Cargando…

Ablation of epidermal RXRα in cooperation with activated CDK4 and oncogenic NRAS generates spontaneous and acute neonatal UVB induced malignant metastatic melanomas

BACKGROUND: Understanding the underlying molecular mechanisms involved in the formation of cutaneous malignant melanoma is critical for improved diagnosis and treatment. Keratinocytic nuclear receptor Retinoid X Receptor α (RXRα) has a protective role against melanomagenesis and is involved in the r...

Descripción completa

Detalles Bibliográficos
Autores principales: Chagani, Sharmeen, Wang, Rong, Carpenter, Evan L., Löhr, Christiane V., Ganguli-Indra, Gitali, Indra, Arup K.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2017
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5679438/
https://www.ncbi.nlm.nih.gov/pubmed/29121869
http://dx.doi.org/10.1186/s12885-017-3714-6
_version_ 1783277581267632128
author Chagani, Sharmeen
Wang, Rong
Carpenter, Evan L.
Löhr, Christiane V.
Ganguli-Indra, Gitali
Indra, Arup K.
author_facet Chagani, Sharmeen
Wang, Rong
Carpenter, Evan L.
Löhr, Christiane V.
Ganguli-Indra, Gitali
Indra, Arup K.
author_sort Chagani, Sharmeen
collection PubMed
description BACKGROUND: Understanding the underlying molecular mechanisms involved in the formation of cutaneous malignant melanoma is critical for improved diagnosis and treatment. Keratinocytic nuclear receptor Retinoid X Receptor α (RXRα) has a protective role against melanomagenesis and is involved in the regulation of keratinocyte and melanocyte homeostasis subsequent acute ultraviolet (UV) irradiation. METHODS: We generated a trigenic mouse model system (RXRα (ep−/−)| Tyr-NRAS (Q61K) | CDK4 (R24C/R24C) ) harboring an epidermal knockout of Retinoid X Receptor α (RXRα (ep−/−)), combined with oncogenic NRAS (Q61K) (constitutively active RAS) and activated CDK4 (R24C/R24C) (constitutively active CDK4). Those mice were subjected to a single neonatal dose of UVB treatment and the role of RXR α was evaluated by characterizing the molecular and cellular changes that took place in the untreated and UVB treated trigenic RXRα (ep−/−) mice compared to the control mice with functional RXRα. RESULTS: Here we report that the trigenic mice develops spontaneous melanoma and exposure to a single neonatal UVB treatment reduces the tumor latency in those mice compared to control mice with functional RXRα. Melanomas from the trigenic RXRα (ep−/−) mice are substantial in size, show increased proliferation, exhibit increased expression of malignant melanoma markers and exhibit enhanced vascularization. Altered expression of several biomarkers including increased expression of activated AKT, p21 and cyclin D1 and reduced expression of pro-apoptotic marker BAX was observed in the tumor adjacent normal (TAN) skin of acute ultraviolet B treated trigenic RXRα (ep−/−) mice. Interestingly, we observed a significant increase in p21 and Cyclin D1 in the TAN skin of un-irradiated trigenic RXRα (ep−/−) mice, suggesting that those changes might be consequences of loss of functional RXRα in the melanoma microenvironment. Loss of RXRα in the epidermal keratinocytes in combination with oncogenic NRAS (Q61K) and CDK4 (R24C/R24C) mutations in trigenic mice led to significant melanoma invasion into the draining lymph nodes as compared to controls with functional RXRα. CONCLUSIONS: Our study demonstrates the protective role of keratinocytic RxRα in (1) suppressing spontaneous and acute UVB-induced melanoma, and (2) preventing progression of the melanoma to malignancy in the presence of driver mutations like activated CDK4 (R24C/R24C) and oncogenic NRAS (Q61K) . ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1186/s12885-017-3714-6) contains supplementary material, which is available to authorized users.
format Online
Article
Text
id pubmed-5679438
institution National Center for Biotechnology Information
language English
publishDate 2017
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-56794382017-11-17 Ablation of epidermal RXRα in cooperation with activated CDK4 and oncogenic NRAS generates spontaneous and acute neonatal UVB induced malignant metastatic melanomas Chagani, Sharmeen Wang, Rong Carpenter, Evan L. Löhr, Christiane V. Ganguli-Indra, Gitali Indra, Arup K. BMC Cancer Research Article BACKGROUND: Understanding the underlying molecular mechanisms involved in the formation of cutaneous malignant melanoma is critical for improved diagnosis and treatment. Keratinocytic nuclear receptor Retinoid X Receptor α (RXRα) has a protective role against melanomagenesis and is involved in the regulation of keratinocyte and melanocyte homeostasis subsequent acute ultraviolet (UV) irradiation. METHODS: We generated a trigenic mouse model system (RXRα (ep−/−)| Tyr-NRAS (Q61K) | CDK4 (R24C/R24C) ) harboring an epidermal knockout of Retinoid X Receptor α (RXRα (ep−/−)), combined with oncogenic NRAS (Q61K) (constitutively active RAS) and activated CDK4 (R24C/R24C) (constitutively active CDK4). Those mice were subjected to a single neonatal dose of UVB treatment and the role of RXR α was evaluated by characterizing the molecular and cellular changes that took place in the untreated and UVB treated trigenic RXRα (ep−/−) mice compared to the control mice with functional RXRα. RESULTS: Here we report that the trigenic mice develops spontaneous melanoma and exposure to a single neonatal UVB treatment reduces the tumor latency in those mice compared to control mice with functional RXRα. Melanomas from the trigenic RXRα (ep−/−) mice are substantial in size, show increased proliferation, exhibit increased expression of malignant melanoma markers and exhibit enhanced vascularization. Altered expression of several biomarkers including increased expression of activated AKT, p21 and cyclin D1 and reduced expression of pro-apoptotic marker BAX was observed in the tumor adjacent normal (TAN) skin of acute ultraviolet B treated trigenic RXRα (ep−/−) mice. Interestingly, we observed a significant increase in p21 and Cyclin D1 in the TAN skin of un-irradiated trigenic RXRα (ep−/−) mice, suggesting that those changes might be consequences of loss of functional RXRα in the melanoma microenvironment. Loss of RXRα in the epidermal keratinocytes in combination with oncogenic NRAS (Q61K) and CDK4 (R24C/R24C) mutations in trigenic mice led to significant melanoma invasion into the draining lymph nodes as compared to controls with functional RXRα. CONCLUSIONS: Our study demonstrates the protective role of keratinocytic RxRα in (1) suppressing spontaneous and acute UVB-induced melanoma, and (2) preventing progression of the melanoma to malignancy in the presence of driver mutations like activated CDK4 (R24C/R24C) and oncogenic NRAS (Q61K) . ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1186/s12885-017-3714-6) contains supplementary material, which is available to authorized users. BioMed Central 2017-11-09 /pmc/articles/PMC5679438/ /pubmed/29121869 http://dx.doi.org/10.1186/s12885-017-3714-6 Text en © The Author(s). 2017 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research Article
Chagani, Sharmeen
Wang, Rong
Carpenter, Evan L.
Löhr, Christiane V.
Ganguli-Indra, Gitali
Indra, Arup K.
Ablation of epidermal RXRα in cooperation with activated CDK4 and oncogenic NRAS generates spontaneous and acute neonatal UVB induced malignant metastatic melanomas
title Ablation of epidermal RXRα in cooperation with activated CDK4 and oncogenic NRAS generates spontaneous and acute neonatal UVB induced malignant metastatic melanomas
title_full Ablation of epidermal RXRα in cooperation with activated CDK4 and oncogenic NRAS generates spontaneous and acute neonatal UVB induced malignant metastatic melanomas
title_fullStr Ablation of epidermal RXRα in cooperation with activated CDK4 and oncogenic NRAS generates spontaneous and acute neonatal UVB induced malignant metastatic melanomas
title_full_unstemmed Ablation of epidermal RXRα in cooperation with activated CDK4 and oncogenic NRAS generates spontaneous and acute neonatal UVB induced malignant metastatic melanomas
title_short Ablation of epidermal RXRα in cooperation with activated CDK4 and oncogenic NRAS generates spontaneous and acute neonatal UVB induced malignant metastatic melanomas
title_sort ablation of epidermal rxrα in cooperation with activated cdk4 and oncogenic nras generates spontaneous and acute neonatal uvb induced malignant metastatic melanomas
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5679438/
https://www.ncbi.nlm.nih.gov/pubmed/29121869
http://dx.doi.org/10.1186/s12885-017-3714-6
work_keys_str_mv AT chaganisharmeen ablationofepidermalrxraincooperationwithactivatedcdk4andoncogenicnrasgeneratesspontaneousandacuteneonataluvbinducedmalignantmetastaticmelanomas
AT wangrong ablationofepidermalrxraincooperationwithactivatedcdk4andoncogenicnrasgeneratesspontaneousandacuteneonataluvbinducedmalignantmetastaticmelanomas
AT carpenterevanl ablationofepidermalrxraincooperationwithactivatedcdk4andoncogenicnrasgeneratesspontaneousandacuteneonataluvbinducedmalignantmetastaticmelanomas
AT lohrchristianev ablationofepidermalrxraincooperationwithactivatedcdk4andoncogenicnrasgeneratesspontaneousandacuteneonataluvbinducedmalignantmetastaticmelanomas
AT ganguliindragitali ablationofepidermalrxraincooperationwithactivatedcdk4andoncogenicnrasgeneratesspontaneousandacuteneonataluvbinducedmalignantmetastaticmelanomas
AT indraarupk ablationofepidermalrxraincooperationwithactivatedcdk4andoncogenicnrasgeneratesspontaneousandacuteneonataluvbinducedmalignantmetastaticmelanomas