首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
The importance of the Cdk4 protein in human cancer became evident following the identification of a germ line mutation in the Cdk4 locus that predisposes humans to melanoma. This mutation results in substitution of argininefirst with cysteine at position 24 (R24C). In an earlier study, we introduced the R24C mutation into the Cdk4 locus of mice using Cre-loxp-mediated “knock-in” technology and observed a very low incidence of spontaneous melanomas in Cdk4R24C/R24C mice. This suggested that additional oncogenic mutations might be required for development of melanomas. Here we report an increased incidence of spontaneous cutaneous melanoma in mice expressing the oncogene HRAS(G12V) in melanocytes on a Cdk4R24C background. Treatment of Tyr-HRas:Cdk4R24C/R24C mice with the carcinogen, DMBA/TPA resulted in a further increase in the number of nevi and melanomas developed when compared with Tyr-HRas:Cdk4+/+ mice. In summary, in Tyr-HRas:Cdk4R24C/R24C mice, we observed that activated Cdk4 cooperates with the oncogenic HRAS(G12V) protein to increase the susceptibility of melanoma development in vivo.Key words: Cdk4R24C, ras, melanoma, skin, carcinogen  相似文献   

2.
To further investigate the use of DNA repair-enhancing agents for skin cancer prevention, we treated Cdk4R24C/R24C/NrasQ61K mice topically with the T4 endonuclease V DNA repair enzyme (known as Dimericine) immediately prior to neonatal ultraviolet radiation (UVR) exposure, which has a powerful effect in exacerbating melanoma development in the mouse model. Dimericine has been shown to reduce the incidence of basal-cell and squamous cell carcinoma. Unexpectedly, we saw no difference in penetrance or age of onset of melanoma after neonatal UVR between Dimericine-treated and control animals, although the drug reduced DNA damage and cellular proliferation in the skin. Interestingly, epidermal melanocytes removed cyclobutane pyrimidine dimers (CPDs) more efficiently than surrounding keratinocytes. Our study indicates that neonatal UVR-initiated melanomas may be driven by mechanisms other than solely that of a large CPD load and/or their inefficient repair. This is further suggestive of different mechanisms by which UVR may enhance the transformation of keratinocytes and melanocytes.  相似文献   

3.
The importance of the CDK4 protein in human cancer first became evident following the identification of a germ line mutation in the Cdk4 locus that predisposes humans to melanoma. This mutation results in substitution of arginine with cysteine at position 24 (R24C). In an earlier study, we introduced the R24C mutation into the Cdk4 locus of mice using Cre-loxP-mediated "knock-in" technology and observed a very low incidence of spontaneous melanomas in Cdk4R24C/R24C mice. This suggested that additional oncogenic mutations might be required for development of melanomas. Here we report an increased incidence of spontaneous cutaneous melanoma in mice expressing the oncogene HRAS(G12V) in melanocytes on a Cdk4R24C background. Treatment of Tyr-HRas:Cdk4R24C/R24C mice with the carcinogen, DMBA/TPA resulted in a further increase in the number of nevi and melanomas developed when compared with Tyr-HRas:Cdk4+/+ mice. In summary, in Tyr-HRas:Cdk4R24C/R24C mice, we observed that activated CDK4 cooperates with the oncogenic HRAS(G12V)protein to increase the susceptibility of melanoma development in vivo.  相似文献   

4.
Ultraviolet radiation (UVR) is a major risk factor for melanoma development, but it has been unclear exactly how UVR leads to melanomagenesis. In a recent publication in Nature, Viros et al. identify TP53/Trp53 as a UVR-target gene in melanoma and show that UVR-induced TP53/Trp53 mutations accelerate BRAF(V600E)-driven melanomagenesis.Melanoma is the deadliest skin cancer, and its incidence has relentlessly increased over recent decades. According to the American Cancer Society''s estimates for melanoma in the United States for 2014, about 76 100 new melanomas will be diagnosed and about 9 710 people are expected to die from melanoma. It is well known that UVR is the major environmental factor contributing to melanomagenesis1. This suggests that there is at least a component of melanoma risk which may be preventable through UVR protective strategies — an issue of immense public health importance due to the availability of sunblocks and sun-safe behaviors. Importantly, while many studies have been conducted to elucidate the link between UVR and melanoma, the precise molecular mechanism(s) by which UVR triggers melanoma formation have remained incompletely understood.Recently, a powerful UVR-induced skin inflammatory response has been shown to provoke metastasis of melanoma2 and the presence of UV signature mutations has also been reported throughout the melanoma exome, including recurrent melanoma genes such as RAC1, PPP6C, and STK193,4. Previous mouse models for UVR-induced melanoma revealed that UVR-induced inflammation promoted melanomagenesis in neonatal mice5,6,7. These studies underlined UVR''s significant contribution to melanoma formation. In a recent study published in Nature, Viros et al.8 address the role of UVR in previously established BRAF(V600E)-expressing melanocytes in vivo, and demonstrate that significantly accelerated melanoma formation often associated with mutations in TP53/Trp53. To mimic both somatic mutation acquisition and mild sunburn in humans, BRAF(V600E) was expressed at physiological levels in adult mice which were subsequently exposed to repeated low doses of UVR. In addition, certain mice were partially covered with UVR-proof cloth or topically treated with SunSense Milk Sunscreen SPF50 (2.2 mg/cm2) 30 min before UVR exposure, to assess the impact of these protective strategies.UVR was seen to significantly accelerate melanoma formation in mice whose melanocytes express BRAF(V600E), but not in BRAF wild-type mice (which unlike BRAF(V600E)-expressing mice do not develop long latency melanomas independently of UVR). Application of UVR-proof cloth or sunscreen delayed the onset of UVR-driven melanoma and partially prevented acceleration of BRAF(V600E)-driven melanomagenesis by UVR, and sunscreen-protected UVR-exposed BRAF(V600E) mice developed a reduced number of melanomas compared with unprotected UVR-exposed BRAF(V600E) mice (Figure 1). More somatic single nucleotide variants and a significantly higher proportion of C-to-T transitions at the 3′ end of pyrimidine dimers were observed in UVR-exposed melanomas, providing direct evidence of UVR-induced DNA damage. In addition, Trp53 mutations (H39Y, S124F, R245C, R270C, C272G) were detected in UVR-exposed BRAF(V600E) mouse melanomas, indicating a direct role of UVR in the induction of Trp53 mutations in melanoma. The mutated corresponding residues (S127F/S124F, R248C/R245C, R273C/R270C, C275G/C272G) were also identified in TP53 mutations in human melanoma, suggesting that TP53 mutations are linked to evidence of UVR-induced DNA damage in human melanoma. These results are consistent with previous reports that p53 deletion accelerates BRAF(V600E)-driven melanomagenesis both in mice9 and in zebrafish10, but demonstrate the ability of UVR to inflict UV signature mutations within the gene as has been widely observed in non-melanoma skin cancers and also in human melanomas.Open in a separate windowFigure 1A diagram depicting feasible routes of BRAF(V600E)-driven melanomagenesis.This elegant study by Viros et al. clearly helps to establish key roles of UVR in melanomagenesis, and further validates the functional importance of TP53 as a UVR-targeted tumor suppressor gene in a fraction of melanomas. The study also raises several intriguing questions worthy of follow-up analysis. For example, through which mechanism(s) did sunscreen or sunshielding delay but not prevent UVR-induced melanoma? Induction of cutaneous inflammatory changes that are less anatomically restricted to UV irradiated fields, would seem to be an attractive mechanism. This may help to explain the known risk of melanoma in both sun-exposed and less-exposed skin of lightly pigmented people. It is also valuable to better understand the role of UVB vs UVA wavelengths in melanomagenesis. Mechanistically, these distinct regions of the UV spectrum inflict largely distinctive chemical alterations on the genome. Efforts to block UVA as well as UVB in commercial sunscreen products are currently being promoted by the US Food and Drug Administration, a welcome improvement to sun protection strategies. Still, the precise role(s) of UVA in melanomagenesis remain incompletely understood and may involve both cell-autonomous and non-cell-autonomous targets. In addition to the acceleration of BRAF(V600E)-driven melanoma formation by UVR, red pigment (pheomelanin) has also been observed to accelerate BRAF(V600E)-driven melanomagenesis even in the absence of UVR11. Pheomelanin has been identified as an intrinsic risk factor for melanoma with the red pigment itself producing reactive oxygen species that cause DNA damage in the skin, and consequently promote melanomagenesis independently of UVR. UVR likely exacerbates red pigment-induced BRAF(V600E)-driven melanoma, and still remains as a major contributor to melanomagenesis. Therefore, along with UV shielding by sunscreens, further preventative strategies should be investigated to diminish UVR-independent melanoma risk mechanisms.Viros et al. provide intriguing answers to several controversial questions regarding melanomagenesis: Does UVR really trigger melanoma? And can sunscreen actually prevent melanoma? The studies by Viros et al. provide experimental evidence for acceleration of BRAF(V600E)-driven melanoma by UVR-induced TP53/Trp53 mutation and demonstrate that sunscreen delayed but did not completely block UVR-driven melanoma. The current study clearly shows that UVR boosts melanoma and sunscreens may provide partial UVR protection against melanoma — evidence which matches human epidemiologic data. Nevertheless, to protect the public from melanoma, Viros et al. advise that sunscreen should be utilized in combination with additional sun avoidance strategies. In addition, measures that may prevent UV-independent melanoma formation will require additional research and may also be needed in order to optimally battle the incidence of this life-threatening malignancy.  相似文献   

5.
Malignant transformation of melanocytes leads to melanoma, the most fatal form of skin cancer. Ultraviolet radiation (UVR)‐induced DNA photoproducts play an important role in melanomagenesis. Cutaneous melanin content represents a major photoprotective mechanism against UVR‐induced DNA damage, and generally correlates inversely with the risk of skin cancer, including melanoma. Melanoma risk is also determined by susceptibility genes, one of which is the melanocortin 1 receptor (MC1R) gene. Certain MC1R alleles are strongly associated with melanoma. We hereby present experimental evidence for the role of two melanoma risk factors, constitutive pigmentation, as assessed by total melanin, eumelanin and pheomelanin contents, and MC1R genotype and function, in determining the induction and repair of DNA photoproducts in cultured human melanocytes after irradiation with increasing doses of UVR. We found that total melanin and eumelanin contents (MC and EC) correlated inversely with the extent of UVR‐induced growth arrest, apoptosis and induction of cyclobutane pyrimidine dimers (CPD), but not with hydrogen peroxide release in melanocytes expressing functional MC1R. In comparison, melanocytes with loss‐of‐function MC1R, regardless of their MC or EC, sustained more UVR‐induced apoptosis and CPD, and exhibited reduced CPD repair. Therefore, MC, mainly EC, and MC1R function are independent determinants of UVR‐induced DNA damage in melanocytes.  相似文献   

6.
7.
Ectopic expression of oncogenes such as Ras induces expression of p19(Arf), which, in turn, activates p53 and growth arrest. Here, we used a multistage model of squamous cell carcinoma development to investigate the functional interactions between Ras, p19(Arf), and p53 during tumor progression in the mouse. Skin tumors were induced in wild-type, p19(Arf)-deficient, and p53-deficient mice using the DMBA/TPA two-step protocol. Activating mutations in Hras were detected in all papillomas and carcinomas examined, regardless of genotype. Relative to wild-type mice, the growth rate of papillomas was greater in p19(Arf)-deficient mice, and reduced in p53-deficient mice. Malignant conversion of papillomas to squamous cell carcinomas, as well as metastasis to lymph nodes and lungs, was markedly accelerated in both p19 (Arf)- and p53-deficient mice. Thus, p19(Arf) inhibits the growth rate of tumors in a p53-independent manner. Through its regulation of p53, p19(Arf) also suppresses malignant conversion and metastasis. p53 expression was upregulated in papillomas from wild-type but not p19( Arf)-null mice, and p53 mutations were more frequently seen in wild-type than in p19( Arf)-null carcinomas. This indicates that selection for p53 mutations is a direct result of signaling from the initiating oncogenic lesion, Hras, acting through p19(Arf).  相似文献   

8.
Cyclin dependent kinase 4 (Cdk4) is a cell cycle regulator involved in early G1 cell cycle progression and has been indirectly implicated in angiogenesis in the Min mouse system, a mouse that harbors a mutation in the Apc gene. Apc+/Min mice when crossed with Ink4a/arf-/- mice, exhibited increased angiogenesis of colorectal tumors suggesting that dysregulation of Cdk4 (due to loss of Ink4a-mediated suppression) may contribute to enhanced angiogenesis. To demonstrate a direct role for Cdk4 in angiogenesis, we crossed mice that have an activated Cdk4, Cdk4R24C/R24C mice, with Apc+/Min mice and examined levels of angiogenesis in intestinal tumors formed. Our results show an increase in the percentage of highly vascularized tumors in Cdk4R24C/R24C:ApcMin/+ and Cdk4+/R24C:ApcMin/+ mice compared to Cdk4+/+:ApcMin/+ mice. In addition immunohistochemical analysis showed an increase in CD-31 staining localized to endothelial cells of Cdk4R24C/R24C:ApcMin/+ mouse tumors, supporting the hypothesis of increased vasculature in these tumors. Further analysis showed an increase in the expression of the E2F1 target proteins Vegf-b and Cyclin A in Cdk4R24C/R24C:Apc+/Min intestinal tumors. Together these data suggest that the dysregulated Cdk4 gene plays an important role in angiogenesis during intestinal tumor formation and may in part act via increasing E2F1 target proteins. This is the first report to show that Cdk4 has a direct role in angiogenesis in vivo and may be an important drug target to reduce or prevent angiogenesis during intestinal tumor formation.  相似文献   

9.
Glioblastoma multiforme (GBM) is the most common and malignant form of glioma with high mortality and no cure. Many human cancers maintain a complex inflammatory program triggering rapid recruitment of inflammatory cells, including mast cells (MCs), to the tumor site. However, the potential contribution of MCs in glioma has not been addressed previously. Here we report for the first time that MCs infiltrate KRas+Akt-induced gliomas, using the RCAS/TV-a system, where KRas and Akt are transduced by RCAS into the brains of neonatal Gtv-a- or Ntv-a transgenic mice lacking Ink4a or Arf. The most abundant MC infiltration was observed in high-grade gliomas of Arf-/- mice. MC accumulation could be localized to the vicinity of glioma-associated vessels but also within the tumor mass. Importantly, proliferating MCs were detected, suggesting that the MC accumulation was caused by local expansion of the MC population. In line with these findings, strong expression of stem cell factor (SCF), i.e. the main MC growth factor, was detected, in particular around tumor blood vessels. Further, glioma cells expressed the MC chemotaxin CXCL12 and MCs expressed the corresponding receptor, i.e. CXCR4, suggesting that MCs could be attracted to the tumor through the CXCL12/CXCR4 axis. Supporting a role for MCs in glioma, strong MC infiltration was detected in human glioma, where GBMs contained significantly higher MC numbers than grade II tumors did. Moreover, human GBMs were positive for CXCL12 and the infiltrating MCs were positive for CXCR4. In conclusion, we provide the first evidence for a role for MCs in glioma.  相似文献   

10.
We have introduced a point mutation in the first coding exon of the locus encoding the cyclin-dependent kinase 4 (Cdk4) by homologous recombination in embryonic stem cells. This mutation (replacement of Arg24 by Cys) was first found in patients with hereditary melanoma and renders Cdk4 insensitive to INK4 inhibitors. Here, we report that primary embryonic fibroblasts expressing the mutant Cdk4R24C kinase are immortal and susceptible to transformation by Ras oncogenes. Moreover, homozygous Cdk4(R24C/R24C) mutant mice develop multiple tumors with almost complete penetrance. The most common neoplasia (endocrine tumors and hemangiosarcomas) are similar to those found in pRb(+/-) and p53(-/-) mice. This Cdk4 mutation cooperates with p53 and p27(Kip1) deficiencies in decreasing tumor latency and favoring development of specific tumor types. These results provide experimental evidence for a central role of Cdk4 regulation in cancer and provide a valuable model for testing the potential anti-tumor effect of Cdk4 inhibitors in vivo.  相似文献   

11.
Whole‐genome sequencing of matched germline and tumour pairs in a well‐characterized cohort of melanoma patients allowed investigation of associations between melanoma body site, age at melanoma onset and MC1R variant status with overall mutation burden and specific base pair changes observed in the corresponding melanoma. We observed statistically significant associations between mutation burden in melanoma and body site, age at onset and MC1R genotype, for both ultraviolet radiation (UVR) signature changes (C>T and CC>TT) and non‐UVR base pair substitutions, as well as with overall variant load.  相似文献   

12.
The status of tumor suppressor genes (TSGs) relevant to human malignant mesothelioma (HMM) pathogenesis was examined in cultures of mesothelioma cells from tumoral ascites developed in mice exposed to asbestos (asb) fibers. The status of the respective hortologous human genes was also investigated in 12 HMM cell cultures. Eleven primary cultures from mice hemizygous for N?2 (asb-Nf2KO3/+) and 4 wild type counterparts (asb-Nf2+/+) were analyzed for mutations in Nf2, p16/Cdkn2a, p19/Arf and Trp53 genes and protein expression of p15/Cdkn2b and Cdk4. TSG alterations in both mouse and human mesothelioma cells consisted in frequent inactivation of p16/Cdkn2a, p19/Arf (or P14/ARF) and p15/Cdkn2b, co-inactivation of p16/Cdkn2a and p15/Cdkn2b and low rate of Trp53 mutations in both asb-Nf2KO3/+ and asb-Nf2+/+ mesothelioma cells. In both mouse and human mesothelioma cells, inactivation of the hortologous genes p16/Cdkn2a or P16/CDKN2A was due to deletions at the Ink4/Arf locus encompassing p19/Arf or P14/ARF, respectively. Loss of heterozygosity at the Nf2 locus was detected in 10 of 11 asb-Nf2KO3/+ cultures and Nf2 gene rearrangement in one asb-Nf2+/+ culture. These data show that the profile of TSG alterations in asbestos-induced mesothelioma is similar in mice and humans. Thus, the mouse mesothelioma model could be useful for human risk assessment, taking into account interindividual variations in genetic sensitivity to carcinogens.  相似文献   

13.
Inactivation of the Arf-Mdm2-p53 tumor suppressor pathway is a necessary event for tumorigenesis. Arf controls Mdm2, which in turn regulates p53, but Arf and Mdm2 also have p53-independent functions that affect tumor development. Moreover, inhibition of oncogene-induced tumorigenesis relies on Arf and p53, but the requirements of Arf and p53 in tumor development initiated in the absence of overt oncogene overexpression and the role of Mdm2 in this process remain unclear. In a series of genetic experiments in mice with defined deficiencies in Arf, Mdm2 and/or p53, we show Mdm2 haploinsufficiency significantly delayed tumorigenesis in mice deficient in Arf and p53. Mdm2 heterozygosity significantly inhibited tumor development in the absence of Arf, and in contrast to Myc oncogene-driven cancer, this delay in tumorigenesis could not be rescued with the presence of one allele of Arf. Notably, Mdm2 haploinsufficieny blocked the accelerated tumor development in Arf deficient mice caused by p53 heterozygosity. However, tumorigenesis was not inhibited in Mdm2 heterozygous mice lacking both alleles of p53 regardless of Arf status. Surprisingly, loss of Arf accelerated tumor development in p53-null mice. Tumor spectrum was largely dictated by Arf and p53 status with Mdm2 haploinsufficiency only modestly altering the tumor type in some of the genotypes and not the number of primary tumors that arose. Therefore, the significant effects of Mdm2 haploinsufficiency on tumor latency were independent of Arf and required at least one allele of p53, and an Mdm2 deficiency had minor effects on the types of tumors that developed. These data also demonstrate that decreased levels of Mdm2 are protective in the presence of multiple genetic events in Arf and p53 genes that normally accelerate tumorigenesis.  相似文献   

14.
MC1R and the response of melanocytes to ultraviolet radiation   总被引:5,自引:0,他引:5  
The constitutive color of our skin plays a dramatic role in our photoprotection from solar ultraviolet radiation (UVR) that reaches the Earth and in minimizing DNA damage that gives rise to skin cancer. More than 120 genes have been identified and shown to regulate pigmentation, one of the key genes being melanocortin 1 receptor (MC1R) that encodes the melanocortin 1 receptor (MC1R), a seven-transmembrane G protein-coupled receptor expressed on the surface of melanocytes. Modulation of MC1R function regulates melanin synthesis by melanocytes qualitatively and quantitatively. The MC1R is regulated by the physiological agonists alpha-melanocyte-stimulating hormone (alphaMSH) and adrenocorticotropic hormone (ACTH), and antagonist agouti signaling protein (ASP). Activation of the MC1R by binding of an agonist stimulates the synthesis of eumelanin primarily via activation of adenylate cyclase. The significance of cutaneous pigmentation lies in the photoprotective effect of melanin, particularly eumelanin, against sun-induced carcinogenesis. Epidermal melanocytes and keratinocytes respond to UVR by increasing their expression of alphaMSH and ACTH, which up-regulate the expression of MC1R, and consequently enhance the response of melanocytes to melanocortins. Constitutive skin pigmentation dramatically affects the incidence of skin cancer. The pigmentary phenotype characterized by red hair, fair complexion, inability to tan and tendency to freckle is an independent risk factor for all skin cancers, including melanoma. The MC1R gene is highly polymorphic in human populations, and allelic variation at this locus accounts, to a large extent, for the variation in pigmentary phenotypes and skin phototypes (SPT) in humans. Several allelic variants of the MC1R gene are associated with the red hair and fair skin (RHC) phenotype, and carrying one of these variants is thought to diminish the ability of the epidermis to respond to DNA damage elicited by UVR. The MC1R gene is considered a melanoma susceptibility gene, and its significance in determining the risk for skin cancer is of tremendous interest.  相似文献   

15.
We previously noted that melanomas developing in Cdk4R24C/R24C::Tyr‐NRAS, Arf?/?::Tyr‐NRAS and Trp53F/F::Tyr‐Cre(ER)::Tyr‐NRAS mice exhibited differences in behaviour in vivo. We investigated this phenomenon using global gene expression profiling of lesions from the respective genotypes. While those from the Cdk4‐ and Arf‐mutant mice exhibited similar profiles, the Trp53F/F::Tyr‐Cre(ER)::Tyr‐NRAS melanomas were strikingly different, showing relative down‐regulation of melanocyte‐related genes, and up‐regulation of genes related to neural differentiation. Specifically, they highly expressed genes representative of the myelin‐producing peripheral oligodendrite (Schwann cell) lineage, although histopathologically the lesions did not exhibit the classical features of schwannoma. As Schwann cell precursors can be a cellular origin of melanocytes, it is unsurprising that plasticity with respect to melanocyte‐neural differentiation can occur in melanoma. What is surprising is the genotype proclivity. Comparison of gene expression signatures revealed that melanomas from the Trp53‐mutant mice show significant similarities with a subset of aggressive human melanomas with relatively low levels of MITF.  相似文献   

16.
Pre-B-cell transformation by Abelson virus (Ab-MLV) is a multistep process in which primary transformants are stimulated to proliferate but subsequently undergo crisis, a period of erratic growth marked by high levels of apoptosis. Inactivation of the p53 tumor suppressor pathway is an important step in this process and can be accomplished by mutation of p53 or down-modulation of p19(Arf), a p53 regulatory protein. Consistent with these data, pre-B cells from either p53 or Ink4a/Arf null mice bypass crisis. However, the Ink4a/Arf locus encodes both p19(Arf) and a second tumor suppressor, p16(Ink4a), that blocks cell cycle progression by inhibiting Cdk4/6. To determine if p16(Ink4a) plays a role in Ab-MLV transformation, primary transformants derived from Arf(-/-) and p16(Ink4a(-/-)) mice were compared. A fraction of those derived from Arf(-/-) animals underwent crisis, and even though all p16(Ink4a(-/-)) primary transformants experienced crisis, these cells became established more readily than cells derived from +/+ mice. Analyses of Ink4a/Arf(-/-) cells infected with a virus that expresses both v-Abl and p16(Ink4a) revealed that p16(Ink4a) expression does not alter cell cycle profiles but does increase the level of apoptosis in primary transformants. These results indicate that both products of the Ink4a/Arf locus influence Ab-MLV transformation and reveal that in addition to its well-recognized effects on the cell cycle, p16(Ink4a) can suppress transformation by inducing apoptosis.  相似文献   

17.
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.  相似文献   

18.
The melanocortin 1 receptor (MC1R) is a G protein‐coupled receptor crucial for the regulation of melanocyte proliferation and function. Upon binding melanocortins, MC1R activates several signaling cascades, notably the cAMP pathway leading to synthesis of photoprotective eumelanin. Polymorphisms in the MC1R gene are a major source of normal variation of human hair color and skin pigmentation, response to ultraviolet radiation (UVR), and skin cancer susceptibility. The identification of a surprisingly high number of MC1R natural variants strongly associated with pigmentary phenotypes and increased skin cancer risk has prompted research on the functional properties of the wild‐type receptor and frequent mutant alleles. We summarize current knowledge on MC1R structural and functional properties, as well as on its intracellular trafficking and signaling. We also review the current knowledge about the function of MC1R as a skin cancer, particularly melanoma, susceptibility gene and how it modulates the response of melanocytes to UVR.  相似文献   

19.
20.
Malignant transformation of melanocytes leads to melanoma, the most fatal form of skin cancer. Ultraviolet radiation (UVR)-induced DNA photoproducts play an important role in melanomagenesis. Cutaneous melanin content represents a major photoprotective mechanism against UVR-induced DNA damage, and generally correlates inversely with the risk of skin cancer, including melanoma. Melanoma risk is also determined by susceptibility genes, one of which is the melanocortin 1 receptor (MC1R) gene. Certain MC1R alleles are strongly associated with melanoma. We hereby present experimental evidence for the role of two melanoma risk factors, constitutive pigmentation, as assessed by total melanin, eumelanin and pheomelanin contents, and MC1R genotype and function, in determining the induction and repair of DNA photoproducts in cultured human melanocytes after irradiation with increasing doses of UVR. We found that total melanin and eumelanin contents (MC and EC) correlated inversely with the extent of UVR-induced growth arrest, apoptosis and induction of cyclobutane pyrimidine dimers (CPD), but not with hydrogen peroxide release in melanocytes expressing functional MC1R. In comparison, melanocytes with loss-of-function MC1R, regardless of their MC or EC, sustained more UVR-induced apoptosis and CPD, and exhibited reduced CPD repair. Therefore, MC, mainly EC, and MC1R function are independent determinants of UVR-induced DNA damage in melanocytes.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号