1
|
Pacheco-Tovar D, Pacheco-Tovar MG, Saavedra-Alonso S, Zapata-Benavides P, Torres-del-Muro FDJ, Bollain-y-Goytia JJ, Herrera-Esparza R, Rodríguez-Padilla C, Avalos-Díaz E. shRNA-Targeting Caspase-3 Inhibits Cell Detachment Induced by Pemphigus Vulgaris Autoantibodies in HaCaT Cells. Int J Mol Sci 2024; 25:8864. [PMID: 39201550 PMCID: PMC11354573 DOI: 10.3390/ijms25168864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Pemphigus is an autoimmune disease that affects the skin and mucous membranes, induced by the deposition of pemphigus IgG, which mainly targets desmogleins 1 and 3 (Dsg1 and 3). This autoantibody causes steric interference between Dsg1 and 3 and the loss of cell adhesion, producing acantholysis. This molecule and its cellular effects are clinically reflected as intraepidermal blistering. Pemphigus vulgaris-IgG (PV-IgG) binding involves p38MAPK-signaling-dependent caspase-3 activation. The present work assessed the in vitro effect of PV-IgG on the adherence of HaCaT cells dependent on caspase-3. PV-IgG induced cell detachment and apoptotic changes, as demonstrated by annexin fluorescent assays. The effect of caspase-3 induced by PV-IgG was suppressed in cells pre-treated with caspase-3-shRNA, and normal IgG (N-IgG) as a control had no relevant effects on the aforementioned parameters. The results demonstrated that shRNA reduces caspase-3 expression, as measured via qRT-PCR and via Western blot and immunofluorescence, and increases cell adhesion. In conclusion, shRNA prevented in vitro cell detachment and the late effects of apoptosis induced by PV-IgG on HaCaT cells, furthering our understanding of the molecular role of caspase-3 cell adhesion dependence in pemphigus disease.
Collapse
Affiliation(s)
- Deyanira Pacheco-Tovar
- Department of Immunology, School of Biological Sciences, UACB, Universidad Autónoma de Zacatecas, Av. de la Revolución Mexicana s/n, Colonia Tierra y Libertad, Guadalupe CP 98615, Zacatecas, Mexico; (D.P.-T.); (M.-G.P.-T.); (F.-d.-J.T.-d.-M.); (J.-J.B.-y.-G.)
- School of Chemistry Sciences, Universidad Autónoma de Zacatecas, Campus Universitario Siglo XXI, Carretera Zacatecas-Guadalajara, Ejido “La Escondida”, Zacatecas CP 98160, Zacatecas, Mexico
- Department of Immunology and Virology, Faculty of Biological Sciences, Universidad Autónoma de Nuevo León, Av. Pedro de Alba s/n, Ciudad Universitaria, San Nicolás de los Garza CP 64450, Nuevo León, Mexico; (S.S.-A.); (C.R.-P.)
| | - María-Guadalupe Pacheco-Tovar
- Department of Immunology, School of Biological Sciences, UACB, Universidad Autónoma de Zacatecas, Av. de la Revolución Mexicana s/n, Colonia Tierra y Libertad, Guadalupe CP 98615, Zacatecas, Mexico; (D.P.-T.); (M.-G.P.-T.); (F.-d.-J.T.-d.-M.); (J.-J.B.-y.-G.)
- School of Chemistry Sciences, Universidad Autónoma de Zacatecas, Campus Universitario Siglo XXI, Carretera Zacatecas-Guadalajara, Ejido “La Escondida”, Zacatecas CP 98160, Zacatecas, Mexico
- Department of Immunology and Virology, Faculty of Biological Sciences, Universidad Autónoma de Nuevo León, Av. Pedro de Alba s/n, Ciudad Universitaria, San Nicolás de los Garza CP 64450, Nuevo León, Mexico; (S.S.-A.); (C.R.-P.)
| | - Santiago Saavedra-Alonso
- Department of Immunology and Virology, Faculty of Biological Sciences, Universidad Autónoma de Nuevo León, Av. Pedro de Alba s/n, Ciudad Universitaria, San Nicolás de los Garza CP 64450, Nuevo León, Mexico; (S.S.-A.); (C.R.-P.)
| | - Pablo Zapata-Benavides
- Department of Immunology and Virology, Faculty of Biological Sciences, Universidad Autónoma de Nuevo León, Av. Pedro de Alba s/n, Ciudad Universitaria, San Nicolás de los Garza CP 64450, Nuevo León, Mexico; (S.S.-A.); (C.R.-P.)
| | - Felipe-de-Jesús Torres-del-Muro
- Department of Immunology, School of Biological Sciences, UACB, Universidad Autónoma de Zacatecas, Av. de la Revolución Mexicana s/n, Colonia Tierra y Libertad, Guadalupe CP 98615, Zacatecas, Mexico; (D.P.-T.); (M.-G.P.-T.); (F.-d.-J.T.-d.-M.); (J.-J.B.-y.-G.)
- School of Chemistry Sciences, Universidad Autónoma de Zacatecas, Campus Universitario Siglo XXI, Carretera Zacatecas-Guadalajara, Ejido “La Escondida”, Zacatecas CP 98160, Zacatecas, Mexico
- Department of Immunology and Virology, Faculty of Biological Sciences, Universidad Autónoma de Nuevo León, Av. Pedro de Alba s/n, Ciudad Universitaria, San Nicolás de los Garza CP 64450, Nuevo León, Mexico; (S.S.-A.); (C.R.-P.)
| | - Juan-José Bollain-y-Goytia
- Department of Immunology, School of Biological Sciences, UACB, Universidad Autónoma de Zacatecas, Av. de la Revolución Mexicana s/n, Colonia Tierra y Libertad, Guadalupe CP 98615, Zacatecas, Mexico; (D.P.-T.); (M.-G.P.-T.); (F.-d.-J.T.-d.-M.); (J.-J.B.-y.-G.)
| | - Rafael Herrera-Esparza
- Department of Immunology, School of Biological Sciences, UACB, Universidad Autónoma de Zacatecas, Av. de la Revolución Mexicana s/n, Colonia Tierra y Libertad, Guadalupe CP 98615, Zacatecas, Mexico; (D.P.-T.); (M.-G.P.-T.); (F.-d.-J.T.-d.-M.); (J.-J.B.-y.-G.)
| | - Cristina Rodríguez-Padilla
- Department of Immunology and Virology, Faculty of Biological Sciences, Universidad Autónoma de Nuevo León, Av. Pedro de Alba s/n, Ciudad Universitaria, San Nicolás de los Garza CP 64450, Nuevo León, Mexico; (S.S.-A.); (C.R.-P.)
| | - Esperanza Avalos-Díaz
- Department of Immunology, School of Biological Sciences, UACB, Universidad Autónoma de Zacatecas, Av. de la Revolución Mexicana s/n, Colonia Tierra y Libertad, Guadalupe CP 98615, Zacatecas, Mexico; (D.P.-T.); (M.-G.P.-T.); (F.-d.-J.T.-d.-M.); (J.-J.B.-y.-G.)
| |
Collapse
|
2
|
Nabeta R, Kanaya A, Elbadawy M, Usui T, Furuya T, Suzuki K, Uchide T. Chemosensitivity of three patient-derived primary cultures of canine pericardial mesothelioma by single-agent and combination treatment. Front Vet Sci 2023; 10:1267359. [PMID: 38026668 PMCID: PMC10653591 DOI: 10.3389/fvets.2023.1267359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/10/2023] [Indexed: 12/01/2023] Open
Abstract
INTRODUCTION Canine mesothelioma is a rare malignant tumor that mostly affects body cavities, such as the pericardial and pleural cavities. Chemotherapy plays a crucial role in the treatment of canine mesotheliomas. We aimed to compare the antitumor effects of single-agent and combination chemotherapeutic agents on patient-derived primary cultures of canine pericardial mesothelioma established in this study. We planned to generate xenograft models for future studies. MATERIAL AND METHODS Effusion samples were collected from three dogs with histologically diagnosed pericardial mesothelioma and used for primary culture. Cultured cells were characterized by immunostaining for pan-cytokeratin AE1/AE3, vimentin, Wilms' tumor suppressor gene 1 (WT1), and cytokeratin 5 (CK5). To assess the tumorigenic properties of cells in the effusion and generate a xenograft model, the cell suspension was injected into a severe combined immunodeficient (SCID) mouse either subcutaneously (SC) or intraperitoneally (IP). Lastly, chemosensitivity of established primary cultures against four drugs, doxorubicin, vinorelbine, carboplatin, and gemcitabine, by single-agent treatment as well as combination treatment of carboplatin at a fixed concentration, either 10 or 100 μM, and gemcitabine at different concentrations ranging from 0-1000 μM was assessed by cell viability assay. RESULTS Primary cultures were successfully generated and characterized by dual positivity for AE1/AE3 and vimentin and positive staining for WT-1 and CK5, confirming the mesothelial origin of the cells. In the xenograft models, SC mouse developed a subcutaneous mass, whereas IP mouse developed multiple intraperitoneal nodules. The masses were histopathologically consistent with mesotheliomas. The chemosensitivity assay revealed that carboplatin had the highest anti-tumor effects among the four tested single-agent treatments. Furthermore, carboplatin at 100 μM combined with gemcitabine at clinically relevant doses demonstrated the augmented anti-tumor effects compared to single-agent treatment. DISCUSSION AND CONCLUSION Primary cultures and xenograft models generated in this study could be useful tools for in vitro and in vivo studies of canine mesothelioma. Carboplatin is a highly effective chemotherapeutic agent against canine mesothelioma when used as a sole agent and in combination with gemcitabine.
Collapse
Affiliation(s)
- Rina Nabeta
- Laboratory of Veterinary Molecular Pathology and Therapeutics, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Tokyo, Japan
| | - Ami Kanaya
- Laboratory of Veterinary Molecular Pathology and Therapeutics, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Tokyo, Japan
| | - Mohamed Elbadawy
- Laboratory of Veterinary Pharmacology, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Tokyo, Japan
- Department of Pharmacology, Faculty of Veterinary Medicine, Benha University, Benha, Egypt
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Tatsuya Usui
- Laboratory of Veterinary Pharmacology, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Tokyo, Japan
| | - Tetsuya Furuya
- Laboratory of Veterinary Infectious Diseases, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Tokyo, Japan
| | - Kazuhiko Suzuki
- Laboratory of Veterinary Toxicology, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Tokyo, Japan
| | - Tsuyoshi Uchide
- Laboratory of Veterinary Molecular Pathology and Therapeutics, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Tokyo, Japan
| |
Collapse
|
3
|
Rangel-Sánchez IY, Salas-Treviño D, Soto-Domínguez A, Garza-Rodríguez OI, Saucedo-Cárdenas O, Zapata-Benavides P, Zarate-Ramos JJ, Cedillo-Rosales S, Zamora-Ávila DE. Expression of the Wilms' tumour gene and its association with PPARβ/δ in healthy skin and melanoma of horses. Acta Vet Hung 2021; 68:374-379. [PMID: 33459615 DOI: 10.1556/004.2020.00045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 04/21/2020] [Indexed: 11/19/2022]
Abstract
The Wilms' tumour gene (WT1) has previously been described as an oncogene in several neoplasms of humans, including melanoma, and its expression increases cancer cell proliferation. Recent reports associate the expression of the PPARβ/δ gene (peroxisome proliferator-activated receptor beta/delta) with the downregulation of WT1 in human melanoma and murine melanoma cell lines. The aim of this work was to analyse the expression of WT1 and its association with PPARβ/δ in samples of healthy and melanoma-affected skin of horses by immunohistochemistry. WT1 protein expression was detected in healthy skin, mainly in the epidermis, hair follicle, sebaceous gland and sweat gland, while no expression was observed in equine melanoma tissues. Moreover, it was observed that PPARβ/δ has a basal expression in healthy skin and that it is overexpressed in melanoma. These results were confirmed by a densitometric analysis, where a significant increase of the WT1-positive area was observed in healthy skin (128.66 ± 19.84 pixels 106) compared with that observed in melanoma (1.94 ± 0.04 pixels 106). On the other hand, a positive area with an expression of PPARβ/δ in healthy skin (214.94 ± 11.85 pixels 106) was significantly decreased compared to melanoma (624.86 ± 181.93 pixels 106). These data suggest that there could be a regulation between WT1 and PPARβ/δ in this disease in horses.
Collapse
Affiliation(s)
- Itzel Y Rangel-Sánchez
- 1Department of Genetics, Faculty of Medicine Veterinary and Zootechnics, Autonomous University of Nuevo León, Av. Francisco Villa s/n, Ex Hacienda el Canadá, 66050 Gral. Escobedo, NL, Mexico
| | - Daniel Salas-Treviño
- 2Department of Histology, Faculty of Medicine, Autonomous University of Nuevo León, Mexico
| | - Adolfo Soto-Domínguez
- 2Department of Histology, Faculty of Medicine, Autonomous University of Nuevo León, Mexico
| | - Oscar I Garza-Rodríguez
- 1Department of Genetics, Faculty of Medicine Veterinary and Zootechnics, Autonomous University of Nuevo León, Av. Francisco Villa s/n, Ex Hacienda el Canadá, 66050 Gral. Escobedo, NL, Mexico
| | - Odila Saucedo-Cárdenas
- 2Department of Histology, Faculty of Medicine, Autonomous University of Nuevo León, Mexico
- 3Northeast Biomedical Research Centre (CIBIN) of the IMSS, Monterrey, NL, Mexico
| | - Pablo Zapata-Benavides
- 4Laboratory of Immunology and Virology, Faculty of Biological Sciences, Autonomous University of Nuevo León, Mexico
| | - Juan J Zarate-Ramos
- 5Department of Parasitology, Faculty of Medicine Veterinary and Zootechnics, Autonomous University of Nuevo León, Mexico
| | - Sibilina Cedillo-Rosales
- 6Department of Virology, Faculty of Medicine Veterinary and Zootechnics, Autonomous University of Nuevo León, Mexico
| | - Diana E Zamora-Ávila
- 1Department of Genetics, Faculty of Medicine Veterinary and Zootechnics, Autonomous University of Nuevo León, Av. Francisco Villa s/n, Ex Hacienda el Canadá, 66050 Gral. Escobedo, NL, Mexico
| |
Collapse
|
4
|
Inhibitory Effect of Cuphea aequipetala Extracts on Murine B16F10 Melanoma In Vitro and In Vivo. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8560527. [PMID: 31275985 PMCID: PMC6560323 DOI: 10.1155/2019/8560527] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 05/07/2019] [Indexed: 12/12/2022]
Abstract
Cuphea aequipetala (C. aequipetala) has been used in Mexican traditional medicine since prehispanic times to treat tumors. In this paper, we evaluated the antiproliferative and apoptotic effect of the methanolic and aqueous extracts of C. aequipetala on several cancer cell lines including the B16F10 cell line of murine melanoma and carried a murine model assay. In vitro assay analyzed the effect in the cellular cycle and several indicators of apoptosis, such as the caspase-3 activity, DNA fragmentation, phosphatidylserine exposure (Annexin-V), and induction of cell membrane permeabilization (propidium iodide) in the B16F10 cells. In vivo, groups of C57BL/6 female mice were subcutaneously injected with 5x105 B16F10 cells and treated with 25 mg/mL of C. aequipetala extracts via oral. Aqueous and methanolic extracts showed a cytotoxic effect in MCF-7, HepG2, and B16F10 cell lines. The methanolic extract showed more antiproliferative effect with less concentration, and for this reason, the in vitro experiments were only continued with it. This extract was able to induce accumulation of cells on G1 phase of the cell cycle; moreover, it was able to induce DNA fragmentation and increase the activity of caspase-3 in B16F10 cells. On the other hand, in the murine model of melanoma, the aqueous extract showed a greater reduction of tumor size in comparison with the methanolic extract, showing an 80% reduction versus one of around 31%, both compared with the untreated control, indicating a better antitumor effect of C. aequipetala aqueous extract via oral administration. In conclusion, the in vitro data showed that both C. aequipetala extracts were able to induce cytotoxicity through the apoptosis pathway in B16F10 cells, and in vivo, the oral administration of aqueous extract reduces the melanoma tumoral mass, suggesting an important antitumoral effect and the perspective to search for effector molecules involved in it.
Collapse
|
5
|
Zapata-Benavides P, Thompson-Armendariz FG, Arellano-Rodríguez M, Franco-Molina MA, Mendoza-Gamboa E, Saavedra-Alonso S, Zacarias-Hernández JL, Trejo-Avila LM, Rodríguez-Padilla C. shRNA-WT1 Potentiates Anticancer Effects of Gemcitabine and Cisplatin Against B16F10 Lung Metastases In Vitro and In Vivo. In Vivo 2019; 33:777-785. [PMID: 31028197 PMCID: PMC6559916 DOI: 10.21873/invivo.11539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/02/2019] [Accepted: 02/07/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND/AIM High expression level of Wilm's tumor gene (WT1) in several types of tumors appears to confer disruption of apoptosis and resistance to chemotherapeutic drugs, and correlate with poor outcome. The aim of this work was to determine if down-regulation of WT1 expression results in decreased cell proliferation and the increased action of different types of drugs, both in vitro in B16F10 cells, and in vivo in C57BL/6 mice. MATERIALS AND METHODS Inhibition of cell proliferation by short hairpin RNA against WT1 (shRNA-WT1), cisplatin, and gemcitabine in B16F10 cells in vitro was determined by the MTT assay and analysis of clonogenic survival. The apoptosis rate was determined by flow cytometry for annexin-V- fluorescein isothiocyante and propidium iodide. RESULTS Compared to treatment with shRNA-WT1 alone, treatment with shRNA-WT1 in combination with drugs had a synergistic inhibitory effect on B16F10 cell proliferation, particularly for the combination of cisplatin and gemcitabine at their 25% cytotoxic concentrations in vitro. Furthermore, mice treated with shRNA-WT1 in combination with cisplatin and gemcitabine were protected in the same way as those treated with the drugs alone, but were in better physical condition. CONCLUSION Decreased WT1 expression induces cell death and potentiates the action of anticancer drugs by inducing synergistic effects both in vitro and in vivo, which may be an attractive strategy in lung cancer therapy.
Collapse
Affiliation(s)
- Pablo Zapata-Benavides
- Department of Microbiology and Immunology, Faculty of Biological Sciences, University Autonomous of Nuevo Leon (UANL), San Nicolas de los Garza, Mexico
| | | | - Mariela Arellano-Rodríguez
- Department of Microbiology and Immunology, Faculty of Biological Sciences, University Autonomous of Nuevo Leon (UANL), San Nicolas de los Garza, Mexico
| | - Moisés Armides Franco-Molina
- Department of Microbiology and Immunology, Faculty of Biological Sciences, University Autonomous of Nuevo Leon (UANL), San Nicolas de los Garza, Mexico
| | - Edgar Mendoza-Gamboa
- Department of Microbiology and Immunology, Faculty of Biological Sciences, University Autonomous of Nuevo Leon (UANL), San Nicolas de los Garza, Mexico
| | - Santiago Saavedra-Alonso
- Department of Microbiology and Immunology, Faculty of Biological Sciences, University Autonomous of Nuevo Leon (UANL), San Nicolas de los Garza, Mexico
| | - José Luis Zacarias-Hernández
- Department of Microbiology and Immunology, Faculty of Biological Sciences, University Autonomous of Nuevo Leon (UANL), San Nicolas de los Garza, Mexico
| | - Laura María Trejo-Avila
- Department of Microbiology and Immunology, Faculty of Biological Sciences, University Autonomous of Nuevo Leon (UANL), San Nicolas de los Garza, Mexico
| | - Cristina Rodríguez-Padilla
- Department of Microbiology and Immunology, Faculty of Biological Sciences, University Autonomous of Nuevo Leon (UANL), San Nicolas de los Garza, Mexico
| |
Collapse
|
6
|
WT1 expression in breast cancer disrupts the epithelial/mesenchymal balance of tumour cells and correlates with the metabolic response to docetaxel. Sci Rep 2017; 7:45255. [PMID: 28345629 PMCID: PMC5366898 DOI: 10.1038/srep45255] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 02/23/2017] [Indexed: 12/15/2022] Open
Abstract
WT1 is a transcription factor which regulates the epithelial-mesenchymal balance during embryonic development and, if mutated, can lead to the formation of Wilms' tumour, the most common paediatric kidney cancer. Its expression has also been reported in several adult tumour types, including breast cancer, and usually correlates with poor outcome. However, published data is inconsistent and the role of WT1 in this malignancy remains unclear. Here we provide a complete study of WT1 expression across different breast cancer subtypes as well as isoform specific expression analysis. Using in vitro cell lines, clinical samples and publicly available gene expression datasets, we demonstrate that WT1 plays a role in regulating the epithelial-mesenchymal balance of breast cancer cells and that WT1-expressing tumours are mainly associated with a mesenchymal phenotype. WT1 gene expression also correlates with CYP3A4 levels and is associated with poorer response to taxane treatment. Our work is the first to demonstrate that the known association between WT1 expression in breast cancer and poor prognosis is potentially due to cancer-related epithelial-to-mesenchymal transition (EMT) and poor chemotherapy response.
Collapse
|
7
|
Fatty Acids of CLA-Enriched Egg Yolks Can Induce Transcriptional Activation of Peroxisome Proliferator-Activated Receptors in MCF-7 Breast Cancer Cells. PPAR Res 2017; 2017:2865283. [PMID: 28458685 PMCID: PMC5385215 DOI: 10.1155/2017/2865283] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 02/02/2017] [Accepted: 02/12/2017] [Indexed: 12/13/2022] Open
Abstract
In our previous study, we showed that fatty acids from CLA-enriched egg yolks (EFA-CLA) reduced the proliferation of breast cancer cells; however, the molecular mechanisms of their action remain unknown. In the current study, we used MCF-7 breast cancer cell line to determine the effect of EFA-CLA, as potential ligands for peroxisome proliferator-activated receptors (PPARs), on identified in silico PPAR-responsive genes: BCAR3, TCF20, WT1, ZNF621, and THRB (transcript TRβ2). Our results showed that EFA-CLA act as PPAR ligands with agonistic activity for all PPAR isoforms, with the highest specificity towards PPARγ. In conclusion, we propose that EFA-CLA-mediated regulation of PPAR-responsive genes is most likely facilitated by cis9,trans11CLA isomer incorporated in egg yolk. Notably, EFA-CLA activated PPAR more efficiently than nonenriched FA as well as synthetic CLA isomers. We also propose that this regulation, at least in part, can be responsible for the observed reduction in the proliferation of MCF-7 cells treated with EFA-CLA.
Collapse
|
8
|
Plönes T, Fischer M, Höhne K, Sato H, Müller-Quernheim J, Zissel G. Turning back the Wheel: Inducing Mesenchymal to Epithelial Transition via Wilms Tumor 1 Knockdown in Human Mesothelioma Cell Lines to Influence Proliferation, Invasiveness, and Chemotaxis. Pathol Oncol Res 2017; 23:723-730. [PMID: 28054314 DOI: 10.1007/s12253-016-0181-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 12/29/2016] [Indexed: 01/21/2023]
Abstract
Malignant pleural mesothelioma (MPM) is a highly aggressive tumor that arises from the surface of the pleura and is associated with a history of asbestos exposure. The tumor is characterized by a strong local invasiveness and a poor response to any single modality therapy. Therefore clinical outcome of patients with MPM is poor and median survival time of untreated patients with MPM is 7 months from initial diagnosis. The Wilms Tumor Protein 1 (WT1) is a transcription factor which is highly expressed by MPM and is involved in cellular development and survival. We evaluated the role of WT1 in two human MPM cell lines (MSTO and H2052) expressing high levels of WT1. We performed a knockdown of WT1 using siRNA. Knockdown of WT1 was confirmed by Westernblotting. After knockdown of WT1 we investigated the effect on proliferation, chemoresistance, chemotaxis and migration. We could demonstrate that knockdown of WT1 suppresses chemoresistance in both cell lines compared with control (scrambled siRNA). Additionally, WT1 knockdown reduces proliferation, chemotaxis and invasiveness of mesothelioma cell lines. WT1 reduces malignancy of malignant mesothelioma cell lines and might be a new molecular target in mesothelioma therapy. Further investigations are needed to discover the mechanisms of chemoresistance depending on WT1.
Collapse
Affiliation(s)
- Till Plönes
- Department of Thoracic Surgery and Thoracic Endoscopy, Ruhrlandklinik, West German Lung Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany.
- Department of Pneumology, Center for Medicine, Medical Center University of Freiburg, Freiburg, Germany.
| | - Mitja Fischer
- Department of Pneumology, Center for Medicine, Medical Center University of Freiburg, Freiburg, Germany
| | - Kerstin Höhne
- Department of Pneumology, Center for Medicine, Medical Center University of Freiburg, Freiburg, Germany
| | - Hiromi Sato
- Graduate School of Pharmaceutical Sciences, Department of Clinical Pharmacology & Pharmacometrics, Chiba University, 1-8-1 Inohana, Chuou-ku, Chiba, 260-8675, Japan
| | - Joachim Müller-Quernheim
- Department of Pneumology, Center for Medicine, Medical Center University of Freiburg, Freiburg, Germany
| | - Gernot Zissel
- Department of Pneumology, Center for Medicine, Medical Center University of Freiburg, Freiburg, Germany
| |
Collapse
|
9
|
Saavedra-Alonso S, Zapata-Benavides P, Chavez-Escamilla AK, Manilla-Muñoz E, Zamora-Avila DE, Franco-Molina MA, Rodriguez-Padilla C. WT1 shRNA delivery using transferrin-conjugated PEG liposomes in an in vivo model of melanoma. Exp Ther Med 2016; 12:3778-3784. [PMID: 28105110 DOI: 10.3892/etm.2016.3851] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 05/31/2016] [Indexed: 12/12/2022] Open
Abstract
The global incidence of melanoma is increasing. Mortality from melanoma is influenced primarily by metastasis in advanced stages of the disease. Current treatments are largely ineffective; thus, novel gene delivery approaches that target tumor-specific markers may be useful for the treatment of melanoma. Systemic administration of encapsulated RNA-interference plasmids targeted against tumor cells is a potential alternative therapy for cancer. Formulations of transferrin (Tf)-conjugated polyethylene glycol (PEG) liposomes loaded with short hairpin RNA (shRNA) against WT1 (Lip + RNAi + Tf), PEG liposomes loaded with shRNA against WT1 (Lip + RNAi), Tf-conjugated PEG liposomes loaded with pEGFP-N3 (Lip + GFP + Tf) and saline solution as negative control (untreated) were administered systemically to C57BL/6 mice implanted subcutaneously with a melanoma cell line. Tumor volume, body weight, tumor weight, survival and relative expression of WT1 were evaluated. No significant differences in net body weight were identified between groups. The tumor volume decreased from 7,871 mm3 (SD±2,087) in the untreated group to 5,981 mm3 (SD±2,099) in the Lip + RNAi + Tf group. The tumor weight was reduced, from 8.8 g (SD±0.30) in the untreated group to 5.5 g (SD±0.87) in the Lip + RNAi + Tf group. An increase of 37% in survival was also observed in the group treated with Lip + RNAi + Tf in comparison to the untreated group. Tumors treated with Lip + RNAi + Tf also showed a decrease in the mean relative expression of WT1 of 0.21 (SD±0.28) folds compared with 1.8 (SD±2.49) folds in untreated group, 1.34 (SD±0.43) folds in Lip + RNAi group and of 1.89 (SD±0.69) folds in Lip + GFP + Tf group. Systemic administration of transferrin-conjugated PEG liposomes loaded with shRNA against WT1 reduced WT1 expression and tumor size and increased survival.
Collapse
Affiliation(s)
- Santiago Saavedra-Alonso
- Department of Microbiology and Immunology, Faculty of Biological Sciences, Autonomous University of Nuevo León (UANL), San Nicolás de los Garza, Nuevo León, México
| | - Pablo Zapata-Benavides
- Department of Microbiology and Immunology, Faculty of Biological Sciences, Autonomous University of Nuevo León (UANL), San Nicolás de los Garza, Nuevo León, México
| | - Ana Karina Chavez-Escamilla
- Department of Microbiology and Immunology, Faculty of Biological Sciences, Autonomous University of Nuevo León (UANL), San Nicolás de los Garza, Nuevo León, México
| | - Edgar Manilla-Muñoz
- Department of Microbiology and Immunology, Faculty of Biological Sciences, Autonomous University of Nuevo León (UANL), San Nicolás de los Garza, Nuevo León, México
| | - Diana Elisa Zamora-Avila
- Department of Genetics, Veterinary Medicine Faculty, Autonomous University of Nuevo León (UANL), Escobedo, Nuevo León, México
| | - Moisés Armides Franco-Molina
- Department of Microbiology and Immunology, Faculty of Biological Sciences, Autonomous University of Nuevo León (UANL), San Nicolás de los Garza, Nuevo León, México
| | - Cristina Rodriguez-Padilla
- Department of Microbiology and Immunology, Faculty of Biological Sciences, Autonomous University of Nuevo León (UANL), San Nicolás de los Garza, Nuevo León, México
| |
Collapse
|
10
|
Downregulation of the WT1 gene expression via TMPyP4 stabilization of promoter G-quadruplexes in leukemia cells. Tumour Biol 2016; 37:9967-77. [PMID: 26815508 DOI: 10.1007/s13277-016-4881-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 01/15/2016] [Indexed: 10/22/2022] Open
Abstract
The WT1 gene is an important oncogene, and its overexpression is considered as an effective target for anticancer therapy. Regulation of its gene transcription is one way for WT1-targeting drug design. Recently, in silico analysis of some oncogene promoters like WT1 showed some guanine-rich regions with the ability to form G-quadruplex structures. Ligands like 5,10,15,20-tetra(N-methyl-4-pyridyl)-porphine (TMPyP4) have predominant effect on G-quadruplex stabilization. The aim of this study was to understand the effect of TMPyP4 on WT1 gene transcription via stabilization of promoter G-quadruplexes. We examined the formation of new G-quadruplex motifs in WT1 promoter in the presence of TMPyP4. In order to understand the nature of its interaction with WT1 promoter quadruplexes, differential pulse voltammetry (DPV), circular dichroism (CD), polyacrylamide gel electrophoresis, electrophoretic mobility shift assay (EMSA), polymerase chain reaction (PCR) stop assays, and quantitative RT-PCR were performed. According to the results, the WT1 promoter can form stable intramolecular parallel G-quadruplexes. In addition, after 48 and 96 h of incubation, 100 μM TMPyP4 reduced the WT1 transcription to 9 and 0.4 %, respectively, compare to control. We report that ligand-mediated stabilization of G-quadruplexes within the WT1 promoter can silence WT1 expression. This study might offer the basis for the reasonable design and improvement of new porphyrin derivatives as effective anti-leukemia agents for cancer therapy.
Collapse
|
11
|
The antioxidant paradox: what are antioxidants and how should they be used in a therapeutic context for cancer. Future Med Chem 2015; 6:1413-22. [PMID: 25329197 DOI: 10.4155/fmc.14.86] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
So-called antioxidants have yet to make a clinical impact on the treatment of human cancer. The reasons for this failure are several. First, many agents that are called antioxidants are truly antioxidants at a given dose, but this dose may not have been given in clinical trials. Second, many agents are not antioxidants at all. Third, not all tumors use reactive oxygen as a signaling mechanism. Finally, reactive oxygen inhibition is often insufficient to kill or regress a tumor cell by itself, but requires sequential introduction of a therapeutic agent for maximal effect. We hope to provide a framework for the logical use of these agents in cancer.
Collapse
|
12
|
Parenti R, Cardile V, Graziano ACE, Parenti C, Venuti A, Bertuccio MP, Furno DL, Magro G. Wilms' tumor gene 1 (WT1) silencing inhibits proliferation of malignant peripheral nerve sheath tumor sNF96.2 cell line. PLoS One 2014; 9:e114333. [PMID: 25474318 PMCID: PMC4256418 DOI: 10.1371/journal.pone.0114333] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 11/06/2014] [Indexed: 12/25/2022] Open
Abstract
Wilms' tumor gene 1 (WT1) plays complex roles in tumorigenesis, acting as tumor suppressor gene or an oncogene depending on the cellular context. WT1 expression has been variably reported in both benign and malignant peripheral nerve sheath tumors (MPNSTs) by means of immunohistochemistry. The aim of the present study was to characterize its potential pathogenetic role in these relatively uncommon malignant tumors. Firstly, immunohistochemical analyses in MPNST sNF96.2 cell line showed strong WT1 staining in nuclear and perinuclear areas of neoplastic cells. Thus, we investigated the effects of silencing WT1 by RNA interference. Through Western Blot analysis and proliferation assay we found that WT1 knockdown leads to the reduction of cell growth in a time- and dose-dependent manner. siWT1 inhibited proliferation of sNF96.2 cell lines likely by influencing cell cycle progression through a decrease in the protein levels of cyclin D1 and inhibition of Akt phosphorylation compared to the control cells. These results indicate that WT1 knockdown attenuates the biological behavior of MPNST cells by decreasing Akt activity, demonstrating that WT1 is involved in the development and progression of MPNSTs. Thus, WT1 is suggested to serve as a potential therapeutic target for MPNSTs.
Collapse
Affiliation(s)
- Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, 95125 Catania, Italy
| | - Venera Cardile
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, 95125 Catania, Italy
| | | | - Carmela Parenti
- Department of Drug Sciences, Pharmacology and Toxicology Section, University of Catania, 95125 Catania, Italy
| | - Assunta Venuti
- Business Unit Oncology, Nerviano Medical Sciences S.r.l., 20014 Nerviano Milano, Italy
| | - Maria Paola Bertuccio
- Business Unit Oncology, Nerviano Medical Sciences S.r.l., 20014 Nerviano Milano, Italy
| | - Debora Lo Furno
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, 95125 Catania, Italy
| | - Gaetano Magro
- Department G.F. Ingrassia, Azienda Ospedaliero-Universitaria “Policlinico-Vittorio Emanuele” Anatomic Pathology, University of Catania, 95125 Catania, Italy
| |
Collapse
|
13
|
Iranparast S, Assarehzadegan MA, Heike Y, Hossienzadeh M, Khodadadi A. Wilms' Tumor Gene (WT1) Expression Correlates with Vascular Epithelial Growth Factor (VEGF) in Newly Acute Leukemia Patients Undergoing Chemotherapy. Asian Pac J Cancer Prev 2014; 15:9217-23. [DOI: 10.7314/apjcp.2014.15.21.9217] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
14
|
Massaoka MH, Matsuo AL, Figueiredo CR, Girola N, Faria CF, Azevedo RA, Travassos LR. A novel cell-penetrating peptide derived from WT1 enhances p53 activity, induces cell senescence and displays antimelanoma activity in xeno- and syngeneic systems. FEBS Open Bio 2014; 4:153-61. [PMID: 24490140 PMCID: PMC3907745 DOI: 10.1016/j.fob.2014.01.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 01/07/2014] [Accepted: 01/14/2014] [Indexed: 01/24/2023] Open
Abstract
The Wilms tumor protein 1 (WT1) transcription factor has been associated in malignant melanoma with cell survival and metastasis, thus emerging as a candidate for targeted therapy. A lysine-arginine rich peptide, WT1-pTj, derived from the ZF domain of WT1 was evaluated as an antitumor agent against A2058 human melanoma cells and B16F10-Nex2 syngeneic murine melanoma. Peptide WT1-pTj quickly penetrated human melanoma cells and induced senescence, recognized by increased SA-β-galactosidase activity, enhanced transcriptional activity of p53, and induction of the cell cycle inhibitors p21 and p27. Moreover, the peptide bound to p53 and competed with WT1 protein for binding to p53. WT1-pTj treatment led to sustained cell growth suppression, abrogation of clonogenicity and G2/M cell cycle arrest. Notably, in vivo studies showed that WT1-pTj inhibited both the metastases and subcutaneous growth of murine melanoma in syngeneic mice, and prolonged the survival of nude mice challenged with human melanoma cells. The 27-amino acid cell-penetrating WT1-derived peptide, depends on C(3) and H(16) for effective antimelanoma activity, inhibits proliferation of WT1-expressing human tumor cell lines, and may have an effective role in the treatment of WT1-expressing malignancies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Luiz R. Travassos
- Experimental Oncology Unit (UNONEX), Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, SP 04023-062, Brazil
| |
Collapse
|
15
|
Zapata-Benavides P, Manilla-Muñoz E, Zamora-Avila DE, Saavedra-Alonso S, Franco-Molina MA, Trejo-Avila LM, Davalos-Aranda G, Rodríguez-Padilla C. WT1 silencing by RNAi synergizes with chemotherapeutic agents and induces chemosensitization to doxorubicin and cisplatin in B16F10 murine melanoma cells. Oncol Lett 2012; 3:751-755. [PMID: 22740987 DOI: 10.3892/ol.2012.578] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Accepted: 11/17/2011] [Indexed: 12/20/2022] Open
Abstract
The Wilm's tumor gene (WT1), encoding a transcription factor that modulates the expression of certain genes that are involved in proliferation and apoptosis, is overexpressed in numerous solid tumors. WT1 is important for cell proliferation and in the diagnosis of melanoma. The objectives of this study were to investigate whether WT1 silencing is capable of synergizing with chemotherapeutic agents and whether this silencing is capable of sensitizing cancer cells to doxorubicin and cisplatin in the B16F10 murine melanoma cell line. In the present study, B16F10 cells were simultaneously treated with median lethal doses (LD50s) of WT1-1 or WT1-2 small hairpin RNAs (shRNAs) and chemotherapeutic agents. A total of 24 h post-transfection, a [3-(4,5-dimethylthiazol-2yl)-2,5- diphenyl tetrazolium bromide assay] MTT assay was performed. To determine whether shRNA interference (shRNAi) is capable of sensitizing B16F10 cells to chemotherapeutic agents, cells were transfected with an LD50 of each of the recombinant plasmids, treated with varying concentrations of doxorubicin or cisplatin 24 h post-transfection, and analyzed 48 h later for inhibition of cell proliferation using the MTT assay. We observed that WT1-RNAi and the two chemotherapeutic agents acted synergistically to inhibit B16F10 cell proliferation. The greatest inhibition of cell proliferation was observed with the WT1-2/cisplatin (91%) and WT1-1/cisplatin combinations (85%). WT1 silencing using shRNAi induced the chemosensitization of cells to doxorubicin and cisplatin, with the greatest inhibition (85%) of cell proliferation being observed in the cells treated with the WT1-2/cisplatin 6 ng/µl combination. Our results provide direct evidence that WT1 gene silencing has a synergistic effect with chemotherapeutic drugs and sensitizes B16F10 melanoma cells to doxorubicin and cisplatin. This suggests that these combination strategies are potentially utilized in melanoma therapy.
Collapse
Affiliation(s)
- Pablo Zapata-Benavides
- Laboratorio de Inmunología y Virología, Departamento de Microbiología e Inmunología, Facultad de Ciencias Biológicas de la Universidad Autónoma de Nuevo León, San Nicolás de los Garza, N.L. México
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Zamora-Avila DE, Zapata-Benavides P, Franco-Molina MA, Saavedra-Alonso S, Trejo-Avila LM, Reséndez-Pérez D, Méndez-Vázquez JL, Isaias-Badillo J, Rodríguez-Padilla C. WT1 gene silencing by aerosol delivery of PEI–RNAi complexes inhibits B16-F10 lung metastases growth. Cancer Gene Ther 2009; 16:892-9. [DOI: 10.1038/cgt.2009.35] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
17
|
Abstract
In melanoma, at least four major signaling abnormalities have been described. They include beta-catenin deregulation (mutation/mislocalization), p16 loss, MAP kinase activation, and Akt activation. In this review, we discuss the role of the fourth pathway, known as the reactive oxygen driven tumor. The role of reactive oxygen in tumorigenesis is likely to relate to virtually all forms of cancer, and lends itself to specific therapies. These include blockade of reactive oxygen, resulting in decreased activation of NF-kappaB, which should sensitize tumors to chemotherapy and radiation. The phenotype of the reactive oxygen driven tumor can be monitored using available markers already in use in most hospital laboratories.
Collapse
Affiliation(s)
- Levi Fried
- Department of Dermatology, Emory University School of Medicine, Atlanta, GA, USA
| | | |
Collapse
|