Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 221
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 81(6): 1276-1291.e9, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33539787

RESUMEN

Aberrant cell proliferation is a hallmark of cancer, including glioblastoma (GBM). Here we report that protein arginine methyltransferase (PRMT) 6 activity is required for the proliferation, stem-like properties, and tumorigenicity of glioblastoma stem cells (GSCs), a subpopulation in GBM critical for malignancy. We identified a casein kinase 2 (CK2)-PRMT6-regulator of chromatin condensation 1 (RCC1) signaling axis whose activity is an important contributor to the stem-like properties and tumor biology of GSCs. CK2 phosphorylates and stabilizes PRMT6 through deubiquitylation, which promotes PRMT6 methylation of RCC1, which in turn is required for RCC1 association with chromatin and activation of RAN. Disruption of this pathway results in defects in mitosis. EPZ020411, a specific small-molecule inhibitor for PRMT6, suppresses RCC1 arginine methylation and improves the cytotoxic activity of radiotherapy against GSC brain tumor xenografts. This study identifies a CK2α-PRMT6-RCC1 signaling axis that can be therapeutically targeted in the treatment of GBM.


Asunto(s)
Neoplasias Encefálicas , Carcinogénesis , Proteínas de Ciclo Celular , Glioblastoma , Factores de Intercambio de Guanina Nucleótido , Mitosis/efectos de la radiación , Proteínas de Neoplasias , Proteínas Nucleares , Proteína-Arginina N-Metiltransferasas , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/radioterapia , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinogénesis/efectos de la radiación , Quinasa de la Caseína II/genética , Quinasa de la Caseína II/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Femenino , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/radioterapia , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células HEK293 , Humanos , Masculino , Ratones , Mitosis/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Transducción de Señal/genética , Transducción de Señal/efectos de la radiación , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Nature ; 575(7783): 519-522, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31666702

RESUMEN

Immunosuppression increases the risk of cancers that are associated with viral infection1. In particular, the risk of squamous cell carcinoma of the skin-which has been associated with beta human papillomavirus (ß-HPV) infection-is increased by more than 100-fold in immunosuppressed patients2-4. Previous studies have not established a causative role for HPVs in driving the development of skin cancer. Here we show that T cell immunity against commensal papillomaviruses suppresses skin cancer in immunocompetent hosts, and the loss of this immunity-rather than the oncogenic effect of HPVs-causes the markedly increased risk of skin cancer in immunosuppressed patients. To investigate the effects of papillomavirus on carcinogen-driven skin cancer, we colonized several strains of immunocompetent mice with mouse papillomavirus type 1 (MmuPV1)5. Mice with natural immunity against MmuPV1 after colonization and acquired immunity through the transfer of T cells from immune mice or by MmuPV1 vaccination were protected against skin carcinogenesis induced by chemicals or by ultraviolet radiation in a manner dependent on CD8+ T cells. RNA and DNA in situ hybridization probes for 25 commensal ß-HPVs revealed a significant reduction in viral activity and load in human skin cancer compared with the adjacent healthy skin, suggesting a strong immune selection against virus-positive malignant cells. Consistently, E7 peptides from ß-HPVs activated CD8+ T cells from unaffected human skin. Our findings reveal a beneficial role for commensal viruses and establish a foundation for immune-based approaches that could block the development of skin cancer by boosting immunity against the commensal HPVs present in all of our skin.


Asunto(s)
Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/prevención & control , Papillomaviridae/inmunología , Infecciones por Papillomavirus/inmunología , Infecciones por Papillomavirus/virología , Neoplasias Cutáneas/prevención & control , Neoplasias Cutáneas/virología , Simbiosis , Anciano , Anciano de 80 o más Años , Animales , Linfocitos T CD8-positivos/inmunología , Carcinogénesis/efectos de la radiación , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/virología , Femenino , Humanos , Huésped Inmunocomprometido/inmunología , Masculino , Ratones , Persona de Mediana Edad , Oncogenes , Papillomaviridae/genética , Papillomaviridae/patogenicidad , ARN Viral/análisis , ARN Viral/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Rayos Ultravioleta
3.
Genes Dev ; 31(8): 724-743, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28512236

RESUMEN

Cutaneous melanoma (CM) and uveal melanoma (UM) derive from cutaneous and uveal melanocytes that share the same embryonic origin and display the same cellular function. However, the etiopathogenesis and biological behaviors of these melanomas are very different. CM and UM display distinct landscapes of genetic alterations and show different metastatic routes and tropisms. Hence, therapeutic improvements achieved in the last few years for the treatment of CM have failed to ameliorate the clinical outcomes of patients with UM. The scope of this review is to discuss the differences in tumorigenic processes (etiologic factors and genetic alterations) and tumor biology (gene expression and signaling pathways) between CM and UM. We develop hypotheses to explain these differences, which might provide important clues for research avenues and the identification of actionable vulnerabilities suitable for the development of new therapeutic strategies for metastatic UM.


Asunto(s)
Melanoma/fisiopatología , Neoplasias Cutáneas/fisiopatología , Neoplasias de la Úvea/fisiopatología , Carcinogénesis/genética , Carcinogénesis/patología , Carcinogénesis/efectos de la radiación , Regulación Neoplásica de la Expresión Génica , Humanos , Melanocitos/patología , Melanocitos/fisiología , Melanoma/clasificación , Melanoma/genética , Investigación/tendencias , Factores de Riesgo , Transducción de Señal/genética , Neoplasias Cutáneas/clasificación , Neoplasias Cutáneas/genética , Rayos Ultravioleta , Neoplasias de la Úvea/clasificación , Neoplasias de la Úvea/genética , Melanoma Cutáneo Maligno
4.
J Biol Chem ; 299(7): 104900, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37301510

RESUMEN

Nucleotide excision repair (NER) eliminates highly genotoxic solar UV-induced DNA photoproducts that otherwise stimulate malignant melanoma development. Here, a genome-wide loss-of-function screen, coupling CRISPR/Cas9 technology with a flow cytometry-based DNA repair assay, was used to identify novel genes required for efficient NER in primary human fibroblasts. Interestingly, the screen revealed multiple genes encoding proteins, with no previously known involvement in UV damage repair, that significantly modulate NER uniquely during S phase of the cell cycle. Among these, we further characterized Dyrk1A, a dual specificity kinase that phosphorylates the proto-oncoprotein cyclin D1 on threonine 286 (T286), thereby stimulating its timely cytoplasmic relocalization and proteasomal degradation, which is required for proper regulation of the G1-S phase transition and control of cellular proliferation. We demonstrate that in UV-irradiated HeLa cells, depletion of Dyrk1A leading to overexpression of cyclin D1 causes inhibition of NER uniquely during S phase and reduced cell survival. Consistently, expression/nuclear accumulation of nonphosphorylatable cyclin D1 (T286A) in melanoma cells strongly interferes with S phase NER and enhances cytotoxicity post-UV. Moreover, the negative impact of cyclin D1 (T286A) overexpression on repair is independent of cyclin-dependent kinase activity but requires cyclin D1-dependent upregulation of p21 expression. Our data indicate that inhibition of NER during S phase might represent a previously unappreciated noncanonical mechanism by which oncogenic cyclin D1 fosters melanomagenesis.


Asunto(s)
Ciclina D1 , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Reparación del ADN , Proteínas Serina-Treonina Quinasas , Proteínas Tirosina Quinasas , Humanos , Ciclina D1/genética , Ciclina D1/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Daño del ADN/efectos de la radiación , Células HeLa , Proteínas Tirosina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Fibroblastos/enzimología , Fibroblastos/metabolismo , Fibroblastos/patología , Fibroblastos/efectos de la radiación , Fase S , Fase G1 , Melanoma/genética , Melanoma/patología , Células Cultivadas , Rayos Ultravioleta/efectos adversos , Carcinogénesis/genética , Carcinogénesis/patología , Carcinogénesis/efectos de la radiación , Quinasas DyrK
5.
Photochem Photobiol Sci ; 23(3): 517-526, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38337129

RESUMEN

Squamous cell carcinoma represents the second most common type of keratinocyte carcinoma with ultraviolet radiation (UVR) making up the primary risk factor. Oral photoprotection aims to reduce incidence rates through oral intake of photoprotective compounds. Recently, drug repurposing has gained traction as an interesting source of chemoprevention. Because of their reported photoprotective properties, we investigated the potential of bucillamine, carvedilol, metformin, and phenformin as photoprotective compounds following oral intake in UVR-exposed hairless mice. Tumour development was observed in all groups in response to UVR, with only the positive control (Nicotinamide) demonstrating a reduction in tumour incidence (23.8%). No change in tumour development was observed in the four repurposed drug groups compared to the UV control group, whereas nicotinamide significantly reduced carcinogenesis (P = 0.00012). Metformin treatment significantly reduced UVR-induced erythema (P = 0.012), bucillamine and phenformin increased dorsal pigmentation (P = 0.0013, and P = 0.0005), but no other photoprotective effect was observed across the repurposed groups. This study demonstrates that oral supplementation with bucillamine, carvedilol, metformin, or phenformin does not affect UVR-induced carcinogenesis in hairless mice.


Asunto(s)
Carcinoma de Células Escamosas , Cisteína/análogos & derivados , Neoplasias Cutáneas , Ratones , Animales , Rayos Ultravioleta , Carvedilol/farmacología , Ratones Pelados , Fenformina/farmacología , Carcinoma de Células Escamosas/prevención & control , Carcinoma de Células Escamosas/etiología , Carcinogénesis/efectos de la radiación , Niacinamida/farmacología , Neoplasias Cutáneas/etiología , Neoplasias Cutáneas/prevención & control , Neoplasias Cutáneas/patología , Piel/efectos de la radiación
6.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34504008

RESUMEN

Chronic ultraviolet (UV) radiation exposure is the greatest risk factor for cutaneous squamous cell carcinoma (cSCC) development, and compromised immunity accelerates this risk. Having previously identified that epidermal Langerhans cells (LC) facilitate the expansion of UV-induced mutant keratinocytes (KC), we sought to more fully elucidate the immune pathways critical to cutaneous carcinogenesis and to identify potential targets of intervention. Herein, we reveal that chronic UV induces and LC enhance a local immune shift toward RORγt+ interleukin (IL)-22/IL-17A-producing cells that occurs in the presence or absence of T cells while identifying a distinct RORγt+ Sca-1+ CD103+ ICOS+ CD2+/- CCR6+ intracellular CD3+ cutaneous innate lymphoid cell type-3 (ILC3) population (uvILC3) that is associated with UV-induced mutant KC growth. We further show that mutant KC clone size is markedly reduced in the absence of RORγt+ lymphocytes or IL-22, both observed in association with expanding KC clones, and find that topical application of a RORγ/γt inhibitor during chronic UV exposure reduces local expression of IL-22 and IL-17A while markedly limiting mutant p53 KC clonal expansion. We implicate upstream Toll-like receptor signaling in driving this immune response to chronic UV exposure, as MyD88/Trif double-deficient mice also show substantially reduced p53 island number and size. These data elucidate key immune components of chronic UV-induced cutaneous carcinogenesis that might represent targets for skin cancer prevention.


Asunto(s)
Interleucinas/metabolismo , Queratinocitos/patología , Linfocitos/patología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Neoplasias Cutáneas/patología , Piel/patología , Rayos Ultravioleta/efectos adversos , Animales , Carcinogénesis/metabolismo , Carcinogénesis/patología , Carcinogénesis/efectos de la radiación , Células Cultivadas , Inmunidad Innata/inmunología , Interleucinas/genética , Queratinocitos/metabolismo , Queratinocitos/efectos de la radiación , Células de Langerhans/inmunología , Células de Langerhans/metabolismo , Células de Langerhans/patología , Células de Langerhans/efectos de la radiación , Linfocitos/inmunología , Linfocitos/metabolismo , Linfocitos/efectos de la radiación , Ratones , Mutación , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Piel/metabolismo , Piel/efectos de la radiación , Neoplasias Cutáneas/etiología , Neoplasias Cutáneas/metabolismo , Interleucina-22
7.
Proc Natl Acad Sci U S A ; 118(4)2021 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-33468664

RESUMEN

We have shown previously that phosphorylation of Mdm2 by ATM and c-Abl regulates Mdm2-p53 signaling and alters the effects of DNA damage in mice, including bone marrow failure and tumorigenesis induced by ionizing radiation. Here, we examine the physiological effects of Mdm2 phosphorylation by Akt, another DNA damage effector kinase. Surprisingly, Akt phosphorylation of Mdm2 does not alter the p53-mediated effects of ionizing radiation in cells or mice but regulates the p53 response to oxidative stress. Akt phosphorylation of Mdm2 serine residue 183 increases nuclear Mdm2 stability, decreases p53 levels, and prevents senescence in primary cells exposed to reactive oxidative species (ROS). Using multiple mouse models of ROS-induced cancer, we show that Mdm2 phosphorylation by Akt reduces senescence to promote KrasG12D-driven lung cancers and carcinogen-induced papilloma and hepatocellular carcinomas. Collectively, we document a unique physiologic role for Akt-Mdm2-p53 signaling in regulating cell growth and tumorigenesis in response to oxidative stress.


Asunto(s)
Neoplasias Inducidas por Radiación/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteína p53 Supresora de Tumor/genética , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Carcinogénesis/efectos de la radiación , Carcinógenos/toxicidad , Carcinoma Hepatocelular/inducido químicamente , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/virología , Proliferación Celular/genética , Senescencia Celular/efectos de los fármacos , Senescencia Celular/efectos de la radiación , Daño del ADN/genética , Daño del ADN/efectos de la radiación , Modelos Animales de Enfermedad , Humanos , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/virología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones , Neoplasias Inducidas por Radiación/patología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/efectos de la radiación , Papillomaviridae/patogenicidad , Fosforilación/efectos de los fármacos , Fosforilación/efectos de la radiación , Radiación Ionizante , Especies Reactivas de Oxígeno/metabolismo
8.
Eur J Immunol ; 51(7): 1799-1808, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33759186

RESUMEN

Mononuclear phagocytes consisting of monocytes, macrophages, and DCs play a complex role in tumor development by either promoting or restricting tumor growth. Cutaneous squamous cell carcinoma (cSCC) is the second most common nonmelanoma skin cancer arising from transformed epidermal keratinocytes. While present at high numbers, the role of tumor-infiltrating and resident myeloid cells in the formation of cSCC is largely unknown. Using transgenic mice and depleting antibodies to eliminate specific myeloid cell types in the skin, we investigated the involvement of mononuclear phagocytes in the development of UV-induced cSCC in K14-HPV8-E6 transgenic mice. Although resident Langerhans cells were enriched in the tumor, their contribution to tumor formation was negligible. Equally, dermal macrophages were dispensable for the development of cSCC. In contrast, mice lacking circulating monocytes were completely resistant to UV-induced cSCC, indicating that monocytes promote tumor development. Collectively, these results demonstrate a critical role for classical monocytes in the initiation of skin cancer.


Asunto(s)
Carcinogénesis/patología , Epidermis/patología , Monocitos/patología , Neoplasias Cutáneas/patología , Rayos Ultravioleta/efectos adversos , Animales , Carcinogénesis/efectos de la radiación , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Epidermis/efectos de la radiación , Femenino , Queratinocitos/patología , Queratinocitos/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Monocitos/efectos de la radiación , Piel/patología , Piel/efectos de la radiación
9.
FASEB J ; 35(7): e21671, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34105803

RESUMEN

Ultraviolet irradiation (UV) exposure is the leading factor underlying the development of skin malignancies. D-dopachrome tautomerase (D-DT), a functional homolog of macrophage migration inhibitory factor (MIF), has functional similarities to MIF. However, its role, unlike the role of MIF in photocarcinogenesis, is unknown. We therefore explored the role of D-DT in photocarcinogenesis by developing D-DT transgenic (D-DT Tg) mice and provided a research model for future studies targeting D-DT. Chronic UVB exposure accelerated tumor development in D-DT Tg mice compared with wild-type (WT) mice, with a higher incidence of tumors observed in D-DT Tg mice than in WT mice. In D-DT Tg irradiated mouse keratinocytes, the p53, PUMA, and Bax expression was lower than that in WT mice. These results indicate that D-DT Tg overexpression confers prevention against UVB-induced apoptosis in keratinocytes. Taken together, these findings support D-DT as a functionally important cytokine in photocarcinogenesis and potential therapeutic target for the prevention of photocarcinogenesis.


Asunto(s)
Carcinogénesis/patología , Oxidorreductasas Intramoleculares/metabolismo , Queratinocitos/patología , Neoplasias Cutáneas/patología , Rayos Ultravioleta/efectos adversos , Animales , Apoptosis , Carcinogénesis/metabolismo , Carcinogénesis/efectos de la radiación , Proliferación Celular , Femenino , Oxidorreductasas Intramoleculares/genética , Queratinocitos/metabolismo , Queratinocitos/efectos de la radiación , Ratones , Ratones Transgénicos , Neoplasias Cutáneas/etiología , Neoplasias Cutáneas/metabolismo
10.
Nucleic Acids Res ; 48(16): 9181-9194, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32789493

RESUMEN

The NAD+-dependent deacetylase and mono-ADP-ribosyl transferase SIRT6 stabilizes the genome by promoting DNA double strand break repair, thereby acting as a tumor suppressor. However, whether SIRT6 regulates nucleotide excision repair (NER) remains unknown. Here, we showed that SIRT6 was recruited to sites of UV-induced DNA damage and stimulated the repair of UV-induced DNA damage. Mechanistic studies further indicated that SIRT6 interacted with DDB2, the major sensor initiating global genome NER (GG-NER), and that the interaction was enhanced upon UV irradiation. SIRT6 deacetylated DDB2 at two lysine residues, K35 and K77, upon UV stress and then promoted DDB2 ubiquitination and segregation from chromatin, thereby facilitating downstream signaling. In addition, we characterized several SIRT6 mutations derived from melanoma patients. These SIRT6 mutants ablated the stimulatory effect of SIRT6 on NER and destabilized the genome due to (i) partial loss of enzymatic activity (P27S or H50Y), (ii) a nonsense mutation (R150*) or (iii) high turnover rates (G134W). Overall, we demonstrate that SIRT6 promotes NER by deacetylating DDB2, thereby preventing the onset of melanomagenesis.


Asunto(s)
Carcinogénesis/genética , Daño del ADN/genética , Proteínas de Unión al ADN/genética , Melanoma/genética , Sirtuinas/genética , Carcinogénesis/efectos de la radiación , Cromatina/genética , Cromatina/efectos de la radiación , Daño del ADN/efectos de la radiación , Reparación del ADN/efectos de la radiación , Regulación de la Expresión Génica/efectos de la radiación , Células HeLa , Humanos , Melanoma/patología , Mutación/efectos de la radiación , Ubiquitinación/efectos de la radiación , Rayos Ultravioleta/efectos adversos
11.
Cancer Sci ; 112(6): 2233-2244, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33743547

RESUMEN

According to TCGA database, mutations in PPP6C (encoding phosphatase PP6) are found in c. 10% of tumors from melanoma patients, in which they coexist with BRAF and NRAS mutations. To assess PP6 function in melanoma carcinogenesis, we generated mice in which we could specifically induce BRAF(V600E) expression and delete Ppp6c in melanocytes. In these mice, melanoma susceptibility following UVB irradiation exhibited the following pattern: Ppp6c semi-deficient (heterozygous) > Ppp6c wild-type > Ppp6c-deficient (homozygous) tumor types. Next-generation sequencing of Ppp6c heterozygous and wild-type melanoma tumors revealed that all harbored Trp53 mutations. However, Ppp6c heterozygous tumors showed a higher Signature 1 (mitotic/mitotic clock) mutation index compared with Ppp6c wild-type tumors, suggesting increased cell division. Analysis of cell lines derived from either Ppp6c heterozygous or wild-type melanoma tissues showed that both formed tumors in nude mice, but Ppp6c heterozygous tumors grew faster compared with those from the wild-type line. Ppp6c knockdown via siRNA in the Ppp6c heterozygous line promoted the accumulation of genomic damage and enhanced apoptosis relative to siRNA controls. We conclude that in the presence of BRAF(V600E) expression and UV-induced Trp53 mutation, Ppp6c haploinsufficiency promotes tumorigenesis.


Asunto(s)
Carcinogénesis/genética , Melanoma/genética , Fosfoproteínas Fosfatasas/genética , Proteínas Proto-Oncogénicas B-raf/genética , Rayos Ultravioleta/efectos adversos , Animales , Carcinogénesis/patología , Carcinogénesis/efectos de la radiación , Exoma/genética , Exoma/efectos de la radiación , Genotipo , Haploinsuficiencia , Humanos , Melanocitos/metabolismo , Melanocitos/patología , Melanocitos/efectos de la radiación , Melanoma/patología , Ratones , Ratones Desnudos , Ratones Transgénicos , Mutación/efectos de la radiación , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética
12.
Mol Carcinog ; 60(1): 60-70, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33283918

RESUMEN

Ultraviolet (UV) B irradiation of the skin induces acute inflammation, as characterized by erythema, edema, and immunosuppression, and is subsequently linked to the progression of skin cancer. Toll-like receptor 4 (TLR4), a component of innate immunity, has been shown to play an important role in cancer. To elucidate the role of TLR4 in UVB-induced tumor development, TLR4-proficient (C3H/HeN) and TLR4-deficient (C3H/HeJ) mice were exposed to multiple doses of UVB radiation (200 mJ/cm2 ) for 40 weeks. Photocarcinogenesis was retarded in terms of tumor incidence, and tumor latency, in mice deficient in TLR4 compared with TLR4-proficient mice, whereas significantly greater numbers of tumors occurred in TLR4-proficient mice. There was significant upregulation of inflammatory markers like COX-2, PGE2 , S100A8, and S100A9 in the skin of TLR4-proficient mice than the skin of TLR4-deficient mice. Furthermore, we found that TLR4-proficient mice had a significantly higher number of Gr1+CD11b+ myeloid cells CD4+CD25+ regulatory T-cells than TLR4-deficient mice. Furthermore, the levels of interferon (IFN)-γ cytokine was increased and the levels of interleukin (IL)-4, IL-10, and IL-17 cytokines were decreased in serum, skin, and tumor lysates of TLR4-deficient mice in comparison with samples from TLR4-proficient mice. Together, our data indicate that TLR4-mediated inflammation may cause suppression of antitumor responses and trigger the development of UVB-induced skin cancers. Thus, strategies to inhibit TLR4-mediated immune suppression may allow us to develop preventive and therapeutic approaches for the management of UVB-induced cutaneous tumors.


Asunto(s)
Neoplasias Cutáneas/etiología , Neoplasias Cutáneas/genética , Receptor Toll-Like 4/genética , Rayos Ultravioleta/efectos adversos , Animales , Carcinogénesis/genética , Carcinogénesis/inmunología , Carcinogénesis/efectos de la radiación , Eliminación de Gen , Inmunidad , Inflamación/etiología , Inflamación/genética , Inflamación/inmunología , Ratones , Neoplasias Cutáneas/inmunología , Receptor Toll-Like 4/inmunología
13.
Nucleic Acids Res ; 47(7): 3536-3549, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-30698791

RESUMEN

UV light induces cyclobutane pyrimidine dimers (CPDs) and pyrimidine-pyrimidone (6-4) photoproducts (6-4PPs), which can result in carcinogenesis and aging, if not properly repaired by nucleotide excision repair (NER). Assays to determine DNA damage load and repair rates are invaluable tools for fundamental and clinical NER research. However, most current assays to quantify DNA damage and repair cannot be performed in real time. To overcome this limitation, we made use of the damage recognition characteristics of CPD and 6-4PP photolyases (PLs). Fluorescently-tagged PLs efficiently recognize UV-induced DNA damage without blocking NER activity, and therefore can be used as sensitive live-cell damage sensors. Importantly, FRAP-based assays showed that PLs bind to damaged DNA in a highly sensitive and dose-dependent manner, and can be used to quantify DNA damage load and to determine repair kinetics in real time. Additionally, PLs can instantly reverse DNA damage by 405 nm laser-assisted photo-reactivation during live-cell imaging, opening new possibilities to study lesion-specific NER dynamics and cellular responses to damage removal. Our results show that fluorescently-tagged PLs can be used as a versatile tool to sense, quantify and repair DNA damage, and to study NER kinetics and UV-induced DNA damage response in living cells.


Asunto(s)
Daño del ADN/genética , ADN/genética , Dímeros de Pirimidina/genética , Carcinogénesis/genética , Carcinogénesis/efectos de la radiación , ADN/efectos de la radiación , Daño del ADN/efectos de la radiación , Reparación del ADN/genética , Reparación del ADN/efectos de la radiación , Desoxirribodipirimidina Fotoliasa/genética , Desoxirribodipirimidina Fotoliasa/efectos de la radiación , Humanos , Dímeros de Pirimidina/efectos de la radiación , Rayos Ultravioleta/efectos adversos
14.
Int J Mol Sci ; 22(11)2021 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-34204077

RESUMEN

Skin cancers are growing in incidence worldwide and are primarily caused by exposures to ultraviolet (UV) wavelengths of sunlight. UV radiation induces the formation of photoproducts and other lesions in DNA that if not removed by DNA repair may lead to mutagenesis and carcinogenesis. Though the factors that cause skin carcinogenesis are reasonably well understood, studies over the past 10-15 years have linked the timing of UV exposure to DNA repair and skin carcinogenesis and implicate a role for the body's circadian clock in UV response and disease risk. Here we review what is known about the skin circadian clock, how it affects various aspects of skin physiology, and the factors that affect circadian rhythms in the skin. Furthermore, the molecular understanding of the circadian clock has led to the development of small molecules that target clock proteins; thus, we discuss the potential use of such compounds for manipulating circadian clock-controlled processes in the skin to modulate responses to UV radiation and mitigate cancer risk.


Asunto(s)
Carcinogénesis/patología , Relojes Circadianos/fisiología , Neoplasias Cutáneas/fisiopatología , Fenómenos Fisiológicos de la Piel , Piel/patología , Piel/fisiopatología , Animales , Carcinogénesis/efectos de la radiación , Relojes Circadianos/efectos de la radiación , Humanos , Factores de Riesgo , Piel/efectos de la radiación , Fenómenos Fisiológicos de la Piel/efectos de la radiación
15.
Carcinogenesis ; 41(11): 1565-1575, 2020 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-32157295

RESUMEN

Space radiation is characterized by high-linear energy transfer (LET) ionizing radiation. The relationships between the early biological effects of space radiation and the probability of cancer in humans are poorly understood. Bcl2 not only functions as a potent antiapoptotic molecule but also as an oncogenic protein that induces DNA replication stress. To test the role and mechanism of Bcl2 in high-LET space radiation-induced lung carcinogenesis, we created lung-targeting Bcl2 transgenic C57BL/6 mice using the CC10 promoter to drive Bcl2 expression selectively in lung tissues. Intriguingly, lung-targeting transgenic Bcl2 inhibits ribonucleotide reductase activity, reduces dNTP pool size and retards DNA replication fork progression in mouse bronchial epithelial cells. After exposure of mice to space radiation derived from 56iron, 28silicon or protons, the incidence of lung cancer was significantly higher in lung-targeting Bcl2 transgenic mice than in wild-type mice, indicating that Bcl2-induced DNA replication stress promotes lung carcinogenesis in response to space radiation. The findings provide some evidence for the relative effectiveness of space radiation and Bcl-2 at inducing lung cancer in mice.


Asunto(s)
Carcinogénesis/patología , Replicación del ADN , Neoplasias Pulmonares/patología , Neoplasias Inducidas por Radiación/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Radiación Ionizante , Estrés Fisiológico , Animales , Carcinogénesis/metabolismo , Carcinogénesis/efectos de la radiación , Neoplasias Pulmonares/etiología , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias Inducidas por Radiación/etiología , Neoplasias Inducidas por Radiación/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética
16.
PLoS Pathog ; 14(1): e1006783, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29324843

RESUMEN

Cutaneous beta human papillomavirus (HPV) types are suspected to be involved, together with ultraviolet (UV) radiation, in the development of non-melanoma skin cancer (NMSC). Studies in in vitro and in vivo experimental models have highlighted the transforming properties of beta HPV E6 and E7 oncoproteins. However, epidemiological findings indicate that beta HPV types may be required only at an initial stage of carcinogenesis, and may become dispensable after full establishment of NMSC. Here, we further investigate the potential role of beta HPVs in NMSC using a Cre-loxP-based transgenic (Tg) mouse model that expresses beta HPV38 E6 and E7 oncogenes in the basal layer of the skin epidermis and is highly susceptible to UV-induced carcinogenesis. Using whole-exome sequencing, we show that, in contrast to WT animals, when exposed to chronic UV irradiation K14 HPV38 E6/E7 Tg mice accumulate a large number of UV-induced DNA mutations, which increase proportionally with the severity of the skin lesions. The mutation pattern detected in the Tg skin lesions closely resembles that detected in human NMSC, with the highest mutation rate in p53 and Notch genes. Using the Cre-lox recombination system, we observed that deletion of the viral oncogenes after development of UV-induced skin lesions did not affect the tumour growth. Together, these findings support the concept that beta HPV types act only at an initial stage of carcinogenesis, by potentiating the deleterious effects of UV radiation.


Asunto(s)
Carcinogénesis/efectos de la radiación , Neoplasias Inducidas por Radiación/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Neoplasias Cutáneas/metabolismo , Piel/efectos de la radiación , Rayos Ultravioleta/efectos adversos , Proteínas Virales/metabolismo , Animales , Betapapillomavirus/metabolismo , Epidermis/metabolismo , Epidermis/patología , Epidermis/efectos de la radiación , Femenino , Eliminación de Gen , Genes p53/efectos de la radiación , Ratones , Ratones Transgénicos , Mutagénesis/efectos de la radiación , Mutación , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Inducidas por Radiación/patología , Proteínas Oncogénicas Virales/genética , Receptores Notch/genética , Receptores Notch/metabolismo , Proteínas Recombinantes/metabolismo , Piel/metabolismo , Piel/patología , Neoplasias Cutáneas/etiología , Neoplasias Cutáneas/patología , Carga Tumoral/efectos de la radiación , Proteínas Virales/genética
17.
Exp Dermatol ; 29(1): 29-38, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31519066

RESUMEN

Cutaneous squamous cell carcinoma (cSCC) is the second most common form of skin cancer and is associated with cumulative UV exposure. Studies have shown that prolonged voriconazole use promotes cSCC formation; however, the biological mechanisms responsible for the increased incidence remain unclear. Here, we show that voriconazole directly increases oxidative stress in human keratinocytes and promotes UV-induced DNA damage as determined by comet assay, 8-oxoguanine immunofluorescence and mass spectrometry. Voriconazole treatment of human keratinocytes potentiates UV-induced apoptosis and activation of the p38 MAP kinase and 53BP1 UV stress response pathways. The p38 MAP kinase activation promoted by voriconazole exposure can be mitigated by pretreating keratinocytes with N-acetylcysteine. Voriconazole increases oxidative stress in keratinocytes by directly inhibiting catalase leading to lower intracellular NADPH levels and the triazole moieties in voriconazole are critical for inhibiting catalase. Furthermore, voriconazole is shown to promote UV-induced dysplasia in an in vivo model. Together, these data demonstrate that voriconazole potentiates oxidative stress in UV-irradiated keratinocytes through catalase inhibition. Use of antioxidants may mitigate the pro-oncogenic effects of voriconazole.


Asunto(s)
Antifúngicos/farmacología , Daño del ADN/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Rayos Ultravioleta/efectos adversos , Voriconazol/farmacología , 8-Hidroxi-2'-Desoxicoguanosina/metabolismo , Acetilcisteína/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Carcinogénesis/efectos de los fármacos , Carcinogénesis/efectos de la radiación , Catalasa/antagonistas & inhibidores , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Daño del ADN/efectos de la radiación , Humanos , Queratinocitos/fisiología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de la radiación , Ratones , Cultivo Primario de Células , Piel/efectos de los fármacos , Piel/metabolismo , Piel/patología , Piel/efectos de la radiación , Terbinafina/farmacología , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
18.
J Immunol ; 201(11): 3269-3281, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30389774

RESUMEN

UV radiation (UVR) causing DNA damage is a well-documented risk factor for nonmelanoma skin cancer. Although poorly understood, UVR may also indirectly contribute to carcinogenesis by promoting immune evasion. To our knowledge, we report the first epidemiological study designed to investigate the association between quantitative measures of UVR, obtained using a spectrophotometer, and circulating T regulatory (Treg) cells. In addition to total Treg cells, the proportion of functionally distinct Treg cell subsets defined by CD45RA and CD27 phenotypic markers, graded expression of FOXP3 and CD25, and those expressing cutaneous lymphocyte-associated Ag and the chemokine receptor CCR4 were enumerated in 350 individuals undergoing routine skin cancer screening exams and determined not to have prevalent skin cancer. No associations were identified for UVR exposure or the overall proportion of circulating Treg cells; however, Treg cell subpopulations with an activation-associated phenotype, CD45RA-/CD27-, and those expressing cutaneous homing receptors were significantly positively associated with UVR. These subpopulations of Treg cells also differed by age, sex, and race. After stratification by natural skin tone, and adjusting for age and sex, we found that spectrophotometer-based measures of UVR exposure, but not self-reported measures of past sun exposure, were positively correlated with the highest levels of these Treg cell subpopulations, particularly among lighter-skinned individuals. Findings from this large epidemiologic study highlight the diversity of human Treg cell subpopulations associated with UVR, thus raising questions about the specific coordinated expression of CD45RA, CD27, CCR4, and cutaneous lymphocyte-associated Ag on Treg cells and the possibility that UVR contributes to nonmelanoma skin cancer carcinogenesis through Treg cell-mediated immune evasion.


Asunto(s)
Exposición a la Radiación/efectos adversos , Neoplasias Cutáneas/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Rayos Ultravioleta/efectos adversos , Carcinogénesis/efectos de la radiación , Estudios de Cohortes , Detección Precoz del Cáncer , Femenino , Factores de Transcripción Forkhead/metabolismo , Humanos , Tolerancia Inmunológica , Inmunofenotipificación , Antígenos Comunes de Leucocito/metabolismo , Masculino , Persona de Mediana Edad , Receptores CCR4/metabolismo , Neoplasias Cutáneas/epidemiología , Fenómenos Fisiológicos de la Piel , Pigmentación de la Piel , Subgrupos de Linfocitos T/efectos de la radiación , Linfocitos T Reguladores/efectos de la radiación , Escape del Tumor , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Estados Unidos/epidemiología
19.
Photodermatol Photoimmunol Photomed ; 36(4): 257-262, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32022939

RESUMEN

Phototherapy is the most commonly used modality for repigmenting vitiligo. Currently, UVB emitting devices, including narrow-band UVB (NBUVB) and excimer laser/light, are considered as the treatment of choice. While emitting wavelengths at close proximity, excimer lights emit higher irradiance (HI; W/m2 ) compared to NBUVB. Clinical reports have shown that excimer light is more efficacious in treating vitiligo compared to NBUVB, and we demonstrated that irradiance plays a critical role in promoting melanoblasts differentiation. UVB radiation from the sun is closely associated with photocarcinogenesis of the skin. Sunscreens were used to protect the skin by reducing UVB irradiance (low irradiance (LI) UVB). Sunscreen use was associated with skin cancer reduction in clinical trials. Paradoxically, sunscreen use was associated with increased sunburn episodes in the real-world settings. It was shown that UVB-induced sunburn depends on fluence (J/m2 ) but not irradiance of UVB radiation. We investigated the significance of irradiance in the context of UVB-induced carcinogenesis of the skin. For mice receiving equivalent fluence of UVB exposure, the LIUVB-treated mice showed earlier tumor development, larger tumor burden, and more epidermal keratinocytes harboring mutant p53 as compared to their HIUVB-treated counterparts. These results suggested that at equivalent fluence, LIUVB radiation has more photocarcinogenic potential on the skin compared to its HI counterpart. Since development of sunburn with or without sunscreen use indicates that certain threshold of UVB fluence has been received by the skin at LI and HI, respectively, sunburn episodes with sunscreen use (LIUVB) are more damaging to the skin compared to that without sunscreen (HIUVB) application. In summary, since irradiance plays an important role determining the biological effects of UVB radiation on the skin, future related studies should take this critical parameter into consideration.


Asunto(s)
Carcinogénesis/efectos de la radiación , Neoplasias Cutáneas/etiología , Rayos Ultravioleta/efectos adversos , Terapia Ultravioleta , Vitíligo/radioterapia , Animales , Humanos , Láseres de Excímeros/uso terapéutico , Ratones , Neoplasias Cutáneas/prevención & control , Protectores Solares/uso terapéutico , Terapia Ultravioleta/efectos adversos , Terapia Ultravioleta/métodos
20.
Molecules ; 25(20)2020 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-33081281

RESUMEN

A 'remedy for all' natural product widely known in the Korean Peninsula is called Panax Ginseng Meyer. Globalization represents a persistent risk to the ozone layer, leading to bountiful amounts of Ultra-Violet B beams (UVB). The variety in human skin hues is ascribed to the characteristic color called Melanin. However, Melanin overproduction due to UVB beams promotes skin staining and tumorigenesis, a process called photo aging, which damages skin quality. To assess the effects of Korean Red Ginseng Oil (KGO) on photo aging, the murine melanoma cell lines B16/F10 were used in vitro and HRM-2 hairless mice exposed to UVB were studied in vivo. Our results revealed that KGO reduced tyrosinase activity and melanin production in B16/F10 cells along with the suppression of upstream factors involved in the melanin production pathway, both transcriptionally and transitionally. In the in vivo studies, KGO suppressed the expression of Matrix Metalloproteinase (MMP) and Interleukins along with a reduction of depth in wrinkle formation and reduced collagen degradation. Moreover, the feed intake and feed efficiency ratio that decreased as a result of UVB exposure was also improved by KGO treatment. In light of our results, we conclude that KGO can have considerable benefits due to its various properties of natural skin enhancement.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Melanoma Experimental/tratamiento farmacológico , Panax/química , Aceites de Plantas/farmacología , Animales , Carcinogénesis/efectos de la radiación , Fibroblastos/efectos de los fármacos , Humanos , Melaninas/biosíntesis , Melaninas/efectos de la radiación , Ratones , Ratones Pelados , Ozono/efectos adversos , Extractos Vegetales/química , Extractos Vegetales/farmacología , Aceites de Plantas/química , Piel/efectos de los fármacos , Piel/metabolismo , Rayos Ultravioleta/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA