Expression of the Embryonic Stem Cell Transcription Factor SOX2 in Human Skin
Alvaro C. Laga,Chiou-Yan Lai,Qian Zhan,Susan S. Huang,Elsa F. Velázquez,Qinghong Yang,Mei‐Yu Hsu,Gëorge F. Murphy
DOI: https://doi.org/10.2353/ajpath.2010.090495
IF: 5.77
2009-01-01
American Journal Of Pathology
Abstract:SOX2 is a gene located on chromosome 3q26.33 that encodes a transcription factor important to maintenance of embryonic neural crest stem cell pluripotency. We have identified rare SOX2-immunoreactive cells in normal human skin at or near the established stem cell niches. Three subsets of SOX2-positive cells were defined in these regions: those expressing only SOX2 and those that co-expressed SOX2 and either CK20 or microphthalmia-associated transcription factor, which are consistent with dichotomous differentiation of SOX2-expressing precursors along neuroendocrine (Merkel cell) or melanocytic lines, respectively. Examination of Merkel cell carcinomas confirmed nuclear SOX2 expression in this tumor type. In human patient melanoma, strong nuclear expression of SOX2 was noted in a subset of tumors, and the ability to detect SOX2 in lesional cells significantly correlated with primary tumor thickness in a survey cohort. To assess the potential role of SOX2 in melanoma growth, an in vivo tumorigenesis assay was used. Whereas SOX2 knockdown failed to influence proliferation of cultured melanoma cells in vitro, tumor xenografts generated with the SOX2-knockdown cell line showed significant decrease in mean tumor volume as compared with controls. In aggregate, these findings suggest that SOX2 is a novel biomarker for subpopulations of normal skin cells that reside in established stem cell niches and that might relate to Merkel cell and melanocyte ontogeny and tumorigenesis. SOX2 is a gene located on chromosome 3q26.33 that encodes a transcription factor important to maintenance of embryonic neural crest stem cell pluripotency. We have identified rare SOX2-immunoreactive cells in normal human skin at or near the established stem cell niches. Three subsets of SOX2-positive cells were defined in these regions: those expressing only SOX2 and those that co-expressed SOX2 and either CK20 or microphthalmia-associated transcription factor, which are consistent with dichotomous differentiation of SOX2-expressing precursors along neuroendocrine (Merkel cell) or melanocytic lines, respectively. Examination of Merkel cell carcinomas confirmed nuclear SOX2 expression in this tumor type. In human patient melanoma, strong nuclear expression of SOX2 was noted in a subset of tumors, and the ability to detect SOX2 in lesional cells significantly correlated with primary tumor thickness in a survey cohort. To assess the potential role of SOX2 in melanoma growth, an in vivo tumorigenesis assay was used. Whereas SOX2 knockdown failed to influence proliferation of cultured melanoma cells in vitro, tumor xenografts generated with the SOX2-knockdown cell line showed significant decrease in mean tumor volume as compared with controls. In aggregate, these findings suggest that SOX2 is a novel biomarker for subpopulations of normal skin cells that reside in established stem cell niches and that might relate to Merkel cell and melanocyte ontogeny and tumorigenesis. Biomarkers for tissue stem cell counterparts of Merkel cells and melanocytes are lacking. In related neoplasms such as melanoma, the identification of initiating/stem cells has relied in part on the observation that highly aggressive melanoma cell subsets are associated with molecular signatures that resemble those of pluripotent physiological stem cells.1Bittner M Meltzer P Chen Y Jiang Y Seftor E Hendrix M Radmacher M Simon Rm Yakhini Z Ben-Dor A Sampas N Dougherty E Wang E Marincola F Gooden C Lueders J Glatfelter A Pollock P Carpten J Gillanders E Leja D Dietrich K Beaudry C Berens M Alberts D Sondak V Molecular classification of cutaneous malignant melanoma by gene expression profiling.Nature. 2000; 406: 536-540Crossref PubMed Scopus (1655) Google Scholar, 2Hendrix MJ Seftor EA Hess AR Seftor RE Molecular plasticity of human melanoma cells.Oncogene. 2003; 22: 3070-3075Crossref PubMed Scopus (184) Google Scholar Several stem and progenitor cell-associated proteins have been detected in melanoma, such as cancer testis antigens,3Simpson AJ Caballero OL Jungbulth A Chen YT Old LJ Cancer/testis antigens, gametogenesis, and cancer.Nat Rev Cancer. 2005; 5: 615-625Crossref PubMed Scopus (1146) Google Scholar bone morphogenetic proteins,4Rothhammer T Wild PJ Meyer S Bataille F Pauer F Pauer A Klinkhammer-Schalke M Hein R Hofstaedter F Bosserhoff AK Bone morphogenic protein 7 (BMP7) expression is a potential novel prognostic marker for recurrence in patients with primary melanoma.Cancer Biomark. 2007; 3: 111-117PubMed Google Scholar, 5Hsu MY Rovinsky SA Lai CY Qasem S Liu X How J Engelhardt JF Murphy GF Aggressive melanoma cells escape from BMP7-mediated autocrine growth inhibition through coordinated Noggin upregulation.Lab Invest. 2008; 88: 842-855Crossref PubMed Scopus (32) Google Scholar, 6Hsu MY Rovinsky S Penmatcha S Herlyn M Muirhead D Bone morphogenetic proteins in melanoma: angel or devil?.Cancer Metastasis Rev. 2005; 24: 251-263Crossref PubMed Scopus (61) Google Scholar Notch receptors,7Balint K Xiao M Pinnix CC Soma A Veres I Juhasz I Brown EJ Capobianco AJ Herlyn M Liu ZJ Activation of Notch1 signaling is required for beta-catenin-mediated human primary melanoma progression.J Clin Invest. 2005; 115: 3166-3176Crossref PubMed Scopus (272) Google Scholar Wnt proteins,8Weeraratna AT Jiang Y Hostetter G Rossenblatt K Duray P Bittner M Trent JM Wnt5a signaling directly affects cell motility and invasion of metastatic melanoma.Cancer Cell. 2002; 1: 279-288Abstract Full Text Full Text PDF PubMed Scopus (739) Google Scholar or the ABCB5, CD133, CD166, CD34, nestin, or c-KIT antigens.2Hendrix MJ Seftor EA Hess AR Seftor RE Molecular plasticity of human melanoma cells.Oncogene. 2003; 22: 3070-3075Crossref PubMed Scopus (184) Google Scholar, 9Frank NY Margaryan A Huang Y Schatton T Waaga-Gasser AM Sayegh MH Sadee W Frank MH ABCB5-mediated doxorubicin transport and chemoresistance in human malignant melanoma.Cancer Res. 2005; 65: 4320-4333Crossref PubMed Scopus (442) Google Scholar, 10Klein WM Wu BP Zhao S Wu H Klein-Szanto AJ Tahan SR Increased expression of stem cell markers in malignant melanoma.Mod Pathol. 2007; 1: 102-107Crossref Scopus (249) Google Scholar, 11van Kempen LC van den Oord JJ van Muijen GN Weidle UH Bloemers HP Swart GW Activated leukocyte cell adhesion molecule/CD166, a marker of tumor progression in primary malignant melanoma of the skin.Am J Pathol. 2000; 156: 769-774Abstract Full Text Full Text PDF PubMed Scopus (173) Google Scholar Although such studies suggest a link between physiological (tissue) and neoplastic stem cell markers, it remains unclear whether melanomas and Merkel cell carcinomas express genes and related proteins typical of embryonic and tissue progenitor stem cells. In the course of gene expression profiling of the rete-tip-associated stem cell niche by immunoguided laser capture microdissection and real time reverse transcription (RT)-PCR,12Zhan Q Signoretti S Whitaker-Menezes D Friedman TM Korngold R Murphy GF Cytokeratin 15-positive basal cells targeted in graft-versus-host disease express a constitutive antiapoptotic phenotype.J Invest Dermatol. 2007; 127: 106-115Crossref PubMed Scopus (23) Google Scholar one of us (Q.Z.) detected significantly increased mRNA expression for SOX2. The SOX2 protein is a transcription factor critical to maintenance of embryonic neural crest stem cell pluripotency and self-renewal. It belongs to the SRY-related high mobility group box proteins that are homologous to the sex-determining gene on chromosome Y.13Bylund M Anderson E Novitch BG Muhr J Vertebrate neurogenesis is counteracted by Sox1–3 activity.Nature Neurosci. 2003; 6: 1162-1168Crossref PubMed Scopus (605) Google Scholar The present study was therefore conducted to characterize the expression of SOX2 in human embryonic and adult skin, and to explore its potential relevance to cutaneous neoplasms of neural crest derivation. Paraffin-embedded sections were obtained from the Department of Pathology, Brigham and Women's Hospital, according to an approved Institutional Review Board protocol in all cases. Samples included discarded human fetal skin (n = 35), adult normal skin (n = 24), nevi (n = 19), Merkel cell carcinoma (n = 9), and melanoma (n = 26). Sections from discarded fetal tissue were obtained from conceptuses of 6 to 24 weeks' estimated gestational age (EGA), and consisted of scalp, acral, truncal, and extremity skin. Sections of adult human skin included acral skin, scalp, lip, trunk, earlobe, face, and thigh. Human melanoma tissue microarrays consisted of: 1) a melanoma progression microarray (Cat# ME1003, US Biomax Inc., Rockville, MD) containing 100 cores of which 94 were technically evaluable (21 banal nevi, 53 primary vertical growth melanomas, and 20 metastatic melanomas); and 2) an outcome-annotated array (Cat# IMH-369, Imgenex, San Diego, CA) containing 59 cores of which 52 were technically evaluable (52 vertical growth phase melanomas—at least 2 mm in Breslow thickness—where 41 patients succumbed to their disease [median follow-up 25 months] and 11 survived [median follow-up 80.5 months]). Human melanoma cell lines A2058 and SK-MEL-5 were obtained from American Type Culture Collection (Manassas, VA) and grown in Dulbecco's modified Eagle's medium (Sigma-Aldrich Inc, St Louis, MO) supplemented with 10% inactivated fetal bovine serum (Hyclone Laboratories Inc, Logan, UT), 200 mmol/L/L l-glutamine, 100 IU/ml penicillin, and 100 μg/ml streptomycin, and maintained at 37°C in a humidified atmosphere containing 5% CO2. Total RNA from human melanoma cell lines A2058 and SK-MEL-5 was extracted from cell cultures using the RNAeasy Mini Kit (Qiagen, Valencia, CA). Total RNA was reverse-transcribed to cDNA using SuperScript III First-strand Synthesis System for RT-PCR (Invitrogen, Carlsbad, CA). cDNA from A2058 and SK-MEL-5 cells was surveyed for the expression of 84 human embryonic stem cell-related genes using the embryonic stem cell RT2Hendrix MJ Seftor EA Hess AR Seftor RE Molecular plasticity of human melanoma cells.Oncogene. 2003; 22: 3070-3075Crossref PubMed Scopus (184) Google Scholar profiler PCR array (Cat. no. PAHS-081, SA Biosciences, Frederic, MD). PCR was performed in a 7300 Real-Time PCR System (Applied Biosystems, Foster City, CA). The glyceraldehyde-3-phosphate dehydrogenase housekeeping gene was used for normalization and data were analyzed using the 2−ΔΔCT method.14Livak KJ Schmittgen TD Analysis of relative gene expression data using real-time quantitative PCR and the 2-ΔΔCT method.Methods. 2001; 25: 402-408Crossref PubMed Scopus (103415) Google Scholar Significant relative gene overexpression was defined as ≥fourfold difference in mRNA transcripts between the two cell lines. Recombinant lentiviral vectors were generated by co-transfecting pLKO.1-SOX2 (Sigma, St. Louis, MO), harboring short hairpin (sh)RNA for human SOX2, or non-target control shRNA (Sigma) with packaging plasmids VSVg and pCMV-ΔR8.2 (Sigma) into 293T packaging cells using Lipofectamine 2000 reagent (Invitrogen, Carlsbad, CA) according to manufacturer's instructions. Culture supernatants containing recombinant lentiviral particles were used to infect A2058 melanoma cells. Two days after infection, cells were selected with puromycin (1 μg/ml) for a period of 7 days. Subconfluent cultures were washed with PBS and extracted in lysis buffer containing 1% Triton X-100, 1% deoxycholic acid, 2 mmol/L CaCl2, and 1.8 mg/ml iodoacetamide, and 1 mmol/L phenylmethyl sulfonyl fluoride) in PBS. Cell lysates were quantified using a bicinchoninic acid protein assay kit (Pierce, Rockford, IL). An equal amount (50 μg) of total protein from each sample was subjected to electrophoresis on NuPAGE 4% to 12% Bis-Tris gels (Invitrogen), transblotted onto nitrocellulose membranes (Pierce), and probed with primary anti-SOX2 antibodies (Millipore, Billerica, MA), followed by a peroxidase-conjugated secondary antibody (Pierce). Subsequent re-probing using anti-β-actin (Abcam, Cambridge, MA) was also performed as internal loading control. Immunoreactive bands were detected using super-Signal West Femto Chemiluminescent substrate (Pierce), captured by a Syngene Chemi Genius Bio Imaging System (Syngene, Frederick, MD), and quantified by densitometry. Experiments were performed at least twice with consistency. Subconfluent cultures were trypsinized and seeded in 35-mm wells at 1 × 104 cells per well. Cells were re-fed twice weekly. At given intervals, cells in quadruplicate wells were harvested and counted in a Coulter counter (Coulter Electronics, Luton, UK). Statistical analyses were performed using the Mann-Whitney U-test. Experiments were repeated twice with similar results and representative data from one experiment is shown. Subconfluent melanoma cells were harvested and suspended in serum-free medium at a density of 108 cells/ml in PBS. One hundred μl of cell suspension (107 cells) were injected subcutaneously in the dorsal skin of each severe combined immunodeficient mouse (C.B-17; Taconic Laboratory, Germantown, NY; eight mice per condition). Tumor volume was monitored twice a week and determined as follows: (maximal dimension × minimal dimension)2Hendrix MJ Seftor EA Hess AR Seftor RE Molecular plasticity of human melanoma cells.Oncogene. 2003; 22: 3070-3075Crossref PubMed Scopus (184) Google Scholar/2. Statistical analyses were performed using analysis of variance following log transformation. The mice were sacrificed at the experimental end point and tumors were dissected, fixed in formalin, and subjected to immunohistochemistry for verification of SOX2 expression. Five-micron sections were deparaffinized in xylene, followed by treatment in 100% ethanol, and by serial hydration through 95% and 75% ethanol, and deionized H2O. Then, sections were placed in 1× target retrieval solution (Dako, Carpenteria, CA) and boiled in a Pascal pressure chamber (Dako) at 125°C for 30 seconds, 90°C for 10 seconds, and then allowed to cool down to room temperature. Immunohistochemistry was performed using a two-step horseradish peroxidase method. Briefly, the sections were first incubated with primary antibodies at 4°C overnight. The following primary antibodies were used: goat anti-human SOX2 (Neuromics, Edina, MN), mouse anti-microphthalmia-associated transcription factor (MITF) (Dako), and mouse anti-melanoma antigen recognized by T-cells (MART-1) (Signet Laboratories, Dedham, MA). After washing out unbound primary antibodies with Tris buffered saline-0.05% Tween 20, the tissue sections were incubated with peroxidase-conjugated secondary antibody at room temperature for 30 minutes, then washed with Tris buffered saline-0.05% Tween 20 three times for 5 minutes each. Immunoreactivity was detected using NovaRed peroxidase substrate (Vector Laboratories, Burlingame, CA). For double-label immunohistochemical staining, sections were deparaffinized and heat-induced antigen retrieval with Target Retrieval Solution (Dako) was used. Sections were incubated with SOX2 antibody (Neuromics) and one of the following: anti-CK20 (Dako), anti-CK15 (Lab Vision Corp., Fremont, CA), or anti-MITF (Dako) at room temperature for 1 hour and then with peroxidase-conjugated horse anti-goat antibody (Vector) and alkaline phosphatase-conjugated horse anti-mouse antibody at room temperature for 30 minutes. SOX2 and MITF staining was detected with NovaRed peroxidase substrate (Vector). CK20 and CK15 staining was detected with nitro blue tetrazolium chloride/5-bromo-4-chloro-3-indolyl phosphate, toluidine salt alkaline phosphatase substrate (Hoffman-La Roche, Nutley, NJ). Cell type(s) with strong nuclear immunoreactivity were recorded for embryonic-fetal skin, adult skin, and tumor samples. For patient tumor whole sections, SOX2 nuclear immunoreactivity was quantified using a semiquantitative scale (0 = negative, 1+ = ≤5% positive cells, 2+ = 6% to 25% positive cells, 3+ = 26% to 50%, and 4+ >50% positive cells). Tissue microarray cores were designated positive when unequivocal nuclear labeling was detected even in a minority of cells. Data were entered into an Excel file and imported into the SAS statistical software program. Descriptive statistics and univariate logistic regression were performed. Survival analysis was performed using the XLSTAT statistical package, using the Log-rank test for comparison of the survival distribution between two groups. For double-label immunofluorescence staining, sections were stained with anti-SOX2 (Neuromics) and one of the following antibodies: nestin (Chemicon, Temecula, CA), SOX10 (R&D, Minneapolis, MN), MITF (Dako), c-KIT (Dako), MART-1 (Signet Laboratories), CD31 (Bethyl Laboratories, Montgomery, TX), and CK20 (Dako). Isotype-matched antibodies (BD Pharmingen Inc., San Diego, CA) were included as controls. Alexa Fluor 594 donkey anti-goat IgG (Invitrogen, Eugene, OR), Alexa Fluor 488 donkey anti-mouse IgG (Invitrogen), and fluorescein isothiocyanate rat anti-mouse IgM (BD Biosciences, San Jose, CA) were used as secondary antibodies. Sections were deparaffinized and heat-induced antigen retrieval with Target Retrieval Solution (Dako) was performed. Slides were blocked for 30 minutes with horse or donkey sera, as appropriate, and incubated with the primary antibodies for 1 hour and then incubated with the appropriate secondary antibody in the dark for 1 hour. SOX2, MITF, and CK20 immunofluorescence triple labeling was performed by incubating sections with SOX2 (Neuromics), MITF (Dako), and CK20 (Dako) antibodies at room temperature for 1 hour and then with Alexa Fluor 594 donkey anti-goat IgG (Invitrogen) and Alexa Fluor 488 donkey anti-mouse IgG (Invitrogen) secondary antibodies. After washing to remove unbound secondary antibodies, slides were coverslipped with ProLong Gold Anti-Fade with DAPI (Invitrogen). Sections were analyzed with a BX51/BX52 microscope (Olympus America Inc, Melville, NY). Images were captured using the CytoVision 3.6 software (Applied Imaging, San Jose, CA). Figure 1 summarizes the localization of SOX2-positive cells at different stages of development in skin of conceptuses ranging from 6 to 24 weeks EGA. The early stages (eg, 6 to 12 weeks) demonstrated scattered single SOX2 immunoreactive cells within the dermis, often in a perivascular array (Figure 1A), and in the stratum germinativum of the epidermis (Figure 1B). Single cells exhibiting SOX2 nuclear staining were also noted in intimate association with forming buds of primary hair germ (Figure 1C), with the underlying mesenchymal condensation of cells also showing positivity that became progressively more prominent as the follicular papillae developed with downward descent to form differentiated lanugo follicles (Figure 1, D−H). SOX2-positive follicular papillae cells with progressive follicular maturation communicated with a thin mantle of positive cells within the adventitial sheath of the hair bulb and isthmus (Figure 1, D−H). A subpopulation of SOX2-positive cells (Figure 1I) within the follicular papilla co-expressed nestin (Figure 1, J−K). This was the only site of SOX2/nestin co-expression in the hair follicle. All SOX2-positive cells in this microdomain (Figure 1L) were negative for SOX10, MITF, c-Kit, and MART-1, all of which were clearly expressed by melanocytes within the basal layer of the adjacent hair bulb epithelium (eg, SOX10, Figure 1, M−N). Within the bulbous hair peg stage of trichogenesis (13 to 24 weeks), single SOX2-positive cells continued to be observed in the basal region of the epidermis as well as in the follicular infundibular epithelium in the vicinity of the forming bulge region. At this juncture, few if any dermal cells retained SOX2 immunoreactivity (Figure 1F). In adult skin, SOX2-positive cells also were present singly within the basal cell layer of the epidermis, with a marked tendency (>90%) to be localized at or near the tips of rete ridges (Figure 2, A−C), without evidence of staining within the underlying dermis (in contrast to embryonic skin, Figure 1A, inset). In the buccal mucosa of the lip, where akin to embryonic skin, rete ridges are not present, small clusters of SOX2-positive cells were present in the superficial submucosa and usually directly subjacent to singly disposed SOX2-positive cells within the epithelial basal layer (Figure 2D). The bulge region of hair follicles, which approximated the insertion site of arrector pili muscles, also consistently contained one to several SOX2-positive cells (Figure 2, E and F). The only mesenchymal localization of SOX2 in adult skin was the persistence of variable numbers of immunolabeled cells in follicular papillae, which otherwise retained a co-labeling profile similar to that of embryonic skin (see above). Specifically, nestin co-expression with SOX2 was restricted to the follicular papilla, as shown in Figure 1. The adventitial mantle at the tips of telogen follicles, presumably residua of follicular papillae, as well as the adjacent persistent follicular bulge regions, also contained positive cells (Figure 2, G−H). Three subsets of SOX2-positive intraepithelial cells were defined in both embryonic and adult skin based on co-expression of markers of melanocytic and Merkel cell lineage. In addition to SOX2-positive cells that failed to co-express nuclear MITF (Figure 2, I−K) or cytoplasmic CK20 (Figure 2, O and Q, arrow), CAM5.2, or the keratinocyte stem cell marker CK15,15Lyle S Christofidou-Solomidou M Liu Y Elder DE Albelda S Cotsarelis G The C8/144B monoclonal antibody recognizes cytokeratin 15 and defines the location of human hair follicle stem cells.J Cell Sci. 1998; 111: 3179-3188PubMed Google Scholar rare cells co-expressed SOX2 and MITF (Figure 2, L−N), and more abundant cells co-expressed SOX2 and either CAM5.2/CK20 (Figure 2, O−Q). Occasionally, SOX2-only and SOX2/CK20-positive cells were clustered in the same microdomain (Figure 2R), and in close proximity to either CK20-only cells (Figure 2S) or MITF-only cells (Figure 2T). Co-expression of SOX2 and MART-1, a marker for more mature melanocytes, was not identified, although numerous MART-1-positive, SOX2-negative melanocytes could be documented in the basal cell layer. Co-expression of SOX2 and c-KIT was not observed in either embryonic of adult skin. Although some Merkel cells had a CK20-positive, SOX2-negative phenotype, the precise ratio of SOX2-positive and negative cells also expressing CK20 could not be determined due to limitations in sampling. Based on the identification of SOX2 and CK20-coexpressing cells in embryonic and adult skin, we next sought to determine the potential presence and expression pattern of SOX2 in Merkel cell carcinoma. Nine samples of human primary and metastatic Merkel cell carcinoma interrogated by immunohistochemistry were all diffusely positive for SOX2, with >50% of cells exhibiting nuclear SOX2 immunopositivity (Figure 3, A−D). One of the cases studied contained areas of squamous differentiation (Figure 3, E and F); the neuroendocrine component of this tumor co-expressed SOX2 and CK20, whereas the malignant squamous component expressed neither (Figure 3, G−K). Double labeling confirmed restriction of CK20 expression to tumor cells that expressed SOX2, and SOX2-immunoreactive cells facilitated detection of intravascular invasion in one case (Figure 3, L−M). Because rare SOX2-positive cells in adult skin also co-expressed MITF, we also examined the immunoreactivity of SOX2 in a survey sample of nevi and melanomas. Table 1 summarizes the characteristics of patients and respective primary or metastatic melanomas and their SOX2 immunoreactivity patterns. Sixteen (62%) melanomas came from male patients and ten (38%) from female patients; eighteen tumors (69%) were primary and eight (31%) were metastatic. The majority of tumors were composed of epithelioid cells, with five (19%) having a prominent spindle cell component. In addition, two melanomas had a small cell component. Overall, zonal SOX2 expression was noted in 9/26 (35%) malignant melanomas. Of these, 1/9 showed 1+ immunopositivity, 3/9 showed 2+ positivity, 2/9 had 3+ positivity, and 3/9 tumors showed 4+ positivity. The remaining lesions had rare isolated SOX2-positive cells, but not zonal positivity as described above. The more intensely stained SOX2-positive cells tended to be concentrated at the interface of tumor and the surrounding stroma (Figure 4, A−C), where they were shown to be intimately associated with CD31-expressing peritumoral and intratumoral blood vessels (Figure 4, C and L−O). Melanomas with epithelioid, spindle, and small cell differentiation all harbored SOX2-positive cells (Figure 4, D−G), although occasional lesions with bimodal differentiation (eg, spindle cells or small cells) sharply juxtaposed with epithelioid cells tended to exhibit qualitatively more intense reactivity in the spindle and small cell components (Figure 4, H−K). In nevi, SOX2 immunoreactivity was present in 3/19 (16%) cases, although staining was qualitatively less intense and accentuations of staining at tumor-stromal interfaces or in association with differences in cell size and shape were not consistently observed (Figure 4C inset).Table 1Characteristics and Sox2 Immunoreactivity Pattern of Tumors from Patients with Primary and Metastatic Melanoma (MM)CaseAgeSexLocationMM typeCell type (s)Breslow (mm)ClarkMits/mm2Sox2166MFlankNevoidEpitheliod, nevoid1.05IV30263MShoulderResidualEpithelioid1.1IV40324MAbdomenSSEpithelioid1.2IV<10445FAnkleALEpithelioid0.85IV<10522MScalpSSEpithelioid1.2III<10662FGreat ToeSpindle cellEpithelioid, spindle6.0V243+777MShoulderMixedEpithelioid6.5IV170842FHeelALEpithelioid, spindle4.0IV23+954MBackSSEpithelioid0.8III201062FChinDesmoplasticSpindle10.0V22+1189MAbdomenNodularEpithelioid2.0IV501252FThighNodularEpithelioid, spindle4.0IV>62+1360MEarlobeUnclassifiedEpithelioid, small cell1.4IV104+1457FMandibleMucosal typeEpithelioid1.05N/AN/A01548FLungPrimary lungEpithelioidN/AN/AN/A01653MChestUnclassifiedEpithelioid0.25IIN/A01782MInguinal RegionMetastaticEpithelioidN/AN/AN/A01849FScalpMetastaticEpithelioidN/AN/AN/A01921MInner ThighNodularEpithelioid2.7IV>602065MPericavalMetastaticEpithelioidN/AN/AN/A1+2158MLymph NodeMetastaticEpithelioidN/AN/AN/A02264MSmall BowelMetastaticEpithelioidN/AN/AN/A02363MChest WallMetastaticEpithelioidN/AN/AN/A4+2459MLymph NodeMetastaticEpithelioid, small cellN/AN/AN/A2+2562FLymph NodeMetastaticEpithelioidN/AN/AN/A02652FShoulderSpindle cellSpindle5.0V124+N/A, not available or not applicable; Mits, mitosis.M, male; F, female; SS, superficial spreading type; AL, acral lentiginous type. Open table in a new tab N/A, not available or not applicable; Mits, mitosis. M, male; F, female; SS, superficial spreading type; AL, acral lentiginous type. An exploratory analysis was undertaken to investigate the clinical significance, if any, of SOX2 immunoreactivity in human melanomas. A univariate logistic regression analysis of the relatively small sample of melanomas evaluated indicated that Breslow thickness was significantly correlated with SOX2 immunopositivity. Via this analysis, melanomas from the cohort that differed by 1 mm in Breslow thickness were on average twice as likely to exhibit presence of SOX2 expression. Conversely stated, SOX2-positive melanomas were significantly associated with a greater likelihood (twice as likely on average) of being 1 mm thicker than their SOX2-negative comparators (OR = 2.01[1.04, 3.92], P = 0.039). To expand the clinical sample size, we also evaluated SOX2 expression in a melanoma progression tissue microarray and in a melanoma outcome-annotated tissue microarray. Of the additional 91 primary melanomas, 41 (45%) expressed SOX2, with positive staining also detected in 3 of 21 nevi (14%), and 13 of 34 metastases (38%). With regard to clinical outcome, 6 of 11 (55%) survivors, and 21 of 41 (51%) nonsurvivors (eg, death of melanoma) expressed SOX2. No statistically significant difference in survival time was found between SOX2-positive and SOX2-negative tumors in this outcome microarray (Log-rank test P = 0.27). Melanoma cell lines were selected based on SOX2 expression; A2058 melanoma cells are uniformly positive for SOX2, whereas SK-MEL-5 cells are largely negative by immunohistochemistry. Real time RT-PCR confirmed that A2058 cells had 247-fold higher expression of SOX2 than to SK-MEL-5 cells. Using a commercially available embryonic stem cell gene expression array (SA Biosciences), we compared the expression profiles in these SOX2-positive and SOX2-negative cells. Of 84 genes represented in the screening array, there was detectable signal for 41 genes in one or both cell lines and no signal for 43 genes in both cell lines. Of the 41 stem cell-related genes detected, 25 (61%) were significantly overexpressed in A2058 cells, and 5 genes (12%) were relatively overexpressed in SK-MEL-5 cells. The remaining 11 genes for which signal was detected showed no significant difference in expression between the two cell lines. Genes overexpressed in A2058 cells corresponded to transcription factors involved in the maintenance of "stemness" (eg, SOX2, FOXD3, GATA6, NANOG, nestin, CDH5 [vascular endothelial-cadherin]), and signaling molecules required for pluripotency and self-renewal (eg, fibroblast growth factors 4 and 5, interferon-induced transmembrane protein 1, telomerase reverse transcription, teratocarcinoma derived growth factor 1). The 5 genes overexpressed in SK-MEL-5 cells were growth differentiation factor 3; laminin-α1; noggin; nuclear receptor subfamily 5, group A, member 2; and transcription factor CP2-like 1. Three of these genes (noggin; nuclear receptor subfamily 5, group A, member 2; and transcription factor CP2-like 1) are related to maintenance of "stemness," and the other two are growth factors. In summary, of embryonic stem cell-related genes that w
What problem does this paper attempt to address?
-
Stem Cells , Tissue Engineering and Hematopoietic Elements Expression of The Embryonic Stem Cell Transcription Factor SOX 2 in Human Skin Relevance to Melanocyte and Merkel Cell Biology
Alvaro C. Laga,Chiou-Yan Lai,Qian Zhan,Susan J. Huang,Elsa F. Velazquez,Qinghong Yang,Mei-Yu Hsu,George F. Murphy
2010-01-01
Abstract:SOX2 is a gene located on chromosome 3q26.33 that encodes a transcription factor important to maintenance of embryonic neural crest stem cell pluripotency. We have identified rare SOX2-immunoreactive cells in normal human skin at or near the established stem cell niches. Three subsets of SOX2-positive cells were defined in these regions: those expressing only SOX2 and those that co-expressed SOX2 and either CK20 or microphthalmia-associated transcription factor , which are consistent with dichotomous differentiation of SOX2-expressing precursors along neuroendocrine (Merkel cell) or melanocytic lines , respectively. Examination of Merkel cell carcinomas confirmed nuclear SOX2 expression in this tumor type. In human patient melanoma, strong nuclear expression of SOX2 was noted in a subset of tumors , and the ability to detect SOX2 in lesional cells significantly correlated with primary tumor thickness in a survey cohort. To assess the potential role of SOX2 in melanoma growth, an in vivo tumorigenesis assay was used. Whereas SOX2 knockdown failed to influence proliferation of cultured melanoma cells in vitro , tumor xenografts generated with the SOX2-knockdown cell line showed significant decrease in mean tumor volume as compared with controls. In aggregate , these findings suggest that SOX2 is a novel biomarker for subpopulations of normal skin cells that reside in established stem cell niches and that might relate to Merkel cell and melanocyte ontogeny and tumorigenesis. (Am J Pathol 2010, 176:903–913; DOI: 10.2353/ajpath.2010.090495) Biomarkers for tissue stem cell counterparts of Merkel cells and melanocytes are lacking. In related neoplasms such as melanoma, the identification of initiating/stem cells has relied in part on the observation that highly aggressive melanoma cell subsets are associated with molecular signatures that resemble those of pluripotent physiological stem cells. Several stem and progenitor cell-associated proteins have been detected in melanoma, such as cancer testis antigens, bone morphogenetic proteins, Notch receptors, Wnt proteins, or the ABCB5, CD133, CD166, CD34, nestin, or c-KIT antigens. Although such studies suggest a link between physiological (tissue) and neoplastic stem cell markers, it remains unclear whether melanomas and Merkel cell carcinomas express genes and related proteins typical of embryonic and tissue progenitor stem cells. In the course of gene expression profiling of the retetip-associated stem cell niche by immunoguided laser capture microdissection and real time reverse transcription (RT)-PCR, one of us (Q.Z.) detected significantly increased mRNA expression for SOX2. The SOX2 protein is a transcription factor critical to maintenance of embryonic neural crest stem cell pluripotency and self-renewal. It belongs to the SRY-related high mobility group box proteins that are homologous to the sex-determining gene on chromosome Y. The present study was therefore conducted to characterize the expression of SOX2 in human embryonic and adult skin, and to explore its potential relevance to cutaneous neoplasms of neural crest derivation.
-
Expression of the embryonic stem cell transcription factor SOX2 in human skin: relevance to melanocyte and merkel cell biology.
Alvaro C Laga,Chiou-Yan Lai,Qian Zhan,Susan J Huang,Elsa F Velazquez,Qinghong Yang,Mei-Yu Hsu,George F Murphy
DOI: https://doi.org/10.2353/ajpath.2010.090495
2010-01-01
Abstract:SOX2 is a gene located on chromosome 3q26.33 that encodes a transcription factor important to maintenance of embryonic neural crest stem cell pluripotency. We have identified rare SOX2-immunoreactive cells in normal human skin at or near the established stem cell niches. Three subsets of SOX2-positive cells were defined in these regions: those expressing only SOX2 and those that co-expressed SOX2 and either CK20 or microphthalmia-associated transcription factor, which are consistent with dichotomous differentiation of SOX2-expressing precursors along neuroendocrine (Merkel cell) or melanocytic lines, respectively. Examination of Merkel cell carcinomas confirmed nuclear SOX2 expression in this tumor type. In human patient melanoma, strong nuclear expression of SOX2 was noted in a subset of tumors, and the ability to detect SOX2 in lesional cells significantly correlated with primary tumor thickness in a survey cohort. To assess the potential role of SOX2 in melanoma growth, an in vivo tumorigenesis assay was used. Whereas SOX2 knockdown failed to influence proliferation of cultured melanoma cells in vitro, tumor xenografts generated with the SOX2-knockdown cell line showed significant decrease in mean tumor volume as compared with controls. In aggregate, these findings suggest that SOX2 is a novel biomarker for subpopulations of normal skin cells that reside in established stem cell niches and that might relate to Merkel cell and melanocyte ontogeny and tumorigenesis.
-
SOX2 and nestin expression in human melanoma: an immunohistochemical and experimental study.
Alvaro C Laga,Qian Zhan,Carsten Weishaupt,Jie Ma,Markus H Frank,George F Murphy
DOI: https://doi.org/10.1111/j.1600-0625.2011.01247.x
2011-01-01
Experimental Dermatology
Abstract:SOX2 is an embryonic neural crest stem-cell transcription factor recently shown to be expressed in human melanoma and to correlate with experimental tumor growth. SOX2 binds to an enhancer region of the gene that encodes for nestin, also a neural progenitor cell biomarker. To define further the potential relationship between SOX2 and nestin, we examined co-expression patterns in 135 melanomas and 37 melanocytic nevi. Immunohistochemical staining in 27 melanoma tissue sections showed an association between SOX2 positivity, spindle cell shape and a peripheral nestin distribution pattern. In contrast, SOX2-negative cells were predominantly epithelioid, and exhibited a cytoplasmic pattern for nestin. In tissue microarrays, co-expression correlated with tumor progression, with only 11% of nevi co-expressing SOX2 and nestin in contrast to 65% of metastatic melanomas, and preliminarily, with clinical outcome. Human melanoma lines that differentially expressed constitutive SOX2 revealed a positive correlation between SOX2 and nestin expression. Experimental melanomas grown from these respective cell lines in murine subcutis and dermis of xenografted human skin maintained the association between SOX2-positivity, spindle cell shape, and peripheral nestin distribution. Moreover, the cytoplasmic pattern of nestin distribution was observed in xenografts generated from SOX2-knockdown A2058 melanoma cells, in contrast to the peripheral nestin pattern seen in tumors grown from A2058 control cells transfected with non-target shRNA. In aggregate, these data further support a biologically significant linkage between SOX2 and nestin expression in human melanoma.
-
Cytoplasmic Skp2 Expression Is Increased In Human Melanoma And Correlated With Patient Survival
Guangdi Chen,Yabin Cheng,Zhizhong Zhang,Magdalena Martinka,Gang Li
DOI: https://doi.org/10.1371/journal.pone.0017578
IF: 3.7
2011-01-01
PLoS ONE
Abstract:Background: S-phase kinase protein 2 (Skp2), an F-box protein, targets cell cycle regulators via ubiquitin-mediated degradation. Skp2 is frequently overexpressed in a variety of cancers and associated with patient survival. In melanoma, however, the prognostic significance of subcellular Skp2 expression remains controversial.Methods: To investigate the role of Skp2 in melanoma development, we constructed tissue microarrays and examined Skp2 expression in melanocytic lesions at different stages, including 30 normal nevi, 61 dysplastic nevi, 290 primary melanomas and 146 metastatic melanomas. The TMA was assessed for cytoplasmic and nuclear Skp2 expression by immunohistochemistry. The Kaplan-Meier method was used to evaluate the patient survival. The univariate and multivariate Cox regression models were performed to estimate the harzard ratios (HR) at five-year follow-up.Results: Cytoplasmic but not nuclear Skp2 expression was gradually increased from normal nevi, dysplastic nevi, primary melanomas to metastatic melanomas. Cytoplasmic Skp2 expression correlated with AJCC stages (I vs II-IV, P < 0.001), tumor thickness (<= 2.00 vs > 2.00 mm, P < 0.001) and ulceration (P = 0.005). Increased cytoplasmic Skp2 expression was associated with a poor five-year disease-specific survival of patients with primary melanoma (P = 0.018) but not metastatic melanoma (P > 0.05).Conclusion: This study demonstrates that cytoplasmic Skp2 plays an important role in melanoma pathogenesis and its expression correlates with patient survival. Our data indicate that cytoplasmic Skp2 may serve as a potential biomarker for melanoma progression and a therapeutic target for this disease.
-
SOX2 Contributes to Melanoma Cell Invasion.
Sasha D Girouard,Alvaro C Laga,Martin C Mihm,Richard A Scolyer,John F Thompson,Qian Zhan,Hans R Widlund,Chung-Wei Lee,George F Murphy
DOI: https://doi.org/10.1038/labinvest.2011.188
IF: 5.511
2012-01-01
Laboratory Investigation
Abstract:The mechanisms of melanoma invasion are poorly understood despite extensive inquiry. SRY (sex determining region Y)-box 2 (SOX2) is an embryonic stem cell transcription factor that has recently been discovered to be expressed in human melanoma where it is associated with dermal invasion and primary tumor thickness. To assess the potential role of SOX2 expression in melanoma invasion, we examined patient melanomas and humanized melanoma xenografts, and noted preferential SOX2 expression in cells that interfaced and infiltrated dermal stroma. Experimental knockdown (KD) of SOX2 mRNA and protein in A2058 melanoma cells with high constitutive SOX2 expression resulted in 4.5-fold decreased invasiveness in vitro compared with controls (P<0.0001). Conversely, when G361 cells that normally express low SOX2 were transduced to overexpress SOX2 mRNA and protein, a 3.8-fold increase in invasiveness was observed (P=0.0004). Among 84 invasion-related genes, RT-PCR screening revealed that SOX2 KD resulted in striking decrease in matrix metalloproteinase-3 (MMP-3), an endopeptidase associated with cleavage of the extracellular matrix. Quantitatively, SOX2 KD diminished MMP-3 mRNA by 87.8%. MMP-3 KD in SOX2-expressing A2058 cells served to inhibit invasion, although to a lesser degree than SOX2 KD. Finally, immunostaining of patient and xenograft melanomas revealed coordinate SOX2 and MMP-3 expression in regions of stromal infiltration. These data implicate SOX2 expression in melanoma invasion, and suggest a role for MMP-3 as one potential mediator of this process.
-
Oct-4 Expression in Epidermal Tumors
SHEN Ying,CAI Sui-qin
DOI: https://doi.org/10.3760/cma.j.issn.1008-1372.2012.05.003
2012-01-01
Abstract:Objective The aim of this study was to understand the role of specific markers of stem cells Oct-4 expression in the development of human epidermal non-melanoma cutaneous tumors.Methods The paraffin-embedded samples were retrieved from files of pathology department at our hospital,including 20 cases of skin squamous cell carcinomas (SCC),20 cases of basal cell carcinomas (BCC),20 cases of seborrhoeic keratosis (SK) and 20 cases of normal skin (from head,face,trunk,extremities).The expression of Oct-4 and PCNA were observed by immunohistochemical staining technique.Results Oct-4 protein was abnormally increased in SCC and BCC comparel to normal skin and SK ( P <0.05 ).However there were no significant difference of Oct-4 protein expression between SCC and BCC ( P >0.05).There were also no significantly different Oct-4 protein expression between Sk and the normal skin ( P > 0.05 ),and no significantly different Oct-4 protein expression between SK and BCC( P >0.05 ).PCNA protein was abnormally increased in SCC and BCC compared to normal skin and SK ( P <0.01 ).There were significantly different PCNA protein expression between SCC and BCC( P <0.05).There were also significantly different PCNA protein expression between SK and the normal skin ( P < 0.05 ).However there were no significant difference of PCNA protein expression between SK and BCC ( P > 0.05 ).There were positive correlation between the expression intensity of Oct-4 and PCNA in SCC and BCC.Conclusions The abnormal expression of Oct-4 may have an important role in the development of BCC and SCC.Positive Oct-4 expression cells may be the tumor stem cell in SCC and BCC.There were positive correlation between the expression intensity of Oct-4 and PCNA in SCC and BCC.The over expression Oct-4 in BCC and SCC may play an important role in proliferation of tumor.
-
Silencing Sox2 Induced Mesenchymal-Epithelial Transition And Its Expression Predicts Liver And Lymph Node Metastasis Of Crc Patients
Xu Han,Xuefeng Fang,Xiaoyan Lou,Dasong Hua,Wenchao Ding,Gregory Foltz,Leroy Hood,Ying Yuan,Biaoyang Lin
DOI: https://doi.org/10.1371/journal.pone.0041335
IF: 3.7
2012-01-01
PLoS ONE
Abstract:SOX2 is an important stem cell marker and plays important roles in development and carcinogenesis. However, the role of SOX2 in Epithelial-Mesenchymal Transition has not been investigated. We demonstrated, for the first time, that SOX2 is involved in the Epithelial-Mesenchymal Transition (EMT) process as knock downof SOX2 in colorectal cancer (CRC) SW620 cells induced a Mesenchymal-Epithelial Transition (MET) process with recognized changes in the expression of key genes involved in the EMT process including E-cadherin and vimentin. In addition, we provided a link between SOX2 activity and the WNT pathway by showing that knock down of SOX2 reduced the WNT pathway activity in colorectal cancer (CRC) cells. We further demonstrated that SOX2 is involved in cell migration and invasion in vitro and in metastasis in vivo for CRC cells, and that the process might be mediated through the MMP2 activity. Finally, an IHC analysis of 44 cases of colorectal cancer patients suggested that SOX2 is a prognosis marker for metastasis of colorectal cancers.
-
Dominant B-cell Epitopes from Cancer/stem Cell Antigen SOX2 Recognized by Serum Samples from Cancer Patients.
Julia Shih,Munira Rahman,Quang T. Luong,Shirley H. Lomeli,Joseph Riss,Robert M. Prins,Ali O. Gure,Gang Zeng
2014-01-01
Abstract:Human sex determining region Y-box 2 (SOX2) is an important transcriptional factor involved in the pluripotency and stemness of human embryonic stem cells. SOX2 plays important roles in maintaining cancer stem cell activities of melanoma and cancers of the brain, prostate, breast, and lung. SOX2 is also a lineage survival oncogene for squamous cell carcinoma of the lung and esophagus. Spontaneous cellular and humoral immune responses against SOX2 present in cancer patients classify it as a tumor-associated antigen (TAA) shared by lung cancer, glioblastoma, and prostate cancer among others. In this study, B-cell epitopes were predicted using computer-assisted algorithms. Synthetic peptides based on the prediction were screened for recognition by serum samples from cancer patients using ELISA. Two dominant B-cell epitopes, SOX2:52-87 and SOX2:98-124 were identified. Prostate cancer, glioblastoma and lung cancer serum samples that recognized the above SOX2 epitopes also recognized the full-length protein based on Western blot. These B-cell epitopes may be used in assessing humoral immune responses against SOX2 in cancer immunotherapy and stem cell-related transplantation.
-
SOX10 requirement for melanoma tumor growth is due, in part, to immune-mediated effects
Sheera R Rosenbaum,Manoela Tiago,Signe Caksa,Claudia Capparelli,Timothy J Purwin,Gaurav Kumar,McKenna Glasheen,Danielle Pomante,Daniel Kotas,Inna Chervoneva,Andrew E Aplin
DOI: https://doi.org/10.1016/j.celrep.2021.110085
2021-12-07
Abstract:Developmental factors may regulate the expression of immune modulatory proteins in cancer, linking embryonic development and cancer cell immune evasion. This is particularly relevant in melanoma because immune checkpoint inhibitors are commonly used in the clinic. SRY-box transcription factor 10 (SOX10) mediates neural crest development and is required for melanoma cell growth. In this study, we investigate immune-related targets of SOX10 and observe positive regulation of herpesvirus entry mediator (HVEM) and carcinoembryonic-antigen cell-adhesion molecule 1 (CEACAM1). Sox10 knockout reduces tumor growth in vivo, and this effect is exacerbated in immune-competent models. Modulation of CEACAM1 expression but not HVEM elicits modest effects on tumor growth. Importantly, Sox10 knockout effects on tumor growth are dependent, in part, on CD8+ T cells. Extending this analysis to samples from patients with cutaneous melanoma, we observe a negative correlation with SOX10 and immune-related pathways. These data demonstrate a role for SOX10 in regulating immune checkpoint protein expression and anti-tumor immunity in melanoma.
-
646 SOX4 is Essential for Melanoma Tumorigenesis
W. Dai,H. Wang,J. Ma,T. Gao,C. Li
DOI: https://doi.org/10.1016/j.jid.2016.02.687
2016-01-01
Abstract:Malignant melanoma is an aggressive skin neoplasm with approximately 20,000 new cases diagnosed in China each year. SOX4, which belongs to the highly conserved group C SOX transcription factors, has been demonstrated to be important for numerous aspects of embryogenesis and various facets of tumor initiation and progression. In this study, we found SOX4 expression is significantly upregulated at the mRNA and protein levels in primary and advanced melanoma patients. We further investigated its function by knockdown SOX4 in melanoma cell lines, which resulted in higher apoptosis rates, shorter cell cycle, attenuated growth and reprogramming metabolism. Mechanistically, knockdown of SOX4 reduced the expression of TERT (human telomerase reverse transcriptase) via NF-B activation, leading to an attenuated invasive phenotype. Collectively, our findings establish that SOX4 is a critical component in melanoma progression and invasion, suggesting the potential implications for combination targeted therapies.
-
Sox10 Regulates Skin Melanocyte Proliferation by Activating the DNA Replication Licensing Factor MCM5.
Zhongyuan Su,Xiaozi Zheng,Xiaobo Zhang,Yipin Wang,Shanpu Zhu,Fan Lu,Jia Qu,Ling Hou
DOI: https://doi.org/10.1016/j.jdermsci.2016.12.002
IF: 5.408
2017-01-01
Journal of Dermatological Science
Abstract:BACKGROUND:The control of cell proliferation is a fundamental aspect of tissue formation in development and regeneration. A cell type that illustrates this point particularly well is the neural crest-derived melanocyte, the pigment cell of vertebrates, as melanocytes can be followed easily during development and their pigment is directly visible in the integument of the adult. In mammals, melanocytes undergo physiological cycles of loss and proliferative regeneration during the hair cycle, and their proliferation is also critical during wound healing, repigmentation of depigmented lesions, and in melanoma formation and progression. Hence, a thorough analysis of the molecular parameters controlling melanocyte proliferation is crucial for our understanding of the physiology of this cell type both in health and disease.OBJECTIVE:SOX10 is a critical regulator in melanocytes and melanoma cells, but its specific role in their proliferation is far from clear. In this study we analyze the role of SOX10 in regulating mammalian melanocyte proliferation in a mouse model.METHODS:The role of SOX10 in melanoblast proliferation was analyzed in Sox10/+ mice by co-staining for melanocyte-specific markers and cell proliferation. In vitro, the role of SOX10 was studied by manipulating its levels using RNAi and analyzing the effects on DNA synthesis and cell growth and on gene expression at the RNA and protein levels.RESULTS:Reduction of Sox10 gene dose led to a reduction in the number of melanoblasts. Knockdown of Sox10 in melanocytes led to inhibition of cell proliferation and a decrease in the expression of the minichromosome maintenance complex component 5 (MCM5). In fact, SOX10 directly activated MCM5 transcription by binding to conserved SOX10 consensus DNA sequences in the MCM5 promoter. Furthermore, the defect in cell proliferation could be rescued partially by overexpression of MCM5 in Sox10 knockdown melanocytes.CONCLUSION:The results suggest that the SOX10-MCM5 axis plays an important role in controlling melanocyte proliferation. Our findings provide novel insights into the regulatory mechanisms of melanocyte proliferation and may have implications for our understanding of the roles of SOX10 and MCM5 in abnormal melanocyte proliferation disorders such as cutaneous melanoma.
-
Dermis‐derived Stem Cells: a Source of Epidermal Melanocytes and Melanoma?
Susan E. Zabierowski,Mizuho Fukunaga-Kalabis,Ling Li,Meenhard Herlyn
DOI: https://doi.org/10.1111/j.1755-148x.2011.00847.x
2011-01-01
Pigment Cell & Melanoma Research
Abstract:SummaryHuman multipotent dermal stem cells (DSCs) have been isolated and propagated from the dermal region of neonatal foreskin. DSCs can self‐renew, express the neural crest stem cell markers NGFRp75 and nestin, and are capable of differentiating into a wide variety of cell types including mesenchymal and neuronal lineages and melanocytes, indicative of their neural crest origin. When placed in the context of reconstructed skin, DSCs migrate to the basement membrane zone and differentiate into melanocytes. These findings, combined with the identification of NGFRp75‐positive cells in the dermis of human foreskin, which are devoid of hair, suggest that DSCs may be a self‐renewing source of extrafollicular epidermal melanocytes. In this review, we discuss the properties of DSCs, the pathways required for melanocyte differentiation, and the value of 3D reconstructed skin to assess the behavior and contribution of DSCs in the naturalized environment of human skin. Potentially, DSCs provide a link to malignant melanoma by being a target of UVA‐induced transformation.
-
Altered HOX Gene Expression in Human Skin and Breast Cancer Cells
Hexin Chen,S. Sukumar
2017-01-01
Abstract:Human HOX genes are expressed in a spatio-temporal fashion during embryogenesis and early development where they act as master transcriptional regulators. HOX genes are also expressed in normal adult cells, potentially in a tissue specific manner involving maintenance of the normal phenotype. In selected oncogenic transformations, mis-expression of many HOX genes have been shown, indicating an involvement of these transcriptional regulators in carcinogenesis and metastasis. Utilising quantitative real-time RT-PCR assays, the expression of 20 HOX genes and two known HOX co-factors, PBX1B and MEIS1, were analysed in human melanoma and breast cancer cell lines, comparing results against non-malignant cells. Alterations in HOX gene expression were observed for all malignant cells examined, with some dysregulated transcript levels seemingly random, and the expression of other HOX genes apparently following the same patterns in both skin and breast cancer establishment. Furthermore, HOX gene expression was correlated with the invasive capacity of the cells. The expression of the HOX co-factors PBX1B and MEIS1 showed no marked changes from the non-malignant to the malignant phenotypes, further indicating that it is dysregulated HOX gene expression, rather than dysregulated gene expression of HOX co-factors, that potentially commit the cell to re-differentiate and undergo oncogenic transformation.
-
Expression and Functional Role of Sox9 in Human Epidermal Keratinocytes
Ge Shi,Kyung-Cheol Sohn,Zhengjun Li,Dae-Kyoung Choi,Young Min Park,Jin-Hwa Kim,Yi-Ming Fan,Yong Hee Nam,Sooyeon Kim,Myung Im,Young Lee,Young-Joon Seo,Chang Deok Kim,Jeung-Hoon Lee
DOI: https://doi.org/10.1371/journal.pone.0054355
IF: 3.7
2013-01-01
PLoS ONE
Abstract:In this study, we investigated the expression and putative role of Sox9 in epidermal keratinocyte. Immunohistochemical staining showed that Sox9 is predominantly expressed in the basal layer of normal human skin epidermis, and highly expressed in several skin diseases including psoriasis, basal cell carcinoma, keratoacanthoma and squamous cell carcinoma. In calcium-induced keratinocyte differentiation model, the expression of Sox9 was decreased in a time dependent manner. When Sox9 was overexpressed using a recombinant adenovirus, cell growth was enhanced, while the expression of differentiation-related genes such as loricrin and involucrin was markedly decreased. Similarly, when rat skin was intradermally injected with the adenovirus expressing Sox9, the epidermis was thickened with increase of PCNA positive cells, while the epidermal differentiation was decreased. Finally, UVB irradiation induced Sox9 expression in cultured human epidermal keratinocytes, and keratinocytes are protected from UVB-induced apoptosis by Sox9 overexpression. Together, these results suggest that Sox9 is an important regulator of epidermal keratinocytes with putative pro-proliferation and/or pro-survival functions, and may be related to several cutaneous diseases that are characterized by abnormal differentiation and hyperproliferation.
-
Low Levels of Sox2 are required for Melanoma Tumor-Repopulating Cell Dormancy.
Qiong Jia,Fang Yang,Wei Huang,Yao Zhang,Binghao Bao,Ke Li,Fuxiang Wei,Cunyu Zhang,Haibo Jia
DOI: https://doi.org/10.7150/thno.29698
IF: 11.6
2019-01-01
Theranostics
Abstract:Tumorigenic cells, when facing a hostile environment, may enter a dormant state, leading to long-term tumor survival, relapse, and metastasis. To date, the molecular mechanism of tumor cell dormancy remains poorly understood. Methods: A soft, 3-dimentional (3D) fibrin gel culture system was used to mechanically select and grow highly malignant and tumorigenic melanoma tumor-repopulating cells (TRCs). We cultured control melanoma TRCs, TRCs with Sox2 knockdown, TRCs with Sox2 knockout, and a 2D control for in vitro and in vivo experiments. Western blotting, immunofluorescence, and flow cytometry analysis were performed to examine TRC dormancy and exit from dormancy. Results: Under a low-expression condition, we show that Sox2, a stemness molecule participates in dormancy regulation of highly tumorigenic cells that can repopulate a tumor (TRCs). Intriguingly, complete depletion of Sox2 via knockout results in dormancy exit and growth resumption of melanoma TRCs in culture and elevation of melanoma TRC apoptosis. Mice that are injected subcutaneously with Sox2-depleted melanoma TRCs do not form tumors and survive much longer than those injected with melanoma TRCs. We found that complete depletion of Sox2 promotes nuclear translocation of phosphorylated STAT3, where it binds to the p53 gene promoter, thus activating the p53-caspase3 cascade. Conclusion: These findings provide a novel insight into the role of the Sox2 gene in tumor cell stemness, tumor dormancy, and apoptosis.
-
Abstract 867: Molecular and functional characterization of melanocyte subpopulations in human epidermis based on single-cell RNA sequencing
Peinan Zhao,Fumihito Noguchi,Christopher Chew,Gamze Kuser Abali,Pacman Szeto,Youfang Zhang,Malaka Ameratunga,Isobel Leece,Jen G. Cheung,Miles Andrews,Nicholas C. Wong,Anthony T. Papenfuss,Mark Shackleton
DOI: https://doi.org/10.1158/1538-7445.am2024-867
IF: 11.2
2024-03-22
Cancer Research
Abstract:Abstract The biological and molecular mechanisms that underpin the malignant transformation of normal melanocytes to melanomas are largely unknown. In part, this is due to the limited understanding of normal human melanocyte homeostasis and how melanocytes respond to oncogenic insults such as ultraviolet radiation (UVR). This is particularly true for interfollicular epidermal melanocytes, which have the highest levels of UVR exposure and from whence most melanomas are thought to arise. These knowledge gaps impede the development of strategies for active, targeted prevention of melanoma formation. We thus evaluated epidermal melanocytes transcriptionally, phenotypically and functionally after isolating them from human skin. Using single-cell RNA sequencing (scRNA-seq), we identified multiple transcriptionally distinct subpopulations within human epidermal melanocytes. RNA velocity analysis revealed subpopulations in different states of melanocytic differentiation, and immunohistochemistry staining demonstrated their distinct anatomical distribution throughout follicular and interfollicular epidermal compartments. Notably, one melanocyte subgroup, marked by increased expression of neurotrophic receptor tyrosine kinase 2 (NTRK2) and genes associated with ribosome biogenesis, exhibited molecular characteristics of progenitor cells. This subpopulation displayed human embryonic stem cell (hESC)-derived melanoblast markers, and their anatomical localization corresponded to that of intermediate melanocyte progenitors. NTRK2+ melanocytes demonstrated enhanced clonogenicity after UVR exposure in primary cell cultures and in ex vivo whole skin explants. In contrast, NTRK2- melanocytes were suppressed by UVR. Furthermore, scRNA-seq data of ex vivo melanocytes after UVR exposure revealed the upregulation of genes associated with cell proliferation within NTRK2-expressing melanocytes. In mouse back skin, the ratio of Ntrk2+ melanocytes increased within 24 hours of UVB irradiation, suggesting a proliferative response in these cells in vivo following UVR. We thus report the discovery in human epidermis of a putative melanocytic cell hierarchy, and of a candidate melanocyte progenitor subpopulation that responds proliferatively to UVR and is thus a candidate cell of origin of melanoma. Citation Format: Peinan Zhao, Fumihito Noguchi, Christopher Chew, Gamze Kuser Abali, Pacman Szeto, Youfang Zhang, Malaka Ameratunga, Isobel Leece, Jen G. Cheung, Miles Andrews, Nicholas C. Wong, Anthony T. Papenfuss, Mark Shackleton. Molecular and functional characterization of melanocyte subpopulations in human epidermis based on single-cell RNA sequencing [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular Abstracts); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl):Abstract nr 867.
oncology
-
SOX2 Is a Marker for Stem-like Tumor Cells in Bladder Cancer.
Fengyu Zhu,Weiqing Qian,Haojie Zhang,Yu Liang,Mingqing Wu,Yingyin Zhang,Xiuhong Zhang,Qian Gao,Yang Li
DOI: https://doi.org/10.1016/j.stemcr.2017.07.004
2017-01-01
Stem Cell Reports
Abstract:It has been reported that functionally distinct cancer stem cells (CSCs) exist in human bladder cancer (BCa). Here, we found that Sox2, a transcription factor that is well characterized as a marker for stem cells, is upregulated in both mouse and human BCa. Sox2 expression is absent in normal urothelial cells, but it begins to be expressed in pre-neoplastic bladder tumors and continues to be expressed in invasive mouse BCa. Using s as a reporter of Sox2 transcriptional expression, we demonstrated that Sox2-expressing cells mark a subpopulation of tumor cells that fuel the growth of established BCa. SOX2-positive cells also expressed other previously reported BCa CSC markers, including Keratin14 (KRT14) and CD44v6. Ablation of Sox2-expressing cells within primary invasive BCa led to enhanced tumor regression, supporting the essential role of SOX2-positive cells in regulating BCa maintenance and progression. Our data show that Sox2 is a marker of bladder CSCs and indicate it as a potential clinical target for BCa therapy.
-
SOX2 boosts major tumor progression genes in prostate cancer and is a functional biomarker of lymph node metastasis
Marco Vincenzo Russo,Silvia Esposito,Maria Grazia Tupone,Lamberto Manzoli,Irma Airoldi,Paolo Pompa,Luca Cindolo,Luigi Schips,Carlo Sorrentino,Emma Di Carlo
DOI: https://doi.org/10.18632/oncotarget.6029
2015-10-19
Oncotarget
Abstract:Critical issues in prostate cancer (PC) are a. identification of molecular drivers of the highly aggressive neuroendocrine differentiation (NED) in adenocarcinoma, and b. early assessment of disease progression. The SRY (sex determining region Y)-box 2 gene, SOX2, is an essential embryonic stem cell gene involved in prostate tumorigenesis. Here we assessed its implications in NED and progression of PC and its diagnostic and prognostic value. Laser microdissection, qRT-PCR, quantitative Methylation-Specific PCR and immunohistochemistry were used to analyze SOX2 gene expression and regulation in 206 PC samples. Results were examined according to the patient's clinical pathological profile and follow-ups. Functional studies were performed using PC cells transfected to overexpress or silence SOX2. SOX2 was consistently downregulated in PC, except in cell clusters lying within lymph node (LN)-positive PC. Multivariate analysis revealed that SOX2 mRNA expression in the primary tumor was significantly associated with LN metastasis. When SOX2 mRNA levels were ≥1.00, relative to (XpressRef) Universal Total RNA, adjusted Odds Ratio was 24.4 (95% CI: 7.54-79.0), sensitivity 0.81 (95% CI: 0.61-0.93) and specificity 0.87 (95% CI: 0.81-0.91). Patients experiencing biochemical recurrence had high median levels of SOX2 mRNA. In both PC and LN metastasis, SOX2 and NED marker, Chromogranin-A, were primarily co-expressed. In PC cells, NED genes were upregulated by SOX2 overexpression and downregulated by its silencing, which also abolished SNAI2/Slug dependent NED. Moreover, SOX2 upregulated neural CAMs, neurotrophins/neurotrophin receptors, pluripotency and epithelial-mesenchymal transition transcription factors, growth, angiogenic and lymphangiogenic factors, and promoted PC cell invasiveness and motility. This study discloses novel SOX2 target genes driving NED and spread of PC and proposes SOX2 as a functional biomarker of LN metastasization for PC.
-
Expression Profile of Embryonic Stem Cell-Associated Genes Oct4, Sox2 and Nanog in Human Gliomas.
Yuji Guo,Shangming Liu,Ping Wang,Shidou Zhao,Fuwu Wang,Lujun Bing,Yanmin Zhang,Eng-Ang Ling,Jiangang Gao,Aijun Hao
DOI: https://doi.org/10.1111/j.1365-2559.2011.03993.x
2011-01-01
Histopathology
Abstract:AIMS:To investigate whether Oct4, Sox2 and Nanog, three core regulatory factors maintaining pluripotency and self-renewal of embryonic stem cells (ESCs), are coexpressed in human gliomas, and whether their expression might be linked to carcinogenesis and the formation of cancer stem cells (CSCs).METHODS AND RESULTS:Forty cases of human glioma were examined. The expression of Oct4, Sox2 and Nanog was analysed by immunohistochemistry, reverse transcription polymerase chain reaction and western blot. We found a positive correlation between the expression levels of Oct4, Sox2 and Nanog and tumour malignancy. Immunohistochemistry showed that Oct4 and Nanog were expressed in both the nuclei and the cytoplasm of glioma cells, whereas Sox2 was expressed only in the nuclei. Double immunofluorescence staining revealed that a majority of Oct4-positive cells coexpressed Sox2 and Nanog. More than 50% of Oct4-positive cells coexpressed the putative CSC markers CD133 and Nestin. Moreover, some cells exhibited Oct4 and Nanog immunoexpression in the cytoplasm, but the frequency of positive cells did not correlate with tumour malignancy.CONCLUSIONS:The present findings suggest that ESC-associated pathways are activated in human gliomas and that these may be involved in glioma progression, a role that is distinct from that in ESCs.
-
Sox2 Promotes Tumorigenicity and Inhibits the Differentiation of I-Type Neuroblastoma Cells
Shaobo Yang,Jicui Zheng,Xianmin Xiao,Ting Xu,Weitao Tang,Haitao Zhu,Lin Yang,Shan Zheng,Kuiran Dong,Guomin Zhou,Yang Wang
DOI: https://doi.org/10.3892/ijo.2014.2713
2014-01-01
International Journal of Oncology
Abstract:SOX2 is a transcription factor associated with the pluripotency, proliferative potential, and self-renewing properties observed with embryonic stem cells and germ cells. SOX2 expression has been reported in several cancers and is implicated in tumorigenesis. We previously found that SOX2 expression was correlated to the clinical stage of neuroblastoma. Recently, we found that SOX2 overexpression occurs in I-type neuroblastoma cells (BE(2)-C cells). To elucidate the tumorigenic function of SOX2, we established a SOX2 overexpressed BE(2)-C cell line. SOX2 overexpressed cells showed higher tumorigenicity than control cells and exhibited decreased expression levels of marker proteins of N- or S-type cells after agent-induced differetiation. By contrast, in cells where SOX2 mRNA expression was knocked down by gene-specific siRNA, tumorigenicty was significantly decreased and the expression levels of marker proteins of N- or S-type cells were upregulated. In conclusion, our findings indicate an important function for SOX2 in promoting tumorigenicity of I-type neuroblastoma cells and in inhibiting their differentiation, suggesting that SOX2 might be a potential therapeutic target in neuroblastoma.