36 results on '"Einav Shoshan"'
Search Results
2. A-to-I miR-378a-3p editing can prevent melanoma progression via regulation of PARVA expression
- Author
-
Guermarie Velazquez-Torres, Einav Shoshan, Cristina Ivan, Li Huang, Enrique Fuentes-Mattei, Harrison Paret, Sun Jin Kim, Cristian Rodriguez-Aguayo, Victoria Xie, Denise Brooks, Steven J. M. Jones, A. Gordon Robertson, George Calin, Gabriel Lopez-Berenstein, Anil Sood, and Menashe Bar-Eli
- Subjects
Science - Abstract
In melanoma, reduced ADAR1 impairs A-to-I microRNA editing. Here, the authors show that miR-378a-3p undergoes this editing in non-metastatic cells and the edited form of miR-378a-3p binds to the PARVA oncogene, inhibiting its expression and preventing melanoma progression and metastasis.
- Published
- 2018
- Full Text
- View/download PDF
3. MIF inhibition as a strategy for overcoming resistance to immune checkpoint blockade therapy in melanoma
- Author
-
Ricardo A. de Azevedo, Einav Shoshan, Shanzhi Whang, Gal Markel, Ashvin R. Jaiswal, Arthur Liu, Michael A. Curran, Luiz R. Travassos, and Menashe Bar-Eli
- Subjects
melanoma ,macrophage migratory inhibition factor ,tumor microenvironment ,immune checkpoint therapy ,combined modality therapy ,Immunologic diseases. Allergy ,RC581-607 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Immune checkpoint blockade (ICB) has demonstrated an impressive outcome in patients with metastatic melanoma, yet, durable complete response; even with Ipilimumab/Nivolumab combo are under 30%. Primary and acquired resistance in response to ICB is commonly due to a tumor immune escape mechanism dictated by the tumor microenvironment (TME). Macrophage Migratory Inhibition Factor (MIF) has emerged as an immunosuppressive factor secreted in the TME. We have previously demonstrated that blockade of the MIF-CD74 signaling on macrophages and dendritic cells restored the anti-tumor immune response against melanoma. Here, we report that inhibition of the MIF-CD74 axis combined with ipilimumab could render resistant melanoma to better respond to anti-CTLA-4 treatment. We provide evidence that blocking the MIF-CD74 signaling potentiates CD8+ T-cells infiltration and drives pro-inflammatory M1 conversion of macrophages in the TME. Furthermore, MIF inhibition resulted in reprogramming the metabolic pathway by reducing lactate production, HIF-1α and PD-L1 expression in the resistant melanoma cells. Melanoma patient data extracted from the TCGA database supports the hypothesis that high MIF expression strongly correlates with poor response to ICB therapy. Our findings provide a rationale for combining anti-CTLA-4 with MIF inhibitors as a potential strategy to overcome resistance to ICB therapy in melanoma, turning a “cold” tumor into a “hot” one mediated by the activation of innate immunity and reprogramming of tumor metabolism and reduced PD-L1 expression in melanoma cells.
- Published
- 2020
- Full Text
- View/download PDF
4. Data from Gene Body Methylation of the Lymphocyte-Specific Gene CARD11 Results in Its Overexpression and Regulates Cancer mTOR Signaling
- Author
-
Anil K. Sood, Keith A. Baggerly, Jing Wang, Menashe Bar-Eli, Eric Jonasch, Sherry Y. Wu, Einav Shoshan, Cristina Ivan, Wencai Ma, Emine Bayraktar, Lingegowda S. Mangala, Yunfei Wen, Hui Yao, Santosh K. Dasari, and Michael H. McGuire
- Abstract
Investigations into the function of nonpromoter DNA methylation have yielded new insights into epigenetic regulation of gene expression. Previous studies have highlighted the importance of distinguishing between DNA methylation in discrete functional regions; however, integrated nonpromoter DNA methylation and gene expression analyses across a wide number of tumor types and corresponding normal tissues have not been performed. Through integrated analysis of gene expression and DNA methylation profiles, we examined 32 tumor types and identified 57 tumor suppressors and oncogenes out of 260 genes exhibiting a correlation of > 0.5 between gene body methylation and gene expression in at least one tumor type. The lymphocyte-specific gene CARD11 exhibits robust association between gene body methylation and expression across 19 of 32 tumor types examined. It is significantly overexpressed in kidney renal cell carcinoma (KIRC) and lung adenocarcinoma (LUAD) tumor tissues in comparison with respective control samples; and is significantly associated with lower overall survival in KIRC. Contrary to its canonical function in lymphocyte NFκB activation, CARD11 activates the mTOR pathway in KIRC and LUAD, resulting in suppressed autophagy. Furthermore, demethylation of a CpG island within the gene body of CARD11 decreases gene expression. Collectively, our study highlights how DNA methylation outside the promoter region can impact tumor progression.Implications:Our study describes a novel regulatory role of gene body DNA methylation–dependent CARD11 expression on mTOR signaling and its impact on tumor progression.
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Table from Gene Body Methylation of the Lymphocyte-Specific Gene CARD11 Results in Its Overexpression and Regulates Cancer mTOR Signaling
- Author
-
Anil K. Sood, Keith A. Baggerly, Jing Wang, Menashe Bar-Eli, Eric Jonasch, Sherry Y. Wu, Einav Shoshan, Cristina Ivan, Wencai Ma, Emine Bayraktar, Lingegowda S. Mangala, Yunfei Wen, Hui Yao, Santosh K. Dasari, and Michael H. McGuire
- Abstract
Supplementary Table from Gene Body Methylation of the Lymphocyte-Specific Gene CARD11 Results in Its Overexpression and Regulates Cancer mTOR Signaling
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Figure 3 from Galectin-3 Contributes to Melanoma Growth and Metastasis via Regulation of NFAT1 and Autotaxin
- Author
-
Menashe Bar-Eli, Avraham Raz, Renduo Song, Aaron K. Mobley, Einav Shoshan, David J. McConkey, Woonyoung Choi, Li Huang, Andrey S. Dobroff, Takafumi Kamiya, Maya Zigler, and Russell R. Braeuer
- Abstract
PDF file, 1717K, qRT-PCR of autotaxin and nfat1 mRNA after silencing nfat1 in A375SM melanoma cells
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Figure 1 from Galectin-3 Contributes to Melanoma Growth and Metastasis via Regulation of NFAT1 and Autotaxin
- Author
-
Menashe Bar-Eli, Avraham Raz, Renduo Song, Aaron K. Mobley, Einav Shoshan, David J. McConkey, Woonyoung Choi, Li Huang, Andrey S. Dobroff, Takafumi Kamiya, Maya Zigler, and Russell R. Braeuer
- Abstract
PDF file, 1628K, Invasion, migration, and soft agar assay of melanoma cells after silencing galectin-3
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Materials and Methods and Legends from Galectin-3 Contributes to Melanoma Growth and Metastasis via Regulation of NFAT1 and Autotaxin
- Author
-
Menashe Bar-Eli, Avraham Raz, Renduo Song, Aaron K. Mobley, Einav Shoshan, David J. McConkey, Woonyoung Choi, Li Huang, Andrey S. Dobroff, Takafumi Kamiya, Maya Zigler, and Russell R. Braeuer
- Abstract
PDF file, 84K
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Table 1 from Galectin-3 Contributes to Melanoma Growth and Metastasis via Regulation of NFAT1 and Autotaxin
- Author
-
Menashe Bar-Eli, Avraham Raz, Renduo Song, Aaron K. Mobley, Einav Shoshan, David J. McConkey, Woonyoung Choi, Li Huang, Andrey S. Dobroff, Takafumi Kamiya, Maya Zigler, and Russell R. Braeuer
- Abstract
PDF file, 61K, Oligos used for experiments
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Figure 2 from NFAT1 Directly Regulates IL8 and MMP3 to Promote Melanoma Tumor Growth and Metastasis
- Author
-
Menashe Bar-Eli, Gabriel J. Villares, Victor Prieto, Cristina Ivan, Nitin Chakravarti, Guermarie Velazquez-Torres, Mayra E. Vasquez, Li Huang, Aaron K. Mobley, Takafumi Kamiya, Russell R. Braeuer, and Einav Shoshan
- Abstract
Relative mRNA expression of NFAT1, IL-8, and MMP-3
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Figure 4 from NFAT1 Directly Regulates IL8 and MMP3 to Promote Melanoma Tumor Growth and Metastasis
- Author
-
Menashe Bar-Eli, Gabriel J. Villares, Victor Prieto, Cristina Ivan, Nitin Chakravarti, Guermarie Velazquez-Torres, Mayra E. Vasquez, Li Huang, Aaron K. Mobley, Takafumi Kamiya, Russell R. Braeuer, and Einav Shoshan
- Abstract
Schematic presentation of the IL-8 and MMP-3 promoters for NFAT1 binding sites
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Figure 4 from Galectin-3 Contributes to Melanoma Growth and Metastasis via Regulation of NFAT1 and Autotaxin
- Author
-
Menashe Bar-Eli, Avraham Raz, Renduo Song, Aaron K. Mobley, Einav Shoshan, David J. McConkey, Woonyoung Choi, Li Huang, Andrey S. Dobroff, Takafumi Kamiya, Maya Zigler, and Russell R. Braeuer
- Abstract
PDF file, 227K, Immunohistochemistry of A375SM tumors after silencing galectin-3
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Figure 6 from Galectin-3 Contributes to Melanoma Growth and Metastasis via Regulation of NFAT1 and Autotaxin
- Author
-
Menashe Bar-Eli, Avraham Raz, Renduo Song, Aaron K. Mobley, Einav Shoshan, David J. McConkey, Woonyoung Choi, Li Huang, Andrey S. Dobroff, Takafumi Kamiya, Maya Zigler, and Russell R. Braeuer
- Abstract
PDF file, 1706K, Expression status of LPA receptors in SB-2, WM2664, and A375SM melanoma cells.
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Figure 5 from Galectin-3 Contributes to Melanoma Growth and Metastasis via Regulation of NFAT1 and Autotaxin
- Author
-
Menashe Bar-Eli, Avraham Raz, Renduo Song, Aaron K. Mobley, Einav Shoshan, David J. McConkey, Woonyoung Choi, Li Huang, Andrey S. Dobroff, Takafumi Kamiya, Maya Zigler, and Russell R. Braeuer
- Abstract
PDF file, 1740K, Microvascular density in A375SM NT and Gal-3 shRNA tumors
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Figure 1 from NFAT1 Directly Regulates IL8 and MMP3 to Promote Melanoma Tumor Growth and Metastasis
- Author
-
Menashe Bar-Eli, Gabriel J. Villares, Victor Prieto, Cristina Ivan, Nitin Chakravarti, Guermarie Velazquez-Torres, Mayra E. Vasquez, Li Huang, Aaron K. Mobley, Takafumi Kamiya, Russell R. Braeuer, and Einav Shoshan
- Abstract
Effect of NFAT1 silencing or overexpression on melanoma cell viability
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Table 1 from NFAT1 Directly Regulates IL8 and MMP3 to Promote Melanoma Tumor Growth and Metastasis
- Author
-
Menashe Bar-Eli, Gabriel J. Villares, Victor Prieto, Cristina Ivan, Nitin Chakravarti, Guermarie Velazquez-Torres, Mayra E. Vasquez, Li Huang, Aaron K. Mobley, Takafumi Kamiya, Russell R. Braeuer, and Einav Shoshan
- Abstract
Top potential genes downregulated after silencing NFAT1
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary Figure 3 from NFAT1 Directly Regulates IL8 and MMP3 to Promote Melanoma Tumor Growth and Metastasis
- Author
-
Menashe Bar-Eli, Gabriel J. Villares, Victor Prieto, Cristina Ivan, Nitin Chakravarti, Guermarie Velazquez-Torres, Mayra E. Vasquez, Li Huang, Aaron K. Mobley, Takafumi Kamiya, Russell R. Braeuer, and Einav Shoshan
- Abstract
Immunohistochemistry of tumor samples after silencing or overexpressing NFAT1 in melanoma cells
- Published
- 2023
- Full Text
- View/download PDF
18. Data from NFAT1 Directly Regulates IL8 and MMP3 to Promote Melanoma Tumor Growth and Metastasis
- Author
-
Menashe Bar-Eli, Gabriel J. Villares, Victor Prieto, Cristina Ivan, Nitin Chakravarti, Guermarie Velazquez-Torres, Mayra E. Vasquez, Li Huang, Aaron K. Mobley, Takafumi Kamiya, Russell R. Braeuer, and Einav Shoshan
- Abstract
Nuclear factor of activated T cell (NFAT1, NFATC2) is a transcription factor that binds and positively regulates IL2 expression during T-cell activation. NFAT1 has important roles in both innate and adaptive immune responses, but its involvement in cancer is not completely understood. We previously demonstrated that NFAT1 contributes to melanoma growth and metastasis by regulating the autotaxin gene (Enpp2). Here, we report a strong correlation between NFAT1 expression and metastatic potential in melanoma cell lines and tumor specimens. To elucidate the mechanisms underlying NFAT1 overexpression during melanoma progression, we conducted a microarray on a highly metastatic melanoma cell line in which NFAT1 expression was stably silenced. We identified and validated two downstream targets of NFAT1, IL8, and MMP3. Accordingly, NFAT1 depletion in metastatic melanoma cell lines was associated with reduced IL8 and MMP3 expression, whereas NFAT1 overexpression in a weakly metastatic cell line induced expression of these targets. Restoration of NFAT1 expression recovered IL8 and MMP3 expression levels back to baseline, indicating that both are direct targets of NFAT1. Moreover, in vivo studies demonstrated that NFAT1 and MMP3 promoted melanoma tumor growth and lung metastasis. Collectively, our findings assign a new role for NFAT1 in melanoma progression, underscoring the multifaceted functions that immunomodulatory factors may acquire in an unpredictable tumor microenvironment. Cancer Res; 76(11); 3145–55. ©2016 AACR.
- Published
- 2023
- Full Text
- View/download PDF
19. Supplementary Figure Legends from NFAT1 Directly Regulates IL8 and MMP3 to Promote Melanoma Tumor Growth and Metastasis
- Author
-
Menashe Bar-Eli, Gabriel J. Villares, Victor Prieto, Cristina Ivan, Nitin Chakravarti, Guermarie Velazquez-Torres, Mayra E. Vasquez, Li Huang, Aaron K. Mobley, Takafumi Kamiya, Russell R. Braeuer, and Einav Shoshan
- Abstract
Supplementary Figure Legends
- Published
- 2023
- Full Text
- View/download PDF
20. Supplementary Figure 2 from Galectin-3 Contributes to Melanoma Growth and Metastasis via Regulation of NFAT1 and Autotaxin
- Author
-
Menashe Bar-Eli, Avraham Raz, Renduo Song, Aaron K. Mobley, Einav Shoshan, David J. McConkey, Woonyoung Choi, Li Huang, Andrey S. Dobroff, Takafumi Kamiya, Maya Zigler, and Russell R. Braeuer
- Abstract
PDF file, 1483K, Transcriptional analysis of autotaxin after silencing galectin-3 by nuclear run-on and dual luciferase assays
- Published
- 2023
- Full Text
- View/download PDF
21. Gene Body Methylation of the Lymphocyte-Specific GeneCARD11Results in Its Overexpression and Regulates Cancer mTOR Signaling
- Author
-
Menashe Bar-Eli, Michael McGuire, Lingegowda S. Mangala, Anil K. Sood, Jing Wang, Yunfei Wen, Santosh K. Dasari, Wencai Ma, Einav Shoshan, Emine Bayraktar, Eric Jonasch, Keith A. Baggerly, Cristina Ivan, Hui Yao, and Sherry Y. Wu
- Subjects
Cancer Research ,Oncology ,CpG site ,Tumor progression ,DNA methylation ,Gene expression ,Cancer research ,Promoter ,Methylation ,Epigenetics ,Biology ,Molecular Biology ,Gene - Abstract
Investigations into the function of nonpromoter DNA methylation have yielded new insights into epigenetic regulation of gene expression. Previous studies have highlighted the importance of distinguishing between DNA methylation in discrete functional regions; however, integrated nonpromoter DNA methylation and gene expression analyses across a wide number of tumor types and corresponding normal tissues have not been performed. Through integrated analysis of gene expression and DNA methylation profiles, we examined 32 tumor types and identified 57 tumor suppressors and oncogenes out of 260 genes exhibiting a correlation of > 0.5 between gene body methylation and gene expression in at least one tumor type. The lymphocyte-specific gene CARD11 exhibits robust association between gene body methylation and expression across 19 of 32 tumor types examined. It is significantly overexpressed in kidney renal cell carcinoma (KIRC) and lung adenocarcinoma (LUAD) tumor tissues in comparison with respective control samples; and is significantly associated with lower overall survival in KIRC. Contrary to its canonical function in lymphocyte NFκB activation, CARD11 activates the mTOR pathway in KIRC and LUAD, resulting in suppressed autophagy. Furthermore, demethylation of a CpG island within the gene body of CARD11 decreases gene expression. Collectively, our study highlights how DNA methylation outside the promoter region can impact tumor progression.Implications:Our study describes a novel regulatory role of gene body DNA methylation–dependent CARD11 expression on mTOR signaling and its impact on tumor progression.
- Published
- 2021
- Full Text
- View/download PDF
22. Lung Regeneration by Transplantation of Allogeneic Lung Progenitors Using a Safer Conditioning Regimen and Clinical-grade Reagents
- Author
-
Irit Milman Krentsis, Ran Orgad, Yangxi Zheng, Esther Bachar Lustig, Chava Rosen, Elias Shezen, Sandeep Yadav, Bar Nathansohn Levi, Miri Assayag, Neville Berkman, Harry Karmouty Quintana, Einav Shoshan, Christa Blagdon, and Yair Reisner
- Subjects
Mice ,Transplantation Chimera ,Transplantation Conditioning ,Hematopoietic Stem Cell Transplantation ,Animals ,Humans ,Indicators and Reagents ,Cell Biology ,General Medicine ,respiratory system ,Cyclophosphamide ,Lung ,Developmental Biology - Abstract
Over the last decades, several studies demonstrated the possibility of lung regeneration through transplantation of various lung progenitor populations. Recently, we showed in mice that fetal or adult lung progenitors could potentially provide donor cells for transplantation, provided that the lung stem cell niche in the recipient is vacated of endogenous lung progenitors by adequate conditioning. Accordingly, marked lung regeneration could be attained following i.v. infusion of a single cell suspension of lung cells into recipient mice conditioned with naphthalene (NA) and 6Gy total body irradiation (TBI). As clinical translation of this approach requires the use of allogenic donors, we more recently developed a novel transplantation modality based on co-infusion of hematopoietic and lung progenitors from the same donor. Thus, by virtue of hematopoietic chimerism, which leads to immune tolerance toward donor antigens, the lung progenitors can be successfully engrafted without any need for post-transplant immune suppression. In the present study, we demonstrate that it is possible to replace NA in the conditioning regimen with Cyclophosphamide (CY), approved for the treatment of many diseases and that a lower dose of 2 GY TBI can successfully enable engraftment of donor-derived hematopoietic and lung progenitors when CY is administered in 2 doses after the stem cell infusion. Taken together, our results suggest a feasible and relatively safe protocol that could potentially be translated to clinical transplantation of lung progenitors across major MHC barriers in patients with terminal lung diseases.
- Published
- 2021
23. Gene Body Methylation of the Lymphocyte-Specific Gene
- Author
-
Michael H, McGuire, Santosh K, Dasari, Hui, Yao, Yunfei, Wen, Lingegowda S, Mangala, Emine, Bayraktar, Wencai, Ma, Cristina, Ivan, Einav, Shoshan, Sherry Y, Wu, Eric, Jonasch, Menashe, Bar-Eli, Jing, Wang, Keith A, Baggerly, and Anil K, Sood
- Subjects
CARD Signaling Adaptor Proteins ,Mice ,TOR Serine-Threonine Kinases ,Animals ,Humans ,Mice, Nude ,Female ,Lymphocytes ,DNA Methylation ,Prognosis ,Transfection ,Article ,Signal Transduction - Abstract
Investigations into the function of non-promoter DNA methylation have yielded new insights into epigenetic regulation of gene expression. Previous studies have highlighted the importance of distinguishing between DNA methylation in discrete functional regions; however, integrated non-promoter DNA methylation and gene expression analyses across a wide number of tumor types and corresponding normal tissues have not been performed. Through integrated analysis of gene expression and DNA methylation profiles, we examined 32 tumor types and identified 57 tumor suppressors and oncogenes out of 260 genes exhibiting a correlation of > 0.5 between gene body methylation and gene expression in at least 1 tumor type. The lymphocyte-specific gene CARD11 exhibits robust association between gene body methylation and expression across 19 of 32 tumor types examined. It is significantly overexpressed in kidney renal cell carcinoma (KIRC) and lung adenocarcinoma (LUAD) tumor tissues in comparison to respective control samples; and is significantly associated with lower overall survival in KIRC. Contrary to its canonical function in lymphocyte NF-kB activation, CARD11 activates the mTOR pathway in KIRC and LUAD, resulting in suppressed autophagy. Furthermore, demethylation of a CpG island within the gene body of CARD11 decreases gene expression. Collectively, our study highlights how DNA methylation outside the promoter region can impact tumor progression.
- Published
- 2020
24. MIF inhibition as a strategy for overcoming resistance to immune checkpoint blockade therapy in melanoma
- Author
-
Michael A. Curran, Shanzhi Whang, Ricardo A. de Azevedo, Menashe Bar-Eli, Luiz R. Travassos, Gal Markel, Einav Shoshan, Arthur Liu, and Ashvin R. Jaiswal
- Subjects
0301 basic medicine ,combined modality therapy ,Immunology ,chemical and pharmacologic phenomena ,Ipilimumab ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Tumor Microenvironment ,Humans ,Immunology and Allergy ,Medicine ,Immune Checkpoint Inhibitors ,Macrophage Migration-Inhibitory Factors ,Melanoma ,Macrophage Migratory Inhibition Factor ,RC254-282 ,Original Research ,Tumor microenvironment ,Innate immune system ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC581-607 ,medicine.disease ,Immune checkpoint ,immune checkpoint therapy ,Blockade ,Intramolecular Oxidoreductases ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Immunologic diseases. Allergy ,Nivolumab ,business ,Research Article ,medicine.drug - Abstract
Immune checkpoint blockade (ICB) has demonstrated an impressive outcome in patients with metastatic melanoma, yet, durable complete response; even with Ipilimumab/Nivolumab combo are under 30%. Primary and acquired resistance in response to ICB is commonly due to a tumor immune escape mechanism dictated by the tumor microenvironment (TME). Macrophage Migratory Inhibition Factor (MIF) has emerged as an immunosuppressive factor secreted in the TME. We have previously demonstrated that blockade of the MIF-CD74 signaling on macrophages and dendritic cells restored the anti-tumor immune response against melanoma. Here, we report that inhibition of the MIF-CD74 axis combined with ipilimumab could render resistant melanoma to better respond to anti-CTLA-4 treatment. We provide evidence that blocking the MIF-CD74 signaling potentiates CD8+ T-cells infiltration and drives pro-inflammatory M1 conversion of macrophages in the TME. Furthermore, MIF inhibition resulted in reprogramming the metabolic pathway by reducing lactate production, HIF-1α and PD-L1 expression in the resistant melanoma cells. Melanoma patient data extracted from the TCGA database supports the hypothesis that high MIF expression strongly correlates with poor response to ICB therapy. Our findings provide a rationale for combining anti-CTLA-4 with MIF inhibitors as a potential strategy to overcome resistance to ICB therapy in melanoma, turning a “cold” tumor into a “hot” one mediated by the activation of innate immunity and reprogramming of tumor metabolism and reduced PD-L1 expression in melanoma cells.
- Published
- 2020
- Full Text
- View/download PDF
25. NFAT1 Directly Regulates IL8 and MMP3 to Promote Melanoma Tumor Growth and Metastasis
- Author
-
Li Huang, Victor G. Prieto, Menashe Bar-Eli, Einav Shoshan, Mayra E. Vasquez, Russell R. Braeuer, Takafumi Kamiya, Gabriel J. Villares, Aaron K. Mobley, Guermarie Velazquez-Torres, Cristina Ivan, and Nitin Chakravarti
- Subjects
0301 basic medicine ,Cancer Research ,Lung Neoplasms ,T cell ,Blotting, Western ,Mice, Nude ,Apoptosis ,Biology ,Real-Time Polymerase Chain Reaction ,Article ,Metastasis ,Immunoenzyme Techniques ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Biomarkers, Tumor ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,RNA, Messenger ,Neoplasm Metastasis ,Melanoma ,Transcription factor ,Cell Proliferation ,Neoplasm Staging ,Mice, Inbred BALB C ,Tumor microenvironment ,NFATC Transcription Factors ,Reverse Transcriptase Polymerase Chain Reaction ,Cell growth ,Interleukin-8 ,Cancer ,Prognosis ,medicine.disease ,Xenograft Model Antitumor Assays ,Survival Rate ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,Female ,Matrix Metalloproteinase 3 ,Autotaxin - Abstract
Nuclear factor of activated T cell (NFAT1, NFATC2) is a transcription factor that binds and positively regulates IL2 expression during T-cell activation. NFAT1 has important roles in both innate and adaptive immune responses, but its involvement in cancer is not completely understood. We previously demonstrated that NFAT1 contributes to melanoma growth and metastasis by regulating the autotaxin gene (Enpp2). Here, we report a strong correlation between NFAT1 expression and metastatic potential in melanoma cell lines and tumor specimens. To elucidate the mechanisms underlying NFAT1 overexpression during melanoma progression, we conducted a microarray on a highly metastatic melanoma cell line in which NFAT1 expression was stably silenced. We identified and validated two downstream targets of NFAT1, IL8, and MMP3. Accordingly, NFAT1 depletion in metastatic melanoma cell lines was associated with reduced IL8 and MMP3 expression, whereas NFAT1 overexpression in a weakly metastatic cell line induced expression of these targets. Restoration of NFAT1 expression recovered IL8 and MMP3 expression levels back to baseline, indicating that both are direct targets of NFAT1. Moreover, in vivo studies demonstrated that NFAT1 and MMP3 promoted melanoma tumor growth and lung metastasis. Collectively, our findings assign a new role for NFAT1 in melanoma progression, underscoring the multifaceted functions that immunomodulatory factors may acquire in an unpredictable tumor microenvironment. Cancer Res; 76(11); 3145–55. ©2016 AACR.
- Published
- 2016
- Full Text
- View/download PDF
26. A-to-I miR-378a-3p editing can prevent melanoma progression via regulation of PARVA expression
- Author
-
Cristina Ivan, A. Gordon Robertson, Victoria K. Xie, Gabriel Lopez-Berenstein, Guermarie Velazquez-Torres, Li Huang, Einav Shoshan, Anil K. Sood, Harrison Paret, George A. Calin, Menashe Bar-Eli, Sun Jin Kim, Steven J.M. Jones, Enrique Fuentes-Mattei, Cristian Rodriguez-Aguayo, and Denise Brooks
- Subjects
0301 basic medicine ,Adenosine ,Skin Neoplasms ,Science ,General Physics and Astronomy ,Mice, Nude ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Metastasis ,Epigenesis, Genetic ,03 medical and health sciences ,Mice ,Cell Line, Tumor ,microRNA ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Cancer epigenetics ,Neoplasm Metastasis ,lcsh:Science ,3' Untranslated Regions ,Melanoma ,Cell Proliferation ,Regulation of gene expression ,Multidisciplinary ,Oncogene ,Three prime untranslated region ,Microfilament Proteins ,General Chemistry ,Oncogenes ,medicine.disease ,Inosine ,3. Good health ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,030104 developmental biology ,RNA editing ,Cancer research ,Disease Progression ,lcsh:Q ,Female ,RNA Editing - Abstract
Previously we have reported that metastatic melanoma cell lines and tumor specimens have reduced expression of ADAR1 and consequently are impaired in their ability to perform A-to-I microRNA (miRNA) editing. The effects of A-to-I miRNAs editing on melanoma growth and metastasis are yet to be determined. Here we report that miR-378a–3p is undergoing A-to-I editing only in the non-metastatic but not in metastatic melanoma cells. The function of the edited form is different from its wild-type counterpart. The edited form of miR-378a-3p preferentially binds to the 3′-UTR of the PARVA oncogene and inhibits its expression, thus preventing the progression of melanoma towards the malignant phenotype. Indeed, edited miR-378a-3p but not its WT form inhibits melanoma metastasis in vivo. These results further emphasize the role of RNA editing in melanoma progression., In melanoma, reduced ADAR1 impairs A-to-I microRNA editing. Here, the authors show that miR-378a-3p undergoes this editing in non-metastatic cells and the edited form of miR-378a-3p binds to the PARVA oncogene, inhibiting its expression and preventing melanoma progression and metastasis.
- Published
- 2017
27. Why is melanoma so metastatic?
- Author
-
Einav Shoshan, Aaron K. Mobley, Chang-Jiun Wu, Ian R. Watson, Takafumi Kamiya, Menashe Bar-Eli, and Russell R. Braeuer
- Subjects
Angiogenesis ,Dermatology ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Metastasis ,Immune system ,Antigen ,Antigens, Neoplasm ,Gene expression ,medicine ,Animals ,Humans ,Neoplasm Metastasis ,Melanoma ,neoplasms ,Neovascularization, Pathologic ,Gene Expression Profiling ,Mesenchymal stem cell ,medicine.disease ,Primary tumor ,Gene Expression Regulation, Neoplastic ,Oncology ,Organ Specificity ,Immunology ,Cancer research - Abstract
Malignant melanoma is one of the most aggressive cancers and can disseminate from a relatively small primary tumor and metastasize to multiple sites, including the lung, liver, brain, bone, and lymph nodes. Elucidating the molecular and genetic changes that take place during the metastatic process has led to a better understanding of why melanoma is so metastatic. Herein, we describe the unique features that distinguish melanoma from other solid tumors and contribute to the malignant phenotype of melanoma cells. For example, although melanoma cells are highly antigenic, they are extremely efficient at evading host immune response. Melanoma cells share numerous cell surface molecules with vascular cells, are highly angiogenic, are mesenchymal in nature, and possess a higher degree of 'stemness' than do other solid tumors. Finally, analysis of melanoma mutations has revealed that the gene expression profile of malignant melanoma is different from that of other cancers. Elucidating these molecular and genetic processes in highly metastatic melanoma can lead to the development of improved treatment and individualized therapy options.
- Published
- 2013
- Full Text
- View/download PDF
28. The sweet and bitter sides of galectins in melanoma progression
- Author
-
Einav Shoshan, Takafumi Kamiya, Russell R. Braeuer, and Menashe Bar-Eli
- Subjects
Angiogenesis ,Melanoma ,Cell migration ,Dermatology ,Biology ,medicine.disease ,General Biochemistry, Genetics and Molecular Biology ,Metastasis ,Oncology ,Galectin-3 ,Tumor progression ,Immunology ,Galectin-1 ,Cancer research ,medicine ,Galectin - Abstract
Melanoma is the leading cause of skin cancer-related deaths, which is due in large part to its aggressive behavior, resistance to therapy, and ability to metastasize to multiple organs such as the lymph nodes, lung, and brain. Melanoma progresses in a stepwise manner from the benign nevus, to radial spreading through the dermis, to a vertical invasive phase, and finally to metastasis. The carbohydrate-binding family of galectins has a strong influence on each phase of melanoma progression through their effects on immune surveillance, angiogenesis, cell migration, tumor cell adhesion, and the cellular response to chemotherapy. Galectins share significant homology in their carbohydrate recognition domain (CRD), which mediates binding to an array of N-glycosylated proteins located on the surface of tumor cells, endothelial cells, T-cells, and to similarly glycosylated extracellular matrix proteins. Galectins are also present within tumor cells where they perform anti-apoptotic functions and enhance intracellular signaling that results in deregulated expression of genes involved in tumor progression. The most extensively studied galectins, galectin-1 and galectin-3, have been shown to have profound effects on melanoma growth and metastasis by influencing many of these biological processes.
- Published
- 2012
- Full Text
- View/download PDF
29. Abstract LB-384: A-to-I miR-378a-3p editing prevents melanoma progression via regulation ofPARVA expression
- Author
-
Guermarie Velazquez-Torres, Einav Shoshan, Menashe Bar-Eli, Enrique Fuentes-Mattei, and Cristina Ivan
- Subjects
Cancer Research ,Oncology ,Expression (architecture) ,Melanoma ,medicine ,Cancer research ,Biology ,medicine.disease - Abstract
Although recent studies have shown that adenosine-to-inosine (A-to-I) RNA editing occurs in microRNAs (miRNAs), the effects of A-to-I miRNAs editing on melanoma growth and metastasis are yet to be determined. Previously we have reported that metastatic melanoma cell lines and tumor specimens have reduced expression of ADAR1 and consequently are impaired in their ability to perform A-to-I microRNA (miRNA) editing. Here we report that miR-378a-3p is undergoing A-to-I editing only in the non-metastatic but not in metastatic melanoma cells. The function of the edited form is different from its wild-type counterpart. The edited form of miR-378a-3p preferentially binds to the 3'UTR of the PARVA oncogene and inhibits its expression, thus preventing the progression of melanoma towards the malignant phenotype. Indeed, edited miR- 378a-3p but not its WT form inhibits melanoma metastasis in vivo. These results further emphasize the role of RNA editing in melanoma progression. Citation Format: Einav Shoshan, Guermarie Velazquez-Torres, Enrique Fuentes-Mattei, Cristina Ivan, Menashe Bar-Eli. A-to-I miR-378a-3p editing prevents melanoma progression via regulation ofPARVA expression [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr LB-384.
- Published
- 2018
- Full Text
- View/download PDF
30. Reduced adenosine-to-inosine miR-455-5p editing promotes melanoma growth and metastasis
- Author
-
Agda Karina Eterovic, Li Huang, George A. Calin, Gal Markel, Denise Brooks, Yu Ye Wen, A. Gordon Robertson, Menashe Bar-Eli, Aaron K. Mobley, Isaiah J. Fidler, Patrick Hwu, Anil K. Sood, Cristina Ivan, Mayra E. Vasquez, Ka Ming Nip, Guermarie Velazquez-Torres, Jeffrey E. Gershenwald, Kelsey Zhu, Einav Shoshan, Ahmad Salameh, Maribel Mosqueda, Sun Jin Kim, Steven J.M. Jones, Inanc Birol, Ho Jeong Lee, Russell R. Braeuer, and Takafumi Kamiya
- Subjects
Adenosine ,Skin Neoplasms ,Adenosine Deaminase ,Molecular Sequence Data ,Mice, Nude ,Biology ,Metastasis ,Mice ,Downregulation and upregulation ,Genes, Reporter ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Neoplasm Metastasis ,Inosine ,Cyclic AMP Response Element-Binding Protein ,Luciferases ,Melanoma ,chemistry.chemical_classification ,mRNA Cleavage and Polyadenylation Factors ,Base Sequence ,RNA-Binding Proteins ,Cell Biology ,medicine.disease ,Cell biology ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,Enzyme ,chemistry ,Cancer research ,Disease Progression ,Female ,RNA Editing ,Neoplasm Transplantation ,medicine.drug ,Transcription Factors - Abstract
Although recent studies have shown that adenosine-to-inosine (A-to-I) RNA editing occurs in microRNAs (miRNAs), its effects on tumour growth and metastasis are not well understood. We present evidence of CREB-mediated low expression of ADAR1 in metastatic melanoma cell lines and tumour specimens. Re-expression of ADAR1 resulted in the suppression of melanoma growth and metastasis in vivo. Consequently, we identified three miRNAs undergoing A-to-I editing in the weakly metastatic melanoma but not in strongly metastatic cell lines. One of these miRNAs, miR-455-5p, has two A-to-I RNA-editing sites. The biological function of edited miR-455-5p is different from that of the unedited form, as it recognizes a different set of genes. Indeed, wild-type miR-455-5p promotes melanoma metastasis through inhibition of the tumour suppressor gene CPEB1. Moreover, wild-type miR-455 enhances melanoma growth and metastasis in vivo, whereas the edited form inhibits these features. These results demonstrate a previously unrecognized role for RNA editing in melanoma progression.
- Published
- 2014
31. Abstract PR05: Control of the melanoma metastatic phenotype by A-to-I microRNA editing
- Author
-
Jeffrey E. Gershenwald, P. Hwu, Cristina Ivan, Li Huang, Gal Markel, Sun Jin Kim, Anil K. Sood, George A. Calin, Isaiah J. Fidler, Ho-Jeong Lee, Menashe Bar-Eli, Einav Shoshan, and Mayra E. Vasquez
- Subjects
Cancer Research ,Tumor microenvironment ,Pathology ,medicine.medical_specialty ,Tumor suppressor gene ,Melanoma ,Cancer ,Biology ,medicine.disease ,Metastasis ,Oncology ,In vivo ,RNA editing ,microRNA ,medicine ,Cancer research - Abstract
Although recent studies have shown that adenosine-to-inosine (A-to-I) RNA editing occurs in microRNAs, its effects on tumor growth and metastasis are not well understood. We present evidence of CREB-mediated low expression of ADAR1 in metastatic melanoma cell lines and tumor specimens. Re-expression of ADAR1 resulted in the suppression of melanoma growth and metastasis in vivo. Consequently, we identified 3 miRs (miR-455-5P, mir-378-3P, miR-324-5P) undergoing A-to-I editing in the low-metastatic melanoma cell lines but not in highly metastatic. One of these miRs, miR-455-5P has two A-to-I RNA editing sites. The biological function of edited miR-455-5P is different from the unedited form. Indeed, w.t. miR-455 promotes melanoma metastasis via inhibition of the tumor suppressor gene CPEB1 . Moreover, w.t. miR-455 enhances melanoma growth and metastasis in vivo while the edited form inhibits these features mainly by affecting the tumor microenvironment. TCGA analysis confirmed accumulation of wild-type miR-455 in metastatic melanoma lesions. These results demonstrate a previously unrecognized role of RNA editing in melanoma progression. Citation Format: Einav Shoshan, Li Huang, Mayra E. Vasquez, Ho-Jeong Lee, Sun-Jin Kim, Cristina Ivan, George A. Calin, Anil K. Sood, Patrick Hwu, Jeffrey E. Gershenwald, Gal Markel, Isaiah J. Fidler, Menashe Bar-Eli. Control of the melanoma metastatic phenotype by A-to-I microRNA editing. [abstract]. In: Proceedings of the AACR Special Conference: Function of Tumor Microenvironment in Cancer Progression; 2016 Jan 7–10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2016;76(15 Suppl):Abstract nr PR05.
- Published
- 2016
- Full Text
- View/download PDF
32. Galectin-3 Contributes to Melanoma Growth and Metastasis via Regulation of NFAT1 and Autotaxin
- Author
-
Li Huang, Menashe Bar-Eli, Takafumi Kamiya, Maya Zigler, Avraham Raz, Woonyoung Choi, Einav Shoshan, Andrey S. Dobroff, David J. McConkey, Aaron K. Mobley, Renduo Song, and Russell R. Braeuer
- Subjects
Cancer Research ,NFATC2 ,Lung Neoplasms ,Angiogenesis ,Galectin 3 ,Mice, Nude ,Biology ,Transfection ,Article ,Metastasis ,Mice ,Cell Line, Tumor ,medicine ,otorhinolaryngologic diseases ,Gene silencing ,Animals ,Humans ,Gene Silencing ,RNA, Messenger ,Neoplasm Metastasis ,RNA, Small Interfering ,Melanoma ,Mice, Inbred BALB C ,NFATC Transcription Factors ,Neovascularization, Pathologic ,Phosphoric Diester Hydrolases ,medicine.disease ,Gene expression profiling ,Oncology ,Galectin-3 ,Cancer research ,lipids (amino acids, peptides, and proteins) ,Female ,Autotaxin - Abstract
Melanoma is the deadliest form of skin cancer in which patients with metastatic disease have a 5-year survival rate of less than 10%. Recently, the overexpression of a β-galactoside binding protein, galectin-3 (LGALS3), has been correlated with metastatic melanoma in patients. We have previously shown that silencing galectin-3 in metastatic melanoma cells reduces tumor growth and metastasis. Gene expression profiling identified the protumorigenic gene autotaxin (ENPP2) to be downregulated after silencing galectin-3. Here we report that galectin-3 regulates autotaxin expression at the transcriptional level by modulating the expression of the transcription factor NFAT1 (NFATC2). Silencing galectin-3 reduced NFAT1 protein expression, which resulted in decreased autotaxin expression and activity. Reexpression of autotaxin in galectin-3 silenced melanoma cells rescues angiogenesis, tumor growth, and metastasis in vivo. Silencing NFAT1 expression in metastatic melanoma cells inhibited tumor growth and metastatic capabilities in vivo. Our data elucidate a previously unidentified mechanism by which galectin-3 regulates autotaxin and assign a novel role for NFAT1 during melanoma progression. Cancer Res; 72(22); 5757–66. ©2012 AACR.
- Published
- 2012
33. Driving transcriptional regulators in melanoma metastasis
- Author
-
Aaron K. Mobley, Russell R. Braeuer, Einav Shoshan, Menashe Bar-Eli, and Takafumi Kamiya
- Subjects
Cancer Research ,TFE3 ,E-box ,Sp3 transcription factor ,Transcription (biology) ,Animals ,Humans ,Cyclic AMP Response Element-Binding Protein ,Transcription factor ,Melanoma ,Activating Transcription Factor 1 ,Microphthalmia-Associated Transcription Factor ,General transcription factor ,biology ,Activating Transcription Factor 2 ,NF-kappa B ,Microphthalmia-associated transcription factor ,Molecular biology ,Activating transcription factor 2 ,Cell biology ,Gene Expression Regulation, Neoplastic ,Oncology ,Transcription Factor AP-2 ,biology.protein ,RNA Editing ,Transcription Factors - Abstract
The progression of melanoma toward the metastatic phenotype occurs in a defined stepwise manner. While many molecular changes take place early in melanoma development, progression toward the malignant phenotype, most notably during the transition from the radial growth phase (RGP) to the vertical growth phase (VGP) involves deregulated expression of several transcription factors. For example, the switch from RGP to VGP is associated with the loss of the transcription factor AP2α and gain of transcriptional activity of cAMP-responsive element binding protein. Together with the upregulation of microphthalmia-associated transcription factor, activating transcription factor 2, nuclear factor kappa B, and other transcription factors, these changes lead to dysregulated expression or function of important cellular adhesion molecules, matrix degrading enzymes, survival factors, as well as other factors leading to metastatic melanoma. Additionally, recent evidence suggests that microRNAs and RNA editing machinery influence the expression of transcription factors or are regulated themselves by transcription factors. Many of the downstream signaling molecules regulated by transcription factors, such as protease activated receptor-1, interleukin-8, and MCAM/MUC18 represent new treatment prospects.
- Published
- 2012
34. The sweet and bitter sides of galectins in melanoma progression
- Author
-
Russell R, Braeuer, Einav, Shoshan, Takafumi, Kamiya, and Menashe, Bar-Eli
- Subjects
Galectins ,Disease Progression ,Humans ,Apoptosis ,Melanoma ,Models, Biological ,Cell Proliferation - Abstract
Melanoma is the leading cause of skin cancer-related deaths, which is due in large part to its aggressive behavior, resistance to therapy, and ability to metastasize to multiple organs such as the lymph nodes, lung, and brain. Melanoma progresses in a stepwise manner from the benign nevus, to radial spreading through the dermis, to a vertical invasive phase, and finally to metastasis. The carbohydrate-binding family of galectins has a strong influence on each phase of melanoma progression through their effects on immune surveillance, angiogenesis, cell migration, tumor cell adhesion, and the cellular response to chemotherapy. Galectins share significant homology in their carbohydrate recognition domain (CRD), which mediates binding to an array of N-glycosylated proteins located on the surface of tumor cells, endothelial cells, T-cells, and to similarly glycosylated extracellular matrix proteins. Galectins are also present within tumor cells where they perform anti-apoptotic functions and enhance intracellular signaling that results in deregulated expression of genes involved in tumor progression. The most extensively studied galectins, galectin-1 and galectin-3, have been shown to have profound effects on melanoma growth and metastasis by influencing many of these biological processes.
- Published
- 2012
35. Role of Increased n-acetylaspartate Levels in Cancer
- Author
-
Rajesha Rupaimoole, Guillermo N. Armaiz-Pena, Michael A. Davies, Menashe Bar Eli, Lifeng Yang, Wei Hu, Alpa M. Nick, Behrouz Zand, Christopher McCullough, Gabriel Lopez-Berestein, Lingegowda S. Mangala, Ying Wang, Archana S. Nagaraja, Michele Guindani, Einav Shoshan, Susan K. Lutgendorf, Keith A. Baggerly, Kshipra M. Gharpure, Takashi Mitamura, Deepak Nagrath, Joelle Baddour, Anil K. Sood, Jinsong Liu, Abhinav Achreja, Chad V. Pecot, Heather J. Dalton, Pratip K. Bhattacharya, Rebecca A. Previs, Niki M. Zacharias, Sherry Y. Wu, Cristian Rodriguez-Aguayo, Sunila Pradeep, and Cristina Ivan
- Subjects
0301 basic medicine ,Cancer Research ,Pathology ,medicine.medical_specialty ,Cell Survival ,Apoptosis ,Kaplan-Meier Estimate ,Biology ,Gene Expression Regulation, Enzymologic ,Article ,Mice ,03 medical and health sciences ,Acetyltransferases ,Tandem Mass Spectrometry ,Uterine cancer ,Cell Line, Tumor ,Biomarkers, Tumor ,medicine ,Animals ,Humans ,Gene silencing ,Chromatography, High Pressure Liquid ,Cell Proliferation ,Ovarian Neoplasms ,Aspartic Acid ,Cell growth ,Melanoma ,Ovary ,medicine.disease ,Cystadenocarcinoma, Serous ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,Cancer cell ,Cancer research ,FOXM1 ,Female ,Neoplasm Grading ,Ovarian cancer - Abstract
The clinical and biological effects of metabolic alterations in cancer are not fully understood.In high-grade serous ovarian cancer (HGSOC) samples (n = 101), over 170 metabolites were profiled and compared with normal ovarian tissues (n = 15). To determine NAT8L gene expression across different cancer types, we analyzed the RNA expression of cancer types using RNASeqV2 data available from the open access The Cancer Genome Atlas (TCGA) website (http://www.cbioportal.org/public-portal/). Using NAT8L siRNA, molecular techniques and histological analysis, we determined cancer cell viability, proliferation, apoptosis, and tumor growth in in vitro and in vivo (n = 6-10 mice/group) settings. Data were analyzed with the Student's t test and Kaplan-Meier analysis. Statistical tests were two-sided.Patients with high levels of tumoral NAA and its biosynthetic enzyme, aspartate N-acetyltransferase (NAT8L), had worse overall survival than patients with low levels of NAA and NAT8L. The overall survival duration of patients with higher-than-median NAA levels (3.6 years) was lower than that of patients with lower-than-median NAA levels (5.1 years, P = .03). High NAT8L gene expression in other cancers (melanoma, renal cell, breast, colon, and uterine cancers) was associated with worse overall survival. NAT8L silencing reduced cancer cell viability (HEYA8: control siRNA 90.61% ± 2.53, NAT8L siRNA 39.43% ± 3.00, P.001; A2780: control siRNA 90.59% ± 2.53, NAT8L siRNA 7.44% ± 1.71, P.001) and proliferation (HEYA8: control siRNA 74.83% ± 0.92, NAT8L siRNA 55.70% ± 1.54, P.001; A2780: control siRNA 50.17% ± 4.13, NAT8L siRNA 26.52% ± 3.70, P.001), which was rescued by addition of NAA. In orthotopic mouse models (ovarian cancer and melanoma), NAT8L silencing reduced tumor growth statistically significantly (A2780: control siRNA 0.52 g ± 0.15, NAT8L siRNA 0.08 g ± 0.17, P.001; HEYA8: control siRNA 0.79 g ± 0.42, NAT8L siRNA 0.24 g ± 0.18, P = .008, A375-SM: control siRNA 0.55 g ± 0.22, NAT8L siRNA 0.21 g ± 0.17 g, P = .001). NAT8L silencing downregulated the anti-apoptotic pathway, which was mediated through FOXM1.These findings indicate that the NAA pathway has a prominent role in promoting tumor growth and represents a valuable target for anticancer therapy.Altered energy metabolism is a hallmark of cancer (1). Proliferating cancer cells have much greater metabolic requirements than nonproliferating differentiated cells (2,3). Moreover, altered cancer metabolism elevates unique metabolic intermediates, which can promote cancer survival and progression (4,5). Furthermore, emerging evidence suggests that proliferating cancer cells exploit alternative metabolic pathways to meet their high demand for energy and to accumulate biomass (6-8).
- Published
- 2016
- Full Text
- View/download PDF
36. Abstract 4969: Hypo adenosine-to-inosine editing of microRNA-455 promotes melanoma metastasis
- Author
-
Anil K. Sood, Isaiah J. Fidler, Li Huang, Sun Jim kim, Einav Shoshan, Russell R. Braeuer, Lee H. Jeong, Mayra E. Vasquez, Gal Markel, George A. Calin, Aaron K. Mobley, Menashe Bar-Eli, and Takafumi Kamiya
- Subjects
Cancer Research ,biology ,business.industry ,Melanoma ,RNA ,Cancer ,CREB ,medicine.disease ,Metastasis ,Adenosine deaminase ,Oncology ,RNA editing ,microRNA ,Immunology ,Cancer research ,biology.protein ,Medicine ,business - Abstract
We have previously reported that activation of CREB in metastatic melanoma cells resulted in reduced expression of adenosine deaminase acting on RNA 1 (ADAR1), an RNA editing enzyme. We hypothesized that disruption of A-to-I editing of microRNAs alters expression of genes regulating melanoma progression. By sequencing miRs in primary vs. metastatic melanoma cells, we show that the overall microRNA editing frequencies were reduced in metastatic melanoma cells. Specifically, we found A-to-I editing in two sites of miR-455 in non-metastatic melanoma cells with accumulation of unedited miR-455 in metastatic melanoma cells. Using two animal models, we demonstrated that the unedited miR-455 promoted melanoma growth and metastasis, while the edited form suppressed these features. Unedited miR-455 promotes melanoma progression via suppression of the tumor suppressor cytoplasmic polyadenilation element binding protein (CPEB1). Thus, miR-455 A-to-I editing alters the selection of its target genes and directs its function to inhibit melanoma growth and metastasis. Taken together, our results provide a previously undescribed mechanism (microRNA editing) contributing to melanoma progression. Citation Format: Einav Shoshan, Aaron Mobley, Russell Braeuer, Takafumi Kamiya, Li Huang, Mayra Vasquez, Lee Ho Jeong, Sun Jim kim, George Calin, Anil Sood, Gal Markel, Isaiah Fidler, Menashe Bar-Eli. Hypo adenosine-to-inosine editing of microRNA-455 promotes melanoma metastasis. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4969. doi:10.1158/1538-7445.AM2014-4969
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.