1
|
Chi H, Su L, Yan Y, Gu X, Su K, Li H, Yu L, Liu J, Wang J, Wu Q, Yang G. Illuminating the immunological landscape: mitochondrial gene defects in pancreatic cancer through a multiomics lens. Front Immunol 2024; 15:1375143. [PMID: 38510247 PMCID: PMC10953916 DOI: 10.3389/fimmu.2024.1375143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 02/16/2024] [Indexed: 03/22/2024] Open
Abstract
This comprehensive review delves into the complex interplay between mitochondrial gene defects and pancreatic cancer pathogenesis through a multiomics approach. By amalgamating data from genomic, transcriptomic, proteomic, and metabolomic studies, we dissected the mechanisms by which mitochondrial genetic variations dictate cancer progression. Emphasis has been placed on the roles of these genes in altering cellular metabolic processes, signal transduction pathways, and immune system interactions. We further explored how these findings could refine therapeutic interventions, with a particular focus on precision medicine applications. This analysis not only fills pivotal knowledge gaps about mitochondrial anomalies in pancreatic cancer but also paves the way for future investigations into personalized therapy options. This finding underscores the crucial nexus between mitochondrial genetics and oncological immunology, opening new avenues for targeted cancer treatment strategies.
Collapse
Affiliation(s)
- Hao Chi
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, and University Hospital, Macau University of Science and Technology, Macau, Macao SAR, China
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Lanqian Su
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Yalan Yan
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Xiang Gu
- Biology Department, Southern Methodist University, Dallas, TX, United States
| | - Ke Su
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Han Li
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Lili Yu
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, and University Hospital, Macau University of Science and Technology, Macau, Macao SAR, China
| | - Jie Liu
- Department of General Surgery, Dazhou Central Hospital, Dazhou, China
| | - Jue Wang
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, and University Hospital, Macau University of Science and Technology, Macau, Macao SAR, China
| | - Qibiao Wu
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, and University Hospital, Macau University of Science and Technology, Macau, Macao SAR, China
| | - Guanhu Yang
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, and University Hospital, Macau University of Science and Technology, Macau, Macao SAR, China
- Department of Specialty Medicine, Ohio University, Athens, OH, United States
| |
Collapse
|
2
|
Elucidated tumorigenic role of MAML1 and TWIST1 in gastric cancer is associated with Helicobacter pylori infection. Microb Pathog 2021; 162:105304. [PMID: 34818576 DOI: 10.1016/j.micpath.2021.105304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND Epithelial-mesenchymal transition (EMT) has a fundamental role in tumor initiation, progression, and metastasis. Helicobacter pylori (HP) induces EMT and thus causes gastric cancer (GC) by deregulating multiple signaling pathways involved in EMT. TWIST1 and MAML1 have been confirmed to be critical inducers of EMT via diverse signaling pathways such as Notch signaling. This study aimed to investigate for the first time possible associations between TWIST1/MAML1 mRNA expression levels, HP infection, and clinicopathological characteristics in GC patients. METHOD TWIST1 and MAML1 mRNA expression levels were evaluated in tumoral and adjacent normal tissues in 73 GC patients using the quantitative reverse transcription PCR (RT-qPCR) method. PCR technique was also applied to examine the infection with HP in GC samples. RESULTS Upregulation of TWIST1 and MAML1 expression was observed in 35 (48%) and 34 (46.6%) of 73 tumor samples, respectively. Co-overexpression of these genes was found in 26 of 73 (35.6%) tumor samples; meanwhile, there was a significant positive correlation between MAML1 and TWIST1 mRNA expression levels (P < 0.001). MAML1 overexpression exhibited meaningful associations with advanced tumor stages (P = 0.006) and nodal metastases (P ˂ 0.001). 34 of 73 (46.6%) tumors tested positive for HP, and meanwhile, MAML1 expression was positively related with T (P = 0.05) and grade (P = 0.0001) in these HP-positive samples. Increased TWIST1 expression was correlated with patient sex (P = 0.035) and advanced tumor grade (P = 0.017) in HP-infected tumors. Furthermore, TWIST1 and MAML1 expression levels were inversely linked with histologic grade in HP-negative tumor samples (P = 0.021 and P = 0.048, respectively). CONCLUSION We propose TWIST1 and MAML1 as potential biomarkers of advanced-stage GC that determine the characteristics and aggressiveness of the disease. Based on accumulating evidence and our findings, they can be introduced as promising therapeutic targets to modify functional abnormalities in cells that promote GC progression. Moreover, HP may enhance GC growth and metastasis by disrupting TWIS1/MAML1 expression patterns and related pathways.
Collapse
|
3
|
Jia M, Su B, Mo L, Qiu W, Ying J, Lin P, Yang B, Li D, Wang D, Xu L, Li H, Zhou Z, Li X, Li J. Circadian clock protein CRY1 prevents paclitaxel‑induced senescence of bladder cancer cells by promoting p53 degradation. Oncol Rep 2021; 45:1033-1043. [PMID: 33650658 PMCID: PMC7860017 DOI: 10.3892/or.2020.7914] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 10/06/2020] [Indexed: 12/19/2022] Open
Abstract
Bladder cancer is a common tumor type of the urinary system, which has high levels of morbidity and mortality. The first‑line treatment is cisplatin‑based combination chemotherapy, but a significant proportion of patients relapse due to the development of drug resistance. Therapy‑induced senescence can act as a 'back‑up' response to chemotherapy in cancer types that are resistant to apoptosis‑based anticancer therapies. The circadian clock serves an important role in drug resistance and cellular senescence. The aim of the present study was to investigate the regulatory effect of the circadian clock on paclitaxel (PTX)‑induced senescence in cisplatin‑resistant bladder cancer cells. Cisplatin‑resistant bladder cancer cells were established via long‑term cisplatin incubation. PTX induced apparent senescence in bladder cancer cells as demonstrated via SA‑β‑Gal staining, but this was not observed in the cisplatin‑resistant cells. The cisplatin‑resistant cells entered into a quiescent state with prolonged circadian rhythm under acute PTX stress. It was identified that the circadian protein cryptochrome1 (CRY1) accumulated in these quiescent cisplatin‑resistant cells, and that CRY1 knockdown restored PTX‑induced senescence. Mechanistically, CRY1 promoted p53 degradation via increasing the binding of p53 with its ubiquitin E3 ligase MDM2 proto‑oncogene. These data suggested that the accumulated CRY1 in cisplatin‑resistant cells could prevent PTX‑induced senescence by promoting p53 degradation.
Collapse
Affiliation(s)
- Min Jia
- Shenzhen Ruipuxun Academy for Stem Cell and Regenerative Medicine, Shenzhen, Guangdong 518122, P.R. China
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Bijia Su
- Shenzhen Ruipuxun Academy for Stem Cell and Regenerative Medicine, Shenzhen, Guangdong 518122, P.R. China
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Lijun Mo
- Shenzhen Ruipuxun Academy for Stem Cell and Regenerative Medicine, Shenzhen, Guangdong 518122, P.R. China
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Wen Qiu
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jiaxu Ying
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Peng Lin
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Bingxuan Yang
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Danying Li
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Dongxia Wang
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Lili Xu
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Hongwei Li
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Zhongxin Zhou
- Department of Vascular Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510630, P.R. China
| | - Xing Li
- Shenzhen Ruipuxun Academy for Stem Cell and Regenerative Medicine, Shenzhen, Guangdong 518122, P.R. China
- Department of Medical Oncology and Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Jinlong Li
- Shenzhen Ruipuxun Academy for Stem Cell and Regenerative Medicine, Shenzhen, Guangdong 518122, P.R. China
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
4
|
Jia M, Zheng D, Wang X, Zhang Y, Chen S, Cai X, Mo L, Hu Z, Li H, Zhou Z, Li J. Cancer Cell enters reversible quiescence through Intracellular Acidification to resist Paclitaxel Cytotoxicity. Int J Med Sci 2020; 17:1652-1664. [PMID: 32669967 PMCID: PMC7359388 DOI: 10.7150/ijms.46034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 06/12/2020] [Indexed: 12/16/2022] Open
Abstract
Cancer cells can enter quiescent or dormant state to resist anticancer agents while maintaining the potential of reactivation. However, the molecular mechanism underlying quiescence entry and reactivation remains largely unknown. In this paper, cancer cells eventually entered a reversible quiescent state to resist long-term paclitaxel (PTX) stress. The quiescent cells were characterized with Na+/H+ exchanger 1 (NHE1) downregulation and showed acidic intracellular pH (pHi). Accordingly, decreasing pHi by NHE1 inhibitor could induce cell enter quiescence. Further, acidic pHi could activate the ubiquitin-proteasome system and inhibiting proteasome activity by MG132 prevented cells entering quiescence. In addition, we show that after partial release, the key G1-S transcription factor E2F1 protein level was not recovered, while MCM7 protein returned to normal level in the reactivated cells. More importantly, MCM7 knockdown inhibited G1/S genes transcription and inhibited the reactivated proliferation. Taken together, this study demonstrates a regulatory function of intracellular acidification and subsequent protein ubiquitination on quiescence entry, and reveals a supportive effect of MCM7 on the quiescence-reactivated proliferation.
Collapse
Affiliation(s)
- Min Jia
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Dianpeng Zheng
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiuyun Wang
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Yongjun Zhang
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Sansan Chen
- Department of Urology, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Xiangsheng Cai
- Clinical Laboratory, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Lijun Mo
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhiming Hu
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Hongwei Li
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhongxin Zhou
- Department of Vascular Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jinlong Li
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
5
|
Cheng H, Chen L, Hu X, Qiu H, Xu X, Gao L, Tang G, Zhang W, Wang J, Yang J, Huang C. Knockdown of MAML1 inhibits proliferation and induces apoptosis of T-cell acute lymphoblastic leukemia cells through SP1-dependent inactivation of TRIM59. J Cell Physiol 2019; 234:5186-5195. [PMID: 30370525 DOI: 10.1002/jcp.27323] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 08/03/2018] [Indexed: 11/06/2022]
Abstract
Notch exerts important functions in cell proliferation, survival, and differentiation, which plays a critical role in tumor development when aberrantly activated. Mastermind-like protein 1 (MAML1) has been functioning as crucial coactivators of Notch receptors and is required for stable formation of Notch transcriptional complexes. However, the mechanism whereby MAML1 induces T-cell acute lymphoblastic leukemia (T-ALL) tumorigenesis is largely unknown. The CCK-8 and flow cytometry assay were performed to examine the effect of MAML1 knockdown on T-ALL cell proliferation, apoptosis, and cell cycle. The expression of MAML1, cell cycle, and apoptosis-related gene, as well as TRIM family members and specific protein 1 (SP1) was measured by western blot analysis and qPCR. Our results showed that MAML1 knockdown significantly inhibited cell proliferation and induced G0/G1 cell cycle arrest and apoptosis in Jurkat and MOLT-4 cells. Cell cycle and apoptosis-related gene expression, including CDK2, Bcl-2, Bax, and Bad, was modified by the MAML1 knockdown. MAML1 knockdown obviously inhibited the CDK2 and Bcl-2 expression and increased the Bax, p53, and Bad expression. Moreover, the TRIM family members, including TRIM13, TRIM32, TRIM44, and TRIM59, were significantly decreased by the MAML1 knockdown, with the highest decrease detected in TRIM59 expression. Interesting, overexpression of SP1 not only increased the expression of MAML1 and TRIM59, but also promoted the promoter activation of TRIM59. Taken together, knockdown of MAML1 inhibits proliferation and induces apoptosis of T-ALL cells through SP1-dependent inactivation of TRIM59, and therefore suggest that MAML1-SP1-TRIM59 axis may serve as potentially interesting therapeutic targets for treatment of T-ALL.
Collapse
Affiliation(s)
- Hui Cheng
- Institute of Hematology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Li Chen
- Institute of Hematology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xiaoxia Hu
- Institute of Hematology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Huiying Qiu
- Institute of Hematology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xiaoqian Xu
- Institute of Hematology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Lei Gao
- Institute of Hematology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Gusheng Tang
- Institute of Hematology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Weiping Zhang
- Institute of Hematology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jianmin Wang
- Institute of Hematology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jianmin Yang
- Institute of Hematology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Chongmei Huang
- Institute of Hematology, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
6
|
Golan T, Levy C. Negative Regulatory Loop between Microphthalmia-Associated Transcription Factor (MITF) and Notch Signaling. Int J Mol Sci 2019; 20:E576. [PMID: 30699982 PMCID: PMC6387231 DOI: 10.3390/ijms20030576] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/24/2019] [Accepted: 01/26/2019] [Indexed: 11/29/2022] Open
Abstract
Melanoma, a melanocyte-origin neoplasm, is a highly metastatic and treatment-resistance cancer. While it is well established that notch signaling activation promotes melanoma progression, little is known about the reciprocal interactions between Notch signaling and melanoma-specific pathways. Here we reveal a negative regulatory loop between Notch signaling and microphthalmia-associated transcription factor (MITF), the central regulator of melanoma progression and the driver of melanoma plasticity. We further demonstrate that Notch signaling activation, in addition to the known competition-based repression mechanism of MITF transcriptional activity, inhibits the transcription of MITF, leading to a decrease in MITF expression. We also found that MITF binds to the promoter of the gene encoding the master regulator of Notch signaling, recombination signal binding protein J kappa (RBPJK), leading to its upregulation. Our findings suggest that, once activated, Notch signaling represses MITF signaling to maintain the melanoma invasiveness and metastatic phenotype.
Collapse
Affiliation(s)
- Tamar Golan
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| | - Carmit Levy
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
7
|
Liu T, Yang T, Xu Z, Tan S, Pan T, Wan N, Li S. MicroRNA-193b-3p regulates hepatocyte apoptosis in selenium-deficient broilers by targeting MAML1. J Inorg Biochem 2018; 186:235-245. [DOI: 10.1016/j.jinorgbio.2018.06.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 06/14/2018] [Accepted: 06/24/2018] [Indexed: 02/08/2023]
|
8
|
Colombo M, Mirandola L, Chiriva-Internati M, Basile A, Locati M, Lesma E, Chiaramonte R, Platonova N. Cancer Cells Exploit Notch Signaling to Redefine a Supportive Cytokine Milieu. Front Immunol 2018; 9:1823. [PMID: 30154786 PMCID: PMC6102368 DOI: 10.3389/fimmu.2018.01823] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 07/24/2018] [Indexed: 12/19/2022] Open
Abstract
Notch signaling is a well-known key player in the communication between adjacent cells during organ development, when it controls several processes involved in cell differentiation. Notch-mediated communication may occur through the interaction of Notch receptors with ligands on adjacent cells or by a paracrine/endocrine fashion, through soluble molecules that can mediate the communication between cells at distant sites. Dysregulation of Notch pathway causes a number of disorders, including cancer. Notch hyperactivation may be caused by mutations of Notch-related genes, dysregulated upstream pathways, or microenvironment signals. Cancer cells may exploit this aberrant signaling to "educate" the surrounding microenvironment cells toward a pro-tumoral behavior. This may occur because of key cytokines secreted by tumor cells or it may involve the microenvironment through the activation of Notch signaling in stromal cells, an event mediated by a direct cell-to-cell contact and resulting in the increased secretion of several pro-tumorigenic cytokines. Up to now, review articles were mainly focused on Notch contribution in a specific tumor context or immune cell populations. Here, we provide a comprehensive overview on the outcomes of Notch-mediated pathological interactions in different tumor settings and on the molecular and cellular mediators involved in this process. We describe how Notch dysregulation in cancer may alter the cytokine network and its outcomes on tumor progression and antitumor immune response.
Collapse
Affiliation(s)
- Michela Colombo
- Department of Health Sciences, Università degli Studi di Milano, Milano, Italy
| | | | - Maurizio Chiriva-Internati
- Kiromic Biopharma Inc., Houston, TX, United States.,Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Andrea Basile
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milano, Italy
| | - Massimo Locati
- Department of Medical Biotechnologies and Translational Medicine, Università degli Studi di Milano, Milano, Italy.,Humanitas Clinical and Research Center, Rozzano, Italy
| | - Elena Lesma
- Department of Health Sciences, Università degli Studi di Milano, Milano, Italy
| | | | - Natalia Platonova
- Department of Health Sciences, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
9
|
Yang Z, Qi Y, Lai N, Zhang J, Chen Z, Liu M, Zhang W, Luo R, Kang S. Notch1 signaling in melanoma cells promoted tumor-induced immunosuppression via upregulation of TGF-β1. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:1. [PMID: 29301578 PMCID: PMC5755139 DOI: 10.1186/s13046-017-0664-4] [Citation(s) in RCA: 235] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 12/11/2017] [Indexed: 01/05/2023]
Abstract
Background The receptors of Notch family play an important role in controlling the development, differentiation, and function of multiple cell types. The aim of this study is to investigate the role of Notch1 signaling upon immune suppression induced by melanoma cells. Methods Melanoma cell line B16 cells were transfected by lentivirus containing mouse Notch1 gene or Notch1 shRNA to generate B16 cell line that highly or lowly expressed Notch1. Notch1 in anti-tumor immune response was comprehensively appraised in murine B16 melanoma tumor model in immunocompetent and immunodeficient mice. The ratios of CD3+CD8+ cytotoxic T cells, CD49b+NK cells, CD4+CD25+FoxP3+ Tregs and Gr1+CD11b+ MDSCs in tumor-DLN or spleen were examined by flow cytometry. After the co-culture of B16 cells and CD8+ T cells, the effects of Notch1 on the proliferation and activation of T cells were assessed by CCK8 assay, CFSE dilution and Chromium-release test. The mRNA expression and supernatant secretion of immunosuppressive cytokines, TGF-β1, VEGF, IL-10 and IFN-γ were measured by RT-PCR and ELISA, respectively. Results Downregulation or overexpression of Notch1 in B16 melanoma cells inhibited or promoted tumor growth in immunocompetent mice, respectively. Notch1 expression in B16 melanoma cells inhibited the infiltration of CD8+ cytotoxic T lymphocytes and NK cells and reduced IFN-γ release in tumor tissue. It could also enhance B16 cell-mediated inhibition of T cell proliferation and activation, and upregulate PD-1 expression on CD4+ and CD8+ T cells. The percentage of CD4+CD25+FoxP3+ Tregs and Gr1+CD11b+MDSCs were significantly increased in tumor microenvironment, and all these were attributed to the upregulation of TGF-β1. Conclusion These findings suggested that Notch1 signaling in B16 melanoma cells might inhibit antitumor immunity by upregulation of TGF-β1.
Collapse
Affiliation(s)
- Zike Yang
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, No.13, Shiliugang Road, Haizhu District, Guangzhou, 510315, Guangdong Province, People's Republic of China
| | - Yanxia Qi
- Cancer Center, The First People's Hospital of Huaihua City, Huaihua, 418000, Hunan Province, People's Republic of China
| | - Nan Lai
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, No.13, Shiliugang Road, Haizhu District, Guangzhou, 510315, Guangdong Province, People's Republic of China
| | - Jiahe Zhang
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, No.13, Shiliugang Road, Haizhu District, Guangzhou, 510315, Guangdong Province, People's Republic of China
| | - Zehong Chen
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, No.13, Shiliugang Road, Haizhu District, Guangzhou, 510315, Guangdong Province, People's Republic of China
| | - Mingyu Liu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, No.13, Shiliugang Road, Haizhu District, Guangzhou, 510315, Guangdong Province, People's Republic of China
| | - Wan Zhang
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, No.13, Shiliugang Road, Haizhu District, Guangzhou, 510315, Guangdong Province, People's Republic of China
| | - Rongcheng Luo
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, No.13, Shiliugang Road, Haizhu District, Guangzhou, 510315, Guangdong Province, People's Republic of China.
| | - Shijun Kang
- Oncology Department, Nanfang Hospital, Southern Medical University, No.1838, North of Guangzhou Avenue, Baiyun District, Guangzhou, Guangdong Province, 510515, People's Republic of China.
| |
Collapse
|
10
|
HES1 in immunity and cancer. Cytokine Growth Factor Rev 2016; 30:113-7. [PMID: 27066918 DOI: 10.1016/j.cytogfr.2016.03.010] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 03/18/2016] [Accepted: 03/18/2016] [Indexed: 01/06/2023]
Abstract
Hairy and enhancer of split homolog-1 (HES1) is a part of an extensive family of basic helix-loop-helix (bHLH) proteins and plays a crucial role in the control and regulation of cell cycle, proliferation, cell differentiation, survival and apoptosis in neuronal, endocrine, T-lymphocyte progenitors as well as various cancers. HES1 is a transcription factor which is regulated by the NOTCH, Hedgehog and Wnt signalling pathways. Aberrant expression of these pathways is a common feature of cancerous cells. There appears to be a fine and complicated crosstalk at the molecular level between the various signalling pathways and HES1, which contributes to its effects on the immune response and cancers such as leukaemia. Several mechanisms have been proposed, including an enhanced invasiveness and metastasis by inducing epithelial mesenchymal transition (EMT), in addition to its strict requirement for tumour cell survival. In this review, we summarize the current biology and molecular mechanisms as well as its use as a clinical target in cancer therapeutics.
Collapse
|
11
|
Fujimoto M, Mano Y, Anai M, Yamamoto S, Fukuyo M, Aburatani H, Kaneda A. Epigenetic alteration to activate Bmp2-Smad signaling in Raf-induced senescence. World J Biol Chem 2016; 7:188-205. [PMID: 26981207 PMCID: PMC4768123 DOI: 10.4331/wjbc.v7.i1.188] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 10/30/2015] [Accepted: 12/04/2015] [Indexed: 02/05/2023] Open
Abstract
AIM: To investigate epigenomic and gene expression alterations during cellular senescence induced by oncogenic Raf.
METHODS: Cellular senescence was induced into mouse embryonic fibroblasts (MEFs) by infecting retrovirus to express oncogenic Raf (RafV600E). RNA was collected from RafV600E cells as well as MEFs without infection and MEFs with mock infection, and a genome-wide gene expression analysis was performed using microarray. The epigenomic status for active H3K4me3 and repressive H3K27me3 histone marks was analyzed by chromatin immunoprecipitation-sequencing for RafV600E cells on day 7 and for MEFs without infection. These data for Raf-induced senescence were compared with data for Ras-induced senescence that were obtained in our previous study. Gene knockdown and overexpression were done by retrovirus infection.
RESULTS: Although the expression of some genes including secreted factors was specifically altered in either Ras- or Raf-induced senescence, many genes showed similar alteration pattern in Raf- and Ras-induced senescence. A total of 841 commonly upregulated 841 genes and 573 commonly downregulated genes showed a significant enrichment of genes related to signal and secreted proteins, suggesting the importance of alterations in secreted factors. Bmp2, a secreted protein to activate Bmp2-Smad signaling, was highly upregulated with gain of H3K4me3 and loss of H3K27me3 during Raf-induced senescence, as previously detected in Ras-induced senescence, and the knockdown of Bmp2 by shRNA lead to escape from Raf-induced senescence. Bmp2-Smad inhibitor Smad6 was strongly repressed with H3K4me3 loss in Raf-induced senescence, as detected in Ras-induced senescence, and senescence was also bypassed by Smad6 induction in Raf-activated cells. Different from Ras-induced senescence, however, gain of H3K27me3 did not occur in the Smad6 promoter region during Raf-induced senescence. When comparing genome-wide alteration between Ras- and Raf-induced senescence, genes showing loss of H3K27me3 during senescence significantly overlapped; genes showing H3K4me3 gain, or those showing H3K4me3 loss, also well-overlapped between Ras- and Raf-induced senescence. However, genes with gain of H3K27me3 overlapped significantly rarely, compared with those with H3K27me3 loss, with H3K4me3 gain, or with H3K4me3 loss.
CONCLUSION: Although epigenetic alterations are partly different, Bmp2 upregulation and Smad6 repression occur and contribute to Raf-induced senescence, as detected in Ras-induced senescence.
Collapse
|
12
|
Lan L, Holland JD, Qi J, Grosskopf S, Rademann J, Vogel R, Györffy B, Wulf-Goldenberg A, Birchmeier W. Shp2 signaling suppresses senescence in PyMT-induced mammary gland cancer in mice. EMBO J 2015; 34:1493-508. [PMID: 25736378 DOI: 10.15252/embj.201489004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 02/04/2015] [Indexed: 12/26/2022] Open
Abstract
In this study, we have used techniques from cell biology, biochemistry, and genetics to investigate the role of the tyrosine phosphatase Shp2 in tumor cells of MMTV-PyMT mouse mammary glands. Genetic ablation or pharmacological inhibition of Shp2 induces senescence, as determined by the activation of senescence-associated β-gal (SA-β-gal), cyclin-dependent kinase inhibitor 1B (p27), p53, and histone 3 trimethylated lysine 9 (H3K9me3). Senescence induction leads to the inhibition of self-renewal of tumor cells and blockage of tumor formation and growth. A signaling cascade was identified that acts downstream of Shp2 to counter senescence: Src, focal adhesion kinase, and Map kinase inhibit senescence by activating the expression of S-phase kinase-associated protein 2 (Skp2), Aurora kinase A (Aurka), and the Notch ligand Delta-like 1 (Dll1), which block p27 and p53. Remarkably, the expression of Shp2 and of selected target genes predicts human breast cancer outcome. We conclude that therapies, which rely on senescence induction by inhibiting Shp2 or controlling its target gene products, may be useful in blocking breast cancer.
Collapse
Affiliation(s)
- Linxiang Lan
- Cancer Research Program, Max-Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Jane D Holland
- Cancer Research Program, Max-Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Jingjing Qi
- Cancer Research Program, Max-Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Stefanie Grosskopf
- Cancer Research Program, Max-Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | | | - Regina Vogel
- Cancer Research Program, Max-Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Balázs Györffy
- MTA TTK Lendület Cancer Biomarker Research Group, Budapest, Hungary 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | | | - Walter Birchmeier
- Cancer Research Program, Max-Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| |
Collapse
|
13
|
Su YX, Hou CC, Yang WX. Control of hair cell development by molecular pathways involving Atoh1, Hes1 and Hes5. Gene 2014; 558:6-24. [PMID: 25550047 DOI: 10.1016/j.gene.2014.12.054] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 11/23/2014] [Accepted: 12/25/2014] [Indexed: 01/14/2023]
Abstract
Atoh1, Hes1 and Hes5 are crucial for normal inner ear hair cell development. They regulate the expression of each other in a complex network, while they also interact with many other genes and pathways, such as Notch, FGF, SHH, WNT, BMP and RA. This paper summarized molecular pathways that involve Atoh1, Hes1, and Hes5. Some of the pathways and gene regulation mechanisms discussed here were studied in other tissues, yet they might inspire studies in inner ear hair cell development. Thereby, we presented a complex regulatory network involving these three genes, which might be crucial for proliferation and differentiation of inner ear hair cells.
Collapse
Affiliation(s)
- Yi-Xun Su
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Cong-Cong Hou
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|