Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
2.
J Clin Invest ; 132(7)2022 04 01.
Article in English | MEDLINE | ID: mdl-35143422

ABSTRACT

Merkel cell carcinoma (MCC) is an aggressive neuroendocrine skin cancer that frequently carries an integrated Merkel cell polyomavirus (MCPyV) genome and expresses viral transforming antigens (TAgs). MCC tumor cells also express signature genes detected in skin-resident, postmitotic Merkel cells, including atonal bHLH transcription factor 1 (ATOH1), which is required for Merkel cell development from epidermal progenitors. We now report the use of in vivo cellular reprogramming, using ATOH1, to drive MCC development from murine epidermis. We generated mice that conditionally expressed MCPyV TAgs and ATOH1 in epidermal cells, yielding microscopic collections of proliferating MCC-like cells arising from hair follicles. Immunostaining of these nascent tumors revealed p53 accumulation and apoptosis, and targeted deletion of transformation related protein 53 (Trp53) led to development of gross skin tumors with classic MCC histology and marker expression. Global transcriptome analysis confirmed the close similarity of mouse and human MCCs, and hierarchical clustering showed conserved upregulation of signature genes. Our data establish that expression of MCPyV TAgs in ATOH1-reprogrammed epidermal cells and their neuroendocrine progeny initiates hair follicle-derived MCC tumorigenesis in adult mice. Moreover, progression to full-blown MCC in this model requires loss of p53, mimicking the functional inhibition of p53 reported in human MCPyV-positive MCCs.


Subject(s)
Carcinoma, Merkel Cell , Merkel cell polyomavirus , Polyomavirus Infections , Skin Neoplasms , Tumor Virus Infections , Animals , Antigens, Viral , Antigens, Viral, Tumor/genetics , Antigens, Viral, Tumor/metabolism , Carcinoma, Merkel Cell/genetics , Carcinoma, Merkel Cell/metabolism , Carcinoma, Merkel Cell/pathology , Cellular Reprogramming , Merkel cell polyomavirus/genetics , Mice , Polyomavirus Infections/genetics , Polyomavirus Infections/pathology , Skin Neoplasms/pathology , Tumor Virus Infections/genetics , Tumor Virus Infections/pathology
3.
Oncogene ; 41(1): 37-45, 2022 01.
Article in English | MEDLINE | ID: mdl-34667274

ABSTRACT

Merkel cell carcinoma (MCC) is a neuroendocrine tumor either induced by integration of the Merkel cell polyomavirus into the cell genome or by accumulation of UV-light-associated mutations (VP-MCC and UV-MCC). Whether VP- and UV-MCC have the same or different cellular origins is unclear; with mesenchymal or epidermal origins discussed. DNA-methylation patterns have a proven utility in determining cellular origins of cancers. Therefore, we used this approach to uncover evidence regarding the cell of origin of classical VP- and UV-MCC cell lines, i.e., cell lines with a neuroendocrine growth pattern (n = 9 and n = 4, respectively). Surprisingly, we observed high global similarities in the DNA-methylation of UV- and VP-MCC cell lines. CpGs of lower methylation in VP-MCC cell lines were associated with neuroendocrine marker genes such as SOX2 and INSM1, or linked to binding sites of EZH2 and SUZ12 of the polycomb repressive complex 2, i.e., genes with an impact on carcinogenesis and differentiation of neuroendocrine cancers. Thus, the observed differences appear to be rooted in viral compared to mutation-driven carcinogenesis rather than distinct cells of origin. To test this hypothesis, we used principal component analysis, to compare DNA-methylation data from different epithelial and non-epithelial neuroendocrine cancers and established a scoring model for epithelial and neuroendocrine characteristics. Subsequently, we applied this scoring model to the DNA-methylation data of the VP- and UV-MCC cell lines, revealing that both clearly scored as epithelial cancers. In summary, our comprehensive analysis of DNA-methylation suggests a common epithelial origin of UV- and VP-MCC cell lines.


Subject(s)
Carcinoma, Merkel Cell/genetics , DNA Methylation/genetics , High-Throughput Screening Assays/methods , Tumor Virus Infections/genetics , Humans
4.
Mod Pathol ; 35(4): 506-514, 2022 04.
Article in English | MEDLINE | ID: mdl-34593967

ABSTRACT

Merkel cell carcinoma (MCC) is an aggressive cutaneous neuroendocrine carcinoma without a known dysplastic precursor. In some cases, MCC is associated with SCCIS in the overlying epidermis; however, the MCC and SCCIS populations display strikingly different morphologies, and thus far a relationship between these components has not been demonstrated. To better understand the relationship between these distinct tumor cell populations, we evaluated 7 pairs of MCC-SCCIS for overlapping genomic alterations by cancer profiling panel. A subset was further characterized by transcriptional profiling and immunohistochemistry. In 6 of 7 MCC-SCCIS pairs there was highly significant mutational overlap including shared TP53 and/or RB1 mutations. In some cases, oncogenic events previously implicated in MCC (MYCL gain, MDM4 gain, HRAS mutation) were detected in both components. Although FBXW7 mutations were enriched in MCC, no gene mutation was unique to the MCC component across all cases. Transcriptome analysis identified 2736 differentially expressed genes between MCC and SCCIS. Genes upregulated in the MCC component included Polycomb repressive complex targets; downregulated transcripts included epidermal markers, and immune genes such as HLA-A. Immunohistochemical studies revealed increased expression of SOX2 in the MCC component, with diminished H3K27Me3, Rb, and HLA-A expression. In summary, MCC-SCCIS pairs demonstrate clonal relatedness. The shift to neuroendocrine phenotype is associated with loss of Rb protein expression, decrease in global H3K27Me3, and increased expression of Merkel cell genes such as SOX2. Our findings suggest an epidermal origin of MCC in this setting, and to our knowledge provide the first molecular evidence that intraepithelial squamous dysplasia may represent a direct precursor for small cell carcinoma.


Subject(s)
Carcinoma, Merkel Cell , Carcinoma, Squamous Cell , Skin Neoplasms , Carcinoma, Merkel Cell/genetics , Carcinoma, Merkel Cell/pathology , Carcinoma, Squamous Cell/genetics , Cell Cycle Proteins , Genomics , Humans , Immunohistochemistry , Proto-Oncogene Proteins , Skin Neoplasms/genetics , Skin Neoplasms/pathology
5.
J Invest Dermatol ; 142(3 Pt A): 641-652, 2022 03.
Article in English | MEDLINE | ID: mdl-34474081

ABSTRACT

Merkel cell carcinoma (MCC) is an aggressive cutaneous neuroendocrine carcinoma that is classified as Merkel cell polyomavirus-positive (virus positive [VP]) or Merkel cell polyomavirus-negative (virus negative [VN]). Epigenetic changes, such as DNA methylation, can alter gene expression and influence cancer progression. However, patterns of DNA methylation and the therapeutic efficacy of hypomethylating agents have not been fully explored in MCC. We characterized genome-wide DNA methylation in 16 MCC cell lines from both molecular subclasses in comparison with other cancer types and found that the overall profile of MCC is similar to that of small-cell lung carcinoma. Comparison of VP MCC with VN MCC revealed 2,260 differentially methylated positions. The hypomethylating agent decitabine upregulated the expression of antigen-presenting machinery in MCC cell lines and stimulated membrane expression of HLA-A in VP and VN MCC xenograft tumors. Decitabine also induced prominent caspase- and large T antigen‒independent cell death in VP MCC, whereas VN MCC cell lines displayed decreased proliferation without increased cell death. In mouse xenografts, decitabine significantly decreased the size of VP tumors but not that of VN tumors. Our findings indicate that viral status predicts genomic methylation patterns in MCC and that decitabine may be therapeutically effective against MCC through antiproliferative effects, cell death, and increased immune recognition.


Subject(s)
Carcinoma, Merkel Cell , Merkel cell polyomavirus , Polyomavirus Infections , Skin Neoplasms , Tumor Virus Infections , Animals , Carcinoma, Merkel Cell/drug therapy , Carcinoma, Merkel Cell/genetics , Carcinoma, Merkel Cell/pathology , DNA Methylation , Decitabine/pharmacology , Decitabine/therapeutic use , Humans , Merkel cell polyomavirus/genetics , Mice , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Tumor Virus Infections/genetics
6.
Carcinogenesis ; 42(8): 1100-1109, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34117865

ABSTRACT

Uncontrolled activation of the Hedgehog (Hh) signaling pathway, operating through GLI transcription factors, plays a central role in the pathogenesis of cutaneous basal cell carcinoma and contributes to the development of several malignancies arising in extracutaneous sites. We now report that K5-tTA;tetO-Gli2 bitransgenic mice develop distinctive epithelial tumors within their jaws. These tumors consist of large masses of highly proliferative, monomorphous, basaloid cells with scattered foci of keratinization and central necrosis, mimicking human basaloid squamous cell carcinoma (BSCC), an aggressive upper aerodigestive tract tumor. Like human BSCC, these tumors express epidermal basal keratins and differentiation-specific keratins within squamous foci. Mouse BSCCs express high levels of Gli2 and Hh target genes, including Gli1 and Ptch1, which we show are also upregulated in a subset of human BSCCs. Mouse BSCCs appear to arise from distinct epithelial sites, including the gingival junctional epithelium and epithelial rests of Malassez, a proposed stem cell compartment. Although Gli2 transgene expression is restricted to epithelial cells, we also detect striking alterations in bone adjacent to BSCCs, with activated osteoblasts, osteoclasts and osteal macrophages, indicative of active bone remodeling. Gli2 transgene inactivation resulted in rapid BSCC regression and reversal of the bone remodeling phenotype. This first-reported mouse model of BSCC supports the concept that uncontrolled Hh signaling plays a central role in the pathogenesis of a subset of human BSCCs, points to Hh/GLI2 signaling as a potential therapeutic target and provides a powerful new tool for probing the mechanistic underpinnings of tumor-associated bone remodeling.


Subject(s)
Bone Remodeling , Carcinoma, Squamous Cell/pathology , Hedgehog Proteins/metabolism , Nuclear Proteins/metabolism , Skin Neoplasms/pathology , Zinc Finger Protein Gli2/metabolism , Animals , Carcinoma, Squamous Cell/metabolism , Humans , Mice , Mice, Transgenic , Skin Neoplasms/metabolism
7.
J Clin Invest ; 131(12)2021 06 15.
Article in English | MEDLINE | ID: mdl-33945506

ABSTRACT

Cutaneous melanoma remains the most lethal skin cancer, and ranks third among all malignancies in terms of years of life lost. Despite the advent of immune checkpoint and targeted therapies, only roughly half of patients with advanced melanoma achieve a durable remission. Sirtuin 5 (SIRT5) is a member of the sirtuin family of protein deacylases that regulates metabolism and other biological processes. Germline Sirt5 deficiency is associated with mild phenotypes in mice. Here we showed that SIRT5 was required for proliferation and survival across all cutaneous melanoma genotypes tested, as well as uveal melanoma, a genetically distinct melanoma subtype that arises in the eye and is incurable once metastatic. Likewise, SIRT5 was required for efficient tumor formation by melanoma xenografts and in an autochthonous mouse Braf Pten-driven melanoma model. Via metabolite and transcriptomic analyses, we found that SIRT5 was required to maintain histone acetylation and methylation levels in melanoma cells, thereby promoting proper gene expression. SIRT5-dependent genes notably included MITF, a key lineage-specific survival oncogene in melanoma, and the c-MYC proto-oncogene. SIRT5 may represent a druggable genotype-independent addiction in melanoma.


Subject(s)
Chromatin/enzymology , Melanoma, Experimental/enzymology , Melanoma/enzymology , Sirtuins/metabolism , Skin Neoplasms/enzymology , Animals , Chromatin/genetics , Melanoma/genetics , Melanoma/pathology , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Sirtuins/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Melanoma, Cutaneous Malignant
8.
JCI Insight ; 4(8)2019 04 18.
Article in English | MEDLINE | ID: mdl-30996136

ABSTRACT

Autoimmune disease is 4 times more common in women than men. This bias is largely unexplained. Female skin is "autoimmunity prone," showing upregulation of many proinflammatory genes, even in healthy women. We previously identified VGLL3 as a putative transcription cofactor enriched in female skin. Here, we demonstrate that skin-directed overexpression of murine VGLL3 causes a severe lupus-like rash and systemic autoimmune disease that involves B cell expansion, autoantibody production, immune complex deposition, and end-organ damage. Excess epidermal VGLL3 drives a proinflammatory gene expression program that overlaps with both female skin and cutaneous lupus. This includes increased B cell-activating factor (BAFF), the only current biologic target in systemic lupus erythematosus (SLE); IFN-κ, a key inflammatory mediator in cutaneous lupus; and CXCL13, a biomarker of early-onset SLE and renal involvement. Our results demonstrate that skin-targeted overexpression of the female-biased factor VGLL3 is sufficient to drive cutaneous and systemic autoimmune disease that is strikingly similar to SLE. This work strongly implicates VGLL3 as a pivotal orchestrator of sex-biased autoimmunity.


Subject(s)
Autoimmunity/genetics , Gene Expression Regulation/immunology , Lupus Erythematosus, Cutaneous/immunology , Lupus Erythematosus, Systemic/immunology , Transcription Factors/metabolism , Animals , Disease Models, Animal , Female , Humans , Lupus Erythematosus, Cutaneous/genetics , Lupus Erythematosus, Cutaneous/pathology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/pathology , Male , Mice , Mice, Transgenic , Sex Factors , Skin/immunology , Skin/pathology , Transcription Factors/genetics
9.
Neoplasia ; 21(3): 322-330, 2019 03.
Article in English | MEDLINE | ID: mdl-30797188

ABSTRACT

Studies on the efficacy of small molecule inhibitors in Merkel cell carcinoma (MCC) have been limited and largely inconclusive. In this study, we investigated the therapeutic potential of a potent BET degrader, BETd-246, in the treatment of MCC. We found that MCC cell lines were significantly more sensitive to BETd-246 than to BET inhibitor treatment. Therapeutic targeting of BET proteins resulted in a loss of "MCC signature" genes but not MYC expression as previously described irrespective of Merkel cell polyomavirus (MCPyV) status. In MCPyV+ MCC cells, BETd-246 alone suppressed downstream targets in the MCPyV-LT Ag axis. We also found enrichment of HOX and cell cycle genes in MCPyV- MCC cell lines that were intrinsically resistant to BETd-246. Our findings uncover a requirement for BET proteins in maintaining MCC lineage identity and point to the potential utility of BET degraders for treating MCC.


Subject(s)
Carcinoma, Merkel Cell/metabolism , Proteins/antagonists & inhibitors , Proteins/metabolism , Skin Neoplasms/metabolism , Acetanilides/pharmacology , Antigens, Viral, Tumor/genetics , Antigens, Viral, Tumor/metabolism , Carcinoma, Merkel Cell/drug therapy , Carcinoma, Merkel Cell/etiology , Carcinoma, Merkel Cell/pathology , Cell Cycle/genetics , Cell Line, Tumor , Dose-Response Relationship, Drug , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genes, Homeobox , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Merkel cell polyomavirus/physiology , Polyomavirus Infections/complications , Polyomavirus Infections/virology , Proteolysis , Skin Neoplasms/drug therapy , Skin Neoplasms/etiology , Skin Neoplasms/pathology , Transcriptome
10.
J Invest Dermatol ; 139(4): 807-817, 2019 04.
Article in English | MEDLINE | ID: mdl-30399362

ABSTRACT

The majority of Merkel cell carcinoma, a highly aggressive neuroendocrine cancer of the skin, is associated with Merkel cell polyomavirus infection. Polyomavirus binding, internalization, and infection are mediated by glycosphingolipids. Besides receptor function, bioactive sphingolipids are increasingly recognized as potent regulators of several hallmarks of cancer. Merkel cell polyomavirus+ and Merkel cell polyomavirus- cells express serine palmitoyl transferase subunits and sphingosine kinase (SK) 1/2 mRNA. Induced expression of Merkel cell polyomavirus-large tumor antigen in human lung fibroblasts resulted in upregulation of SPTLC1-3 and SK 1/2 expression. Therefore, we exploited pharmacological inhibition of sphingolipid metabolism as an option to interfere with proliferation of Merkel cell polyomavirus+ Merkel cell carcinoma cell lines. We used myriocin (a serine palmitoyl transferase antagonist) and two SK inhibitors (SKI-II and ABC294640). In MKL-1 and WaGa cells myriocin decreased cellular ceramide, sphingomyelin, and sphingosine-1-phosphate content. SKI-II increased ceramide species but decreased sphingomyelin and sphingosine-1-phosphate concentrations. Aberrant sphingolipid homeostasis was associated with reduced cell viability, increased necrosis, procaspase-3 and PARP processing, caspase-3 activity, and decreased AKTS473 phosphorylation. Myriocin and SKI-II decreased tumor size and Ki-67 staining of xenografted MKL-1 and WaGa tumors on the chorioallantoic membrane. Our data suggest that pharmacological inhibition of sphingolipid synthesis could represent a potential therapeutic approach in Merkel cell carcinoma.


Subject(s)
Carcinoma, Merkel Cell/drug therapy , Fatty Acids, Monounsaturated/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Polyomavirus Infections/drug therapy , Serine C-Palmitoyltransferase/antagonists & inhibitors , Skin Neoplasms/drug therapy , Tumor Virus Infections/drug therapy , Carcinoma, Merkel Cell/metabolism , Carcinoma, Merkel Cell/pathology , Cell Count , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunosuppressive Agents/pharmacology , Merkel cell polyomavirus/immunology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Polyomavirus Infections/metabolism , Polyomavirus Infections/pathology , RNA, Neoplasm/genetics , Serine C-Palmitoyltransferase/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tumor Virus Infections/metabolism , Tumor Virus Infections/pathology
11.
Clin Cancer Res ; 24(23): 5873-5882, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30061360

ABSTRACT

PURPOSE: Merkel cell carcinoma (MCC) is an aggressive skin cancer with neuroendocrine differentiation. There is an unmet need for MCC-specific blood-based surrogate biomarkers of tumor burden; circulating cell-free miRNA may serve this purpose. EXPERIMENTAL DESIGN: Expression of miR-375 was quantified in 24 MCC and 23 non-MCC cell lines, 67 MCC and 58 non-MCC tumor tissues, sera of 2 preclinical MCC models, and sera of 109 patients with MCC and 30 healthy controls by nCounter human-v2-miRNA expression or miR-375-specific real-time PCR assays. The patients' sera consisted of two retrospective (discovery and training) and two prospective (validation) cohorts. RESULTS: miR-375 expression was high in MCC cell lines and tissues compared with non-MCCs. It was readily detected in MCC-conditioned medium and sera of preclinical models bearing MCC xenografts. miR-375 levels were higher in sera from tumor-bearing patients with MCC than in tumor-free patients or healthy controls (P < 0.0005). Moreover, miR-375 serum levels correlated with tumor stage in tumor-bearing (P = 0.037) but not in tumor-free (P = 0.372) patients with MCC. miR-375 serum level showed high diagnostic accuracy to discriminate tumor-bearing and tumor-free patients with MCC as demonstrated by ROC curve analysis in the retrospective cohorts (AUC = 0.954 and 0.800) as well as in the prospective cohorts (AUC = 0.929 and 0.959). miR-375 serum level reflected dynamic changes in tumor burden of patients with MCC during therapeutic interventions. CONCLUSIONS: Circulating cell-free miR-375 proved as a surrogate marker for tumor burden in MCC without restriction to polyomavirus positivity; it thus appears to be useful for therapy monitoring and the follow-up of patients with MCC.


Subject(s)
Biomarkers, Tumor , Carcinoma, Merkel Cell/blood , Carcinoma, Merkel Cell/genetics , Circulating MicroRNA , MicroRNAs/genetics , Animals , Carcinoma, Merkel Cell/diagnosis , Case-Control Studies , Cell Line, Tumor , Chick Embryo , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Positron Emission Tomography Computed Tomography , Prognosis , ROC Curve , Real-Time Polymerase Chain Reaction , Tumor Burden , Xenograft Model Antitumor Assays
12.
Cancer Cell ; 33(2): 229-243.e4, 2018 02 12.
Article in English | MEDLINE | ID: mdl-29395868

ABSTRACT

Hedgehog (Hh) pathway inhibitors such as vismodegib are highly effective for treating basal cell carcinoma (BCC); however, residual tumor cells frequently persist and regenerate the primary tumor upon drug discontinuation. Here, we show that BCCs are organized into two molecularly and functionally distinct compartments. Whereas interior Hh+/Notch+ suprabasal cells undergo apoptosis in response to vismodegib, peripheral Hh+++/Notch- basal cells survive throughout treatment. Inhibiting Notch specifically promotes tumor persistence without causing drug resistance, while activating Notch is sufficient to regress already established lesions. Altogether, these findings suggest that the three-dimensional architecture of BCCs establishes a natural hierarchy of drug response in the tumor and that this hierarchy can be overcome, for better or worse, by modulating Notch.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Basal Cell/drug therapy , Receptors, Notch/drug effects , Signal Transduction/drug effects , Skin Neoplasms/drug therapy , Apoptosis/drug effects , Carcinoma, Basal Cell/pathology , Hedgehog Proteins/drug effects , Hedgehog Proteins/metabolism , Humans , Receptors, Notch/metabolism , Skin Neoplasms/pathology
13.
Cancer Res ; 77(12): 3151-3157, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28512245

ABSTRACT

Merkel cell carcinoma (MCC) tumor cells express several markers detected in normal Merkel cells, a nonproliferative population of neuroendocrine cells that arise from epidermis. MCCs frequently contain Merkel cell polyomavirus (MCPyV) DNA and express viral transforming antigens, sT and tLT, but the role of these putative oncogenes in MCC development, and this tumor's cell of origin, are unknown. Using a panel of preterm transgenic mice, we show that epidermis-targeted coexpression of sT and the cell fate-determinant atonal bHLH transcription factor 1 (ATOH1) leads to development of widespread cellular aggregates, with histology and marker expression mimicking that of human intraepidermal MCC. The MCC-like tumor phenotype was dependent on the FBXW7-binding domain of sT, but not the sT-PP2A binding domain. Coexpression of MCPyV tLT did not appreciably alter the phenotype driven by either sT or sT combined with ATOH1. MCPyV sT, when coexpressed with ATOH1, is thus sufficient to initiate development of epidermis-derived MCC-like tumors in mice. Cancer Res; 77(12); 3151-7. ©2017 AACR.


Subject(s)
Antigens, Viral, Tumor/metabolism , Carcinoma, Merkel Cell/virology , Polyomavirus Infections/virology , Skin Neoplasms/virology , Tumor Virus Infections/virology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Merkel Cell/metabolism , Disease Models, Animal , Humans , Immunohistochemistry , Merkel cell polyomavirus/immunology , Mice , Mice, Transgenic , Polymerase Chain Reaction , Polyomavirus Infections/metabolism , Skin Neoplasms/metabolism , Tumor Virus Infections/metabolism
14.
Mod Pathol ; 29(3): 240-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26743471

ABSTRACT

Merkel cell carcinoma is a rare but highly aggressive cutaneous neuroendocrine carcinoma. Cytokeratin 20 (CK20) is expressed in ~95% of Merkel cell carcinomas and is useful for distinction from morphologically similar entities including metastatic small-cell lung carcinoma. Lack of CK20 expression may make diagnosis of Merkel cell carcinoma more challenging, and has unknown biological significance. Approximately 80% of CK20-positive Merkel cell carcinomas are associated with the oncogenic Merkel cell polyomavirus. Merkel cell carcinomas lacking Merkel cell polyomavirus display distinct genetic changes from Merkel cell polyomavirus-positive Merkel cell carcinoma, including RB1 inactivating mutations. Unlike CK20-positive Merkel cell carcinoma, the majority of CK20-negative Merkel cell carcinomas are Merkel cell polyomavirus-negative, suggesting CK20-negative Merkel cell carcinomas predominantly arise through virus-independent pathway(s) and may harbor additional genetic differences from conventional Merkel cell carcinoma. Hence, we analyzed 15 CK20-negative Merkel cell carcinoma tumors (10 Merkel cell polyomavirus-negative, four Merkel cell polyomavirus-positive, and one undetermined) using the Ion Ampliseq Comprehensive Cancer Panel, which assesses copy number alterations and mutations in 409 cancer-relevant genes. Twelve tumors displayed prioritized high-level chromosomal gains or losses (average 1.9 per tumor). Non-synonymous high-confidence somatic mutations were detected in 14 tumors (average 11.9 per tumor). Assessing all somatic coding mutations, an ultraviolet-signature mutational profile was present, and more prevalent in Merkel cell polyomavirus-negative tumors. Recurrent deleterious tumor suppressor mutations affected TP53 (9/15, 60%), RB1 (3/15, 20%), and BAP1 (2/15, 13%). Oncogenic activating mutations included PIK3CA (3/15, 20%), AKT1 (1/15, 7%) and EZH2 (1/15, 7%). In conclusion, CK20-negative Merkel cell carcinoma display overlapping genetic changes with CK20-positive Merkel cell carcinoma, including RB1 mutations restricted to Merkel cell polyomavirus-negative tumors. However, some CK20-negative Merkel cell carcinomas harbor mutations not previously described in Merkel cell carcinoma. Hence, CK20-negative Merkel cell carcinomas harbor diverse oncogenic drivers which may represent therapeutic targets in individual tumors.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Merkel Cell/genetics , Mutation , Skin Neoplasms/genetics , Aged , Aged, 80 and over , DNA Mutational Analysis/methods , Female , High-Throughput Nucleotide Sequencing/methods , Humans , Keratin-20/genetics , Male , Middle Aged , Retinoblastoma Binding Proteins/genetics , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/genetics
15.
Cell Stem Cell ; 16(4): 400-12, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25842978

ABSTRACT

Basal cell carcinoma (BCC) is characterized by frequent loss of PTCH1, leading to constitutive activation of the Hedgehog pathway. Although the requirement for Hedgehog in BCC is well established, the identity of disease-initiating cells and the compartments in which they reside remain controversial. By using several inducible Cre drivers to delete Ptch1 in different cell compartments in mice, we show here that multiple hair follicle stem cell populations readily develop BCC-like tumors. In contrast, stem cells within the interfollicular epidermis do not efficiently form tumors. Notably, we observed that innervated Gli1-expressing progenitors within mechanosensory touch dome epithelia are highly tumorigenic. Sensory nerves activate Hedgehog signaling in normal touch domes, while denervation attenuates touch dome-derived tumors. Together, our studies identify varying tumor susceptibilities among different stem cell populations in the skin, highlight touch dome epithelia as "hot spots" for tumor formation, and implicate cutaneous nerves as mediators of tumorigenesis.


Subject(s)
Carcinoma, Basal Cell/metabolism , Epithelial Cells/physiology , Hair Follicle/physiology , Merkel Cells/physiology , Musculocutaneous Nerve/physiology , Pluripotent Stem Cells/physiology , Receptors, Cell Surface/metabolism , Skin Neoplasms/metabolism , Animals , Carcinogenesis/genetics , Carcinoma, Basal Cell/pathology , Denervation , Hair Follicle/innervation , Hedgehog Proteins/metabolism , Humans , Kruppel-Like Transcription Factors/metabolism , Mechanoreceptors/metabolism , Mechanotransduction, Cellular/genetics , Mice , Mice, Inbred Strains , Mice, Mutant Strains , Mice, Transgenic , Musculocutaneous Nerve/surgery , Patched Receptors , Patched-1 Receptor , Receptors, Cell Surface/genetics , Skin Neoplasms/pathology , Zinc Finger Protein GLI1
16.
J Invest Dermatol ; 135(5): 1415-1424, 2015 May.
Article in English | MEDLINE | ID: mdl-25313532

ABSTRACT

Merkel cell carcinoma (MCC) is a rare and deadly neuroendocrine skin tumor frequently associated with clonal integration of a polyomavirus, Merkel cell polyomavirus (MCPyV), and MCC tumor cells express putative polyomavirus oncoprotein small T antigen (sTAg) and truncated large T antigen. Here, we show robust transforming activity of sTAg in vivo in a panel of transgenic mouse models. Epithelia of preterm sTAg-expressing embryos exhibited hyperplasia, impaired differentiation, increased proliferation, and apoptosis, and activation of a DNA damage response. Epithelial transformation did not require sTAg interaction with the protein phosphatase 2A protein complex, a tumor suppressor in some other polyomavirus transformation models, but was strictly dependent on a recently described sTAg domain that binds Fbxw7, the substrate-binding component of the Skp1/Cullin1/F-box protein ubiquitin ligase complex. Postnatal induction of sTAg using a Cre-inducible transgene also led to epithelial transformation with development of lesions resembling squamous cell carcinoma in situ and elevated expression of Fbxw7 target proteins. Our data establish that expression of MCPyV sTAg alone is sufficient for rapid neoplastic transformation in vivo, implicating sTAg as an oncogenic driver in MCC and perhaps other human malignancies. Moreover, the loss of transforming activity following mutation of the sTAg Fbxw7 binding domain identifies this domain as crucial for in vivo transformation.


Subject(s)
Antigens, Viral, Tumor/physiology , Carcinogenesis/pathology , Carcinoma, Merkel Cell/physiopathology , Merkel cell polyomavirus/immunology , Polyomavirus Infections/immunology , Skin Neoplasms/physiopathology , Tumor Virus Infections/immunology , Animals , Antigens, Viral, Tumor/immunology , Apoptosis/physiology , Carcinogenesis/immunology , Carcinoma, Merkel Cell/immunology , Carcinoma, Merkel Cell/pathology , Cell Proliferation/physiology , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/pathology , DNA Damage/physiology , Disease Models, Animal , Merkel Cells/immunology , Merkel Cells/pathology , Mice , Mice, Transgenic , Skin Neoplasms/immunology , Skin Neoplasms/pathology
17.
J Invest Dermatol ; 134(8): 2241-2250, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24614157

ABSTRACT

Merkel cell carcinoma (MCC), a rare but aggressive cutaneous neoplasm with high metastatic potential, has a poor prognosis at late stages of disease with no proven chemotherapeutic regimens. Using an enriched culture medium, we established and characterized 11 MCC cell lines for Bcl-2 family profiling and functional studies. Immunoblot analysis revealed collectively high protein levels of prosurvival Bcl-2 members in cell lines and a panel of MCC tumors. Downregulation of individual Bcl-2 proteins by RNAi promoted death in a subset of MCC cell lines, whereas simultaneous inhibition of multiple family members by using the small-molecule antagonist ABT-263 led to a marked induction of cell death in 10 of 11 lines. ABT-263 induced Bax-dependent apoptosis with rapid cleavage of caspase-3 and PARP, regardless of Bcl-2 family profile or the presence of Merkel cell polyomavirus. Furthermore, ABT-263 treatment led to rapid and sustained growth suppression of MCC xenografts from a representative cell line, accompanied by a striking increase in apoptosis. Our results establish that concurrent inhibition of multiple prosurvival Bcl-2 proteins leads to effective induction of apoptosis, and strongly support the concept that targeting MCC dependence on these molecules may be useful therapeutically by reversing an intrinsic resistance to cell death.


Subject(s)
Carcinoma, Merkel Cell/pathology , Proto-Oncogene Proteins c-bcl-2/physiology , Skin Neoplasms/pathology , Aged , Aged, 80 and over , Aniline Compounds/pharmacology , Animals , Apoptosis/drug effects , Carcinoma, Merkel Cell/virology , Cell Line, Tumor , Cell Survival , Female , Humans , Male , Merkel cell polyomavirus/isolation & purification , Mice , Middle Aged , Myeloid Cell Leukemia Sequence 1 Protein/analysis , Skin Neoplasms/virology , Sulfonamides/pharmacology , bcl-2-Associated X Protein/physiology
18.
J Invest Dermatol ; 133(4): 936-45, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23223137

ABSTRACT

Merkel cell carcinoma (MCC) is an aggressive cutaneous neuroendocrine tumor with high mortality rates. Merkel cell polyomavirus (MCPyV), identified in the majority of MCCs, may drive tumorigenesis via viral T antigens. However, the mechanisms underlying pathogenesis in MCPyV-negative MCCs remain poorly understood. To nominate genes contributing to the pathogenesis of MCPyV-negative MCCs, we performed DNA microarray analysis on 30 MCCs. The MCPyV status of MCCs was determined by PCR for viral DNA and RNA. A total of 1,593 probe sets were differentially expressed between MCPyV-negative and MCPyV-positive MCCs, with significant differential expression defined as at least a 2-fold change in either direction and a P-value 0.05. MCPyV-negative tumors showed decreased RB1 expression, whereas MCPyV-positive tumors were enriched for immune response genes. Validation studies included immunohistochemistry demonstration of decreased RB protein expression in MCPyV-negative tumors and increased peritumoral CD8+ T lymphocytes surrounding MCPyV-positive tumors. In conclusion, our data suggest that loss of RB1 expression may have an important role in the tumorigenesis of MCPyV-negative MCCs. Functional and clinical validation studies are needed to determine whether this tumor-suppressor pathway represents an avenue for targeted therapy.


Subject(s)
Carcinoma, Merkel Cell/genetics , Merkel cell polyomavirus/genetics , Polyomavirus Infections/genetics , Skin Neoplasms/genetics , Transcriptome , Tumor Virus Infections/genetics , Aged , Aged, 80 and over , Carcinoma, Merkel Cell/mortality , Carcinoma, Merkel Cell/virology , Carcinoma, Squamous Cell/genetics , Female , Gene Expression Regulation, Neoplastic/physiology , Humans , Incidence , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Lymphocytes/physiology , Male , Middle Aged , Neoplasms, Basal Cell/genetics , Polyomavirus Infections/mortality , Prevalence , Retinoblastoma Protein/genetics , Skin Neoplasms/mortality , Skin Neoplasms/virology , Tumor Virus Infections/mortality
19.
EMBO Mol Med ; 4(3): 218-33, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22294553

ABSTRACT

Inhibition of Hedgehog (HH)/GLI signalling in cancer is a promising therapeutic approach. Interactions between HH/GLI and other oncogenic pathways affect the strength and tumourigenicity of HH/GLI. Cooperation of HH/GLI with epidermal growth factor receptor (EGFR) signalling promotes transformation and cancer cell proliferation in vitro. However, the in vivo relevance of HH-EGFR signal integration and the critical downstream mediators are largely undefined. In this report we show that genetic and pharmacologic inhibition of EGFR signalling reduces tumour growth in mouse models of HH/GLI driven basal cell carcinoma (BCC). We describe HH-EGFR cooperation response genes including SOX2, SOX9, JUN, CXCR4 and FGF19 that are synergistically activated by HH-EGFR signal integration and required for in vivo growth of BCC cells and tumour-initiating pancreatic cancer cells. The data validate EGFR signalling as drug target in HH/GLI driven cancers and shed light on the molecular processes controlled by HH-EGFR signal cooperation, providing new therapeutic strategies based on combined targeting of HH-EGFR signalling and selected downstream target genes.


Subject(s)
Carcinoma, Basal Cell/metabolism , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/metabolism , Pancreatic Neoplasms/metabolism , Animals , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/pathology , Cell Line, Tumor , ErbB Receptors/genetics , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Hedgehog Proteins/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phenotype , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Burden , Zinc Finger Protein GLI1
20.
J Invest Dermatol ; 131(10): 2113-20, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21697888

ABSTRACT

Merkel cell carcinoma (MCC) is a neoplasm thought to originate from the neuroendocrine Merkel cells of the skin. Although the prevalence of MCC has been increasing, treatments for this disease remain limited because of a paucity of information regarding MCC biology. We have found that the endocytic oncoprotein Huntingtin-interacting protein 1 (HIP1) is expressed at high levels in ∼90% of MCC tumors and serves as a more reliable histological cytoplasmic stain than the gold standard, cytokeratin 20. Furthermore, high anti-HIP1 antibody reactivity in the sera of a cohort of MCC patients predicts the presence of metastases. Another protein that is frequently expressed at high levels in MCC tumors is the stem cell factor (SCF) receptor tyrosine kinase, c-Kit. In working toward an understanding of how HIP1 might contribute to MCC tumorigenesis, we have discovered that HIP1 interacts with SCF-activated c-Kit. These data not only identify HIP1 as a molecular marker for management of MCC patients but also show that HIP1 interacts with and slows the degradation of c-Kit.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Merkel Cell/metabolism , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Aged , Animals , Cytoplasm/metabolism , Endocytosis , Female , Humans , Male , Mice , Middle Aged , Neoplasm Metastasis , Prevalence , Stem Cell Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...