Microbiota, Oxidative Stress, and Skin Cancer: An Unexpected Triangle
Abstract
:1. Introduction
2. Skin Cancer
2.1. Cutaneous Melanoma
2.2. Keratinocyte Carcinomas
3. Microbiota and Skin Cancer
3.1. Human Microbiota and Cutaneous Melanoma
3.2. Microbiota and Keratinocyte Skin Cancer
4. Microbiota and Oxidative Stress
4.1. Oxidative Stress in the Skin
4.2. Regulation of Redox Skin Level from Microbiota
5. Oxidative Stress and Skin Cancer
6. The Interplay between Microbiota, Oxidative Stress, and Skin Cancer
7. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
- Olsen, C.M.; Green, A.C.; Pandeya, N.; Whiteman, D.C. Trends in Melanoma Incidence Rates in Eight Susceptible Populations through 2015. J. Investig. Dermatol. 2019, 139, 1392–1395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Blazek, K.; Furestad, E.; Ryan, D.; Damian, D.; Fernandez-Penas, P.; Tong, S. The impact of skin cancer prevention efforts in New South Wales, Australia: Generational trends in melanoma incidence and mortality. Cancer Epidemiol. 2022, 26, 102263. [Google Scholar] [CrossRef] [PubMed]
- Ali, Z.; Yousaf, N.; Larkin, J. Melanoma epidemiology, biology and prognosis. EJC Suppl. 2013, 11, 81–91. [Google Scholar] [CrossRef] [Green Version]
- Kalaora, S.; Nagler, A.; Wargo, J.A.; Samuels, Y. Mechanisms of immune activation and regulation: Lessons from melanoma. Nat. Rev. Cancer 2022, 22, 195–207. [Google Scholar] [CrossRef] [PubMed]
- Saltman, B.E.; Ganly, I.; Patel, S.G.; Coit, D.G.; Brady, M.S.; Wong, R.J.; Boyle, J.O.; Singh, B.; Shaha, A.R.; Shah, J.P.; et al. Prognostic Implication of sentinel Lymph Node Biopsy in Cutaneous Head and Neck Melanoma. Head Neck. 2010, 32, 1686–1692. [Google Scholar] [CrossRef]
- Cherobin, A.C.F.P.; Wainstein, A.J.A.; Colosimo, E.A.; Goulart, E.M.A.; Bittencourt, F.V. Prognostic Factors for Metastasis in Cutaneous Melanoma. Bras. Dermatol. 2018, 93, 19–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Scolyer, R.A.; Long, G.V.; Thompson, J.F. Evolving Concepts in Melanoma Classification and Their Relevance to Multidisciplinary Melanoma Patient Care. Mol. Oncol. 2011, 5, 124–136. [Google Scholar] [CrossRef] [Green Version]
- Davis, L.E.; Shalin, S.C.; Tackett, A.J. Current State of Melanoma Diagnosis and Treatment. Cancer Biol. Ther. 2019, 20, 1366–1379. [Google Scholar] [CrossRef] [Green Version]
- Curtin, J.A.; Fridlyand, J.; Kageshita, T.; Patel, H.N.; Busam, K.J.; Kutzner, H.; Cho, K.H.; Aiba, S.; Bröcker, E.B.; LeBoit, P.E.; et al. Distinct sets of genetic alterations in melanoma. N. Engl. J. Med. 2005, 353, 2135–2147. [Google Scholar] [CrossRef]
- Fedorenko, I.V.; Gibney, G.T.; Smalley, K.S. NRAS mutant melanoma: Biological behavior and future strategies for therapeutic management. Oncogene 2013, 32, 3009–3018. [Google Scholar] [CrossRef] [Green Version]
- Yuan, T.L.; Cantley, L.C. PI3K pathway alterations in cancer: Variations on a theme. Oncogene 2008, 27, 5497–5510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Davies, M.A. The role of the PI3K-AKT pathway in melanoma. Cancer J. 2012, 18, 142–147. [Google Scholar] [CrossRef]
- Chiriboga, L.; Meehan, S.; Osman, I.; Glick, M.; de la Cruz, G.; Howell, B.S.; Friedman-Jiménez, G.; Schneider, R.J.; Jamal, S. Endothelin-1 in the tumor microenvironment correlates with melanoma invasion. Melanoma Res. 2016, 26, 236–244. [Google Scholar] [CrossRef] [PubMed]
- Moro, N.; Mauch, C.; Zigrino, P. Metalloproteinases in melanoma. Eur. J. Cell. Biol. 2014, 93, 23–29. [Google Scholar] [CrossRef] [PubMed]
- Sandri, S.; Faião-Flores, F.; Tiago, M.; Pennacchi, P.C.; Massaro, R.R.; Alves-Fernandes, D.K.; Berardinelli, G.N.; Evangelista, A.F.; de Lima Vazquez, V.; Reis, R.M.; et al. Vemurafenib resistance increases melanoma invasiveness and modulates the tumor microenvironment by MMP-2 upregulation. Pharmacol. Res. 2016, 111, 523–533. [Google Scholar] [CrossRef]
- Falzone, L.; Salemi, R.; Travali, S.; Scalisi, A.; McCubrey, J.A.; Candido, S.; Libra, M. MMP-9 overexpression is associated with intragenic hypermethylation of MMP9 gene in melanoma. Aging 2016, 8, 933–944. [Google Scholar] [CrossRef] [Green Version]
- Lee, K.R.; Lee, J.S.; Kim, Y.R.; Song, I.G.; Hong, E.K. Polysaccharide from Inonotus obliquus inhibits migration and invasion in B16-F10 cells by suppressing MMP-2 and MMP-9 via downregulation of NF-κB signaling pathway. Oncol. Rep. 2014, 31, 2447–2453. [Google Scholar] [CrossRef] [Green Version]
- Guarneri, C.; Bevelacqua, V.; Polesel, J.; Falzone, L.; Cannavò, P.S.; Spandidos, D.A.; Malaponte, G.; Libra, M. NF-κB inhibition is associated with OPN/MMP 9 downregulation in cutaneous melanoma. Oncol. Rep. 2017, 37, 737–746. [Google Scholar] [CrossRef] [Green Version]
- Hao, L.; Ha, J.R.; Kuzel, P.; Garcia, E.; Persad, S. Cadherin switch from E to N-cadherin in melanoma progression is regulated by the PI3K/PTEN pathway through twist and snail. Br. J. Derm. 2012, 166, 1184–1197. [Google Scholar] [CrossRef]
- Blass, E.; Ott, P.A. Advances in the development of personalized neoantigen-based therapeutic cancer vaccines. Nat. Rev. Clin. Oncol. 2021, 18, 215–229. [Google Scholar] [CrossRef] [PubMed]
- Croce, L.; Coperchini, F.; Magri, F.; Chiovato, L.; Rotondi, M. The multifaceted anti-cancer effects of BRAF-inhibitors. Oncotarget 2019, 10, 6623–6640. [Google Scholar] [CrossRef] [Green Version]
- Kuske, M.; Westphal, D.; Wehner, R.; Schmitz, M.; Beissert, S.; Praetorius, C.; Meier, F. Immunomodulatory effects of BRAF and MEK inhibitors: Implications for Melanoma therapy. Pharmacol. Res. 2018, 136, 151–159. [Google Scholar] [CrossRef]
- Proietti, I.; Skroza, N.; Michelini, S.; Mambrin, A.; Balduzzi, V.; Bernardini, N.; Marchesiello, A.; Tolino, E.; Volpe, S.; Maddalena, P.; et al. BRAF Inhibitors: Molecular Targeting and Immunomodulatory Actions. Cancers 2020, 12, 1823. [Google Scholar] [CrossRef] [PubMed]
- Walunas, T.L.; Lenschow, D.J.; Bakker, C.Y.; Linsley, P.S.; Freeman, G.J.; Green, J.M.; Thompson, C.B.; Bluestone, J.A. CTLA-4 can function as a negative regulator of T cell activation. Immunity 1994, 1, 405–413. [Google Scholar] [CrossRef] [PubMed]
- Krummel, M.F.; Allison, J.P. CTLA-4 engagement inhibits IL-2 accumulation and cell cycle progression upon activation of resting T cells. J. Exp. Med. 1996, 183, 2533–2540. [Google Scholar] [CrossRef]
- Wei, S.C.; Duffy, C.R.; Allison, J.P. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. 2018, 8, 1069–1086. [Google Scholar] [CrossRef] [Green Version]
- Robert, C.; Schachter, J.; Long, G.V.; Arance, A.; Grob, J.J.; Mortier, L.; Daud, A.; Carlino, M.S.; McNeil, C.; Lotem, M.; et al. KEYNOTE-006 investigators. Pembrolizumab versus ipilimumab in advanced melanoma. N. Engl. J. Med. 2015, 372, 2521–2532. [Google Scholar] [CrossRef] [PubMed]
- Willsmore, Z.N.; Coumbe, B.G.T.; Crescioli, S.; Reci, S.; Gupta, A.; Harris, R.J.; Chenoweth, A.; Chauhan, J.; Bax, H.J.; McCraw, A.; et al. Combined anti-PD-1 and anti-CTLA-4 checkpoint blockade: Treatment of melanoma and immune mechanisms of action. Eur. J. Immunol. 2021, 51, 544–556. [Google Scholar] [CrossRef]
- Czarnecka, A.M.; Bartnik, E.; Fiedorowicz, M.; Rutkowski, P. Targeted Therapy in Melanoma and Mechanisms of Resistance. Int. J. Mol. Sci. 2020, 21, 4576. [Google Scholar] [CrossRef]
- Thornton, J.; Chhabra, G.; Singh, C.K.; Guzmán-Pérez, G.; Shirley, C.A.; Ahmad, N. Mechanisms of Immunotherapy Resistance in Cutaneous Melanoma: Recognizing a Shapeshifter. Front. Oncol. 2022, 12, 880876. [Google Scholar] [CrossRef] [PubMed]
- Cives, M.; Mannavola, F.; Lospalluti, L.; Sergi, M.C.; Cazzato, G.; Filoni, E.; Cavallo, F.; Giudice, G.; Stucci, L.S.; Porta, C.; et al. Non-Melanoma Skin Cancers: Biological and Clinical Features. Int. J. Mol. Sci. 2020, 21, 5394. [Google Scholar] [CrossRef] [PubMed]
- Didona, D.; Paolino, G.; Bottoni, U.; Cantisani, C. Non Melanoma Skin Cancer Pathogenesis Overview. Biomedicines 2018, 6, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dourmishev, L.; Rusinova, D.; Botev, I. Clinical variants, stages, and management of basal cell carcinoma. Indian Dermatol. Online J. 2013, 4, 12. [Google Scholar] [CrossRef]
- Polak-Witka, K.; Rudnicka, L.; Blume-Peytavi, U.; Vogt, A. The role of the microbiome in scalp hair follicle biology and disease. Exp. Dermatol. 2020, 29, 286–294. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rubin, A.I.; Chen, E.H.; Ratner, D. Basal-cell carcinoma. N. Engl. J. Med. 2005, 353, 2262–2269. [Google Scholar] [CrossRef] [PubMed]
- Thieu, K.; Ruiz, M.E.; Owens, D.M. Cells of origin and tumor-initiating cells for nonmelanoma skin cancers. Cancer Lett. 2013, 338, 82–88. [Google Scholar] [CrossRef] [Green Version]
- Elder, D.E. Lever’s Histopathology of the Skin, 10th ed.; Wolters Kluwer/Lippincott Williams & Williams: Philadelphia, PA, USA, 2008. [Google Scholar]
- Yanofsky, V.R.; Mercer, S.E.; Phelps, R.G. Histopathological Variants of Cutaneous Squamous Cell Carcinoma: A Review. J. Skin Cancer 2011, 2011, 1–13. [Google Scholar] [CrossRef] [Green Version]
- Wood, D.L.A.; Lachner, N.; Tan, J.M.; Tang, S.; Angel, N.; Laino, A.; Linedale, R.; Lê Cao, K.A.; Morrison, M.; Frazer, I.H.; et al. A Natural History of Actinic Keratosis and Cutaneous Squamous Cell Carcinoma Microbiomes. mBio 2018, 9, e01432-18. [Google Scholar] [CrossRef] [Green Version]
- Josiah, A.J.; Twilley, D.; Pillai, S.K.; Ray, S.S.; Lall, N. Pathogenesis of Keratinocyte Carcinomas and the Therapeutic Potential of Medicinal Plants and Phytochemicals. Molecules 2021, 26, 1979. [Google Scholar] [CrossRef]
- Hawrot, A.; Alam, M.; Ratner, D. Squamous cell carcinoma. Curr. Probl. Dermatol. 2003, 15, 91–133. [Google Scholar] [CrossRef]
- Ogawa, T.; Kiuru, M.; Konia, T.H.; Fung, M.A. Acantholytic squamous cell carcinoma is usually associated with hair follicles, not acantholytic actinic keratosis, and is not “high risk”: Diagnosis, management, and clinical outcomes in a series of 115 cases. J. Am. Acad. Dermatol. 2017, 76, 327–333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kuo, T. Clear cell carcinoma of the skin. A variant of the squamous cell carcinoma that simulates sebaceous carcinoma. Am. J. Surg. Pathol. 1980, 4, 573–583. [Google Scholar] [CrossRef]
- Ko, C.J.; McNiff, J.M.; Glusac, E.J. Squamous cell carcinomas with single cell infiltration: A potential diagnostic pitfall and the utility of MNF116 and p63. J. Cutan. Pathol. 2008, 35, 353–357. [Google Scholar] [CrossRef]
- Pogorzelska-Dyrbus, J.; Szepietowski, J.C. Adhesion Molecules in Non-melanoma Skin Cancers: A Comprehensive Review. In Vivo 2021, 35, 1327–1336. [Google Scholar] [CrossRef]
- Leiter, U.; Keim, U.; Garbe, C. Epidemiology of Skin Cancer: Update 2019. Adv. Exp. Med. Biol. 2020, 1268, 123–139. [Google Scholar] [CrossRef]
- Cameron, M.C.; Lee, E.; Hibler, B.P.; Barker, C.A.; Mori, S.; Cordova, M.; Nehal, K.S.; Rossi, A.M. Basal cell carcinoma: Epidemiology; pathophysiology; clinical and histological subtypes; and disease associations. J. Am. Acad. Dermatol. 2019, 80, 303–317. [Google Scholar] [CrossRef] [Green Version]
- Godar, D.E. UV and Reactive Oxygen Species Activate Human Papillomaviruses Causing Skin Cancers. Curr. Probl. Dermatol. 2021, 55, 339–353. [Google Scholar] [CrossRef] [PubMed]
- Onajin, O.; Brewer, J.D. Skin cancer in patients with chronic lymphocytic leukemia and non-Hodgkin lymphoma. Clin. Adv. Hematol. Oncol. 2012, 10, 571–576. [Google Scholar] [PubMed]
- Li, C.; Athar, M. Ionizing radiation exposure and basal cell carcinoma pathogenesis. Radiat. Res. 2016, 185, 217–228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tang, L.; Wang, K. Chronic inflammation in skin malignancies. J. Mol. Signal. 2016, 11, 2. [Google Scholar] [CrossRef] [Green Version]
- Choquet, H.; Ashrafzadeh, S.; Kim, Y.; Asgari, M.M.; Jorgenson, E. Genetic and environmental factors underlying keratinocyte carcinoma risk. JCI Insight 2020, 5, e134783. [Google Scholar] [CrossRef]
- Asgari, M.M.; Warton, E.M.; Whittemore, A.S. Family history of skin cancer is associated with increased risk of cutaneous squamous cell carcinoma. Dermatol. Surg. 2015, 41, 481–486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chahal, H.S.; Lin, Y.; Ransohoff, K.J.; Hinds, D.A.; Wu, W.; Dai, H.J.; Qureshi, A.A.; Li, W.Q.; Kraft, P.; Tang, J.Y.; et al. Genome-wide association study identifies 14 novel risk alleles associated with basal cell carcinoma. Nat. Commun. 2016, 7, 12048. [Google Scholar] [CrossRef] [Green Version]
- Asgari, M.M.; Wang, W.; Ioannidis, N.M.; Itnyre, J.; Hoffmann, T.; Jorgenson, E.; Whittemore, A.S. Identification of susceptibility loci for cutaneous squamous cell carcinoma. J. Invest Dermatol. 2016, 136, 930–937. [Google Scholar] [CrossRef]
- Albert, M.R.; Weinstock, M.A. Keratinocyte carcinoma. CA Cancer J. Clin. 2003, 53, 292–302. [Google Scholar] [CrossRef]
- Gibbs, P.; Gonzalez, R.; Lee, L.A.; Walsh, P. Medical management of cutaneous malignancies. Clin. Dermatol. 2001, 19, 298–304. [Google Scholar] [CrossRef]
- Firnhaber, J.M. Basal Cell and Cutaneous Squamous Cell Carcinomas: Diagnosis and Treatment. Am. Fam. Physician. 2020, 102, 339–346. [Google Scholar]
- Boutros, A.; Cecchi, F.; Tanda, E.T.; Croce, E.; Gili, R.; Arecco, L.; Spagnolo, F.; Queirolo, P. Immunotherapy for the Treatment of Cutaneous Squamous Cell Carcinoma. Front. Oncol. 2021, 11, 733917. [Google Scholar] [CrossRef]
- Mrázek, J.; Mekadim, C.; Kučerová, P.; Švejstil, R.; Salmonová, H.; Vlasáková, J.; Tarasová, R.; Čížková, J.; Červinková, M. Melanoma-related changes in skin microbiome. Folia Microbiol. 2019, 64, 435–442. [Google Scholar] [CrossRef] [PubMed]
- Vitali, F.; Colucci, R.; Di Paola, M.; Pindo, M.; De Filippo, C.; Moretti, S.; Cavalieri, D. Early melanoma invasivity correlates with gut fungal and bacterial profiles. Br. J. Dermatol. 2022, 186, 106–116. [Google Scholar] [CrossRef] [PubMed]
- Mizuhashi, S.; Kajihara, I.; Sawamura, S.; Kanemaru, H.; Makino, K.; Aoi, J.; Makino, T.; Masuguchi, S.; Fukushima, S.; Ihn, H. Skin microbiome in acral melanoma: Corynebacterium is associated with advanced melanoma. J. Dermatol. 2021, 48, e15–e16. [Google Scholar] [CrossRef] [PubMed]
- Wang, L.; Yi, T.; Kortylewski, M.; Pardoll, D.M.; Zeng, D.; Yu, H. IL-17 can promote tumor growth through an IL-6-Stat3 signaling pathway. J. Exp. Med. 2009, 206, 1457–1464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ridaura, V.K.; Bouladoux, N.; Claesen, J.; Chen, Y.E.; Byrd, A.L.; Constantinides, M.G.; Merrill, E.D.; Tamoutounour, S.; Fischbach, M.A.; Belkaid, Y. Contextual control of skin immunity and inflammation by Corynebacterium. J. Exp. Med. 2018, 215, 785–799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tsuda, K.; Yamanaka, K.; Linan, W.; Miyahara, Y.; Akeda, T.; Nakanishi, T.; Kitagawa, H.; Kakeda, M.; Kurokawa, I.; Shiku, H.; et al. Intratumoral injection of Propionibacterium acnes suppresses malignant melanoma by enhancing Th1 immune responses. PLoS ONE 2011, 6, e29020. [Google Scholar] [CrossRef] [Green Version]
- Wang, Z.; Choi, J.E.; Wu, C.C.; Di Nardo, A. Skin commensal bacteria Staphylococcus epidermidis promote survival of melanocytes bearing UVB-induced DNA damage, while bacteria Propionibacterium acnes inhibit survival of melanocytes by increasing apoptosis. Photodermatol. Photoimmunol. Photomed. 2018, 34, 405–414. [Google Scholar] [CrossRef] [PubMed]
- Nakatsuji, T.; Chen, T.H.; Butcher, A.M.; Trzoss, L.L.; Nam, S.J.; Shirakawa, K.T.; Zhou, W.; Oh, J.; Otto, M.; Fenical, W.; et al. A commensal strain of Staphylococcus epidermidis protects against skin neoplasia. Sci. Adv. 2018, 4, eaao4502. [Google Scholar] [CrossRef] [Green Version]
- Zhou, Z.; Chen, J.; Yao, H.; Hu, H. Fusobacterium and Colorectal Cancer. Front. Oncol. 2018, 8, 371. [Google Scholar] [CrossRef] [Green Version]
- Fujiwara, N.; Kitamura, N.; Yoshida, K.; Yamamoto, T.; Ozaki, K.; Kudo, Y. Involvement of Fusobacterium Species in Oral Cancer Progression: A Literature Review Including Other Types of Cancer. Int. J. Mol. Sci. 2020, 21, 6207. [Google Scholar] [CrossRef]
- Gur, C.; Ibrahim, Y.; Isaacson, B.; Yamin, R.; Abed, J.; Gamliel, M.; Enk, J.; Bar-On, Y.; Stanietsky-Kaynan, N.; Coppenhagen-Glazer, S.; et al. Binding of the Fap2 protein of Fusobacterium nucleatum to human inhibitory receptor TIGIT protects tumors from immune cell attack. Immunity 2015, 42, 344–355. [Google Scholar] [CrossRef] [Green Version]
- Wang, S.; Liu, Y.; Li, J.; Zhao, L.; Yan, W.; Lin, B.; Guo, X.; Wei, Y. Fusobacterium nucleatum Acts as a Pro-carcinogenic Bacterium in Colorectal Cancer: From Association to Causality. Front. Cell. Dev. Biol. 2021, 9, 710165. [Google Scholar] [CrossRef]
- Casasanta, M.A.; Yoo, C.C.; Udayasuryan, B.; Sanders, B.E.; Umaña, A.; Zhang, Y.; Peng, H.; Duncan, A.J.; Wang, Y.; Li, L.; et al. Fusobacterium nucleatum host-cell binding and invasion induces IL-8 and CXCL1 secretion that drives colorectal cancer cell migration. Sci. Signal. 2020, 13, eaba9157. [Google Scholar] [CrossRef] [PubMed]
- Robinson, C.; Xu, M.M.; Nair, S.K.; Beasley, G.M.; Rhodin, K.E. Oncolytic viruses in melanoma. Front. Biosci. 2022, 27, 63. [Google Scholar] [CrossRef] [PubMed]
- Chen, M.L.; Wang, S.H.; Wei, J.C.; Yip, H.T.; Hung, Y.M.; Chang, R. The Impact of Human Papillomavirus Infection on Skin Cancer: A Population-Based Cohort Study. Oncologist 2021, 26, e473–e483. [Google Scholar] [CrossRef]
- Dréau, D.; Culberson, C.; Wyatt, S.; Holder, W.D., Jr. Human papilloma virus in melanoma biopsy specimens and its relation to melanoma progression. Ann. Surg. 2000, 231, 664–671. [Google Scholar] [CrossRef]
- Roussaki-Schulze, A.V.; Kouskoukis, C.; Rammos, C.; Rallis, E.; Kontos, F.; Zafiriou, E.; Gross, G. Identification of human papillomavirus DNA in melanoma biopsy specimens of Greek population. Int. J. Clin. Pharmacol. Res. 2005, 25, 145–150. [Google Scholar]
- Ambretti, S.; Venturoli, S.; Mirasoli, M.; La Placa, M.; Bonvicini, F.; Cricca, M.; Zerbini, M.; Roda, A.; Musiani, M. Assessment of the presence of mucosal human papillomaviruses in malignant melanomas using combined fluorescent in situ hybridization and chemiluminescent immunohistochemistry. Br. J. Dermatol. 2007, 156, 38–44. [Google Scholar] [CrossRef]
- La Placa, M.; Ambretti, S.; Bonvicini, F.; Venturoli, S.; Bianchi, T.; Varotti, C.; Zerbini, M.; Musiani, M. Presence of high-risk mucosal human papillomavirus genotypes in primary melanoma and in acquired dysplastic melanocytic naevi. Br. J. Dermatol. 2005, 152, 909–914. [Google Scholar] [CrossRef]
- Cun, B.; Song, X.; Jia, R.; Wang, H.; Zhao, X.; Liu, B.; Ge, S.; Fan, X. Cell growth inhibition in HPV 18 positive uveal melanoma cells by E6/E7 siRNA. Tumour Biol. 2013, 34, 1801–1806. [Google Scholar] [CrossRef]
- Ruer, J.B.; Pépin, L.; Gheit, T.; Vidal, C.; Kantelip, B.; Tommasino, M.; Prétet, J.L.; Mougin, C.; Aubin, F. Detection of alpha- and beta-human papillomavirus (HPV) in cutaneous melanoma: A matched and controlled study using specific multiplex PCR combined with DNA microarray primer extension. Exp. Dermatol. 2009, 18, 857–862. [Google Scholar] [CrossRef]
- Koburger, I.; Meckbach, D.; Metzler, G.; Fauser, U.; Garbe, C.; Bauer, J. Absence of merkel cell polyoma virus in cutaneous melanoma. Exp. Dermatol. 2011, 20, 78–79. [Google Scholar] [CrossRef]
- Mokánszki, A.; Méhes, G.; Csoma, S.L.; Kollár, S.; Chang Chien, Y.C. Molecular Profiling of Merkel Cell Polyomavirus-Associated Merkel Cell Carcinoma and Cutaneous Melanoma. Diagnostics 2021, 11, 212. [Google Scholar] [CrossRef] [PubMed]
- Singh, M.; Cai, H.; Bunse, M.; Feschotte, C.; Izsvák, Z. Human Endogenous Retrovirus K Rec forms a Regulatory Loop with MITF that Opposes the Progression of Melanoma to an Invasive Stage. Viruses 2020, 12, 1303. [Google Scholar] [CrossRef] [PubMed]
- Oey, O.; Liu, Y.Y.; Sunjaya, A.F.; Simadibrata, D.M.; Khattak, M.A.; Gray, E. Gut microbiota diversity and composition in predicting immunotherapy response and immunotherapy-related colitis in melanoma patients: A systematic review. World J. Clin. Oncol. 2022, 13, 929–942. [Google Scholar] [CrossRef] [PubMed]
- Spencer, C.N.; McQuade, J.L.; Gopalakrishnan, V.; McCulloch, J.A.; Vetizou, M.; Cogdill, A.P.; Khan, M.A.W.; Zhang, X.; White, M.G.; Peterson, C.B.; et al. Dietary fiber and probiotics influence the gut microbiome and melanoma immunotherapy response. Science 2021, 374, 1632–1640. [Google Scholar] [CrossRef]
- Squarzanti, D.F.; Zavattaro, E.; Pizzimenti, S.; Amoruso, A.; Savoia, P.; Azzimonti, B. Non-Melanoma Skin Cancer: News from microbiota research. Crit. Rev. Microbiol. 2020, 46, 433–449. [Google Scholar] [CrossRef] [PubMed]
- Rademacher, F.; Simanski, M.; Gläser, R.; Harder, J. Skin microbiota and human 3D skin models. Exp. Dermatol. 2018, 27, 489–494. [Google Scholar] [CrossRef] [Green Version]
- Wanke, I.; Steffen, H.; Christ, C.; Krismer, B.; Götz, F.; Peschel, A.; Schaller, M.; Schittek, B. Skin commensals amplify the innate immune response to pathogens by activation of distinct signaling pathways. J. Investig. Dermatol. 2011, 131, 382–390. [Google Scholar] [CrossRef] [Green Version]
- Grice, E.A.; Segre, J.A. The skin microbiome. Nat. Rev. Microbiol. 2011, 9, 244–253. [Google Scholar] [CrossRef]
- Chen, Y.E.; Fischbach, M.A.; Belkaid, Y. Skin microbiota-host interactions. Nature 2018, 553, 427–436. [Google Scholar] [CrossRef]
- Woo, Y.R.; Cho, S.H.; Lee, J.D.; Kim, H.S. The Human Microbiota and Skin Cancer. Int. J. Mol. Sci. 2022, 23, 1813. [Google Scholar] [CrossRef] [PubMed]
- Kullander, J.; Forslund, O.; Dillner, J. Staphylococcus aureus and squamous cell carcinoma of the skin. Cancer Epidemiol. Biomarkers Prev. 2009, 18, 472–478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aggarwal, B.B.; Shishodia, S.; Sandur, S.K.; Pandey, M.K.; Sethi, G. Inflammation and cancer: How hot is the link? Biochem. Pharmacol. 2006, 72, 1605–1621. [Google Scholar] [CrossRef] [PubMed]
- Voigt, A.Y.; Emiola, A.; Johnson, J.S.; Fleming, E.S.; Nguyen, H.; Zhou, W.; Tsai, K.Y.; Fink, C.; Oh, J. Skin Microbiome Variation with Cancer Progression in Human Cutaneous Squamous Cell Carcinoma. J. Investig. Dermatol. 2022, 14, 2773–2782.e16. [Google Scholar] [CrossRef]
- Hata, T.R.; Gallo, R.L. Antimicrobial peptides, skin infections, and atopic dermatitis. Semin. Cutan. Med. Surg. 2008, 27, 144–150. [Google Scholar] [CrossRef] [Green Version]
- Nakagawa, S.; Matsumoto, M.; Katayama, Y.; Oguma, R.; Wakabayashi, R.; Nygaard, T.; Saijo, S.; Inohara, N.; Otto, M.; Matsue, H. Staphylococcus aureus Virulent PSMα Peptides Induce Keratinocyte Alarmin Release to Orchestrate IL-17-Dependent Skin Inflammation. Cell Host Microbe. 2017, 22, 667–677.e5. [Google Scholar] [CrossRef] [Green Version]
- Damour, A.; Robin, B.; Deroche, L.; Broutin, L.; Bellin, N.; Verdon, J.; Lina, G.; Leclère, F.M.; Garcia, M.; Cremniter, J.; et al. Phenol-soluble modulins α are major virulence factors of Staphylococcus aureus secretome promoting inflammatory response in human epidermis. Virulence 2021, 12, 2474–2492. [Google Scholar] [CrossRef]
- Stewart, O.A.; Wu, F.; Chen, Y. The role of gastric microbiota in gastric cancer. Gut Microbes. 2020, 11, 1220–1230. [Google Scholar] [CrossRef]
- Tourelle, K.M.; Boutin, S.; Weigand, M.A.; Schmitt, F.C.F. The Association of Gut Microbiota and Complications in Gastrointestinal-Cancer Therapies. Biomedicines 2021, 9, 1305. [Google Scholar] [CrossRef]
- Khalil, C. Human skin explants an in vitro approach for assessing UVB induced damage. Toxicol. In Vitro 2018, 53, 193–199. [Google Scholar] [CrossRef]
- Conteville, L.C.; Vicente, A.C.P. Skin exposure to sunlight: A factor modulating the human gut microbiome composition. Gut Microbes 2020, 11, 1135–1138. [Google Scholar] [CrossRef] [PubMed]
- Di Meo, S.; Reed, T.T.; Venditti, P.; Victor, V.M. Role of ROS and RNS Sources in Physiological and Pathological Conditions. Oxid. Med. Cell. Longev. 2016, 2016, 1245049. [Google Scholar] [CrossRef] [PubMed]
- Juan, C.A.; Pérez de la Lastra, J.M.; Plou, F.J.; Pérez-Lebeña, E. The Chemistry of Reactive Oxygen Species (ROS) Revisited: Outlining Their Role in Biological Macromolecules (DNA, Lipids and Proteins) and Induced Pathologies. Int. J. Mol. Sci. 2021, 22, 4642. [Google Scholar] [CrossRef] [PubMed]
- Espinosa-Diez, C.; Miguel, V.; Mennerich, D.; Kietzmann, T.; Sánchez-Pérez, P.; Cadenas, S.; Lamas, S. Antioxidant responses and cellular adjustments to oxidative stress. Redox Biol. 2015, 6, 183–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rojo de la Vega, M.; Chapman, E.; Zhang, D.D. NRF2 and the Hallmarks of Cancer. Cancer Cell 2018, 34, 21–43. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.; Liu, Y.; Zhao, Z.; Qiu, J. Oxidative stress in the skin: Impact and related protection. Int. J. Cosmet. Sci. 2021, 43, 495–509. [Google Scholar] [CrossRef] [PubMed]
- Krutmann, J.; Bouloc, A.; Sore, G.; Bernard, B.A.; Passeron, T. The skin aging exposome. J. Dermatol. Sci. 2017, 85, 152–161. [Google Scholar] [CrossRef] [Green Version]
- Xian, D.; Lai, R.; Song, J.; Xiong, X.; Zhong, J. Emerging Perspective: Role of Increased ROS and Redox Imbalance in Skin Carcinogenesis. Oxid. Med. Cell. Longev. 2019, 2019, 8127362. [Google Scholar] [CrossRef] [Green Version]
- Hanson, K.M.; Simon, J.D. Epidermal trans-urocanic acid and the UV-A-induced photoaging of the skin. Proc. Natl. Acad. Sci. USA 1998, 95, 10576–10578. [Google Scholar] [CrossRef] [Green Version]
- Buettner, G.R. The pecking order of free radicals and antioxidants: Lipid peroxidation, alpha-tocopherol, and ascorbate. Arch. Biochem. Biophys. 1993, 300, 535–543. [Google Scholar] [CrossRef]
- Valencia, A.; Kochevar, I.E. Nox1-based NADPH oxidase is the major source of UVA-induced reactive oxygen species in human keratinocytes. J. Investig. Dermatol. 2008, 128, 214–222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Soeur, J.; Belaïdi, J.P.; Chollet, C.; Denat, L.; Dimitrov, A.; Jones, C.; Perez, P.; Zanini, M.; Zobiri, O.; Mezzache, S.; et al. Photo-pollution stress in skin: Traces of pollutants (PAH and particulate matter) impair redox homeostasis in keratinocytes exposed to UVA1. J. Dermatol. Sci. 2017, 86, 162–169. [Google Scholar] [CrossRef] [PubMed]
- Ryu, Y.S.; Kang, K.A.; Piao, M.J.; Ahn, M.J.; Yi, J.M.; Hyun, Y.M.; Kim, S.H.; Ko, M.K.; Park, C.O.; Hyun, J.W. Particulate matter induces inflammatory cytokine production via activation of NFκB by TLR5-NOX4-ROS signaling in human skin keratinocyte and mouse skin. Redox Biol. 2019, 21, 101080. [Google Scholar] [CrossRef]
- Szelest, M.; Walczak, K.; Plech, T. A New Insight into the Potential Role of Tryptophan-Derived AhR Ligands in Skin Physiological and Pathological Processes. Int. J. Mol. Sci. 2021, 22, 1104. [Google Scholar] [CrossRef] [PubMed]
- Denison, M.S.; Soshilov, A.A.; He, G.; DeGroot, D.E.; Zhao, B. Exactly the same but different: Promiscuity and diversity in the molecular mechanisms of action of the aryl hydrocarbon (dioxin) receptor. Toxicol. Sci. 2011, 124, 1–22. [Google Scholar] [CrossRef] [Green Version]
- Grishanova, A.Y.; Perepechaeva, M.L. Aryl Hydrocarbon Receptor in Oxidative Stress as a Double Agent and Its Biological and Therapeutic Significance. Int. J. Mol. Sci. 2022, 23, 6719. [Google Scholar] [CrossRef]
- Tsuji, G.; Takahara, M.; Uchi, H.; Takeuchi, S.; Mitoma, C.; Moroi, Y.; Furue, M. An environmental contaminant, benzo(a)pyrene, induces oxidative stress-mediated interleukin-8 production in human keratinocytes via the aryl hydrocarbon receptor signaling pathway. J. Dermatol. Sci. 2011, 62, 42–49. [Google Scholar] [CrossRef]
- Lee, C.W.; Lin, Z.C.; Hu, S.C.; Chiang, Y.C.; Hsu, L.F.; Lin, Y.C.; Lee, I.T.; Tsai, M.H.; Fang, J.Y. Urban particulate matter down-regulates filaggrin via COX2 expression/PGE2 production leading to skin barrier dysfunction. Sci. Rep. 2016, 6, 27995. [Google Scholar] [CrossRef] [Green Version]
- Denat, L.; Kadekaro, A.L.; Marrot, L.; Leachman, S.A.; Abdel-Malek, Z.A. Melanocytes as instigators and victims of oxidative stress. J. Investig. Dermatol. 2014, 134, 1512–1518. [Google Scholar] [CrossRef] [Green Version]
- Ni, Q.; Zhang, P.; Li, Q.; Han, Z. Oxidative Stress and Gut Microbiome in Inflammatory Skin Diseases. Front. Cell. Dev. Biol. 2022, 10, 849985. [Google Scholar] [CrossRef]
- Tsuboi, H.; Kouda, K.; Takeuchi, H.; Takigawa, M.; Masamoto, Y.; Takeuchi, M.; Ochi, H. 8-hydroxydeoxyguanosine in urine as an index of oxidative damage to DNA in the evaluation of atopic dermatitis. Br. J. Dermatol. 1998, 138, 1033–1035. [Google Scholar] [CrossRef]
- Omata, N.; Tsukahara, H.; Ito, S.; Ohshima, Y.; Yasutomi, M.; Yamada, A.; Jiang, M.; Hiraoka, M.; Nambu, M.; Deguchi, Y.; et al. Increased Oxidative Stress in Childhood Atopic Dermatitis. Life Sci. 2001, 69, 223–228. [Google Scholar] [CrossRef] [PubMed]
- Tsukahara, H.; Shibata, R.; Ohta, N.; Sato, S.; Hiraoka, M.; Ito, S.; Noiri, E.; Mayumi, M. High levels of urinary pentosidine, an advanced glycation end product, in children with acute exacerbation of atopic dermatitis: Relationship with oxidative stress. Metabolism 2003, 52, 1601–1605. [Google Scholar] [CrossRef] [PubMed]
- Simonetti, O.; Bacchetti, T.; Ferretti, G.; Molinelli, E.; Rizzetto, G.; Bellachioma, L.; Offidani, A. Oxidative Stress and Alterations of Paraoxonases in Atopic Dermatitis. Antioxidants 2021, 10, 697. [Google Scholar] [CrossRef] [PubMed]
- Sivaranjani, N.; Rao, S.V.; Rajeev, G. Role of reactive oxygen species and antioxidants in atopic dermatitis. J. Clin. Diagn. Res. 2013, 7, 2683–2685. [Google Scholar] [CrossRef]
- Amin, M.N.; Liza, K.F.; Sarwar, M.S.; Ahmed, J.; Adnan, M.T.; Chowdhury, M.I.; Hossain, M.Z.; Islam, M.S. Effect of lipid peroxidation, antioxidants, macro minerals and trace elements on eczema. Arch. Dermatol. Res. 2015, 307, 617–623. [Google Scholar] [CrossRef] [PubMed]
- Pietrzak, A.; Michalak-Stoma, A.; Chodorowska, G.; Szepietowski, J.C. Lipid Disturbances in Psoriasis: An Update. Mediators Inflamm. 2010, 2010, 535612. [Google Scholar] [CrossRef] [Green Version]
- Khmaladze, I.; Kelkka, T.; Guerard, S.; Wing, K.; Pizzolla, A.; Saxena, A.; Lundqvist, K.; Holmdahl, M.; Nandakumar, K.S.; Holmdahl, R. Mannan Induces ROS-Regulated, IL-17A-dependent Psoriasis Arthritis-like Disease in Mice. Proc. Natl. Acad. Sci. USA 2014, 111, E3669–E3678. [Google Scholar] [CrossRef] [Green Version]
- Barygina, V.; Becatti, M.; Prignano, F.; Lotti, T.; Taddei, N.; Fiorillo, C. Fibroblasts to Keratinocytes Redox Signaling: The Possible Role of ROS in Psoriatic Plaque Formation. Antioxidants 2019, 8, 566. [Google Scholar] [CrossRef] [Green Version]
- Hoffmann, J.H.O.; Schaekel, K.; Hartl, D.; Enk, A.H.; Hadaschik, E.N. Dimethyl Fumarate Modulates Neutrophil Extracellular Trap Formation in a Glutathione- and Superoxide-dependent Manner. Br. J. Dermatol. 2018, 178, 207–214. [Google Scholar] [CrossRef]
- Li, S.; Dai, W.; Wang, S.; Kang, P.; Ye, Z.; Han, P.; Zeng, K.; Li, C. Clinical Significance of Serum Oxidative Stress Markers to Assess Disease Activity and Severity in Patients with Non-segmental Vitiligo. Front. Cel Dev. Biol. 2021, 9, 739413. [Google Scholar] [CrossRef] [PubMed]
- Mitra, S.; De Sarkar, S.; Pradhan, A.; Pati, A.K.; Pradhan, R.; Mondal, D.; Sen, S.; Ghosh, A.; Chatterjee, S.; Chatterjee, M. Levels of Oxidative Damage and Proinflammatory Cytokines Are Enhanced in Patients with Active Vitiligo. Free Radic. Res. 2017, 51, 986–994. [Google Scholar] [CrossRef] [PubMed]
- Mittal, M.; Siddiqui, M.R.; Tran, K.; Reddy, S.P.; Malik, A.B. Reactive oxygen species in inflammation and tissue injury. Antioxid. Redox Signal. 2014, 20, 1126–1167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bourgonje, A.R.; Feelisch, M.; Faber, K.N.; Pasch, A.; Dijkstra, G.; van Goor, H. Oxidative Stress and Redox-Modulating Therapeutics in Inflammatory Bowel Disease. Trends Mol. Med. 2020, 26, 1034–1046. [Google Scholar] [CrossRef]
- Lingappan, K. NF-κB in Oxidative Stress. Curr. Opin. Toxicol. 2018, 7, 81–86. [Google Scholar] [CrossRef]
- Ji, H.; Li, X.K. Oxidative Stress in Atopic Dermatitis. Oxid. Med. Cell. Longev. 2016, 2016, 2721469. [Google Scholar] [CrossRef]
- Yao, W.; Tepper, R.S.; Kaplan, M.H. Predisposition to the Development of IL-9-secreting T Cells in Atopic Infants. J. Allergy Clin. Immunol. 2011, 128, 1357–1360.e1355. [Google Scholar] [CrossRef] [Green Version]
- Stefanovic, N.; Irvine, A.D.; Flohr, C. The role of the environment and exposome in atopic dermatitis. Curr. Treat. Options Allergy 2021, 8, 222–241. [Google Scholar] [CrossRef]
- Grange, P.A.; Chéreau, C.; Raingeaud, J.; Nicco, C.; Weill, B.; Dupin, N.; Batteux, F. Production of superoxide anions by keratinocytes initiates P. acnes-induced inflammation of the skin. PLoS Pathog. 2009, 5, e1000527. [Google Scholar] [CrossRef]
- Krueger, A.; Mohamed, A.; Kolka, C.M.; Stoll, T.; Zaugg, J.; Linedale, R.; Morrison, M.; Soyer, H.P.; Hugenholtz, P.; Frazer, I.H.; et al. Skin Cancer-Associated S. aureus Strains Can Induce DNA Damage in Human Keratinocytes by Downregulating DNA Repair and Promoting Oxidative Stress. Cancers 2022, 14, 2143. [Google Scholar] [CrossRef]
- Magiatis, P.; Pappas, P.; Gaitanis, G.; Mexia, N.; Melliou, E.; Galanou, M.; Vlachos, C.; Stathopoulou, K.; Skaltsounis, A.L.; Marselos, M.; et al. Malassezia yeasts produce a collection of exceptionally potent activators of the Ah (dioxin) receptor detected in diseased human skin. J. Investig. Dermatol. 2013, 133, 2023–2030.e12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, J.; Luo, Y.; Zhu, Z.; Zhou, Y.; Sun, L.; Gao, J.; Sun, J.; Wang, G.; Yao, X.; Li, W. A tryptophan metabolite of the skin microbiota attenuates inflammation in patients with atopic dermatitis through the aryl hydrocarbon receptor. J. Allergy Clin. Immunol. 2019, 143, 2108–2119.e8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Uberoi, A.; Bartow-McKenney, C.; Zheng, Q.; Flowers, L.; Campbell, A.; Knight, S.A.B.; Chan, N.; Wei, M.; Lovins, V.; Bugayev, J.; et al. Commensal microbiota regulates skin barrier function and repair via signaling through the aryl hydrocarbon receptor. Cell. Host Microbe. 2021, 29, 1235–1248. [Google Scholar] [CrossRef] [PubMed]
- Napolitano, M.; Fabbrocini, G.; Martora, F.; Picone, V.; Morelli, P.; Patruno, C. Role of Aryl Hydrocarbon Receptor Activation in Inflammatory Chronic Skin Diseases. Cells 2021, 10, 3559. [Google Scholar] [CrossRef]
- Bock, K.W. Human AHR functions in vascular tissue: Pro- and anti-inflammatory responses of AHR agonists in atherosclerosis. Biochem. Pharmacol. 2019, 159, 116–120. [Google Scholar] [CrossRef]
- Wang, Y.; Wu, Y.; Wang, Y.; Xu, H.; Mei, X.; Yu, D.; Wang, Y.; Li, W. Antioxidant Properties of Probiotic Bacteria. Nutrients 2017, 9, 521. [Google Scholar] [CrossRef] [Green Version]
- Ron-Doitch, S.; Soroka, Y.; Frusic-Zlotkin, M.; Barasch, D.; Steinberg, D.; Kohen, R. Saturated and aromatic aldehydes originating from skin and cutaneous bacteria activate the Nrf2-keap1 pathway in human keratinocytes. Exp. Dermatol. 2021, 30, 1381–1387. [Google Scholar] [CrossRef]
- Lee, E.; Ahn, H.; Park, S.; Kim, G.; Kim, H.; Noh, M.G.; Kim, Y.; Yeon, J.S.; Park, H. Staphylococcus epidermidis WF2R11 Suppresses PM2.5-Mediated Activation of the Aryl Hydrocarbon Receptor in HaCaT Keratinocytes. Probiotics Antimicrob. Proteins 2022, 14, 915–933. [Google Scholar] [CrossRef]
- Andersson, T.; Ertürk Bergdahl, G.; Saleh, K.; Magnúsdóttir, H.; Stødkilde, K.; Andersen, C.B.F.; Lundqvist, K.; Jensen, A.; Brüggemann, H.; Lood, R. Common skin bacteria protect their host from oxidative stress through secreted antioxidant RoxP. Sci. Rep. 2019, 9, 3596. [Google Scholar] [CrossRef] [Green Version]
- Ellis, S.R.; Nguyen, M.; Vaughn, A.R.; Notay, M.; Burney, W.A.; Sandhu, S.; Sivamani, R.K. The Skin and Gut Microbiome and Its Role in Common Dermatologic Conditions. Microorganisms 2019, 7, 550. [Google Scholar] [CrossRef] [Green Version]
- Mahmud, M.R.; Akter, S.; Tamanna, S.K.; Mazumder, L.; Esti, I.Z.; Banerjee, S.; Akter, S.; Hasan, M.R.; Acharjee, M.; Hossain, M.S.; et al. Impact of gut microbiome on skin health: Gut-skin axis observed through the lenses of therapeutics and skin diseases. Gut Microbes 2022, 14, 2096995. [Google Scholar] [CrossRef]
- De Pessemier, B.; Grine, L.; Debaere, M.; Maes, A.; Paetzold, B.; Callewaert, C. Gut-Skin Axis: Current Knowledge of the Interrelationship between Microbial Dysbiosis and Skin Conditions. Microorganisms 2021, 9, 353. [Google Scholar] [CrossRef] [PubMed]
- Lin, W.Y.; Lin, J.H.; Kuo, Y.; Chiang, P.R.; Ho, H.H. Probiotics and their Metabolites Reduce Oxidative Stress in Middle-Aged Mice. Curr. Microbiol. 2022, 79, 104. [Google Scholar] [CrossRef] [PubMed]
- Litus, O.; Derkach, N.; Litus, V.; Bisyuk, Y.; Lytvynenko, B. Efficacy of Probiotic Therapy on Atopic Dermatitis in Adults Depends on the C-159T Polymorphism of the CD14 Receptor Gene—A Pilot Study. Open Access Maced J. Med. Sci. 2019, 7, 1053–1058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moludi, J.; Khedmatgozar, H.; Saiedi, S.; Razmi, H.; Alizadeh, M.; Ebrahimi, B. Probiotic supplementation improves clinical outcomes and quality of life indicators in patients with plaque psoriasis: A randomized double-blind clinical trial. Clin. Nutr. ESPEN 2021, 46, 33–39. [Google Scholar] [CrossRef]
- Barrera, G. Oxidative stress and lipid peroxidation products in cancer progression and therapy. ISRN Oncol. 2012, 2012, 137289. [Google Scholar] [CrossRef] [Green Version]
- Moloney, J.N.; Cotter, T.G. ROS signalling in the biology of cancer. Semin. Cell. Dev. Biol. 2018, 80, 50–64. [Google Scholar] [CrossRef]
- Snezhkina, A.V.; Kudryavtseva, A.V.; Kardymon, O.L.; Savvateeva, M.V.; Melnikova, N.V.; Krasnov, G.S.; Dmitriev, A.A. ROS Generation and Antioxidant Defense Systems in Normal and Malignant Cells. Oxid. Med. Cell. Longev. 2019, 2019, 6175804. [Google Scholar] [CrossRef] [Green Version]
- Venza, M.; Visalli, M.; Beninati, C.; De Gaetano, G.V.; Teti, D.; Venza, I. Cellular Mechanisms of Oxidative Stress and Action in Melanoma. Oxid. Med. Cell. Longev. 2015, 2015, 481782. [Google Scholar] [CrossRef] [Green Version]
- Ikehata, H.; Yamamoto, M. Roles of the KEAP1-NRF2 system in mammalian skin exposed to UV radiation. Toxicol. Appl. Pharmacol. 2018, 360, 69–77. [Google Scholar] [CrossRef]
- Dinkova-Kostova, A.T.; Jenkins, S.N.; Fahey, J.W.; Ye, L.; Wehage, S.L.; Liby, K.T.; Stephenson, K.K.; Wade, K.L.; Talalay, P. Protection against UV-light-induced skin carcinogenesis in SKH-1 high-risk mice by sulforaphane-containing broccoli sprout extracts. Cancer Lett. 2006, 240, 243–252. [Google Scholar] [CrossRef]
- Knatko, E.V.; Ibbotson, S.H.; Zhang, Y.; Higgins, M.; Fahey, J.W.; Talalay, P.; Dawe, R.S.; Ferguson, J.; Huang, J.T.; Clarke, R.; et al. Nrf2 Activation Protects against Solar-Simulated Ultraviolet Radiation in Mice and Humans. Cancer Prev. Res. 2015, 8, 475–486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Le Gal, K.; Ibrahim, M.X.; Wiel, C.; Sayin, V.I.; Akula, M.K.; Karlsson, C.; Dalin, M.G.; Akyürek, L.-M.; Lindahl, P.; Nilsson, J.; et al. Antioxidants can increase melanoma metastasis in mice. Sci. Transl. Med. 2015, 7, 308re8. [Google Scholar] [CrossRef] [PubMed]
- Mahamat-Saleh, Y.; Savoye, I.; Cervenka, I.; Rahmoun, M.A.; Mancini, F.R.; Boutron-Ruault, M.C.; Kvaskoff, M. Intake of Antioxidant Supplements and Risk of Keratinocytes Cancers in Women: A Prospective Cohort Study. Curr. Dev. Nutr. 2020, 4, 335. [Google Scholar] [CrossRef]
- Obrador, E.; Liu-Smith, F.; Dellinger, R.W.; Salvador, R.; Meyskens, F.L.; Estrela, J.M. Oxidative stress and antioxidants in the pathophysiology of malignant melanoma. Biol. Chem. 2019, 400, 589–612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Emanuelli, M.; Sartini, D.; Molinelli, E.; Campagna, R.; Pozzi, V.; Salvolini, E.; Simonetti, O.; Campanati, A.; Offidani, A. The Double-Edged Sword of Oxidative Stress in Skin Damage and Melanoma: From Physiopathology to Therapeutical Approaches. Antioxidants 2022, 11, 612. [Google Scholar] [CrossRef]
- Menegon, S.; Columbano, A.; Giordano, S. The Dual Roles of NRF2 in Cancer. Trends Mol. Med. 2016, 22, 578–593. [Google Scholar] [CrossRef]
- Barrera, G.; Cucci, M.A.; Grattarola, M.; Dianzani, C.; Muzio, G.; Pizzimenti, S. Control of Oxidative Stress in Cancer Chemoresistance: Spotlight on Nrf2 Role. Antioxidants 2021, 10, 510. [Google Scholar] [CrossRef]
- Pizzimenti, S.; Ribero, S.; Cucci, M.A.; Grattarola, M.; Monge, C.; Dianzani, C.; Barrera, G.; Muzio, G. Oxidative Stress-Related Mechanisms in Melanoma and in the Acquired Resistance to Targeted Therapies. Antioxidants 2021, 10, 1942. [Google Scholar] [CrossRef]
- Hintsala, H.R.; Jokinen, E.; Haapasaari, K.M.; Moza, M.; Ristimäki, A.; Soini, Y.; Koivunen, J.; Karihtala, P. Nrf2/Keap1 Pathway and Expression of Oxidative Stress Lesions 8-hydroxy-2′-deoxyguanosine and Nitrotyrosine in Melanoma. Anticancer Res. 2016, 36, 1497–1506. [Google Scholar]
- Kim, Y.R.; Oh, J.E.; Kim, M.S.; Kang, M.R.; Park, S.W.; Han, J.Y.; Eom, H.S.; Yoo, N.J.; Lee, S.H. Oncogenic NRF2 mutations in squamous cell carcinomas of oesophagus and skin. J. Pathol. 2010, 220, 446–451. [Google Scholar] [CrossRef]
- Chaisiriwong, L.; Wanitphakdeedecha, R.; Sitthinamsuwan, P.; Sampattavanich, S.; Chatsiricharoenkul, S.; Manuskiatti, W.; Panich, U. A Case-Control Study of Involvement of Oxidative DNA Damage and Alteration of Antioxidant Defense System in Patients with Basal Cell Carcinoma: Modulation by Tumor Removal. Oxid. Med. Cell. Longev. 2016, 2016, 5934024. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Matsuoka, Y.; Yoshida, R.; Kawahara, K.; Sakata, J.; Arita, H.; Nkashima, H.; Takahashi, N.; Hirayama, M.; Nagata, M.; Hirosue, A.; et al. The antioxidative stress regulator Nrf2 potentiates radioresistance of oral squamous cell carcinoma accompanied with metabolic modulation. Lab. Investig. 2022, 102, 896–907. [Google Scholar] [CrossRef] [PubMed]
- Akdemir, B.; Nakajima, Y.; Inazawa, J.; Inoue, J. miR-432 Induces NRF2 Stabilization by Directly Targeting KEAP1. Mol. Cancer Res. 2017, 15, 1570–1578. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Khamari, R.; Trinh, A.; Gabert, P.E.; Corazao-Rozas, P.; Riveros-Cruz, S.; Balayssac, S.; Malet-Martino, M.; Dekiouk, S.; Joncquel Chevalier Curt, M.; Maboudou, P.; et al. Glucose metabolism and NRF2 coordinate the antioxidant response in melanoma resistant to MAPK inhibitors. Cell Death Dis. 2018, 9, 325. [Google Scholar] [CrossRef] [Green Version]
- Matsuzaki, T.; Yokokura, T.; Azuma, I. Antimetastatic effect of Lactobacillus casei YIT9018 (LC 9018) on a highly metastatic variant of B16 melanoma in C57BL/6J mice. Cancer Immunol. Immunother. 1987, 24, 99–105. [Google Scholar] [CrossRef]
- Luo, M.; Hu, M.; Feng, X.; Xiao Li, W.; Dong, D.; Wang, W. Preventive effect of Lactobacillus reuteri on melanoma. Biomed. Pharmacother. 2020, 126, 109929. [Google Scholar] [CrossRef]
- Gaitanis, G.; Velegraki, A.; Magiatis, P.; Pappas, P.; Bassukas, I.D. Could Malassezia yeasts be implicated in skin carcinogenesis through the production of aryl-hydrocarbon receptor ligands? Med. Hypotheses 2011, 77, 47–51. [Google Scholar] [CrossRef]
- Williams, V.M.; Filippova, M.; Filippov, V.; Payne, K.J.; Duerksen-Hughes, P. Human papillomavirus type 16 E6* induces oxidative stress and DNA damage. J. Virol. 2014, 88, 6751–6761. [Google Scholar] [CrossRef] [Green Version]
- Lesiów, M.K.; Pietrzyk, P.; Kyzioł, A.; Komarnicka, U.K. Correction to “Cu(II) Complexes with FomA Protein Fragments of Fusobacterium Nucleatum Increase Oxidative Stress and Malondialdehyde Level”. Chem Res Toxicol. 2020, 33, 2218. [Google Scholar] [CrossRef]
- Leung, M.H.Y.; Tong, X.; Bastien, P.; Guinot, F.; Tenenhaus, A.; Appenzeller, B.M.R.; Betts, R.J.; Mezzache, S.; Li, J.; Bourokba, N.; et al. Changes of the human skin microbiota upon chronic exposure to polycyclic aromatic hydrocarbon pollutants. Microbiome 2020, 8, 100. [Google Scholar] [CrossRef] [PubMed]
- Eshak, G.A.; Malak, H.M.; Ahmed, M.I. Polycyclic aromatic hydrocarbons: Role of apoptosis in dermatotoxic and carcinogenic effect in asphalt road paving workers. J. Clin. Toxicol. 2012, 2, 5. [Google Scholar] [CrossRef] [Green Version]
- Später, S.; Hipler, U.C.; Haustein, U.F.; Nenoff, P. Generation of reactive oxygen species in vitro by Malassezia yeasts. Hautarzt 2009, 60, 122–127. [Google Scholar] [CrossRef] [PubMed]
- Marlowe, J.L.; Puga, A. Aryl hydrocarbon receptor, cell cycle regulation, toxicity, and tumorigenesis. J. Cell. Biochem. 2005, 96, 1174–1184. [Google Scholar] [CrossRef]
- Ciążyńska, M.; Olejniczak-Staruch, I.; Sobolewska-Sztychny, D.; Narbutt, J.; Skibińska, M.; Lesiak, A. Ultraviolet Radiation and Chronic Inflammation-Molecules and Mechanisms Involved in Skin Carcinogenesis: A Narrative Review. Life 2021, 11, 326. [Google Scholar] [CrossRef]
- Klucky, B.; Mueller, R.; Vogt, I.; Teurich, S.; Hartenstein, B.; Breuhahn, K.; Flechtenmacher, C.; Angel, P.; Hess, J. Kallikrein 6 Induces E-Cadherin Shedding and Promotes Cell Proliferation, Migration, and Invasion. Cancer Res. 2007, 67, 8198–8206. [Google Scholar] [CrossRef] [Green Version]
- Kuo, P.T.; Zeng, Z.; Salim, N.; Mattarollo, S.; Wells, J.W.; Leggatt, G.R. The Role of CXCR3 and Its Chemokine Ligands in Skin Disease and Cancer. Front Med. 2018, 5, 271. [Google Scholar] [CrossRef] [Green Version]
- Kerkelä, E.; Saarialho-Kere, U. Matrix metalloproteinases in tumor progression: Focus on basal and squamous cell skin cancer. Exp. Dermatol. 2003, 12, 109–125. [Google Scholar] [CrossRef]
- Carvalho, L.A.C.; Queijo, R.G.; Baccaro, A.L.B.; Siena, Á.D.D.; Silva, W.A., Jr.; Rodrigues, T.; Maria-Engler, S.S. Redox-Related Proteins in Melanoma Progression. Antioxidants 2022, 11, 438. [Google Scholar] [CrossRef]
- Pieniz, S.; Andreazza, R.; Okeke, B.C.; Camargo, F.A.; Brandelli, A. Antimicrobial and antioxidant activities of Enterococcus species isolated from meat and dairy products. Braz. J. Biol. 2015, 75, 923–931. [Google Scholar] [CrossRef] [Green Version]
Trial Number | Study Title | Trial Description | Phase/ Location |
---|---|---|---|
Fecal Microbiota Transplantation (FMT) | |||
NCT03353402 | FMT in Metastatic Melanoma Pts Who Failed Immunotherapy | Pts with metastatic melanoma who responded to immunotherapy serve as the fecal implant donors in melanoma pts resistant to CPI. Primary outcomes: incidence of FMT-related AE and proper implant engraftment. Secondary outcomes: changes in composition and activity of the immune cell population. | Phase 1 Israel |
NCT03341143 | FMT in Melanoma Pts | The donors are pts with advanced melanoma who have been treated successfully with CPI. CPI-resistant melanoma pts received FMT and treatment with Pembrolizumab, a PD-1 inhibitor. Primary outcomes: ORR: number of pts with objective responses (PR, CR). Secondary outcomes: AE, PFS, OS, changes in immune cell composition and functionalities. | Phase 2 United States |
NCT05251389 | FMT to Convert Response to Immunotherapy | This is a randomized double-blind intervention phase Ib/IIa trial in CPI refractory metastatic melanoma pts receiving either FMT of a CPI responding or FMT from a CPI non-responding donor, in combination with CPI. After both types of FMT, melanoma pts will continue their anti-PD-1 treatment. Primary outcomes: efficacy, defined as CBR: SD, PR, CR. Secondary outcomes: AE, PFS, changes in the gut microbiome, metabolome, and immune cell populations. | Phase 1 Phase 2 Netherlands |
NCT05286294 | Microbiota Transplant to Cancer Patients Who Have Failed Immunotherapy Using Feces from Clinical Responders | FMT in CPI non-responding pts with melanoma or SCC. Primary outcomes: AE, ORR. Secondary outcomes: OS, PFS, DRR, implant engraftment estimation, evaluation of the effect of therapy on quality of life, fatigue, and pain. | Phase 2 Norway |
Live Biotherapeutic Product (LBP) | |||
NCT05354102 | A First-in-human (FIH) Combination Treatment Study With a Single Dose Level of BMC128 | Melanoma pts receive a combination treatment of BMC128, a live bio-therapeutic product composed of 4 commensal bacterial strains, natural inhabitants of the human intestinal tract, in combination with anti-PD-1 Nivolumab. Primary outcomes: AE. Secondary outcomes: ORR, CBR: SD, PR, CR, DRR. | Phase 1 Israel |
NCT04208958 | Study of VE800 and Nivolumab in Patients With Selected Types of Advanced or Metastatic Cancer | Melanoma pts receive a combination treatment of VE800, a live bio-therapeutic product consisting of 11 distinct non-pathogenic, non-toxigenic, commensal bacterial strains, in combination with anti-PD-1 Nivolumab. Primary outcomes: AE, ORR. Secondary outcomes: DRR, CBR, PFS, OS, pharmacokinetics studies of VE800. | Phase 1 Phase 2 United States |
NCT03934827 | MRx0518 in Patients With Solid Tumors Waiting Surgical Removal of the Tumor | Melanoma pts amenable to surgical resection receive MRx0518, a proprietary strain of a bacterium belonging to Enterococcus species, which is found in the gastrointestinal tract of approx. 25% of humans and is predicted, from preclinical studies, to produce beneficial effects in humans. The first part of the study is devoted to measuring the safety and tolerability of MRx0518; in case of successful evaluation, the study will continue as a randomized double-blinded with a placebo. Primary outcomes: AE, general health assessment. Secondary outcomes: tumor markers, OS. | Phase 1 United Kingdom |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Azzimonti, B.; Ballacchino, C.; Zanetta, P.; Cucci, M.A.; Monge, C.; Grattarola, M.; Dianzani, C.; Barrera, G.; Pizzimenti, S. Microbiota, Oxidative Stress, and Skin Cancer: An Unexpected Triangle. Antioxidants 2023, 12, 546. https://doi.org/10.3390/antiox12030546
Azzimonti B, Ballacchino C, Zanetta P, Cucci MA, Monge C, Grattarola M, Dianzani C, Barrera G, Pizzimenti S. Microbiota, Oxidative Stress, and Skin Cancer: An Unexpected Triangle. Antioxidants. 2023; 12(3):546. https://doi.org/10.3390/antiox12030546
Chicago/Turabian StyleAzzimonti, Barbara, Chiara Ballacchino, Paola Zanetta, Marie Angele Cucci, Chiara Monge, Margherita Grattarola, Chiara Dianzani, Giuseppina Barrera, and Stefania Pizzimenti. 2023. "Microbiota, Oxidative Stress, and Skin Cancer: An Unexpected Triangle" Antioxidants 12, no. 3: 546. https://doi.org/10.3390/antiox12030546