1
|
Guida S, Puig S, DI Resta C, Sallustio F, Mangano E, Stabile G, Longo C, Pellacani G, Guida G, Rongioletti F. Melanocortin-1 receptor (MC1R): a review for dermatologists. Ital J Dermatol Venerol 2024; 159:285-293. [PMID: 38376504 DOI: 10.23736/s2784-8671.24.07839-3] [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: 02/21/2024]
Abstract
Melanocortin-1 receptor (MC1R) and its variants have a pivotal role in melanin synthesis. However, MC1R has been associated to non-pigmentary pathways related to DNA-repair activities and inflammation. The aim of this review is to provide an up-to-date overview about the role of MC1R in the skin. Specifically, after summarizing the current knowledge about MC1R structure and polymorphisms, we report data concerning the correlation between MC1R, phenotypic traits, skin aging, other diseases and skin cancers and their risk assessment through genetic testing.
Collapse
Affiliation(s)
- Stefania Guida
- Dermatology Clinic, IRCCS San Raffaele Hospital, Milan, Italy -
- Faculty of Medicine, Vita-Salute San Raffaele University, Milan, Italy -
| | - Susana Puig
- Melanoma Unit, Department of Dermatology, Hospital Clínic de Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunye, University of Barcelona, Barcelona, Spain
| | - Chiara DI Resta
- Faculty of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Genomic Unit for the Diagnosis of Human Pathologies, IRCCS San Raffaele Hospital, Milan, Italy
| | - Fabio Sallustio
- Department of Interdisciplinary Medicine, Aldo Moro University of Bari, Bari, Italy
| | - Eleonora Mangano
- Institute of Biomedical Technologies (ITB), National Research Center (CNR), Segrate, Milan, Italy
| | - Giorgio Stabile
- Dermatology Clinic, IRCCS San Raffaele Hospital, Milan, Italy
- Faculty of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| | - Caterina Longo
- Department of Dermatology, University of Modena and Reggio Emilia, Modena, Italy
- Skin Cancer Center, Azienda Unità Sanitaria Locale, IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | | | - Gabriella Guida
- Section of Molecular Biology, Department of Basic Medical Sciences, Neurosciences and Sense Organs, Aldo Moro University of Bari, Bari, Italy
| | - Franco Rongioletti
- Dermatology Clinic, IRCCS San Raffaele Hospital, Milan, Italy
- Faculty of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
2
|
Coutant K, Magne B, Ferland K, Fuentes-Rodriguez A, Chancy O, Mitchell A, Germain L, Landreville S. Melanocytes in regenerative medicine applications and disease modeling. J Transl Med 2024; 22:336. [PMID: 38589876 PMCID: PMC11003097 DOI: 10.1186/s12967-024-05113-x] [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: 11/08/2023] [Accepted: 03/20/2024] [Indexed: 04/10/2024] Open
Abstract
Melanocytes are dendritic cells localized in skin, eyes, hair follicles, ears, heart and central nervous system. They are characterized by the presence of melanosomes enriched in melanin which are responsible for skin, eye and hair pigmentation. They also have different functions in photoprotection, immunity and sound perception. Melanocyte dysfunction can cause pigmentary disorders, hearing and vision impairments or increased cancer susceptibility. This review focuses on the role of melanocytes in homeostasis and disease, before discussing their potential in regenerative medicine applications, such as for disease modeling, drug testing or therapy development using stem cell technologies, tissue engineering and extracellular vesicles.
Collapse
Affiliation(s)
- Kelly Coutant
- Department of Ophthalmology and Otorhinolaryngology-Cervico-Facial Surgery, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
- Regenerative Medicine Division, CHU de Québec-Université Laval Research Centre, Quebec City, QC, Canada
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Quebec City, QC, Canada
- Université Laval Cancer Research Center, Quebec City, QC, Canada
| | - Brice Magne
- Regenerative Medicine Division, CHU de Québec-Université Laval Research Centre, Quebec City, QC, Canada
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Quebec City, QC, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Karel Ferland
- Regenerative Medicine Division, CHU de Québec-Université Laval Research Centre, Quebec City, QC, Canada
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Quebec City, QC, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Aurélie Fuentes-Rodriguez
- Department of Ophthalmology and Otorhinolaryngology-Cervico-Facial Surgery, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
- Regenerative Medicine Division, CHU de Québec-Université Laval Research Centre, Quebec City, QC, Canada
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Quebec City, QC, Canada
- Université Laval Cancer Research Center, Quebec City, QC, Canada
| | - Olivier Chancy
- Department of Ophthalmology and Otorhinolaryngology-Cervico-Facial Surgery, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
- Regenerative Medicine Division, CHU de Québec-Université Laval Research Centre, Quebec City, QC, Canada
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Quebec City, QC, Canada
- Université Laval Cancer Research Center, Quebec City, QC, Canada
| | - Andrew Mitchell
- Department of Ophthalmology and Otorhinolaryngology-Cervico-Facial Surgery, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
- Regenerative Medicine Division, CHU de Québec-Université Laval Research Centre, Quebec City, QC, Canada
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Quebec City, QC, Canada
- Université Laval Cancer Research Center, Quebec City, QC, Canada
| | - Lucie Germain
- Regenerative Medicine Division, CHU de Québec-Université Laval Research Centre, Quebec City, QC, Canada.
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Quebec City, QC, Canada.
- Department of Surgery, Faculty of Medicine, Université Laval, Quebec City, QC, Canada.
| | - Solange Landreville
- Department of Ophthalmology and Otorhinolaryngology-Cervico-Facial Surgery, Faculty of Medicine, Université Laval, Quebec City, QC, Canada.
- Regenerative Medicine Division, CHU de Québec-Université Laval Research Centre, Quebec City, QC, Canada.
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Quebec City, QC, Canada.
- Université Laval Cancer Research Center, Quebec City, QC, Canada.
| |
Collapse
|
3
|
Bhattacharya K, Khanal P, Patil VS, Dwivedi PSR, Chanu NR, Chaudhary RK, Deka S, Chakraborty A. Computational pharmacology profiling of borapetoside C against melanoma. J Biomol Struct Dyn 2024; 42:3233-3248. [PMID: 37203884 DOI: 10.1080/07391102.2023.2213333] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 05/04/2023] [Indexed: 05/20/2023]
Abstract
Melanoma,also known as a 'black tumor', begins in the melanocytes when cells (that produce pigment) grows out of control. Immunological dysregulation, which raises the risk for multiple illnesses, including melanoma, may be influenced by stress tiggered through viral infection, long term effects of ultraviolet radiation, environmental pollutants etc. Borapetoside C is one of the phytoconstituents from Tinospora crispa, and its biological source has been reported for its antistress property. Network pharmacology and KEGG pathway analysis of borapetoside C-regulated proteins were conducted to identify the hub genes involved in melanoma development. Further, a molecular docking was performed between borapetoside C and targets involved in melanoma. Further, the top 3 complexes were selected based on the binding energy to conduct molecular dynamics simulations to evaluate the stability of ligand-protein complex followed by principal component analysis and dynamic cross-correlation matrix. In addition, borapetoside C was also screened for its pharmacokinetics and toxicity profile. Network Pharmacology studies and KEGG pathway analysis revealed 8 targets involved in melanoma. Molecular docking between borapetoside C and targets involved in melanoma identified 3 complexes with minimum binding i.e. borapetoside C- MAP2K1, MMP9, and EGFR. Further, molecular dynamics simulations showed a stable complex of borapetoside C with MMP9 and EGFR. The present study suggested that borapetoside C may target MMP9 and EGFR to possess an anti-melanoma property. This finding can be useful in developing a novel therapeutic agent against melanoma from a natural source.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Kunal Bhattacharya
- Pratiksha Institute of Pharmaceutical Sciences, Guwahati, Assam, India
- Royal School of Pharmacy, The Assam Royal Global University, Guwahati, Assam, India
| | - Pukar Khanal
- Department of Pharmacology, NGSM Institute of Pharmaceutical Sciences (NGSMIPS), Nitte (Deemed to be University), Mangalore, India
| | - Vishal S Patil
- KLE College of Pharmacy, KLE Academy of Higher Education and Research, Belagavi, India
| | - Prarambh S R Dwivedi
- Department of Pharmacology, NGSM Institute of Pharmaceutical Sciences (NGSMIPS), Nitte (Deemed to be University), Mangalore, India
| | - Nongmaithem Randhoni Chanu
- Pratiksha Institute of Pharmaceutical Sciences, Guwahati, Assam, India
- Faculty of Pharmaceutical Science, Assam Downtown University, Guwahati, Assam, India
| | - Raushan Kumar Chaudhary
- Department of Pharmacy Practice, NGSM Institute of Pharmaceutical Sciences (NGSMIPS), Nitte (Deemed to be University), Mangalore, India
| | - Satyendra Deka
- Pratiksha Institute of Pharmaceutical Sciences, Guwahati, Assam, India
| | - Arup Chakraborty
- Pratiksha Institute of Pharmaceutical Sciences, Guwahati, Assam, India
| |
Collapse
|
4
|
Flores-Torres MH, Bjornevik K, Zhang X, Gao X, Hung AY, Schwarzschild MA, Chen X, Ascherio A. Hair color, family history of melanoma, and the risk of Parkinson's disease: An analysis update. Parkinsonism Relat Disord 2024; 119:105965. [PMID: 38142631 PMCID: PMC10843649 DOI: 10.1016/j.parkreldis.2023.105965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/24/2023] [Accepted: 12/17/2023] [Indexed: 12/26/2023]
Abstract
BACKGROUND A shared biological component between melanoma and Parkinson's disease (PD) has been suggested. Yet, epidemiological evidence is scarce. OBJECTIVE To examine the association of hair color and family history of melanoma, two strong predictors of melanoma risk, with the occurrence of PD. METHODS We followed 131,342 women and men for ∼30 years for the development of PD. We calculated the cumulative incidence of PD from ages 40 to 90 according to hair color, and estimated the hazard ratio of PD according to hair color and family history of melanoma. RESULTS Hair color was not strongly associated with the risk of PD, especially at advanced ages. In contrast, individuals with a family history of melanoma had a 1.4-fold higher risk of PD compared to those without a history. CONCLUSIONS Our results support the hypothesis of a shared biological component between PD and melanoma. Both pigmentary and non-pigmentary pathways may play a role.
Collapse
Affiliation(s)
- Mario H Flores-Torres
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Kjetil Bjornevik
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Xinyuan Zhang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Xiang Gao
- Department of Nutrition and Food Hygiene, School of Public Health, Institute of Nutrition, Fudan University, Shanghai, China
| | - Albert Y Hung
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Michael A Schwarzschild
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Xiqun Chen
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Alberto Ascherio
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| |
Collapse
|
5
|
Dana PM, Hallajzadeh J, Asemi Z, Mansournia MA, Yousefi B. Advances in Chitosan-based Drug Delivery Systems in Melanoma: A Narrative Review. Curr Med Chem 2024; 31:3488-3501. [PMID: 37202890 DOI: 10.2174/0929867330666230518143654] [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: 12/04/2022] [Revised: 03/24/2023] [Accepted: 04/10/2023] [Indexed: 05/20/2023]
Abstract
Melanoma accounts for the minority of skin cancer cases. However, it has the highest mortality rate among the subtypes of skin cancer. At the early stages of the disease, patients show a good prognosis after the surgery, but developing metastases leads to a remarkable drop in patients' 5-year survival rate. Despite the advances made in the therapeutic approaches to this disease, melanoma treatment is still facing several obstacles. Systemic toxicity, water insolubility, instability, lack of proper biodistribution, inadequate cellular penetration, and rapid clearance are some of the challenges that should be addressed in the field of melanoma treatment. While various delivery systems have been developed to circumvent these challenges, chitosan-based delivery platforms have indicated significant success. Chitosan that is produced by the deacetylation of chitin can be formulated into different materials (e.g., nanoparticle, film, and hydrogel) due to its characteristics. Both in vitro and in vivo studies have reported that chitosan-based materials can be used in drug delivery systems while offering a solution for the common problems in this area, such as enhancing biodistribution and skin penetration as well as the sustained release of the drugs. Herein, we reviewed the studies concerning the role of chitosan as a drug delivery system in melanoma and discussed how these drug systems are used for delivering chemotherapeutic drugs (e.g., doxorubicin and paclitaxel), genes (e.g., TRAIL), and RNAs (e.g., miRNA199a and STAT3 siRNA) successfully. Furthermore, we take a look into the role of chitosan-based nanoparticles in neutron capture therapy.
Collapse
Affiliation(s)
- Parisa Maleki Dana
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Jamal Hallajzadeh
- Department of Biochemistry and Nutrition, Research Center for Evidence-based Health Management, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Mohammad Ali Mansournia
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
6
|
Srivastava P, Nishiyama S, Zhou F, Lin SH, Srivastava A, Su C, Xu Y, Peng W, Levy M, Schwarzschild M, Chen X. Peripheral MC1R Activation Modulates Immune Responses and is Neuroprotective in a Mouse Model of Parkinson's Disease. J Neuroimmune Pharmacol 2023; 18:704-717. [PMID: 38110615 PMCID: PMC10769915 DOI: 10.1007/s11481-023-10094-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 10/17/2023] [Indexed: 12/20/2023]
Abstract
BACKGROUND Melanocortin 1 receptor (MC1R) is a key pigmentation gene, and loss-of-function of MC1R variants that produce red hair may be associated with Parkinson's disease (PD). We previously reported compromised dopaminergic neuron survival in Mc1r mutant mice and dopaminergic neuroprotective effects of local injection of a MC1R agonist to the brain or a systemically administered MC1R agonist with appreciable central nervous system (CNS) permeability. Beyond melanocytes and dopaminergic neurons, MC1R is expressed in other peripheral tissues and cell types, including immune cells. The present study investigates the impact of NDP-MSH, a synthetic melanocortin receptor (MCR) agonist that does not cross BBB, on the immune system and the nigrostriatal dopaminergic system in mouse model of PD. METHODS C57BL/6 mice were treated systemically with MPTP.HCl (20 mg/kg) and LPS (1 mg/kg) from day 1 to day 4 and NDP-MSH (400 µg/kg) or vehicle from day 1 to day 12 following which the mice were sacrificed. Peripheral and CNS immune cells were phenotyped and inflammatory markers were measured. The nigrostriatal dopaminergic system was assessed behaviorally, chemically, immunologically, and pathologically. To understand the role of regulatory T cells (Tregs) in this model, CD25 monoclonal antibody was used to deplete CD25 + Tregs. RESULTS Systemic NDP-MSH administration significantly attenuated striatal dopamine depletion and nigral dopaminergic neuron loss induced by MPTP + LPS. It improved the behavioral outcomes in the pole test. Mc1r mutant mice injected with NDP-MSH in the MPTP and LPS paradigm showed no changes in striatal dopamine levels suggesting that the NDP-MSH acts through the MC1R pathway. Although no NDP-MSH was detected in the brain, peripheral, NDP-MSH attenuated neuroinflammation as observed by diminished microglial activation in the nigral region, along with reduced TNF-α and IL1β levels in the ventral midbrain. Depletion of Tregs was associated with diminished neuroprotective effects of NDP-MSH. CONCLUSIONS Our study demonstrates that peripherally acting NDP-MSH confers protection on dopaminergic nigrostriatal neurons and reduces hyperactivated microglia. NDP-MSH modulates peripheral immune responses, and Tregs may be involved in the neuroprotective effect of NDP-MSH.
Collapse
Affiliation(s)
- Pranay Srivastava
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Shuhei Nishiyama
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Fang Zhou
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Sonia H Lin
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Akriti Srivastava
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Chienwen Su
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Yuehang Xu
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Weiyi Peng
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Michael Levy
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Michael Schwarzschild
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Xiqun Chen
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
7
|
Soares de Sá BC, Moredo LF, Torrezan GT, Fidalgo F, de Araújo ÉSS, Formiga MN, Duprat JP, Carraro DM. Characterization of Potential Melanoma Predisposition Genes in High-Risk Brazilian Patients. Int J Mol Sci 2023; 24:15830. [PMID: 37958811 PMCID: PMC10649559 DOI: 10.3390/ijms242115830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Increased genetic risk for melanoma can occur in the context of germline pathogenic variants in high-penetrance genes, such as CDKN2A and CDK4, risk variants in low- to moderate-penetrance genes (MC1R and MITF), and possibly due to variants in emerging genes, such as ACD, TERF2IP, and TERT. We aimed to identify germline variants in high- and low- to moderate-penetrance melanoma risk genes in Brazilian patients with clinical criteria for familial melanoma syndrome. We selected patients with three or more melanomas or melanoma patients from families with three tumors (melanoma and pancreatic cancer) in first- or second-degree relatives. Genetic testing was performed with a nine-gene panel (ACD, BAP1, CDK4, CDKN2A, POT1, TERT, TERF2IP, MC1R, and MITF). In 36 patients, we identified 2 (5.6%) with germline pathogenic variants in CDKN2A and BAP1 and 4 (11.1%) with variants of uncertain significance in the high-penetrance genes. MC1R variants were found in 86.5%, and both red hair color variants and unknown risk variants were enriched in patients compared to a control group. The low frequency of germline pathogenic variants in the high-penetrance genes and the high prevalence of MC1R variants found in our cohort show the importance of the MC1R genotype in determining the risk of melanoma in the Brazilian melanoma-prone families.
Collapse
Affiliation(s)
- Bianca Costa Soares de Sá
- Skin Cancer Department, A.C. Camargo Cancer Center, São Paulo 01529-001, Brazil; (B.C.S.d.S.); (L.F.M.); (J.P.D.)
| | - Luciana Facure Moredo
- Skin Cancer Department, A.C. Camargo Cancer Center, São Paulo 01529-001, Brazil; (B.C.S.d.S.); (L.F.M.); (J.P.D.)
| | - Giovana Tardin Torrezan
- Clinical and Functional Genomics Group, International Research Center/CIPE, A.C. Camargo Cancer Center, 440 Taguá St., São Paulo 01508-010, Brazil; (G.T.T.); (F.F.); (É.S.S.d.A.)
- National Institute of Science and Technology in Oncogenomics and Therapeutic Innovation, 440 Taguá St., São Paulo 01508-010, Brazil
| | - Felipe Fidalgo
- Clinical and Functional Genomics Group, International Research Center/CIPE, A.C. Camargo Cancer Center, 440 Taguá St., São Paulo 01508-010, Brazil; (G.T.T.); (F.F.); (É.S.S.d.A.)
| | - Érica Sara Souza de Araújo
- Clinical and Functional Genomics Group, International Research Center/CIPE, A.C. Camargo Cancer Center, 440 Taguá St., São Paulo 01508-010, Brazil; (G.T.T.); (F.F.); (É.S.S.d.A.)
| | | | - João Pereira Duprat
- Skin Cancer Department, A.C. Camargo Cancer Center, São Paulo 01529-001, Brazil; (B.C.S.d.S.); (L.F.M.); (J.P.D.)
| | - Dirce Maria Carraro
- Clinical and Functional Genomics Group, International Research Center/CIPE, A.C. Camargo Cancer Center, 440 Taguá St., São Paulo 01508-010, Brazil; (G.T.T.); (F.F.); (É.S.S.d.A.)
- National Institute of Science and Technology in Oncogenomics and Therapeutic Innovation, 440 Taguá St., São Paulo 01508-010, Brazil
| |
Collapse
|
8
|
Wądzyńska J, Simiczyjew A, Pietraszek-Gremplewicz K, Kot M, Ziętek M, Matkowski R, Nowak D. The impact of cellular elements of TME on melanoma biology and its sensitivity to EGFR and MET targeted therapy. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119549. [PMID: 37506884 DOI: 10.1016/j.bbamcr.2023.119549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/18/2023] [Accepted: 07/22/2023] [Indexed: 07/30/2023]
Abstract
Microenvironment of the melanoma consists of cellular elements like fibroblasts, adipocytes, and keratinocytes as well as extracellular matrix and physicochemical conditions. In our previous research, we have established that melanoma influences strongly above mentioned cells present in the tumor niche and recruits them to support cancer progression. In this work, we evaluated the impact of cancer-associated cells, namely fibroblasts (CAFs), adipocytes (CAAs), and keratinocytes (CAKs) on melanoma proliferation, signaling pathways activation, metabolism as well as the effectiveness of used anti-cancer therapy. Obtained results indicated elevated phosphorylation of STAT3, upregulated GLUT1 and GLUT3 as well as downregulated of MCT-1 expression level in melanoma cells under the influence of all examined cells present in the tumor niche. The proliferation of melanoma cells was increased after co-culture with CAFs and CAKs, while epithelial-mesenchymal transition markers' expression level was raised in the presence of CAFs and CAAs. The level of perilipin 2 and lipid content was elevated in melanoma cells under the influence of CAAs. Moreover, increased expression of CYP1A1, gene encoding drug metabolizing protein, in melanoma cells co-cultured with CAFs and CAKs prompted us to verify the effectiveness of the previously proposed by us anti-melanoma therapy based on combination of EGFR and MET inhibitors. Obtained results indicate that the designed therapy is still efficient, even if the fibroblasts, adipocytes, and keratinocytes, are present in the melanoma vicinity.
Collapse
Affiliation(s)
- Justyna Wądzyńska
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| | - Aleksandra Simiczyjew
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | | | - Magdalena Kot
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Marcin Ziętek
- Department of Oncology and Division of Surgical Oncology, Wroclaw Medical University, Plac Hirszfelda 12, 53-413 Wroclaw, Poland; Lower Silesian Oncology, Pulmonology, and Hematology Center, Plac Hirszfelda 12, 53-413 Wroclaw, Poland
| | - Rafał Matkowski
- Department of Oncology and Division of Surgical Oncology, Wroclaw Medical University, Plac Hirszfelda 12, 53-413 Wroclaw, Poland; Lower Silesian Oncology, Pulmonology, and Hematology Center, Plac Hirszfelda 12, 53-413 Wroclaw, Poland
| | - Dorota Nowak
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| |
Collapse
|
9
|
Cerdido S, Sánchez-Beltrán J, Lambertos A, Abrisqueta M, Padilla L, Herraiz C, Olivares C, Jiménez-Cervantes C, García-Borrón JC. A Side-by-Side Comparison of Wildtype and Variant Melanocortin 1 Receptor Signaling with Emphasis on Protection against Oxidative Damage to DNA. Int J Mol Sci 2023; 24:14381. [PMID: 37762683 PMCID: PMC10532403 DOI: 10.3390/ijms241814381] [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: 07/26/2023] [Revised: 09/11/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Common variants of the MC1R gene coding the α-melanocyte stimulating hormone receptor are associated with light skin, poor tanning, blond or red hair, and increased melanoma risk, due to pigment-dependent and -independent effects. This complex phenotype is usually attributed to impaired activation of cAMP signaling. However, several MC1R variants show significant residual coupling to cAMP and efficiently activate mitogenic extracellular signal-regulated kinase 1 and 2 (ERK1/2) signaling. Yet, residual signaling and the key actions of wildtype and variant MC1R have never been assessed under strictly comparable conditions in melanocytic cells of identical genetic background. We devised a strategy based on CRISPR-Cas9 knockout of endogenous MC1R in a human melanoma cell line wildtype for BRAF, NRAS and NF1, followed by reconstitution with epitope-labeled MC1R constructs, and functional analysis of clones expressing comparable levels of wildtype, R151C or D294H MC1R. The proliferation rate, shape, adhesion, motility and sensitivity to oxidative DNA damage were compared. The R151C and D294H RHC variants displayed impaired cAMP signaling, intracellular stability similar to the wildtype, triggered ERK1/2 activation as effectively as the wildtype, and afforded partial protection against oxidative DNA damage, although less efficiently than the wildtype. Therefore, common melanoma-associated MC1R variants display biased signaling and significant genoprotective activity.
Collapse
Affiliation(s)
- Sonia Cerdido
- Department of Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Murcia, 30120 Murcia, Spain; (S.C.); (J.S.-B.); (A.L.); (M.A.); (L.P.); (C.H.); (C.O.); (C.J.-C.)
- Instituto Murciano de Investigación Biosanitaria IMIB-LAIB, El Palmar, 30120 Murcia, Spain
| | - José Sánchez-Beltrán
- Department of Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Murcia, 30120 Murcia, Spain; (S.C.); (J.S.-B.); (A.L.); (M.A.); (L.P.); (C.H.); (C.O.); (C.J.-C.)
- Instituto Murciano de Investigación Biosanitaria IMIB-LAIB, El Palmar, 30120 Murcia, Spain
| | - Ana Lambertos
- Department of Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Murcia, 30120 Murcia, Spain; (S.C.); (J.S.-B.); (A.L.); (M.A.); (L.P.); (C.H.); (C.O.); (C.J.-C.)
- Instituto Murciano de Investigación Biosanitaria IMIB-LAIB, El Palmar, 30120 Murcia, Spain
| | - Marta Abrisqueta
- Department of Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Murcia, 30120 Murcia, Spain; (S.C.); (J.S.-B.); (A.L.); (M.A.); (L.P.); (C.H.); (C.O.); (C.J.-C.)
- Instituto Murciano de Investigación Biosanitaria IMIB-LAIB, El Palmar, 30120 Murcia, Spain
| | - Lidia Padilla
- Department of Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Murcia, 30120 Murcia, Spain; (S.C.); (J.S.-B.); (A.L.); (M.A.); (L.P.); (C.H.); (C.O.); (C.J.-C.)
| | - Cecilia Herraiz
- Department of Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Murcia, 30120 Murcia, Spain; (S.C.); (J.S.-B.); (A.L.); (M.A.); (L.P.); (C.H.); (C.O.); (C.J.-C.)
- Instituto Murciano de Investigación Biosanitaria IMIB-LAIB, El Palmar, 30120 Murcia, Spain
| | - Conchi Olivares
- Department of Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Murcia, 30120 Murcia, Spain; (S.C.); (J.S.-B.); (A.L.); (M.A.); (L.P.); (C.H.); (C.O.); (C.J.-C.)
- Instituto Murciano de Investigación Biosanitaria IMIB-LAIB, El Palmar, 30120 Murcia, Spain
| | - Celia Jiménez-Cervantes
- Department of Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Murcia, 30120 Murcia, Spain; (S.C.); (J.S.-B.); (A.L.); (M.A.); (L.P.); (C.H.); (C.O.); (C.J.-C.)
- Instituto Murciano de Investigación Biosanitaria IMIB-LAIB, El Palmar, 30120 Murcia, Spain
| | - José C. García-Borrón
- Department of Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Murcia, 30120 Murcia, Spain; (S.C.); (J.S.-B.); (A.L.); (M.A.); (L.P.); (C.H.); (C.O.); (C.J.-C.)
- Instituto Murciano de Investigación Biosanitaria IMIB-LAIB, El Palmar, 30120 Murcia, Spain
| |
Collapse
|
10
|
Srivastava P, Nishiyama S, Lin SH, Srivastava A, Su C, Peng W, Levy M, Schwarzschild M, Xu Y, Chen X. Peripheral MC1R activation modulates immune responses and is neuroprotective in a mouse model of Parkinson's disease. RESEARCH SQUARE 2023:rs.3.rs-3042571. [PMID: 37398302 PMCID: PMC10312952 DOI: 10.21203/rs.3.rs-3042571/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Background Melanocortin 1 receptor (MC1R) is a key pigmentation gene, and loss-of-function of MC1R variants that produce red hair may be associated with Parkinson's disease (PD). We previously reported compromised dopaminergic neuron survival in Mc1r mutant mice and dopaminergic neuroprotective effects of local injection of a MC1R agonist to the brain or a systemically administered MC1R agonist with appreciable CNS permeability. Beyond melanocytes and dopaminergic neurons, MC1R is expressed in other peripheral tissues and cell types, including immune cells. The present study investigates the impact of NDP-MSH, a synthetic melanocortin receptor (MCR) agonist that does not cross BBB, on the immune system and the nigrostriatal dopaminergic system in mouse model of PD. Methods C57BL/6 mice were treated systemically with MPTP.HCl (20 mg/kg) and LPS (1 mg/kg) from day 1 to day 4 and NDP-MSH (400 μg/kg) or vehicle from day 1 to day 12 following which the mice were sacrificed. Peripheral and CNS immune cells were phenotyped and inflammatory markers were measured. The nigrostriatal dopaminergic system was assessed behaviorally, chemically, immunologically, and pathologically. To understand the role of regulatory T cells (Tregs) in this model, CD25 monoclonal antibody was used to deplete CD25+ Tregs. Results Systemic NDP-MSH administration significantly attenuated striatal dopamine depletion and nigral dopaminergic neuron loss induced by MPTP+LPS. It improved the behavioral outcomes in the pole test. Mc1r mutant mice injected with NDP-MSH in the MPTP and LPS paradigm showed no changes in striatal dopamine levels suggesting that the NDP-MSH acts through the MC1R pathway. Although no NDP-MSH was detected in the brain, peripheral, NDP-MSH attenuated neuroinflammation as observed by diminished microglial activation in the nigral region, along with reduced TNF-α and IL1β levels in the ventral midbrain. Depletion of Tregs limited the neuroprotective effects of NDP-MSH. Conclusions Our study demonstrates that peripherally acting NDP-MSH confers protection on dopaminergic nigrostriatal neurons and reduces hyperactivated microglia. NDP-MSH modulates peripheral immune responses, and Tregs may be involved in the neuroprotective effect of NDP-MSH.
Collapse
Affiliation(s)
- Pranay Srivastava
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| | - Shuhei Nishiyama
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| | - Sonia H Lin
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| | - Akriti Srivastava
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| | - Chienwen Su
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| | - Weiyi Peng
- Department of Biology and Biochemistry, University of Houston
| | - Michael Levy
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| | - Michael Schwarzschild
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| | - Yuehang Xu
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| | - Xiqun Chen
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| |
Collapse
|
11
|
Wallingford CK, Demeshko A, Krishnakripa AK, Smit D, Duffy DL, Betz-Stablein B, Pflugfelder A, Jagirdar K, Holland E, Mann GJ, Primiero CA, Yanes T, Malvehy J, Badenas C, Carrera C, Aguilera P, Olsen C, Ward SV, Haass NK, Sturm RA, Puig S, Whiteman D, Law MH, Cust AE, Potrony M, Soyer H P, McInerney-Leo AM. The MC1R r allele does not increase melanoma risk in MITF E318K carriers. Br J Dermatol 2023; 188:770-776. [PMID: 36879448 PMCID: PMC10230961 DOI: 10.1093/bjd/ljad041] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 02/18/2023] [Indexed: 03/08/2023]
Abstract
BACKGROUND Population-wide screening for melanoma is not cost-effective, but genetic characterization could facilitate risk stratification and targeted screening. Common Melanocortin-1 receptor (MC1R) red hair colour (RHC) variants and Microphthalmia-associated transcription factor (MITF) E318K separately confer moderate melanoma susceptibility, but their interactive effects are relatively unexplored. OBJECTIVES To evaluate whether MC1R genotypes differentially affect melanoma risk in MITF E318K+ vs. E318K- individuals. MATERIALS AND METHODS Melanoma status (affected or unaffected) and genotype data (MC1R and MITF E318K) were collated from research cohorts (five Australian and two European). In addition, RHC genotypes from E318K+ individuals with and without melanoma were extracted from databases (The Cancer Genome Atlas and Medical Genome Research Bank, respectively). χ2 and logistic regression were used to evaluate RHC allele and genotype frequencies within E318K+/- cohorts depending on melanoma status. Replication analysis was conducted on 200 000 general-population exomes (UK Biobank). RESULTS The cohort comprised 1165 MITF E318K- and 322 E318K+ individuals. In E318K- cases MC1R R and r alleles increased melanoma risk relative to wild type (wt), P < 0.001 for both. Similarly, each MC1R RHC genotype (R/R, R/r, R/wt, r/r and r/wt) increased melanoma risk relative to wt/wt (P < 0.001 for all). In E318K+ cases, R alleles increased melanoma risk relative to the wt allele [odds ratio (OR) 2.04 (95% confidence interval 1.67-2.49); P = 0.01], while the r allele risk was comparable with the wt allele [OR 0.78 (0.54-1.14) vs. 1.00, respectively]. E318K+ cases with the r/r genotype had a lower but not significant melanoma risk relative to wt/wt [OR 0.52 (0.20-1.38)]. Within the E318K+ cohort, R genotypes (R/R, R/r and R/wt) conferred a significantly higher risk compared with non-R genotypes (r/r, r/wt and wt/wt) (P < 0.001). UK Biobank data supported our findings that r did not increase melanoma risk in E318K+ individuals. CONCLUSIONS RHC alleles/genotypes modify melanoma risk differently in MITF E318K- and E318K+ individuals. Specifically, although all RHC alleles increase risk relative to wt in E318K- individuals, only MC1R R increases melanoma risk in E318K+ individuals. Importantly, in the E318K+ cohort the MC1R r allele risk is comparable with wt. These findings could inform counselling and management for MITF E318K+ individuals.
Collapse
Affiliation(s)
- Courtney K Wallingford
- Frazer Institute, University of Queensland, Dermatology Research Centre, Brisbane, Australia
| | - Anastassia Demeshko
- Frazer Institute, University of Queensland, Dermatology Research Centre, Brisbane, Australia
| | | | - Darren J Smit
- Frazer Institute, University of Queensland, Dermatology Research Centre, Brisbane, Australia
| | - David L Duffy
- Frazer Institute, University of Queensland, Dermatology Research Centre, Brisbane, Australia
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland, Australia
| | - Brigid Betz-Stablein
- Frazer Institute, University of Queensland, Dermatology Research Centre, Brisbane, Australia
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland, Australia
| | - Annette Pflugfelder
- Center of Dermato-Oncology, Department of Dermatology, University of Tübingen, Tübingen, Germany
| | - Kasturee Jagirdar
- Frazer Institute, University of Queensland, Dermatology Research Centre, Brisbane, Australia
- Biochemistry and Molecular Biology Department, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Elizabeth Holland
- The Daffodil Centre, The University of Sydney, a joint venture with Cancer Council NSW, Sydney, Australia
| | - Graham J Mann
- The Melanoma Institute Australia, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Clare A Primiero
- Frazer Institute, University of Queensland, Dermatology Research Centre, Brisbane, Australia
| | - Tatiane Yanes
- Frazer Institute, University of Queensland, Dermatology Research Centre, Brisbane, Australia
| | - Josep Malvehy
- Dermatology Department, Hospital Clínic de Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| | - Cèlia Badenas
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
- Biochemistry and Molecular Genetics Department, Hospital Clínic de Barcelona, IDIBAPS, Barcelona, Spain
| | - Cristina Carrera
- Dermatology Department, Hospital Clínic de Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| | - Paula Aguilera
- Dermatology Department, Hospital Clínic de Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| | - Catherine M Olsen
- Frazer Institute, University of Queensland, Dermatology Research Centre, Brisbane, Australia
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland, Australia
| | - Sarah V Ward
- School of Population and Global Health, The University of Western Australia, Perth, WA, Australia
| | - Nikolas K Haass
- Frazer Institute, University of Queensland, Dermatology Research Centre, Brisbane, Australia
| | - Richard A Sturm
- Frazer Institute, University of Queensland, Dermatology Research Centre, Brisbane, Australia
| | - Susana Puig
- Dermatology Department, Hospital Clínic de Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| | - David C Whiteman
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland, Australia
| | - Matthew H Law
- Statistical Genetics, QIMR Berghofer Medical Research Institute, 300 Herston Rd, Herston, QLD, 4006, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
- School of Biomedical Sciences, University of Queensland, Brisbane, Australia
| | - Anne E Cust
- The Daffodil Centre, The University of Sydney, a joint venture with Cancer Council NSW, Sydney, Australia
- The Melanoma Institute Australia, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Miriam Potrony
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
- Biochemistry and Molecular Genetics Department, Hospital Clínic de Barcelona, IDIBAPS, Barcelona, Spain
| | - H Peter Soyer
- Frazer Institute, University of Queensland, Dermatology Research Centre, Brisbane, Australia
- Dermatology Department, Princess Alexandra Hospital, Brisbane, Australia
| | - Aideen M McInerney-Leo
- Frazer Institute, University of Queensland, Dermatology Research Centre, Brisbane, Australia
| |
Collapse
|
12
|
Manzari Tavakoli G, Mirzapour MH, Razi S, Rezaei N. Targeting ferroptosis as a cell death pathway in Melanoma: From molecular mechanisms to skin cancer treatment. Int Immunopharmacol 2023; 119:110215. [PMID: 37094541 DOI: 10.1016/j.intimp.2023.110215] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 03/30/2023] [Accepted: 04/17/2023] [Indexed: 04/26/2023]
Abstract
Melanoma, the most aggressive form of human skin cancer, has been under investigation to reach the most efficient treatment. Surgical resection for early-diagnosed primary melanoma, targeted therapies, and immune checkpoint inhibitors for advanced/metastatic melanoma is the best clinical approach. Ferroptosis, a newly identified iron-dependent cell death pathway, which is morphologically and biochemically different from apoptosis and necrosis, has been reported to be involved in several cancers. Ferroptosis inducers could provide therapeutic options in case of resistance to conventional therapies for advanced/metastatic melanoma. Recently developed ferroptosis inducers, MEK and BRAF inhibitors, miRNAs such as miR-137 and miR-9, and novel strategies for targeting major histocompatibility complex (MHC) class II in melanoma can provide new opportunities for melanoma treatment. Combining ferroptosis inducers with targeted therapies or immune checkpoint inhibitors increases patient response rates. Here we review the mechanisms of ferroptosis and its environmental triggers. We also discuss the pathogenesis and current treatments of melanoma. Moreover, we aim to elucidate the relationship between ferroptosis and melanoma and ferroptosis implications to develop new therapeutic strategies against melanoma.
Collapse
Affiliation(s)
- Gita Manzari Tavakoli
- Department of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran; Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohammad Hossein Mirzapour
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran; School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sepideh Razi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran; School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Stockholm, Sweden.
| |
Collapse
|
13
|
Melanogenesis and the Targeted Therapy of Melanoma. Biomolecules 2022; 12:biom12121874. [PMID: 36551302 PMCID: PMC9775438 DOI: 10.3390/biom12121874] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 11/30/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
Pigment production is a unique character of melanocytes. Numerous factors are linked with melanin production, including genetics, ultraviolet radiation (UVR) and inflammation. Understanding the mechanism of melanogenesis is crucial to identify new preventive and therapeutic strategies in the treatment of melanoma. Here, we reviewed the current available literatures on the mechanisms of melanogenesis, including the signaling pathways of UVR-induced pigment production, MC1R's central determinant roles and MITF as a master transcriptional regulator in melanogenesis. Moreover, we further highlighted the role of targeting BRAF, NRAS and MC1R in melanoma prevention and treatment. The combination therapeutics of immunotherapy and targeted kinase inhibitors are becoming the newest therapeutic option in advanced melanoma.
Collapse
|
14
|
Russo T, Piccolo V, Moscarella E, Tschandl P, Kittler H, Paoli J, Lallas A, Braun RP, Thomas L, Soyer HP, Malvehy J, Puig S, Marghoob A, Scope A, Blum A, Halpern AC, Cabo H, Menzies S, Stolz W, Tanaka M, Rabinovitz H, Hofmann-Wellenhof R, Bakos RM, Zalaudek I, Pellacani G, Veiga AV, Maceiras LR, de las Heras-Sotos C, Argenziano G. Indications for Digital Monitoring of Patients With Multiple Nevi: Recommendations from the International Dermoscopy Society. Dermatol Pract Concept 2022; 12:e2022182. [PMID: 36534527 PMCID: PMC9681223 DOI: 10.5826/dpc.1204a182] [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] [Accepted: 01/13/2022] [Indexed: 11/06/2022] Open
Abstract
Introduction In patients with multiple nevi, sequential imaging using total body skin photography (TBSP) coupled with digital dermoscopy (DD) documentation reduces unnecessary excisions and improves the early detection of melanoma. Correct patient selection is essential for optimizing the efficacy of this diagnostic approach. Objectives The purpose of the study was to identify, via expert consensus, the best indications for TBSP and DD follow-up. Methods This study was performed on behalf of the International Dermoscopy Society (IDS). We attained consensus by using an e-Delphi methodology. The panel of participants included international experts in dermoscopy. In each Delphi round, experts were asked to select from a list of indications for TBSP and DD. Results Expert consensus was attained after 3 rounds of Delphi. Participants considered a total nevus count of 60 or more nevi or the presence of a CDKN2A mutation sufficient to refer the patient for digital monitoring. Patients with more than 40 nevi were only considered an indication in case of personal history of melanoma or red hair and/or a MC1R mutation or history of organ transplantation. Conclusions Our recommendations support clinicians in choosing appropriate follow-up regimens for patients with multiple nevi and in applying the time-consuming procedure of sequential imaging more efficiently. Further studies and real-life data are needed to confirm the usefulness of this list of indications in clinical practice.
Collapse
Affiliation(s)
- Teresa Russo
- Dermatology Unit, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Vincenzo Piccolo
- Dermatology Unit, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Elvira Moscarella
- Dermatology Unit, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Philipp Tschandl
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Harald Kittler
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - John Paoli
- Department of Dermatology and Venereology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Aimilios Lallas
- First Department of Dermatology, Aristotle University, Thessaloniki, Greece
| | - Ralph P. Braun
- Department of Dermatology, University Hospital Zürich, Zürich, Switzerland
| | - Luc Thomas
- Department of Dermatology, Lyon-1 University, and Cancer research center Lyon, Lyon, France
| | - H. Peter Soyer
- The University of Queensland Diamantina Institute, The University of Queensland, Dermatology Research Centre, Brisbane, Australia
| | - Josep Malvehy
- Melanoma Unit, Dermatology Department, Hospital Clinic Barcelona, Universitat de Barcelona & IDIBAPS & CIBERER, Barcelona, Spain
| | - Susana Puig
- Melanoma Unit, Dermatology Department, Hospital Clinic Barcelona, Universitat de Barcelona & IDIBAPS & CIBERER, Barcelona, Spain
| | - Ashfaq Marghoob
- Memorial Sloan Kettering Cancer Center, Hauppauge, New York, USA
| | - Alon Scope
- The Kittner Skin Cancer Screening and Research Institute, Sheba Medical Center and Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Andreas Blum
- Public, Private and Teaching Practice of Dermatology, Konstanz, Germany
| | - Allan C. Halpern
- Memorial Sloan Kettering Cancer Center, Hauppauge, New York, USA
| | - Horacio Cabo
- Dermatology Institute of Medical Research, University of Buenos Aires, Buenos Aires, Argentina
| | - Scott Menzies
- Discipline of Dermatology, Sydney Medical School, The University of Sydney and Sydney Melanoma Diagnostic Centre, Royal Prince Alfred Hospital, Camperdown, NSW Australia
| | - Wilhelm Stolz
- Department of Dermatology, Allergology, and Environmental Medicine Clinic Thalkirchen, Hospital Munich, Munich, Germany
| | - Masaru Tanaka
- Department of Dermatology, Tokyo Women’s Medical University Medical Center East, Japan
| | - Harold Rabinovitz
- Department of Dermatology Medical College of Georgia, Augusta, United States
| | | | - Renato Marchiori Bakos
- Department of Dermatology, Hospital de Clınicas de Porto Alegre - Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Iris Zalaudek
- Department of Dermatology, University of Trieste, Trieste, Italy
| | | | - Ana Varela Veiga
- Department of Dermatology, University Hospital Complex of Ferrol, A Coruña, Spain
| | | | | | | |
Collapse
|
15
|
Wu M, Huang X, Gao L, Zhou G, Xie F. The application of photodynamic therapy in plastic and reconstructive surgery. Front Chem 2022; 10:967312. [PMID: 35936104 PMCID: PMC9353173 DOI: 10.3389/fchem.2022.967312] [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: 06/12/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
Photodynamic therapy (PDT) is a modern clinical treatment paradigm with the advantages of high selectivity, non-invasiveness, rare side-effect, no obvious drug resistance and easy combination with other therapies. These features have endowed PDT with high focus and application prospects. Studies of photodynamic therapy have been expanded in a lot of biomedical and clinical fields, especially Plastic and Reconstructive Surgery (PRS) the author major in. In this review, we emphasize the mechanism and advances in PDT related to the PRS applications including benign pigmented lesions, vascular malformations, inflammatory lesions, tumor and others. Besides, combined with clinical data analysis, the limitation of PDT and current issues that need to be addressed in the field of PRS have also been discussed. At last, a comprehensive discussion and outlooking represent future progress of PDT in PRS.
Collapse
Affiliation(s)
- Min Wu
- Department of Plastic and Reconstructive Surgery, School of Medicine, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Min Wu, ; Feng Xie,
| | - Xiaoyu Huang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Lu Gao
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Guoyu Zhou
- Department of Oral and Maxillofacial-Head Neck Oncology, School of Medicine, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Feng Xie
- Department of Plastic and Reconstructive Surgery, School of Medicine, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Min Wu, ; Feng Xie,
| |
Collapse
|
16
|
Lacson JCA, Forgas SM, Doyle SH, Qian L, Del Rio J, Valavanis S, Carvajal R, Gonzalez-Calderon G, Kim Y, Roetzheim RG, Vadaparampil ST, Kanetsky PA. Assessment of melanoma precision prevention materials incorporating MC1R genetic risk information. Transl Behav Med 2022; 12:683-687. [PMID: 35552458 PMCID: PMC9274978 DOI: 10.1093/tbm/ibac034] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Few studies have examined cognitive responses to mailed precision prevention materials. MC1R is a robust, well-described melanoma susceptibility marker. The purpose was to assess cognitive responses to generic or precision prevention materials incorporating MC1R genetic risk. Non-Hispanic White participants (n = 1134) enrolled in a randomized controlled trial received either precision prevention materials incorporating MC1R genetic risk (higher/average) or generic prevention (standard) materials. Six months after baseline, 808 (71.3%) participants reported on the amount of prevention materials read (5-point scale); believability and clarity of materials; intention to change preventive behaviors (7-point Likert scale); and recall of their MC1R genetic risk. Comparisons were conducted using Kruskal-Wallis and chi-squared tests. Overall, participants read most to all (Mdn = 4, IQR = 2) of the prevention materials, reported high believability (Mdn = 7, IQR = 1) and clarity (Mdn = 7, IQR = 1), and moderate intention to change preventive behaviors (Mdn = 5, IQR = 2). Higher-risk participants reported slightly less clarity (Mdn = 6, IQR = 2) than either average-risk (Mdn = 6, IQR = 1, p = 2.50 × 10-3) or standard participants (Mdn = 7, IQR = 1, p = 2.30 × 10-5); and slightly less believability (Mdn = 6, IQR = 1) than standard participants (Mdn = 7, IQR = 1, p = .005). Higher-risk participants were 2.21 times as likely (95% CI = 1.43-3.43) to misremember or forget their risk compared to average-risk participants; misremembering was observed only among higher-risk participants (14%). Mailed precision prevention information were mostly read, highly believable and clear, and resulted in moderate levels of intention to change sun protection behaviors, bolstering the feasibility of population-level precision prevention. Defensive reactions may explain lower clarity, believability, and higher incorrect risk recall among higher-risk participants.
Collapse
Affiliation(s)
- John Charles A Lacson
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Stephanie M Forgas
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Scarlet H Doyle
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Lu Qian
- SWOG Statistics and Data Management Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Jocelyn Del Rio
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Stella Valavanis
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Rodrigo Carvajal
- Biostatistics and Bioinformatics Shared Resource, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Guillermo Gonzalez-Calderon
- Biostatistics and Bioinformatics Shared Resource, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Youngchul Kim
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Richard G Roetzheim
- Department of Family Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Susan T Vadaparampil
- Department of Health and Behavioral Outcomes, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Peter A Kanetsky
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
17
|
Mazurkiewicz J, Simiczyjew A, Dratkiewicz E, Pietraszek-Gremplewicz K, Majkowski M, Kot M, Ziętek M, Matkowski R, Nowak D. Melanoma cells with diverse invasive potential differentially induce the activation of normal human fibroblasts. Cell Commun Signal 2022; 20:63. [PMID: 35538545 PMCID: PMC9092709 DOI: 10.1186/s12964-022-00871-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 04/01/2022] [Indexed: 12/15/2022] Open
Abstract
Background The tumor microenvironment consists of stromal cells, extracellular matrix, and physicochemical properties (e.g., oxygenation, acidification). An important element of the tumor niche are cancer-associated fibroblasts (CAFs). They may constitute up to 80% of the tumor mass and share some features with myofibroblasts involved in the process of wound healing. CAFs can facilitate cancer progression. However, their interaction with melanoma cells is still poorly understood.
Methods We obtained CAFs using conditioned media derived from primary and metastatic melanoma cells, and via co-culture with melanoma cells on Transwell inserts. Using 2D and 3D wound healing assays and Transwell invasion method we evaluated CAFs’ motile activities, while coverslips with FITC-labeled gelatin, gelatin zymography, and fluorescence-based activity assay were employed to determine the proteolytic activity of the examined cells. Western Blotting method was used for the identification of CAFs’ markers as well as estimation of the mediators of MMPs’ (matrix metalloproteinases) expression levels. Lastly, CAFs’ secretome was evaluated with cytokine and angiogenesis proteomic arrays, and lactate chemiluminescence-based assay. Results Acquired FAP-α/IL6-positive CAFs exhibited elevated motility expressed as increased migration and invasion ratio, as well as higher proteolytic activity (area of digestion, MMP2, MMP14). Furthermore, fibroblasts activated by melanoma cells showed upregulation of the MMPs’ expression mediators’ levels (pERK, p-p38, CD44, RUNX), enhanced secretion of lactate, several cytokines (IL8, IL6, CXCL1, CCL2, ICAM1), and proteins related to angiogenesis (GM-CSF, DPPIV, VEGFA, PIGF). Conclusions Observed changes in CAFs’ biology were mainly driven by highly aggressive melanoma cells (A375, WM9, Hs294T) compared to the less aggressive WM1341D cells and could promote melanoma invasion, as well as impact inflammation, angiogenesis, and acidification of the tumor niche. Interestingly, different approaches to CAFs acquisition seem to complement each other showing interactions between studied cells. Video Abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-022-00871-x.
Collapse
Affiliation(s)
- Justyna Mazurkiewicz
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383, Wrocław, Poland.
| | - Aleksandra Simiczyjew
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383, Wrocław, Poland
| | - Ewelina Dratkiewicz
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383, Wrocław, Poland
| | | | - Michał Majkowski
- Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383, Wrocław, Poland
| | - Magdalena Kot
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383, Wrocław, Poland
| | - Marcin Ziętek
- Department of Oncology and Division of Surgical Oncology, Wroclaw Medical University, Plac Hirszfelda 12, 53-413, Wrocław, Poland.,Wroclaw Comprehensive Cancer Center, Plac Hirszfelda 12, 53-413, Wrocław, Poland
| | - Rafał Matkowski
- Department of Oncology and Division of Surgical Oncology, Wroclaw Medical University, Plac Hirszfelda 12, 53-413, Wrocław, Poland.,Wroclaw Comprehensive Cancer Center, Plac Hirszfelda 12, 53-413, Wrocław, Poland
| | - Dorota Nowak
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383, Wrocław, Poland
| |
Collapse
|
18
|
Cai W, Srivastava P, Feng D, Lin Y, Vanderburg CR, Xu Y, Mclean P, Frosch MP, Fisher DE, Schwarzschild MA, Chen X. Melanocortin 1 receptor activation protects against alpha-synuclein pathologies in models of Parkinson's disease. Mol Neurodegener 2022; 17:16. [PMID: 35197079 PMCID: PMC8867846 DOI: 10.1186/s13024-022-00520-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 02/11/2022] [Indexed: 12/02/2022] Open
Abstract
Background Epidemiological studies suggest a link between the melanoma-related pigmentation gene melanocortin 1 receptor (MC1R) and risk of Parkinson’s disease (PD). We previously showed that MC1R signaling can facilitate nigrostriatal dopaminergic neuron survival. The present study investigates the neuroprotective potential of MC1R against neurotoxicity induced by alpha-synuclein (αSyn), a key player in PD genetics and pathogenesis. Methods Nigral dopaminergic neuron toxicity induced by local overexpression of aSyn was assessed in mice that have an inactivating mutation of MC1R, overexpress its wild-type transgene, or were treated with MC1R agonists. The role of nuclear factor erythroid 2-related factor 2 (Nrf2) in MC1R-mediated protection against αSyn was characterized in vitro. Furthermore, MC1R expression was determined in human postmortem midbrain from patients with PD and unaffected subjects. Results Targeted expression of αSyn in the nigrostriatal pathway induced exacerbated synuclein pathologies in MC1R mutant mice, which were accompanied by neuroinflammation and altered Nrf2 responses, and reversed by the human MC1R transgene. Two MC1R agonists were neuroprotective against αSyn-induced dopaminergic neurotoxicity. In vitro experiments showed that Nrf2 was a necessary mediator of MC1R effects. Lastly, MC1R was present in dopaminergic neurons in the human substantia nigra and appeared to be reduced at the tissue level in PD patients. Conclusion Our study supports an interaction between MC1R and αSyn that can be mediated by neuronal MC1R possibly through Nrf2. It provides evidence for MC1R as a therapeutic target and a rationale for development of MC1R-activating strategies for PD. Supplementary Information The online version contains supplementary material available at 10.1186/s13024-022-00520-4.
Collapse
Affiliation(s)
- Waijiao Cai
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA.,Department of Integrative Medicine, HuaShan Hospital, Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Pranay Srivastava
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA.,Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Towson, MD, USA
| | - Danielle Feng
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Yue Lin
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Charles R Vanderburg
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA.,Harvard NeuroDiscovery Advanced Tissue Resource Center, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Yuehang Xu
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | | | - Matthew P Frosch
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA.,Harvard NeuroDiscovery Advanced Tissue Resource Center, Massachusetts General Hospital, Harvard Medical School, Boston, USA.,Neuropathology Service, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - David E Fisher
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Michael A Schwarzschild
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA.,Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Towson, MD, USA
| | - Xiqun Chen
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA. .,Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Towson, MD, USA.
| |
Collapse
|
19
|
Nistorescu S, Udrea AM, Badea MA, Lungu I, Boni M, Tozar T, Dumitrache F, Maraloiu VA, Popescu RG, Fleaca C, Andronescu E, Dinischiotu A, Staicu A, Balas M. Low Blue Dose Photodynamic Therapy with Porphyrin-Iron Oxide Nanoparticles Complexes: In Vitro Study on Human Melanoma Cells. Pharmaceutics 2021; 13:2130. [PMID: 34959411 PMCID: PMC8705854 DOI: 10.3390/pharmaceutics13122130] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/01/2021] [Accepted: 12/03/2021] [Indexed: 01/10/2023] Open
Abstract
The purpose of this study was to investigate the effectiveness in photodynamic therapy of iron oxide nanoparticles (γ-Fe2O3 NPs), synthesized by laser pyrolysis technique, functionalized with 5,10,15,20-(Tetra-4-sulfonatophenyl) porphyrin tetraammonium (TPPS) on human cutaneous melanoma cells, after only 1 min blue light exposure. The efficiency of porphyrin loading on the iron oxide nanocarriers was estimated by using absorption and FTIR spectroscopy. The singlet oxygen yield was determined via transient characteristics of singlet oxygen phosphorescence at 1270 nm both for porphyrin functionalized nanoparticles and rose bengal used as standard. The irradiation was performed with a LED (405 nm, 1 mW/cm2) for 1 min after melanoma cells were treated with TPPS functionalized iron oxide nanoparticles (γ-Fe2O3 NPs_TPPS) and incubated for 24 h. Biological tests revealed a high anticancer effect of γ-Fe2O3 NPs_TPPS complexes indi-cated by the inhibition of tumor cell proliferation, reduction of cell adhesion, and induction of cell death through ROS generated by TPPS under light exposure. The biological assays were combined with the pharmacokinetic prediction of the porphyrin.
Collapse
Affiliation(s)
- Simona Nistorescu
- National Institute of Laser, Plasma and Radiation Physics, 409 Atomistilor Str., 077125 Magurele, Romania; (S.N.); (A.-M.U.); (I.L.); (M.B.); (T.T.); (F.D.); (C.F.)
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (M.A.B.); (R.G.P.); (A.D.)
| | - Ana-Maria Udrea
- National Institute of Laser, Plasma and Radiation Physics, 409 Atomistilor Str., 077125 Magurele, Romania; (S.N.); (A.-M.U.); (I.L.); (M.B.); (T.T.); (F.D.); (C.F.)
- Research Institute of the University of Bucharest, Earth, Environmental and Life Sciences, Section-ICUB, 050663 Bucharest, Romania
| | - Madalina Andreea Badea
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (M.A.B.); (R.G.P.); (A.D.)
| | - Iulia Lungu
- National Institute of Laser, Plasma and Radiation Physics, 409 Atomistilor Str., 077125 Magurele, Romania; (S.N.); (A.-M.U.); (I.L.); (M.B.); (T.T.); (F.D.); (C.F.)
- Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 1-7 Gh. Polizu Str., 011061 Bucharest, Romania;
| | - Mihai Boni
- National Institute of Laser, Plasma and Radiation Physics, 409 Atomistilor Str., 077125 Magurele, Romania; (S.N.); (A.-M.U.); (I.L.); (M.B.); (T.T.); (F.D.); (C.F.)
| | - Tatiana Tozar
- National Institute of Laser, Plasma and Radiation Physics, 409 Atomistilor Str., 077125 Magurele, Romania; (S.N.); (A.-M.U.); (I.L.); (M.B.); (T.T.); (F.D.); (C.F.)
| | - Florian Dumitrache
- National Institute of Laser, Plasma and Radiation Physics, 409 Atomistilor Str., 077125 Magurele, Romania; (S.N.); (A.-M.U.); (I.L.); (M.B.); (T.T.); (F.D.); (C.F.)
| | | | - Roua Gabriela Popescu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (M.A.B.); (R.G.P.); (A.D.)
| | - Claudiu Fleaca
- National Institute of Laser, Plasma and Radiation Physics, 409 Atomistilor Str., 077125 Magurele, Romania; (S.N.); (A.-M.U.); (I.L.); (M.B.); (T.T.); (F.D.); (C.F.)
| | - Ecaterina Andronescu
- Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 1-7 Gh. Polizu Str., 011061 Bucharest, Romania;
| | - Anca Dinischiotu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (M.A.B.); (R.G.P.); (A.D.)
| | - Angela Staicu
- National Institute of Laser, Plasma and Radiation Physics, 409 Atomistilor Str., 077125 Magurele, Romania; (S.N.); (A.-M.U.); (I.L.); (M.B.); (T.T.); (F.D.); (C.F.)
| | - Mihaela Balas
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (M.A.B.); (R.G.P.); (A.D.)
| |
Collapse
|
20
|
Morkavuk ŞB, Çulcu S, Esen E, Ünal AE. The diagnostic value of modified systemic ınflammation score in predicting post-operative outcomes of cutaneous melanoma patients who underwent ısolated limb perfusion. World J Surg Oncol 2021; 19:327. [PMID: 34781987 PMCID: PMC8594072 DOI: 10.1186/s12957-021-02437-6] [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: 07/09/2021] [Accepted: 11/01/2021] [Indexed: 11/18/2022] Open
Abstract
Background In-transit metastasis is considered a locoregional disease in cutaneous melanoma (CM) patients. Isolated limb perfusion (ILP) is among the treatment options in selected cases. The aim of this study was to determine the success of pre- and post-perfusion mSIS values in predicting the potential complications and the prognosis of the disease by investigating the early and long-term results of mSIS values calculated before and after ILP in CM cases with in-transit metastases. Materials and methods Patients who underwent ILP within the period from 2014 to 2020 in our department were retrospectively scanned. A total of 20 patients were found to undergo ILP. The scores obtained from modified inflammation score (mSIS) were formulated according to albumin (Alb) and lymphocyte to monocyte ratio (LMR) scores. Results The mean follow-up time was 20.47 months. Complications requiring surgical intervention developed in three patients. According to the Wieberdink local toxicity classification, the majority (70%) of the patients were found to be grade II. Based on pre-perfusion mSIS values, 8 patients were classified as mSIS 0 while six patients were classified as mSIS 1 and 2. Based on post-perfusion mSIS values, 14 patients and one patient were classified as mSIS 2 (70%) and mSIS 0, respectively. Accordingly, univariate analysis showed that mSIS 1 and mSIS 2 were negative prognostic factors for mean survival in the pre-perfusion period (HR 0.162, 95% CI 0.036–0.729; p = 0.018 and HR: 0.223, 95% CI 0.049–1.019; p = 0.053) whereas albumin (Alb) and lymphocyte to monocyte ratio (LMR) were not independent prognostic factors for mean survival. Conclusion The mSIS values calculated in the pre-perfusion period can give an opinion about the OS of the patients whereas post-perfusion mSIS values may predict potential surgical complications and local toxicities.
Collapse
Affiliation(s)
| | - Serdar Çulcu
- Department of Surgical Oncology, Dr. Abdurrahman Yurtaslan Research and Training Hospital, Ankara, Turkey
| | - Ebru Esen
- Department of Surgical Oncology, Ankara Gülhane Research and Training Hospital, Ankara, Turkey
| | - Ali Ekrem Ünal
- Department of Surgical Oncology, Ankara University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
21
|
Guida S, Guida G, Goding CR. MC1R Functions, Expression, and Implications for Targeted Therapy. J Invest Dermatol 2021; 142:293-302.e1. [PMID: 34362555 DOI: 10.1016/j.jid.2021.06.018] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/14/2021] [Accepted: 06/17/2021] [Indexed: 12/23/2022]
Abstract
The G protein-coupled MC1R is expressed in melanocytes and has a pivotal role in human skin pigmentation, with reduced function in human genetic variants exhibiting a red hair phenotype and increased melanoma predisposition. Beyond its role in pigmentation, MC1R is increasingly recognized as promoting UV-induced DNA damage repair. Consequently, there is mounting interest in targeting MC1R for therapeutic benefit. However, whether MC1R expression is restricted to melanocytes or is more widely expressed remains a matter of debate. In this paper, we review MC1R function and highlight that unbiased analysis suggests that its expression is restricted to melanocytes, granulocytes, and the brain.
Collapse
Affiliation(s)
- Stefania Guida
- Dermatology Unit, Surgical, Medical and Dental Department of Morphological Sciences Related to Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy.
| | - Gabriella Guida
- Molecular Biology Section, Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Colin Ronald Goding
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
22
|
Manganelli M, Guida S, Ferretta A, Pellacani G, Porcelli L, Azzariti A, Guida G. Behind the Scene: Exploiting MC1R in Skin Cancer Risk and Prevention. Genes (Basel) 2021; 12:1093. [PMID: 34356109 PMCID: PMC8305013 DOI: 10.3390/genes12071093] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/14/2021] [Accepted: 07/16/2021] [Indexed: 02/06/2023] Open
Abstract
Melanoma and non-melanoma skin cancers (NMSCs) are the most frequent cancers of the skin in white populations. An increased risk in the development of skin cancers has been associated with the combination of several environmental factors (i.e., ultraviolet exposure) and genetic background, including melanocortin-1 receptor (MC1R) status. In the last few years, advances in the diagnosis of skin cancers provided a great impact on clinical practice. Despite these advances, NMSCs are still the most common malignancy in humans and melanoma still shows a rising incidence and a poor prognosis when diagnosed at an advanced stage. Efforts are required to underlie the genetic and clinical heterogeneity of melanoma and NMSCs, leading to an optimization of the management of affected patients. The clinical implications of the impact of germline MC1R variants in melanoma and NMSCs' risk, together with the additional risk conferred by somatic mutations in other peculiar genes, as well as the role of MC1R screening in skin cancers' prevention will be addressed in the current review.
Collapse
Affiliation(s)
- Michele Manganelli
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari-“Aldo Moro”, 70125 Bari, Italy; (M.M.); (A.F.)
- DMMT-Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Stefania Guida
- Department of Surgical-Medical-Dental and Morphological Science with Interest Transplant-Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, 41124 Modena, Italy;
| | - Anna Ferretta
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari-“Aldo Moro”, 70125 Bari, Italy; (M.M.); (A.F.)
| | - Giovanni Pellacani
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Dermatology Clinic, Sapienza University of Rome, 00161 Rome, Italy;
| | - Letizia Porcelli
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy; (L.P.); (A.A.)
| | - Amalia Azzariti
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy; (L.P.); (A.A.)
| | - Gabriella Guida
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari-“Aldo Moro”, 70125 Bari, Italy; (M.M.); (A.F.)
| |
Collapse
|
23
|
Chatterjee S, Chakraborty R, Hasija Y. Polymorphisms at site 469 of B-RAF protein associated with skin melanoma may be correlated with dabrafenib resistance: An in silico study. J Biomol Struct Dyn 2021; 40:10862-10877. [PMID: 34278963 DOI: 10.1080/07391102.2021.1950571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 06/28/2021] [Indexed: 12/24/2022]
Abstract
Melanoma is a type of skin cancer. Numerous genes and their proteins are strongly associated with melanoma susceptibility. This study aims to use an in silico method to identify genetic variants in the melanoma susceptibility gene. The COSMIC database was queried for genes and cross-referenced with three environment-gene interaction databases (EGP, SeattleSNPs and CTD) to identify shared genes. The majority of approved skin melanoma drugs were found to act on the protein serine/threonine-protein kinase (B-RAF) encoded by the BRAF gene, which was also present in all three referenced databases. Comprehensive computational analysis was performed to predict deleterious genetic variants associated with skin melanoma, and the nsSNPs G469V and G469E were prioritized based on their predicted deleterious effects. Molecular dynamic simulation analysis of the B-RAF protein mutants G469V and G469E reveals that variations in the amino acid conformation at the drug binding site result in inconsistency in drug interaction. Additionally, this analysis showed that the G469V and G469E mutants have lower binding energy for dabrafenib than the wild type. The population with the highest frequency of each deleterious and pathogenic variant has been determined. The study's findings would support the development of more effective treatment strategies for skin melanoma. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | | | - Yasha Hasija
- Department of Biotechnology, Delhi Technological University, Delhi, India
| |
Collapse
|
24
|
Abrahamian C, Grimm C. Endolysosomal Cation Channels and MITF in Melanocytes and Melanoma. Biomolecules 2021; 11:biom11071021. [PMID: 34356645 PMCID: PMC8301777 DOI: 10.3390/biom11071021] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 01/25/2023] Open
Abstract
Microphthalmia-associated transcription factor (MITF) is the principal transcription factor regulating pivotal processes in melanoma cell development, growth, survival, proliferation, differentiation and invasion. In recent years, convincing evidence has been provided attesting key roles of endolysosomal cation channels, specifically TPCs and TRPMLs, in cancer, including breast cancer, glioblastoma, bladder cancer, hepatocellular carcinoma and melanoma. In this review, we provide a gene expression profile of these channels in different types of cancers and decipher their roles, in particular the roles of two-pore channel 2 (TPC2) and TRPML1 in melanocytes and melanoma. We specifically discuss the signaling cascades regulating MITF and the relationship between endolysosomal cation channels, MAPK, canonical Wnt/GSK3 pathways and MITF.
Collapse
|
25
|
Molecular landscape of Hereditary Melanoma. Crit Rev Oncol Hematol 2021; 164:103425. [PMID: 34245855 DOI: 10.1016/j.critrevonc.2021.103425] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 05/20/2021] [Accepted: 07/04/2021] [Indexed: 12/27/2022] Open
Abstract
Melanoma is considered the most lethal skin cancer and its incidence has increased during the past decades. About 10 % of cases are classified as hereditary melanoma. Genetic predisposition usually manifests itself clinically as early onset and multiple cutaneous melanomas. Several genes have been identified as involved to melanoma susceptibility, some of them still with unknown clinical relevance. Beyond melanoma, the affected families are also more prone to develop other malignancies, such as pancreatic cancer. The identification of risk families and involved genes is of great importance, since different forms of oncological surveillance are recommended. However, well established guidelines to standardize both the selection of individuals and the genetic panel to be requested are still lacking. Given the importance of the genetic counseling and testing in the context of clinical suspicion of hereditary melanoma, this paper aims to review the literature regarding genetic panel indications worldwide.
Collapse
|
26
|
Anderson JL, Kruisselbrink TM, Lisi EC, Hughes TM, Steyermark JM, Winkler EM, Berg CM, Vierkant RA, Gupta R, Ali AH, Faubion SS, Aoudia SL, McAllister TM, Farrugia G, Stewart AK, Lazaridis KN. Clinically Actionable Findings Derived From Predictive Genomic Testing Offered in a Medical Practice Setting. Mayo Clin Proc 2021; 96:1407-1417. [PMID: 33890576 DOI: 10.1016/j.mayocp.2020.08.051] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 08/28/2020] [Accepted: 08/31/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To assess the presence of clinically actionable results and other genetic findings in an otherwise healthy population of adults seen in a medical practice setting and offered "predictive" genomic testing. PATIENTS AND METHODS In 2014, a predictive genomics clinic for generally healthy adults was launched through the Mayo Clinic Executive Health Program. Self-identified interested patients met with a genomic nurse and genetic counselor for pretest advice and education. Two genome sequencing platforms and one gene panel-based health screen were offered. Posttest genetic counseling was available for patients who elected testing. From March 1, 2014, through June 1, 2019, 1281 patients were seen and 301 (23.5%) chose testing. Uptake rates increased to 36.3% [70 of 193]) in 2019 from 11.8% [2 of 17] in 2014. Clinically actionable results and genetic findings were analyzed using descriptive statistics. RESULTS Clinically actionable results were detected in 11.6% of patients (35 of 301), and of those, 51.7% (15 of 29) with a cancer or cardiovascular result = did not have a personal or family history concerning for a hereditary disorder. The most common actionable results were in the BCHE, BRCA2, CHEK2, LDLR, MUTYH, and MYH7 genes. A carrier of at least one recessive condition was found in 53.8% of patients (162 of 301). At least one variant associated with multifactorial disease was found in 44.5% (134 of 301) (eg, 25 patients were heterozygous for the F5 factor V Leiden variant associated with thrombophilia risk). CONCLUSION Our predictive screening revealed that 11.6% of individuals will test positive for a clinically actionable, likely pathogenic/pathogenic variant. This finding suggests that wider knowledge and adoption of predictive genomic services could be beneficial in medical practice, although additional studies are needed.
Collapse
Affiliation(s)
| | | | - Emily C Lisi
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | | | | | - Erin M Winkler
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Corinne M Berg
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Robert A Vierkant
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN
| | - Ruchi Gupta
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN
| | - Ahmad H Ali
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | | | - Stacy L Aoudia
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | | | - Gianrico Farrugia
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - A Keith Stewart
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Konstantinos N Lazaridis
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN; Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN.
| |
Collapse
|
27
|
Podlipnik S, Potrony M, Puig S. Genetic markers for characterization and prediction of prognosis of melanoma subtypes: a 2021 update. Ital J Dermatol Venerol 2021; 156:322-330. [PMID: 33982545 DOI: 10.23736/s2784-8671.21.06957-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In this article we examined the most important genetic markers involved in melanoma susceptibility, initiation and progression, and their impact on the prognosis of the disease. Current knowledge in melanoma genetics identifies distinct pathways to the development of different melanoma subtypes characterized by specific clinico-pathological features and partially known genetic markers, modulated by high, low or absence of cumulative sun damage. The most prevalent somatic mutations are related to the activation of the MAPK pathway, which are classified into four major subtypes: BRAF mutant, NRAS mutant, NF1 mutant and triple wild type. Moreover, germinal mutations are also involved in the characterization and predictions of prognosis in melanoma. Currently, CDKN2A is seen as the main high-risk gene involved in melanoma susceptibility being mutated in around 20% of melanoma-prone families. Other high-risk susceptibility genes described include CDK4, POT1, BAP1, TERT promoter, ACD, and TERF2IP. Melanoma is one of the most genetically predisposed among all cancers in humans, and ultraviolet light from the sun is the main environmental factor. This genetic predisposition is starting to be understood, impacting not only on the risk of developing melanoma but also on the risk of developing other types of cancer, as well as on the prognosis of the disease.
Collapse
Affiliation(s)
- Sebastian Podlipnik
- Department of Dermatology, University of Barcelona, Hospital of Barcelona, Barcelona, Spain.,Unit of Melanoma, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Miriam Potrony
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain.,Department of Biochemistry and Molecular Genetics, Hospital of Barcelona, Barcelona, Spain
| | - Susana Puig
- Department of Dermatology, University of Barcelona, Hospital of Barcelona, Barcelona, Spain - .,Unit of Melanoma, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| |
Collapse
|
28
|
Hubert JN, Suybeng V, Vallée M, Delhomme TM, Maubec E, Boland A, Bacq D, Deleuze JF, Jouenne F, Brennan P, McKay JD, Avril MF, Bressac-de Paillerets B, Chanudet E. The PI3K/mTOR Pathway Is Targeted by Rare Germline Variants in Patients with Both Melanoma and Renal Cell Carcinoma. Cancers (Basel) 2021; 13:2243. [PMID: 34067022 PMCID: PMC8125037 DOI: 10.3390/cancers13092243] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 12/24/2022] Open
Abstract
Background: Malignant melanoma and RCC have different embryonic origins, no common lifestyle risk factors but intriguingly share biological properties such as immune regulation and radioresistance. An excess risk of malignant melanoma is observed in RCC patients and vice versa. This bidirectional association is poorly understood, and hypothetic genetic co-susceptibility remains largely unexplored. Results: We hereby provide a clinical and genetic description of a series of 125 cases affected by both malignant melanoma and RCC. Clinical germline mutation testing identified a pathogenic variant in a melanoma and/or RCC predisposing gene in 17/125 cases (13.6%). This included mutually exclusive variants in MITF (p.E318K locus, N = 9 cases), BAP1 (N = 3), CDKN2A (N = 2), FLCN (N = 2), and PTEN (N = 1). A subset of 46 early-onset cases, without underlying germline variation, was whole-exome sequenced. In this series, thirteen genes were significantly enriched in mostly exclusive rare variants predicted to be deleterious, compared to 19,751 controls of similar ancestry. The observed variation mainly consisted of novel or low-frequency variants (<0.01%) within genes displaying strong evolutionary mutational constraints along the PI3K/mTOR pathway, including PIK3CD, NFRKB, EP300, MTOR, and related epigenetic modifier SETD2. The screening of independently processed germline exomes from The Cancer Genome Atlas confirmed an association with melanoma and RCC but not with cancers of established differing etiology such as lung cancers. Conclusions: Our study highlights that an exome-wide case-control enrichment approach may better characterize the rare variant-based missing heritability of multiple primary cancers. In our series, the co-occurrence of malignant melanoma and RCC was associated with germline variation in the PI3K/mTOR signaling cascade, with potential relevance for early diagnostic and clinical management.
Collapse
Affiliation(s)
- Jean-Noël Hubert
- Section of Genetics, International Agency for Research on Cancer (IARC-WHO), 69372 Lyon, France; (J.-N.H.); (M.V.); (T.M.D.); (P.B.); (J.D.M.)
| | - Voreak Suybeng
- Gustave Roussy, Département de Biopathologie, 94805 Villejuif, France; (V.S.); (F.J.)
| | - Maxime Vallée
- Section of Genetics, International Agency for Research on Cancer (IARC-WHO), 69372 Lyon, France; (J.-N.H.); (M.V.); (T.M.D.); (P.B.); (J.D.M.)
| | - Tiffany M. Delhomme
- Section of Genetics, International Agency for Research on Cancer (IARC-WHO), 69372 Lyon, France; (J.-N.H.); (M.V.); (T.M.D.); (P.B.); (J.D.M.)
| | - Eve Maubec
- Department of Dermatology, AP-HP, Hôpital Avicenne, University Paris 13, 93000 Bobigny, France;
- UMRS-1124, Campus Paris Saint-Germain-des-Prés, University of Paris, 75006 Paris, France
| | - Anne Boland
- Centre National de Recherche en Génomique Humaine, Université Paris-Saclay, CEA, 91057 Evry, France; (A.B.); (D.B.); (J.-F.D.)
| | - Delphine Bacq
- Centre National de Recherche en Génomique Humaine, Université Paris-Saclay, CEA, 91057 Evry, France; (A.B.); (D.B.); (J.-F.D.)
| | - Jean-François Deleuze
- Centre National de Recherche en Génomique Humaine, Université Paris-Saclay, CEA, 91057 Evry, France; (A.B.); (D.B.); (J.-F.D.)
| | - Fanélie Jouenne
- Gustave Roussy, Département de Biopathologie, 94805 Villejuif, France; (V.S.); (F.J.)
| | - Paul Brennan
- Section of Genetics, International Agency for Research on Cancer (IARC-WHO), 69372 Lyon, France; (J.-N.H.); (M.V.); (T.M.D.); (P.B.); (J.D.M.)
| | - James D. McKay
- Section of Genetics, International Agency for Research on Cancer (IARC-WHO), 69372 Lyon, France; (J.-N.H.); (M.V.); (T.M.D.); (P.B.); (J.D.M.)
| | | | - Brigitte Bressac-de Paillerets
- Gustave Roussy, Département de Biopathologie, 94805 Villejuif, France; (V.S.); (F.J.)
- INSERM U1279, Tumor Cell Dynamics, 94805 Villejuif, France
| | - Estelle Chanudet
- Section of Genetics, International Agency for Research on Cancer (IARC-WHO), 69372 Lyon, France; (J.-N.H.); (M.V.); (T.M.D.); (P.B.); (J.D.M.)
| |
Collapse
|
29
|
Herraiz C, Martínez-Vicente I, Maresca V. The α-melanocyte-stimulating hormone/melanocortin-1 receptor interaction: A driver of pleiotropic effects beyond pigmentation. Pigment Cell Melanoma Res 2021; 34:748-761. [PMID: 33884776 DOI: 10.1111/pcmr.12980] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/25/2021] [Accepted: 04/13/2021] [Indexed: 12/21/2022]
Abstract
Melanocortin-1 Receptor (MC1R), when stimulated by alpha-melanocyte-stimulating hormone (α-MSH), is a driver of eumelanogenesis. Brown/black eumelanin is an effective filter against ultraviolet radiation (UVR) and is a scavenger of free radicals. Several polymorphic variants of MC1R are frequent in red-head people. These polymorphisms reduce the ability of MC1R to promote eumelanogenesis after its activation and spontaneous pheomelanogenesis take place. Since pheomelanin can act as an endogenous photosensitizer, people carrying MC1R polymorphisms are more susceptible to skin cancer. Here, we summarize current knowledge on the biology of MC1R beyond its ability to drive eumelanogenesis. We analyze its capacity to cope with oxidative insult and consequent DNA damage. We describe its ability to transduce through different pathways. We start from the canonical pathway, the cAMP/protein kinase A (PKA) pathway mainly involved in promoting eumelanogenesis, and protection from oxidative damage, and we then move on to describe more recent knowledge concerning ERK pathways, phosphoinositide 3-kinase (PI3K) pathway/AKT, and α-MSH/Peroxisome proliferators activated receptor-γ (PPAR-γ) connection. We describe MC1R polymorphic variants associated with melanoma risk which represent an open window of clinical relevance.
Collapse
Affiliation(s)
- Cecilia Herraiz
- Department of Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Murcia and Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Idoya Martínez-Vicente
- Department of Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Murcia and Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Vittoria Maresca
- Laboratory of Cutaneous Physiopathology, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| |
Collapse
|
30
|
Mazurkiewicz J, Simiczyjew A, Dratkiewicz E, Ziętek M, Matkowski R, Nowak D. Stromal Cells Present in the Melanoma Niche Affect Tumor Invasiveness and Its Resistance to Therapy. Int J Mol Sci 2021; 22:E529. [PMID: 33430277 PMCID: PMC7825728 DOI: 10.3390/ijms22020529] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 12/29/2020] [Accepted: 01/05/2021] [Indexed: 12/14/2022] Open
Abstract
Malignant melanoma is a highly metastatic type of cancer, which arises frequently from transformed pigment cells and melanocytes as a result of long-term UV radiation exposure. In recent years, the incidence of newly diagnosed melanoma patients reached 5% of all cancer cases. Despite the development of novel targeted therapies directed against melanoma-specific markers, patients' response to treatment is often weak or short-term due to a rapid acquisition of drug resistance. Among the factors affecting therapy effectiveness, elements of the tumor microenvironment play a major role. Melanoma niche encompasses adjacent cells, such as keratinocytes, cancer-associated fibroblasts (CAFs), adipocytes, and immune cells, as well as components of the extracellular matrix and tumor-specific physicochemical properties. In this review, we summarize the current knowledge concerning the influence of cancer-associated cells (keratinocytes, CAFs, adipocytes) on the process of melanomagenesis, tumor progression, invasiveness, and the emergence of drug resistance in melanoma. We also address how melanoma can alter the differentiation and activation status of cells present in the tumor microenvironment. Understanding these complex interactions between malignant and cancer-associated cells could improve the development of effective antitumor therapeutic strategies.
Collapse
Affiliation(s)
- Justyna Mazurkiewicz
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland; (A.S.); (E.D.); (D.N.)
| | - Aleksandra Simiczyjew
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland; (A.S.); (E.D.); (D.N.)
| | - Ewelina Dratkiewicz
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland; (A.S.); (E.D.); (D.N.)
| | - Marcin Ziętek
- Department of Oncology and Division of Surgical Oncology, Wroclaw Medical University, Plac Hirszfelda 12, 53-413 Wroclaw, Poland; (M.Z.); (R.M.)
- Wroclaw Comprehensive Cancer Center, Plac Hirszfelda 12, 53-413 Wroclaw, Poland
| | - Rafał Matkowski
- Department of Oncology and Division of Surgical Oncology, Wroclaw Medical University, Plac Hirszfelda 12, 53-413 Wroclaw, Poland; (M.Z.); (R.M.)
- Wroclaw Comprehensive Cancer Center, Plac Hirszfelda 12, 53-413 Wroclaw, Poland
| | - Dorota Nowak
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland; (A.S.); (E.D.); (D.N.)
| |
Collapse
|
31
|
Lavelle TJ, Alver TN, Heintz KM, Wernhoff P, Nygaard V, Nakken S, Øy GF, Bøe SL, Urbanucci A, Hovig E. Dysregulation of MITF Leads to Transformation in MC1R-Defective Melanocytes. Cancers (Basel) 2020; 12:cancers12071719. [PMID: 32605315 PMCID: PMC7408466 DOI: 10.3390/cancers12071719] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/20/2020] [Accepted: 06/24/2020] [Indexed: 12/12/2022] Open
Abstract
The MC1R/cAMP/MITF pathway is a key determinant for growth, differentiation, and survival of melanocytes and melanoma. MITF-M is the melanocyte-specific isoform of Microphthalmia-associated Transcription Factor (MITF) in human melanoma. Here we use two melanocyte cell lines to show that forced expression of hemagglutinin (HA) -tagged MITF-M through lentiviral transduction represents an oncogenic insult leading to consistent cell transformation of the immortalized melanocyte cell line Hermes 4C, being a melanocortin-1 receptor (MC1R) compound heterozygote, while not causing transformation of the MC1R wild type cell line Hermes 3C. The transformed HA-tagged MITF-M transduced Hermes 4C cells form colonies in soft agar and tumors in mice. Further, Hermes 4C cells display increased MITF chromatin binding, and transcriptional reprogramming consistent with an invasive melanoma phenotype. Mechanistically, forced expression of MITF-M drives the upregulation of the AXL tyrosine receptor kinase (AXL), with concomitant downregulation of phosphatase and tensin homolog (PTEN), leading to increased activation of the PI3K/AKT pathway. Treatment with AXL inhibitors reduces growth of the transformed cells by reverting AKT activation. In conclusion, we present a model system of melanoma development, driven by MITF-M in the context of MC1R loss of function, and independent of UV exposure. This model provides a basis for further studies of critical changes in the melanocyte transformation process.
Collapse
Affiliation(s)
- Timothy J. Lavelle
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, 0424 Oslo, Norway; (T.J.L.); (T.N.A.); (K.-M.H.); (P.W.); (V.N.); (S.N.); (G.F.Ø.)
| | - Tine Norman Alver
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, 0424 Oslo, Norway; (T.J.L.); (T.N.A.); (K.-M.H.); (P.W.); (V.N.); (S.N.); (G.F.Ø.)
| | - Karen-Marie Heintz
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, 0424 Oslo, Norway; (T.J.L.); (T.N.A.); (K.-M.H.); (P.W.); (V.N.); (S.N.); (G.F.Ø.)
| | - Patrik Wernhoff
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, 0424 Oslo, Norway; (T.J.L.); (T.N.A.); (K.-M.H.); (P.W.); (V.N.); (S.N.); (G.F.Ø.)
| | - Vegard Nygaard
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, 0424 Oslo, Norway; (T.J.L.); (T.N.A.); (K.-M.H.); (P.W.); (V.N.); (S.N.); (G.F.Ø.)
| | - Sigve Nakken
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, 0424 Oslo, Norway; (T.J.L.); (T.N.A.); (K.-M.H.); (P.W.); (V.N.); (S.N.); (G.F.Ø.)
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0424 Oslo, Norway
| | - Geir Frode Øy
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, 0424 Oslo, Norway; (T.J.L.); (T.N.A.); (K.-M.H.); (P.W.); (V.N.); (S.N.); (G.F.Ø.)
| | - Sigurd Leinæs Bøe
- Department of Medical Biochemistry, Oslo University Hospital, Radiumhospitalet, 0424 Oslo, Norway;
| | - Alfonso Urbanucci
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, 0424 Oslo, Norway; (T.J.L.); (T.N.A.); (K.-M.H.); (P.W.); (V.N.); (S.N.); (G.F.Ø.)
- Correspondence: (A.U.); (E.H.)
| | - Eivind Hovig
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, 0424 Oslo, Norway; (T.J.L.); (T.N.A.); (K.-M.H.); (P.W.); (V.N.); (S.N.); (G.F.Ø.)
- Department of Informatics, University of Oslo, 0316 Oslo, Norway
- Correspondence: (A.U.); (E.H.)
| |
Collapse
|
32
|
Calbet-Llopart N, Pascini-Garrigos M, Tell-Martí G, Potrony M, Martins da Silva V, Barreiro A, Puig S, Captier G, James I, Degardin N, Carrera C, Malvehy J, Etchevers HC, Puig-Butillé JA. Melanocortin-1 receptor (MC1R) genotypes do not correlate with size in two cohorts of medium-to-giant congenital melanocytic nevi. Pigment Cell Melanoma Res 2020; 33:685-694. [PMID: 32323445 DOI: 10.1111/pcmr.12883] [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: 01/21/2020] [Revised: 04/07/2020] [Accepted: 04/16/2020] [Indexed: 01/29/2023]
Abstract
Congenital melanocytic nevi (CMN) are cutaneous malformations whose prevalence is inversely correlated with projected adult size. CMN are caused by somatic mutations, but epidemiological studies suggest that germline genetic factors may influence CMN development. In CMN patients from the U.K., genetic variants in MC1R, such as p.V92M and loss-of-function variants, have been previously associated with larger CMN. We analyzed the association of MC1R variants with CMN characteristics in two distinct cohorts of medium-to-giant CMN patients from Spain (N = 113) and from France, Norway, Canada, and the United States (N = 53), similar at the clinical and phenotypical level except for the number of nevi per patient. We found that the p.V92M or loss-of-function MC1R variants either alone or in combination did not correlate with CMN size, in contrast to the U.K. CMN patients. An additional case-control analysis with 259 unaffected Spanish individuals showed a higher frequency of MC1R compound heterozygous or homozygous variant genotypes in Spanish CMN patients compared to the control population (15.9% vs. 9.3%; p = .075). Altogether, this study suggests that MC1R variants are not associated with CMN size in these non-UK cohorts. Additional studies are required to define the potential role of MC1R as a risk factor in CMN development.
Collapse
Affiliation(s)
- Neus Calbet-Llopart
- Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain
| | - Mirella Pascini-Garrigos
- Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Gemma Tell-Martí
- Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain
| | - Miriam Potrony
- Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain
| | - Vanessa Martins da Silva
- Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Alicia Barreiro
- Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Susana Puig
- Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain
| | - Guillaume Captier
- Plastic pediatric surgery, University of Montpellier Hospital, Montpellier, France
| | - Isabelle James
- Service de Chirurgie Réparatrice de l'Enfant, Clinique du Val d'Ouest, Ecully, France
| | - Nathalie Degardin
- Service de Chirurgie Plastique Réparatrice, Hôpital de la Timone Enfants, Marseille, France.,Faculté de Médecine, Marseille Medical Genetics, Aix-Marseille Univ, INSERM, U1251, Marseille, France
| | - Cristina Carrera
- Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain
| | - Josep Malvehy
- Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain
| | - Heather C Etchevers
- Faculté de Médecine, Marseille Medical Genetics, Aix-Marseille Univ, INSERM, U1251, Marseille, France
| | - Joan Anton Puig-Butillé
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain.,Molecular Biology CORE, Biomedical Diagnostic Center (CDB), Melanoma Unit, Hospital Clínic de Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain
| |
Collapse
|
33
|
Yang K, Oak AS, Slominski RM, Brożyna AA, Slominski AT. Current Molecular Markers of Melanoma and Treatment Targets. Int J Mol Sci 2020; 21:ijms21103535. [PMID: 32429485 PMCID: PMC7278971 DOI: 10.3390/ijms21103535] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/08/2020] [Accepted: 05/13/2020] [Indexed: 12/17/2022] Open
Abstract
Melanoma is a deadly skin cancer that becomes especially difficult to treat after it metastasizes. Timely identification of melanoma is critical for effective therapy, but histopathologic diagnosis can frequently pose a significant challenge to this goal. Therefore, auxiliary diagnostic tools are imperative to facilitating prompt recognition of malignant lesions. Melanoma develops as result of a number of genetic mutations, with UV radiation often acting as a mutagenic risk factor. Novel methods of genetic testing have improved detection of these molecular alterations, which subsequently revealed important information for diagnosis and prognosis. Rapid detection of genetic alterations is also significant for choosing appropriate treatment and developing targeted therapies for melanoma. This review will delve into the understanding of various mutations and the implications they may pose for clinical decision making.
Collapse
Affiliation(s)
- Kevin Yang
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (K.Y.); (A.S.O.)
| | - Allen S.W. Oak
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (K.Y.); (A.S.O.)
| | - Radomir M. Slominski
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Anna A. Brożyna
- Department of Human Biology, Institute of Biology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100 Toruń, Poland;
| | - Andrzej T. Slominski
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (K.Y.); (A.S.O.)
- Comprehensive Cancer Center, Cancer Chemoprevention Program, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Veteran Administration Medical Center, Birmingham, AL 35294, USA
- Correspondence:
| |
Collapse
|
34
|
Toussi A, Mans N, Welborn J, Kiuru M. Germline mutations predisposing to melanoma. J Cutan Pathol 2020; 47:606-616. [PMID: 32249949 DOI: 10.1111/cup.13689] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/03/2020] [Accepted: 03/06/2020] [Indexed: 12/11/2022]
Abstract
Nearly 15% of melanomas occur in patients with a family history and a subset of these patients have a germline mutation in a melanoma predisposing gene. CDKN2A mutations are responsible for the majority of hereditary melanoma, but many other susceptibility genes have been discovered in recent years, including CDK4, TERT, ACD, TERF2IP, POT1, MITF, MC1R, and BAP1. Additionally, melanoma risk is increased in mixed cancer syndromes caused by mutations in PTEN, BRCA2, BRCA1, RB1, and TP53. While early onset, multiple tumors, and family cancer history remain the most valuable clinical clues for hereditary melanoma, characteristic epithelioid cytology of melanocytic tumors may suggest an underlying BAP1 mutation. Herein, we review the clinical and histopathologic characteristics of melanocytic tumors associated with these germline mutations and discuss the role of genetic counseling.
Collapse
Affiliation(s)
- Atrin Toussi
- Department of Dermatology, University of California, Davis, Sacramento, California, USA
| | - Nicole Mans
- Hereditary Cancer Program, Comprehensive Cancer Center, University of California, Davis, Sacramento, California, USA
| | - Jeanna Welborn
- Hereditary Cancer Program, Comprehensive Cancer Center, University of California, Davis, Sacramento, California, USA
| | - Maija Kiuru
- Department of Dermatology, University of California, Davis, Sacramento, California, USA.,Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, California, USA
| |
Collapse
|
35
|
Therapeutic targeting of protein S-acylation for the treatment of disease. Biochem Soc Trans 2019; 48:281-290. [DOI: 10.1042/bst20190707] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/04/2019] [Accepted: 12/09/2019] [Indexed: 12/13/2022]
Abstract
The post-translational modification protein S-acylation (commonly known as palmitoylation) plays a critical role in regulating a wide range of biological processes including cell growth, cardiac contractility, synaptic plasticity, endocytosis, vesicle trafficking, membrane transport and biased-receptor signalling. As a consequence, zDHHC-protein acyl transferases (zDHHC-PATs), enzymes that catalyse the addition of fatty acid groups to specific cysteine residues on target proteins, and acyl proteins thioesterases, proteins that hydrolyse thioester linkages, are important pharmaceutical targets. At present, no therapeutic drugs have been developed that act by changing the palmitoylation status of specific target proteins. Here, we consider the role that palmitoylation plays in the development of diseases such as cancer and detail possible strategies for selectively manipulating the palmitoylation status of specific target proteins, a necessary first step towards developing clinically useful molecules for the treatment of disease.
Collapse
|
36
|
Ozola A, Ruklisa D, Pjanova D. The complementary effect of rs1042522 in TP53 and rs1805007 in MC1R is associated with an elevated risk of cutaneous melanoma in Latvian population. Oncol Lett 2019; 18:5225-5234. [PMID: 31612033 PMCID: PMC6781780 DOI: 10.3892/ol.2019.10906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 08/30/2019] [Indexed: 11/06/2022] Open
Abstract
Genetic factors serve important roles in melanoma susceptibility. Although much genetic variation has been associated with cutaneous melanoma (CM), little is known about the interactions between genetic variants. The current study investigated the joint effect of rs1042522 in the tumour protein 53 (TP53) gene, rs2279744 in the murine double minute-2 (MDM2) gene and several single nucleotide polymorphisms (SNPs) in the melanocortin 1 receptor (MC1R) gene. All of these genes are interconnected in a single signalling pathway that regulates pigmentation. The current study included 479 individuals, of which, 255 were patients with CM and 224 were controls from the Latvian population. Multifaceted analyses of potential interactions between SNPs were performed, whilst taking into account the pigmentation phenotypes of individuals and tumour characteristics (Breslow thickness and ulceration). Univariate analyses revealed a borderline significant association between rs1042522 in the TP53 gene and CM risk. The results also confirmed a known association with rs1805007 in the MC1R gene. The rs1042522 was also selected as a CM risk factor in multivariate models, suggesting an effect that is independent from and complementary to that of rs1805007. The results indicated that these SNPs need to be taken into account when determining melanoma risk. A strong association between CM and red hair was identified for rs1805007, and rs1805008 in the MC1R gene was mainly associated with red hair. An association was also determined between rs2279744 in the MDM2 gene and brown eye colour. No convincing associations were identified between the analysed SNPs and Breslow thickness of tumours or ulcerations.
Collapse
Affiliation(s)
- Aija Ozola
- Latvian Biomedical Research and Study Centre, Riga LV-1067, Latvia
| | - Dace Ruklisa
- Newnham College, University of Cambridge, Cambridge CB3 9DF, United Kingdom
| | - Dace Pjanova
- Latvian Biomedical Research and Study Centre, Riga LV-1067, Latvia
| |
Collapse
|
37
|
Swope VB, Starner RJ, Rauck C, Abdel-Malek ZA. Endothelin-1 and α-melanocortin have redundant effects on global genome repair in UV-irradiated human melanocytes despite distinct signaling pathways. Pigment Cell Melanoma Res 2019; 33:293-304. [PMID: 31505093 DOI: 10.1111/pcmr.12823] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 08/19/2019] [Accepted: 08/27/2019] [Indexed: 12/26/2022]
Abstract
Human melanocyte homeostasis is sustained by paracrine factors that reduce the genotoxic effects of ultraviolet radiation (UV), the major etiological factor for melanoma. The keratinocyte-derived endothelin-1 (End-1) and α-melanocyte-stimulating hormone (α-MSH) regulate human melanocyte function, proliferation and survival, and enhance repair of UV-induced DNA photoproducts by binding to the Gq - and Gi -protein-coupled endothelin B receptor (EDNRB), and the Gs -protein-coupled melanocortin 1 receptor (MC1R), respectively. We hereby report that End-1 and α-MSH regulate common effectors of the DNA damage response to UV, despite distinct signaling pathways. Both factors activate the two DNA damage sensors ataxia telangiectasia and Rad3-related and ataxia telangiectasia mutated, enhance DNA damage recognition by reducing soluble nuclear and chromatin-bound DNA damage binding protein 2, and increase total and chromatin-bound xeroderma pigmentosum (XP) C. Additionally, α-MSH and End-1 increase total levels and chromatin localization of the damage verification protein XPA, and the levels of γH2AX, which facilitates recruitment of DNA repair proteins to DNA lesions. Activation of EDNRB compensates for MC1R loss of function, thereby reducing the risk of malignant transformation of these vulnerable melanocytes. Therefore, MC1R and EDNRB signaling pathways represent redundant mechanisms that inhibit the genotoxic effects of UV and melanomagenesis.
Collapse
Affiliation(s)
- Viki B Swope
- Department of Dermatology, University of Cincinnati, Cincinnati, OH, USA
| | - Renny J Starner
- Department of Dermatology, University of Cincinnati, Cincinnati, OH, USA
| | - Corinne Rauck
- Department of Dermatology, University of Cincinnati, Cincinnati, OH, USA
| | | |
Collapse
|
38
|
Diagnostic Performance of a Support Vector Machine for Dermatofluoroscopic Melanoma Recognition: The Results of the Retrospective Clinical Study on 214 Pigmented Skin Lesions. Diagnostics (Basel) 2019; 9:diagnostics9030103. [PMID: 31450697 PMCID: PMC6787620 DOI: 10.3390/diagnostics9030103] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 08/20/2019] [Accepted: 08/22/2019] [Indexed: 11/17/2022] Open
Abstract
The need for diagnosing malignant melanoma in its earliest stages results in an increasing number of unnecessary excisions. Objective criteria beyond the visual inspection are needed to distinguish between benign and malignant melanocytic tumors in vivo. Fluorescence spectra collected during the prospective, multicenter observational study (“FLIMMA”) were retrospectively analyzed by the newly developed machine learning algorithm. The formalin-fixed paraffin-embedded (FFPE) tissue samples of 214 pigmented skin lesions (PSLs) from 144 patients were examined by two independent pathologists in addition to the first diagnosis from the FLIMMA study, resulting in three histopathological results per sample. The support vector machine classifier was trained on 17,918 fluorescence spectra from 49 lesions labeled as malignant (1) and benign (0) by three histopathologists. A scoring system that scales linearly with the number of the “malignant spectra” was designed to classify the lesion as malignant melanoma (score > 28) or non-melanoma (score ≤ 28). Finally, the scoring algorithm was validated on 165 lesions to ensure model prediction power and to estimate the diagnostic accuracy of dermatofluoroscopy in melanoma detection. The scoring algorithm revealed a sensitivity of 91.7% and a specificity of 83.0% in diagnosing malignant melanoma. Using additionally the image segmentation for normalization of lesions’ region of interest, a further improvement of sensitivity of 95.8% was achieved, with a corresponding specificity of 80.9%.
Collapse
|
39
|
Davis LE, Shalin SC, Tackett AJ. Current state of melanoma diagnosis and treatment. Cancer Biol Ther 2019; 20:1366-1379. [PMID: 31366280 PMCID: PMC6804807 DOI: 10.1080/15384047.2019.1640032] [Citation(s) in RCA: 465] [Impact Index Per Article: 93.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 05/23/2019] [Accepted: 06/23/2019] [Indexed: 12/13/2022] Open
Abstract
Melanoma is the deadliest form of skin cancer. In the early stages, melanoma can be treated successfully with surgery alone and survival rates are high, but after metastasis survival rates drop significantly. Therefore, early and correct diagnosis is key for ensuring patients have the best possible prognosis. Melanoma misdiagnosis accounts for more pathology and dermatology malpractice claims than any cancer other than breast cancer, as an early misdiagnosis can significantly reduce a patient's chances of survival. As far as treatment for metastatic melanoma goes, there have been several new drugs developed over the last 10 years that have greatly improved the prognosis of patients with metastatic melanoma, however, a majority of patients do not show a lasting response to these treatments. Thus, new biomarkers and drug targets are needed to improve the accuracy of melanoma diagnosis and treatment. This article will discuss the major advancements of melanoma diagnosis and treatment from antiquity to the present day.
Collapse
Affiliation(s)
- Lauren E. Davis
- University of Arkansas for Medical Sciences, Department of Biochemistry and Molecular Biology, Little Rock, AR, USA
| | - Sara C. Shalin
- University of Arkansas for Medical Sciences, Department of Pathology, Little Rock, AR, USA
| | - Alan J. Tackett
- University of Arkansas for Medical Sciences, Department of Biochemistry and Molecular Biology, Little Rock, AR, USA
| |
Collapse
|
40
|
Cust AE, Mishra K, Berwick M. Melanoma - role of the environment and genetics. Photochem Photobiol Sci 2018; 17:1853-1860. [PMID: 30113042 DOI: 10.1039/c7pp00411g] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Melanoma rates have increased in populations that are mainly European. The main etiologic factor is ultraviolet radiation, from the sun as well as artificial tanning devices. Host factors such as skin color, number of nevi, hair and eye color and tanning ability are critical factors in modifying an individual's response to the sun. Genetic factors interact with host factors and environmental factors to increase risk. This review summarizes our current knowledge of environment and genetics on melanoma risk and on gene-environment interaction.
Collapse
Affiliation(s)
- Anne E Cust
- Cancer Epidemiology and Prevention Research, Sydney School of Public Health, The University of Sydney, Australia
| | | | | |
Collapse
|
41
|
Ozola A, Ruklisa D, Pjanova D. Association of the 16q24.3 region gene variants rs1805007 and rs4785763 with heightened risk of melanoma in Latvian population. Meta Gene 2018. [DOI: 10.1016/j.mgene.2018.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
42
|
Karagianni F, Njauw CN, Kypreou KP, Stergiopoulou A, Plaka M, Polydorou D, Chasapi V, Pappas L, Stratigos IA, Champsas G, Panagiotou P, Gogas H, Evangelou E, Tsao H, Stratigos AJ, Stefanaki I. CDKN2A/CDK4 Status in Greek Patients with Familial Melanoma and Association with Clinico-epidemiological Parameters. Acta Derm Venereol 2018; 98:862-866. [PMID: 29774366 PMCID: PMC6572781 DOI: 10.2340/00015555-2969] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Approximately 5–10% of melanoma cases occur in a familial context. CDKN2A/CDK4 were the first high- penetrance melanoma genes identified. The aims of this study were to evaluate CDKN2A/CDK4 variants in Greek familial melanoma patients and to correlate the mutational status with specific clinico-epidemiological characteristics. A cross-sectional study was conducted by genotyping CDKN2A/CDK4 variants and selected MC1R polymorphisms in 52 melanoma-prone families. Descriptive statistics were calculated and comparisons were made using the X2 test, Fisher’s exact test and Student’s t-test for statistical analysis, as appropriate. CDKN2A variants were detected in 46.2% of melanoma-prone families, while a CDK4 variant was found in only one family. This study confirmed that, in the Greek population, the age at melanoma diagnosis was lower in patients carrying a variant in CDKN2A compared with wild-type patients. No statistically significant associations were found between CDKN2A mutational status and MC1R polymorphisms.
Collapse
Affiliation(s)
- Fani Karagianni
- 1st Department of Dermatology, Andreas Sygros Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Swope VB, Abdel-Malek ZA. MC1R: Front and Center in the Bright Side of Dark Eumelanin and DNA Repair. Int J Mol Sci 2018; 19:E2667. [PMID: 30205559 PMCID: PMC6163888 DOI: 10.3390/ijms19092667] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 08/31/2018] [Accepted: 09/03/2018] [Indexed: 12/17/2022] Open
Abstract
Melanin, the pigment produced by specialized cells, melanocytes, is responsible for skin and hair color. Skin pigmentation is an important protective mechanism against the DNA damaging and mutagenic effects of solar ultraviolet radiation (UV). It is acknowledged that exposure to UV is the main etiological environmental factor for all forms of skin cancer, including melanoma. DNA repair capacity is another major factor that determines the risk for skin cancer. Human melanocytes synthesize eumelanin, the dark brown form of melanin, as well as pheomelanin, which is reddish-yellow in color. The relative rates of eumelanin and pheomelanin synthesis by melanocytes determine skin color and the sensitivity of skin to the drastic effects of solar UV. Understanding the complex regulation of melanocyte function and how it responds to solar UV has a huge impact on developing novel photoprotective strategies to prevent skin cancer, particularly melanoma, the most fatal form, which originates from melanocytes. This review provides an overview of the known differences in the photoprotective effects of eumelanin versus pheomelanin, how these two forms of melanin are regulated genetically and biochemically, and their impact on the DNA damaging effects of UV exposure. Additionally, this review briefly discusses the role of paracrine factors, focusing on α-melanocortin (α-melanocyte stimulating hormone; α-MSH), in regulating melanogenesis and the response of melanocytes to UV, and describes a chemoprevention strategy based on targeting the melanocortin 1 receptor (MC1R) by analogs of its physiological agonist α-MSH.
Collapse
Affiliation(s)
- Viki B Swope
- Department of Dermatology, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA.
| | - Zalfa A Abdel-Malek
- Department of Dermatology, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA.
| |
Collapse
|
44
|
Hernando B, Swope VB, Guard S, Starner RJ, Choi K, Anwar A, Cassidy P, Leachman S, Kadekaro AL, Bennett DC, Abdel-Malek ZA. In vitro behavior and UV response of melanocytes derived from carriers of CDKN2A mutations and MC1R variants. Pigment Cell Melanoma Res 2018; 32:259-268. [PMID: 30117292 DOI: 10.1111/pcmr.12732] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/03/2018] [Accepted: 08/09/2018] [Indexed: 12/19/2022]
Abstract
Coinheritance of germline mutation in cyclin-dependent kinase inhibitor 2A (CDKN2A) and loss-of-function (LOF) melanocortin 1 receptor (MC1R) variants is clinically associated with exaggerated risk for melanoma. To understand the combined impact of these mutations, we established and tested primary human melanocyte cultures from different CDKN2A mutation carriers, expressing either wild-type MC1R or MC1RLOF variant(s). These cultures expressed the CDKN2A product p16 (INK4A) and functional MC1R. Except for 32ins24 mutant melanocytes, the remaining cultures showed no detectable aberrations in proliferation or capacity for replicative senescence. Additionally, the latter cultures responded normally to ultraviolet radiation (UV) by cell cycle arrest, JNK, p38, and p53 activation, hydrogen peroxide generation, and repair of DNA photoproducts. We propose that malignant transformation of melanocytes expressing CDKN2A mutation and MC1RLOF allele(s) requires acquisition of somatic mutations facilitated by MC1R genotype or aberrant microenvironment due to CDKN2A mutation in keratinocytes and fibroblasts.
Collapse
Affiliation(s)
- Barbara Hernando
- Department of Medicine, Jaume I University of Castellon, Castellon, Spain
| | - Viki B Swope
- Department of Dermatology, University of Cincinnati, Cincinnati, Ohio
| | - Steven Guard
- Department of Dermatology, University of Cincinnati, Cincinnati, Ohio
| | - Renny J Starner
- Department of Dermatology, University of Cincinnati, Cincinnati, Ohio
| | - Kevin Choi
- Department of Dermatology, University of Cincinnati, Cincinnati, Ohio
| | - Ayesha Anwar
- Department of Dermatology, University of Cincinnati, Cincinnati, Ohio
| | - Pamela Cassidy
- Department of Dermatology, Oregon Health and Sciences University, Portland, Oregon
| | - Sancy Leachman
- Department of Dermatology, Oregon Health and Sciences University, Portland, Oregon
| | | | - Dorothy C Bennett
- Molecular & Clinical Sciences Research Institute, St George's, University of London, London, UK
| | | |
Collapse
|
45
|
Nguyen NT, Fisher DE. MITF and UV responses in skin: From pigmentation to addiction. Pigment Cell Melanoma Res 2018; 32:224-236. [PMID: 30019545 DOI: 10.1111/pcmr.12726] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 06/21/2018] [Accepted: 06/25/2018] [Indexed: 12/15/2022]
Abstract
Ultraviolet radiation (UVR) has numerous effects on skin, including DNA damage, tanning, vitamin D synthesis, carcinogenesis, and immunomodulation. Keratinocytes containing damaged DNA secrete both α-melanocyte-stimulating hormone (α-MSH), which stimulates pigment production by melanocytes, and the opioid β-endorphin, which can trigger addiction-like responses to UVR. The pigmentation (tanning) response is an adaptation that provides some delayed protection against further DNA damage and carcinogenesis, while the opioid response may be an evolutionary adaptation for promoting sun-seeking behavior to prevent vitamin D deficiency. Here, we review the pigmentation response to UVR, driven by melanocytic microphthalmia-associated transcription factor (MITF), and evidence for UVR-induced melanomagenesis and addiction. We also discuss potential applications of a novel approach to generate protective pigmentation in the absence of UVR (sunless tanning) using a topical small-molecule inhibitor of the salt-inducible kinase (SIK) family.
Collapse
Affiliation(s)
- Nhu T Nguyen
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, and Harvard Medical School, Boston, Massachusetts
| | - David E Fisher
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
46
|
Tagliabue E, Gandini S, Bellocco R, Maisonneuve P, Newton-Bishop J, Polsky D, Lazovich D, Kanetsky PA, Ghiorzo P, Gruis NA, Landi MT, Menin C, Fargnoli MC, García-Borrón JC, Han J, Little J, Sera F, Raimondi S. MC1R variants as melanoma risk factors independent of at-risk phenotypic characteristics: a pooled analysis from the M-SKIP project. Cancer Manag Res 2018; 10:1143-1154. [PMID: 29795986 PMCID: PMC5958947 DOI: 10.2147/cmar.s155283] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
PURPOSE Melanoma represents an important public health problem, due to its high case-fatality rate. Identification of individuals at high risk would be of major interest to improve early diagnosis and ultimately survival. The aim of this study was to evaluate whether MC1R variants predicted melanoma risk independently of at-risk phenotypic characteristics. MATERIALS AND METHODS Data were collected within an international collaboration - the M-SKIP project. The present pooled analysis included data on 3,830 single, primary, sporadic, cutaneous melanoma cases and 2,619 controls from seven previously published case-control studies. All the studies had information on MC1R gene variants by sequencing analysis and on hair color, skin phototype, and freckles, ie, the phenotypic characteristics used to define the red hair phenotype. RESULTS The presence of any MC1R variant was associated with melanoma risk independently of phenotypic characteristics (OR 1.60; 95% CI 1.36-1.88). Inclusion of MC1R variants in a risk prediction model increased melanoma predictive accuracy (area under the receiver-operating characteristic curve) by 0.7% over a base clinical model (P=0.002), and 24% of participants were better assessed (net reclassification index 95% CI 20%-30%). Subgroup analysis suggested a possibly stronger role of MC1R in melanoma prediction for participants without the red hair phenotype (net reclassification index: 28%) compared to paler skinned participants (15%). CONCLUSION The authors suggest that measuring the MC1R genotype might result in a benefit for melanoma prediction. The results could be a valid starting point to guide the development of scientific protocols assessing melanoma risk prediction tools incorporating the MC1R genotype.
Collapse
Affiliation(s)
- Elena Tagliabue
- Clinical Trial Center, Scientific Directorate, Fondazione IRCCS Istituto Nazionale dei Tumori
| | - Sara Gandini
- Division of Epidemiology and Biostatistics, European Institute of Oncology, Milan, Italy
| | - Rino Bellocco
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Statistics and Quantitative Methods, University of Milano-Bicocca, Milan, Italy
| | - Patrick Maisonneuve
- Division of Epidemiology and Biostatistics, European Institute of Oncology, Milan, Italy
| | - Julia Newton-Bishop
- Section of Epidemiology and Biostatistics, Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - David Polsky
- Ronald O. Perelman Department of Dermatology, New York University School of Medicine, NYU Langone Medical Center, New York, NY
| | - DeAnn Lazovich
- Division of Epidemiology and Community Health, University of Minnesota, MN
| | - Peter A Kanetsky
- Department of Cancer Epidemiology, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Paola Ghiorzo
- Department of Internal Medicine and Medical Specialties, University of Genoa
- IRCCS AOU San Martino-IST, Genoa, Italy
| | - Nelleke A Gruis
- Department of Dermatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Chiara Menin
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS, Padua
| | | | - Jose Carlos García-Borrón
- Department of Biochemistry, Molecular Biology, and Immunology, University of Murcia
- IMIB-Arrixaca, Murcia, Spain
| | - Jiali Han
- Department of Epidemiology, Richard M Fairbanks School of Public Health, Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, IN, USA
| | - Julian Little
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
| | - Francesco Sera
- Department of Social and Environmental Health Research, London School of Hygiene and Tropical Medicine, London, UK
| | - Sara Raimondi
- Division of Epidemiology and Biostatistics, European Institute of Oncology, Milan, Italy
| |
Collapse
|
47
|
de Assis LVM, Moraes MN, Magalhães-Marques KK, Castrucci AMDL. Melanopsin and rhodopsin mediate UVA-induced immediate pigment darkening: Unravelling the photosensitive system of the skin. Eur J Cell Biol 2018; 97:150-162. [PMID: 29395480 DOI: 10.1016/j.ejcb.2018.01.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/12/2018] [Accepted: 01/19/2018] [Indexed: 11/25/2022] Open
Abstract
The mammalian skin has a photosensitive system comprised by several opsins, including rhodopsin (OPN2) and melanopsin (OPN4). Recently, our group showed that UVA (4.4 kJ/m2) leads to immediate pigment darkening (IPD) in murine normal and malignant melanocytes. We show the role of OPN2 and OPN4 as UVA sensors: UVA-induced IPD was fully abolished when OPN4 was pharmacologically inhibited by AA9253 or when OPN2 and OPN4 were knocked down by siRNA in both cell lines. Our data, however, demonstrate that phospholipase C/protein kinase C pathway, a classical OPN4 pathway, is not involved in UVA-induced IPD in either cell line. Nonetheless, in both cell types we have shown that: a) intracellular calcium signal is necessary for UVA-induced IPD; b) the involvement of CaMK II, whose inhibition, abolished the UVA-induced IPD; c) the role of CAMK II/NOS/sGC/cGMP pathway in the process since inhibition of either NOS or sGC abolished the UVA-induced IPD. Taken altogether, we show that OPN2 and OPN4 participate in IPD induced by UVA in murine normal and malignant melanocytes through a conserved common pathway. Interestingly, upon knockdown of OPN2 or OPN4, the UVA-driven IPD is completely lost, which suggests that both opsins are required and cooperatively signal in murine both cell lines. The participation of OPN2 and OPN4 system in UVA radiation-induced response, if proven to take place in human skin, may represent an interesting pharmacological target for the treatment of depigmentary disorders and skin-related cancer.
Collapse
Affiliation(s)
- Leonardo Vinícius Monteiro de Assis
- Laboratory of Comparative Physiology of Pigmentation, Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - Maria Nathalia Moraes
- Laboratory of Comparative Physiology of Pigmentation, Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - Keila Karoline Magalhães-Marques
- Laboratory of Comparative Physiology of Pigmentation, Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - Ana Maria de Lauro Castrucci
- Laboratory of Comparative Physiology of Pigmentation, Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil; Department of Biology, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
48
|
Bruno W, Martinuzzi C, Dalmasso B, Andreotti V, Pastorino L, Cabiddu F, Gualco M, Spagnolo F, Ballestrero A, Queirolo P, Grillo F, Mastracci L, Ghiorzo P. Combining molecular and immunohistochemical analyses of key drivers in primary melanomas: interplay between germline and somatic variations. Oncotarget 2018; 9:5691-5702. [PMID: 29464027 PMCID: PMC5814167 DOI: 10.18632/oncotarget.23204] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 11/15/2017] [Indexed: 01/15/2023] Open
Abstract
Due to the high mutational somatic burden of Cutaneous Malignant Melanoma (CMM) a thorough profiling of the driver mutations and their interplay is necessary to explain the timing of tumorigenesis or for the identification of actionable genetic events. The aim of this study was to establish the mutation rate of some of the key drivers in melanoma tumorigenesis combining molecular analyses and/or immunohistochemistry in 93 primary CMMs from an Italian cohort also characterized for germline status, and to investigate an interplay between germline and somatic variants. BRAF mutations were present in 68% of cases, while CDKN2A germline mutations were found in 16 % and p16 loss in tissue was found in 63%. TERT promoter somatic mutations were detected in 38% of cases while the TERT -245T>C polymorphism was found in 51% of cases. NRAS mutations were found in 39% of BRAF negative or undetermined cases. NF1 was expressed in all cases analysed. MC1R variations were both considered as a dichotomous variable or scored. While a positive, although not significant association between CDKN2A germline mutations, but not MC1R variants, and BRAF somatic mutation was found, we did not observe other associations between germline and somatic events. A yet undescribed inverse correlation between TERT -245T>C polymorphism and the presence of BRAF mutation was found. It is possible to hypothesize that -245T>C polymorphism could be included in those genotypes which may influence the occurrence of BRAF mutations. Further studies are needed to investigate the role of -245T>C polymorphism as a germline predictor of BRAF somatic mutation status.
Collapse
Affiliation(s)
- William Bruno
- Department of Internal Medicine and Medical Specialties (DiMI), University of Genoa and Ospedale Policlinico San Martino, Genoa, Italy
| | - Claudia Martinuzzi
- Department of Internal Medicine and Medical Specialties (DiMI), University of Genoa and Ospedale Policlinico San Martino, Genoa, Italy
| | - Bruna Dalmasso
- Department of Internal Medicine and Medical Specialties (DiMI), University of Genoa and Ospedale Policlinico San Martino, Genoa, Italy
| | - Virginia Andreotti
- Department of Internal Medicine and Medical Specialties (DiMI), University of Genoa and Ospedale Policlinico San Martino, Genoa, Italy
| | - Lorenza Pastorino
- Department of Internal Medicine and Medical Specialties (DiMI), University of Genoa and Ospedale Policlinico San Martino, Genoa, Italy
| | | | - Marina Gualco
- Pathology Unit, Ospedale Policlinico San Martino, Genoa, Italy
| | - Francesco Spagnolo
- Department of Medical Oncology, Ospedale Policlinico San Martino, Genoa, Italy
| | - Alberto Ballestrero
- Department of Internal Medicine and Medical Specialties (DiMI), University of Genoa and Ospedale Policlinico San Martino, Genoa, Italy
| | - Paola Queirolo
- Department of Medical Oncology, Ospedale Policlinico San Martino, Genoa, Italy
| | - Federica Grillo
- Department of Surgical and Diagnostic Sciences, Pathology Unit, University of Genoa and Ospedale Policlinico San Martino, Genoa, Italy
| | - Luca Mastracci
- Department of Surgical and Diagnostic Sciences, Pathology Unit, University of Genoa and Ospedale Policlinico San Martino, Genoa, Italy
| | - Paola Ghiorzo
- Department of Internal Medicine and Medical Specialties (DiMI), University of Genoa and Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
49
|
Li X, Lee KJ, Duffy DL, Xu D, Basude MER, Hu Y, Zhang H, Jagirdar K, Soyer HP, Dong H, Sturm RA. Acquired melanocytic naevus phenotypes and MC1R genotypes in Han Chinese: a cross-sectional study. PeerJ 2018; 5:e4168. [PMID: 29340229 PMCID: PMC5741976 DOI: 10.7717/peerj.4168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 11/24/2017] [Indexed: 12/02/2022] Open
Abstract
Background Early detection and treatment are the most important elements in reducing the incidence of melanoma deaths. Acquired melanocytic naevi (AMN) are well-known precursors of melanoma but most of our knowledge on the clinico-dermoscopic phenotypes of AMN is based on studies in European-background populations, particularly American and Australian populations. There has been little research in Chinese Han populations on clinico-dermoscopic variability of naevi or how naevi are affected by melanoma-linked variants of the melanocortin 1 receptor (MC1R) gene. Methods Clinical and dermoscopic features of 448 AMN in 115 patients from the Han ethnic group in mainland China were described. Germline polymorphisms in MC1R were determined for 98 of these patients. Results AMN were predominantly found on the head and neck. Dermoscopic patterns observed were nonspecific, reticular, globular, and parallel furrow, with most AMN having a nonspecific pattern. There were no associations between MC1R polymorphisms and clinical or dermoscopic features of AMN. Discussion Our results provide evidence that AMN in the Han population in China have similar dermoscopic patterns to those in European populations, but are present in much lower numbers. As there were no associations between clinical or dermoscopic features of AMN and MC1R polymorphisms, further studies should focus on candidate gene associations with AMN features and the risk of melanoma, with larger sample sizes and comparisons to AMN in other populations.
Collapse
Affiliation(s)
- Xiaohong Li
- Department of Dermatology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Key-Disciplines Laboratory, Clinical-Medicine Henan, Zhengzhou, Henan, China
| | - Katie J Lee
- Dermatology Research Centre, The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
| | - David L Duffy
- Dermatology Research Centre, The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia.,QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Dandan Xu
- Department of Dermatology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Key-Disciplines Laboratory, Clinical-Medicine Henan, Zhengzhou, Henan, China.,Department of Dermatology, Huaihe Hospital of Henan University, Kaifeng, Henan, China
| | - Madhur Eshwar Rao Basude
- Department of Dermatology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Key-Disciplines Laboratory, Clinical-Medicine Henan, Zhengzhou, Henan, China.,Department of Dermatology, Vinayaka Missions Medical College and Hospital, Karaikal, Puducherry, India
| | - Ying Hu
- Department of Dermatology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Department of Dermatology, Central Hospital of Zhengzhou, Zhengzhou, Henan, China
| | - Hang Zhang
- Department of Dermatology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Kasturee Jagirdar
- Dermatology Research Centre, The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
| | - H Peter Soyer
- Dermatology Research Centre, The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
| | - Huiting Dong
- Department of Dermatology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Key-Disciplines Laboratory, Clinical-Medicine Henan, Zhengzhou, Henan, China
| | - Richard A Sturm
- Dermatology Research Centre, The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
| |
Collapse
|
50
|
Kansal RG, McCravy MS, Basham JH, Earl JA, McMurray SL, Starner CJ, Whitt MA, Albritton LM. Inhibition of melanocortin 1 receptor slows melanoma growth, reduces tumor heterogeneity and increases survival. Oncotarget 2018; 7:26331-45. [PMID: 27028866 PMCID: PMC5041983 DOI: 10.18632/oncotarget.8372] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/14/2016] [Indexed: 11/25/2022] Open
Abstract
Melanoma risk is increased in patients with mutations of melanocortin 1 receptor (MC1R) yet the basis for the increased risk remains unknown. Here we report in vivo evidence supporting a critical role for MC1R in regulating melanoma tumor growth and determining overall survival time. Inhibition of MC1R by its physiologically relevant competitive inhibitor, agouti signaling protein (ASIP), reduced melanin synthesis and morphological heterogeneity in murine B16-F10 melanoma cells. In the lungs of syngeneic C57BL/6 mice, mCherry-marked, ASIP-secreting lung tumors inhibited MC1R on neighboring tumors lacking ASIP in a dose dependent manner as evidenced by a proportional loss of pigment in tumors from mice injected with 1:1, 3:1 and 4:1 mixtures of parental B16-F10 to ASIP-expressing tumor cells. ASIP-expressing B16-F10 cells formed poorly pigmented tumors in vivo that correlated with a 20% longer median survival than those bearing parental B16-F10 tumors (p=0.0005). Mice injected with 1:1 mixtures also showed survival benefit (p=0.0054), whereas injection of a 4:1 mixture showed no significant difference in survival. The longer survival time of mice bearing ASIP-expressing tumors correlated with a significantly slower growth rate than parental B16-F10 tumors as judged by quantification of numbers of tumors and total tumor load (p=0.0325), as well as a more homogeneous size and morphology of ASIP-expressing lung tumors. We conclude that MC1R plays an important role in regulating melanoma growth and morphology. Persistent inhibition of MC1R provided a significant survival advantage resulting in part from slower tumor growth, establishing MC1R as a compelling new molecular target for metastatic melanoma.
Collapse
Affiliation(s)
- Rita G Kansal
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Matthew S McCravy
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jacob H Basham
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Joshua A Earl
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Stacy L McMurray
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Chelsey J Starner
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Michael A Whitt
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Lorraine M Albritton
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|