Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Clin Cancer Res ; 29(1): 100-109, 2023 01 04.
Article in English | MEDLINE | ID: mdl-36112545

ABSTRACT

PURPOSE: Intratumoral oncolytic virotherapy may overcome anti-PD(L)-1 resistance by triggering pro-inflammatory remodeling of the tumor microenvironment. This pilot study investigated ONCOS-102 (oncolytic adenovirus expressing GM-CSF) plus anti-programmed cell death protein 1 (PD)-1 therapy in anti-PD-1-resistant melanoma. PATIENTS AND METHODS: Patients with advanced melanoma progressing after prior PD-1 blockade received intratumoral ONCOS-102 either as priming with 3 doses (3 × 1011 viral particles) during Week 1 [Part 1 (sequential treatment)] or as 4-dose priming and 8 booster doses every 3 weeks [Part 2 (combination treatment)]. From Week 3, all patients received pembrolizumab every 3 weeks (≤8 doses). The primary endpoint was safety. Objective response rate (ORR), progression-free survival, and immunologic activation in repeat biopsies were also investigated. RESULTS: In 21 patients (Part 1, n = 9; Part 2, n = 12) ONCOS-102 plus pembrolizumab was well tolerated: most adverse events (AE) were mild/moderate in severity. Pyrexia (43%), chills (43%), and nausea (28%) were the most common ONCOS-102-related AEs. There were no dose-limiting toxicities. ORR was 35% [response evaluation in solid tumors (RECIST) 1.1, irRECIST]. Reduction in size of ≥1 non-injected lesions observed in 53% patients indicated a systemic effect. In injected tumors, persistent immune-related gene expression and T-cell infiltration were associated with clinical benefit. Viral persistence and efficacy in injected and non-injected lesions without additional toxicity supported Part 2 dosing regimen in future studies. CONCLUSIONS: ONCOS-102 plus pembrolizumab was well tolerated and led to objective responses in patients with anti-PD-1-resistant advanced melanoma. ONCOS-102 promoted T-cell infiltration, particularly cytotoxic CD8+ T cells, which persisted at Week 9, driving clinical benefit. Further investigation of ONCOS-102 plus PD-1 blockade is warranted. See related commentary by Levi and Boland, p. 3.


Subject(s)
Melanoma , Tumor Microenvironment , Humans , Pilot Projects , Antibodies, Monoclonal, Humanized/administration & dosage , Melanoma/drug therapy
3.
Pigment Cell Melanoma Res ; 34(5): 905-917, 2021 09.
Article in English | MEDLINE | ID: mdl-33544968

ABSTRACT

Melanocyte stem cells (McSCs) are key components of the hair follicle (HF) stem cell system that regenerate differentiated melanocytes during successive HF cycles. To facilitate continued research on melanocyte development and differentiation and McSCs, we backcrossed inducible Dct-H2BGFP mice into the C57BL/6J background (B6-Dct-H2BGFP). We compared the expression pattern of B6-Dct-H2BGFP to that of Dct-H2BGFP mice on a mixed genetic background reported previously. To characterize B6-Dct-H2BGFP mice, we confirmed not only the expression of GFP in all melanocyte lineage cells, but also doxycycline regulation of GFP expression. Furthermore, ex vivo culture of the McSC subsets isolated by fluorescence-activated cell sorting (FACS) showed the propensity of bulge/CD34+ McSCs to differentiate with expression of non-melanocytic, neural crest lineage markers including glia (Gfap and CNPase, 73 ± 1% and 77 ± 2%, respectively), neurons (Tuj1 26 ± 5%), and smooth muscle (α-Sma, 31 ± 9%). In contrast, CD34-/secondary hair germ (SHG) McSCs differentiated into pigmented melanocytes, with higher expression of melanogenic markers Tyr (71 ± 1%), Tyrp1 (68 ± 4%), and Mitf (75 ± 7%). These results establish the utility of B6-Dct-H2BGFP bitransgenic mice for future in vivo studies of melanocytes requiring a defined genetic background.


Subject(s)
Cell Differentiation , Gene Expression Regulation , Melanocytes/metabolism , Membrane Glycoproteins/biosynthesis , Microphthalmia-Associated Transcription Factor/biosynthesis , Models, Biological , Oxidoreductases/biosynthesis , Stem Cells/metabolism , Animals , Membrane Glycoproteins/genetics , Mice , Mice, Transgenic , Microphthalmia-Associated Transcription Factor/genetics , Oxidoreductases/genetics
4.
J Invest Dermatol ; 140(4): 743-745, 2020 04.
Article in English | MEDLINE | ID: mdl-32200877

ABSTRACT

Wenzina et al. (2020) explore the potential role of E-cadherin (CDH1) as a marker for invasive behavior in melanoma. The authors show that CDH1 expression is modulated by p38 signaling, and that manipulation of this pathway can impede endothelial disruption and lung dissemination in vivo and in vitro. The downstream markers PODXL and DEL of the invasive phenotype are associated with a poor prognosis.


Subject(s)
Melanoma , Skin Neoplasms , Adaptation, Physiological , Cadherins/genetics , Cell Line, Tumor , Humans , Melanoma/genetics , Signal Transduction , Skin Neoplasms/genetics
6.
Oncologist ; 24(12): 1510-e1265, 2019 12.
Article in English | MEDLINE | ID: mdl-31350329

ABSTRACT

LESSONS LEARNED: This is the first human interventional study in patients with Cowden syndrome that is driven by inactivation of germline PTEN gene.Single-agent sirolimus, a mTOR inhibitor, suppressed mTOR signaling in surrogate human tissues without significant toxicity. BACKGROUND: Cowden syndrome is characterized by inactivating germline PTEN mutations, which can lead to activation of the PI3K-Akt-mTOR pathway. METHODS: Adult subjects with germline PTEN mutation who met international diagnostic criteria for Cowden syndrome and who had Eastern Cooperative Oncology Group (ECOG) performance status 0-2 and adequate organ function were enrolled. Subjects were treated with a 56-day course of daily oral sirolimus. In addition to symptom assessment and physical examination, dermatologic, endoscopic, neurologic (cerebellar), and radiographic assessments were conducted. Inhibition of the mTOR pathway in benign skin and gastrointestinal (GI) lesion was assessed by immunohistochemistry. RESULTS: A total of 18 patients and 16 families were enrolled. PTEN mutations were located at exons 1-8. Regression of skin and GI lesions was observed by dermoscopy or endoscopy. Neurological evaluation showed improvement in cerebellar function score at 1 month. Immunohistochemistry (IHC) analysis in skin and GI benign lesions showed a decrease in the ratio of phosphorylated (p)S6 to total S6 in response to sirolimus. Ratios of pS6K to total S6 at days 14 and 56 were significantly lower than at baseline (p = .0026, p = .00391, respectively). A 56-day course of sirolimus was well tolerated. CONCLUSION: A 56-day course of sirolimus was well tolerated in subjects with Cowden syndrome and was associated with some evidence of improvement in symptoms, skin and GI lesions, cerebellar function, and decreased mTOR signaling.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Hamartoma Syndrome, Multiple/drug therapy , PTEN Phosphohydrolase/genetics , Sirolimus/therapeutic use , Adult , Aged , Anti-Bacterial Agents/pharmacology , Female , Germ-Line Mutation , Humans , Male , Middle Aged , Pilot Projects , Sirolimus/pharmacology , Young Adult
7.
PLoS Genet ; 15(4): e1008034, 2019 04.
Article in English | MEDLINE | ID: mdl-31017901

ABSTRACT

Melanocyte stem cells (McSCs) are the undifferentiated melanocytic cells of the mammalian hair follicle (HF) responsible for recurrent generation of a large number of differentiated melanocytes during each HF cycle. HF McSCs reside in both the CD34+ bulge/lower permanent portion (LPP) and the CD34- secondary hair germ (SHG) regions of the HF during telogen. Using Dct-H2BGFP mice, we separate bulge/LPP and SHG McSCs using FACS with GFP and anti-CD34 to show that these two subsets of McSCs are functionally distinct. Genome-wide expression profiling results support the distinct nature of these populations, with CD34- McSCs exhibiting higher expression of melanocyte differentiation genes and with CD34+ McSCs demonstrating a profile more consistent with a neural crest stem cell. In culture and in vivo, CD34- McSCs regenerate pigmentation more efficiently whereas CD34+ McSCs selectively exhibit the ability to myelinate neurons. CD34+ McSCs, and their counterparts in human skin, may be useful for myelinating neurons in vivo, leading to new therapeutic opportunities for demyelinating diseases and traumatic nerve injury.


Subject(s)
Antigens, CD34/metabolism , Melanocytes/immunology , Melanocytes/physiology , Stem Cells/immunology , Stem Cells/physiology , Animals , Cell Differentiation , Cells, Cultured , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hair Color/physiology , Hair Follicle/cytology , Hair Follicle/physiology , Melanocytes/classification , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Nude , Mice, Transgenic , Myelin Basic Protein/deficiency , Myelin Basic Protein/genetics , Neural Crest/cytology , Neural Crest/immunology , Neural Crest/physiology , Pigmentation/physiology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Regeneration/physiology , Stem Cells/classification
8.
J Invest Dermatol ; 138(7): 1470-1472, 2018 07.
Article in English | MEDLINE | ID: mdl-29941098

ABSTRACT

Mann et al. (2018) use recombinant human tyrosinase to screen for novel inhibitors of pigmentation. They develop thiamidol, a new thiazolyl-resorcinol derivative, that is a submicromolar tyrosinase inhibitor and effective for treating solar lentigines. Thiamidol and established inhibitors of pigmentation exhibit substantially different activities on human and mushroom tyrosinase, supporting use of the human enzyme in high-throughput screens.


Subject(s)
Agaricales , Lentigo , Humans , Monophenol Monooxygenase , Pigmentation
9.
PLoS Biol ; 16(5): e2003648, 2018 05.
Article in English | MEDLINE | ID: mdl-29723194

ABSTRACT

Melanocyte stem cells (McSCs) and mouse models of hair graying serve as useful systems to uncover mechanisms involved in stem cell self-renewal and the maintenance of regenerating tissues. Interested in assessing genetic variants that influence McSC maintenance, we found previously that heterozygosity for the melanogenesis associated transcription factor, Mitf, exacerbates McSC differentiation and hair graying in mice that are predisposed for this phenotype. Based on transcriptome and molecular analyses of Mitfmi-vga9/+ mice, we report a novel role for MITF in the regulation of systemic innate immune gene expression. We also demonstrate that the viral mimic poly(I:C) is sufficient to expose genetic susceptibility to hair graying. These observations point to a critical suppressor of innate immunity, the consequences of innate immune dysregulation on pigmentation, both of which may have implications in the autoimmune, depigmenting disease, vitiligo.


Subject(s)
Adult Stem Cells , Hair Color/immunology , Immunity, Innate , Melanocytes , Microphthalmia-Associated Transcription Factor/physiology , Animals , Female , Gene Expression Regulation , Genetic Predisposition to Disease , Hair Color/genetics , Interferon Type I/metabolism , Mice , Mice, Transgenic , Poly I-C
10.
PLoS One ; 13(2): e0191264, 2018.
Article in English | MEDLINE | ID: mdl-29481571

ABSTRACT

Heat shock protein 90 (HSP90) is a molecular chaperone which stabilizes client proteins with important roles in tumor growth. 17-allylamino-17-demethoxygeldanamycin (17-AAG), an inhibitor of HSP90 ATPase activity, occupies the ATP binding site of HSP90 causing a conformational change which destabilizes client proteins and directs them towards proteosomal degradation. Malignant melanomas have active RAF-MEK-ERK signaling which can occur either through an activating mutation in BRAF (BRAFV600E) or through activation of signal transduction upstream of BRAF. Prior work showed that 17-AAG inhibits cell growth in BRAFV600E and BRAF wildtype (BRAFWT) melanomas, although there were conflicting reports about the dependence of BRAFV600E and BRAFWT upon HSP90 activity for stability. Here, we demonstrate that BRAFWT and CRAF are bound by HSP90 in BRAFWT, NRAS mutant melanoma cells. HSP90 inhibition by 17-AAG inhibits ERK signaling and cell growth by destabilizing CRAF but not BRAFWT in the majority of NRAS mutant melanoma cells. The highly-selective BRAFV600E inhibitor, PLX4032 (vemurafenib), inhibits ERK signaling and cell growth in mutant BRAF melanoma cells, but paradoxically enhances signaling in cells with wild-type BRAF. In our study, we examined whether 17-AAG could inhibit PLX4032-enhanced ERK signaling in BRAFWT melanoma cells. As expected, PLX4032 alone enhanced ERK signaling in the BRAFWT melanoma cell lines Mel-Juso, SK-Mel-2, and SK-Mel-30, and inhibited signaling and cell growth in BRAFV600E A375 cells. However, HSP90 inhibition by 17-AAG inhibited PLX4032-enhanced ERK signaling and inhibited cell growth by destabilizing CRAF. Surprisingly, 17-AAG also stimulated melanin production in SK-Mel-30 cells and enhanced TYRP1 and DCT expression without stimulating TYR production in all three BRAFWT cell lines studied as well as in B16F10 mouse melanoma cells. In vivo, the combination of 17-AAG and cellular immunotherapy directed against Tyrp1 enhanced the inhibition of tumor growth compared to either therapy alone. Our studies support a role for 17-AAG and HSP90 inhibition in enhancing cellular immunotherapy for melanoma.


Subject(s)
Benzoquinones/therapeutic use , Immunotherapy, Adoptive , Indoles/therapeutic use , Lactams, Macrocyclic/therapeutic use , MAP Kinase Signaling System/drug effects , Melanoma, Experimental/drug therapy , Melanoma, Experimental/therapy , Sulfonamides/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Combined Modality Therapy , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Melanoma, Experimental/metabolism , Mice , Mutation , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Vemurafenib
11.
Gene Expr Patterns ; 27: 76-84, 2018 01.
Article in English | MEDLINE | ID: mdl-29061525

ABSTRACT

Melanocytes are neural crest-derived cells that are responsible for mammalian hair follicle (HF) pigmentation. The Dct-LacZ transgenic mouse is extensively used to study melanocyte biology but lacks conditionally-inducible labelling and fluorescent labelling, enabling specific, viable isolation of melanocytes using fluorescence-activated cell sorting (FACS). Here, we have generated a Tet-off bitransgenic mouse model, Dct-H2BGFP, containing Dct-tTA and TRE-H2BGFP transgenes. Characterization of Dct-H2BGFP mice confirmed a pattern of Dct-H2BGFP expression in melanoblasts, melanocyte stem cells (McSCs), and terminally differentiated melanocytes similar to the expression pattern of previously published mouse models Dct-LacZ and iDct-GFP. GFP expression is regulated by doxycycline. GFP is shown to co-localize with melanocyte label-retaining cells (LRCs) identified through BrdU retention. The GFP-expressing cells identified in vivo in the bulge and the secondary hair germ of telogen HFs of Dct-H2BGFP mice express the melanocyte and melanocyte stem cell markers Dct and Kit. Using Dct-H2BGFP mice, we separated GFP-expressing cells from the telogen HF based on FACS and showed that GFP-expressing cells express high levels of Kit and Dct, and lower levels of HF epithelial keratin genes. We also show that GFP-expressing cells express high levels of the melanocyte differentiation genes Tyr, Tyrp1, and Pmel17, further substantiating their identity within the melanocyte lineage. Thus, Dct-H2BGFP mice are not only useful for the in vivo identification of melanocytic cells, but also for isolating them viably and studying their molecular and biological properties.


Subject(s)
Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental/drug effects , Hair Follicle/cytology , Melanocytes/cytology , Neural Crest/cytology , Tetracycline/pharmacology , Animals , Cell Lineage , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Female , Flow Cytometry/methods , Hair Follicle/drug effects , Hair Follicle/metabolism , Lac Operon , Male , Melanocytes/drug effects , Melanocytes/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Neural Crest/drug effects , Neural Crest/metabolism , Oxidoreductases/genetics , Oxidoreductases/metabolism , Pigmentation , Promoter Regions, Genetic , Protein Synthesis Inhibitors/pharmacology , gp100 Melanoma Antigen/genetics , gp100 Melanoma Antigen/metabolism
12.
Dermatol Clin ; 35(1): 85-93, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27890240

ABSTRACT

Melanocytic nevi represent benign clonal proliferations of the melanocytes in the skin that usually remain stable in size and behavior or disappear during life. Infrequently, melanocytic nevi undergo malignant transformation to melanoma. Understanding molecular and cellular mechanisms underlying oncogene-induced senescence should help identify pathways underlying melanoma development, leading to the development of new strategies for melanoma prevention and early detection.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cellular Senescence/genetics , Epigenesis, Genetic/genetics , Melanoma/genetics , Nevus, Pigmented/genetics , Oncogenes/genetics , Skin Neoplasms/genetics , Humans
13.
Pigment Cell Melanoma Res ; 28(3): 330-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25475071

ABSTRACT

Melanocytes undergo rapid and significant changes in their gene expression programs at regular intervals during development and the hair follicle cycle. In melanoma, the gene expression pattern found in normal melanocytes is disrupted. These gene expression patterns are regulated in part by post-translational histone modifications catalyzed by Polycomb group (PcG) proteins, which play a major role in many developmental processes and are often altered in cancer. In this review, we discuss the role of the PcG proteins in stem cell and cancer biology, in general, as well as in melanocyte development and melanomagenesis. Highlights include the discussion of newly identified treatments that target the activity of PcG proteins as well as new developments in the understanding of the role that these proteins play in melanocyte biology.


Subject(s)
Epigenesis, Genetic , Melanocytes/metabolism , Melanoma/metabolism , Polycomb-Group Proteins/metabolism , Animals , Humans , Models, Biological , Molecular Targeted Therapy
14.
Pigment Cell Melanoma Res ; 27(3): 454-64, 2014 May.
Article in English | MEDLINE | ID: mdl-24483290

ABSTRACT

We examined nevi and melanomas in 10 xeroderma pigmentosum (XP) patients with defective DNA repair. The lesions had a lentiginous appearance with markedly increased numbers of melanocytes. Using laser capture microdissection, we performed DNA sequencing of 18 benign and atypical nevi and 75 melanomas (melanoma in situ and invasive melanomas). The nevi had a similar high frequency of PTEN mutations as melanomas [61% (11/18) versus 53% (39/73)]. Both had a very high proportion of UV-type mutations (occurring at adjacent pyrimidines) [91% (10/11) versus 92% (36/39)]. In contrast to melanomas in the general population, the frequency of BRAF mutations (11%, 7/61), NRAS mutations (21%, 13/62), and KIT mutations (21%, 6/28) in XP melanomas was lower than for PTEN. Phospho-S6 immunostaining indicated activation of the mTOR pathway in the atypical nevi and melanomas. Thus, the clinical and histological appearances and the molecular pathology of these UV-related XP nevi and melanomas were different from nevi and melanomas in the general population.


Subject(s)
Melanoma/genetics , Mutation , Nevus, Pigmented/genetics , PTEN Phosphohydrolase/genetics , Skin Neoplasms/genetics , Xeroderma Pigmentosum/complications , Adult , DNA Mutational Analysis , DNA, Neoplasm/genetics , Dermoscopy , Female , GTP Phosphohydrolases/genetics , Humans , Loss of Heterozygosity , Male , Melanoma/etiology , Melanoma/pathology , Membrane Proteins/genetics , Middle Aged , Neoplasm Proteins/genetics , Neoplasms, Radiation-Induced/genetics , Neoplasms, Radiation-Induced/pathology , Nevus, Pigmented/etiology , Nevus, Pigmented/pathology , Oncogenes , Precancerous Conditions/etiology , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-kit/genetics , Skin Neoplasms/etiology , Skin Neoplasms/pathology , Sunlight/adverse effects , TOR Serine-Threonine Kinases/physiology , Ultraviolet Rays/adverse effects , Xeroderma Pigmentosum/genetics , Young Adult
15.
AAPS J ; 15(4): 1064-71, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23904153

ABSTRACT

Epigenetics is an important emerging area for study of mechanisms of cancer prevention. In recent years, it has been realized that cancer prevention agents, derived from natural dietary sources, impact cancer cell survival by modulating epigenetic processes. In the present manuscript, we review key epigenetic regulatory mechanisms and examine the impact of sulforaphane and green tea polyphenols on these processes. We also discuss available information on the epigenetics in the context of skin cancer. These studies indicate that diet-derived chemopreventive agents modulate DNA methylation status and histone modification via multiple processes and point to additional areas for study of epigenetic mechanisms in skin cancer.


Subject(s)
Anticarcinogenic Agents/administration & dosage , Epigenesis, Genetic/physiology , Skin Neoplasms/diet therapy , Skin Neoplasms/prevention & control , Animals , Anticarcinogenic Agents/isolation & purification , Humans , Isothiocyanates/administration & dosage , Isothiocyanates/isolation & purification , Polyphenols/administration & dosage , Polyphenols/isolation & purification , Skin Neoplasms/genetics , Sulfoxides
16.
Pigment Cell Melanoma Res ; 26(1): 78-87, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23020089

ABSTRACT

The human deafness-pigmentation syndromes, Waardenburg syndrome (WS) type 2a, and Tietz syndrome are characterized by profound deafness but only partial cutaneous pigmentary abnormalities. Both syndromes are caused by mutations in MITF. To illuminate differences between cutaneous and otic melanocytes in these syndromes, their development and survival in heterozygous Microphthalmia-White (Mitf(Mi-wh) /+) mice were studied and hearing function of these mice characterized. Mitf(Mi-wh) /+ mice have a profound hearing deficit, characterized by elevated auditory brainstem response thresholds, reduced distortion product otoacoustic emissions, absent endocochlear potential, loss of outer hair cells, and stria vascularis abnormalities. Mitf(Mi-wh) /+ embryos have fewer melanoblasts during embryonic development than their wild-type littermates. Although cochlear melanocytes are present at birth, they disappear from the Mitf(Mi-wh) /+ cochlea between P1 and P7. These findings may provide insight into the mechanism of melanocyte and hearing loss in human deafness-pigmentation syndromes such as WS and Tietz syndrome and illustrate differences between otic and follicular melanocytes.


Subject(s)
Albinism, Oculocutaneous/physiopathology , Deafness/physiopathology , Hearing/physiology , Heterozygote , Microphthalmia-Associated Transcription Factor/genetics , Waardenburg Syndrome/physiopathology , Action Potentials/physiology , Albinism, Oculocutaneous/genetics , Albinism, Oculocutaneous/pathology , Animals , Animals, Newborn , Deafness/genetics , Deafness/pathology , Disease Models, Animal , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Evoked Potentials, Auditory, Brain Stem/physiology , Hair Cells, Auditory, Outer/metabolism , Hair Cells, Auditory, Outer/pathology , Humans , Melanocytes/metabolism , Melanocytes/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Otoacoustic Emissions, Spontaneous/physiology , Stria Vascularis/metabolism , Stria Vascularis/pathology , Waardenburg Syndrome/genetics , Waardenburg Syndrome/pathology
17.
Mol Cancer Res ; 9(4): 418-29, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21383005

ABSTRACT

Polycomb group (PcG) proteins such as Enhancer of zeste homolog 2 (EZH2) are epigenetic transcriptional repressors that function through recognition and modification of histone methylation and chromatin structure. Targets of PcG include cell cycle regulatory proteins which govern cell cycle progression and cellular senescence. Senescence is a characteristic of melanocytic nevi, benign melanocytic proliferations that can be precursors of malignant melanoma. In this study, we report that EZH2, which we find absent in melanocytic nevi but expressed in many or most metastatic melanoma cells, functionally suppresses the senescent state in human melanoma cells. EZH2 depletion in melanoma cells inhibits cell proliferation, restores features of a cellular senescence phenotype, and inhibits growth of melanoma xenografts in vivo. p21/CDKN1A is activated upon EZH2 knockdown in a p53-independent manner and contributes substantially to cell cycle arrest and induction of a senescence phenotype. EZH2 depletion removes histone deacetylase 1 (HDAC1) from the CDKN1A transcriptional start site and downstream region, enhancing histone 3 acetylation globally and at CDKN1A. This results in recruitment of RNA polymerase II, leading to p21/CDKN1A activation. Depletion of EZH2 synergistically activates p21/CDKN1A expression in combination with the HDAC inhibitor trichostatin A. Since melanomas often retain wild-type p53 function activating p21, our findings describe a novel mechanism whereby EZH2 activation during tumor progression represses p21, leading to suppression of cellular senescence and enhanced tumorigenicity.


Subject(s)
Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p21/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Melanoma/pathology , Nevus, Pigmented/pathology , Skin Neoplasms/pathology , Transcription Factors/metabolism , Animals , Cell Cycle , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA-Binding Proteins/genetics , Enhancer of Zeste Homolog 2 Protein , Histone Deacetylase 1/metabolism , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Melanoma/genetics , Melanoma/metabolism , Mice , Nevus, Pigmented/genetics , Nevus, Pigmented/metabolism , Polycomb Repressive Complex 2 , RNA Polymerase II/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Transcription Factors/genetics
19.
Nature ; 469(7331): 548-53, 2011 Jan 27.
Article in English | MEDLINE | ID: mdl-21248750

ABSTRACT

Cutaneous malignant melanoma is a highly aggressive and frequently chemoresistant cancer, the incidence of which continues to rise. Epidemiological studies show that the major aetiological melanoma risk factor is ultraviolet (UV) solar radiation, with the highest risk associated with intermittent burning doses, especially during childhood. We have experimentally validated these epidemiological findings using the hepatocyte growth factor/scatter factor transgenic mouse model, which develops lesions in stages highly reminiscent of human melanoma with respect to biological, genetic and aetiological criteria, but only when irradiated as neonatal pups with UVB, not UVA. However, the mechanisms underlying UVB-initiated, neonatal-specific melanomagenesis remain largely unknown. Here we introduce a mouse model permitting fluorescence-aided melanocyte imaging and isolation following in vivo UV irradiation. We use expression profiling to show that activated neonatal skin melanocytes isolated following a melanomagenic UVB dose bear a distinct, persistent interferon response signature, including genes associated with immunoevasion. UVB-induced melanocyte activation, characterized by aberrant growth and migration, was abolished by antibody-mediated systemic blockade of interferon-γ (IFN-γ), but not type-I interferons. IFN-γ was produced by macrophages recruited to neonatal skin by UVB-induced ligands to the chemokine receptor Ccr2. Admixed recruited skin macrophages enhanced transplanted melanoma growth by inhibiting apoptosis; notably, IFN-γ blockade abolished macrophage-enhanced melanoma growth and survival. IFN-γ-producing macrophages were also identified in 70% of human melanomas examined. Our data reveal an unanticipated role for IFN-γ in promoting melanocytic cell survival/immunoevasion, identifying a novel candidate therapeutic target for a subset of melanoma patients.


Subject(s)
Interferon-gamma/metabolism , Melanocytes/metabolism , Melanoma/physiopathology , Ultraviolet Rays , Animals , Disease Models, Animal , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental/radiation effects , Humans , Macrophages/metabolism , Macrophages/radiation effects , Male , Melanocytes/radiation effects , Mice
20.
Clin Plast Surg ; 37(1): 169-76, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19914467

ABSTRACT

The future of melanoma research is promising. Specific mechanisms leading to oncogenic transformation in melanoma development have been identified, and are likely to produce new targets for melanoma therapy. Also, advances in melanoma research will result from melanoma investigators co-opting approaches used to study other malignancies in which progress has been made more rapidly. Systematic roadblocks limiting advances in melanoma research relative to other malignancies are being addressed in a formal manner. The public and public officials are increasingly becoming aware of the need for more dedicated efforts to address the challenges of research on this malignancy.


Subject(s)
Biomedical Research , Melanoma , Skin Neoplasms , Comparative Genomic Hybridization , Gene Expression Profiling , Genetic Predisposition to Disease , Humans , Melanoma/genetics , Melanoma/pathology , Mutation , Neoplastic Stem Cells/physiology , Skin Neoplasms/genetics , Skin Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...