engineering, Chulalongkorn University, 254 Phayathai Road, Pathumwan, Bangkok, 10330, Thailand
| | - Tanatorn Saisavoey
- Center of Excellence in Bioconversion and Bioseparation for Platform Chemical Production, Institute of Biotechnology and Genetic Engineering, Chulalongkorn University, 254 Phayathai Road, Pathumwan, Bangkok, 10330, Thailand
| | - Songchan Puthong
- Center of Excellence in Bioconversion and Bioseparation for Platform Chemical Production, Institute of Biotechnology and Genetic Engineering, Chulalongkorn University, 254 Phayathai Road, Pathumwan, Bangkok, 10330, Thailand
| | - Anumart Buakeaw
- Center of Excellence in Bioconversion and Bioseparation for Platform Chemical Production, Institute of Biotechnology and Genetic Engineering, Chulalongkorn University, 254 Phayathai Road, Pathumwan, Bangkok, 10330, Thailand
| | - Rutairat Suttisuwan
- Biodiversity and Sustainable Utilization Research Unit, Department of Biology, Faculty of Science and Technology, Rajamangala University of Technology Krungthep, 2 Nang linchi Road, Sathorn, Bangkok, 10120, Thailand
| | - Marisa Jatupornpipat
- Department of Biology, Faculty of Science, King Mongkut's Institute of Technology, Ladkrabang, Chalongkrung Road, Ladkrabang, Bangkok, 10520, Thailand
| | - Wittaya Pimtong
- Nano Environmental and Health Safety Research Team, National Science and Technology Development Agency (NSTDA), 111 Thailand Science Park, Phahonyothin Road, Khlong Nueng, Khlong Luang, 12120, Pathum Thani, Thailand
| | - Onrapak Reamtong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Aphichart Karnchanatat
- Center of Excellence in Bioconversion and Bioseparation for Platform Chemical Production, Institute of Biotechnology and Genetic Engineering, Chulalongkorn University, 254 Phayathai Road, Pathumwan, Bangkok, 10330, Thailand.
| |
Collapse
|
3
|
Lee WH, Ha Y, Park JI, Joh WB, Park M, Kim JK, Jeon HK, Kim YJ. Triglochin maritima Extracts Exert Anti-Melanogenic Properties via the CREB/MAPK Pathway in B16F10 Cells. Mar Drugs 2024; 22:532. [PMID: 39728107 DOI: 10.3390/md22120532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/28/2024] Open
Abstract
Triglochin maritima, a salt-tolerant plant, has demonstrated antioxidant effects, the ability to prevent prostate enlargement, antifungal properties, and skin moisturizing benefits. This study aimed to explore the anti-melanogenic potential of the 70% ethanol extract of T. maritima (TME) along with its ethyl acetate (TME-EA) and water (TME-A) fractions. TME (10-200 µg/mL), TME-EA (1-15 µg/mL), and TME-A (100-1000 µg/mL) were prepared and applied to B16F10 cells with or without α-MSH for 72 h. MTT assays were used to assess cytotoxicity, and anti-melanogenesis activity was determined by measuring melanin content, conducting a tyrosinase activity assay, and evaluating the expression of melanogenesis-related genes and proteins via RT-PCR and Western blotting. HPLC-PDA was used to analyze TME and TME-EA. The IC20 cytotoxicity values of TME, TME-A, and TME-EA without α-MSH, were 198.426 μg/mL, 1000 μg/mL, and 18.403 μg/mL, respectively. TME and TME-EA significantly decreased melanin and tyrosinase activity in α-MSH-stimulated B16F10 cells, with TME-EA showing comparable effects to arbutin, while TME-A showed no influence. TME-EA down-regulated melanogenesis genes (Tyr, Trp1, Dct, Mitf, Mc1r) and reduced CREB, p-38, and JNK phosphorylation while increasing ERK phosphorylation, suggesting the CREB/MAPK pathway's role in the anti-melanogenic effect. Luteolin was identified as a potential active ingredient. TME-EA may serve as an effective cosmeceutical for hyperpigmentation improvement due to its anti-melanogenic properties.
Collapse
Affiliation(s)
- Won-Hwi Lee
- Research Institute of Basic Sciences, Incheon National University, Incheon 22012, Republic of Korea
| | - Yuna Ha
- Research Institute of Basic Sciences, Incheon National University, Incheon 22012, Republic of Korea
| | - Jeong-In Park
- Research Institute of Basic Sciences, Incheon National University, Incheon 22012, Republic of Korea
- Department of Marine Sciences, Incheon National University, Incheon 22012, Republic of Korea
| | - Won Bae Joh
- Research Institute of Basic Sciences, Incheon National University, Incheon 22012, Republic of Korea
- Department of Marine Sciences, Incheon National University, Incheon 22012, Republic of Korea
| | - Mira Park
- Research Institute of Basic Sciences, Incheon National University, Incheon 22012, Republic of Korea
| | - Jang Kyun Kim
- Research Institute of Basic Sciences, Incheon National University, Incheon 22012, Republic of Korea
- Department of Marine Sciences, Incheon National University, Incheon 22012, Republic of Korea
| | - Hee-Kyung Jeon
- Advanced Energy Materials and Components R&D Group, Korea Institute of Industrial Technology, Busan 46938, Republic of Korea
| | - Youn-Jung Kim
- Research Institute of Basic Sciences, Incheon National University, Incheon 22012, Republic of Korea
- Department of Marine Sciences, Incheon National University, Incheon 22012, Republic of Korea
| |
Collapse
|
4
|
Boira C, Chapuis E, Lapierre L, Auriol D, Jarrin C, Robe P, Tiguemounine J, Scandolera A, Reynaud R. Epigallocatechin Gallate Enzymatic Alpha Glucosylation Potentiates Its Skin-Lightening Activity-Involvement of Skin Microbiota. Molecules 2024; 29:5391. [PMID: 39598779 PMCID: PMC11597265 DOI: 10.3390/molecules29225391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/11/2024] [Accepted: 10/18/2024] [Indexed: 11/29/2024] Open
Abstract
(1) Background: Ultraviolet radiation takes part in photoaging and pigmentation disorders on skin. Epigallocatechin gallate (EGCG) is a well-known brightening and photoprotective compound but it faces limitations in terms of stability and solubility. (2) Methods: A more stable and water-soluble glucoside called EGCG-G1 was obtained by enzymatic glucosylation of EGCG. In vitro and ex vivo experiments evaluated EGCG-G1 skin penetration, antioxidant activity, and antimelanogenic properties compared to EGCG. This gene expression study characterized the pathways impacted by EGCG-G1. Four clinical studies covering phototypes I to V, at various ages, and different skin areas, using several tools, were conducted to assess the effect of EGCG-G1 on skin hyperpigmentation and tone. The impact of glucoside on skin microbiota, especially Lactobacillus sp., was assessed through in vitro and in vivo investigations. (3) Results: EGCG-G1 better penetrated the epidermis than EGCG due to a possible interaction with GLUT1. EGCG-G1 presented similar antioxidant activity to that of EGCG and decreased melanogenesis through the inhibition of 13 genes, including MITF. The skin Lactobacillus population increased with EGCG-G1, which promoted bacterial growth in vitro as prebiotic, and induced the release of a microbial brightening metabolite. Clinical trials demonstrated EGCG-G1 to decrease hyperpigmented spots and increase skin brightness and homogeneity in a large panel of phototypes, outperforming EGCG and vitamin C. (4) Conclusions: Glucosylation of EGCG maintained its photoprotective antioxidant properties and enhanced penetration across the epidermis. EGCG-G1 demonstrated brightening properties on all skin types by down-regulation of melanogenesis pathways and indirectly by skin microbiota stimulation.
Collapse
Affiliation(s)
- Cloé Boira
- Givaudan Active Beauty, R&D, 51110 Pomacle, France
| | | | | | | | | | - Patrick Robe
- Givaudan Active Beauty, R&D, 31400 Toulouse, France
| | | | | | | |
Collapse
|
5
|
Park NY, Jo DS, Park HJ, Bae JE, Kim YH, Kim JB, Lee HJ, Kim SH, Choi H, Lee HS, Yoshimori T, Lee DS, Lee JA, Kim P, Cho DH. Deciphering melanophagy: role of the PTK2-ITCH-MLANA-OPTN cascade on melanophagy in melanocytes. Autophagy 2024:1-10. [PMID: 39477686 DOI: 10.1080/15548627.2024.2421695] [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: 01/16/2024] [Revised: 10/10/2024] [Accepted: 10/22/2024] [Indexed: 11/13/2024] Open
Abstract
Melanosomes play a pivotal role in skin color and photoprotection. In contrast to the well-elucidated pathway of melanosome biogenesis, the process of melanosome degradation, referred to as melanophagy, is largely unexplored. Previously, we discovered that 3,4,5-trimethoxycinnamate thymol ester (TCTE) effectively inhibits skin pigmentation by activating melanophagy. In this study, we discovered a new regulatory signaling cascade that controls melanophagy in TCTE-treated melanocytes. ITCH (itchy E3 ubiquitin protein ligase) facilitates ubiquitination of the melanosome membrane protein MLANA (melan-A) during TCTE-induced melanophagy. This ubiquitinated MLANA is then recognized by an autophagy receptor protein, OPTN (optineurin). Additionally, a phospho-kinase antibody array revealed that TCTE activates PTK2 (protein tyrosine kinase 2), which phosphorylates ITCH, enhancing the ubiquitination of MLANA. Furthermore, inhibition of either PTK2 or ITCH disrupts the ubiquitination of MLANA and the MLANA-OPTN interaction in TCTE-treated cells. Taken together, our findings highlight the critical role of the PTK2-ITCH-MLANA-OPTN cascade in orchestrating melanophagy progression.Abbreviations: α-MSH: alpha-melanocyte-stimulating hormone; dichlone: 2,3-dichloro-1,4-naphthoquinone; ITCH: itchy E3 ubiquitin protein ligase; MITF: melanocyte inducing transcription factor; MLANA: melan-A; NBR1: NBR1 autophagy cargo receptor; OPTN: optineurin; PINK1: PTEN induced kinase 1; PTK2: protein tyrosine kinase 2; SQSTM1/p62: sequestosome 1; TCTE: 3,4,5-trimethoxycinnamate thymol ester; TPC2: two pore segment channel 2; VDAC1: voltage dependent anion channel 1.
Collapse
Affiliation(s)
- Na Yeon Park
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Doo Sin Jo
- ORGASIS Corp, Suwon, Gyeonggi-do, Republic of Korea
| | - Hyun Jun Park
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Ji-Eun Bae
- KNU G-LAMP Project Group, KNU Institute of Basic Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Yong Hwan Kim
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Joon Bum Kim
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Ha Jung Lee
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
- ORGASIS Corp, Suwon, Gyeonggi-do, Republic of Korea
| | - Sung Hyun Kim
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Hyunjung Choi
- R&D Unit, AmorePacific Corporation, Yongin, Gyeonggi-Do, Republic of Korea
| | - Hyun-Shik Lee
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
- KNU G-LAMP Project Group, KNU Institute of Basic Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Tamotsu Yoshimori
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Dong-Seok Lee
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
- Organelle Institute, KNU, Daegu, Republic of Korea
| | - Jin-A Lee
- Department of Biological Sciences and Biotechnology, College of Life Sciences and Nanotechnology, Hannam University, Daejeon, Republic of Korea
| | - Pansoo Kim
- ORGASIS Corp, Suwon, Gyeonggi-do, Republic of Korea
| | - Dong-Hyung Cho
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
- ORGASIS Corp, Suwon, Gyeonggi-do, Republic of Korea
- Organelle Institute, KNU, Daegu, Republic of Korea
| |
Collapse
|
6
|
Misiąg P, Molik K, Kisielewska M, Typek P, Skowron I, Karwowska A, Kuźnicki J, Wojno A, Ekiert M, Choromańska A. Amelanotic Melanoma-Biochemical and Molecular Induction Pathways. Int J Mol Sci 2024; 25:11502. [PMID: 39519055 PMCID: PMC11546312 DOI: 10.3390/ijms252111502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/19/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Amelanotic melanoma (AM) is a subtype of hypomelanotic or completely amelanotic melanoma. AM is a rare subtype of melanoma that exhibits a higher recurrence rate and aggressiveness as well as worse surveillance than typical melanoma. AM shows a dysregulation of melanin production, cell cycle control, and apoptosis pathways. Knowing these pathways has an application in medicine due to targeted therapies based on the inhibiting elements of the abovementioned pathways. Therefore, we summarized and discussed AM biochemical and molecular induction pathways and personalized medicine approaches, clinical management, and future directions due to the fact that AM is relatively rare. AM is commonly misdiagnosed. Hence, the role of biomarkers is becoming significant. Nonetheless, there is a shortage of biomarkers specific to AM. BRAF, NRAS, and c-KIT genes are the main targets of therapy. However, the role of BRAF and KIT in AM varied among studies. BRAF inhibitors combined with MAK inhibitors demonstrate better results. Immune checkpoint inhibitors targeting CTLA-4 combined with a programmed death receptor 1 (PD-1) show better outcomes than separately. Fecal microbiota transplantation may overcome resistance to immune checkpoint therapy of AM. Immune-modulatory vaccines against indoleamine 2,3-dioxygenase (IDO) and PD ligand (PD-L1) combined with nivolumab may be efficient in melanoma treatment.
Collapse
Affiliation(s)
- Piotr Misiąg
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (P.M.); (K.M.); (M.K.); (P.T.); (I.S.); (A.K.); (J.K.); (A.W.)
- Students Scientific Group No. 148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Klaudia Molik
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (P.M.); (K.M.); (M.K.); (P.T.); (I.S.); (A.K.); (J.K.); (A.W.)
- Students Scientific Group No. 148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Monika Kisielewska
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (P.M.); (K.M.); (M.K.); (P.T.); (I.S.); (A.K.); (J.K.); (A.W.)
- Students Scientific Group No. 148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Paulina Typek
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (P.M.); (K.M.); (M.K.); (P.T.); (I.S.); (A.K.); (J.K.); (A.W.)
- Students Scientific Group No. 148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Izabela Skowron
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (P.M.); (K.M.); (M.K.); (P.T.); (I.S.); (A.K.); (J.K.); (A.W.)
- Students Scientific Group No. 148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Anna Karwowska
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (P.M.); (K.M.); (M.K.); (P.T.); (I.S.); (A.K.); (J.K.); (A.W.)
- Students Scientific Group No. 148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Jacek Kuźnicki
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (P.M.); (K.M.); (M.K.); (P.T.); (I.S.); (A.K.); (J.K.); (A.W.)
- Students Scientific Group No. 148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Aleksandra Wojno
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (P.M.); (K.M.); (M.K.); (P.T.); (I.S.); (A.K.); (J.K.); (A.W.)
- Students Scientific Group No. 148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Marcin Ekiert
- Department of Oncology, Wroclaw Medical University, pl. L. Hirszfelda 12, 53-413 Wroclaw, Poland;
| | - Anna Choromańska
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| |
Collapse
|
7
|
Byun KA, Lee SY, Oh S, Batsukh S, Jang JW, Lee BJ, Rheu KM, Li S, Jeong MS, Son KH, Byun K. Fermented Fish Collagen Attenuates Melanogenesis via Decreasing UV-Induced Oxidative Stress. Mar Drugs 2024; 22:421. [PMID: 39330302 PMCID: PMC11433465 DOI: 10.3390/md22090421] [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: 07/05/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 09/28/2024] Open
Abstract
Excessive melanogenesis leads to hyperpigmentation-related cosmetic problems. UV exposure increases oxidative stress, which promotes melanogenesis-related signal pathways such as the PKA, microphthalmia-associated transcription factor (MITF), tyrosinase (TYR), tyrosinase-related protein-1 (TRP1), and tyrosinase-related protein-2 (TRP2) pathways. Glycine is a source of endogenous antioxidants, including glutathione. Fermented fish collagen (FC) contains glycine; thus, we evaluated the effect of FC on decreasing melanogenesis via decreasing oxidative stress. The glycine receptor (GlyR) and glycine transporter-1 (GlyT1) levels were decreased in UV-irradiated keratinocytes; however, the expression levels of these proteins increased upon treatment with FC. The FC decreased oxidative stress, as indicated by the decreasing expression of NOX1/2/4, increased expression of GSH/GSSG, increased SOD activity, and decreased 8-OHdG expression in UV-irradiated keratinocytes. Administration of conditioned media from FC-treated keratinocytes to melanocytes led to decreased p38, PKC, MITF, TRP1, and TRP2 expression. These changes induced by the FC were also observed in UV-irradiated animal skin. FC treatment increased the expression of GlyR and GlyT, which was accompanied by decreased oxidative stress in the UV-irradiated skin. Moreover, the FC negatively regulated the melanogenesis signaling pathways, leading to decreased melanin content in the UV-irradiated skin. In conclusion, FC decreased UV-induced oxidative stress and melanogenesis in melanocytes and animal skin. FC could be used in the treatment of UV-induced hyperpigmentation problems.
Collapse
Affiliation(s)
- Kyung-A Byun
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
- LIBON Inc., Incheon 22006, Republic of Korea
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - So Young Lee
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, Gachon University, Incheon 21565, Republic of Korea
| | - Seyeon Oh
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Sosorburam Batsukh
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Jong-Won Jang
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health & Sciences and Technology (GAIHST), Gachon University, Incheon 21999, Republic of Korea
| | - Bae-Jin Lee
- Marine Bioprocess Co., Ltd., Busan 46048, Republic of Korea
| | | | - Sichao Li
- Marine Bioprocess Co., Ltd., Busan 46048, Republic of Korea
| | - Min-Seok Jeong
- Marine Bioprocess Co., Ltd., Busan 46048, Republic of Korea
| | - Kuk Hui Son
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, Gachon University, Incheon 21565, Republic of Korea
| | - Kyunghee Byun
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health & Sciences and Technology (GAIHST), Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
8
|
Capasso C, Supuran CT. Overview on tyrosinases: Genetics, molecular biology, phylogenetic relationship. Enzymes 2024; 56:1-30. [PMID: 39304284 DOI: 10.1016/bs.enz.2024.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Tyrosinases (TYRs) are enzymes found in various organisms that are crucial for melanin biosynthesis, coloration, and UV protection. They play vital roles in insect cuticle sclerotization, mollusk shell formation, fungal and bacterial pigmentation, biofilm formation, and virulence. Structurally, TYRs feature copper-binding sites that are essential for catalytic activity, facilitating substrate oxidation via interactions with conserved histidine residues. TYRs exhibit diversity across animals, plants, fungi, mollusks, and bacteria, reflecting their roles and function. Eukaryotic TYRs undergo post-translational modifications, such as glycosylation, which affect protein folding and activity. Bacterial TYRs are categorized into five types based on their structural variation, domain organization and enzymatic properties, showing versatility across bacterial species. Moreover, bacterial TYRs, akin to fungal TYRs, have been implicated in the synthesis of secondary metabolites with antimicrobial properties. TYRs share significant sequence homology with hemocyanins, oxygen-carrier proteins in mollusks and arthropods, highlighting their evolutionary relationships. The evolution of TYRs underscores the dynamic nature of these enzymes and reflects adaptive strategies across diverse taxa.
Collapse
Affiliation(s)
- Clemente Capasso
- Department of Biology, Agriculture and Food Sciences, Institute of Biosciences and Bioresources, CNR, Napoli, Italy.
| | - Claudiu T Supuran
- Neurofarba Department, Pharmaceutical and Nutraceutical Section, University of Florence, Sesto Fiorentino, Florence, Italy
| |
Collapse
|
9
|
Seong SH, Oh SH. Up-and-Coming Drugs for the Treatment of Vitiligo. Ann Dermatol 2024; 36:197-208. [PMID: 39082655 PMCID: PMC11291099 DOI: 10.5021/ad.24.038] [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: 03/25/2024] [Revised: 05/07/2024] [Accepted: 06/02/2024] [Indexed: 08/03/2024] Open
Abstract
Vitiligo is a chronic autoimmune disease that causes depigmented patches on the skin. It affects 0.5%-2.0% of the global population. It goes beyond physical appearance, often leading to stigmatization, low self-esteem, and depression, burdening patients with psychosocial challenges. The pathogenesis of vitiligo involves the loss of melanocytes due to autoreactive CD8+ T cells, triggered by environmental stressors and exacerbated by cellular vulnerabilities and immune responses. The release of danger signals and pro-inflammatory factors initiates an immune cascade perpetuating melanocyte destruction, mainly driven by interferon-γ and the C-X-C motif chemokine ligand 9/10-chemokine receptor 3 axis. Long-lasting tissue-resident memory T cells (Trms) and cytokines contribute to lesion persistence. Current treatments focus on topical steroids and tacrolimus, systemic steroids, and phototherapies, but their efficacy remains suboptimal, necessitating the development of new therapeutic options. Building on recent advancements in understanding the immunological mechanisms in vitiligo pathogenesis, with the initiation of Food and Drug Administration approval of topical ruxolitinib, various potential treatment options such as JAK inhibitors, cytokine blockers, and Trm or regulatory T cell targeting agents are being clinically researched and anticipated for vitiligo based on both preclinical and clinical data. This review aims to categorize and summarize the diverse investigational drugs currently undergoing clinical trials for vitiligo. By examining clinical outcomes, it is anticipated that this review will bring hope to dermatologists and patients regarding vitiligo, a condition that has historically posed challenges and transform it into a realm of potential possibilities.
Collapse
Affiliation(s)
- Seol Hwa Seong
- Department of Dermatology, Kosin University College of Medicine, Busan, Korea
| | - Sang Ho Oh
- Department of Dermatology and Cutaneous Biology Research Institute, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
10
|
Beregoi M, Oprea D, Bunea MC, Enculescu M, Enache TA. Electrospun fibrillary scaffold for electrochemical cell biomarkers detection. Mikrochim Acta 2024; 191:435. [PMID: 38949689 PMCID: PMC11217050 DOI: 10.1007/s00604-024-06523-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 06/21/2024] [Indexed: 07/02/2024]
Abstract
A novel scaffold for in situ electrochemical detection of cell biomarkers was developed using electrospun nanofibers and commercial adhesive polymeric membranes. The electrochemical sensing of cell biomarkers requires the cultivation of the cells on/near the (bio)sensor surface in a manner to preserve an appropriate electroactive available surface and to avoid the surface passivation and sensor damage. This can be achieved by employing biocompatible nanofiber meshes that allow the cells to have a normal behavior and do not alter the electrochemical detection. For a better mechanical stability and ease of handling, nylon 6/6 nanofibers were collected on commercial polymeric membranes, at an optimal fiber density, obtaining a double-layered platform. To demonstrate the functionality of the fabricated scaffold, the screening of cellular stress has been achieved integrating melanoma B16-F10 cells and the (bio)sensor components on the transducer whereas the melanin exocytosis was successfully quantified using a commercial electrode. Either directly on the surface of the (bio)sensor or spatially detached from it, the integration of cell cultures in biosensing platforms based on electrospun nanofibers represents a powerful bioanalytical tool able to provide real-time information about the biomarker release, enzyme activity or inhibition, and monitoring of various cellular events.
Collapse
Affiliation(s)
- Mihaela Beregoi
- Functional Nanostructures Laboratory, National Institute of Materials Physics, Atomistilor Str. 405A, 077125, Magurele, Romania
| | - Daniela Oprea
- Functional Nanostructures Laboratory, National Institute of Materials Physics, Atomistilor Str. 405A, 077125, Magurele, Romania
- Faculty of Physics, University of Bucharest, Atomistilor Str. 405, 077125, Magurele, Romania
| | - Mihaela Cristina Bunea
- Functional Nanostructures Laboratory, National Institute of Materials Physics, Atomistilor Str. 405A, 077125, Magurele, Romania
| | - Monica Enculescu
- Functional Nanostructures Laboratory, National Institute of Materials Physics, Atomistilor Str. 405A, 077125, Magurele, Romania
| | - Teodor Adrian Enache
- Functional Nanostructures Laboratory, National Institute of Materials Physics, Atomistilor Str. 405A, 077125, Magurele, Romania.
| |
Collapse
|
11
|
Xie W, Dhinojwala A, Gianneschi NC, Shawkey MD. Interactions of Melanin with Electromagnetic Radiation: From Fundamentals to Applications. Chem Rev 2024; 124:7165-7213. [PMID: 38758918 DOI: 10.1021/acs.chemrev.3c00858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2024]
Abstract
Melanin, especially integumentary melanin, interacts in numerous ways with electromagnetic radiation, leading to a set of critical functions, including radiation protection, UV-protection, pigmentary and structural color productions, and thermoregulation. By harnessing these functions, melanin and melanin-like materials can be widely applied to diverse applications with extraordinary performance. Here we provide a unified overview of the melanin family (all melanin and melanin-like materials) and their interactions with the complete electromagnetic radiation spectrum (X-ray, Gamma-ray, UV, visible, near-infrared), which until now has been absent from the literature and is needed to establish a solid fundamental base to facilitate their future investigation and development. We begin by discussing the chemistries and morphologies of both natural and artificial melanin, then the fundamentals of melanin-radiation interactions, and finally the exciting new developments in high-performance melanin-based functional materials that exploit these interactions. This Review provides both a comprehensive overview and a discussion of future perspectives for each subfield of melanin that will help direct the future development of melanin from both fundamental and applied perspectives.
Collapse
Affiliation(s)
- Wanjie Xie
- Department of Biology, Evolution and Optics of Nanostructure Group, University of Ghent, Gent 9000, Belgium
| | - Ali Dhinojwala
- School of Polymer Science and Polymer Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Nathan C Gianneschi
- Department of Chemistry, Department of Materials Science and Engineering, Department of Biomedical Engineering, Simpson-Querrey Institute, Chemistry of Life Processes Institute, Lurie Cancer Center, and International Institute of Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
| | - Matthew D Shawkey
- Department of Biology, Evolution and Optics of Nanostructure Group, University of Ghent, Gent 9000, Belgium
| |
Collapse
|
12
|
Rzepka Z, Rok J, Zdybel M, Pilawa B, Beberok A, Wrześniok D. Streptomycin generates oxidative stress in melanin-producing cells: In vitro study with EPR spectroscopy evidence. Toxicol In Vitro 2024; 98:105844. [PMID: 38740103 DOI: 10.1016/j.tiv.2024.105844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/12/2024] [Accepted: 05/08/2024] [Indexed: 05/16/2024]
Abstract
Streptomycin (STR) is an aminoglycoside antibiotic with a broad-spectrum of activity and ototoxic potential. The mechanism of STR-induced inner ear damage has not been fully elucidated. It was previously found that STR binds to melanin, which may result in the accumulation of the drug in melanin-containing tissues. Melanin pigment is present in various parts of the inner ear, including the cochlea and vestibular organ. The present study aimed to assess if streptomycin generates oxidative stress and affects melanogenesis in normal human melanocytes. Moreover the variation of free radical concentration in STR-treated melanocytes was examined by electron paramagnetic resonance spectroscopy (EPR). We found that STR decreases cell metabolic activity and reduces melanin content. The observed changes in the activity of antioxidant enzymes activity in HEMn-DPs treated with streptomycin may suggest that the drug affects redox homeostasis in melanocytes. In this work EPR study expanded knowledge about free radicals in interactions of STR and melanin in melanocytes. The results may help elucidate the mechanisms of STR toxicity on pigment cells, including melanin-producing cells in the inner ear. This is important because understanding the mechanism of STR-induced ototoxicity would be helpful in developing new therapeutic strategies to protect patients' hearing.
Collapse
Affiliation(s)
- Zuzanna Rzepka
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 4 Jagiellońska Str., 41-200 Sosnowiec, Poland
| | - Jakub Rok
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 4 Jagiellońska Str., 41-200 Sosnowiec, Poland
| | - Magdalena Zdybel
- Department of Biophysics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 8 Jedności Str., 41-200 Sosnowiec, Poland
| | - Barbara Pilawa
- Department of Biophysics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 8 Jedności Str., 41-200 Sosnowiec, Poland
| | - Artur Beberok
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 4 Jagiellońska Str., 41-200 Sosnowiec, Poland
| | - Dorota Wrześniok
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 4 Jagiellońska Str., 41-200 Sosnowiec, Poland.
| |
Collapse
|
13
|
Liu Z, Jin S, Cheng D, Chen H, Wang Y, Ji C, Yan Z, Fang X, Xiao S, Bi X. Modified human skin cell isolation protocol and its influence on keratinocyte and melanocyte culture. Regen Ther 2024; 26:203-212. [PMID: 38948130 PMCID: PMC11214264 DOI: 10.1016/j.reth.2024.05.014] [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: 04/28/2024] [Revised: 05/21/2024] [Accepted: 05/26/2024] [Indexed: 07/02/2024] Open
Abstract
Introduction With the increasing emphasis on the use of nonanimal ingredients in clinical care, studies have proposed the use of TrypLE™ as an alternative to trypsin. However, previous research has reported insufficient cell yield and viability when using TrypLE to isolate skin cells compared to the dispase/trypsin-EDTA method. This study aimed to propose an improved method for increasing the yield and viability of cells isolated by TrypLE and to evaluate isolated keratinocytes and melanocytes. Methods Foreskin tissues were isolated to keratinocytes and melanocytes using the trypsin-EDTA protocol and our modified TrypLE protocol. The yield and viability of freshly isolated cells were compared, the epidermal residue after cell suspension filtration was analyzed histologically, and the expression of cytokeratin 14 (CK14) and Melan-A was detected by flow cytometry. After cultivation, keratinocytes and melanocytes were further examined for marker expression and proliferation. A coculture model of melanocytes and HaCaT cells was used to evaluate melanin transfer. Results The yield, viability of total cells and expression of the keratinocyte marker CK14 were similar for freshly isolated cells from both protocols. No differences were observed in the histologic analysis of epidermal residues. Moreover, no differences in keratinocyte marker expression or melanocyte melanin transfer function were observed after culture. However, melanocytes generated using the TrypLE protocol exhibited increased Melan-A expression and proliferation in culture. Conclusion Our TrypLE protocol not only solved the problems of insufficient cell yield and viability in previous studies but also preserved normal cell morphology and function, which enables the clinical treatment of depigmentation diseases.
Collapse
Affiliation(s)
- Zhi Liu
- Department of Dermatology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Shunxin Jin
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Dapeng Cheng
- Department of Dermatology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Hao Chen
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
- Department of Hand Surgery, The 971st Hospital of the CPLA Navy, Qingdao, China
| | - Yuxiang Wang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Chao Ji
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Zhenzhen Yan
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xiao Fang
- Department of Urology, The Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Shichu Xiao
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xinling Bi
- Department of Dermatology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
14
|
Chen H, Jian M, Teng H, Li Z, Xu X, Li X, Jiang R, Zhao D, Sun L, Liu J. Ginsenoside Rf in wild ginseng adventitious roots extract inhibits melanogenesis via cAMP/PKA and NO/cGMP signalling pathways in α-melanocyte-stimulating hormone-stimulated B16F10 mouse melanoma cells and zebrafish. Nat Prod Res 2024:1-8. [PMID: 38318698 DOI: 10.1080/14786419.2024.2312541] [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: 10/11/2023] [Accepted: 01/27/2024] [Indexed: 02/07/2024]
Abstract
The scarcity of more effective wild ginseng has severely limited its use, culturing of adventitious roots from wild ginseng were its good substitute. In this study, we found ginsenoside Rf as the special component in adventitious roots extract significantly decreased melanin levels and tyrosinase activity in B16F10 cells and zebrafish, and suppressed the expression of microphthalmia-associated transcription factor and melanogenic enzymes in B16F10 cells. Notably, Rf treatment of B16F10 cells led to reduced cell levels of adenosine cyclic 3', 5'-monophosphate (cAMP), nitric oxide (NO), and guanoside cyclic 3', 5'-monophosphate (cGMP), and reduced activities of adenylate cyclase (AC), protein kinase A (PKA), guanylate cyclase (GC), and protein kinase G (PKG), which suggest Rf anti-melanogenic activity potentially involved inhibition of AC/cAMP/PKA and NO/GC/cGMP/PKG signalling pathway. This work provides experimental basis for skin-lightening effect of wild ginseng adventitious roots and their functional part.
Collapse
Affiliation(s)
- Hong Chen
- School of Mechanical Engineering, Shanghai Jiao Tong University, Shanghai, China
- Tonghua Herbal Biotechnology CO., Ltd, Tonghua, Jilin Province, China
| | - Mengqiong Jian
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Huanhuan Teng
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Zhenzhuo Li
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Xiaohao Xu
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Xiangzhu Li
- Tonghua Herbal Biotechnology CO., Ltd, Tonghua, Jilin Province, China
| | - Rui Jiang
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Daqing Zhao
- Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin Province, China
- Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Liwei Sun
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin Province, China
- Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Jianzeng Liu
- Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| |
Collapse
|
15
|
Montero P, Sanz C, Pérez-Fidalgo JA, Pérez-Leal M, Milara J, Cortijo J. Paclitaxel alters melanogenesis and causes pigmentation in the skin of gynecological cancer patients. Fundam Clin Pharmacol 2024; 38:183-191. [PMID: 37483143 DOI: 10.1111/fcp.12943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/09/2023] [Accepted: 07/10/2023] [Indexed: 07/25/2023]
Abstract
BACKGROUND Paclitaxel (PTX) is a microtubule-stabilizing antineoplastic that has been shown to damage healthy tissues like the skin. Hyperpigmentation can be found among the adverse effects caused by PTX, but the literature is limited and the mechanisms driving PTX-induced pigmentary alterations are unknown. OBJECTIVES This study aimed to describe the pigmentary alterations caused by PTX and to determine the effects of PTX on melanocytes. METHODS Pigmentary skin alterations were measured in 20 gynecological cancer patients under PTX treatment by using specific probes, which determine the melanin index and the pigmentation level. Melanocytes were incubated with paclitaxel to analyze melanogenesis markers gene expression, melanin content, and transcription factors activation. RESULTS Paclitaxel induced alterations in the skin pigmentation with no visible clinical manifestations. Gynecological cancer patients under paclitaxel treatment had an increase in the melanin index and pigmentation levels. In vitro, PTX exposure to melanocytes increased the expression of melanogenesis markers, melanin content, and induced activation of ERK and MITF. CONCLUSIONS The results suggest that PTX alters pigmentation in patients with no clinically visible manifestations, and these alterations might be driven by its capacity to stimulate melanogenesis on melanocytes through the MITF activation pathway.
Collapse
Affiliation(s)
- Paula Montero
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
- Faculty of Health Sciences, Universidad Europea de Valencia, Valencia, Spain
| | - Celia Sanz
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Jose Alejandro Pérez-Fidalgo
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, Valencia, Spain
- Biomedical Research Networking Centre on Cancer (CIBERONC), Health Institute Carlos III, Madrid, Spain
- INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Martín Pérez-Leal
- Faculty of Health Sciences, Universidad Europea de Valencia, Valencia, Spain
| | - Javier Milara
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Health Institute Carlos III, Madrid, Spain
- Pharmacy Unit, University General Hospital Consortium, Valencia, Spain
| | - Julio Cortijo
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Health Institute Carlos III, Madrid, Spain
- Research and Teaching Unit, University General Hospital Consortium, Valencia, Spain
| |
Collapse
|
16
|
Lv J, Zhang X, An X, Cao Y, Meng D, Zou K, Gao R, Zhang R. The inhibition of VDAC1 oligomerization promotes pigmentation through the CaMK-CRTCs/CREB-MITF pathway. Exp Cell Res 2024; 434:113874. [PMID: 38070860 DOI: 10.1016/j.yexcr.2023.113874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/30/2023] [Accepted: 12/03/2023] [Indexed: 12/17/2023]
Abstract
The voltage-dependent anion channel 1 (VDAC1) forms an oligomeric structure on the mitochondrial outer membrane, which plays critical roles in many physiological processes. Research studies have demonstrated that the knockout of VDAC1 increases pigment content and up-regulates the expression of melanogenic genes. Due to its involvement in various physiological processes, the depletion of VDAC1 has significant detrimental effects on cellular functions and the inhibition of VDAC1 oligomerization has recently emerged as a promising strategy for the treatment of several diseases. In this study, we found that VDAC1 oligomerization inhibitors, VBIT-12 and NSC-15364, promote melanogenesis, dendrite formation and melanosome transport in human epidermal melanocytes (HEMCs). Mechanistically, treatment of HEMCs with an oligomerization inhibitor increased the level of cytoplasmic calcium ions, which activated calcium-calmodulin dependent protein kinase (CaMK) and led to the phosphorylation of CREB and the nuclear translocation of CREB-regulated transcription coactivators (CRTCs). Subsequently, CRTCs, p-CREB and CREB-binding protein (CBP) in the nucleus cooperatively recruit the transcription machinery to initiate the transcription of MITF thus promoting pigmentation. Importantly, our study also demonstrates that VDAC1 oligomerization inhibitors increase pigmentation in zebrafish and in human skin explants, highlighting their potential as a therapeutic strategy for skin pigmentation disorders.
Collapse
Affiliation(s)
- Jinpeng Lv
- School of Pharmacy, Changzhou University, Changzhou, 213000, China; Department of Dermatology, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
| | - Ximei Zhang
- School of Pharmacy, Changzhou University, Changzhou, 213000, China
| | - Xiaohong An
- Yunnan Characteristic Plant Extraction Laboratory, Yunnan Yunke Characteristic Plant Extraction Laboratory Co., Ltd., Kunming, 650106, China; Yunnan Botanee Bio-technology Group Co., Ltd., Kunming, 650106, China
| | - Yan Cao
- Department of Dermatology, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, 213000, China
| | - Duo Meng
- School of Pharmacy, Changzhou University, Changzhou, 213000, China
| | - Kun Zou
- School of Pharmacy, Changzhou University, Changzhou, 213000, China
| | - Rongyin Gao
- Department of Dermatology, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
| | - Ruzhi Zhang
- Department of Dermatology, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, China.
| |
Collapse
|
17
|
Yamashita T, Hatakeyama T, Hashimoto S, Inenaga T, Kashimura A, Matsumoto H. PMEL p.L18del associates with beef quality of Kumamoto sub-breed of Japanese Brown cattle. Anim Sci J 2024; 95:e14003. [PMID: 39318113 DOI: 10.1111/asj.14003] [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: 07/01/2024] [Revised: 08/21/2024] [Accepted: 08/29/2024] [Indexed: 09/26/2024]
Abstract
Japanese Brown cattle is the second most popular breed among Wagyu breeds and raised mainly in Kumamoto and Kochi Prefectures. Typical coat color of the Kumamoto sub-breed is solid brown, but individuals with diluted coat color are sometimes born. We previously detected four SNPs in PMEL gene and identified p.L18del as the causative polymorphism of this diluted phenotype. The current study examined the association between the SNPs in PMEL gene and carcass traits of the Kumamoto sub-breed. Our association analysis revealed that p.L18del had significant effects on BMS (p = 0.0263), meat brightness (p = 0.0179), meat firmness (p = 0.0102), and meat texture (p = 0.0252) and that del allele of this SNP might be useful to improve these carcass traits.
Collapse
Affiliation(s)
- Taito Yamashita
- Course of Agricultural Sciences, Graduate School of Agriculture, Tokai University, Kumamoto, Japan
| | - Toko Hatakeyama
- Department of Animal Science, School of Agriculture, Tokai University, Kumamoto, Japan
| | - Syun Hashimoto
- Department of Animal Science, School of Agriculture, Tokai University, Kumamoto, Japan
| | - Toshiaki Inenaga
- Course of Agricultural Sciences, Graduate School of Agriculture, Tokai University, Kumamoto, Japan
- Department of Animal Science, School of Agriculture, Tokai University, Kumamoto, Japan
| | - Atsushi Kashimura
- Course of Agricultural Sciences, Graduate School of Agriculture, Tokai University, Kumamoto, Japan
- Department of Animal Science, School of Agriculture, Tokai University, Kumamoto, Japan
| | - Hirokazu Matsumoto
- Course of Agricultural Sciences, Graduate School of Agriculture, Tokai University, Kumamoto, Japan
- Department of Animal Science, School of Agriculture, Tokai University, Kumamoto, Japan
| |
Collapse
|
18
|
Kim S, Na GH, Jung HM, Han SH, Han J, Koo YK. Enzyme-treated caviar extract ameliorates melanogenesis in UVB-induced SKH-1 hairless mice. Biochem Biophys Res Commun 2023; 673:81-86. [PMID: 37364389 DOI: 10.1016/j.bbrc.2023.05.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/17/2023] [Accepted: 05/03/2023] [Indexed: 06/28/2023]
Abstract
This study investigated anti-melanogenesis effects of enzyme-treated caviar extract (CV) in murine melanoma B16F10 cells and SKH-1 hairless mice. To induce melanin production in vitro and in vivo studies, B16F10 cells were treated with 3-Isobutyl-1-methylxanthine (IBMX), and SKH-1 hairless mice were irradiated with UVB, respectively. The expression of melnogenesis-related factors and signaling molecules were analyzed by ELISA and western blotting. 50, 100 and 200 μg/mL of CV significantly decreased the melanin contents and the activities of tyrosinase, nitric oxide, glutathione, and cAMP, melanogenesis factor, in B16F10 cells treated IBMX. In addition, CV significantly suppressed the expression of melanogenesis proteins such as pPKA, pCREB, MITF, TRP-1and TRP-2. Similarly, results of oral administration of CV (20, 50 and 100 mg/kg) for 8 weeks in UVB-Induced SKH-1 hairless mice, the expression of melanogenesis-related factor tyrosinase, nitric oxide, and cAMP and protein expression of pPKA, pCREBa, MITF, TRP-1and TRP-2 was significantly reduced. In particular, 100 mg/kg of CV exhibited an excellent effect similar to control group. Therefore, we suggest the possibility of developing CV as a food supplement having skin whitening effects by ameliorating melanogenesis.
Collapse
Affiliation(s)
- SukJin Kim
- Department of R&I Center, COSMAXBIO, Seongnam, 13486, Republic of Korea
| | - Gwi Hwan Na
- Department of R&I Center, COSMAXBIO, Seongnam, 13486, Republic of Korea
| | - Hyun Mook Jung
- Department of R&I Center, COSMAXBIO, Seongnam, 13486, Republic of Korea
| | - Sang Hun Han
- Almas Caviar, Hwaseoung-si, 18553, Republic of Korea
| | - Jehee Han
- Almas Caviar, Hwaseoung-si, 18553, Republic of Korea
| | - Yean Kyoung Koo
- Department of R&I Center, COSMAXBIO, Seongnam, 13486, Republic of Korea.
| |
Collapse
|
19
|
Shahid NH, Rashid H, Kumar S, Archoo S, Umar SA, Nazir LA, Parvinder SP, Tasduq SA. Inhibition of melanogenesis by 3-(1'-methyltetrahydropyridinyl)-2,4-6-trihydroxy acetophenone via suppressing the activity of cAMP response element-binding protein (CREB) and nuclear exclusion of CREB-regulated transcription coactivator 1 (CRTC1). Eur J Pharmacol 2023:175734. [PMID: 37080332 DOI: 10.1016/j.ejphar.2023.175734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/03/2023] [Accepted: 04/17/2023] [Indexed: 04/22/2023]
Abstract
Exposure to Ultraviolet radiation or α-melanocyte-stimulating hormone (α-MSH) stimulates the Cyclic Adenosine Monophosphate/Protein Kinase A signalling pathway, which leads to the synthesis and deposition of melanin granules in the epidermis. Skin pigmentation is the major physiological defence against inimical effects of sunlight. However, excessive melanin production and accumulation can cause various skin hyperpigmentation disorders. The present study involved the identification of 3-(1'-methyltetrahydropyridinyl)-2,4-6-trihydroxy acetophenone (IIIM-8) as an inhibitor of melanogenesis, IIIM-8 significantly inhibited pigment production both invitro and invivowithout incurring any cytotoxicity in Human Adult Epidermal Melanocytes (HAEM). IIIM-8 repressed melanin synthesis and secretion both at basal levels and in α-MSH stimulated cultured HAEM cells by decreasing the levels of Cyclic Adenosine Monophosphate (cAMP) and inhibiting the phosphorylation of cAMP response element-binding (CREB) protein, coupled with restoring the phosphorylation of CREB-regulated transcription coactivator 1 (CRTC1) and its nuclear exclusion in HAEM cells. This impeding effect correlates with diminished expression of master melanogenic proteins including microphthalmia-associated transcription factor (MITF), Tyrosinase (TYR), Tyrosinase related protein 1 (TRP1), and Tyrosinase related protein 2 (TRP2). Additionally, topical application of IIIM-8 induced tail depigmentation in C57BL/6 J mice. Furthermore, IIIM-8 efficiently mitigated the effect of ultraviolet-B radiation on melanin synthesis in the auricles of C57BL/6 J mice. This study demonstrates that IIIM-8 is an active anti-melanogenic agent against ultraviolet radiation-induced melanogenesis and other hyperpigmentation disorders.
Collapse
Affiliation(s)
- Naikoo H Shahid
- Pharmacology Division, Council of Scientific and Industrial Research-Indian Institute of Integrative Medicine, Canal Road, Jammu, Jammu and Kashmir, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Haroon Rashid
- Sher-e-KashmirInstitute of Medical Sciences, Soura, Srinagar, 190011, Jammu and Kashmir, India
| | - Sanjay Kumar
- Natural Product and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu Tawi, Jammu and Kashmir, India
| | - Sajida Archoo
- Pharmacology Division, Council of Scientific and Industrial Research-Indian Institute of Integrative Medicine, Canal Road, Jammu, Jammu and Kashmir, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sheikh A Umar
- Pharmacology Division, Council of Scientific and Industrial Research-Indian Institute of Integrative Medicine, Canal Road, Jammu, Jammu and Kashmir, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Lone A Nazir
- Pharmacology Division, Council of Scientific and Industrial Research-Indian Institute of Integrative Medicine, Canal Road, Jammu, Jammu and Kashmir, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Singh P Parvinder
- Natural Product and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu Tawi, Jammu and Kashmir, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sheikh A Tasduq
- Pharmacology Division, Council of Scientific and Industrial Research-Indian Institute of Integrative Medicine, Canal Road, Jammu, Jammu and Kashmir, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
20
|
Tamura Y, Ito A, Wakamatsu K, Torigoe T, Honda H, Ito S, Jimbow K. A Sulfur Containing Melanogenesis Substrate, N-Pr-4-S-CAP as a Potential Source for Selective Chemoimmunotherapy of Malignant Melanoma. Int J Mol Sci 2023; 24:ijms24065235. [PMID: 36982309 PMCID: PMC10049105 DOI: 10.3390/ijms24065235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/04/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
N-propionyl-4-S-cysteaminylphenol (N-Pr-4-S-CAP) is a substrate for tyrosinase, which is a melanin biosynthesis enzyme and has been shown to be selectively incorporated into melanoma cells. It was found to cause selective cytotoxicity against melanocytes and melanoma cells after selective incorporation, resulting in the induction of anti-melanoma immunity. However, the underlying mechanisms for the induction of anti-melanoma immunity remain unclear. This study aimed to elucidate the cellular mechanism for the induction of anti-melanoma immunity and clarify whether N-Pr-4-S-CAP administration could be a new immunotherapeutic approach against melanoma, including local recurrence and distant metastasis. A T cell depletion assay was used for the identification of the effector cells responsible for N-Pr-4-S-CAP-mediated anti-melanoma immunity. A cross-presentation assay was carried out by using N-Pr-4-S-CAP-treated B16-OVA melanoma-loaded bone marrow-derived dendritic cells (BMDCs) and OVA-specific T cells. Administration of N-Pr-4-S-CAP induced CD8+ T cell-dependent anti-melanoma immunity and inhibited the growth of challenged B16F1 melanoma cells, indicating that the administration of N-Pr-4-S-CAP can be a prophylactic therapy against recurrence and metastasis of melanoma. Moreover, intratumoral injection of N-Pr-4-S-CAP in combination with BMDCs augmented the tumor growth inhibition when compared with administration of N-Pr-4-S-CAP alone. BMDCs cross-presented a melanoma-specific antigen to CD8+ T cells through N-Pr-4-S-CAP-mediated melanoma cell death. Combination therapy using N-Pr-4-S-CAP and BMDCs elicited a superior anti-melanoma effect. These results suggest that the administration of N-Pr-4-S-CAP could be a new strategy for the prevention of local recurrence and distant metastasis of melanoma.
Collapse
Affiliation(s)
- Yasuaki Tamura
- Department of Pathology, Sapporo Medical University School of Medicine, South 1 West 16, Chuo-ku, Sapporo 060-8556, Hokkaido, Japan;
- Correspondence: ; Tel.: +81-(0)11-611-2111; Fax: +81-(0)11-643-2310
| | - Akira Ito
- Department of Chemical Systems Engineering, School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Aichi, Japan;
| | - Kazumasa Wakamatsu
- Institute for Melanin Chemistry, Fujita Health University, 1-98 Degakugakubo, Kutsukake-cho, Toyoake 470-1192, Aichi, Japan; (K.W.); (S.I.)
| | - Toshihiko Torigoe
- Department of Pathology, Sapporo Medical University School of Medicine, South 1 West 16, Chuo-ku, Sapporo 060-8556, Hokkaido, Japan;
| | - Hiroyuki Honda
- Department of Biomolecular Engineering, School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Aichi, Japan;
| | - Shosuke Ito
- Institute for Melanin Chemistry, Fujita Health University, 1-98 Degakugakubo, Kutsukake-cho, Toyoake 470-1192, Aichi, Japan; (K.W.); (S.I.)
| | - Kowichi Jimbow
- Institute of Dermatology & Cutaneous Sciences, 1-27 Odori West 17, Chuo-ku, Sapporo 060-0042, Hokkaido, Japan;
| |
Collapse
|
21
|
Anti-melanogenic effect of Moju through inhibition of tyrosinase activity. Mol Cell Toxicol 2023. [DOI: 10.1007/s13273-022-00329-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Abstract
Background
Moju is a traditional rice beverage local to Jeonju with an alcohol content of 1–2%. Moju is made by boiling makgeolli with several kinds of medicinal herbs, such as ginger, jujube and cinnamon. The raw materials used in Moju are well known for their physiological and functional effects. Although Moju is made with functional raw materials, the operational role of Moju has not yet been reported.
Objectives
The aim of this study was to identify the anti-melanogenic effects of Moju in B16F10 melanoma cells and explore the potential mechanisms.
Results
In this study, we investigated the antioxidant activity and anti-melanogenic effect of Moju. Moju showed no toxicity to HEK293T or B16F10 cells. The antioxidant activity of Moju was confirmed by its ability to increase radical scavenging activity. Moju decreased tyrosinase activity in a concentration-dependent manner. At the cellular level, Moju reduced melanin synthesis and the expression of proteins involved in melanin synthesis at concentrations of 100, 250, and 500 μg/mL in B16F10 cells. In addition, Moju inhibited the phosphorylation of extracellular signal-regulated kinase (ERK).
Conclusions
These results provide evidence that Moju has antioxidant activity and anti-melanogenic effect that occur through regulation of the ERK pathway. Although further research is needed to elucidate the specific mechanism and functional components, the ability of Moju to inhibit melanin synthesis by altering tyrosinase activation suggest that it can be used as a functional whitening ingredient.
Collapse
|
22
|
Athapaththu AMGK, Sanjaya SS, Lee KT, Karunarathne WAHM, Choi YH, Hur SP, Kim GY. Pinostrobin Suppresses the α-Melanocyte-Stimulating Hormone-Induced Melanogenic Signaling Pathway. Int J Mol Sci 2023; 24:ijms24010821. [PMID: 36614262 PMCID: PMC9821324 DOI: 10.3390/ijms24010821] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/27/2022] [Accepted: 12/27/2022] [Indexed: 01/05/2023] Open
Abstract
Pinostrobin is a dietary flavonoid found in several plants that possesses pharmacological properties, such as anti-cancer, anti-virus, antioxidant, anti-ulcer, and anti-aromatase effects. However, it is unclear if pinostrobin exerts anti-melanogenic properties and, if so, what the underlying molecular mechanisms comprise. Therefore, we, in this study, investigated whether pinostrobin inhibits melanin biosynthesis in vitro and in vivo, as well as the potential associated mechanism. Pinostrobin reduced mushroom tyrosinase activity in vitro in a concentration-dependent manner, with an IC50 of 700 μM. Molecular docking simulations further revealed that pinostrobin forms a hydrogen bond, as well as other non-covalent interactions, between the C-type lectin-like fold and polyphenol oxidase chain, rather than the previously known copper-containing catalytic center. Additionally, pinostrobin significantly decreased α-melanocyte-stimulating hormone (α-MSH)-induced extracellular and intracellular melanin production, as well as tyrosinase activity, in B16F10 melanoma cells. More specifically, pinostrobin inhibited the α-MSH-induced melanin biosynthesis signaling pathway by suppressing the cAMP-CREB-MITF axis. In fact, pinostrobin also attenuated pigmentation in α-MSH-stimulated zebrafish larvae without causing cardiotoxicity. The findings suggest that pinostrobin effectively inhibits melanogenesis in vitro and in vivo via regulation of the cAMP-CREB-MITF axis.
Collapse
Affiliation(s)
| | | | - Kyoung Tae Lee
- Forest Bioresources Department, Forest Microbiology Division, National Institute of Forest Science, Suwon 16631, Republic of Korea
| | | | - Yung Hyun Choi
- Department of Biochemistry, College of Korean Medicine, Dong-Eui University, Busan 47227, Republic of Korea
| | - Sung-Pyo Hur
- Department of Marine Life Science, Jeju National University, Jeju 63243, Republic of Korea
| | - Gi-Young Kim
- Department of Marine Life Science, Jeju National University, Jeju 63243, Republic of Korea
- Correspondence:
| |
Collapse
|
23
|
Study on the Mechanism of miR-125b-5p Affecting Melanocyte Biological Behavior and Melanogenesis in Vitiligo through Regulation of MITF. DISEASE MARKERS 2022; 2022:6832680. [DOI: 10.1155/2022/6832680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/18/2022] [Accepted: 08/23/2022] [Indexed: 11/18/2022]
Abstract
Objective. The goal was to confirm the mechanism by which miR-125b-5p influences melanocyte biological behavior and melanogenesis in vitiligo by regulating MITF. Methods. oe-MITF, sh-MITF, miR-125b-5p mimic, NC-mimic, NC-inhibitor, and miR-125b-5p inhibitor were transfected into cells by cell transfection. Western blotting was used to detect the related protein expression, qRT–PCR was used to detect miR-125b-5p and MITF expression, immunohistochemistry was used to detect the MITF-positive cells in vitiligo patients tissues, and a dual-luciferase reporter system was used to detect the target of miR-125b-5p and MITF. PIG1 and PIG3V cell proliferation by the CCK-8 method, cell cycle progression and apoptosis by flow cytometry, apoptosis was detected by TUNEL, Tyr activity and melanin content were measured using Tyr and melanin content assay kits. Results. Compared with the healthy control group, the expression of miR-125b-5p in the tissues and serum of vitiligo patients was upregulated, and the expression of MITF was downregulated; compared with PIG1 cells, the expression of miR-125b-5p and MITF in the PIG3V group was consistent with the above. Compared with the NC-minic group, the cell proliferation activity of the miR-125b-5p mimic group decreased, apoptosis increased, and the expression levels of melanogenesis-related proteins Tyr, Tyrp1, Tyrp2, and DCT were downregulated. Compared with the NC-inhibitor group, the above indices in the miR-125b-5p inhibitor group were all opposite to those in the miR-125b-5p mimic group. Transfection of oe-MITF into the miR-125b-5p mimic group reversed the effect of the miR-125b-5p mimic, while transfection of sh-MITF enhanced the effect of the miR-125b-5p mimic. Conclusion. miR-125b-5p affects vitiligo melanocyte biological behavior and melanogenesis by downregulating MITF expression.
Collapse
|
24
|
Uto T, Tung NH, Ohta T, Shoyama Y. (+)-Magnolin Enhances Melanogenesis in Melanoma Cells and Three-Dimensional Human Skin Equivalent; Involvement of PKA and p38 MAPK Signaling Pathways. PLANTA MEDICA 2022; 88:1199-1208. [PMID: 35211932 DOI: 10.1055/a-1740-7325] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Magnoliae Flos is a traditional herbal medicine used to treat nasal congestion associated with headache, empyema, and allergic rhinitis. In our preliminary screening of crude drugs used in Japanese Kampo formulas for melanin synthesis, the methanol extract of Magnoliae Flos was found to exhibit strong melanin synthesis activity. However, there have been no studies evaluating the effects of Magnoliae Flos or its constituents on melanogenesis. The present study aimed to isolate the active compounds from Magnoliae Flos that activate melanin synthesis in melanoma cells and three-dimensional human skin equivalent, and to investigate the molecular mechanism underlying melanin induction. The methanol extract of Magnoliae Flos induced an increase of melanin content in both B16-F1 and HMV-II cells. A comparison of melanin induction by three fractions prepared from the extract showed that the ethyl acetate fraction markedly induced melanin synthesis. Bioassay-guided separation of the ethyl acetate fraction resulted in the isolation of seven lignans (1: - 7: ). Among them, (+)-magnolin (5: ) strongly induced melanin synthesis and intracellular tyrosinase activity. Furthermore, the ethyl acetate fraction and 5: clearly induced melanin content in a three-dimensional human skin equivalent. Molecular analysis revealed that 5: triggered the protein expression of tyrosinase, tyrosinase-related protein-1, and tyrosinase-related protein-2. Further analysis of transcriptional factors and signaling pathways demonstrated that 5: induces the protein expression of tyrosinase, tyrosinase-related protein-1, and tyrosinase-related protein-2 activated by the protein kinase A- and p38 mitogen-activated protein kinase-dependent pathways, leading to cAMP-responsive element-binding protein phosphorylation and microphthalmia-associated transcription factor expression. These findings demonstrate the potential of 5: as a potent therapeutic agent for hypopigmentation.
Collapse
Affiliation(s)
- Takuhiro Uto
- Department of Pharmacognosy, Faculty of Pharmaceutical Sciences, Nagasaki International University, Sasebo, Nagasaki, Japan
| | - Nguyen Huu Tung
- Department of Pharmacognosy, Faculty of Pharmaceutical Sciences, Nagasaki International University, Sasebo, Nagasaki, Japan
- Faculty of Pharmacy, Phenikaa University, Hanoi, Vietnam
| | - Tomoe Ohta
- Department of Pharmacognosy, Faculty of Pharmaceutical Sciences, Nagasaki International University, Sasebo, Nagasaki, Japan
| | - Yukihiro Shoyama
- Department of Pharmacognosy, Faculty of Pharmaceutical Sciences, Nagasaki International University, Sasebo, Nagasaki, Japan
| |
Collapse
|
25
|
Immunomodulation of Melanoma by Chemo-Thermo-Immunotherapy Using Conjugates of Melanogenesis Substrate NPrCAP and Magnetite Nanoparticles: A Review. Int J Mol Sci 2022; 23:ijms23126457. [PMID: 35742905 PMCID: PMC9223671 DOI: 10.3390/ijms23126457] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 05/31/2022] [Accepted: 06/07/2022] [Indexed: 12/30/2022] Open
Abstract
A major advance in drug discovery and targeted therapy directed at cancer cells may be achieved by the exploitation and immunomodulation of their unique biological properties. This review summarizes our efforts to develop novel chemo-thermo-immunotherapy (CTI therapy) by conjugating a melanogenesis substrate, N-propionyl cysteaminylphenol (NPrCAP: amine analog of tyrosine), with magnetite nanoparticles (MNP). In our approach, NPrCAP provides a unique drug delivery system (DDS) because of its selective incorporation into melanoma cells. It also functions as a melanoma-targeted therapeutic drug because of its production of highly reactive free radicals (melanoma-targeted chemotherapy). Moreover, the utilization of MNP is a platform to develop thermo-immunotherapy because of heat shock protein (HSP) expression upon heat generation in MNP by exposure to an alternating magnetic field (AMF). This comprehensive review covers experimental in vivo and in vitro mouse melanoma models and preliminary clinical trials with a limited number of advanced melanoma patients. We also discuss the future directions of CTI therapy.
Collapse
|
26
|
Casalou C, Moreiras H, Mayatra JM, Fabre A, Tobin DJ. Loss of 'Epidermal Melanin Unit' Integrity in Human Skin During Melanoma-Genesis. Front Oncol 2022; 12:878336. [PMID: 35574390 PMCID: PMC9097079 DOI: 10.3389/fonc.2022.878336] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/09/2022] [Indexed: 12/13/2022] Open
Abstract
Cutaneous melanoma can be a most challenging neoplasm of high lethality, in part due to its extreme heterogeneity and characteristic aggressive and invasive nature. Indeed, its moniker 'the great masquerader' reflects that not all melanomas are created equal in terms of their originating cellular contexts, but also that melanoma cells in the malignant tumor can adopt a wide range of different cell states and variable organotropism. In this review, we focus on the early phases of melanomagenesis by discussing how the originating pigment cell of the melanocyte lineage can be influenced to embark on a wide range of tumor fates with distinctive microanatomical pathways. In particular, we assess how cells of the melanocyte lineage can differ by maturation status (stem cell; melanoblast; transiently amplifying cell; differentiated; post-mitotic; terminally-differentiated) as well as by micro-environmental niche (in the stratum basale of the epidermis; within skin appendages like hair follicle, eccrine gland, etc). We discuss how the above variable contexts may influence the susceptibility of the epidermal-melanin unit (EMU) to become unstable, which may presage cutaneous melanoma development. We also assess how unique features of follicular-melanin unit(s) (FMUs) can, by contrast, protect melanocytes from melanomagenesis. Lastly, we postulate how variable melanocyte fates in vitiligo, albinism, psoriasis, and alopecia areata may provide new insights into immune-/non immune-mediated outcomes for melanocytes in cutaneous melanin units.
Collapse
Affiliation(s)
- Cristina Casalou
- The Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin, Ireland
| | - Hugo Moreiras
- The Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin, Ireland
| | - Jay M Mayatra
- The Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin, Ireland
| | - Aurelie Fabre
- Department of Histopathology, St Vincent's University Hospital, Dublin, Ireland.,UCD School of Medicine, University College Dublin, Dublin, Ireland.,The Conway Institute of Biomedical and Biomolecular Science, University College Dublin, Dublin, Ireland
| | - Desmond J Tobin
- The Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin, Ireland.,The Conway Institute of Biomedical and Biomolecular Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
27
|
Joyjamras K, Netcharoensirisuk P, Roytrakul S, Chanvorachote P, Chaotham C. Recycled Sericin Hydrolysates Modified by Alcalase ® Suppress Melanogenesis in Human Melanin-Producing Cells via Modulating MITF. Int J Mol Sci 2022; 23:3925. [PMID: 35409284 PMCID: PMC8999004 DOI: 10.3390/ijms23073925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 03/27/2022] [Accepted: 03/31/2022] [Indexed: 11/17/2022] Open
Abstract
Because available depigmenting agents exhibit short efficacy and serious side effects, sericin, a waste protein from the silk industry, was hydrolyzed using Alcalase® to evaluate its anti-melanogenic activity in human melanin-producing cells. Sericin hydrolysates consisted of sericin-related peptides in differing amounts and smaller sizes compared with unhydrolyzed sericin, as respectively demonstrated by peptidomic and SDS-PAGE analysis. The lower half-maximum inhibitory concentration (9.05 ± 0.66 mg/mL) compared with unhydrolyzed sericin indicated a potent effect of sericin hydrolysates on the diminution of melanin content in human melanoma MNT1 cells. Not only inhibiting enzymatic activity but also a downregulated expression level of tyrosinase was evident in MNT1 cells incubated with 20 mg/mL sericin hydrolysates. Quantitative RT-PCR revealed the decreased mRNA level of microphthalmia-associated transcription factor (MITF), a tyrosinase transcription factor, which correlated with the reduction of pCREB/CREB, an upstream cascade, as assessed by Western blot analysis in MNT1 cells cultured with 20 mg/mL sericin hydrolysates for 12 h. Interestingly, treatment with sericin hydrolysates for 6-24 h also upregulated pERK, a molecule that triggers MITF degradation, in human melanin-producing cells. These results warrant the recycling of wastewater from the silk industry for further development as a safe and effective treatment of hyperpigmentation disorders.
Collapse
Affiliation(s)
- Keerati Joyjamras
- Graduate Program of Pharmaceutical Sciences and Technology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10300, Thailand;
| | - Ponsawan Netcharoensirisuk
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Sittiruk Roytrakul
- Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani 12120, Thailand;
| | - Pithi Chanvorachote
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Chatchai Chaotham
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand;
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
28
|
Moon SY, Akter KM, Ahn MJ, Kim KD, Yoo J, Lee JH, Lee JH, Hwangbo C. Fraxinol Stimulates Melanogenesis in B16F10 Mouse Melanoma Cells through CREB/MITF Signaling. Molecules 2022; 27:1549. [PMID: 35268650 PMCID: PMC8911637 DOI: 10.3390/molecules27051549] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/18/2022] [Accepted: 02/22/2022] [Indexed: 12/10/2022] Open
Abstract
Melanin pigment produced in melanocytes plays a protective role against ultraviolet radiation. Selective destruction of melanocytes causes chronic depigmentation conditions such as vitiligo, for which there are very few specific medical treatments. Here, we found that fraxinol, a natural coumarin from Fraxinus plants, effectively stimulated melanogenesis. Treatment of B16-F10 cells with fraxinol increased the melanin content and tyrosinase activity in a concentration-dependent manner without causing cytotoxicity. Additionally, fraxinol enhanced the mRNA expression of melanogenic enzymes such as tyrosinase, tyrosinase-related protein-1, and tyrosinase-related protein-2. Fraxinol also increased the expression of microphthalmia-associated transcription factor at both mRNA and protein levels. Fraxinol upregulated the phosphorylation of cyclic adenosine monophosphate (cAMP) response element-binding protein (CREB). Furthermore, H89, a cAMP-dependent protein kinase A inhibitor, decreased fraxinol-induced CREB phosphorylation and microphthalmia-associated transcription factor expression and significantly attenuated the fraxinol-induced melanin content and intracellular tyrosinase activity. These results suggest that fraxinol enhances melanogenesis via a protein kinase A-mediated mechanism, which may be useful for developing potent melanogenesis stimulators.
Collapse
Affiliation(s)
- Sun Young Moon
- Division of Applied Life Science (BK21), PMBBRC and Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, Korea; (S.Y.M.); (K.D.K.); (J.Y.)
- Division of Life Science, College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea
| | - Kazi-Marjahan Akter
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Korea; (K.-M.A.); (M.-J.A.)
| | - Mi-Jeong Ahn
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Korea; (K.-M.A.); (M.-J.A.)
| | - Kwang Dong Kim
- Division of Applied Life Science (BK21), PMBBRC and Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, Korea; (S.Y.M.); (K.D.K.); (J.Y.)
- Division of Life Science, College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea
| | - Jiyun Yoo
- Division of Applied Life Science (BK21), PMBBRC and Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, Korea; (S.Y.M.); (K.D.K.); (J.Y.)
- Division of Life Science, College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea
| | - Joon-Hee Lee
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52828, Korea;
| | - Jeong-Hyung Lee
- Department of Biochemistry (BK21), College of Natural Sciences, Kangwon National University, Chuncheon 24341, Korea;
| | - Cheol Hwangbo
- Division of Applied Life Science (BK21), PMBBRC and Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, Korea; (S.Y.M.); (K.D.K.); (J.Y.)
- Division of Life Science, College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea
| |
Collapse
|
29
|
Yan J, Ma LP, Liu F, Sun B, Tian M, Lu X, Liu HX, Gao L, Liu QJ. Effect of Ultraviolet B Irradiation on Melanin Content Accompanied by the Activation of p62/GATA4-Mediated Premature Senescence in HaCaT Cells. Dose Response 2022; 20:15593258221075321. [PMID: 35185418 PMCID: PMC8848062 DOI: 10.1177/15593258221075321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 01/01/2021] [Indexed: 11/17/2022] Open
Abstract
Objective To explore the effect and mechanism of ultraviolet B (UVB) on melanin synthesis and premature senescence in human immortalized keratinocytes (HaCaT) cells. Methods HaCaT cells were irradiated with 0, 20, 50, 80, 100, 150, and 200 mJ/cm2 of UVB. NaOH method was used for melanin content assay, cellular tyrosinase (TYR) activity was determined by 3,4-Dihydroxy-L-phenylalanine (L-DOPA) oxidation to dopachrome, premature senescence was analyzed by senescence-associated beta-galactosidase (SA-β-gal) staining kit, and the levels of p21, p16, p62, and GATA4 proteins were detected by Western blotting. Premature senescence was inhibited by the inhibitors of ataxia telangiectasia mutated (ATM) or ataxia telangiectasia and Rad3–related (ATR), and the p53 signaling pathway was activated by Nutlin-3. The mRNA levels of senescence-associated secretory phenotype (SASP) factors including tumor necrosis factor alpha (TNF-α), vascular endothelial growth factor A (VEGF-A), and interleukin-8 (IL-8) were measured by real-time quantitative polymerase chain reaction in HaCaT cells after 80 mJ/cm2 of UVB irradiation. Results The melanin level increased significantly with the elevation of irradiation dose ( F = 28.19, 43.82, 143.60, P < .05), reaching the peak at the dose of 80 mJ/cm2. The tyrosinase activity increased significantly ( F = 84.50, P < .05), the percentage of premature senescence increased ( F = 16.31, P < .05), the levels of p62 decreased, and the level of GATA4 increased obviously with the increase of UVB dose after irradiation. The UVB-induced promotion of GATA4 level was significantly inhibited by being treated with ATM or ATR inhibitor. However, this did not occur in the Nutlin-3-treated group. The mRNA and protein expression of TNF-α increased significantly at 72 h at 80 mJ/cm2 of UVB irradiation. Conclusions Melanin contents increased first and decreased afterward with the increasing of UVB irradiation. The decrease of p62-mediated selective autophagy was accompanied by the accumulation of GATA4 after different doses of UVB irradiation. Activation of this p62/GATA4 pathway depends on the ATM and ATR but is independent of p53, and the SASP factor was activated in HaCaT cells at 80 mJ/cm2 of UVB irradiation.
Collapse
Affiliation(s)
- Juan Yan
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, People’s Republic of China
| | - Li-Ping Ma
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, People’s Republic of China
| | - Fang Liu
- Department of Dermatology, Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Bo Sun
- PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Mei Tian
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, People’s Republic of China
| | - Xue Lu
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, People’s Republic of China
| | - Hai-Xiang Liu
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, People’s Republic of China
| | - Ling Gao
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, People’s Republic of China
| | - Qing-Jie Liu
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, People’s Republic of China
| |
Collapse
|
30
|
Matias M, Pinho JO, Penetra MJ, Campos G, Reis CP, Gaspar MM. The Challenging Melanoma Landscape: From Early Drug Discovery to Clinical Approval. Cells 2021; 10:3088. [PMID: 34831311 PMCID: PMC8621991 DOI: 10.3390/cells10113088] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/02/2021] [Accepted: 11/06/2021] [Indexed: 02/06/2023] Open
Abstract
Melanoma is recognized as the most dangerous type of skin cancer, with high mortality and resistance to currently used treatments. To overcome the limitations of the available therapeutic options, the discovery and development of new, more effective, and safer therapies is required. In this review, the different research steps involved in the process of antimelanoma drug evaluation and selection are explored, including information regarding in silico, in vitro, and in vivo experiments, as well as clinical trial phases. Details are given about the most used cell lines and assays to perform both two- and three-dimensional in vitro screening of drug candidates towards melanoma. For in vivo studies, murine models are, undoubtedly, the most widely used for assessing the therapeutic potential of new compounds and to study the underlying mechanisms of action. Here, the main melanoma murine models are described as well as other animal species. A section is dedicated to ongoing clinical studies, demonstrating the wide interest and successful efforts devoted to melanoma therapy, in particular at advanced stages of the disease, and a final section includes some considerations regarding approval for marketing by regulatory agencies. Overall, considerable commitment is being directed to the continuous development of optimized experimental models, important for the understanding of melanoma biology and for the evaluation and validation of novel therapeutic strategies.
Collapse
Affiliation(s)
- Mariana Matias
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (M.M.); (J.O.P.); (M.J.P.)
| | - Jacinta O. Pinho
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (M.M.); (J.O.P.); (M.J.P.)
| | - Maria João Penetra
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (M.M.); (J.O.P.); (M.J.P.)
| | - Gonçalo Campos
- CICS–UBI–Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6201-506 Covilhã, Portugal;
| | - Catarina Pinto Reis
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (M.M.); (J.O.P.); (M.J.P.)
| | - Maria Manuela Gaspar
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (M.M.); (J.O.P.); (M.J.P.)
| |
Collapse
|
31
|
Yin SJ, Qian GY, Yang JM, Lee J, Park YD. Detection of melanogenesis- and anti-apoptosis-associated melanoma factors: Array CGH and PPI mapping integrating study. Protein Pept Lett 2021; 28:1408-1424. [PMID: 34749602 DOI: 10.2174/0929866528666211105112927] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 08/02/2021] [Accepted: 08/26/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND We investigated melanogenesis- and anti-apoptosis-related melanoma factors in melanoma cells (TXM1, TXM18, A375P, and A375SM). OBJECTIVE To find melanoma associated hub factor, high-throughput screening-based techniques integrating with bioinformatics were investigated. METHODS Array CGH analysis was conducted with a commercial system. Total genomic DNAs prepared individually from each cell line with control DNA were properly labeled with Cy3-dCTP and Cy5-dCTP and hybridizations and subsequently performed data treatment by the log2 green (G; test) to red (R; reference) fluorescence ratios (G/R). Gain or loss of copy number was judged by spots with log2-transformed ratios. PPI mapping analysis of detected candidate genes based on the array CGH results was conducted using the human interactome in the STRING database. Energy minimization and a short molecular dynamics (MD) simulation using the implicit solvation model in CHARMM were performed to analyze the interacting residues between YWHAZ and YWHAB. RESULTS Three genes (BMP-4, BFGF, LEF-1) known to be involved in melanogenesis were found to lose chromosomal copy numbers, and Chr. 6q23.3 was lost in all tested cell lines. Ten hub genes (CTNNB1, PEX13, PEX14, PEX5, IFNG, EXOSC3, EXOSC1, EXOSC8, UBC, and PEX10) were predicted to be functional interaction factors in the network of the 6q23.3 locus. The apoptosis-associated genes E2F1, p50, BCL2L1, and BIRC7 gained, and FGF2 lost chromosomal copy numbers in the tested melanoma cell lines. YWHAB, which gained chromosomal copy numbers, was predicted to be the most important hub protein in melanoma cells. Molecular dynamics simulations for binding YWHAB and YWHAZ were conducted, and the complex was predicted to be energetically and structurally stable through its 3 hydrogen-bond patterns. The number of interacting residues is 27. CONCLUSION Our study compares genome-wide screening interactomics predictions for melanoma factors and offers new information for understanding melanogenesis- and anti-apoptosis-associated mechanisms in melanoma. Especially, YWHAB was newly detected as a core factor in melanoma cells.
Collapse
Affiliation(s)
- Shang-Jun Yin
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo 315100. China
| | - Guo-Ying Qian
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo 315100. China
| | - Jun-Mo Yang
- Department of Dermatology, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul 135-710. Korea
| | - Jinhyuk Lee
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Gwahak-ro, Yuseong-gu, Daejeon, 34141. Korea
| | - Yong-Doo Park
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo 315100. China
| |
Collapse
|
32
|
Cajanin Suppresses Melanin Synthesis through Modulating MITF in Human Melanin-Producing Cells. Molecules 2021; 26:molecules26196040. [PMID: 34641584 PMCID: PMC8512678 DOI: 10.3390/molecules26196040] [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: 07/27/2021] [Revised: 10/03/2021] [Accepted: 10/03/2021] [Indexed: 11/17/2022] Open
Abstract
Despite its classification as a non-life-threatening disease, increased skin pigmentation adversely affects quality of life and leads to loss of self-confidence. Until now, there are no recommended remedies with high efficacy and human safety for hyperpigmentation. This study aimed to investigate anti-melanogenic activity and underlying mechanism of cajanin, an isoflavonoid extracted from Dalbergia parviflora Roxb. (Leguminosae) in human melanin-producing cells. Culture with 50 μM cajanin for 48–72 h significantly suppressed proliferation in human melanoma MNT1 cells assessed via MTT viability assay. Interestingly, cajanin also efficiently diminished melanin content in MNT1 cells with the half maximum inhibitory concentration (IC50) at 77.47 ± 9.28 μM. Instead of direct inactivating enzymatic function of human tyrosinase, down-regulated mRNA and protein expression levels of MITF and downstream melanogenic enzymes, including tyrosinase, TRP-1 and Dct (TRP-2) were observed in MNT1 cells treated with 50 μM cajanin for 24–72 h. Correspondingly, treatment with cajanin modulated the signaling pathway of CREB and ERK which both regulate MITF expression level. Targeted suppression on MITF-related proteins in human melanin-producing cells strengthens the potential development of cajanin as an effective treatment for human hyperpigmented disorders.
Collapse
|
33
|
Synergistic Effect of 300 μm Needle-Depth Fractional Microneedling Radiofrequency on the Treatment of Senescence-Induced Aging Hyperpigmentation of the Skin. Int J Mol Sci 2021; 22:ijms22147480. [PMID: 34299100 PMCID: PMC8305123 DOI: 10.3390/ijms22147480] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 01/08/2023] Open
Abstract
Aging-associated dermatological pigmentary diseases are associated with accumulation of senescence cells and the disruption of basement membrane due to chronic ultraviolet radiation (UVR) exposure. Our study is on the synergistic effect of the novel 300 μm needle-depth fractional microneedling radiofrequency (FMR) treatment and conventional Q-switched ND:YAG laser on aging-associated hyperpigmentation of the skin. The prospective controlled clinical trial of 25 Asian women revealed significantly higher improvements not only on wrinkles, but also on hyperpigmentation. Additional ex vivo study revealed significant reduction of pro-melanogenic markers as well as senescent keratinocytes, while increased expression of collagen type IV on the epidermal basement membrane, after additional FMR treatment on UV-irradiated human tissues. These results demonstrate that 300 μm needle-depth FMR might effectively remove senescent keratinocytes that secrete pro-melanogenic markers, and repair disrupted basement membrane, therefore preventing constant hyperpigmentation of the aged skin.
Collapse
|
34
|
Targeting Melanoma-Initiating Cells by Caffeine: In Silico and In Vitro Approaches. Molecules 2021; 26:molecules26123619. [PMID: 34199192 PMCID: PMC8231844 DOI: 10.3390/molecules26123619] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 12/17/2022] Open
Abstract
The beneficial effects of coffee on human diseases are well documented, but the molecular mechanisms of its bioactive compounds on cancer are not completely elucidated. This is likely due to the large heterogeneity of coffee preparations and different coffee-based beverages, but also to the choice of experimental models where proliferation, differentiation and immune responses are differently affected. The aim of the present study was to investigate the effects of one of the most interesting bioactive compounds in coffee, i.e., caffeine, using a cellular model of melanoma at a defined differentiation level. A preliminary in silico analysis carried out on public gene-expression databases identified genes potentially involved in caffeine’s effects and suggested some specific molecular targets, including tyrosinase. Proliferation was investigated in vitro on human melanoma initiating cells (MICs) and cytokine expression was measured in conditioned media. Tyrosinase was revealed as a key player in caffeine’s mechanisms of action, suggesting a crucial role in immunomodulation through the reduction in IL-1β, IP-10, MIP-1α, MIP-1β and RANTES secretion onto MICs conditioned media. The potent antiproliferative effects of caffeine on MICs are likely to occur by promoting melanin production and reducing inflammatory signals’ secretion. These data suggest tyrosinase as a key player mediating the effects of caffeine on melanoma.
Collapse
|
35
|
Hushcha Y, Blo I, Oton-Gonzalez L, Mauro GD, Martini F, Tognon M, Mattei MD. microRNAs in the Regulation of Melanogenesis. Int J Mol Sci 2021; 22:ijms22116104. [PMID: 34198907 PMCID: PMC8201055 DOI: 10.3390/ijms22116104] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/21/2021] [Accepted: 06/03/2021] [Indexed: 12/11/2022] Open
Abstract
Melanogenesis is the process leading to the synthesis of melanin, the main substance that influences skin color and plays a pivotal role against UV damage. Altered melanogenesis is observed in several pigmentation disorders. Melanogenesis occurs in specialized cells called melanocytes, physically and functionally related by means of autocrine and paracrine interplay to other skin cell types. Several external and internal factors control melanin biosynthesis and operate through different intracellular signaling pathways, which finally leads to the regulation of microphthalmia-associated transcription factor (MITF), the key transcription factor involved in melanogenesis and the expression of the main melanogenic enzymes, including TYR, TYRP-1, and TYRP-2. Epigenetic factors, including microRNAs (miRNAs), are involved in melanogenesis regulation. miRNAs are small, single-stranded, non-coding RNAs, of approximately 22 nucleotides in length, which control cell behavior by regulating gene expression, mainly by binding the 3′ untranslated region (3′-UTR) of target mRNAs. This review collects data on the miRNAs involved in melanogenesis and how these miRNAs can modulate target gene expression. Bringing to light the biological function of miRNAs could lead to a wider understanding of epigenetic melanogenesis regulation and its dysregulation. This knowledge may constitute the basis for developing innovative treatment approaches for pigmentation dysregulation.
Collapse
Affiliation(s)
| | - Irene Blo
- Department of Medical Sciences, Section of Experimental Medicine, School of Medicine, University of Ferrara, 64b, Fossato di Mortara Street, 44121 Ferrara, Italy; (I.B.); (L.O.-G.); (G.D.M.); (F.M.); (M.T.)
| | - Lucia Oton-Gonzalez
- Department of Medical Sciences, Section of Experimental Medicine, School of Medicine, University of Ferrara, 64b, Fossato di Mortara Street, 44121 Ferrara, Italy; (I.B.); (L.O.-G.); (G.D.M.); (F.M.); (M.T.)
| | - Giulia Di Mauro
- Department of Medical Sciences, Section of Experimental Medicine, School of Medicine, University of Ferrara, 64b, Fossato di Mortara Street, 44121 Ferrara, Italy; (I.B.); (L.O.-G.); (G.D.M.); (F.M.); (M.T.)
| | - Fernanda Martini
- Department of Medical Sciences, Section of Experimental Medicine, School of Medicine, University of Ferrara, 64b, Fossato di Mortara Street, 44121 Ferrara, Italy; (I.B.); (L.O.-G.); (G.D.M.); (F.M.); (M.T.)
- Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Mauro Tognon
- Department of Medical Sciences, Section of Experimental Medicine, School of Medicine, University of Ferrara, 64b, Fossato di Mortara Street, 44121 Ferrara, Italy; (I.B.); (L.O.-G.); (G.D.M.); (F.M.); (M.T.)
| | - Monica De Mattei
- Department of Medical Sciences, Section of Experimental Medicine, School of Medicine, University of Ferrara, 64b, Fossato di Mortara Street, 44121 Ferrara, Italy; (I.B.); (L.O.-G.); (G.D.M.); (F.M.); (M.T.)
- Correspondence: ; Tel.: +39-0532-455534
| |
Collapse
|
36
|
Cellular Senescence and Inflammaging in the Skin Microenvironment. Int J Mol Sci 2021; 22:ijms22083849. [PMID: 33917737 PMCID: PMC8068194 DOI: 10.3390/ijms22083849] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/05/2021] [Accepted: 04/06/2021] [Indexed: 01/07/2023] Open
Abstract
Cellular senescence and aging result in a reduced ability to manage persistent types of inflammation. Thus, the chronic low-level inflammation associated with aging phenotype is called “inflammaging”. Inflammaging is not only related with age-associated chronic systemic diseases such as cardiovascular disease and diabetes, but also skin aging. As the largest organ of the body, skin is continuously exposed to external stressors such as UV radiation, air particulate matter, and human microbiome. In this review article, we present mechanisms for accumulation of senescence cells in different compartments of the skin based on cell types, and their association with skin resident immune cells to describe changes in cutaneous immunity during the aging process.
Collapse
|
37
|
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
|
38
|
Tan S, Zhao Z, Qiao Y, Zhang B, Zhang T, Zhang M, Qi J, Wang X, Meng M, Zhou Q. Activation of the tumor suppressive Hippo pathway by triptonide as a new strategy to potently inhibit aggressive melanoma cell metastasis. Biochem Pharmacol 2021; 185:114423. [PMID: 33476574 DOI: 10.1016/j.bcp.2021.114423] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/02/2021] [Accepted: 01/13/2021] [Indexed: 02/08/2023]
Abstract
Metastatic melanoma has a very high mortality rate despite the availability of chemotherapy, radiotherapy, and immunotherapy; therefore, more effective therapeutics are needed. The Hippo pathway plays an inhibitory role in melanoma progression, but the tumor suppressors Salvador homolog-1 (SAV1) and large tumor suppressor 1 (LATS1) in this pathway are down-regulated in melanoma. As a result, the downstream oncogenic Yes-associated protein (YAP) is active, resulting in uncontrolled melanoma growth and metastasis. Therapeutics for remedying SAV1 and LATS1 deficiency in melanoma have not yet been reported in the literature. Here, we show that the small molecule triptonide (MW 358 Da) robustly suppressed melanoma cell tumorigenicity, migration, and invasion. Furthermore, triptonide markedly reduced tumor growth and melanoma lung metastasis in tumor-bearing mice with low toxicity. Molecular mechanistic studies revealed that triptonide promoted SAV1 and LATS1 expression, strongly activated the tumor-suppressive Hippo pathway, degraded oncogenic YAP via the lysosomal pathway, and reduced levels of tumorigenic microphthalmia-associated transcription factor (MITF) in melanoma cells. Triptonide also strongly inhibited activation of AKT, a SAV1-binding signaling protein. Collectively, our results conceptually demonstrate that induction of SAV1 and LATS1 expression and activation of the tumor-suppressive Hippo pathway by triptonide potently inhibits aggressive melanoma cell growth and metastasis. These findings suggest a new strategy for developing therapeutics to treat metastatic melanoma and highlight a novel drug candidate against aggressive melanoma.
Collapse
Affiliation(s)
- Shijie Tan
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Zhe Zhao
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China; CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Jiangsu 215123, PR China
| | - Yingnan Qiao
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Bin Zhang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China; Center of Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, PR China; Suzhou Institute of Systems Medicine, Suzhou 215123, PR China
| | - Tong Zhang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Mengli Zhang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Jindan Qi
- School of Nursing, Soochow University, Suzhou, Jiangsu 215006, PR China
| | - Xiaohua Wang
- School of Nursing, Soochow University, Suzhou, Jiangsu 215006, PR China
| | - Mei Meng
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China.
| | - Quansheng Zhou
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China; State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, Jiangsu 215123, PR China; National Clinical Research Center for Hematology Diseases, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, PR China.
| |
Collapse
|
39
|
Yin SJ, Lee JR, Hahn MJ, Yang JM, Qian GY, Park YD. Tyrosinase-mediated melanogenesis in melanoma cells: Array comparative genome hybridization integrating proteomics and bioinformatics studies. Int J Biol Macromol 2020; 170:150-163. [PMID: 33359255 DOI: 10.1016/j.ijbiomac.2020.12.146] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 12/16/2020] [Accepted: 12/18/2020] [Indexed: 12/17/2022]
Abstract
We investigated the tyrosinase-associated melanogenesis in melanoma cells by using OMICS techniques. We characterized the chromosome copy numbers, including Chr 11q21 where the tyrosinase gene is located, from several melanoma cell lines (TXM13, G361, and SK-MEL-28) by using array CGH. We revealed that 11q21 is stable in TXM13 cells, which is directly related to a spontaneous high melanin pigment production. Meanwhile, significant loss of copy number of 11q21 was found in G361 and SK-MEL-28. We further profiled the proteome of TXM13 cells by LC-ESI-MSMS and detected more than 900 proteins, then predicted 11 hub proteins (YWHAZ; HSP90AA1; HSPA5; HSPA1L; HSPA9; HSP90B1; HSPA1A; HSPA8; FKSG30; ACTB; DKFZp686DQ972) by using an interactomic algorithm. YWHAZ (25% interaction in the network) is thought to be a most important protein as a linking factor between tyrosinase-triggered melanogenesis and melanoma growth. Bioinformatic tools were further applied for revealing various physiologic mechanisms and functional classification. The results revealed clues for the spontaneous pigmentation capability of TXM13 cells, contrary to G361 and SK-MEL-28 cells, which commonly have depigmentation properties during subculture. Our study comparatively conducted the genome-wide screening and proteomic profiling integrated interactomics prediction for TXM13 cells and suggests new insights for studying both melanogenesis and melanoma.
Collapse
Affiliation(s)
- Shang-Jun Yin
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo 315100, PR China
| | - Jae-Rin Lee
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Myong-Joon Hahn
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Jun-Mo Yang
- Department of Dermatology, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul 135-710, South Korea
| | - Guo-Ying Qian
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo 315100, PR China.
| | - Yong-Doo Park
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo 315100, PR China; Skin Diseases Research Center, Yangtze Delta Region Institute of Tsinghua University, 705 Yatai Road, Jiaxing 314006, PR China; Zhejiang Provincial Key Laboratory of Applied Enzymology, Yangtze Delta Region Institute of Tsinghua University, 705 Yatai Road, Jiaxing 314006, PR China.
| |
Collapse
|