1
|
Oka T, Smith SS, Oliver-Garcia VS, Lee T, Son HG, Mortaja M, Azin M, Garza-Mayers AC, Huang JT, Nazarian RM, Horn TD, Demehri S. Epigenomic regulation of stemness contributes to the low immunogenicity of the most mutated human cancer. Cell Rep 2025:115561. [PMID: 40250424 DOI: 10.1016/j.celrep.2025.115561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 12/02/2024] [Accepted: 03/24/2025] [Indexed: 04/20/2025] Open
Abstract
Despite harboring the highest tumor mutational burden of all cancers, basal cell carcinoma (BCC) has low immunogenicity. Here, we demonstrate that BCC's low immunogenicity is associated with epigenomic suppression of antigen presentation machinery reminiscent of its cell of origin. Primary BCC had low T cell infiltrates and low human leukocyte antigen class I (HLA-I) expression compared with cutaneous squamous cell carcinoma (SCC) and normal keratinocytes. Forkhead box C1 (Foxc1), a regulator of quiescence in hair follicle stem cells, was expressed in BCC. Foxc1 bound to promoter of interferon regulatory factor 1 and HLA-I genes, leading to their deacetylation and reduced expression. A histone deacetylase inhibitor, entinostat, overcame Foxc1's effect and upregulated HLA-I in BCC. Topical entinostat plus imiquimod immunotherapy blocked BCC development in mice. Collectively, our findings demonstrate that low BCC immunogenicity is associated with a stem-like quiescent program preserved in the tumor cells, which can be blocked to enable BCC immunotherapy.
Collapse
Affiliation(s)
- Tomonori Oka
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Sabrina S Smith
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Valeria S Oliver-Garcia
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Truelian Lee
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Heehwa G Son
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Mahsa Mortaja
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Marjan Azin
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Anna C Garza-Mayers
- Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jennifer T Huang
- Dermatology Section, Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Rosalynn M Nazarian
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Thomas D Horn
- Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Shadmehr Demehri
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Chen W, Lai F, Xu J. Synthesis and Evaluation of Aromatic A-Ring 23-Oxavitamin D 3 Analogues as Hedgehog Pathway Inhibitors. Int J Mol Sci 2025; 26:1631. [PMID: 40004093 PMCID: PMC11855207 DOI: 10.3390/ijms26041631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/08/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
The Hedgehog (Hh) signaling pathway plays a crucial role in the initiation and progression of tumors, and Hh inhibitors have been used as potential chemotherapeutic agents for the treatment of basal cell carcinomas (BCCs). Vitamin D3 (VD3) and its derivatives have been identified as potent Hh inhibitors. However, the selectivity of VD3 derivatives to vitamin D receptor (VDR) and the Hh signaling pathway still needs optimization. In this study, a series of aromatic A-ring mimics VD3 analogues that contain a C-23 oxygen atom or incorporate C-25 hydroxyl on side chains were designed and synthesized. These compounds were tested in various cell lines for anti-Hh activity, with analogues 3j and 4i identified as potent inhibitors. Mechanism studies showed their anti-Hh effects are mainly due to targeting Smoothened (Smo) without binding to the cyclopamine site. Structure-activity relationship (SAR) studies revealed that VD3-based inhibitors enhance anti-Hh activity by adding a hydroxyl group at C25 while reducing VDR activity by incorporating an oxygen atom into the side chain.
Collapse
Affiliation(s)
- Wang Chen
- School of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong 723000, China; (F.L.); (J.X.)
- Shaanxi Engineering Research Center of Natural Active Products Industrialization, Hanzhong 723000, China
| | - Feifan Lai
- School of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong 723000, China; (F.L.); (J.X.)
- Shaanxi Engineering Research Center of Natural Active Products Industrialization, Hanzhong 723000, China
| | - Jianghe Xu
- School of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong 723000, China; (F.L.); (J.X.)
- Shaanxi Engineering Research Center of Natural Active Products Industrialization, Hanzhong 723000, China
| |
Collapse
|
3
|
Haensel D, Daniel B, Gaddam S, Pan C, Fabo T, Bjelajac J, Jussila AR, Gonzalez F, Li NY, Chen Y, Hou J, Patel T, Aasi S, Satpathy AT, Oro AE. Skin basal cell carcinomas assemble a pro-tumorigenic spatially organized and self-propagating Trem2+ myeloid niche. Nat Commun 2023; 14:2685. [PMID: 37164949 PMCID: PMC10172319 DOI: 10.1038/s41467-023-37993-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 04/07/2023] [Indexed: 05/12/2023] Open
Abstract
Cancer immunotherapies have revolutionized treatment but have shown limited success as single-agent therapies highlighting the need to understand the origin, assembly, and dynamics of heterogeneous tumor immune niches. Here, we use single-cell and imaging-based spatial analysis to elucidate three microenvironmental neighborhoods surrounding the heterogeneous basal cell carcinoma tumor epithelia. Within the highly proliferative neighborhood, we find that TREM2+ skin cancer-associated macrophages (SCAMs) support the proliferation of a distinct tumor epithelial population through an immunosuppression-independent manner via oncostatin-M/JAK-STAT3 signaling. SCAMs represent a unique tumor-specific TREM2+ population defined by VCAM1 surface expression that is not found in normal homeostatic skin or during wound healing. Furthermore, SCAMs actively proliferate and self-propagate through multiple serial tumor passages, indicating long-term potential. The tumor rapidly drives SCAM differentiation, with intratumoral injections sufficient to instruct naive bone marrow-derived monocytes to polarize within days. This work provides mechanistic insights into direct tumor-immune niche dynamics independent of immunosuppression, providing the basis for potential combination tumor therapies.
Collapse
Affiliation(s)
- Daniel Haensel
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Bence Daniel
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, 94158, USA
| | - Sadhana Gaddam
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Cory Pan
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Tania Fabo
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jeremy Bjelajac
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Anna R Jussila
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Fernanda Gonzalez
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Nancy Yanzhe Li
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yun Chen
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - JinChao Hou
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Tiffany Patel
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sumaira Aasi
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ansuman T Satpathy
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, 94158, USA
- Parker Institute of Cancer Immunotherapy, San Francisco, CA, 94305, USA
| | - Anthony E Oro
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
4
|
Wen J, Charan Dash R, Zaino AM, Harrahill NJ, Calhoun JT, Dusek CO, Morel SR, Russolillo M, Kyle Hadden M. 8-Hydroxyquinoline derivatives suppress GLI1-mediated transcription through multiple mechanisms. Bioorg Chem 2023; 132:106387. [PMID: 36724660 DOI: 10.1016/j.bioorg.2023.106387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/03/2023] [Accepted: 01/17/2023] [Indexed: 01/22/2023]
Abstract
Aberrant activation of the Hedgehog (Hh) signaling pathway has been observed in various human malignancies. Glioma-associated oncogene transcription factor 1 (GLI1) is the ultimate effector of the canonical Hh pathway and has also been identified as a common regulator of several tumorigenic pathways prevalent in Hh-independent cancers. The anti-cancer potential of GLI1 antagonism with small molecule inhibitors has demonstrated initial promise; however, the continued development of GLI1 inhibitors is still needed. We previously identified a scaffold containing an 8-hydroxyquinoline as a promising lead GLI1 inhibitor (compound 1). To further develop this scaffold, we performed a systematic structure-activity relationship study to map the structural requirements of GLI1 inhibition by this chemotype. A series of biophysical and cellular experiments identified compound 39 as an enhanced GLI1 inhibitor with improved activity. In addition, our studies on this scaffold suggest a potential role for SRC family kinases in regulating oncogenic GLI1 transcriptional activity.
Collapse
Affiliation(s)
- Jiachen Wen
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Rd, Unit 3092, Storrs, CT 06029-3092, United States
| | - Radha Charan Dash
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Rd, Unit 3092, Storrs, CT 06029-3092, United States
| | - Angela M Zaino
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Rd, Unit 3092, Storrs, CT 06029-3092, United States
| | - Noah J Harrahill
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Rd, Unit 3092, Storrs, CT 06029-3092, United States
| | - Jackson T Calhoun
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Rd, Unit 3092, Storrs, CT 06029-3092, United States
| | - Christopher O Dusek
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Rd, Unit 3092, Storrs, CT 06029-3092, United States
| | - Shana R Morel
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Rd, Unit 3092, Storrs, CT 06029-3092, United States
| | - Matthew Russolillo
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Rd, Unit 3092, Storrs, CT 06029-3092, United States
| | - M Kyle Hadden
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Rd, Unit 3092, Storrs, CT 06029-3092, United States.
| |
Collapse
|
5
|
Haensel D, Gaddam S, Li NY, Gonzalez F, Patel T, Cloutier JM, Sarin KY, Tang JY, Rieger KE, Aasi SZ, Oro AE. LY6D marks pre-existing resistant basosquamous tumor subpopulations. Nat Commun 2022; 13:7520. [PMID: 36473848 PMCID: PMC9726704 DOI: 10.1038/s41467-022-35020-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 11/15/2022] [Indexed: 12/12/2022] Open
Abstract
Improved response to canonical therapies requires a mechanistic understanding of dynamic tumor heterogeneity by identifying discrete cellular populations with enhanced cellular plasticity. We have previously demonstrated distinct resistance mechanisms in skin basal cell carcinomas, but a comprehensive understanding of the cellular states and markers associated with these populations remains poorly understood. Here we identify a pre-existing resistant cellular population in naive basal cell carcinoma tumors marked by the surface marker LY6D. LY6D+ tumor cells are spatially localized and possess basal cell carcinoma and squamous cell carcinoma-like features. Using computational tools, organoids, and spatial tools, we show that LY6D+ basosquamous cells represent a persister population lying on a central node along the skin lineage-associated spectrum of epithelial states with local environmental and applied therapies determining the kinetics of accumulation. Surprisingly, LY6D+ basosquamous populations exist in many epithelial tumors, such as pancreatic adenocarcinomas, which have poor outcomes. Overall, our results identify the resistant LY6D+ basosquamous population as an important clinical target and suggest strategies for future therapeutic approaches to target them.
Collapse
Affiliation(s)
- Daniel Haensel
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sadhana Gaddam
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Nancy Y Li
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Fernanda Gonzalez
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Tiffany Patel
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jeffrey M Cloutier
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kavita Y Sarin
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jean Y Tang
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kerri E Rieger
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sumaira Z Aasi
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Anthony E Oro
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
6
|
Slominski AT, Brożyna AA, Kim TK, Elsayed MM, Janjetovic Z, Qayyum S, Slominski RM, Oak AS, Li C, Podgorska E, Li W, Jetten AM, Tuckey RC, Tang EK, Elmets C, Athar M. CYP11A1‑derived vitamin D hydroxyderivatives as candidates for therapy of basal and squamous cell carcinomas. Int J Oncol 2022; 61:96. [PMID: 35775377 PMCID: PMC9262157 DOI: 10.3892/ijo.2022.5386] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/23/2022] [Indexed: 12/14/2022] Open
Abstract
Hydroxyderivatives of vitamin D3, including classical 1,25(OH)2D3 and novel CYP11A1‑derived hydroxyderivatives, exert their biological activity by acting as agonists on the vitamin D receptor (VDR) and inverse agonists on retinoid‑related orphan receptors (ROR)α and γ. The anticancer activities of CYP11A1‑derived hydroxyderivatives were tested using cell biology, tumor biology and molecular biology methods in human A431 and SCC13 squamous (SCC)‑ and murine ASZ001 basal (BCC)‑cell carcinomas, in comparison with classical 1,25(OH)2D3. Vitamin D3‑hydroxyderivatives with or without a C1α(OH) inhibited cell proliferation in a dose‑dependent manner. While all the compounds tested had similar effects on spheroid formation by A431 and SCC13 cells, those with a C1α(OH) group were more potent in inhibiting colony and spheroid formation in the BCC line. Potent anti‑tumorigenic activity against the BCC line was exerted by 1,25(OH)2D3, 1,20(OH)2D3, 1,20,23(OH)3D3, 1,20,24(OH)3D3, 1,20,25(OH)3D3 and 1,20,26(OH)3D3, with smaller effects seen for 25(OH)D3, 20(OH)D3 and 20,23(OH)2D3. 1,25(OH)2D3, 1,20(OH)2D3 and 20(OH)D3 inhibited the expression of GLI1 and β‑catenin in ASZ001 cells. In A431 cells, these compounds also decreased the expression of GLI1 and stimulated involucrin expression. VDR, RORγ, RORα and CYP27B1 were detected in A431, SCC13 and ASZ001 lines, however, with different expression patterns. Immunohistochemistry performed on human skin with SCC and BCC showed nuclear expression of all three of these receptors, as well as megalin (transmembrane receptor for vitamin D‑binding protein), the level of which was dependent on the type of cancer and antigen tested in comparison with normal epidermis. Classical and CYP11A1‑derived vitamin D3‑derivatives exhibited anticancer‑activities on skin cancer cell lines and inhibited GLI1 and β‑catenin signaling in a manner that was dependent on the position of hydroxyl groups. The observed expression of VDR, RORγ, RORα and megalin in human SCC and BCC suggested that they might provide targets for endogenously produced or exogenously applied vitamin D hydroxyderivatives and provide excellent candidates for anti‑cancer therapy.
Collapse
Affiliation(s)
- Andrzej T. Slominski
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35292, USA
- VA Medical Center, Birmingham, AL 35233, USA
| | - Anna A. Brożyna
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35292, USA
- Department of Human Biology, Institute of Biology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Toruń 87-100, Poland
| | - Tae-Kang Kim
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35292, USA
| | - Mahmoud M. Elsayed
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35292, USA
| | - Zorica Janjetovic
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35292, USA
| | - Shariq Qayyum
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35292, USA
| | - Radomir M. Slominski
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35292, USA
| | - Allen S.W. Oak
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35292, USA
| | - Changzhao Li
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35292, USA
| | - Ewa Podgorska
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35292, USA
| | - Wei Li
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Anton M. Jetten
- Cell Biology Section, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Robert C. Tuckey
- School of Molecular Sciences, The University of Western Australia, Perth, Western Australia 6009, Australia
| | - Edith K.Y. Tang
- School of Molecular Sciences, The University of Western Australia, Perth, Western Australia 6009, Australia
| | - Craig Elmets
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35292, USA
| | - Mohammad Athar
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35292, USA
| |
Collapse
|
7
|
Nicolás-Morala J, Portillo-Esnaola M, Terrén S, Gutiérrez-Pérez M, Gilaberte Y, González S, Juarranz Á. In vitro 5-Fluorouracil resistance produces enhanced photodynamic therapy damage in SCC and tumor resistance in BCC. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2022; 233:112483. [PMID: 35679749 DOI: 10.1016/j.jphotobiol.2022.112483] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 05/10/2022] [Accepted: 05/25/2022] [Indexed: 06/15/2023]
Abstract
Non-melanoma skin cancer (NMSC) is the most common malignancy worldwide, with rising incidence in the recent years. It includes basal cell carcinoma (BCC), and squamous cell carcinoma (SCC). Several non-invasive therapies have been developed for its treatment such as topical 5-Fluorouracil (5FU) and photodynamic therapy (PDT), among others. Despite both are appropriated for NMSC treatment, recurrence cases have been reported. To prevent this, in this work we explore the potential of the combination of PDT and 5FU to treat SCC and BCC. First we evaluate the efficacy of PDT in cells resistant to 5FU. For this purpose, we use SCC-13 and CSZ-1 cells, obtained from a human SCC and a murine BCC, respectively. We first induced 5FU resistance in these cell lines by repeated treatments with the drug and then, the efficacy to PDT was evaluated. The results obtained indicated that SCC-5FU resistant cells were sensible to PDT administration, whereas BCC-5FU resistant cells were also resistant to PDT. The observed responses in both cell lines are in concordance to Protoporphyrin IX (PpIX) and reactive oxygen species (ROS) levels produced after the incubation with MAL and subsequent light exposure. The obtained data support the fact that PDT seems to be an appropriate therapeutic option to be administered after 5FU resistance in SCC. However, PDT would not be a choice therapy for resistant BCC cells to 5FU.
Collapse
Affiliation(s)
- Jimena Nicolás-Morala
- Department of Biology, Faculty of Sciences, Autónoma University of Madrid (UAM), Madrid 28049, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid 28034, Spain.
| | - Mikel Portillo-Esnaola
- Department of Biology, Faculty of Sciences, Autónoma University of Madrid (UAM), Madrid 28049, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid 28034, Spain
| | - Samuel Terrén
- Department of Biology, Faculty of Sciences, Autónoma University of Madrid (UAM), Madrid 28049, Spain
| | - María Gutiérrez-Pérez
- Department of Biology, Faculty of Sciences, Autónoma University of Madrid (UAM), Madrid 28049, Spain
| | | | - Salvador González
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid 28034, Spain; Department of Medicine and Medical Specialties, Alcalá de Henares University, 28805 Madrid, Spain.
| | - Ángeles Juarranz
- Department of Biology, Faculty of Sciences, Autónoma University of Madrid (UAM), Madrid 28049, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid 28034, Spain.
| |
Collapse
|
8
|
Mendoza M, Tran U, Zhang GC, Leister J, To K, Malepeai-Tofaeono T, Ondrus AE, Billingsley KL. Indolactam Dipeptides as Nanomolar Gli Inhibitors. ACS Med Chem Lett 2022; 13:1036-1042. [DOI: 10.1021/acsmedchemlett.1c00562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Affiliation(s)
- Manuel Mendoza
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, California 92831, United States
| | - UyenPhuong Tran
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, California 92831, United States
| | - Grace C. Zhang
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125, United States
| | - Jeffrey Leister
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, California 92831, United States
| | - Kyle To
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, California 92831, United States
| | - Theodore Malepeai-Tofaeono
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, California 92831, United States
| | - Alison E. Ondrus
- Department of Chemistry, University of Illinois Chicago, Chicago, Illinois 60607, United States
| | - Kelvin L. Billingsley
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, California 92831, United States
- Department of Chemistry and Biochemistry, Loyola University Chicago, Chicago, Illinois 60660, United States
| |
Collapse
|
9
|
Wnt Inhibitory Factor 1 Binds to and Inhibits the Activity of Sonic Hedgehog. Cells 2021; 10:cells10123496. [PMID: 34944004 PMCID: PMC8699845 DOI: 10.3390/cells10123496] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/07/2021] [Accepted: 12/09/2021] [Indexed: 12/17/2022] Open
Abstract
The hedgehog (Hh) and Wnt pathways, crucial for the embryonic development and stem cell proliferation of Metazoa, have long been known to have similarities that argue for their common evolutionary origin. A surprising additional similarity of the two pathways came with the discovery that WIF1 proteins are involved in the regulation of both the Wnt and Hh pathways. Originally, WIF1 (Wnt Inhibitory Factor 1) was identified as a Wnt antagonist of vertebrates, but subsequent studies have shown that in Drosophila, the WIF1 ortholog serves primarily to control the distribution of Hh. In the present, work we have characterized the interaction of the human WIF1 protein with human sonic hedgehog (Shh) using Surface Plasmon Resonance spectroscopy and reporter assays monitoring the signaling activity of human Shh. Our studies have shown that human WIF1 protein binds human Shh with high affinity and inhibits its signaling activity efficiently. Our observation that the human WIF1 protein is a potent antagonist of human Shh suggests that the known tumor suppressor activity of WIF1 may not be ascribed only to its role as a Wnt inhibitor.
Collapse
|
10
|
Kuonen F, Li NY, Haensel D, Patel T, Gaddam S, Yerly L, Rieger K, Aasi S, Oro AE. c-FOS drives reversible basal to squamous cell carcinoma transition. Cell Rep 2021; 37:109774. [PMID: 34610301 PMCID: PMC8515919 DOI: 10.1016/j.celrep.2021.109774] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/28/2021] [Accepted: 09/08/2021] [Indexed: 01/22/2023] Open
Abstract
While squamous transdifferentiation within subpopulations of adenocarcinomas represents an important drug resistance problem, its underlying mechanism remains poorly understood. Here, using surface markers of resistant basal cell carcinomas (BCCs) and patient single-cell and bulk transcriptomic data, we uncover the dynamic roadmap of basal to squamous cell carcinoma transition (BST). Experimentally induced BST identifies activator protein 1 (AP-1) family members in regulating tumor plasticity, and we show that c-FOS plays a central role in BST by regulating the accessibility of distinct AP-1 regulatory elements. Remarkably, despite prominent changes in cell morphology and BST marker expression, we show using inducible model systems that c-FOS-mediated BST demonstrates reversibility. Blocking EGFR pathway activation after c-FOS induction partially reverts BST in vitro and prevents BST features in both mouse models and human tumors. Thus, by identifying the molecular basis of BST, our work reveals a therapeutic opportunity targeting plasticity as a mechanism of tumor resistance.
Collapse
MESH Headings
- Animals
- Carcinoma, Basal Cell/metabolism
- Carcinoma, Basal Cell/pathology
- Carcinoma, Basal Cell/veterinary
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/veterinary
- Cell Transdifferentiation/drug effects
- Chromatin Assembly and Disassembly
- Drug Resistance, Neoplasm/genetics
- Humans
- Male
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Mucin-1/metabolism
- Protein Kinase Inhibitors/pharmacology
- Proto-Oncogene Proteins c-fos/antagonists & inhibitors
- Proto-Oncogene Proteins c-fos/genetics
- Proto-Oncogene Proteins c-fos/metabolism
- RNA Interference
- RNA, Small Interfering/metabolism
- Signal Transduction/drug effects
- Transcription Factor AP-1/metabolism
- Transforming Growth Factor beta/antagonists & inhibitors
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/metabolism
- ras Proteins/genetics
- ras Proteins/metabolism
Collapse
Affiliation(s)
- François Kuonen
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA; Department of Dermatology and Venereology, Hôpital de Beaumont, Lausanne University Hospital Center, 1011 Lausanne, Switzerland.
| | - Nancy Yanzhe Li
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Daniel Haensel
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Tiffany Patel
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sadhana Gaddam
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Laura Yerly
- Department of Dermatology and Venereology, Hôpital de Beaumont, Lausanne University Hospital Center, 1011 Lausanne, Switzerland
| | - Kerri Rieger
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sumaira Aasi
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Anthony E Oro
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
11
|
Patmanathan SN, Tong BT, Jackie Teo JH, Jonathan Ting YZ, Tan NS, Kenice Sim SH, Ta YC, Woo WM. A PDZ Protein GIPC3 Positively Modulates Hedgehog Signaling and Melanoma Growth. J Invest Dermatol 2021; 142:179-188.e4. [PMID: 34224745 DOI: 10.1016/j.jid.2021.04.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 04/23/2021] [Accepted: 04/28/2021] [Indexed: 12/30/2022]
Abstract
The Hedgehog (Hh) pathway is essential for animal development but aberrant activation promotes cancer growth. Here we show that GIPC3, a PDZ domain-containing protein with putative adaptor protein function, positively modulates Hh target gene expression in normal fibroblasts and melanoma cells and supports melanoma tumor growth. Using overexpression and epistasis studies, we show that Gipc3 potentiates Hh transcriptional output and it modulates GLI-dependent transcription independently of Sufu. While we find GIPC3 protein does not interact with Hh pathway components, Ingenuity Pathway Analyses of GIPC3-interacting proteins identified by co-immunoprecipitation and mass spectrometry show an association with cancer pathogenesis. Subsequent interrogation of TCGA and The Human Protein Atlas databases reveals GIPC3 upregulation in many cancers. Using expression screens in selected groups of GIPC3-upregulated cancers with reported Hh pathway activation, we find a significant positive correlation of GIPC3 expression with Hh pathway components GLI1, GLI2, and GPR161, in melanoma lines. Consistently, GIPC3 knockdown in melanoma lines significantly reduces GLI1 and GLI2 expression, cell viability, colony formation, and allograft tumor growth. Our findings highlight previously unidentified roles of Gipc3 in potentiating Hh response and melanoma tumorigenesis, and suggest that GIPC3 modulation on Hh signaling may be targeted to reduce melanoma growth.
Collapse
Affiliation(s)
| | - Bing Teck Tong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; School of Chemical and Life Sciences, Singapore Polytechnic, Singapore
| | - Jia Hao Jackie Teo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore
| | | | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore
| | | | - Yng-Cun Ta
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Wei-Meng Woo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.
| |
Collapse
|
12
|
Chow RY, Jeon US, Levee TM, Kaur G, Cedeno DP, Doan LT, Atwood SX. PI3K Promotes Basal Cell Carcinoma Growth Through Kinase-Induced p21 Degradation. Front Oncol 2021; 11:668247. [PMID: 34268113 PMCID: PMC8276170 DOI: 10.3389/fonc.2021.668247] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 06/14/2021] [Indexed: 12/26/2022] Open
Abstract
Basal cell carcinoma (BCC) is a locally invasive epithelial cancer that is primarily driven by the Hedgehog (HH) pathway. Advanced BCCs are a critical subset of BCCs that frequently acquire resistance to Smoothened (SMO) inhibitors and identifying pathways that bypass SMO could provide alternative treatments for patients with advanced or metastatic BCC. Here, we use a combination of RNA-sequencing analysis of advanced human BCC tumor-normal pairs and immunostaining of human and mouse BCC samples to identify a PI3K pathway expression signature in BCC. Pharmacological inhibition of PI3K activity in BCC cells significantly reduces cell proliferation and HH signaling. However, treatment of Ptch1fl/fl; Gli1-CreERT2 mouse BCCs with the PI3K inhibitor BKM120 results in a reduction of tumor cell growth with no significant effect on HH signaling. Downstream PI3K components aPKC and Akt1 showed a reduction in active protein, whereas their substrate, cyclin-dependent kinase inhibitor p21, showed a concomitant increase in protein stability. Our results suggest that PI3K promotes BCC tumor growth by kinase-induced p21 degradation without altering HH signaling.
Collapse
Affiliation(s)
- Rachel Y Chow
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, United States
| | - Ung Seop Jeon
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, United States
| | - Taylor M Levee
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, United States
| | - Gurleen Kaur
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, United States
| | - Daniel P Cedeno
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, United States
| | - Linda T Doan
- Department of Dermatology, University of California, Irvine, Irvine, CA, United States
| | - Scott X Atwood
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, United States.,Department of Dermatology, University of California, Irvine, Irvine, CA, United States.,Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
13
|
Abstract
The development of molecular nanostructures with well-defined particle size and shape is of eminent interest in biomedicine. Among many studied nanostructures, dendrimers represent the group of those most thoroughly characterized ones. Due to their unique structure and properties, dendrimers are very attractive for medical and pharmaceutical applications. Owing to the controllable cavities inside the dendrimer, guest molecules may be encapsulated, and highly reactive terminal groups are susceptible to further modifications, e.g., to facilitate target delivery. To understand the potential of these nanoparticles and to predict and avoid any adverse cellular reactions, it is necessary to know the mechanisms responsible for an efficient dendrimer uptake and the destination of their intracellular journey. In this article, we summarize the results of studies describing the dendrimer uptake, traffic, and efflux mechanisms depending on features of specific nanoparticles and cell types. We also present mechanisms of dendrimers responsible for toxicity and alteration in signal transduction pathways at the cellular level.
Collapse
Affiliation(s)
- Barbara Ziemba
- Department of Clinical and Laboratory Genetics, Medical University of Lodz, Lodz, Poland
| | - Maciej Borowiec
- Department of Clinical and Laboratory Genetics, Medical University of Lodz, Lodz, Poland
| | - Ida Franiak-Pietryga
- Department of Clinical and Laboratory Genetics, Medical University of Lodz, Lodz, Poland.,Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
14
|
AP-1 and TGFß cooperativity drives non-canonical Hedgehog signaling in resistant basal cell carcinoma. Nat Commun 2020; 11:5079. [PMID: 33033234 PMCID: PMC7546632 DOI: 10.1038/s41467-020-18762-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 09/01/2020] [Indexed: 12/17/2022] Open
Abstract
Tumor heterogeneity and lack of knowledge about resistant cell states remain a barrier to targeted cancer therapies. Basal cell carcinomas (BCCs) depend on Hedgehog (Hh)/Gli signaling, but can develop mechanisms of Smoothened (SMO) inhibitor resistance. We previously identified a nuclear myocardin-related transcription factor (nMRTF) resistance pathway that amplifies noncanonical Gli1 activity, but characteristics and drivers of the nMRTF cell state remain unknown. Here, we use single cell RNA-sequencing of patient tumors to identify three prognostic surface markers (LYPD3, TACSTD2, and LY6D) which correlate with nMRTF and resistance to SMO inhibitors. The nMRTF cell state resembles transit-amplifying cells of the hair follicle matrix, with AP-1 and TGFß cooperativity driving nMRTF activation. JNK/AP-1 signaling commissions chromatin accessibility and Smad3 DNA binding leading to a transcriptional program of RhoGEFs that facilitate nMRTF activity. Importantly, small molecule AP-1 inhibitors selectively target LYPD3+/TACSTD2+/LY6D+ nMRTF human BCCs ex vivo, opening an avenue for improving combinatorial therapies.
Collapse
|
15
|
Mascaraque M, Delgado-Wicke P, Nuevo-Tapioles C, Gracia-Cazaña T, Abarca-Lachen E, González S, Cuezva JM, Gilaberte Y, Juarranz Á. Metformin as an Adjuvant to Photodynamic Therapy in Resistant Basal Cell Carcinoma Cells. Cancers (Basel) 2020; 12:cancers12030668. [PMID: 32183017 PMCID: PMC7139992 DOI: 10.3390/cancers12030668] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/04/2020] [Accepted: 03/09/2020] [Indexed: 01/08/2023] Open
Abstract
Photodynamic Therapy (PDT) with methyl-aminolevulinate (MAL-PDT) is being used for the treatment of Basal Cell Carcinoma (BCC), although resistant cells may appear. Normal differentiated cells depend primarily on mitochondrial oxidative phosphorylation (OXPHOS) to generate energy, but cancer cells switch this metabolism to aerobic glycolysis (Warburg effect), influencing the response to therapies. We have analyzed the expression of metabolic markers (β-F1-ATPase/GAPDH (glyceraldehyde-3-phosphate dehydrogenase) ratio, pyruvate kinase M2 (PKM2), oxygen consume ratio, and lactate extracellular production) in the resistance to PDT of mouse BCC cell lines (named ASZ and CSZ, heterozygous for ptch1). We have also evaluated the ability of metformin (Metf), an antidiabetic type II compound that acts through inhibition of the AMP-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) pathway to sensitize resistant cells to PDT. The results obtained indicated that resistant cells showed an aerobic glycolysis metabolism. The treatment with Metf induced arrest in the G0/G1 phase and a reduction in the lactate extracellular production in all cell lines. The addition of Metf to MAL-PDT improved the cytotoxic effect on parental and resistant cells, which was not dependent on the PS protoporphyrin IX (PpIX) production. After Metf + MAL-PDT treatment, activation of pAMPK was detected, suppressing the mTOR pathway in most of the cells. Enhanced PDT-response with Metf was also observed in ASZ tumors. In conclusion, Metf increased the response to MAL-PDT in murine BCC cells resistant to PDT with aerobic glycolysis.
Collapse
Affiliation(s)
- Marta Mascaraque
- Departamento de Biología, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.M.); (P.D.-W.)
- Instituto Ramón y Cajal de Investigaciones Sanitarias, IRYCIS, 28034 Madrid, Spain
| | - Pablo Delgado-Wicke
- Departamento de Biología, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.M.); (P.D.-W.)
- Instituto Ramón y Cajal de Investigaciones Sanitarias, IRYCIS, 28034 Madrid, Spain
| | - Cristina Nuevo-Tapioles
- Centro de Biología Molecular-Severo Ochoa (CBMSO/CSIC) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER-ISCIII), Universidad Autónoma de Madrid, 28049 Madrid, Spain; (C.N.-T.); (J.M.C.)
| | | | - Edgar Abarca-Lachen
- Facultad de Ciencias de la Salud, Universidad San Jorge, 50830 Villanueva de Gállego, Spain;
| | - Salvador González
- Departmento de Medicina y Especialidades Médicas, Universidad de Alcalá, 28801 Madrid, Spain;
| | - José M. Cuezva
- Centro de Biología Molecular-Severo Ochoa (CBMSO/CSIC) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER-ISCIII), Universidad Autónoma de Madrid, 28049 Madrid, Spain; (C.N.-T.); (J.M.C.)
| | - Yolanda Gilaberte
- Servicio de Dermatología, Hospital Miguel Servet, 50009 Zaragoza, Spain;
| | - Ángeles Juarranz
- Departamento de Biología, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.M.); (P.D.-W.)
- Instituto Ramón y Cajal de Investigaciones Sanitarias, IRYCIS, 28034 Madrid, Spain
- Correspondence:
| |
Collapse
|
16
|
Becker M, Bauer J, Pyczek J, König S, Müllen A, Rabe H, Schön MP, Uhmann A, Hahn H. WIF1 Suppresses the Generation of Suprabasal Cells in Acanthotic Skin and Growth of Basal Cell Carcinomas upon Forced Overexpression. J Invest Dermatol 2020; 140:1556-1565.e11. [PMID: 31987884 DOI: 10.1016/j.jid.2019.11.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 11/06/2019] [Accepted: 11/25/2019] [Indexed: 12/20/2022]
Abstract
We analyzed the role of WIF1 in normal and acanthotic epidermis of 12-O-tetradecanoylphorbol-13-acetate (TPA) or all-trans-retinoic acid (ATRA)-treated and basal cell carcinoma (BCC)-bearing mice. WIF1 protein is located in the follicular infundibulum and interfollicular epidermis (IFE) in murine back skin. Within the hyperplastic epidermis of TPA- or ATRA-treated or BCC-bearing murine skin, WIF1 and Keratin 10 overlap in Ki67⁻ suprabasal layers, while basal epidermal layers expressing Ki67, and BCCs expressing Wif1 mRNA, are free of WIF1 protein. This is similar in human skin, with the exception that WIF1 protein is found in single Ki67⁻ basal epidermal cells in normal skin and additionally in Ki67+ cells in acanthotic skin. Wif1-deficiency enhances acanthosis of the murine BCC-associated epidermis, which is accompanied by an increase of Ki67+ and of Sca-1+ basal cells. WIF1 overexpression in allografted BCC-derived keratinocytes prevents growth and keratinization, involving enhanced phosphorylation of protein kinase C and extracellular signal-regulated kinase 1 and arguably factors secreted by the in vivo environment. In summary, WIF1 protein marks suprabasal layers in the normal IFE. It is also present in the epidermis overlaying BCCs where it diminishes proliferation of basal cells and production of differentiating suprabasal cells. In addition, WIF1 can prevent proliferation and keratinization of BCC-related keratinocytes.
Collapse
Affiliation(s)
- Marco Becker
- Department of Human Genetics, University Medical Center Goettingen, Goettingen, Germany
| | - Julia Bauer
- Department of Human Genetics, University Medical Center Goettingen, Goettingen, Germany
| | - Joanna Pyczek
- Department of Human Genetics, University Medical Center Goettingen, Goettingen, Germany
| | - Simone König
- Department of Human Genetics, University Medical Center Goettingen, Goettingen, Germany
| | - Anna Müllen
- Department of Human Genetics, University Medical Center Goettingen, Goettingen, Germany
| | - Hanna Rabe
- Department of Human Genetics, University Medical Center Goettingen, Goettingen, Germany
| | - Michael P Schön
- Department of Dermatology, Venereology and Allergology, University Medical Center Goettingen, Goettingen, Germany
| | - Anja Uhmann
- Department of Human Genetics, University Medical Center Goettingen, Goettingen, Germany
| | - Heidi Hahn
- Department of Human Genetics, University Medical Center Goettingen, Goettingen, Germany.
| |
Collapse
|
17
|
Biomarkers of basal cell carcinoma resistance to methyl-aminolevulinate photodynamic therapy. PLoS One 2019; 14:e0215537. [PMID: 31017970 PMCID: PMC6481917 DOI: 10.1371/journal.pone.0215537] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 04/03/2019] [Indexed: 01/15/2023] Open
Abstract
Background Methyl-aminolevulinate photodynamic therapy (MAL-PDT) is an excellent option for the treatment of basal cell carcinoma (BCC). However, up to 25% of cases are resistant to this treatment modality. Objective The aim of this study was to identify potential biomarkers of BCC response to MAL-PDT. Material and methods Clinical, histological, and immunohistochemical (p53, Ki-67, CD-31, COX2, β-catenin, EGFR, and survivin) variables were analyzed in a retrospective study of consecutive BCC patients treated with MAL-PDT at the San Jorge Hospital, Huesca, Spain between January 2006 and December 2015. To deepen on these markers, the effects on p53 and cyclin D1 expression, in vitro response to MAL-PDT of 2 murine BCC cell lines (ASZ and BSZ), was also evaluated. Results The retrospective study examined the response to MAL-PDT of 390 BCCs from 182 patients. The overall clinical response rate was 82.8%, with a mean follow-up time of 35.96 months (SD = 23.46). Immunohistochemistry revealed positive p53 in 84.6% of responders but only 15.4% of nonresponsive tumors (p = 0.011). Tumors with increased peripheral palisading of basal cell islands to immunostaining β-catenin responded poorly to PDT (p = 0.01). In line with our findings in patients, in vitro studies revealed a better response to PDT in the p53-positive ASZ cell line than the p53-negative BSZ cell line (p<0.01). Multivariate analysis revealed that the following variables were significantly associated with response to PDT: age, nBCC, presence of peritumoral inflammatory infiltrate, and p53 immunopositivity. Patients with positive p53 immunostaining were 68.54 times more likely to achieve cure than p53-negative patients (CI95% 2.94–159.8) Conclusion Our finding suggest that certain clinicopathological and immunohistochemical variables, particularly p53 expression, may serve as indicators of BCC response to MAL-PDT, and thus facilitate the selection of patients who are most likely to benefit from this therapy.
Collapse
|
18
|
Lucena SR, Zamarrón A, Carrasco E, Marigil MA, Mascaraque M, Fernández-Guarino M, Gilaberte Y, González S, Juarranz A. Characterisation of resistance mechanisms developed by basal cell carcinoma cells in response to repeated cycles of Photodynamic Therapy. Sci Rep 2019; 9:4835. [PMID: 30886381 PMCID: PMC6423284 DOI: 10.1038/s41598-019-41313-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 03/04/2019] [Indexed: 02/06/2023] Open
Abstract
Photodynamic Therapy (PDT) with methyl-aminolevulinate acid (MAL-PDT) is being used for the treatment of Basal cell carcinoma (BCC), but recurrences have been reported. In this work, we have evaluated resistance mechanisms to MAL-PDT developed by three BCC cell lines (ASZ, BSZ and CSZ), derived from mice on a ptch+/- background and with or without p53 expression, subjected to 10 cycles of PDT (10thG). The resistant populations showed mesenchymal-like structure and diminished proliferative capacity and size compared to the parental (P) cells. The resistance was dependent on the production of the endogenous photosensitiser protoporphyrin IX in the CSZ cell line and on its cellular localisation in ASZ and BSZ cells. Moreover, resistant cells expressing the p53 gene presented lower proliferation rate and increased expression levels of N-cadherin and Gsk3β (a component of the Wnt/β-catenin pathway) than P cells. In contrast, 10thG cells lacking the p53 gene showed lower levels of expression of Gsk3β in the cytoplasm and of E-cadherin and β-catenin in the membrane. In addition, resistant cells presented higher tumorigenic ability in immunosuppressed mice. Altogether, these results shed light on resistance mechanisms of BCC to PDT and may help to improve the use of this therapeutic approach.
Collapse
Affiliation(s)
- Silvia Rocio Lucena
- Biology Department, Faculty of Sciences, Autonomous University of Madrid, Madrid, Spain
| | - Alicia Zamarrón
- Biology Department, Faculty of Sciences, Autonomous University of Madrid, Madrid, Spain
| | - Elisa Carrasco
- Molecular Biology Department, Faculty of Sciences, Autonomous University of Madrid, Madrid, Spain
| | | | - Marta Mascaraque
- Biology Department, Faculty of Sciences, Autonomous University of Madrid, Madrid, Spain
| | | | | | - Salvador González
- Medicine and Medical Specialties Department, Alcalá de Henares University, Madrid, Spain
| | - Angeles Juarranz
- Biology Department, Faculty of Sciences, Autonomous University of Madrid, Madrid, Spain.
- Instituto Ramón y Cajal de Investigaciones Sanitarias, IRYCIS, Madrid, Spain.
| |
Collapse
|
19
|
Teske KA, Dash RC, Morel SR, Chau LQ, Wechsler-Reya RJ, Hadden MK. Development of posaconazole-based analogues as hedgehog signaling pathway inhibitors. Eur J Med Chem 2019; 163:320-332. [PMID: 30529635 PMCID: PMC6358021 DOI: 10.1016/j.ejmech.2018.11.056] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 11/13/2018] [Accepted: 11/22/2018] [Indexed: 01/20/2023]
Abstract
Inhibition of the hedgehog (Hh) signaling pathway has been validated as a therapeutic strategy to treat basal cell carcinoma and holds potential for several other forms of human cancer. Itraconazole and posaconazole are clinically useful triazole anti-fungals that are being repurposed as anti-cancer agents based on their ability to inhibit the Hh pathway. We have previously demonstrated that removal of the triazole from itraconazole does not affect its ability to inhibit the Hh pathway while abolishing its primary side effect, potent inhibition of Cyp3A4. To develop structure-activity relationships for the related posaconazole scaffold, we synthesized and evaluated a series of des-triazole analogues designed through both ligand- and structure-based methods. These compounds demonstrated improved anti-Hh properties compared to posaconazole and enhanced stability without inhibiting Cyp3A4. In addition, we utilized a series of molecular dynamics and binding energy studies to probe specific interactions between the compounds and their proposed binding site on Smoothened. These studies strongly suggest that the tetrahydrofuran region of the scaffold projects out of the binding site and that π-π interactions between the compound and Smoothened play a key role in stabilizing the bound analogues.
Collapse
Affiliation(s)
- Kelly A Teske
- Department of Pharmaceutical Sciences, University of Connecticut, 69 N Eagleville Rd, Unit 3092, Storrs, CT, 06269, USA
| | - Radha Charan Dash
- Department of Pharmaceutical Sciences, University of Connecticut, 69 N Eagleville Rd, Unit 3092, Storrs, CT, 06269, USA
| | - Shana R Morel
- Department of Pharmaceutical Sciences, University of Connecticut, 69 N Eagleville Rd, Unit 3092, Storrs, CT, 06269, USA
| | - Lianne Q Chau
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, United States
| | - Robert J Wechsler-Reya
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, United States
| | - M Kyle Hadden
- Department of Pharmaceutical Sciences, University of Connecticut, 69 N Eagleville Rd, Unit 3092, Storrs, CT, 06269, USA.
| |
Collapse
|
20
|
Kuonen F, Huskey NE, Shankar G, Jaju P, Whitson RJ, Rieger KE, Atwood SX, Sarin KY, Oro AE. Loss of Primary Cilia Drives Switching from Hedgehog to Ras/MAPK Pathway in Resistant Basal Cell Carcinoma. J Invest Dermatol 2019; 139:1439-1448. [PMID: 30707899 DOI: 10.1016/j.jid.2018.11.035] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/08/2018] [Accepted: 11/09/2018] [Indexed: 12/20/2022]
Abstract
Basal cell carcinomas (BCCs) rely on Hedgehog (HH) pathway growth signal amplification by the microtubule-based organelle, the primary cilium. Despite naive tumor responsiveness to Smoothened inhibitors (Smoi), resistance in advanced tumors remains common. Although the resistant BCCs usually maintain HH pathway activation, squamous cell carcinomas with Ras/MAPK pathway activation also arise, and the molecular basis of tumor type and pathway selection are still obscure. Here, we identify the primary cilium as a critical determinant controlling tumor pathway switching. Strikingly, Smoothened inhibitor-resistant BCCs have an increased mutational load in ciliome genes, resulting in reduced primary cilia and HH pathway activation compared with naive or Gorlin syndrome patient BCCs. Gene set enrichment analysis of resistant BCCs with a low HH pathway signature showed increased Ras/MAPK pathway activation. Tissue analysis confirmed an inverse relationship between primary cilia presence and Ras/MAPK activation, and primary cilia removal in BCCs potentiated Ras/MAPK pathway activation. Moreover, activating Ras in HH-responsive cell lines conferred resistance to both canonical (vismodegib) and noncanonical (atypical protein kinase C and MRTF inhibitors) HH pathway inhibitors and conferred sensitivity to MAPK inhibitors. Our results provide insights into BCC treatment and identify the primary cilium as an important lineage gatekeeper, preventing HH-to-Ras/MAPK pathway switching.
Collapse
Affiliation(s)
- François Kuonen
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA; Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Noelle E Huskey
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA; Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Gautam Shankar
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA
| | - Prajakta Jaju
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA
| | - Ramon J Whitson
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA
| | - Kerri E Rieger
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA
| | - Scott X Atwood
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA
| | - Kavita Y Sarin
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA
| | - Anthony E Oro
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA.
| |
Collapse
|
21
|
Mirza AN, McKellar SA, Urman NM, Brown AS, Hollmig T, Aasi SZ, Oro AE. LAP2 Proteins Chaperone GLI1 Movement between the Lamina and Chromatin to Regulate Transcription. Cell 2019; 176:198-212.e15. [PMID: 30503211 PMCID: PMC6379078 DOI: 10.1016/j.cell.2018.10.054] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/13/2018] [Accepted: 10/26/2018] [Indexed: 12/20/2022]
Abstract
Understanding transcription factor navigation through the nucleus remains critical for developing targeted therapeutics. The GLI1 transcription factor must maintain maximal Hedgehog pathway output in basal cell carcinomas (BCCs), and we have previously shown that resistant BCCs increase GLI1 deacetylation through atypical protein kinase Cι/λ (aPKC) and HDAC1. Here we identify a lamina-associated polypeptide 2 (LAP2) isoform-dependent nuclear chaperoning system that regulates GLI1 movement between the nuclear lamina and nucleoplasm to achieve maximal activation. LAP2β forms a two-site interaction with the GLI1 zinc-finger domain and acetylation site, stabilizing an acetylation-dependent reserve on the inner nuclear membrane (INM). By contrast, the nucleoplasmic LAP2α competes with LAP2β for GLI1 while scaffolding HDAC1 to deacetylate the secondary binding site. aPKC functions to promote GLI1 association with LAP2α, promoting egress off the INM. GLI1 intranuclear trafficking by LAP2 isoforms represents a powerful signal amplifier in BCCs with implications for zinc finger-based signal transduction and therapeutics.
Collapse
Affiliation(s)
- Amar N Mirza
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Siegen A McKellar
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nicole M Urman
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alexander S Brown
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tyler Hollmig
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sumaira Z Aasi
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Anthony E Oro
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
22
|
Overexpression of Desmoglein 2 in a Mouse Model of Gorlin Syndrome Enhances Spontaneous Basal Cell Carcinoma Formation through STAT3-Mediated Gli1 Expression. J Invest Dermatol 2018; 139:300-307. [PMID: 30291846 DOI: 10.1016/j.jid.2018.09.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 09/03/2018] [Accepted: 09/04/2018] [Indexed: 02/06/2023]
Abstract
Activation of the hedgehog pathway is causative of virtually all sporadic and Gorlin syndrome-related basal cell carcinomas (BCCs), with loss of function of Ptc1 being the most common genomic lesion. Sporadic BCCs also overexpress Dsg2, a desmosomal cadherin normally found in the basal layer. Using a mouse model of Gorlin syndrome (Ptc1+/lacZ mice), we found that overexpressing Dsg2 in the basal layer (K14-Dsg2/Ptc1+/lacZ mice) or the superficial epidermis (Inv-Dsg2/Ptc1+/lacZ mice) resulted in increased spontaneous BCC formation at 3 and 6 months, respectively. The tumors did not show loss of heterozygosity of Ptc1, despite high levels of Gli1 and phosphorylated Stat3. A panel of sporadic human BCCs showed increased staining of both Dsg2 and phosphorylated Stat3 in all nine samples. Overexpression of Dsg2 in ASZ001 cells, a Ptc1-/- BCC cell line, induced Stat3 phosphorylation and further increased Gli1 levels, in both an autocrine and paracrine manner. Three different Stat3 inhibitors reduced viability and Gli1 expression in ASZ001 cells but not in HaCaT cells. Conversely, stimulation of Stat3 in ASZ001 cells with IL-6 increased Gli1 expression. Our results indicate that Dsg2 enhances canonical hedgehog signaling downstream of Ptc1 to promote BCC development through the activation of phosphorylated Stat3 and regulation of Gli1 expression.
Collapse
|
23
|
Burns MA, Liao ZW, Yamagata N, Pouliot GP, Stevenson KE, Neuberg DS, Thorner AR, Ducar M, Silverman EA, Hunger SP, Loh ML, Winter SS, Dunsmore KP, Wood B, Devidas M, Harris MH, Silverman LB, Sallan SE, Gutierrez A. Hedgehog pathway mutations drive oncogenic transformation in high-risk T-cell acute lymphoblastic leukemia. Leukemia 2018; 32:2126-2137. [PMID: 29654263 PMCID: PMC6148437 DOI: 10.1038/s41375-018-0097-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 01/16/2018] [Accepted: 02/19/2018] [Indexed: 02/07/2023]
Abstract
The role of Hedgehog signaling in normal and malignant T-cell development is controversial. Recently, Hedgehog pathway mutations have been described in T-ALL, but whether mutational activation of Hedgehog signaling drives T-cell transformation is unknown, hindering the rationale for therapeutic intervention. Here, we show that Hedgehog pathway mutations predict chemotherapy resistance in human T-ALL, and drive oncogenic transformation in a zebrafish model of the disease. We found Hedgehog pathway mutations in 16% of 109 childhood T-ALL cases, most commonly affecting its negative regulator PTCH1. Hedgehog mutations were associated with resistance to induction chemotherapy (P = 0.009). Transduction of wild-type PTCH1 into PTCH1-mutant T-ALL cells induced apoptosis (P = 0.005), a phenotype that was reversed by downstream Hedgehog pathway activation (P = 0.007). Transduction of most mutant PTCH1, SUFU, and GLI alleles into mammalian cells induced aberrant regulation of Hedgehog signaling, indicating that these mutations are pathogenic. Using a CRISPR/Cas9 system for lineage-restricted gene disruption in transgenic zebrafish, we found that ptch1 mutations accelerated the onset of notch1-induced T-ALL (P = 0.0001), and pharmacologic Hedgehog pathway inhibition had therapeutic activity. Thus, Hedgehog-activating mutations are driver oncogenic alterations in high-risk T-ALL, providing a molecular rationale for targeted therapy in this disease.
Collapse
Affiliation(s)
- Melissa A Burns
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Zi Wei Liao
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Natsuko Yamagata
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Gayle P Pouliot
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Kristen E Stevenson
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Donna S Neuberg
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Aaron R Thorner
- Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Matthew Ducar
- Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Emily A Silverman
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Stephen P Hunger
- Division of Oncology and the Center for Childhood Cancer Research, The Children's Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Mignon L Loh
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Stuart S Winter
- Department of Pediatrics, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Kimberly P Dunsmore
- Division of Oncology, University of Virginia Children's Hospital, Charlottesville, VA, 22903, USA
| | - Brent Wood
- Department of Laboratory Medicine, University of Washington, Seattle, 98195, WA, USA
| | - Meenakshi Devidas
- Department of Biostatistics, University of Florida, Gainesville, FL, 32611, USA
| | - Marian H Harris
- Department of Pathology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Lewis B Silverman
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Stephen E Sallan
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Alejandro Gutierrez
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, 02115, USA.
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
| |
Collapse
|
24
|
Maglic D, Schlegelmilch K, Dost AF, Panero R, Dill MT, Calogero RA, Camargo FD. YAP-TEAD signaling promotes basal cell carcinoma development via a c-JUN/AP1 axis. EMBO J 2018; 37:embj.201798642. [PMID: 30037824 DOI: 10.15252/embj.201798642] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 06/13/2018] [Accepted: 06/14/2018] [Indexed: 12/29/2022] Open
Abstract
The mammalian Hippo signaling pathway, through its effectors YAP and TAZ, coerces epithelial progenitor cell expansion for appropriate tissue development or regeneration upon damage. Its ability to drive rapid tissue growth explains why many oncogenic events frequently exploit this pathway to promote cancer phenotypes. Indeed, several tumor types including basal cell carcinoma (BCC) show genetic aberrations in the Hippo (or YAP/TAZ) regulators. Here, we uncover that while YAP is dispensable for homeostatic epidermal regeneration, it is required for BCC development. Our clonal analyses further demonstrate that the few emerging Yap-null dysplasia have lower fitness and thus are diminished as they progress to invasive BCC Mechanistically, YAP depletion in BCC tumors leads to effective impairment of the JNK-JUN signaling, a well-established tumor-driving cascade. Importantly, in this context, YAP does not influence canonical Wnt or Hedgehog signaling. Overall, we reveal Hippo signaling as an independent promoter of BCC pathogenesis and thereby a viable target for drug-resistant BCC.
Collapse
Affiliation(s)
- Dejan Maglic
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | | | | | - Riccardo Panero
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy
| | - Michael T Dill
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Raffaele A Calogero
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy
| | - Fernando D Camargo
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA .,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| |
Collapse
|
25
|
Kim AL, Back JH, Chaudhary SC, Zhu Y, Athar M, Bickers DR. SOX9 Transcriptionally Regulates mTOR-Induced Proliferation of Basal Cell Carcinomas. J Invest Dermatol 2018; 138:1716-1725. [PMID: 29550418 DOI: 10.1016/j.jid.2018.01.040] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 01/17/2018] [Accepted: 01/28/2018] [Indexed: 01/20/2023]
Abstract
Currently available smoothened targeted therapies in patients with basal cell nevus syndrome are associated with substantial tumor recurrence and clinical resistance. Strategies bypassing smoothened and/or identifying additional downstream components of the Hedgehog pathway could provide novel antitumor targets with a better therapeutic index. Sry-related high mobility group box 9 (SOX9) is a Hedgehog/glioma-associated oncogene homolog-regulated transcription factor known to be overexpressed in basal cell carcinomas (BCCs). A sequence motif search for SOX9-responsive elements identified three motifs in the promoter region of mammalian target of rapamycin (mTOR). In murine BCC cells, SOX9 occupies the mTOR promoter and induces its transcriptional activity. Short hairpin RNA (shRNA)-mediated knockdown of SOX9, as well as smoothened inhibition by itraconazole and vismodegib, reduces mTOR expression and the phosphorylation of known downstream mTOR targets. These effects culminate in diminishing the proliferative capacity of BCC cells, demonstrating a direct mechanistic link between the Hedgehog and mTOR pathways capable of driving BCC growth. Furthermore, rapamycin, a pharmacologic mTOR inhibitor, suppressed the growth of UV-induced BCCs in Ptch1+/-/SKH-1 mice, a model that closely mimics the accelerated BCC growth pattern of patients with basal cell nevus syndrome. Our data demonstrate that Hedgehog signaling converges on mTOR via SOX9, and highlight the SOX9-mTOR axis as a viable additional target downstream of smoothened that could enhance tumor elimination in patients with BCC.
Collapse
Affiliation(s)
- Arianna L Kim
- Department of Dermatology, Columbia University Medical Center, New York, New York, USA.
| | - Jung Ho Back
- Department of Dermatology, Columbia University Medical Center, New York, New York, USA
| | - Sandeep C Chaudhary
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yucui Zhu
- Department of Dermatology, Columbia University Medical Center, New York, New York, USA
| | - Mohammad Athar
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - David R Bickers
- Department of Dermatology, Columbia University Medical Center, New York, New York, USA
| |
Collapse
|
26
|
Mirza AN, Fry MA, Urman NM, Atwood SX, Roffey J, Ott GR, Chen B, Lee A, Brown AS, Aasi SZ, Hollmig T, Ator MA, Dorsey BD, Ruggeri BR, Zificsak CA, Sirota M, Tang JY, Butte A, Epstein E, Sarin KY, Oro AE. Combined inhibition of atypical PKC and histone deacetylase 1 is cooperative in basal cell carcinoma treatment. JCI Insight 2017; 2:97071. [PMID: 29093271 DOI: 10.1172/jci.insight.97071] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 09/29/2017] [Indexed: 01/09/2023] Open
Abstract
Advanced basal cell carcinomas (BCCs) circumvent Smoothened (SMO) inhibition by activating GLI transcription factors to sustain the high levels of Hedgehog (HH) signaling required for their survival. Unfortunately, there is a lack of efficacious therapies. We performed a gene expression-based drug repositioning screen in silico and identified the FDA-approved histone deacetylase (HDAC) inhibitor, vorinostat, as a top therapeutic candidate. We show that vorinostat only inhibits proliferation of BCC cells in vitro and BCC allografts in vivo at high dose, limiting its usefulness as a monotherapy. We leveraged this in silico approach to identify drug combinations that increase the therapeutic window of vorinostat and identified atypical PKC Ɩ/ʎ (aPKC) as a HDAC costimulator of HH signaling. We found that aPKC promotes GLI1-HDAC1 association in vitro, linking two positive feedback loops. Combination targeting of HDAC1 and aPKC robustly inhibited GLI1, lowering drug doses needed in vitro, in vivo, and ex vivo in patient-derived BCC explants. We identified a bioavailable and selective small-molecule aPKC inhibitor, bringing the pharmacological blockade of aPKC and HDAC1 into the realm of clinical possibility. Our findings provide a compelling rationale and candidate drugs for combined targeting of HDAC1 and aPKC in HH-dependent cancers.
Collapse
Affiliation(s)
- Amar N Mirza
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA
| | - Micah A Fry
- Children's Hospital Oakland Research Institute, Oakland, California, USA
| | - Nicole M Urman
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA
| | - Scott X Atwood
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA
| | - Jon Roffey
- CRUK Therapeutic Discovery Laboratories, London Bioscience Innovation Centre, London, United Kingdom
| | - Gregory R Ott
- Teva Branded Pharmaceutical Products R&D, West Chester, Pennsylvania, USA
| | - Bin Chen
- Institute for Computational Health Sciences, UCSF, San Francisco, California, USA
| | - Alex Lee
- Children's Hospital Oakland Research Institute, Oakland, California, USA
| | - Alexander S Brown
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA
| | - Sumaira Z Aasi
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA
| | - Tyler Hollmig
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA
| | - Mark A Ator
- Teva Branded Pharmaceutical Products R&D, West Chester, Pennsylvania, USA
| | - Bruce D Dorsey
- Teva Branded Pharmaceutical Products R&D, West Chester, Pennsylvania, USA
| | - Bruce R Ruggeri
- Teva Branded Pharmaceutical Products R&D, West Chester, Pennsylvania, USA
| | - Craig A Zificsak
- Teva Branded Pharmaceutical Products R&D, West Chester, Pennsylvania, USA
| | - Marina Sirota
- Institute for Computational Health Sciences, UCSF, San Francisco, California, USA
| | - Jean Y Tang
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA.,Children's Hospital Oakland Research Institute, Oakland, California, USA
| | - Atul Butte
- Institute for Computational Health Sciences, UCSF, San Francisco, California, USA
| | - Ervin Epstein
- Children's Hospital Oakland Research Institute, Oakland, California, USA
| | - Kavita Y Sarin
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA
| | - Anthony E Oro
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
27
|
Dabrzalska M, Janaszewska A, Zablocka M, Mignani S, Majoral JP, Klajnert-Maculewicz B. Cationic Phosphorus Dendrimer Enhances Photodynamic Activity of Rose Bengal against Basal Cell Carcinoma Cell Lines. Mol Pharm 2017; 14:1821-1830. [PMID: 28350966 DOI: 10.1021/acs.molpharmaceut.7b00108] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
In the last couple of decades, photodynamic therapy emerged as a useful tool in the treatment of basal cell carcinoma. However, it still meets limitations due to unfavorable properties of photosensitizers such as poor solubility or lack of selectivity. Dendrimers, polymers widely studied in biomedical field, may play a role as photosensitizer carriers and improve the efficacy of photodynamic treatment. Here, we describe the evaluation of an electrostatic complex of cationic phosphorus dendrimer and rose bengal in such aspects as singlet oxygen production, cellular uptake, and phototoxicity against three basal cell carcinoma cell lines. Rose bengal-cationic dendrimer complex in molar ratio 5:1 was compared to free rose bengal. Obtained results showed that the singlet oxygen production in aqueous medium was significantly higher for the complex than for free rose bengal. The cellular uptake of the complex was 2-7-fold higher compared to a free photosensitizer. Importantly, rose bengal, rose bengal-dendrimer complex, and dendrimer itself showed no dark toxicity against all three cell lines. Moreover, we observed that phototoxicity of the complex was remarkably enhanced presumably due to high cellular uptake. On the basis of the obtained results, we conclude that rose bengal-cationic dendrimer complex has a potential in photodynamic treatment of basal cell carcinoma.
Collapse
Affiliation(s)
- Monika Dabrzalska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz , Pomorska 141/143, 90-236 Lodz, Poland
| | - Anna Janaszewska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz , Pomorska 141/143, 90-236 Lodz, Poland
| | - Maria Zablocka
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences , Sienkiewicza 112, 90-363 Lodz, Poland
| | - Serge Mignani
- Laboratoire de Chimie et de Biochimie pharmacologiques et toxicologique, Université Paris Descartes, PRES Sorbonne Paris Cité, CNRS UMR 860 , 45 rue des Saints Pères, 75006 Paris, France
| | - Jean Pierre Majoral
- Laboratoire de Chimie de Coordination CNRS, 205 route de Narbonne, 31077 Toulouse, France.,Université de Toulouse, UPS, INPT , 31077 Toulouse Cedex 4, France
| | - Barbara Klajnert-Maculewicz
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz , Pomorska 141/143, 90-236 Lodz, Poland.,Leibniz-Institut für Polymerforschung Dresden e.V. , Hohe Strasse 6, 01069 Dresden, Germany
| |
Collapse
|
28
|
Dabrzalska M, Janaszewska A, Zablocka M, Mignani S, Majoral JP, Klajnert-Maculewicz B. Complexing Methylene Blue with Phosphorus Dendrimers to Increase Photodynamic Activity. Molecules 2017; 22:E345. [PMID: 28241491 PMCID: PMC6155338 DOI: 10.3390/molecules22030345] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 02/01/2017] [Accepted: 02/20/2017] [Indexed: 11/20/2022] Open
Abstract
The efficiency of photodynamic therapy is limited mainly due to low selectivity, unfavorable biodistribution of photosensitizers, and long-lasting skin sensitivity to light. However, drug delivery systems based on nanoparticles may overcome the limitations mentioned above. Among others, dendrimers are particularly attractive as carriers, because of their globular architecture and high loading capacity. The goal of the study was to check whether an anionic phosphorus dendrimer is suitable as a carrier of a photosensitizer-methylene blue (MB). As a biological model, basal cell carcinoma cell lines were used. We checked the influence of the MB complexation on its singlet oxygen production ability using a commercial fluorescence probe. Next, cellular uptake, phototoxicity, reactive oxygen species (ROS) generation, and cell death were investigated. The MB-anionic dendrimer complex (MB-1an) was found to generate less singlet oxygen; however, the complex showed higher cellular uptake and phototoxicity against basal cell carcinoma cell lines, which was accompanied with enhanced ROS production. Owing to the obtained results, we conclude that the photodynamic activity of MB complexed with an anionic dendrimer is higher than free MB against basal cell carcinoma cell lines.
Collapse
Affiliation(s)
- Monika Dabrzalska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland.
| | - Anna Janaszewska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland.
| | - Maria Zablocka
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, 90-363 Lodz, Poland.
| | - Serge Mignani
- Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologique, Université Paris Descartes, PRES Sorbonne Paris Cité, CNRS UMR 860, 45 Rue Des Saints Pères, 75006 Paris, France.
| | - Jean Pierre Majoral
- Laboratoire de Chimie de Coordination CNRS, 205 Route de Narbonne, 31077 Toulouse CEDEX 4, France.
- Institut National Polytechnique de Toulouse, Université de Toulouse, UPS, 31077 Toulouse CEDEX 4, France.
| | - Barbara Klajnert-Maculewicz
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland.
- Institut für Polymerforschung Dresden e.V., Hohe Strasse 6, 01069 Dresden, Germany.
| |
Collapse
|
29
|
Urman NM, Mirza A, Atwood SX, Whitson RJ, Sarin KY, Tang JY, Oro AE. Tumor-Derived Suppressor of Fused Mutations Reveal Hedgehog Pathway Interactions. PLoS One 2016; 11:e0168031. [PMID: 28030567 PMCID: PMC5193403 DOI: 10.1371/journal.pone.0168031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 11/23/2016] [Indexed: 11/18/2022] Open
Abstract
The Hedgehog pathway is a potent regulator of cellular growth and plays a central role in the development of many cancers including basal cell carcinoma (BCC). The majority of BCCs arise from mutations in the Patched receptor resulting in constitutive activation of the Hedgehog pathway. Secondary driver mutations promote BCC oncogenesis and occur frequently due to the high mutational burden resulting from sun exposure of the skin. Here, we uncover novel secondary mutations in Suppressor of Fused (SUFU), the major negative regulator of the Hedgehog pathway. SUFU normally binds to a Hedgehog transcriptional activator, GLI1, in order to prevent it from initiating transcription of Hedgehog target genes. We sequenced tumor-normal pairs from patients with early sporadic BCCs. This resulted in the discovery of nine mutations in SUFU, which were functionally investigated to determine whether they help drive BCC formation. Our results show that four of the SUFU mutations inappropriately activate the Hedgehog pathway, suggesting they may act as driver mutations for BCC development. Indeed, all four of the loss of function SUFU variants were found to disrupt its binding to GLI, leading to constitutive pathway activation. Our results from functional characterization of these mutations shed light on SUFU’s role in Hedgehog signaling, tumor progression, and highlight a way in which BCCs can arise.
Collapse
Affiliation(s)
- Nicole M. Urman
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Amar Mirza
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Scott X. Atwood
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, CA, United States of America
- Department of Developmental and Cell Biology, Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA, United States of America
| | - Ramon J. Whitson
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Kavita Y. Sarin
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Jean Y. Tang
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Anthony E. Oro
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, CA, United States of America
- * E-mail:
| |
Collapse
|
30
|
Kim AL, Back JH, Zhu Y, Tang X, Yardley NP, Kim KJ, Athar M, Bickers DR. AKT1 Activation is Obligatory for Spontaneous BCC Tumor Growth in a Murine Model that Mimics Some Features of Basal Cell Nevus Syndrome. Cancer Prev Res (Phila) 2016; 9:794-802. [PMID: 27388747 DOI: 10.1158/1940-6207.capr-16-0066] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 06/28/2016] [Indexed: 01/14/2023]
Abstract
Patients with basal cell nevus syndrome (BCNS), also known as Gorlin syndrome, develop numerous basal cell carcinomas (BCC) due to germline mutations in the tumor suppressor PTCH1 and aberrant activation of Hedgehog (Hh) signaling. Therapies targeted at components of the Hh pathway, including the smoothened (SMO) inhibitor vismodegib, can ablate these tumors clinically, but tumors recur upon drug discontinuation. Using SKH1-Ptch1+/- as a model that closely mimics the spontaneous and accelerated growth pattern of BCCs in patients with BCNS, we show that AKT1, a serine/threonine protein kinase, is intrinsically activated in keratinocytes derived from the skin of newborn Ptch1+/- mice in the absence of carcinogenic stimuli. Introducing Akt1 haplodeficiency in Ptch1+/- mice (Akt1+/- Ptch1+/-) significantly abrogated BCC growth. Similarly, pharmacological inhibition of AKT with perifosine, an alkyl phospholipid AKT inhibitor, diminished the growth of spontaneous and UV-induced BCCs. Our data demonstrate an obligatory role for AKT1 in BCC growth, and targeting AKT may help reduce BCC tumor burden in BCNS patients. Cancer Prev Res; 9(10); 794-802. ©2016 AACR.
Collapse
Affiliation(s)
- Arianna L Kim
- Department of Dermatology, Columbia University Medical Center, New York, New York.
| | - Jung Ho Back
- Department of Dermatology, Columbia University Medical Center, New York, New York
| | - Yucui Zhu
- Department of Dermatology, Columbia University Medical Center, New York, New York
| | - Xiuwei Tang
- Department of Dermatology, Columbia University Medical Center, New York, New York
| | - Nathan P Yardley
- Department of Dermatology, Columbia University Medical Center, New York, New York
| | - Katherine J Kim
- Department of Dermatology, Columbia University Medical Center, New York, New York
| | - Mohammad Athar
- University of Alabama at Birmingham, Birmingham, Alabama.
| | - David R Bickers
- Department of Dermatology, Columbia University Medical Center, New York, New York.
| |
Collapse
|
31
|
Pace JR, DeBerardinis AM, Sail V, Tacheva-Grigorova SK, Chan KA, Tran R, Raccuia DS, Wechsler-Reya RJ, Hadden MK. Repurposing the Clinically Efficacious Antifungal Agent Itraconazole as an Anticancer Chemotherapeutic. J Med Chem 2016; 59:3635-49. [PMID: 27014922 DOI: 10.1021/acs.jmedchem.5b01718] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Itraconazole (ITZ) is an FDA-approved member of the triazole class of antifungal agents. Two recent drug repurposing screens identified ITZ as a promising anticancer chemotherapeutic that inhibits both the angiogenesis and hedgehog (Hh) signaling pathways. We have synthesized and evaluated first- and second-generation ITZ analogues for their anti-Hh and antiangiogenic activities to probe more fully the structural requirements for these anticancer properties. Our overall results suggest that the triazole functionality is required for ITZ-mediated inhibition of angiogenesis but that it is not essential for inhibition of Hh signaling. The synthesis and evaluation of stereochemically defined des-triazole ITZ analogues also provides key information as to the optimal configuration around the dioxolane ring of the ITZ scaffold. Finally, the results from our studies suggest that two distinct cellular mechanisms of action govern the anticancer properties of the ITZ scaffold.
Collapse
Affiliation(s)
- Jennifer R Pace
- Department of Pharmaceutical Sciences, University of Connecticut , 69 North Eagleville Road, Unit 3092, Storrs, Connecticut 06269-3092, United States
| | - Albert M DeBerardinis
- Department of Pharmaceutical Sciences, University of Connecticut , 69 North Eagleville Road, Unit 3092, Storrs, Connecticut 06269-3092, United States
| | - Vibhavari Sail
- Department of Pharmaceutical Sciences, University of Connecticut , 69 North Eagleville Road, Unit 3092, Storrs, Connecticut 06269-3092, United States
| | - Silvia K Tacheva-Grigorova
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute , 2880 Torrey Pines Scenic Drive, La Jolla, California 92037, United States
| | - Kelly A Chan
- Department of Pharmaceutical Sciences, University of Connecticut , 69 North Eagleville Road, Unit 3092, Storrs, Connecticut 06269-3092, United States
| | - Raymond Tran
- Department of Pharmaceutical Sciences, University of Connecticut , 69 North Eagleville Road, Unit 3092, Storrs, Connecticut 06269-3092, United States
| | - Daniel S Raccuia
- Department of Pharmaceutical Sciences, University of Connecticut , 69 North Eagleville Road, Unit 3092, Storrs, Connecticut 06269-3092, United States
| | - Robert J Wechsler-Reya
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute , 2880 Torrey Pines Scenic Drive, La Jolla, California 92037, United States
| | - M Kyle Hadden
- Department of Pharmaceutical Sciences, University of Connecticut , 69 North Eagleville Road, Unit 3092, Storrs, Connecticut 06269-3092, United States
| |
Collapse
|
32
|
Brennan-Crispi DM, Hossain C, Sahu J, Brady M, Riobo NA, Mahoney MG. Crosstalk between Desmoglein 2 and Patched 1 accelerates chemical-induced skin tumorigenesis. Oncotarget 2016; 6:8593-605. [PMID: 25871385 PMCID: PMC4496169 DOI: 10.18632/oncotarget.3309] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 02/08/2015] [Indexed: 01/12/2023] Open
Abstract
Aberrant activation of Hedgehog (Hh) signaling is causative of BCCs and has been associated with a fraction of SCCs. Desmoglein 2 (Dsg2) is an adhesion protein that is upregulated in many cancers and overexpression of Dsg2 in the epidermis renders mice more susceptible to squamous-derived neoplasia. Here we examined a potential crosstalk between Dsg2 and Hh signaling in skin tumorigenesis. Our findings show that Dsg2 modulates Gli1 expression, in vitro and in vivo. Ectopic expression of Dsg2 on Ptc1(+/lacZ) background enhanced epidermal proliferation and interfollicular activation of the Hh pathway. Furthermore, in response to DMBA/TPA, the Dsg2/Ptc1+/lacZ mice developed squamous lessons earlier than the WT, Ptc1(+/lacZ), and Inv-Dsg2 littermates. Additionally, DMBA/TPA induced BCC formation in all mice harboring the Ptc1(+/lacZ) gene and the presence of Dsg2 in Dsg2/Ptc1(+/lacZ) mice doubled the BCC tumor burden. Reporter analysis revealed activation of the Hh pathway in the BCC tumors. However, in the SCCs we observed Hh activity only in the underlying dermis of the tumors. Furthermore, Dsg2/Ptc1(+/lacZ) mice demonstrated enhanced MEK/Erk1/2 activation within the tumors and expression of Shh in the dermis. In summary, our results demonstrate that Dsg2 modulates Hh signaling, and this synergy may accelerate skin tumor development by different mechanisms.
Collapse
Affiliation(s)
- Donna M Brennan-Crispi
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA, USA.,Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA.,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Claudia Hossain
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Joya Sahu
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Mary Brady
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Natalia A Riobo
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA.,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Mỹ G Mahoney
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA, USA.,Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA.,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
33
|
Gruber W, Hutzinger M, Elmer DP, Parigger T, Sternberg C, Cegielkowski L, Zaja M, Leban J, Michel S, Hamm S, Vitt D, Aberger F. DYRK1B as therapeutic target in Hedgehog/GLI-dependent cancer cells with Smoothened inhibitor resistance. Oncotarget 2016; 7:7134-48. [PMID: 26784250 PMCID: PMC4872774 DOI: 10.18632/oncotarget.6910] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 01/04/2016] [Indexed: 12/26/2022] Open
Abstract
A wide range of human malignancies displays aberrant activation of Hedgehog (HH)/GLI signaling, including cancers of the skin, brain, gastrointestinal tract and hematopoietic system. Targeting oncogenic HH/GLI signaling with small molecule inhibitors of the essential pathway effector Smoothened (SMO) has shown remarkable therapeutic effects in patients with advanced and metastatic basal cell carcinoma. However, acquired and de novo resistance to SMO inhibitors poses severe limitations to the use of SMO antagonists and urgently calls for the identification of novel targets and compounds.Here we report on the identification of the Dual-Specificity-Tyrosine-Phosphorylation-Regulated Kinase 1B (DYRK1B) as critical positive regulator of HH/GLI signaling downstream of SMO. Genetic and chemical inhibition of DYRK1B in human and mouse cancer cells resulted in marked repression of HH signaling and GLI1 expression, respectively. Importantly, DYRK1B inhibition profoundly impaired GLI1 expression in both SMO-inhibitor sensitive and resistant settings. We further introduce a novel small molecule DYRK1B inhibitor, DYRKi, with suitable pharmacologic properties to impair SMO-dependent and SMO-independent oncogenic GLI activity. The results support the use of DYRK1B antagonists for the treatment of HH/GLI-associated cancers where SMO inhibitors fail to demonstrate therapeutic efficacy.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Blotting, Western
- Carcinoma, Basal Cell/drug therapy
- Carcinoma, Basal Cell/genetics
- Carcinoma, Basal Cell/metabolism
- Carcinoma, Basal Cell/pathology
- Cell Proliferation/drug effects
- Cells, Cultured
- Drug Resistance, Neoplasm
- Forkhead Transcription Factors/physiology
- Hedgehog Proteins/antagonists & inhibitors
- Hedgehog Proteins/genetics
- Hedgehog Proteins/metabolism
- Humans
- Mice
- Mice, Nude
- NIH 3T3 Cells
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Protein-Tyrosine Kinases/antagonists & inhibitors
- Protein-Tyrosine Kinases/genetics
- Protein-Tyrosine Kinases/metabolism
- RNA, Messenger/genetics
- RNA, Small Interfering/genetics
- Real-Time Polymerase Chain Reaction
- Reverse Transcriptase Polymerase Chain Reaction
- Skin Neoplasms/drug therapy
- Skin Neoplasms/genetics
- Skin Neoplasms/metabolism
- Skin Neoplasms/pathology
- Smoothened Receptor/antagonists & inhibitors
- Smoothened Receptor/genetics
- Smoothened Receptor/metabolism
- Xenograft Model Antitumor Assays
- Zinc Finger Protein GLI1/antagonists & inhibitors
- Zinc Finger Protein GLI1/genetics
- Zinc Finger Protein GLI1/metabolism
- Dyrk Kinases
Collapse
Affiliation(s)
- Wolfgang Gruber
- Cancer Cluster Salzburg, Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - Martin Hutzinger
- Cancer Cluster Salzburg, Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - Dominik Patrick Elmer
- Cancer Cluster Salzburg, Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - Thomas Parigger
- Cancer Cluster Salzburg, Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - Christina Sternberg
- Cancer Cluster Salzburg, Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - Lukasz Cegielkowski
- Cancer Cluster Salzburg, Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - Mirko Zaja
- 4SC Discovery GmbH, Planegg-Martinsried, Germany
| | - Johann Leban
- Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | | | | | - Daniel Vitt
- 4SC Discovery GmbH, Planegg-Martinsried, Germany
- 4SC AG, Planegg-Martinsried, Germany
| | - Fritz Aberger
- Cancer Cluster Salzburg, Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| |
Collapse
|
34
|
Chen B, Trang V, Lee A, Williams NS, Wilson AN, Epstein EH, Tang JY, Kim J. Posaconazole, a Second-Generation Triazole Antifungal Drug, Inhibits the Hedgehog Signaling Pathway and Progression of Basal Cell Carcinoma. Mol Cancer Ther 2016; 15:866-76. [PMID: 26823493 DOI: 10.1158/1535-7163.mct-15-0729-t] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 12/30/2015] [Indexed: 12/12/2022]
Abstract
Deregulation of Hedgehog (Hh) pathway signaling has been associated with the pathogenesis of various malignancies, including basal cell carcinomas (BCC). Inhibitors of the Hh pathway currently available or under clinical investigation all bind and antagonize Smoothened (SMO), inducing a marked but transient clinical response. Tumor regrowth and therapy failure were attributed to mutations in the binding site of these small-molecule SMO antagonists. The antifungal itraconazole was demonstrated to be a potent SMO antagonist with a distinct mechanism of action from that of current SMO inhibitors. However, itraconazole represents a suboptimal therapeutic option due to its numerous drug-drug interactions. Here, we show that posaconazole, a second-generation triazole antifungal with minimal drug-drug interactions and a favorable side-effect profile, is also a potent inhibitor of the Hh pathway that functions at the level of SMO. We demonstrate that posaconazole inhibits the Hh pathway by a mechanism distinct from that of cyclopamine and other cyclopamine-competitive SMO antagonists but, similar to itraconazole, has robust activity against drug-resistant SMO mutants and inhibits the growth of Hh-dependent BCC in vivo Our results suggest that posaconazole, alone or in combination with other Hh pathway antagonists, may be readily tested in clinical studies for the treatment of Hh-dependent cancers. Mol Cancer Ther; 15(5); 866-76. ©2016 AACR.
Collapse
Affiliation(s)
- Baozhi Chen
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern, Dallas, Texas
| | - Vinh Trang
- Department of Internal Medicine, University of Texas Southwestern, Dallas, Texas
| | - Alex Lee
- Children's Hospital Oakland Research Institute, Oakland, California
| | - Noelle S Williams
- Department of Biochemistry, University of Texas Southwestern, Dallas, Texas
| | - Alexandra N Wilson
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern, Dallas, Texas
| | - Ervin H Epstein
- Children's Hospital Oakland Research Institute, Oakland, California
| | - Jean Y Tang
- Children's Hospital Oakland Research Institute, Oakland, California. Department of Dermatology, Stanford University, Stanford, California
| | - James Kim
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern, Dallas, Texas. Department of Internal Medicine, University of Texas Southwestern, Dallas, Texas.
| |
Collapse
|
35
|
Ra H, González-González E, Uddin MJ, King BL, Lee A, Ali-Khan I, Marnett LJ, Tang JY, Contag CH. Detection of non-melanoma skin cancer by in vivo fluorescence imaging with fluorocoxib A. Neoplasia 2015; 17:201-7. [PMID: 25748239 PMCID: PMC4351298 DOI: 10.1016/j.neo.2014.12.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Accepted: 12/23/2014] [Indexed: 11/16/2022] Open
Abstract
Non-melanoma skin cancer (NMSC) is the most common form of cancer in the US and its incidence is increasing. The current standard of care is visual inspection by physicians and/or dermatologists, followed by skin biopsy and pathologic confirmation. We have investigated the use of in vivo fluorescence imaging using fluorocoxib A as a molecular probe for early detection and assessment of skin tumors in mouse models of NMSC. Fluorocoxib A targets the cyclooxygenase-2 (COX-2) enzyme that is preferentially expressed by inflamed and tumor tissue, and therefore has potential to be an effective broadly active molecular biomarker for cancer detection. We tested the sensitivity of fluorocoxib A in a BCC allograft SCID hairless mouse model using a wide-field fluorescence imaging system. Subcutaneous allografts comprised of 1000 BCC cells were detectable above background. These BCC allograft mice were imaged over time and a linear correlation (R2 = 0.8) between tumor volume and fluorocoxib A signal levels was observed. We also tested fluorocoxib A in a genetically engineered spontaneous BCC mouse model (Ptch1+/− K14-Cre-ER2 p53fl/fl), where sequential imaging of the same animals over time demonstrated that early, microscopic lesions (100 μm size) developed into visible macroscopic tumor masses over 11 to 17 days. Overall, for macroscopic tumors, the sensitivity was 88% and the specificity was 100%. For microscopic tumors, the sensitivity was 85% and specificity was 56%. These results demonstrate the potential of fluorocoxib A as an in vivo imaging agent for early detection, margin delineation and guided biopsies of NMSCs.
Collapse
Affiliation(s)
- Hyejun Ra
- Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA; Dept. of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Emilio González-González
- Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA; Dept. of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Md Jashim Uddin
- A.B. Hancock Jr. Memorial Laboratory for Cancer Research, Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN; A.B. Hancock Jr. Memorial Laboratory for Cancer Research, Department of Chemistry, Vanderbilt University School of Medicine, Nashville, TN; A.B. Hancock Jr. Memorial Laboratory for Cancer Research, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN
| | - Bonnie L King
- Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA; Dept. of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Alex Lee
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA
| | - Irfan Ali-Khan
- Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA; Dept. of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Lawrence J Marnett
- A.B. Hancock Jr. Memorial Laboratory for Cancer Research, Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN; A.B. Hancock Jr. Memorial Laboratory for Cancer Research, Department of Chemistry, Vanderbilt University School of Medicine, Nashville, TN; A.B. Hancock Jr. Memorial Laboratory for Cancer Research, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN
| | - Jean Y Tang
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA
| | - Christopher H Contag
- Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA; Dept. of Pediatrics, Stanford University School of Medicine, Stanford, CA; Dept. of Radiology, Stanford University School of Medicine, Stanford, CA; Dept. of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA.
| |
Collapse
|
36
|
Brennan-Crispi DM, Mahoney MG, Riobo NA. Methods for Detection of Ptc1-Driven LacZ Expression in Adult Mouse Skin. Methods Mol Biol 2015; 1322:167-185. [PMID: 26179048 DOI: 10.1007/978-1-4939-2772-2_15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The Ptc-lacZ reporter mice are a highly utilized animal model for studying both normal tissue development and cancer. Identifying cell specific activation of Hedgehog (Hh) signaling is essential to understand the effects of this critical and complex signaling pathway. β-gal detection in tissues can be difficult, with various staining procedures yielding differential results. Thus, detailed information on staining protocols is essential for determining the ideal method for a given study. Furthermore, immunohistochemical staining of X-Gal stained tissues can provide further insight into other key players in Hh signaling activation.
Collapse
Affiliation(s)
- Donna M Brennan-Crispi
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | | | | |
Collapse
|
37
|
Banerjee U, DeBerardinis AM, Hadden MK. Design, synthesis, and evaluation of hybrid vitamin D3 side chain analogues as hedgehog pathway inhibitors. Bioorg Med Chem 2014; 23:548-55. [PMID: 25549899 DOI: 10.1016/j.bmc.2014.12.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 11/24/2014] [Accepted: 12/03/2014] [Indexed: 10/24/2022]
Abstract
Vitamin D3 (VD3) is a moderately potent and non-selective inhibitor of the Hedgehog (Hh) signaling cascade. Previous studies have established that the CD-ring region of VD3 serves as the Hh inhibitory pharmacophore. Subsequently, compound 3, an ester linked aromatic A-ring and CD-ring derivative was identified as an improved and selective Hh inhibitor. Herein, we report modifications of the CD-ring side chain that afford enhancement of selectivity for Hh modulation thereby diminishing the detrimental effects of concomitant vitamin D receptor activation. In general, linear or moderately branched alkyl chains of five or six carbons were optimal for potent and selective inhibition of Hh signaling. Moreover, hybrid VD3 side chain derivative 20 demonstrated 4-fold improvement in Hh antagonistic activity over VD3(IC50=1.1-1.6 μM) while gaining greater than a 1000-fold selectivity for Hh signaling over canonical activation of the vitamin D receptor pathway.
Collapse
Affiliation(s)
- Upasana Banerjee
- Department of Pharmaceutical Sciences, University of Connecticut, 69 N Eagleville Rd, Storrs, CT 06269, USA
| | - Albert M DeBerardinis
- Department of Pharmaceutical Sciences, University of Connecticut, 69 N Eagleville Rd, Storrs, CT 06269, USA
| | - M Kyle Hadden
- Department of Pharmaceutical Sciences, University of Connecticut, 69 N Eagleville Rd, Storrs, CT 06269, USA.
| |
Collapse
|
38
|
König S, Nitzki F, Uhmann A, Dittmann K, Theiss-Suennemann J, Herrmann M, Reichardt HM, Schwendener R, Pukrop T, Schulz-Schaeffer W, Hahn H. Depletion of cutaneous macrophages and dendritic cells promotes growth of basal cell carcinoma in mice. PLoS One 2014; 9:e93555. [PMID: 24691432 PMCID: PMC3972151 DOI: 10.1371/journal.pone.0093555] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 03/06/2014] [Indexed: 01/17/2023] Open
Abstract
Basal cell carcinoma (BCC) belongs to the group of non-melanoma skin tumors and is the most common tumor in the western world. BCC arises due to mutations in the tumor suppressor gene Patched1 (Ptch). Analysis of the conditional Ptch knockout mouse model for BCC reveals that macrophages and dendritic cells (DC) of the skin play an important role in BCC growth restraining processes. This is based on the observation that a clodronate-liposome mediated depletion of these cells in the tumor-bearing skin results in significant BCC enlargement. The depletion of these cells does not modulate Ki67 or K10 expression, but is accompanied by a decrease in collagen-producing cells in the tumor stroma. Together, the data suggest that cutaneous macrophages and DC in the tumor microenvironment exert an antitumor effect on BCC.
Collapse
Affiliation(s)
- Simone König
- Institute of Human Genetics, University Medical Center, Goettingen, Germany
| | - Frauke Nitzki
- Institute of Human Genetics, University Medical Center, Goettingen, Germany
| | - Anja Uhmann
- Institute of Human Genetics, University Medical Center, Goettingen, Germany
| | - Kai Dittmann
- Institute of Cellular and Molecular Immunology, University Medical Center, Goettingen, Germany
| | | | - Markus Herrmann
- Department of Radiation Oncology, University Medical Center, Goettingen, Germany
| | - Holger M. Reichardt
- Institute of Cellular and Molecular Immunology, University Medical Center, Goettingen, Germany
| | - Reto Schwendener
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Tobias Pukrop
- Department of Hematology and Oncology, University Medical Center, Goettingen, Germany
| | | | - Heidi Hahn
- Institute of Human Genetics, University Medical Center, Goettingen, Germany
- * E-mail:
| |
Collapse
|
39
|
So PL, Wang GY, Wang K, Chuang M, Chiueh VC, Kenny PA, Epstein EH. PI3K-AKT signaling is a downstream effector of retinoid prevention of murine basal cell carcinogenesis. Cancer Prev Res (Phila) 2014; 7:407-17. [PMID: 24449057 DOI: 10.1158/1940-6207.capr-13-0304] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Basal cell carcinoma (BCC) is the most common human cancer. We have demonstrated previously that topical application of the retinoid prodrug tazarotene profoundly inhibits murine BCC carcinogenesis via retinoic acid receptor γ-mediated regulation of tumor cell transcription. Because topical retinoids can cause adverse cutaneous effects and because tumors can develop resistance to retinoids, we have investigated mechanisms downstream of tazarotene's antitumor effect in this model. Specifically we have used (i) global expression profiling to identify and (ii) functional cell-based assays to validate the phosphoinositide 3-kinase (PI3K)/AKT/mTOR pathway as a downstream target pathway of tazarotene's action. Crucially, we have demonstrated that pharmacologic inhibition of this downstream pathway profoundly reduces murine BCC cell proliferation and tumorigenesis both in vitro and in vivo. These data identify PI3K/AKT/mTOR signaling as a highly attractive target for BCC chemoprevention and indicate more generally that this pathway may be, in some contexts, an important mediator of retinoid anticancer effects.
Collapse
Affiliation(s)
- Po-Lin So
- Jr., Children's Hospital Oakland Research Institute, 5700 Martin Luther King Jr. Way, Oakland, CA 94609. ; and Po-Lin So,
| | | | | | | | | | | | | |
Collapse
|
40
|
König S, Regen T, Dittmann K, Engelke M, Wienands J, Schwendener R, Hanisch UK, Pukrop T, Hahn H. Empty liposomes induce antitumoral effects associated with macrophage responses distinct from those of the TLR1/2 agonist Pam3CSK 4 (BLP). Cancer Immunol Immunother 2013; 62:1587-97. [PMID: 23917775 PMCID: PMC11028801 DOI: 10.1007/s00262-013-1444-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 05/24/2013] [Indexed: 12/31/2022]
Abstract
Liposomes are frequently used in cancer therapy to encapsulate and apply anticancer drugs. Here, we show that a systemic treatment of mice bearing skin tumors with empty phosphatidylcholine liposomes (PCL) resulted in inhibition of tumor growth, which was similar to that observed with the synthetic bacterial lipoprotein and TLR1/2 agonist Pam(3)CSK(4) (BLP). Both compounds led to a substantial decrease of macrophages in spleen and in the tumor-bearing skin. Furthermore, both treatments induced the expression of typical macrophage markers in the tumor-bearing tissue. As expected, BLP induced the expression of the M1 marker genes Cxcl10 and iNOS, whereas PCL, besides inducing iNOS, also increased the M2 marker genes Arg1 and Trem2. In vitro experiments demonstrated that neither PCL nor BLP influenced proliferation or survival of tumor cells, whereas both compounds inhibited proliferation and survival and increased the migratory capacity of bone marrow-derived macrophages (BMDM). However, in contrast to BLP, PCL did not activate cytokine secretion and induced a different BMDM phenotype. Together, the data suggest that similar to BLP, PCL induce an antitumor response by influencing the tumor microenvironment, in particular by functional alterations of macrophages, however, in a distinct manner from those induced by BLP.
Collapse
Affiliation(s)
- Simone König
- Institute of Human Genetics, University Medical Center, Heinrich-Düker-Weg 12, 37073 Göttingen, Germany
| | - Tommy Regen
- Department of Neuropathology, University Medical Center, Göttingen, Germany
- Present Address: Institute for Molecular Medicine, University of Mainz, Mainz, Germany
| | - Kai Dittmann
- Department of Cellular and Molecular Immunology, University Medical Center, Göttingen, Germany
| | - Michael Engelke
- Department of Cellular and Molecular Immunology, University Medical Center, Göttingen, Germany
| | - Jürgen Wienands
- Department of Cellular and Molecular Immunology, University Medical Center, Göttingen, Germany
| | - Reto Schwendener
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | | | - Tobias Pukrop
- Department of Hematology and Oncology, University Medical Center, Göttingen, Germany
| | - Heidi Hahn
- Institute of Human Genetics, University Medical Center, Heinrich-Düker-Weg 12, 37073 Göttingen, Germany
| |
Collapse
|
41
|
Imiquimod directly inhibits Hedgehog signalling by stimulating adenosine receptor/protein kinase A-mediated GLI phosphorylation. Oncogene 2013; 32:5574-81. [PMID: 23995793 PMCID: PMC3898320 DOI: 10.1038/onc.2013.343] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 06/27/2013] [Accepted: 07/19/2013] [Indexed: 12/20/2022]
Abstract
Imiquimod (IMQ), a nucleoside analogue of the imidazoquinoline family, is used in the topical treatment of basal cell carcinoma (BCC) and other skin diseases. It is reported to be a TLR7 and TLR8 agonist and, as such, initiates a Th1 immune response by activating sentinel cells in the vicinity of the tumour. BCC is a hedgehog (HH)-driven malignancy with oncogenic glioma-associated oncogene (GLI) signalling activated in a ligand-independent manner. Here we show that IMQ can also directly repress HH signalling by negatively modulating GLI activity in BCC and medulloblastoma cells. Further, we provide evidence that the repressive effect of IMQ on HH signalling is not dependent on TLR/MYD88 signalling. Our results suggest a mechanism for IMQ engaging adenosine receptors (ADORAs) to control GLI signalling. Pharmacological activation of ADORA with either an ADORA agonist or IMQ resulted in a protein kinase A (PKA)-mediated GLI phosphorylation and reduction in GLI activator levels. The activation of PKA and HH pathway target gene downregulation in response to IMQ were abrogated by ADORA inhibition. Furthermore, activated Smoothened signalling, which positively signals to GLI transcription factors, could be effectively counteracted by IMQ. These results reveal a previously unknown mode of action of IMQ in the treatment of BCC and also suggest a role for ADORAs in the regulation of oncogenic HH signalling.
Collapse
|
42
|
DeBerardinis AM, Banerjee U, Hadden MK. Identification of vitamin d3-based hedgehog pathway inhibitors that incorporate an aromatic a-ring isostere. ACS Med Chem Lett 2013; 4:590-5. [PMID: 24900716 DOI: 10.1021/ml400014t] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 05/14/2013] [Indexed: 01/21/2023] Open
Abstract
Previous structure-activity relationship studies for vitamin D3 (VD3) inhibition of Hedgehog (Hh) signaling directed the design, synthesis, and evaluation of a series of VD3-based analogues that contain an aromatic A-ring mimic. Characterization of these compounds in a series of cellular assays demonstrated their ability to potently and selectively down-regulate Hh pathway signaling. The most active of these, 17, inhibited pathway signaling in Hh-dependent mouse fibroblasts (IC50 = 0.74 ± 0.1 μM) and cultured cancer cells (IC50 values 3.8 ± 0.1 to 5.2 ± 0.2 μM). In addition, 17 demonstrated reduced activation of the vitamin D receptor (VDR) compared to VD3 in these cellular models. These results suggest that VD3-based analogues with an aromatic A-ring are a valid scaffold for the development of more selective and potent Hh pathway inhibitors and identify 17 as an intriguing lead from this class of compounds for further development. In addition, our analysis of Hh pathway inhibitors in cancer cells suggests that the murine basal cell carcinoma cell line ASZ001 and the human medulloblastoma cell line DAOY are appropriate in vitro cancer models for early stage evaluation of pathway inhibition.
Collapse
Affiliation(s)
- Albert M. DeBerardinis
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road,
Unit 3092, Storrs, Connecticut 06269-3092, United States
| | - Upasana Banerjee
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road,
Unit 3092, Storrs, Connecticut 06269-3092, United States
| | - M. Kyle Hadden
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road,
Unit 3092, Storrs, Connecticut 06269-3092, United States
| |
Collapse
|
43
|
Gallinari P, Filocamo G, Jones P, Pazzaglia S, Steinkühler C. Smoothened antagonists: a promising new class of antitumor agents. Expert Opin Drug Discov 2013; 4:525-44. [PMID: 23485085 DOI: 10.1517/17460440902852686] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Hedgehog signaling is essential for the development of most metazoans. In recent years, evidence has accumulated showing that many human tumors aberrantly re-activate this developmental signaling pathway and that interfering with it may provide a new strategy for the development of novel anti-cancer therapeutics. Smoothened is a G-protein coupled receptor-like protein that is essentially involved in hedgehog signal transduction and small molecule antagonists of Smoothened have started to show antitumor activity in preclinical models and in clinical trials. OBJECTIVE We critically review the role of hedgehog signaling in normal development and in human malignancies, the available drug discovery tools and the classes of small molecule inhibitors that are in development. We further aim to address the potential impact that pathway antagonists may have on the treatment options of cancer patients. METHODS Literature, patents and clinical trial results from the past 5 years were analyzed. CONCLUSIONS 1) A large body of evidence suggests a frequent reactivation of hedgehog signaling in human cancer. 2) Smoothened is an attractive, highly druggable target with extensive preclinical and initial clinical validation in basal cell carcinoma. Several promising novel classes of Smoothened antagonists have been discovered and are being developed as anticancer agents. 3) Our knowledge of the biology of hedgehog signaling in cancer is still very incomplete and significant efforts will be required to understand how to use the emerging novel agents in the clinic.
Collapse
Affiliation(s)
- Paola Gallinari
- Istituto di Ricerche di Biologia Molecolare P. Angeletti, Department of Oncology, IRBM- Merck Research Laboratories Rome, Via Pontina Km 30,600, 00040 Pomezia, Italy +39 06 91093232 ; +39 06 91093549 ;
| | | | | | | | | |
Collapse
|
44
|
Small molecule inhibitors of Smoothened ciliary localization and ciliogenesis. Proc Natl Acad Sci U S A 2012; 109:13644-9. [PMID: 22864913 DOI: 10.1073/pnas.1207170109] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Vertebrate Hedgehog (Hh) signals involved in development and some forms of cancer, such as basal cell carcinoma, are transduced by the primary cilium, a microtubular projection found on many cells. A critical step in vertebrate Hh signal transduction is the regulated movement of Smoothened (Smo), a seven-transmembrane protein, to the primary cilium. To identify small molecules that interfere with either the ciliary localization of Smo or ciliogenesis, we undertook a high-throughput, microscopy-based screen for compounds that alter the ciliary localization of YFP-tagged Smo. This screen identified 10 compounds that inhibit Hh pathway activity. Nine of these Smo antagonists (SA1-9) bind Smo, and one (SA10) does not. We also identified two compounds that inhibit ciliary biogenesis, which block microtubule polymerization or alter centrosome composition. Differential labeling of cell surface and intracellular Smo pools indicates that SA1-7 and 10 specifically inhibit trafficking of intracellular Smo to cilia. In contrast, SA8 and 9 recruit endogenous Smo to the cilium in some cell types. Despite these different mechanisms of action, all of the SAs inhibit activation of the Hh pathway by an oncogenic form of Smo, and abrogate the proliferation of basal cell carcinoma-like cancer cells. The SA compounds may provide alternative means of inhibiting pathogenic Hh signaling, and our study reveals that different pools of Smo move into cilia through distinct mechanisms.
Collapse
|
45
|
Eberl M, Klingler S, Mangelberger D, Loipetzberger A, Damhofer H, Zoidl K, Schnidar H, Hache H, Bauer HC, Solca F, Hauser-Kronberger C, Ermilov AN, Verhaegen ME, Bichakjian CK, Dlugosz AA, Nietfeld W, Sibilia M, Lehrach H, Wierling C, Aberger F. Hedgehog-EGFR cooperation response genes determine the oncogenic phenotype of basal cell carcinoma and tumour-initiating pancreatic cancer cells. EMBO Mol Med 2012; 4:218-33. [PMID: 22294553 PMCID: PMC3305999 DOI: 10.1002/emmm.201100201] [Citation(s) in RCA: 135] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Revised: 12/09/2011] [Accepted: 12/12/2011] [Indexed: 12/20/2022] Open
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.
Collapse
Affiliation(s)
- Markus Eberl
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Establishment of murine basal cell carcinoma allografts: a potential model for preclinical drug testing and for molecular analysis. J Invest Dermatol 2011; 131:2298-305. [PMID: 21833014 PMCID: PMC3193585 DOI: 10.1038/jid.2011.204] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Dysregulated hedgehog (HH) signaling has been found in numerous cancers, suggesting that therapeutic targeting of this pathway may be useful vs. a wide range of cancers. Basal cell carcinoma (BCC) is an excellent model system for studying the influence of the HH pathway on carcinogenesis because aberrant activation of HH signaling not only is crucial for the development but also for the maintenance of BCC. Genetically engineered BCC mouse models provide one important tool for the study of the biology of human BCCs and for evaluating therapeutic interventions since these mice produce multiple genetically defined tumors within a relatively short period of time. However, these models remain expensive and cumbersome to use for large-scale pre-clinical drug testing. Here we report a method for growing allografts from murine BCC tumors in NOD/SCID mice. These allografts develop faster and reproduce the histology, immunophenotypes and response to at least one anti-BCC drug of the parental autochthonous tumors from which they arise. Therefore, the allograft model provides a practical model for (i) studying BCC carcinogenesis and (ii) initial pre-clinical screening for anti-HH pathway and other anti-BCC drugs.
Collapse
|
47
|
Kuphal S, Shaw-Hallgren G, Eberl M, Karrer S, Aberger F, Bosserhoff AK, Massoumi R. GLI1-dependent transcriptional repression of CYLD in basal cell carcinoma. Oncogene 2011; 30:4523-30. [PMID: 21577203 DOI: 10.1038/onc.2011.163] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
CYLD is a deubiquitination enzyme that regulates different cellular processes, such as cell proliferation and cell survival. Mutation and loss of heterozygosity of the CYLD gene causes development of cylindromatosis, a benign tumour originating from the skin. Our study shows that CYLD expression is dramatically downregulated in basal cell carcinoma (BCC), the most common cancer in humans. Reduced CYLD expression in basal cell carcinoma was mediated by GLI1-dependent activation of the transcriptional repressor Snail. Inhibition of GLI1 restored the CYLD expression-mediated Snail signaling pathway, and caused a significant delay in the G1 to S phase transition, as well as proliferation. Our data suggest that GLI1-mediated suppression of CYLD has a significant role in basal cell carcinoma progression.
Collapse
Affiliation(s)
- S Kuphal
- Institute of Pathology, University of Regensburg, Germany
| | | | | | | | | | | | | |
Collapse
|
48
|
Tang JY, Xiao TZ, Oda Y, Chang KS, Shpall E, Wu A, So PL, Hebert J, Bikle D, Epstein EH. Vitamin D3 inhibits hedgehog signaling and proliferation in murine Basal cell carcinomas. Cancer Prev Res (Phila) 2011; 4:744-51. [PMID: 21436386 PMCID: PMC3088781 DOI: 10.1158/1940-6207.capr-10-0285] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Constitutive Hedgehog (HH) signaling underlies several human tumors, including basal cell carcinoma (BCC). Recently, Bijlsma and colleagues reported a new biologic function for vitamin D3 in suppressing HH signaling in an in vitro model system. On the basis of that work, we have assessed effects of vitamin D3 on HH signaling and proliferation of murine BCCs in vitro and in vivo. We find that indeed in BCC cells, vitamin D3 blocks both proliferation and HH signaling as assessed by mRNA expression of the HH target gene Gli1. These effects of vitamin D3 on Gli1 expression and on BCC cell proliferation are comparable to the effects of cyclopamine, a known inhibitor of the HH pathway. These results are specific for vitamin D3, because the precursor 7-dehydrocholesterol and the downstream products 25-hydroxy vitamin D3 [25(OH)D] and 1,25-dihydroxy vitamin D3 [1,25(OH)(2)D] are considerably less effective in reducing either Gli1 mRNA or cellular proliferation. Moreover, these effects seem to be independent of the vitamin D receptor (VDR) because short hairpin RNA knockdown of VDR does not abrogate the anti-HH effects of D3 despite reducing expression of the VDR target gene 24-hydroxylase. Finally, topical vitamin D3 treatment of existing murine BCC tumors significantly decreases Gli1 and Ki67 staining. Thus, topical vitamin D3 acting via its HH inhibiting effect may hold promise as an effective anti-BCC agent.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Bone Density Conservation Agents/pharmacology
- Carcinoma, Basal Cell/genetics
- Carcinoma, Basal Cell/metabolism
- Carcinoma, Basal Cell/pathology
- Cell Differentiation/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Cerebellar Neoplasms/genetics
- Cerebellar Neoplasms/metabolism
- Cerebellar Neoplasms/pathology
- Cholecalciferol/pharmacology
- Hedgehog Proteins/genetics
- Hedgehog Proteins/metabolism
- Immunoenzyme Techniques
- Keratinocytes/cytology
- Keratinocytes/metabolism
- Kruppel-Like Transcription Factors/genetics
- Kruppel-Like Transcription Factors/metabolism
- Medulloblastoma/genetics
- Medulloblastoma/metabolism
- Medulloblastoma/pathology
- Mice
- RNA, Messenger/genetics
- RNA, Small Interfering/genetics
- Receptors, Calcitriol/antagonists & inhibitors
- Receptors, Calcitriol/genetics
- Receptors, Calcitriol/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Steroid Hydroxylases/genetics
- Steroid Hydroxylases/metabolism
- Vitamin D3 24-Hydroxylase
- Zinc Finger Protein GLI1
Collapse
Affiliation(s)
- Jean Y Tang
- Department of Dermatology, Stanford University School of Medicine, Redwood City, CA 94063-5334, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Kurokawa I, Takahashi K, Moll I, Moll R. Expression of keratins in cutaneous epithelial tumors and related disorders - distribution and clinical significance. Exp Dermatol 2011; 20:217-28. [DOI: 10.1111/j.1600-0625.2009.01006.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
50
|
So PL, Tang JY, Epstein EH. Novel investigational drugs for basal cell carcinoma. Expert Opin Investig Drugs 2011; 19:1099-112. [PMID: 20662553 DOI: 10.1517/13543784.2010.504714] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
IMPORTANCE OF THE FIELD In the United States, the annual incidence of basal cell carcinoma (BCC) is close to 1 million. Ultraviolet radiation exposure is the main risk factor; however, the availability of ever more potent sunscreens and education have not prevented the rise in BCC incidence. Therefore, concerted effects to identify novel preventive and therapeutic strategies are necessary. AREAS COVERED IN THIS REVIEW This article summarizes our current understanding of the etiology and molecular mechanisms of BCC tumorigenesis and discusses the preclinical and clinical studies to identify agents with anti-BCC efficacy. WHAT THE READER WILL GAIN The discovery that hyperactive Hh pathway signaling causes several cancers, including BCC, has spawned the development of many pharmacologic inhibitors of Hh signaling. Early clinical testing of the most advanced, GDC-0449, demonstrated impressive efficacy in patients with advanced BCC. Other promising anti-BCC chemopreventive strategies include drugs that are already FDA-approved for treating other diseases. TAKE HOME MESSAGE Preclinical and clinical trials with pre-existing FDA-approved drugs suggest novel uses for BCC chemoprevention and treatment. Also, new chemical entities that inhibit the Hh pathway show promise, and in combination with other drugs may provide a nonsurgical cure for this most common cancer.
Collapse
Affiliation(s)
- Po-Lin So
- Children's Hospital Oakland Research Institute, Cancer Division, 5700 Martin Luther King Jr Way, Oakland, CA 94609, USA.
| | | | | |
Collapse
|