1
|
Niu Y, Tang Y, Ma F, Zhou X, Chen Y, Wang Y, Xu Y, Sun L, Liang S, Yang J, Wang K, Zhang F, Su S, Guo L. Super-enhancer MYCNOS-SE promotes chemoresistance in small cell lung cancer by recruiting transcription factors CTCF and KLF15. Oncogene 2025; 44:255-268. [PMID: 39511411 PMCID: PMC11746145 DOI: 10.1038/s41388-024-03202-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 10/07/2024] [Accepted: 10/16/2024] [Indexed: 11/15/2024]
Abstract
Small cell lung cancer (SCLC) is an aggressive form of lung cancer that often becomes resistant to chemotherapy. Understanding the molecular mechanisms of chemoresistance is crucial for identifying effective therapeutic targets. In this study, we used RNA-Seq to identify highly expressed molecules associated with chemoresistance. We also performed H3K27Ac and ATAC-Seq binding analyses to identify super-enhancers (SE) and their corresponding transcription factors. Both in vitro and in vivo experiments were conducted to examine the impact of these molecules and clinical samples were collected to establish their prognostic value. Our findings revealed elevated expression of MYCNOS, which exhibited chemoresistant properties in both in vitro and in vivo models of SCLC. We identified MYCNOS-SE as a significant SE in SCLC that regulates the distal target gene MYCNOS. This SE recruits transcription factors CTCF and KLF15 to regulate MYCNOS expression. Additionally, MYCNOS, an antisense of MYCN, was found to modulate chemotherapy sensitivity through the NOTCH pathway. This study highlights the significance of SE -regulated target genes as markers for chemoresistance in SCLC. Furthermore, it suggests that MYCNOS could serve as a predictor to identify patients who may benefit from NOTCH inhibitors. These findings provide valuable insights for future studies aimed at developing therapeutic strategies targeting these identified pathways.
Collapse
Affiliation(s)
- Yuchun Niu
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Radiation Oncology, The First People's Hospital of Foshan, Foshan, China
| | - Yichun Tang
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Feng Ma
- Department of Radiation Oncology, The First People's Hospital of Foshan, Foshan, China
| | - Xuyang Zhou
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yi Chen
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yu Wang
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yue Xu
- Department of Oncology, Guangzhou Chest Hospital, Guangzhou, China
| | - Lei Sun
- Department of Oncology, The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, China
| | - Shaoqiang Liang
- Department of Radiation Oncology, The First People's Hospital of Foshan, Foshan, China
| | - Jianqi Yang
- Department of Orthopedics, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Kai Wang
- Department of Orthopedics, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Fan Zhang
- Department of Pathology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China.
| | - Shan Su
- Department of Oncology, Guangzhou Chest Hospital, Guangzhou, China.
| | - Linlang Guo
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
2
|
Ma J, Zhang P, Wang Y, Lu M, Cao K, Wei S, Qi C, Ling X, Zhu J. LncRNA HAR1A inhibits non-small cell lung cancer growth by downregulating c-MYC transcripts and facilitating its proteasomal degradation. Int Immunopharmacol 2024; 142:113264. [PMID: 39340992 DOI: 10.1016/j.intimp.2024.113264] [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: 06/06/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 09/30/2024]
Abstract
Non-small cell lung cancer (NSCLC) is a primary cause of cancer-related mortality on a global scale. Research increasingly shows that long non-coding RNAs (lncRNAs) play crucial regulatory roles and serve as biomarkers for diagnosis, prognosis, therapy monitoring, and druggable targets in NSCLC. We previously identified HAR1A as a tumor-suppressing lncRNA in NSCLC, with its loss also observed in oral and hepatocellular carcinoma. This study aimed to expand the understanding of the functional role of HAR1A in NSCLC and uncover its underlying mechanisms. Our results demonstrated that elevating HAR1A levels impeded NSCLC cell proliferation and migration but promoted apoptosis, thereby boosting their susceptibility to cisplatin. Subsequently, we discovered that HAR1A enhanced cisplatin's cytotoxicity in NSCLC cells by curbing adaptive autophagy through the downregulation of MYC. Further analysis revealed that HAR1A suppresses MYC by both lowering its transcript levels and promoting protein ubiquitination and degradation, thereby restricting tumor cell proliferation, migration, and adaptive autophagy. In exploring MYC's targets, we observed that MYC upregulated the transcription of heat shock protein 90 alpha family class B member 1 (HSP90AB1/HSP90β) gene. Rescue experiments verified that HAR1A mitigated NSCLC cell proliferation and migration and induced apoptosis through the MYC/HSP90β axis. Finally, we confirmed that HAR1A overexpression increased cisplatin efficacy in nude mouse NSCLC xenograft models.In conclusion, the findings suggest that HAR1A could be a promising therapeutic target in treating NSCLC and biomarkers for predicting chemotherapy outcomes. This study provides new insights into the molecular mechanisms of chemoresistance in NSCLC and underscores the potential of lncRNA-based strategies in cancer therapy.
Collapse
Affiliation(s)
- Jianqun Ma
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150040, Heilongjiang, China
| | - Ping Zhang
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150040, Heilongjiang, China
| | - Yuning Wang
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150040, Heilongjiang, China
| | - Mengdi Lu
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150040, Heilongjiang, China
| | - Kui Cao
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150040, Heilongjiang, China
| | - Shenshui Wei
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150040, Heilongjiang, China
| | - Cuicui Qi
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150040, Heilongjiang, China
| | - Xiaodong Ling
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150040, Heilongjiang, China
| | - Jinhong Zhu
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150040, Heilongjiang, China; Biobank, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150040, Heilongjiang, China.
| |
Collapse
|
3
|
Huang X, Qi L, Lu W, Li Z, Li W, Li F. Retracted article: MYCN contributes to the sensitization of acute myelogenous leukemia cells to cisplatin by targeting SRY-box transcription factor 4. Bioengineered 2024; 15:1997697. [PMID: 34709111 PMCID: PMC10841026 DOI: 10.1080/21655979.2021.1997697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 10/20/2022] Open
Abstract
Xianbao Huang, Ling Qi, Wei Lu, Ziye Li, Wuping Li and Fei Li. MYCN contributes to the sensitization of acute myelogenous leukemia cells to cisplatin by targeting SRY-box transcription factor 4. Bioengineered. 2021 Oct. doi: 10.1080/21655979.2021.1997697.Since publication, significant concerns have been raised about the compliance with ethical policies for human research and the integrity of the data reported in the article.When approached for an explanation, the authors provided some original data but were not able to provide all the necessary supporting information. As verifying the validity of published work is core to the scholarly record's integrity, we are retracting the article. All authors listed in this publication have been informed.We have been informed in our decision-making by our editorial policies and the COPE guidelines.The retracted article will remain online to maintain the scholarly record, but it will be digitally watermarked on each page as 'Retracted.'
Collapse
Affiliation(s)
- Xianbao Huang
- Department of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, China
| | - Ling Qi
- Department of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, China
| | - Wei Lu
- Department of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, China
| | - Ziye Li
- Department of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, China
| | - Wuping Li
- Lymphoma and Myeloma Department, Jiangxi Cancer Hospital, Qingshan Lake District, Nanchang City, Jiangxi Province, China
| | - Fei Li
- Department of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, China
| |
Collapse
|
4
|
Gao R, Zhang X, Chen X, Chen X, Jin L, Zheng H, Yu X. Clinicopathological Characteristics and Prognosis Analysis of Lung Carcinoma With p40/TTF1 Coexpression and Lung Adenosquamous Carcinoma: Lung Carcinoma With p40/TTF1 Coexpression Is a Rare Tumor With High Metastatic Potential. Int J Surg Pathol 2024; 32:1286-1291. [PMID: 38321785 DOI: 10.1177/10668969241229343] [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] [Indexed: 02/08/2024]
Abstract
Background. Lung carcinoma with p40/TTF1 coexpression (LC-PTC) is a very rare tumor with poor prognosis, and few cases have been reported to date. Objectives. To better understand biological behavior and prognosis of LC-PTC. Methods. We collected 9 examples of LC-PTC and compared them with 36 lung adenosquamous carcinomas during the same period in clinicopathologic characteristics, biologic behaviour, and prognosis. Results. Lung carcinoma with p40/TTF1 coexpression mainly occurred in middle-aged and elderly men; 8 tumors belonged to the peripheral type, and 1 belonged to the central type. The rates of lymph node and distant metastasis were 88% (7/8) and 50% (4/8), respectively; 2 patients died during follow-up. Histologically, the LC-PTC showed nest-like growth pattern without glandular growth pattern; the surface of 2 tumors was covered with ciliated columnar epithelium and tumor cells grew under the columnar epithelium. In all patients, tumor cells diffusely coexpressed p40 and TTF1. Although there was no significant difference in the maximum diameter of tumor with lymph node metastasis or with distant metastasis between LC-PTC and lung adenosquamous carcinoma, LC-PTC had a higher rate of lymph node metastasis and distant metastasis. There was no significant difference in overall survival of patients between LC-PTC and lung adenosquamous carcinoma. Additional histologic evaluation of normal pulmonary structures revealed that p40/TTF1 coexpression cells existed in bronchial mucosa and the number of cells coexpressing p40/TTF1 increased gradually from proximal bronchus to distal bronchus. Conclusions. Lung carcinoma with p40/TTF1 coexpression is a rare tumor with high metastatic potential and may originate from p40/TTF1 coexpression cells in distal bronchial mucosa.
Collapse
Affiliation(s)
- Rui Gao
- Department of Pathology, Fujian Provincial Hospital, Provincial Clinical Medical College of Fujian Medical University, P.R. China
| | - Xi Zhang
- Department of Gastroenterology, Fujian Provincial Hospital, Provincial Clinical Medical College of Fujian Medical University, P.R. China
| | - Xiaoyan Chen
- Department of Pathology, Fujian Provincial Hospital, Provincial Clinical Medical College of Fujian Medical University, P.R. China
| | - Xin Chen
- Department of Pathology, Fujian Provincial Hospital, Provincial Clinical Medical College of Fujian Medical University, P.R. China
| | - Long Jin
- Department of Pathology, Fujian Provincial Hospital, Provincial Clinical Medical College of Fujian Medical University, P.R. China
| | - Huawei Zheng
- Department of Pathology, Fujian Provincial Hospital, Provincial Clinical Medical College of Fujian Medical University, P.R. China
| | - Xunbin Yu
- Department of Pathology, Fujian Provincial Hospital, Provincial Clinical Medical College of Fujian Medical University, P.R. China
| |
Collapse
|
5
|
Shen J, Zhao Y, Man Y, Sun X. CLK3 promotes tumor proliferation by activating MYC signaling. Cell Div 2024; 19:28. [PMID: 39289754 PMCID: PMC11409627 DOI: 10.1186/s13008-024-00132-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 09/09/2024] [Indexed: 09/19/2024] Open
Abstract
Colorectal cancer (CRC) ranks among the leading causes of cancer-related mortality worldwide, posing a significant public health challenge. Despite advancements in treatment strategies, prognosis for advanced CRC remains poor. Here, we investigate the role of CLK3 and its interaction with the c-Myc signaling pathway in CRC progression. Our study reveals significant overexpression of CLK3 in CRC tumor tissues, correlating with disease advancement, and demonstrates that CLK3 promotes CRC cell proliferation, mediated by its activation of MYC signaling through upregulation of c-MYC expression. In vivo experiments confirm the oncogenic role of CLK3, with its loss resulting in decreased tumor growth and c-MYC expression. These findings highlight CLK3 as a potential therapeutic target in CRC, offering insights into novel treatment strategies.
Collapse
Affiliation(s)
- Jing Shen
- Department of Neurology, The First Affiliated Hospital of Shihezi University, Shihezi, 832000, Xinjiang, P.R. China
| | - Yu Zhao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Shihezi University, Shihezi, 832000, Xinjiang, P.R. China
| | - Yang Man
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Shihezi University, Shihezi, 832000, Xinjiang, P.R. China
| | - Xuling Sun
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Shihezi University, Shihezi, 832000, Xinjiang, P.R. China.
| |
Collapse
|
6
|
Kuşoğlu A, Örnek D, Dansık A, Uzun C, Nur Özkan S, Sarıca S, Yangın K, Özdinç Ş, Sorhun DT, Solcan N, Doğanalp EC, Arlov Ø, Cunningham K, Karaoğlu IC, Kizilel S, Solaroğlu I, Bulutay P, Fırat P, Erus S, Tanju S, Dilege Ş, Vunjak‐Novakovic G, Tuncbag N, Öztürk E. Extracellular Matrix Sulfation in the Tumor Microenvironment Stimulates Cancer Stemness and Invasiveness. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309966. [PMID: 39083319 PMCID: PMC11423251 DOI: 10.1002/advs.202309966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 07/08/2024] [Indexed: 09/26/2024]
Abstract
Tumor extracellular matrices (ECM) exhibit aberrant changes in composition and mechanics compared to normal tissues. Proteoglycans (PG) are vital regulators of cellular signaling in the ECM with the ability to modulate receptor tyrosine kinase (RTK) activation via their sulfated glycosaminoglycan (sGAG) side chains. However, their role on tumor cell behavior is controversial. Here, it is demonstrated that PGs are heavily expressed in lung adenocarcinoma (LUAD) patients in correlation with invasive phenotype and poor prognosis. A bioengineered human lung tumor model that recapitulates the increase of sGAGs in tumors in an organotypic matrix with independent control of stiffness, viscoelasticity, ligand density, and porosity, is developed. This model reveals that increased sulfation stimulates extensive proliferation, epithelial-mesenchymal transition (EMT), and stemness in cancer cells. The focal adhesion kinase (FAK)-phosphatidylinositol 3-kinase (PI3K) signaling axis is identified as a mediator of sulfation-induced molecular changes in cells upon activation of a distinct set of RTKs within tumor-mimetic hydrogels. The study shows that the transcriptomic landscape of tumor cells in response to increased sulfation resembles native PG-rich patient tumors by employing integrative omics and network modeling approaches.
Collapse
|
7
|
Han Q, Fernandez J, Rajczewski AT, Kono TJY, Weirath NA, Rahim A, Lee AS, Seabloom D, Tretyakova NY. A Multi-Omics Study of Epigenetic Changes in Type II Alveolar Cells of A/J Mice Exposed to Environmental Tobacco Smoke. Int J Mol Sci 2024; 25:9365. [PMID: 39273313 PMCID: PMC11394788 DOI: 10.3390/ijms25179365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Lung cancer remains a major contributor to cancer fatalities, with cigarette smoking known to be responsible for up to 80% of cases. Based on the ability of cigarette smoke to induce inflammation in the lungs and increased lung cancer incidence in smokers with inflammatory conditions such as COPD, we hypothesized that inflammation plays an important role in the carcinogenicity of cigarette smoke. To test this hypothesis, we performed multi-omic analyses of Type II pneumocytes of A/J mice exposed to cigarette smoke for various time periods. We found that cigarette smoke exposure resulted in significant changes in DNA methylation and hydroxymethylation, gene expression patterns, and protein abundance that were partially reversible and contributed to an inflammatory and potentially oncogenic phenotype.
Collapse
Affiliation(s)
- Qiyuan Han
- Department of Biochemistry, Biophysics and Molecular Biology, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA; (Q.H.); (A.T.R.)
| | - Jenna Fernandez
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA; (J.F.); (N.A.W.); (A.R.)
| | - Andrew T. Rajczewski
- Department of Biochemistry, Biophysics and Molecular Biology, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA; (Q.H.); (A.T.R.)
| | - Thomas J. Y. Kono
- Minnesota Supercomputing Institute, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA;
| | - Nicholas A. Weirath
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA; (J.F.); (N.A.W.); (A.R.)
| | - Abdur Rahim
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA; (J.F.); (N.A.W.); (A.R.)
| | - Alexander S. Lee
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Evanston, IL 60611, USA;
| | - Donna Seabloom
- AeroCore Testing Services, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA;
| | - Natalia Y. Tretyakova
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA; (J.F.); (N.A.W.); (A.R.)
| |
Collapse
|
8
|
Casacuberta-Serra S, González-Larreategui Í, Capitán-Leo D, Soucek L. MYC and KRAS cooperation: from historical challenges to therapeutic opportunities in cancer. Signal Transduct Target Ther 2024; 9:205. [PMID: 39164274 PMCID: PMC11336233 DOI: 10.1038/s41392-024-01907-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 06/05/2024] [Accepted: 06/24/2024] [Indexed: 08/22/2024] Open
Abstract
RAS and MYC rank amongst the most commonly altered oncogenes in cancer, with RAS being the most frequently mutated and MYC the most amplified. The cooperative interplay between RAS and MYC constitutes a complex and multifaceted phenomenon, profoundly influencing tumor development. Together and individually, these two oncogenes regulate most, if not all, hallmarks of cancer, including cell death escape, replicative immortality, tumor-associated angiogenesis, cell invasion and metastasis, metabolic adaptation, and immune evasion. Due to their frequent alteration and role in tumorigenesis, MYC and RAS emerge as highly appealing targets in cancer therapy. However, due to their complex nature, both oncogenes have been long considered "undruggable" and, until recently, no drugs directly targeting them had reached the clinic. This review aims to shed light on their complex partnership, with special attention to their active collaboration in fostering an immunosuppressive milieu and driving immunotherapeutic resistance in cancer. Within this review, we also present an update on the different inhibitors targeting RAS and MYC currently undergoing clinical trials, along with their clinical outcomes and the different combination strategies being explored to overcome drug resistance. This recent clinical development suggests a paradigm shift in the long-standing belief of RAS and MYC "undruggability", hinting at a new era in their therapeutic targeting.
Collapse
Affiliation(s)
| | - Íñigo González-Larreategui
- Models of cancer therapies Laboratory, Vall d'Hebron Institute of Oncology, Cellex Centre, Hospital University Vall d'Hebron Campus, Barcelona, Spain
| | - Daniel Capitán-Leo
- Models of cancer therapies Laboratory, Vall d'Hebron Institute of Oncology, Cellex Centre, Hospital University Vall d'Hebron Campus, Barcelona, Spain
| | - Laura Soucek
- Peptomyc S.L., Barcelona, Spain.
- Models of cancer therapies Laboratory, Vall d'Hebron Institute of Oncology, Cellex Centre, Hospital University Vall d'Hebron Campus, Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain.
- Department of Biochemistry and Molecular Biology, Universitat Autonoma de Barcelona, Bellaterra, Spain.
| |
Collapse
|
9
|
Lei T, Shen Z, Shen M, Du L, Shi Y, Peng Y, Zhou Z, Da W, Chen X, Li Q. Clinicopathological and genetic characterization of radiotherapy-induced undifferentiated pleomorphic sarcoma following breast cancer: a case series of three tumors and comprehensive literature review. Diagn Pathol 2024; 19:110. [PMID: 39143618 PMCID: PMC11325744 DOI: 10.1186/s13000-024-01534-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 07/30/2024] [Indexed: 08/16/2024] Open
Abstract
AIMS Compared to primary breast sarcoma (BSs), radiotherapy-induced sarcoma (RIS) is a less frequent type of secondary breast sarcoma. Undifferentiated pleomorphic sarcoma (UPS) is an even rarer occurrence within the RIS category. This study aimed to present the clinicopathologic and molecular features of breast radiotherapy-induced UPS. METHODS A retrospective study was conducted at the Third Affiliated Hospital of Soochow University to analyze three patients with radiation-induced undifferentiated pleomorphic sarcoma (UPS) following breast cancer, spanning from 2006 to 2023. The clinical and pathological variables were extracted from the medical records, while immunohistochemistry was employed to analyze the immunophenotypes of these tumors. Genomic characteristics were assessed through DNA and RNA sequencing techniques. Another 15 cases from the literature were also reviewed to better characterize the tumor. RESULTS The affected areas encompass the chest wall and breasts, with an incubation period ranging from 6 to 17 years. The tumor cells exhibit pleomorphism and demonstrate a high degree of pathological mitosis. Notably, two cases displayed an accelerated disease progression, characterized by recurrent tumors and metastases occurring within short intervals of 48 and 7 months respectively subsequent to the initial diagnosis. The two prevailing identified genes were TP53 (2/3, 66.7%) and RB1 (1/3, 33.3%). Through analysis of somatic copy number variation (CNV), it was discovered that two oncogenes, MCL1 (1/3, 33.3%) and MYC (1/3, 33.3%), had experienced gains in CNV. The Tumor Mutational Burden (TMB) values for case 1, case 2, and case 3 were 5.9 mut/Mb, 1.0 mut/Mb, and 3.0 mut/Mb, respectively. Moreover, the analysis of RNA-NGS (next-generation sequencing) revealed the presence of a novel gene fusion, named COL3A1-GULP1, in case 2. CONCLUSIONS Based on our thorough analysis of research findings and previous reports, it is evident that radiotherapy-induced UPS exhibits a highly diverse and frequently severe clinical and biological behavior. Identifying tumor formation using genome sequencing can help understand its biological behavior and determine personalized treatments.
Collapse
Affiliation(s)
- Ting Lei
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, P.R. China
| | - Zhiyi Shen
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, P.R. China
| | - Mengjia Shen
- Department of Pathology, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, 610041, Sichuan, China
| | - Lingfang Du
- Clinical Medical Research Center, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, P.R. China
| | - Yongqiang Shi
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, P.R. China
| | - Yan Peng
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, P.R. China
| | - Zidi Zhou
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, P.R. China
| | - Wenyue Da
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, P.R. China
| | - Xi Chen
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, P.R. China
| | - Qing Li
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, P.R. China.
| |
Collapse
|
10
|
Wang H, Zhou Y, Lu L, Cen J, Wu Z, Yang B, Zhu C, Cao J, Yu Y, Chen W. Identification of 5-Thiocyanatothiazol-2-amines Disrupting WDR5-MYC Protein-Protein Interactions. ACS Med Chem Lett 2024; 15:1143-1150. [PMID: 39015274 PMCID: PMC11247650 DOI: 10.1021/acsmedchemlett.4c00220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/18/2024] Open
Abstract
MYC amplification is frequently observed in approximately 50% of human cancers, rendering it a highly desired anticancer target. Given the challenge of direct pharmacological inhibiting of MYC, impairing the interaction of MYC and its key cofactor WDR5 has been proposed as a promising strategy for MYC-driven cancer treatment. Herein, we report the discovery of 5-thiocyanatothiazol-2-amines that disrupt the WDR5-MYC interaction. Hit fragments were initially identified in a fluorescence polarization (FP)-based screening of an in-house library, and structural-activity relationship exploration resulted in the lead compounds 4m and 4o with potent inhibitory activities on WDR5-MYC interaction (K i = 2.4 μM for 4m; K i = 1.0 μM for 4o). These compounds were further validated via differential scanning fluorimetry (DSF) and coimmunoprecipitation (Co-IP). Moreover, 4m and 4o exhibited good cellular activities with the IC50 values at the micromolar level (IC50 = 0.71-7.40 μM) against multiple MYC-driven cancer cell lines. Our findings afforded a potential small molecule blocking the WDR5-MYC interaction.
Collapse
Affiliation(s)
- Haiyang Wang
- Zhejiang
Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical
Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yihui Zhou
- Zhejiang
Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical
Sciences, Zhejiang University, Hangzhou 310058, China
- Department
of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Li Lu
- Zhejiang
Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical
Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jie Cen
- Zhejiang
Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical
Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhenying Wu
- Zhejiang
Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical
Sciences, Zhejiang University, Hangzhou 310058, China
| | - Bo Yang
- Zhejiang
Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical
Sciences, Zhejiang University, Hangzhou 310058, China
- Engineering
Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou 310000, China
- Center
for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, Hangzhou 310020, China
| | - Chengliang Zhu
- Zhejiang
Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical
Sciences, Zhejiang University, Hangzhou 310058, China
- Engineering
Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou 310000, China
- Center
for Drug Safety Evaluation and Research of ZJU, Hangzhou 310058, China
- Cancer Center, Zhejiang University, Hangzhou City 310058, China
| | - Ji Cao
- Zhejiang
Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical
Sciences, Zhejiang University, Hangzhou 310058, China
- Engineering
Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou 310000, China
- Center
for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, Hangzhou 310020, China
- Jinhua Institute
of Zhejiang University, Jinhua 321299, China
| | - Yongping Yu
- Zhejiang
Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical
Sciences, Zhejiang University, Hangzhou 310058, China
- School
of Pharmacy, Xinjiang Medical University, Urumqi 830054, China
- Jinhua Institute
of Zhejiang University, Jinhua 321299, China
| | - Wenteng Chen
- Zhejiang
Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical
Sciences, Zhejiang University, Hangzhou 310058, China
- Jinhua Institute
of Zhejiang University, Jinhua 321299, China
| |
Collapse
|
11
|
Nishio Y, Kato K, Oishi H, Takahashi Y, Saitoh S. MYCN in human development and diseases. Front Oncol 2024; 14:1417607. [PMID: 38884091 PMCID: PMC11176553 DOI: 10.3389/fonc.2024.1417607] [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: 04/15/2024] [Accepted: 05/15/2024] [Indexed: 06/18/2024] Open
Abstract
Somatic mutations in MYCN have been identified across various tumors, playing pivotal roles in tumorigenesis, tumor progression, and unfavorable prognoses. Despite its established notoriety as an oncogenic driver, there is a growing interest in exploring the involvement of MYCN in human development. While MYCN variants have traditionally been associated with Feingold syndrome type 1, recent discoveries highlight gain-of-function variants, specifically p.(Thr58Met) and p.(Pro60Leu), as the cause for megalencephaly-polydactyly syndrome. The elucidation of cellular and murine analytical data from both loss-of-function (Feingold syndrome model) and gain-of-function models (megalencephaly-polydactyly syndrome model) is significantly contributing to a comprehensive understanding of the physiological role of MYCN in human development and pathogenesis. This review discusses the MYCN's functional implications for human development by reviewing the clinical characteristics of these distinct syndromes, Feingold syndrome, and megalencephaly-polydactyly syndrome, providing valuable insights into the understanding of pathophysiological backgrounds of other syndromes associated with the MYCN pathway and the overall comprehension of MYCN's role in human development.
Collapse
Affiliation(s)
- Yosuke Nishio
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Genetics, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Kohji Kato
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Genetics, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Hisashi Oishi
- Department of Comparative and Experimental Medicine, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yoshiyuki Takahashi
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinji Saitoh
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
12
|
Liu S, Liu Y, Qiu X, Suhail Y, Kshitiz. Tissue-of-origin for cancers determines HIF-1 activation induced phenotypic heterogeneity. Mol Carcinog 2024; 63:834-848. [PMID: 38372346 PMCID: PMC11013563 DOI: 10.1002/mc.23691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 01/02/2024] [Accepted: 01/16/2024] [Indexed: 02/20/2024]
Abstract
Hypoxia-inducible factor-1 (HIF-1) is the master regulator of cellular response to hypoxia, and is activated in many cancers contributing to many steps in the metastatic cascade by acting as a key transcription co-regulator for a large number of downstream genes. Presence of hypoxia within a tumor is spatially nonuniform, and can also by dynamic. Further, although HIF-1 is primarily stabilized and activated by lack of molecular O2, its stability is also affected by other factors present in the tumor microenvironment. HIF-1 also crosstalks with other transcription factors in co-regulating gene expression. Consequently, it is nontrivial to predict the gene expression patterns in cells in response to hypoxia, or HIF-1 activation. Additionally, cancers originating from tissue origins with different basal level of partial oxygen tension may activate HIF-1 at different threshold of hypoxia. We analyzed large published single cell RNAseq data for colorectal, lung, and pancreatic cancers to investigate the phenotypic outcome of HIF-1 activation in cancer cells. We found that cancers from tissues with different partial O2 tension levels exhibit HIF-1 activation at different stages of metastasis, and phenotypically respond differently to HIF-1 activation, likely by contextual co-option of different transcription factors. We experimentally confirmed these predictions by using cell lines representative of colorectal, lung, and pancreatic cancers, finding that while hypoxia enhances growth of colorectal cancer, it induces increased invasion of lung, and pancreatic cancers. Our analysis suggest that HIF-1 activation may act as a rheostat regulating downstream gene expression towards phenotypic outcomes differently in various cancers.
Collapse
Affiliation(s)
- Shaofei Liu
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, Connecticut, USA
| | - Yamin Liu
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, Connecticut, USA
| | - Xihua Qiu
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, Connecticut, USA
| | - Yasir Suhail
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, Connecticut, USA
| | - Kshitiz
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, Connecticut, USA
- Center for Cell Analysis and Modeling, University of Connecticut Health, Farmington, Connecticut, USA
| |
Collapse
|
13
|
Chen L, Hu M, Chen L, Peng Y, Zhang C, Wang X, Li X, Yao Y, Song Q, Li J, Pei H. Targeting O-GlcNAcylation in cancer therapeutic resistance: The sugar Saga continues. Cancer Lett 2024; 588:216742. [PMID: 38401884 DOI: 10.1016/j.canlet.2024.216742] [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: 11/28/2023] [Revised: 02/03/2024] [Accepted: 02/19/2024] [Indexed: 02/26/2024]
Abstract
O-linked-N-acetylglucosaminylation (O-GlcNAcylation), a dynamic post-translational modification (PTM), holds profound implications in controlling various cellular processes such as cell signaling, metabolism, and epigenetic regulation that influence cancer progression and therapeutic resistance. From the therapeutic perspective, O-GlcNAc modulates drug efflux, targeting and metabolism. By integrating signals from glucose, lipid, amino acid, and nucleotide metabolic pathways, O-GlcNAc acts as a nutrient sensor and transmits signals to exerts its function on genome stability, epithelial-mesenchymal transition (EMT), cell stemness, cell apoptosis, autophagy, cell cycle. O-GlcNAc also attends to tumor microenvironment (TME) and the immune response. At present, several strategies aiming at targeting O-GlcNAcylation are under mostly preclinical evaluation, where the newly developed O-GlcNAcylation inhibitors markedly enhance therapeutic efficacy. Here we systematically outline the mechanisms through which O-GlcNAcylation influences therapy resistance and deliberate on the prospects and challenges associated with targeting O-GlcNAcylation in future cancer treatments.
Collapse
Affiliation(s)
- Lulu Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, 20057, USA.
| | - Mengxue Hu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Luojun Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yihan Peng
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - Cai Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xin Wang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiangpan Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yi Yao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qibin Song
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jing Li
- Beijing Key Laboratory of DNA Damage Response and College of Life Sciences, Capital Normal University, Beijing, 100048, China.
| | - Huadong Pei
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, 20057, USA.
| |
Collapse
|
14
|
Li R, He J, Ni Z, Zhang J, Chi X, Kang C, Li Z, Li X. Mining and exploration of rehabilitation nursing targets for colorectal cancer. Aging (Albany NY) 2024; 16:7022-7042. [PMID: 38637125 PMCID: PMC11087124 DOI: 10.18632/aging.205739] [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: 09/28/2023] [Accepted: 11/20/2023] [Indexed: 04/20/2024]
Abstract
BACKGROUND There are often subtle early symptoms of colorectal cancer, a common malignancy of the intestinal tract. However, it is not yet clear how MYC and NCAPG2 are involved in colorectal cancer. METHOD We obtained colorectal cancer datasets GSE32323 and GSE113513 from the Gene Expression Omnibus (GEO). After downloading, we identified differentially expressed genes (DEGs) and performed Weighted Gene Co-expression Network Analysis (WGCNA). We then undertook functional enrichment assay, gene set enrichment assay (GSEA) and immune infiltration assay. Protein-protein interaction (PPI) network construction and analysis were undertaken. Survival analysis and Comparative Toxicogenomics Database (CTD) analysis were conducted. A gene expression heat map was generated. We used TargetScan to identify miRNAs that are regulators of DEGs. RESULTS 1117 DEGs were identified. Their predominant enrichment in activities like the cellular phase of the cell cycle, in cell proliferation, in nuclear and cytoplasmic localisation and in binding to protein-containing complexes was revealed by Gene Ontology (GO). When the enrichment data from GSE32323 and GSE113513 colon cancer datasets were merged, the primary enriched DEGs were linked to the cell cycle, protein complex, cell cycle control, calcium signalling and P53 signalling pathways. In particular, MYC, MAD2L1, CENPF, UBE2C, NUF2 and NCAPG2 were identified as highly expressed in colorectal cancer samples. Comparative Toxicogenomics Database (CTD) demonstrated that the core genes were implicated in the following processes: colorectal neoplasia, tumour cell transformation, inflammation and necrosis. CONCLUSIONS High MYC and NCAPG2 expression has been observed in colorectal cancer, and increased MYC and NCAPG2 expression correlates with worse prognosis.
Collapse
Affiliation(s)
- Ruipu Li
- Gastrointestinal Rehabilitation Center, Beijing Rehabilitation Hospital Affiliated to Capital Medical University, Shijingshan 100144, Beijing, China
| | - Jie He
- Department of Colorectal Surgery, China Aerospace Science and Industry Corporation 731 Hospital, Fengtai, Beijing, China
| | - Zhijie Ni
- Department of Colorectal Surgery, China Aerospace Science and Industry Corporation 731 Hospital, Fengtai, Beijing, China
| | - Jie Zhang
- Gastrointestinal Rehabilitation Center, Beijing Rehabilitation Hospital Affiliated to Capital Medical University, Shijingshan 100144, Beijing, China
| | - Xiaoqian Chi
- Gastrointestinal Rehabilitation Center, Beijing Rehabilitation Hospital Affiliated to Capital Medical University, Shijingshan 100144, Beijing, China
| | - Chunbo Kang
- Gastrointestinal Rehabilitation Center, Beijing Rehabilitation Hospital Affiliated to Capital Medical University, Shijingshan 100144, Beijing, China
| | - Zhongbo Li
- Department of Colorectal Surgery, China Aerospace Science and Industry Corporation 731 Hospital, Fengtai, Beijing, China
| | - Xubin Li
- Gastrointestinal Rehabilitation Center, Beijing Rehabilitation Hospital Affiliated to Capital Medical University, Shijingshan 100144, Beijing, China
| |
Collapse
|
15
|
Zacarías-Fluck MF, Soucek L, Whitfield JR. MYC: there is more to it than cancer. Front Cell Dev Biol 2024; 12:1342872. [PMID: 38510176 PMCID: PMC10952043 DOI: 10.3389/fcell.2024.1342872] [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: 11/22/2023] [Accepted: 02/20/2024] [Indexed: 03/22/2024] Open
Abstract
MYC is a pleiotropic transcription factor involved in multiple cellular processes. While its mechanism of action and targets are not completely elucidated, it has a fundamental role in cellular proliferation, differentiation, metabolism, ribogenesis, and bone and vascular development. Over 4 decades of research and some 10,000 publications linking it to tumorigenesis (by searching PubMed for "MYC oncogene") have led to MYC becoming a most-wanted target for the treatment of cancer, where many of MYC's physiological functions become co-opted for tumour initiation and maintenance. In this context, an abundance of reviews describes strategies for potentially targeting MYC in the oncology field. However, its multiple roles in different aspects of cellular biology suggest that it may also play a role in many additional diseases, and other publications are indeed linking MYC to pathologies beyond cancer. Here, we review these physiological functions and the current literature linking MYC to non-oncological diseases. The intense efforts towards developing MYC inhibitors as a cancer therapy will potentially have huge implications for the treatment of other diseases. In addition, with a complementary approach, we discuss some diseases and conditions where MYC appears to play a protective role and hence its increased expression or activation could be therapeutic.
Collapse
Affiliation(s)
- Mariano F. Zacarías-Fluck
- Models of Cancer Therapies Laboratory, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Laura Soucek
- Models of Cancer Therapies Laboratory, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Peptomyc S.L., Barcelona, Spain
| | - Jonathan R. Whitfield
- Models of Cancer Therapies Laboratory, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| |
Collapse
|
16
|
Ye W, Fan J, Wu W, Chen Z, Huang Q, Qian L. Effects of fecal microbiota transplantation on metabolic health of DBA mice. Front Microbiol 2024; 15:1352555. [PMID: 38444807 PMCID: PMC10912182 DOI: 10.3389/fmicb.2024.1352555] [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: 12/08/2023] [Accepted: 01/31/2024] [Indexed: 03/07/2024] Open
Abstract
Introduction Numerous studies have demonstrated that C57BL/6 mice exhibit superior growth rates and overall growth performance compared to DBA mice. To investigate whether this discrepancy in growth performance is linked to the composition of gut microorganisms, we conducted fecal microbiome transplantation (FMT) experiments. Methods Specifically, we transplanted fecal fluids from adult C57BL/6 mice, high-fat C57BL/6 mice, and Wistar rats into weaned DBA mice (0.2mL/d), and subsequently analyzed their gut contents and gene expression through 16S rRNA sequencing and transcriptome sequencing. During the test period, C57BL/6 mice and Wistar rats were provided with a normal diet, and high-fat C57BL/6 mice were provided with a high-fat diet. Results The results of our study revealed that mice receiving FMT from all three donor groups exhibited significantly higher daily weight gain and serum triglyceride (TG) levels compared to mice of CK group. 16S rRNA sequensing unveiled substantial differences in the abundance and function of the gut microbiota between the FMT groups and the CK group. Transcriptome analysis revealed a total of 988 differential genes, consisting of 759 up-regulated genes and 187 down-regulated genes, between the three experimental groups and the CK group. Functional Gene Ontology (GO) annotation suggested that these genes were primarily linked to lipid metabolism, coagulation, and immunity. Pearson correlation analysis was performed on the differential genes and clusters, and it revealed significant correlations, mainly related to processes such as fatty acid metabolism, fat digestion and absorption, and cholesterol metabolism. Discussion In summary, FMT from dominant strains improved the growth performance of DBA mice, including body weight gain, institutional growth, and immune performance. This change may be due to the increase of probiotic content in the intestinal tract by FMT and subsequent alteration of intestinal gene expression. However, the effects of cross-species fecal transplantation on the intestinal flora and gene expression of recipient mice were not significant.
Collapse
Affiliation(s)
- Wenxin Ye
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Jinghui Fan
- Hangzhou Academy of Agricultural Sciences, Hangzhou, China
| | - Wenzi Wu
- Hainan Institute of Zhejiang University, Sanya, China
| | - Zhuo Chen
- Hainan Institute of Zhejiang University, Sanya, China
| | - Qixin Huang
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Lichun Qian
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
17
|
Wang Y, Dong A, Jin M, Li S, Duan Y. TEP RNA: a new frontier for early diagnosis of NSCLC. J Cancer Res Clin Oncol 2024; 150:97. [PMID: 38372784 PMCID: PMC10876732 DOI: 10.1007/s00432-024-05620-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 01/10/2024] [Indexed: 02/20/2024]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is the most common type of lung cancer (LC), which is the leading cause of tumor mortality. In recent years, compared with tissue biopsy, which is the diagnostic gold standard for tumor diagnosis, Liquid biopsy (LB) is considered to be a more minimally invasive, sensitive, and safer alternative or auxiliary diagnostic method. However, the current value of LB in early diagnosis of LC is not ideal, so it is particularly important to study the changes in blood composition during the process of tumorigenesis and find more sensitive biomarkers. PURPOSE Platelets are a type of abundant blood cells that carry a large amount of RNA. In the LC regulatory network, activated platelets play an important role in the process of tumorigenesis, development, and metastasis. In order to identify predictive liquid biopsy biomarkers for the diagnosis of NSCLC, we summarized the development and function of platelets, the interaction between platelets and tumors, the value of TEP RNA in diagnosis, prognosis, and treatment of NSCLC, and the method for detecting TEP RNA of NSCLC in this article. CONCLUSION The application of platelets in the diagnosis and treatment of NSCLC remains at a nascent stage. In addition to the drawbacks of low platelet count and complex experimental processes, the diagnostic accuracy of TEP RNA-seq for cancer in different populations still needs to be improved and validated. At present, a large number of studies have confirmed significant differences in the expression of TEP RNA in platelets between NSCLC patients and healthy individuals. Continuous exploration of the diagnostic value of TEP RNA in NSCLC is of utmost importance. The integration of NSCLC platelet-related markers with other NSCLC markers can improve current tumor diagnosis and prognostic evaluation systems, providing broad prospects in tumor screening, disease monitoring, and prognosis assessment.
Collapse
Affiliation(s)
- Yuan Wang
- Clinical Laboratory, The First Affiliated Hospital of Weifang Medical University (Weifang People's Hospital), Weifang Medical University, Weifang, 261000, Shandong, China
- Department of Clinical Laboratory Science, Weifang Medical University, Weifang, 261000, Shandong, China
| | - Aiping Dong
- Clinical Laboratory, The First Affiliated Hospital of Weifang Medical University (Weifang People's Hospital), Weifang Medical University, Weifang, 261000, Shandong, China
| | - Minhan Jin
- Clinical Laboratory, The First Affiliated Hospital of Weifang Medical University (Weifang People's Hospital), Weifang Medical University, Weifang, 261000, Shandong, China
- Department of Clinical Laboratory Science, Weifang Medical University, Weifang, 261000, Shandong, China
| | - Shirong Li
- Clinical Laboratory, The First Affiliated Hospital of Weifang Medical University (Weifang People's Hospital), Weifang Medical University, Weifang, 261000, Shandong, China.
| | - Yang Duan
- Clinical Laboratory, The First Affiliated Hospital of Weifang Medical University (Weifang People's Hospital), Weifang Medical University, Weifang, 261000, Shandong, China.
| |
Collapse
|
18
|
Hamilton G, Stickler S, Rath B. Bromodomain Protein-directed Agents and MYC in Small Cell Lung Cancer. Curr Cancer Drug Targets 2024; 24:930-940. [PMID: 38275056 DOI: 10.2174/0115680096272757231211113206] [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: 08/16/2023] [Revised: 10/17/2023] [Accepted: 11/13/2023] [Indexed: 01/27/2024]
Abstract
Small cell lung cancer (SCLC) has a dismal prognosis. In addition to the inactivation of the tumor suppressors TP53 and RB1, tumor-promoting MYC and paralogs are frequently overexpressed in this neuroendocrine carcinoma. SCLC exhibits high resistance to second-line chemotherapy and all attempts of novel drugs and targeted therapy have failed so far to achieve superior survival. MYC and paralogs have key roles in the oncogenic process, orchestrating proliferation, apoptosis, differentiation, and metabolism. In SCLC, MYC-L and MYC regulate the neuroendocrine dedifferentiation of SCLC cells from Type A (ASCL1 expression) to the other SCLC subtypes. Targeting MYC to suppress tumor growth is difficult due to the lack of suitable binding pockets and the most advanced miniprotein inhibitor Omomyc exhibits limited efficacy. MYC may be targeted indirectly via the bromodomain (BET) protein BRD4, which activates MYC transcription, by specific BET inhibitors that reduce the expression of this oncogenic driver. Here, novel BET-directed Proteolysis Targeting Chimeras (PROTACs) are discussed that show high antiproliferative activity in SCLC. Particularly, ARV-825, targeting specifically BRD4, exhibits superior cytotoxic effects on SCLC cell lines and may become a valuable adjunct to SCLC combination chemotherapy.
Collapse
Affiliation(s)
- Gerhard Hamilton
- Institute of Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Sandra Stickler
- Institute of Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Barbara Rath
- Institute of Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
19
|
Li S, Xue P, Diao X, Fan QY, Ye K, Tang XM, Liu J, Huang ZY, Tang QH, Jia CY, Xin R, Lv ZW, Liu JB, Ma YS, Fu D. Identification and validation of functional roles for three MYC-associated genes in hepatocellular carcinoma. J Adv Res 2023; 54:133-146. [PMID: 36716957 DOI: 10.1016/j.jare.2023.01.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 11/29/2022] [Accepted: 01/11/2023] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Aberrations in MYC underlie a large proportion of liver hepatocellular carcinoma (LIHC) cases; however, MYC is difficult to target because of its undruggable structure. We aimed to uncover MYC-associated molecular targets to provide new strategies for LIHC treatment. METHODS LIHC transcriptome datasets and clinical information were obtained from The Cancer Genome Atlas. A series of bioinformatics analyses were performed for 370 patients who were stratified based on the median MYC expression level (high-MYC group and low-MYC group). Correlation analysis was performed to determine relationships between the expression of key MYC-associated genes and prognosis, DNA promotor methylation, and immune cell infiltration. Gene ontology and Kyoto Encyclopedia of Genes and Genomes Pathway enrichment analyses were performed to elucidate the functions of these genes in LIHC. Their expression and functions in LIHC were further verified using transgenic mice overexpressing c-Myc under control of the hepatocyte-specific promoter (Alb-Cre). RESULTS AURKB, CCNB2, and CDKN3 were overexpressed in LIHC patients with high MYC expression and were associated with poor prognosis. Upregulation of these 3 genes was significantly correlated with hypomethylated promoter status, advanced T stage, metastasis, and immune cell infiltration in LIHC patients. Functional enrichment analyses indicated that these genes participate in the "p53 signaling pathway" and "cell cycle". Furthermore, RT-PCR and IHC analysis revealed that their mRNA and protein expression levels were upregulated in an Alb-Cre;cMYClsl/- mouse model. Drugs that target these 3 MYC-related genes were identified. CONCLUSION Taken together, our results identify biomarkers of potential utility for managing liver cancer therapy owing to their significance in tumorigenesis, proliferation, and tumor immunity.
Collapse
Affiliation(s)
- Sha Li
- Institute of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong 226631, Jiangsu Province, China; Department of Head and Neck Surgery, Central Laboratory, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan Province, China
| | - Pei Xue
- Division of Gastrointestinal and Colorectal Surgery, Ruijin Hospital, Department of General Surgery, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | - Xun Diao
- Institute of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong 226631, Jiangsu Province, China
| | - Qi-Yu Fan
- Institute of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong 226631, Jiangsu Province, China
| | - Kun Ye
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China; Uro-Oncology Institute of Central South University, Changsha 410011, Hunan Province, China
| | - Xiao-Mei Tang
- General Surgery, Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Jia Liu
- General Surgery, Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Zhong-Yan Huang
- General Surgery, Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Qing-Hai Tang
- Hunan Key Laboratory for Conservation and Utilization of Biological Resources in the Nanyue Mountainous Region and College of Life Sciences and Environment, Hengyang Normal University, Hengyang 421008, Hunan Province, China
| | - Cheng-You Jia
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Rui Xin
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Zhong-Wei Lv
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Ji-Bin Liu
- Institute of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong 226631, Jiangsu Province, China.
| | - Yu-Shui Ma
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Da Fu
- General Surgery, Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
20
|
Kong Y, Liu Y, Li X, Rao M, Li D, Ruan X, Li S, Jiang Z, Zhang Q. Palmitoylation landscapes across human cancers reveal a role of palmitoylation in tumorigenesis. J Transl Med 2023; 21:826. [PMID: 37978524 PMCID: PMC10655258 DOI: 10.1186/s12967-023-04611-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 10/10/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Protein palmitoylation, which is catalyzed by palmitoyl-transferase and de-palmitoyl-transferase, plays a crucial role in various biological processes. However, the landscape and dynamics of protein palmitoylation in human cancers are not well understood. METHODS We utilized 23 palmitoyl-acyltransferases and seven de-palmitoyl-acyltransferases as palmitoylation-related genes for protein palmitoylation analysis. Multiple publicly available datasets were employed to conduct pan-cancer analysis, examining the transcriptome, genomic alterations, clinical outcomes, and correlation with c-Myc (Myc) for palmitoylation-related genes. Real-time quantitative PCR and immunoblotting were performed to assess the expression of palmitoylation-related genes and global protein palmitoylation levels in cancer cells treated with Myc depletion or small molecule inhibitors. Protein docking and drug sensitivity analyses were employed to predict small molecules that target palmitoylation-related genes. RESULTS We identified associations between palmitoylation and cancer subtype, stage, and patient survival. We discovered that abnormal DNA methylation and oncogenic Myc-driven transcriptional regulation synergistically contribute to the dysregulation of palmitoylation-related genes. This dysregulation of palmitoylation was closely correlated with immune infiltration in the tumor microenvironment and the response to immunotherapy. Importantly, dysregulated palmitoylation was found to modulate canonical cancer-related pathways, thus influencing tumorigenesis. To support our findings, we performed a proof-of-concept experiment showing that depletion of Myc led to reduced expression of most palmitoylation-related genes, resulting in decreased global protein palmitoylation levels. Through mass spectrometry and enrichment analyses, we also identified palmitoyl-acyltransferases ZDHHC7 and ZDHHC23 as significant contributors to mTOR signaling, DNA repair, and immune pathways, highlighting their potential roles in tumorigenesis. Additionally, our study explored the potential of three small molecular (BI-2531, etoposide, and piperlongumine) to modulate palmitoylation by targeting the expression or activity of palmitoylation-related genes or enzymes. CONCLUSIONS Overall, our findings underscore the critical role of dysregulated palmitoylation in tumorigenesis and the response to immunotherapy, mediated through classical cancer-related pathways and immune cell infiltration. Additionally, we propose that the aforementioned three small molecule hold promise as potential therapeutics for modulating palmitoylation, thereby offering novel avenues for cancer therapy.
Collapse
Affiliation(s)
- Yue Kong
- Department of Microbiology and Immunology, Basic Medicine College, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangdong, China
- Key Laboratory of Ministry of Education for Viral Pathogenesis and Infection Prevention and Control, Jinan University, Guangzhou, 510632, China
| | - Yugeng Liu
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China
| | - Xianzhe Li
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Menglan Rao
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, 510632, China
| | - Dawei Li
- Zhumadian Central Hospital, Huanghuai University, Zhumadian, 463000, China
| | - Xiaolan Ruan
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, 510632, China
| | - Shanglin Li
- Department of Microbiology and Immunology, Basic Medicine College, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangdong, China
- Key Laboratory of Ministry of Education for Viral Pathogenesis and Infection Prevention and Control, Jinan University, Guangzhou, 510632, China
| | - Zhenyou Jiang
- Department of Microbiology and Immunology, Basic Medicine College, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangdong, China.
- Key Laboratory of Ministry of Education for Viral Pathogenesis and Infection Prevention and Control, Jinan University, Guangzhou, 510632, China.
| | - Qiang Zhang
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, Guangdong, China.
| |
Collapse
|
21
|
Hamilton G, Stickler S, Rath B. Integration of signaling pathway and bromodomain and extra-terminal domain inhibition for the treatment of mutant Kirsten rat sarcoma viral oncogene homolog cancer. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:1027-1038. [PMID: 38023987 PMCID: PMC10651355 DOI: 10.37349/etat.2023.00178] [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: 03/29/2023] [Accepted: 09/04/2023] [Indexed: 12/01/2023] Open
Abstract
Mutant Kirsten rat sarcoma viral oncogene homolog (KRAS) is now a drugable oncogenic driver and the KRAS G12C variant responds clinically to sotorasib and adagrasib that covalently block the cysteine of the active center and inhibit downstream signaling and proliferation. Unfortunately, progression-free survival (PFS) of lung cancer patients is only 5-6 months and no survival advantage has been found for sotorasib in comparison to docetaxel chemotherapy. Increased responses to KRAS inhibitors are tested in combination with the son of sevenless 1 (SOS1) inhibitors, upstream and downstream signaling modulators as well as chemotherapeutics. Some of these approaches are limited by toxicity to normal tissues and by diverse mechanisms of resistance. In essence, most of these attempts are directed to the inhibition of proliferation by impairment of the signal transduction pathways. The final target of KRAS-mediated growth stimulation is MYC in the cell nucleus that stimulates transcription of a host of genes. In detail, MYC alters genomic enhancer and super-enhancers of transcription that are frequently deregulated in cancer. Such enhancers can be targeted by bromodomain and extra-terminal (BET) inhibitors (BETi) or degraders and this review discusses whether integrated SOS1 inhibition and BET targeting of MYC synergizes against mutant KRAS tumor growth. BET degraders in the form of proteolysis-targeting chimeras (PROTACs) combined with BAY-293-mediated SOS1 inhibition revealed marked cytotoxic synergy against mutant KRAS cancer cells and may constitute a promising option for clinical treatment.
Collapse
Affiliation(s)
- Gerhard Hamilton
- Department of Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Sandra Stickler
- Department of Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Barbara Rath
- Department of Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| |
Collapse
|
22
|
Qin X, Chen B. Comprehensive analysis and validation reveal potential MYCN regulatory biomarkers associated with neuroblastoma prognosis. J Biomol Struct Dyn 2023; 41:8902-8917. [PMID: 36300516 DOI: 10.1080/07391102.2022.2138977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 10/16/2022] [Indexed: 10/31/2022]
Abstract
Neuroblastoma (NB) is an embryonic malignant tumor that occurs in the sympathetic nervous system. The treatment results of patients in the high-risk group are poor, and relapse and treatment failure can occur even with multiple combination treatments. The proto-oncogene MYCN is a BHLH Transcription Factor used as an independent prognostic factor for NB. The proportion of MYCN amplification in tumor tissues of high-risk patients reaches 40-50%. Hence, exploring new MYCN target genes is a meaningful approach in developing treatment for high-risk NB patients. The microarray datasets were obtained from Gene Expression Omnibus (GEO), and differentially expressed genes (DEGs) were identified. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and miRPathDB were used for enrichment analysis. STRING and Cytoscape were used to construct a protein-protein interaction (PPI) network and for modular analysis. The miRNet and NetworkAnalyst databases were used to predict and construct gene-miRNA and gene-TFs networks. The R2 database was used for expression, correlation, and prognostic analyses. The diagnostic value of the biomarkers was predicted by ROC analysis, and RT-qPCR was used to validate the identified hub genes. Finally, using specific MYCN siRNA and overexpressing plasmids, the correlation between the identified hub genes and MYCN was investigated. Our results showed that FBXO9, HECW2, MIB2, RNF19B, RNF213, TRIM36, and ZBTB16 are novel biomarkers that affect the prognosis of the NB patients. In addition, FBXO9, RNF19B, and TRIM36 were preliminarily confirmed as potential target genes of MYCN. Overall, FBXO9, HECW2, MIB2, RNF19B, RNF213, TRIM36, and ZBTB16 are expected to become novel biomarkers for the treatment of high-risk NB patients.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Xiuni Qin
- Guangzhou Concord Cancer Center, Guangzhou, China
| | - Bo Chen
- Medical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
23
|
Xiao W, Xu Y, Baak JP, Dai J, Jing L, Zhu H, Gan Y, Zheng S. Network module analysis and molecular docking-based study on the mechanism of astragali radix against non-small cell lung cancer. BMC Complement Med Ther 2023; 23:345. [PMID: 37770919 PMCID: PMC10537544 DOI: 10.1186/s12906-023-04148-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 08/31/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND Most lung cancer patients worldwide (stage IV non-small cell lung cancer, NSCLC) have a poor survival: 25%-30% patients die < 3 months. Yet, of those surviving > 3 months, 10%-15% patients survive (very) long. Astragali radix (AR) is an effective traditional Chinese medicine widely used for non-small cell lung cancer (NSCLC). However, the pharmacological mechanisms of AR on NSCLC remain to be elucidated. METHODS Ultra Performance Liquid Chromatography system coupled with Q-Orbitrap HRMS (UPLC-Q-Orbitrap HRMS) was performed for the qualitative analysis of AR components. Then, network module analysis and molecular docking-based approach was conducted to explore underlying mechanisms of AR on NSCLC. The target genes of AR were obtained from four databases including TCMSP (Traditional Chinese Medicine Systems Pharmacology) database, ETCM (The Encyclopedia of TCM) database, HERB (A high-throughput experiment- and reference-guided database of TCM) database and BATMAN-TCM (a Bioinformatics Analysis Tool for Molecular mechanism of TCM) database. NSCLC related genes were screened by GEO (Gene Expression Omnibus) database. The STRING database was used for protein interaction network construction (PIN) of AR-NSCLC shared target genes. The critical PIN were further constructed based on the topological properties of network nodes. Afterwards the hub genes and network modules were analyzed, and enrichment analysis were employed by the R package clusterProfiler. The Autodock Vina was utilized for molecular docking, and the Gromacs was utilized for molecular dynamics simulations Furthermore, the survival analysis was performed based on TCGA (The Cancer Genome Atlas) database. RESULTS Seventy-seven AR components absorbed in blood were obtained. The critical network was constructed with 1447 nodes and 28,890 edges. Based on topological analysis, 6 hub target genes and 7 functional modules were gained. were obtained including TP53, SRC, UBC, CTNNB1, EP300, and RELA. After module analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis showed that AR may exert therapeutic effects on NSCLC by regulating JAK-STAT signaling pathway, PI3K-AKT signaling pathway, ErbB signaling pathway, as well as NFkB signaling pathway. After the intersection calculation of the hub targets and the proteins participated in the above pathways, TP53, SRC, EP300, and RELA were obtained. These proteins had good docking affinity with astragaloside IV. Furthermore, RELA was associated with poor prognosis of NSCLC patients. CONCLUSIONS This study could provide chemical component information references for further researches. The potential pharmacological mechanisms of AR on NSCLC were elucidated, promoting the clinical application of AR in treating NSCLC. RELA was selected as a promising candidate biomarker affecting the prognosis of NSCLC patients.
Collapse
Affiliation(s)
- Wenke Xiao
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yaxin Xu
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jan P Baak
- Stavanger University Hospital, Stavanger, 4068, Norway
- Dr. Med Jan Baak AS, Tananger, 4056, Norway
| | - Jinrong Dai
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Lijia Jing
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Hongxia Zhu
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yanxiong Gan
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Shichao Zheng
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
24
|
Sharifi S, Pakdel A, Pakdel MH, Tabashiri R, Bakhtiarizadeh MR, Tahmasebi A. Integrated co-expression analysis of regulatory elements (miRNA, lncRNA, and TFs) in bovine monocytes induced by Str. uberis. Sci Rep 2023; 13:15076. [PMID: 37699972 PMCID: PMC10497586 DOI: 10.1038/s41598-023-42067-4] [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: 04/18/2023] [Accepted: 09/05/2023] [Indexed: 09/14/2023] Open
Abstract
Non-coding RNAs, including long non-coding RNAs (lncRNAs) and microRNAs (miRNAs), together with transcription factors, are critical pre-, co-, and post-transcriptional regulators. In addition to their criteria as ideal biomarkers, they have great potential in disease prognosis, diagnosis, and treatment of complex diseases. Investigation of regulatory mechanisms in the context of bovine mastitis, as most common and economic disease in the dairy industry, to identify elements influencing the expression of candidate genes as key regulators of the mammary immune response is not yet fully understood. Transcriptome profiles (50 RNA-Seq and 50 miRNA-Seq samples) of bovine monocytes induced by Str. uberis were used for co-expression module detection and preservation analysis using the weighted gene co-expression network analysis (WGCNA) approach. Assigned mi-, lnc-, and m-modules used to construct the integrated regulatory networks and miRNA-lncRNA-mRNA regulatory sub-networks. Remarkably, we have identified 18 miRNAs, five lncRNAs, and seven TFs as key regulators of str. uberis-induced mastitis. Most of the genes introduced here, mainly involved in immune response, inflammation, and apoptosis, were new to mastitis. These findings may help to further elucidate the underlying mechanisms of bovine mastitis, and the discovered genes may serve as signatures for early diagnosis and treatment of the disease.
Collapse
Affiliation(s)
- Somayeh Sharifi
- Department of Animal Science, College of Agriculture, Isfahan University of Technology, Isfahan, 84156-83111, Islamic Republic of Iran.
| | - Abbas Pakdel
- Department of Animal Science, College of Agriculture, Isfahan University of Technology, Isfahan, 84156-83111, Islamic Republic of Iran.
| | - Mohammad Hossein Pakdel
- Department of Plant Molecular Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Islamic Republic of Iran
| | - Raana Tabashiri
- Agricultural Biotechnology Department, Faculty of Agriculture, Tarbiat Modares University, Tehran, Islamic Republic of Iran
| | - Mohammad Reza Bakhtiarizadeh
- Department of Animal and Poultry Science, College of Aburaihan, University of Tehran, Tehran, 3391653755, Islamic Republic of Iran
| | - Ahmad Tahmasebi
- Institute of Biotechnology, Shiraz University, Shiraz, 71946-84334, Islamic Republic of Iran
| |
Collapse
|
25
|
Zhu K, Xia Y, Tian X, He Y, Zhou J, Han R, Guo H, Song T, Chen L, Tian X. Characterization and therapeutic perspectives of differentiation-inducing therapy in malignant tumors. Front Genet 2023; 14:1271381. [PMID: 37745860 PMCID: PMC10514561 DOI: 10.3389/fgene.2023.1271381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 08/30/2023] [Indexed: 09/26/2023] Open
Abstract
Cancer is a major public health issue globally and is one of the leading causes of death. Although available treatments improve the survival rate of some cases, many advanced tumors are insensitive to these treatments. Cancer cell differentiation reverts the malignant phenotype to its original state and may even induce differentiation into cell types found in other tissues. Leveraging differentiation-inducing therapy in high-grade tumor masses offers a less aggressive strategy to curb tumor progression and heightens chemotherapy sensitivity. Differentiation-inducing therapy has been demonstrated to be effective in a variety of tumor cells. For example, differentiation therapy has become the first choice for acute promyelocytic leukemia, with the cure rate of more than 90%. Although an appealing concept, the mechanism and clinical drugs used in differentiation therapy are still in their nascent stage, warranting further investigation. In this review, we examine the current differentiation-inducing therapeutic approach and discuss the clinical applications as well as the underlying biological basis of differentiation-inducing agents.
Collapse
Affiliation(s)
- Kangwei Zhu
- Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yuren Xia
- Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xindi Tian
- Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yuchao He
- Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jun Zhou
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Chiyoda, Japan
| | - Ruyu Han
- Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Hua Guo
- Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Tianqiang Song
- Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Lu Chen
- Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xiangdong Tian
- Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
26
|
Xu H, Xiao L, Chen Y, Liu Y, Zhang Y, Gao Y, Man S, Yan N, Zhang M. Effect of CDK7 inhibitor on MYCN-amplified retinoblastoma. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2023; 1866:194964. [PMID: 37536559 DOI: 10.1016/j.bbagrm.2023.194964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 07/08/2023] [Accepted: 07/19/2023] [Indexed: 08/05/2023]
Abstract
Retinoblastoma (RB) is a common malignancy that primarily affects pediatric populations. Although a well-known cause of RB is RB1 mutation, MYCN amplification can also lead to the disease, which is a poor prognosis factor. Studies conducted in various tumor types have shown that MYCN inhibition is an effective approach to impede tumor growth. Various indirect approaches have been developed to overcome the difficulty of directly targeting MYCN, such as modulating the super enhancer (SE) upstream of MYCN. The drug used in this study to treat MYCN-amplified RB was THZ1, a CDK7 inhibitor that can effectively suppress transcription by interfering with the activity of SEs. The study findings confirmed the anticancer activity of THZ1 against RB in both in vitro and in vivo experiments. Therapy with THZ1 was found to affect numerous genes in RB according to the RNA-seq analysis. Moreover, the gene expression changes induced by THZ1 treatment were enriched in ribosome, endocytosis, cell cycle, apoptosis, etc. Furthermore, the combined analysis of ChIP-Seq and RNA-seq data suggested a potential role of SEs in regulating the expression of critical transcription factors, such as MYCN, OTX2, and SOX4. Moreover, ChIP-qPCR experiments were conducted to confirm the interaction between MYCN and SEs. In conclusion, THZ1 caused substantial changes in gene transcription in RB, resulting in inhibited cell proliferation, interference with the cell cycle, and increased apoptosis. The efficacy of THZ1 is positively correlated with the degree of MYCN amplification and is likely exerted by interfering with MYCN upstream SEs.
Collapse
Affiliation(s)
- Hanyue Xu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China; Research Laboratory of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Lirong Xiao
- Research Laboratory of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Yi Chen
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China; Research Laboratory of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Yilin Liu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Yifan Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Yuzhu Gao
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Shulei Man
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Naihong Yan
- Research Laboratory of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China.
| | - Ming Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China.
| |
Collapse
|
27
|
Chakraborty S, Coleman C, Manoj P, Demircioglu D, Shah N, de Stanchina E, Rudin CM, Hasson D, Sen T. De Novo and Histologically Transformed Small-Cell Lung Cancer Is Sensitive to Lurbinectedin Treatment Through the Modulation of EMT and NOTCH Signaling Pathways. Clin Cancer Res 2023; 29:3526-3540. [PMID: 37382635 PMCID: PMC10901109 DOI: 10.1158/1078-0432.ccr-23-0471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/21/2023] [Accepted: 06/27/2023] [Indexed: 06/30/2023]
Abstract
PURPOSE Small-cell lung cancer (SCLC) is a high-grade neuroendocrine tumor with dismal prognosis and limited treatment options. Lurbinectedin, conditionally approved as a second-line treatment for metastatic SCLC, drives clinical responses in about 35% of patients, and the overall survival (OS) of those who benefit from it remains very low (∼9.3 months). This finding highlights the need to develop improved mechanistic insight and predictive biomarkers of response. EXPERIMENTAL DESIGN We used human and patient-derived xenograft (PDX)-derived SCLC cell lines to evaluate the effect of lurbinectedin in vitro. We also demonstrate the antitumor effect of lurbinectedin in multiple de novo and transformed SCLC PDX models. Changes in gene and protein expression pre- and post-lurbinectedin treatment was assessed by RNA sequencing and Western blot analysis. RESULTS Lurbinectedin markedly reduced cell viability in the majority of SCLC models with the best response on POU2F3-driven SCLC cells. We further demonstrate that lurbinectedin, either as a single agent or in combination with osimertinib, causes an appreciable antitumor response in multiple models of EGFR-mutant lung adenocarcinoma with histologic transformation to SCLC. Transcriptomic analysis identified induction of apoptosis, repression of epithelial-mesenchymal transition, modulation of PI3K/AKT, NOTCH signaling associated with lurbinectedin response in de novo, and transformed SCLC models. CONCLUSIONS Our study provides a mechanistic insight into lurbinectedin response in SCLC and the first demonstration that lurbinectedin is a potential therapeutic target after SCLC transformation.
Collapse
Affiliation(s)
- Subhamoy Chakraborty
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Charles Coleman
- Tisch Cancer Institute, Mount Sinai, New York, New York
- Bioinformatics for Next Generation Sequencing (BiNGS) Core, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Parvathy Manoj
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Deniz Demircioglu
- Tisch Cancer Institute, Mount Sinai, New York, New York
- Bioinformatics for Next Generation Sequencing (BiNGS) Core, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Nisargbhai Shah
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elisa de Stanchina
- Antitumor Assessment Core, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Charles M Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Dan Hasson
- Tisch Cancer Institute, Mount Sinai, New York, New York
- Bioinformatics for Next Generation Sequencing (BiNGS) Core, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Triparna Sen
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Tisch Cancer Institute, Mount Sinai, New York, New York
| |
Collapse
|
28
|
Pan Y, van der Watt PJ, Kay SA. E-box binding transcription factors in cancer. Front Oncol 2023; 13:1223208. [PMID: 37601651 PMCID: PMC10437117 DOI: 10.3389/fonc.2023.1223208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 06/27/2023] [Indexed: 08/22/2023] Open
Abstract
E-boxes are important regulatory elements in the eukaryotic genome. Transcription factors can bind to E-boxes through their basic helix-loop-helix or zinc finger domain to regulate gene transcription. E-box-binding transcription factors (EBTFs) are important regulators of development and essential for physiological activities of the cell. The fundamental role of EBTFs in cancer has been highlighted by studies on the canonical oncogene MYC, yet many EBTFs exhibit common features, implying the existence of shared molecular principles of how they are involved in tumorigenesis. A comprehensive analysis of TFs that share the basic function of binding to E-boxes has been lacking. Here, we review the structure of EBTFs, their common features in regulating transcription, their physiological functions, and their mutual regulation. We also discuss their converging functions in cancer biology, their potential to be targeted as a regulatory network, and recent progress in drug development targeting these factors in cancer therapy.
Collapse
Affiliation(s)
- Yuanzhong Pan
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Pauline J. van der Watt
- Division of Medical Biochemistry and Structural Biology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Steve A. Kay
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
29
|
Huang Y, Qian Y, Xing Y, Pei Y, Zhang B, Li T, Pan X, Zhong A, Du J, Zhou T, Shi M. POLRMT over-expression is linked to WNT/beta-catenin signaling, immune infiltration, and unfavorable outcomes in lung adenocarcinoma patients. Cancer Med 2023; 12:15691-15703. [PMID: 37283308 PMCID: PMC10417304 DOI: 10.1002/cam4.6174] [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: 11/09/2022] [Revised: 04/10/2023] [Accepted: 05/19/2023] [Indexed: 06/08/2023] Open
Abstract
BACKGROUND Mitochondrial RNA polymerase (POLRMT) is essential for the expression of mitochondrial genes. In recent studies, POLRMT expression promoted non-small cell cancer cell proliferation in cell lines and xenografts. The present study investigated the impact of POLRMT expression and function on lung adenocarcinoma (LUAD) patients. METHOD Multi-omics data (genomics, transcriptomics, and proteomics) from publicly available databases were used to assess the role of POLRMT expression and function in LUAD. These findings were further verified using cancer tissues from clinical samples. RESULTS POLRMT was over-expressed in LUADs, with mutation frequencies ranging from 1.30% to 5.71%. Over-expression of POLRMT was associated with an abnormal clinicopathological condition resulting in a decreased lifespan. Furthermore, gene sets enrich analysis revealed that POLRMT expression was linked to WNT/beta-catenin signaling; the expression of downstream target genes was positively correlated with POLRMT expression. Also, POLRMT expression was positively correlated with immunosuppressive genes, thereby affecting immune infiltration. CONCLUSION POLRMT is over-expressed in LUAD, thereby impacting patient survival. It is also involved in WNT/beta-catenin signaling and may affect tumor infiltration.
Collapse
Affiliation(s)
- Yongkang Huang
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Yajuan Qian
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Yufei Xing
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Yongjian Pei
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Beilei Zhang
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Ting Li
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Xue Pan
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Anyuan Zhong
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Juan Du
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Tong Zhou
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Minhua Shi
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| |
Collapse
|
30
|
Arshadi A, Tolomeo D, Venuto S, Storlazzi CT. Advancements in Focal Amplification Detection in Tumor/Liquid Biopsies and Emerging Clinical Applications. Genes (Basel) 2023; 14:1304. [PMID: 37372484 PMCID: PMC10298061 DOI: 10.3390/genes14061304] [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: 05/17/2023] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Focal amplifications (FAs) are crucial in cancer research due to their significant diagnostic, prognostic, and therapeutic implications. FAs manifest in various forms, such as episomes, double minute chromosomes, and homogeneously staining regions, arising through different mechanisms and mainly contributing to cancer cell heterogeneity, the leading cause of drug resistance in therapy. Numerous wet-lab, mainly FISH, PCR-based assays, next-generation sequencing, and bioinformatics approaches have been set up to detect FAs, unravel the internal structure of amplicons, assess their chromatin compaction status, and investigate the transcriptional landscape associated with their occurrence in cancer cells. Most of them are tailored for tumor samples, even at the single-cell level. Conversely, very limited approaches have been set up to detect FAs in liquid biopsies. This evidence suggests the need to improve these non-invasive investigations for early tumor detection, monitoring disease progression, and evaluating treatment response. Despite the potential therapeutic implications of FAs, such as, for example, the use of HER2-specific compounds for patients with ERBB2 amplification, challenges remain, including developing selective and effective FA-targeting agents and understanding the molecular mechanisms underlying FA maintenance and replication. This review details a state-of-the-art of FA investigation, with a particular focus on liquid biopsies and single-cell approaches in tumor samples, emphasizing their potential to revolutionize the future diagnosis, prognosis, and treatment of cancer patients.
Collapse
Affiliation(s)
| | | | | | - Clelia Tiziana Storlazzi
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70125 Bari, Italy; (A.A.); (D.T.); (S.V.)
| |
Collapse
|
31
|
Iqbal S, Begum F, Nyamai DW, Jalal N, Shaw P. An Integrated Computational Analysis of High-Risk SNPs in Angiopoietin-like Proteins (ANGPTL3 and ANGPTL8) Reveals Perturbed Protein Dynamics Associated with Cancer. Molecules 2023; 28:4648. [PMID: 37375208 DOI: 10.3390/molecules28124648] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/01/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Angiopoietin-like proteins (ANGPTL) constitute a family of eight proteins (1-8) which play a pivotal role in the regulation of various pathophysiological processes. The current study sought to identify high-risk, "non-synonymous, single-nucleotide polymorphisms" (nsSNPs) in both ANGPTL3 and ANGPTL8 to evaluate the role that these nsSNPs play in various types of cancer. We retrieved a total of 301 nsSNPs from various databases; 79 of these candidates constitute high-risk nsSNPs. Moreover, we identified eleven high-risk nsSNPs that cause various types of cancer: seven candidates for ANGPTL3 (L57H, F295L, L309F, K329M, R332L, S348C, and G409R) and four candidates for ANGPTL8 (P23L, R85W, R138S, and E148D). Protein-protein interaction analysis revealed a strong association of ANGPTL proteins with several tumor-suppressor proteins such as ITGB3, ITGAV, and RASSF5. 'Gene-expression profiling interactive analysis' (GEPIA) showed that expression of ANGPTL3 is significantly downregulated in five cancers: sarcoma (SARC); cholangio carcinoma (CHOL); kidney chromophobe carcinoma (KICH); kidney renal clear cell carcinoma (KIRC); and kidney renal papillary cell carcinoma (KIRP). GEPIA also showed that expression of ANGPTL8 remains downregulated in three cancers: CHOL; glioblastoma (GBM); and breast invasive carcinoma (BRCA). Survival rate analysis indicated that both upregulation and downregulation of ANGPTL3 and ANGPTL8 leads to low survival rates in various types of cancer. Overall, the current study revealed that both ANGPTL3 and ANGPTL8 constitute potential prognostic biomarkers for cancer; moreover, nsSNPs in these proteins might lead to the progression of cancer. However, further in vivo investigation will be helpful to validate the role of these proteins in the biology of cancer.
Collapse
Affiliation(s)
- Sajid Iqbal
- Oujiang Laboratory (Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health), Wenzhou 325000, China
| | - Farida Begum
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
| | - Dorothy Wavinya Nyamai
- Oujiang Laboratory (Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health), Wenzhou 325000, China
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology, Nairobi 00200, Kenya
| | - Nasir Jalal
- Oujiang Laboratory (Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health), Wenzhou 325000, China
| | - Peter Shaw
- Oujiang Laboratory (Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health), Wenzhou 325000, China
| |
Collapse
|
32
|
Yu J, Zhang Y, Zhu H. Pleiotropic effects of cell competition between normal and transformed cells in mammalian cancers. J Cancer Res Clin Oncol 2023; 149:1607-1619. [PMID: 35796779 PMCID: PMC9261164 DOI: 10.1007/s00432-022-04143-6] [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: 04/09/2022] [Accepted: 06/13/2022] [Indexed: 11/23/2022]
Abstract
PURPOSE In the course of tumor progression, cancer clones interact with host normal cells, and these interactions make them under selection pressure all the time. Cell competition, which can eliminate suboptimal cells and optimize organ development via comparison of cell fitness information, is found to take place between host cells and transformed cells in mammals and play important roles in different phases of tumor progression. The aim of this study is to summarize the current knowledge about the roles and corresponding mechanisms of different cell competition interactions between host normal cells and transformed cells involved in mammalian tumor development. METHODS We reviewed the published relevant articles in the Pubmed. RESULTS So far, the role of several cell competition interactions have been well described in the different phases of mammalian tumor genesis and development. While cell competitions for trophic factors and epithelial defense against cancer (EDAC) prevent the emergence of transformed cells and suppress carcinogenesis, fitness-fingerprints-comparison system and Myc supercompetitors promote the local expansion of transformed cells after the early tumor lesion is formatted. In addition, various preclinical tumor-suppression models which based on the molecular mechanisms of these competition interactions show potential clinical value of boosting the fitness of host normal cells. CONCLUSION Cell competition between host and transformed cells has pleiotropic effects in mammalian tumor genesis and development. The clarification of specific molecular mechanisms shed light on novel ideas for the prevention and treatment of cancer.
Collapse
Affiliation(s)
- Jing Yu
- Department of Oral and Maxillofacial Surgery, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, Zhejiang, China
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yamin Zhang
- Department of Oral and Maxillofacial Surgery, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, Zhejiang, China
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Huiyong Zhu
- Department of Oral and Maxillofacial Surgery, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, Zhejiang, China.
| |
Collapse
|
33
|
Chen D, Zhang H, Zhao L, Liu X, Xue S, Wu P, Jiang H. Prognostic value of RILPL2 and its correlation with tumor immune microenvironment and glycolysis in non-small cell lung cancer. Cell Cycle 2023; 22:841-857. [PMID: 36536539 PMCID: PMC10026870 DOI: 10.1080/15384101.2022.2159203] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Rab-interacting lysosomal protein - like 2 (RILPL2) has been reported to be associated with prognosis and tumor biological functions in breast cancer and endometrial carcinoma. However, its expression and functional role in non-small cell lung cancer (NSCLC) remain unclear. The expression and clinical data of lung adenocarcinoma (LUAD) and lung squamous carcinoma (LUSC) were downloaded from the TCGA database. The expression of RILPL2 in NSCLC cell lines was verified by the Western blot. We used online databases and bioinformatics analysis tools to explore its prognostic value, potential biological functions, and correlations with tumor immune microenvironment.The expression of RILPL2 was significantly lower in NSCLC compared with adjacent normal tissues. Low RILPL2 expression was associated with worse overall survival (OS) in NSCLC. The GO analysis showed RILPL2 was comprehensively involved in immune activity. RILPL2 expression was significantly positively correlated with the infiltration levels of B cells, CD8+T cells, CD4+T cells, macrophages, neutrophils, dendritic cells (P < 0.001), and it was also significantly positively correlated with programmed cell death ligand 1 (PD-L1/CD274) (P < 0.001). High RILPL2 expression could predict better immunotherapy response and prognosis in the immunotherapy cohort. The GSEA analysis showed low RILPL2 expression was associated with glycolysis process in LUAD, which was verified in vitro.These results showed RILPL2 expression was correlated with prognosis, tumor microenvironment, and immunotherapy response in NSCLC. Besides, RILPL2 may regulate glycolysis in LUAD.
Collapse
Affiliation(s)
- Dongfang Chen
- Department of Respiratory and Critical Care Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Hongyan Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Lifang Zhao
- Department of Respiratory and Critical Care Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Xueqing Liu
- Department of Respiratory and Critical Care Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Shan Xue
- Department of Respiratory and Critical Care Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Peiling Wu
- Department of Respiratory and Critical Care Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Handong Jiang
- Department of Respiratory and Critical Care Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| |
Collapse
|
34
|
Huang D, Wang X, Qian Y, Wu J, Chen B, Zhang D, Dong F, Li Y. MAX transcriptionally enhances PD-L1 to inhibit CD8+ T cell-mediated killing of lung adenocarcinoma cells. Cell Immunol 2023; 386:104706. [PMID: 36931054 DOI: 10.1016/j.cellimm.2023.104706] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 03/04/2023] [Accepted: 03/04/2023] [Indexed: 03/12/2023]
Abstract
Immune checkpoint blockade (ICB) therapies, such as monoclonal antibodies against the PD-1/PD-L1 immune checkpoint pathway, have been a major breakthrough in the treatment of lung cancer especially lung adenocarcinoma (LUAD), but their effectiveness is limited. High expression of PD-L1 in tumor cells is one of the key reasons evading immune surveillance, yet the mechanisms that regulate PD-L1 expression are not fully understood. By analyzing the chromatin immunoprecipitation sequencing data of MYC-associated X-factor (MAX) based on lung cancer cell lines, we found that the transcriptional regulator MAX is able to bind to the promoter region of the PD-L1 gene. Further, we performed several molecular biology experiments to determine that MAX promotes PD-L1 transcription in LUAD cells, which in turn assists LUAD cells to evade killing by CD8+ T cells, an effect that can be reversed by anti-PD-L1 antibody. In LUAD, the expression of MAX is positively correlated with PD-L1 and the infiltration of CD8+ T cells. Importantly, we further identified that high expression of the MAX/PD-L1 axis is associated with poor overall survival and fist progression of patients with LUAD. Thus, this study sheds light on the mechanism by which MAX inhibits CD8+ T cell-mediated killing of LUAD cells by activating PD-L1 transcription, and MAX may serve as a potential combinatorial target for ICB therapies that block the PD-1/PD-L1 pathway in LUAD.
Collapse
Affiliation(s)
- Dongwei Huang
- Third District of Cadre Ward, General Hospital of Southern Theater Command of PLA, Guangzhou, Guangdong, China
| | - Xueni Wang
- Third District of Cadre Ward, General Hospital of Southern Theater Command of PLA, Guangzhou, Guangdong, China
| | - Yunfeng Qian
- Third District of Cadre Ward, General Hospital of Southern Theater Command of PLA, Guangzhou, Guangdong, China
| | - Jun Wu
- Third District of Cadre Ward, General Hospital of Southern Theater Command of PLA, Guangzhou, Guangdong, China
| | - Binzhuan Chen
- Third District of Cadre Ward, General Hospital of Southern Theater Command of PLA, Guangzhou, Guangdong, China
| | - Deming Zhang
- Third District of Cadre Ward, General Hospital of Southern Theater Command of PLA, Guangzhou, Guangdong, China
| | - Fengying Dong
- Third District of Cadre Ward, General Hospital of Southern Theater Command of PLA, Guangzhou, Guangdong, China.
| | - Yongqiang Li
- Department of Respiratory and Critical Care Medicine, General Hospital of Southern Theater Command of PLA, Guangzhou, Guangdong, China.
| |
Collapse
|
35
|
Geng Q, Zhao H, Zhou Z, Chen X, Zhao H, Chen N, Li Y, Guo C, Yang A, Beer DG, Chen G. CircSMARCA5 silencing impairs cell proliferation and invasion via the miR-17-3p-EGFR signaling in lung adenocarcinoma. Life Sci 2023; 320:121560. [PMID: 36893940 DOI: 10.1016/j.lfs.2023.121560] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 02/15/2023] [Accepted: 03/02/2023] [Indexed: 03/09/2023]
Abstract
AIMS Circular RNAs are widely expressed in various cancers and play important roles in tumorigenesis and tumor progression. The function and mechanism of circSMARCA5 in lung adenocarcinoma however remains unclear. MAIN METHODS QRT-PCR analysis was applied for determining circSMARCA5 expression in lung adenocarcinoma patient tumor tissues and cells. Molecular biological assays were used for investigating the role of circSMARCA5 in lung adenocarcinoma progression. Luciferase reporter and bioinformatics assays were used for identifying the underlying mechanism. KEY FINDINGS In this study, we observed that circSMARCA5 expression was decreased in lung adenocarcinoma tissues but silencing of circSMARCA5 in lung adenocarcinoma cells inhibited cell proliferation, colony formation, migration and invasion. Mechanistically, we found EGFR, c-MYC and p21 were down-regulated upon circSMARCA5 knockdown. MiR-17-3p efficiently down- regulated EGFR expression via directly binding to EGFR mRNA. SIGNIFICANCE These studies suggest that circSMARCA5 functions as an oncogene via targeting miR-17-3p-EGFR axis and may represent a promising therapeutic target for lung adenocarcinoma.
Collapse
Affiliation(s)
- Qianqian Geng
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Department of Surgery, University of Michigan, Ann Arbor, USA
| | - Huijie Zhao
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Zhiqing Zhou
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Xiuyuan Chen
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
| | - Honglin Zhao
- Department of Thoracic Surgery, Tianjin Medical University, Tianjin, China
| | - Nanzheng Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yu Li
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Chunfang Guo
- Department of Surgery, University of Michigan, Ann Arbor, USA
| | - Aimin Yang
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - David G Beer
- Department of Surgery, University of Michigan, Ann Arbor, USA
| | - Guoan Chen
- School of Medicine, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
36
|
Xie J, Ni J, Shi H, Wang K, Ma X, Li W, Peng B. LncRNA SNHG3 enhances BMI1 mRNA stability by binding and regulating c-MYC: Implications for the carcinogenic role of SNHG3 in bladder cancer. Cancer Med 2023; 12:5718-5735. [PMID: 36208024 PMCID: PMC10028137 DOI: 10.1002/cam4.5316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/13/2022] [Accepted: 09/20/2022] [Indexed: 11/06/2022] Open
Abstract
The transformation of nonmuscle-invasive bladder cancer (BLCa) to muscle-invasive type and distant metastasis are the two major threats to patients after surgery. Thus, it is important to identify the key genes of BLCa cell invasion and metastasis. Long noncoding RNA (lncRNA) is a potential clinical tool for cancer diagnosis and treatment. Herein, we verified that lncRNA SNHG3 is upregulated in human BLCa specimens and is proportional to poor clinical prognosis via a combination of bioinformatic analyses and wet bench experiments. Then, we constructed SNHG3 knockdown and overexpression cell models via lentiviral packaging and CRISPR-Cas9 technique. Fluorescence in situ hybridization assay showed that SNHG3 is distributed in both the nucleus and cytoplasm of BLCa cell lines. In vitro assays including CCK-8, EdU, colony formation, wound healing, transwell, and tube formation demonstrated that SNHG3 knockdown and overexpression potently inhibited and enhanced BLCa cell proliferation, migration, invasion, and angiogenesis. In addition, IVIS imaging revealed that SNHG3 knockdown could significantly inhibit M-NSG mice xenograft tumor growth. Next, RNA sequencing, bioinformatics analyses and western blots indicated that SNHG3 could promote c-MYC expression. RNA immunoprecipitation, actinomycin D assay and western blot assays suggested that SNHG3 could also bind c-MYC protein which subsequently facilitate the stabilization of BMI1 mRNA, thus enhancing BMI1 protein level. However, SNHG3 knockdown had a slightly weaker inhibitory effect on BMI1 expression than c-MYC knockdown. Further, in vitro assays demonstrated that BMI1 knockdown could suppress the SNHG3 activation-induced tumor promoting effect in BLCa cells. Overall, this study has provided new insights into the potential implication of lncRNA SNHG3 in the pathogenesis of BLCa. Importantly, SNHG3/c-MYC/BMI1 axis may be a novel target for regulating tumor growth and metastasis in BLCa patients.
Collapse
Affiliation(s)
- Jinbo Xie
- Department of Urology, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Jinliang Ni
- Shanghai Clinical College, Anhui Medical University, Shanghai, China
| | - Huajuan Shi
- Department of Urology, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Keyi Wang
- Department of Urology, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaoying Ma
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Wei Li
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bo Peng
- Department of Urology, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
37
|
Antigene MYCN Silencing by BGA002 Inhibits SCLC Progression Blocking mTOR Pathway and Overcomes Multidrug Resistance. Cancers (Basel) 2023; 15:cancers15030990. [PMID: 36765949 PMCID: PMC9913109 DOI: 10.3390/cancers15030990] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 01/27/2023] [Accepted: 01/30/2023] [Indexed: 02/08/2023] Open
Abstract
Small-cell lung cancer (SCLC) is the most aggressive lung cancer type, and is associated with smoking, low survival rate due to high vascularization, metastasis and drug resistance. Alterations in MYC family members are biomarkers of poor prognosis for a large number of SCLC. In particular, MYCN alterations define SCLC cases with immunotherapy failure. MYCN has a highly restricted pattern of expression in normal cells and is an ideal target for cancer therapy but is undruggable by traditional approaches. We propose an innovative approach to MYCN inhibition by an MYCN-specific antigene-PNA oligonucleotide (BGA002)-as a new precision medicine for MYCN-related SCLC. We found that BGA002 profoundly and specifically inhibited MYCN expression in SCLC cells, leading to cell-growth inhibition and apoptosis, while also overcoming multidrug resistance. These effects are driven by mTOR pathway block in concomitance with autophagy reactivation, thus avoiding the side effects of targeting mTOR in healthy cells. Moreover, we identified an MYCN-related SCLC gene signature comprehending CNTFR, DLX5 and TNFAIP3, that was reverted by BGA002. Finally, systemic treatment with BGA002 significantly increased survival in MYCN-amplified SCLC mouse models, including in a multidrug-resistant model in which tumor vascularization was also eliminated. These findings warrant the clinical testing of BGA002 in MYCN-related SCLC.
Collapse
|
38
|
Weber LI, Hartl M. Strategies to target the cancer driver MYC in tumor cells. Front Oncol 2023; 13:1142111. [PMID: 36969025 PMCID: PMC10032378 DOI: 10.3389/fonc.2023.1142111] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 02/10/2023] [Indexed: 03/29/2023] Open
Abstract
The MYC oncoprotein functions as a master regulator of cellular transcription and executes non-transcriptional tasks relevant to DNA replication and cell cycle regulation, thereby interacting with multiple proteins. MYC is required for fundamental cellular processes triggering proliferation, growth, differentiation, or apoptosis and also represents a major cancer driver being aberrantly activated in most human tumors. Due to its non-enzymatic biochemical functions and largely unstructured surface, MYC has remained difficult for specific inhibitor compounds to directly address, and consequently, alternative approaches leading to indirect MYC inhibition have evolved. Nowadays, multiple organic compounds, nucleic acids, or peptides specifically interfering with MYC activities are in preclinical or early-stage clinical studies, but none of them have been approved so far for the pharmacological treatment of cancer patients. In addition, specific and efficient delivery technologies to deliver MYC-inhibiting agents into MYC-dependent tumor cells are just beginning to emerge. In this review, an overview of direct and indirect MYC-inhibiting agents and their modes of MYC inhibition is given. Furthermore, we summarize current possibilities to deliver appropriate drugs into cancer cells containing derailed MYC using viral vectors or appropriate nanoparticles. Finding the right formulation to target MYC-dependent cancers and to achieve a high intracellular concentration of compounds blocking or attenuating oncogenic MYC activities could be as important as the development of novel MYC-inhibiting principles.
Collapse
|
39
|
Liu Y, Cheng W, Xin H, Liu R, Wang Q, Cai W, Peng X, Yang F, Xin H. Nanoparticles advanced from preclinical studies to clinical trials for lung cancer therapy. Cancer Nanotechnol 2023; 14:28. [PMID: 37009262 PMCID: PMC10042676 DOI: 10.1186/s12645-023-00174-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 03/06/2023] [Indexed: 03/30/2023] Open
Abstract
Lung cancer is the leading cause of cancer mortality. As a heterogeneous disease, it has different subtypes and various treatment modalities. In addition to conventional surgery, radiotherapy and chemotherapy, targeted therapy and immunotherapy have also been applied in the clinics. However, drug resistance and systemic toxicity still cannot be avoided. Based on the unique properties of nanoparticles, it provides a new idea for lung cancer therapy, especially for targeted immunotherapy. When nanoparticles are used as carriers of drugs with special physical properties, the nanodrug delivery system ensures the accuracy of targeting and the stability of drugs while increasing the permeability and the aggregation of drugs in tumor tissues, showing good anti-tumor effects. This review introduces the properties of various nanoparticles including polymer nanoparticles, liposome nanoparticles, quantum dots, dendrimers, and gold nanoparticles and their applications in tumor tissues. In addition, the specific application of nanoparticle-based drug delivery for lung cancer therapy in preclinical studies and clinical trials is discussed.
Collapse
Affiliation(s)
- Yifan Liu
- grid.410654.20000 0000 8880 6009Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023 Hubei China
- grid.410654.20000 0000 8880 6009Jingzhou Hospital Affiliated to Yangtze University, Yangtze University, Jingzhou, 434023 Hubei China
| | - Wenxu Cheng
- grid.410654.20000 0000 8880 6009Jingzhou Hospital Affiliated to Yangtze University, Yangtze University, Jingzhou, 434023 Hubei China
| | - HongYi Xin
- The Doctoral Scientific Research Center, People’s Hospital of Lianjiang, Guangdong, 524400 China
- grid.410560.60000 0004 1760 3078The Doctoral Scientific Research Center, People’s Hospital of Lianjiang, Affiliated to Guangdong Medical University, Guangdong, 524400 China
| | - Ran Liu
- grid.410654.20000 0000 8880 6009Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023 Hubei China
- grid.410654.20000 0000 8880 6009Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, 434023 Hubei China
| | - Qinqi Wang
- grid.410654.20000 0000 8880 6009Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023 Hubei China
- grid.410654.20000 0000 8880 6009Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, 434023 Hubei China
| | - Wenqi Cai
- grid.49470.3e0000 0001 2331 6153Xinzhou Traditional Chinese Medicine Hospital, Zhongnan Hospital of Wuhan University (Xinzhou), Hubei, 430000 China
| | - Xiaochun Peng
- grid.410654.20000 0000 8880 6009Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023 Hubei China
- grid.410654.20000 0000 8880 6009Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, 434023 Hubei China
| | - Fuyuan Yang
- grid.410654.20000 0000 8880 6009Department of Physiology, School of Basic Medicine, Health Science Center, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023 Hubei China
| | - HongWu Xin
- grid.410654.20000 0000 8880 6009Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023 Hubei China
- grid.410654.20000 0000 8880 6009Department of Biochemistry and Molecular Biology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, 434023 Hubei China
- grid.443353.60000 0004 1798 8916Research Center of Molecular Medicine, Medical College of Chifeng University, Inner Mongolian Autonomous Region, Chifeng, 024000 China
| |
Collapse
|
40
|
Tolomeo D, Traversa D, Venuto S, Ebbesen KK, García Rodríguez JL, Tamma G, Ranieri M, Simonetti G, Ghetti M, Paganelli M, Visci G, Liso A, Kok K, Muscarella LA, Fabrizio FP, Frassanito MA, Lamanuzzi A, Saltarella I, Solimando AG, Fatica A, Ianniello Z, Marsano RM, Palazzo A, Azzariti A, Longo V, Tommasi S, Galetta D, Catino A, Zito A, Mazza T, Napoli A, Martinelli G, Kjems J, Kristensen LS, Vacca A, Storlazzi CT. circPVT1 and PVT1/AKT3 show a role in cell proliferation, apoptosis, and tumor subtype-definition in small cell lung cancer. Genes Chromosomes Cancer 2022; 62:377-391. [PMID: 36562080 DOI: 10.1002/gcc.23121] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/06/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Small cell lung cancer (SCLC) is treated as a homogeneous disease, although the expression of NEUROD1, ASCL1, POU2F3, and YAP1 identifies distinct molecular subtypes. The MYC oncogene, amplified in SCLC, was recently shown to act as a lineage-specific factor to associate subtypes with histological classes. Indeed, MYC-driven SCLCs show a distinct metabolic profile and drug sensitivity. To disentangle their molecular features, we focused on the co-amplified PVT1, frequently overexpressed and originating circular (circRNA) and chimeric RNAs. We analyzed hsa_circ_0001821 (circPVT1) and PVT1/AKT3 (chimPVT1) as examples of such transcripts, respectively, to unveil their tumorigenic contribution to SCLC. In detail, circPVT1 activated a pro-proliferative and anti-apoptotic program when over-expressed in lung cells, and knockdown of chimPVT1 induced a decrease in cell growth and an increase of apoptosis in SCLC in vitro. Moreover, the investigated PVT1 transcripts underlined a functional connection between MYC and YAP1/POU2F3, suggesting that they contribute to the transcriptional landscape associated with MYC amplification. In conclusion, we have uncovered a functional role of circular and chimeric PVT1 transcripts in SCLC; these entities may prove useful as novel biomarkers in MYC-amplified tumors.
Collapse
Affiliation(s)
- Doron Tolomeo
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Debora Traversa
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Santina Venuto
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Karoline K Ebbesen
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, Aarhus, Denmark
| | | | - Grazia Tamma
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Marianna Ranieri
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Giorgia Simonetti
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola (FC), Italy
| | - Martina Ghetti
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola (FC), Italy
| | - Matteo Paganelli
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola (FC), Italy
| | - Grazia Visci
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Arcangelo Liso
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Klaas Kok
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Lucia Anna Muscarella
- Laboratory of Oncology, Fondazione IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo (FG), Italy
| | - Federico Pio Fabrizio
- Laboratory of Oncology, Fondazione IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo (FG), Italy
| | - Maria Antonia Frassanito
- Department of Precision and Regenerative Medicine and Ionian Area - (DiMePRe-J), School of Medicine, Aldo Moro University of Bari, Bari, Italy
| | - Aurelia Lamanuzzi
- Department of Precision and Regenerative Medicine and Ionian Area - (DiMePRe-J), School of Medicine, Aldo Moro University of Bari, Bari, Italy
| | - Ilaria Saltarella
- Department of Precision and Regenerative Medicine and Ionian Area - (DiMePRe-J), School of Medicine, Aldo Moro University of Bari, Bari, Italy
| | - Antonio Giovanni Solimando
- Department of Precision and Regenerative Medicine and Ionian Area - (DiMePRe-J), School of Medicine, Aldo Moro University of Bari, Bari, Italy
| | - Alessandro Fatica
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Zaira Ianniello
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | | | - Antonio Palazzo
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Amalia Azzariti
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Vito Longo
- Medical Thoracic Oncology Unit, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Stefania Tommasi
- Molecular Diagnostics and Pharmacogenetics Unit, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Domenico Galetta
- Medical Thoracic Oncology Unit, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Annamaria Catino
- Medical Thoracic Oncology Unit, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Alfredo Zito
- Pathology Unit, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Tommaso Mazza
- Bioinformatics Unit, Fondazione IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo (FG), Italy
| | - Alessandro Napoli
- Bioinformatics Unit, Fondazione IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo (FG), Italy
| | - Giovanni Martinelli
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola (FC), Italy
| | - Jørgen Kjems
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, Aarhus, Denmark
| | | | - Angelo Vacca
- Department of Precision and Regenerative Medicine and Ionian Area - (DiMePRe-J), School of Medicine, Aldo Moro University of Bari, Bari, Italy
| | - Clelia Tiziana Storlazzi
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
41
|
Eymin B. Inspiratory hyperoxia: a new way to prevent metastasis through metabolism reprogramming in non-small cell lung cancer. Eur Respir J 2022; 60:60/6/2201357. [PMID: 36455962 DOI: 10.1183/13993003.01357-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/01/2022] [Indexed: 12/03/2022]
Affiliation(s)
- Beatrice Eymin
- Univ. Grenoble Alpes, INSERM 1209, CNRS 5309, Institute For Advanced Biosciences, Team RNA Splicing, Cell Signaling and Response to Therapies, Grenoble, France
| |
Collapse
|
42
|
Xu JQ, Fu YL, Zhang J, Zhang KY, Ma J, Tang JY, Zhang ZW, Zhou ZY. Targeting glycolysis in non-small cell lung cancer: Promises and challenges. Front Pharmacol 2022; 13:1037341. [PMID: 36532721 PMCID: PMC9748442 DOI: 10.3389/fphar.2022.1037341] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 11/04/2022] [Indexed: 08/17/2023] Open
Abstract
Metabolic disturbance, particularly of glucose metabolism, is a hallmark of tumors such as non-small cell lung cancer (NSCLC). Cancer cells tend to reprogram a majority of glucose metabolism reactions into glycolysis, even in oxygen-rich environments. Although glycolysis is not an efficient means of ATP production compared to oxidative phosphorylation, the inhibition of tumor glycolysis directly impedes cell survival and growth. This review focuses on research advances in glycolysis in NSCLC and systematically provides an overview of the key enzymes, biomarkers, non-coding RNAs, and signaling pathways that modulate the glycolysis process and, consequently, tumor growth and metastasis in NSCLC. Current medications, therapeutic approaches, and natural products that affect glycolysis in NSCLC are also summarized. We found that the identification of appropriate targets and biomarkers in glycolysis, specifically for NSCLC treatment, is still a challenge at present. However, LDHB, PDK1, MCT2, GLUT1, and PFKM might be promising targets in the treatment of NSCLC or its specific subtypes, and DPPA4, NQO1, GAPDH/MT-CO1, PGC-1α, OTUB2, ISLR, Barx2, OTUB2, and RFP180 might be prognostic predictors of NSCLC. In addition, natural products may serve as promising therapeutic approaches targeting multiple steps in glycolysis metabolism, since natural products always present multi-target properties. The development of metabolic intervention that targets glycolysis, alone or in combination with current therapy, is a potential therapeutic approach in NSCLC treatment. The aim of this review is to describe research patterns and interests concerning the metabolic treatment of NSCLC.
Collapse
Affiliation(s)
- Jia-Qi Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan-Li Fu
- Department of Oncology, Shenzhen (Fu Tian) Hospital, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Jing Zhang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Kai-Yu Zhang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jie Ma
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing-Yi Tang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhi-Wei Zhang
- Department of Oncology, Shenzhen (Fu Tian) Hospital, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Zhong-Yan Zhou
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
43
|
Sturm N, Schuhbaur JS, Hüttner F, Perkhofer L, Ettrich TJ. Gallbladder Cancer: Current Multimodality Treatment Concepts and Future Directions. Cancers (Basel) 2022; 14:5580. [PMID: 36428670 PMCID: PMC9688543 DOI: 10.3390/cancers14225580] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
Gallbladder cancer (GBC) is the most common primary tumor site of biliary tract cancer (BTC), accounting for 0.6% of newly diagnosed cancers and 0.9% of cancer-related deaths. Risk factors, including female sex, age, ethnic background, and chronic inflammation of the gallbladder, have been identified. Surgery is the only curative option for early-stage GBC, but only 10% of patients are primary eligible for curative treatment. After neoadjuvant treatment, up to one-third of locally advanced GBC patients could benefit from secondary surgical treatment. After surgery, only a high-risk subset of patients benefits from adjuvant treatment. For advanced-stage GBC, palliative chemotherapy with gemcitabine and cisplatin is the current standard of care in line with other BTCs. After the failure of gemcitabine and cisplatin, data for second-line treatment in non-resectable GBC is poor, and the only recommended chemotherapy regimen is FOLFOX (5-FU/folinic acid and oxaliplatin). Recent advances with the PD-L1 inhibitor durvalumab open the therapy landscape for immune checkpoint inhibition in GBC. Meanwhile, targeted therapy approaches are a cornerstone of GBC therapy based on molecular profiling and new evidence of molecular differences between different BTC forms and might further improve the prognosis of GBC patients.
Collapse
Affiliation(s)
- Niklas Sturm
- Department of Internal Medicine I, Ulm University Hospital, 89081 Ulm, Germany
| | | | - Felix Hüttner
- Department of General and Visceral Surgery, Ulm University Hospital, 89081 Ulm, Germany
| | - Lukas Perkhofer
- Department of Internal Medicine I, Ulm University Hospital, 89081 Ulm, Germany
| | - Thomas Jens Ettrich
- Department of Internal Medicine I, Ulm University Hospital, 89081 Ulm, Germany
| |
Collapse
|
44
|
Hao L, Chen Q, Chen X, Zhou Q. Integrated analysis of bulk and single-cell RNA-seq reveals the role of MYC signaling in lung adenocarcinoma. Front Genet 2022; 13:1021978. [PMID: 36299592 PMCID: PMC9589149 DOI: 10.3389/fgene.2022.1021978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/26/2022] [Indexed: 11/22/2022] Open
Abstract
MYC is one of the well-known oncogenes, and its important role in cancer still remains largely unknown. We obtained lung adenocarcinoma (LUAD) multi-omics data including genome, transcriptome, and single-cell sequencing data from multiple cohorts. We calculated the GSVA score of the MYC target v1 using the ssGSEA method, and obtained the genes highly correlated with this score by Spearman correlation analysis. Subsequent hierarchical clustering divided these genes into two gene sets highly associated with MYC signaling (S1 and S2). Unsupervised clustering based on these genes divided the LUAD samples into two distinct subgroups, namely, the MYC signaling inhibition group (C1) and activation group (C2). The MCP counter package in R was used to assess tumor immune cell infiltration abundance and ssGSEA was used to calculate gene set scores. The scRNA-seq was used to verify the association of MYC signaling to cell differentiation. We observed significant differences in prognosis, clinical characteristics, immune microenvironment, and genomic alterations between MYC signaling inhibition and MYC signaling activation groups. MYC-signaling is associated with genomic instability and can mediate the immunosuppressive microenvironment and promote cell proliferation, tumor stemness. Moreover, MYC-signaling activation is also subject to complex post-transcriptional regulation and is highly associated with cell differentiation. In conclusion, MYC signaling is closely related to the genomic instability, genetic alteration and regulation, the immune microenvironment landscape, cell differentiation, and disease survival in LUAD. The findings of this study provide a valuable reference to revealing the mechanism of cancer-promoting action of MYC in LUAD.
Collapse
Affiliation(s)
- Lu Hao
- Science and Education Department, Shenzhen Baoan Shiyan People’s Hospital, Shenzhen, China
| | - Qiuyan Chen
- Science and Education Department, Shenzhen Baoan Shiyan People’s Hospital, Shenzhen, China
| | - Xi Chen
- Central Laboratory, The People’s Hospital of Baoan Shenzhen, The Second Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Qing Zhou
- Central Laboratory, The People’s Hospital of Baoan Shenzhen, The Second Affiliated Hospital of Shenzhen University, Shenzhen, China
- *Correspondence: Qing Zhou,
| |
Collapse
|
45
|
Zou J, Lin Y, Hu M, Wan M, Tan X, Xu X, Xu F. N-Myc transcriptionally activates Skp2 to suppress p27 expression in small cell lung cancer. Pathol Res Pract 2022; 238:154083. [PMID: 36027654 DOI: 10.1016/j.prp.2022.154083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 08/12/2022] [Accepted: 08/18/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Small cell lung cancer (SCLC) is characterized by a high proliferative rate, a strong predilection for early metastasis and poor prognosis. Novel SCLC biomarkers are urgently required to improve current diagnostic and treatment modalities. MYCN encodes the proto-oncogene N-Myc that is overexpressed in SCLC, but its downstream effectors are poorly characterized. Here, we investigated the role of the N-Myc/Skp2/p27 axis during SCLC progression. METHODS Immunohistochemistry (IHC) and western blotting were performed to evaluate N-Myc/Skp2/p27 expression. SCLC cell apoptosis was investigated through TUNEL staining. Wound healing and transwell assays were performed to detect the migratory and invasive potential of SCLC cells. N-Myc and Skp2 binding was confirmed through luciferase reporter and ChIP assays. Xenograft models were developed to investigate the function of Skp2 during SCLC tumor growth in vivo. RESULTS N-Myc and Skp2 were overexpressed in SCLC, whilst p27 expression was suppressed. Skp2 facilitated SCLC progression by protecting cells from apoptosis and facilitating cell migration and invasion. N-Myc was found to bind to the promoter region of Skp2 to enhance its expression. Skp2 enhanced tumor growth in vivo through the suppression of p27. Skp2 silencing reversed the pro-oncogenic effects of N-myc in SCLC tumors. CONCLUSION We show that N-Myc enhances Skp2 to regulate p27 expression during SCLC progression. We therefore highlight the N-Myc/Skp2/p27 axis as a novel diagnostic and much-needed therapeutic target in SCLC.
Collapse
Affiliation(s)
- Juntao Zou
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 1519, Dongyue Avenue, Dongxin Township, Nanchang County, Nanchang 330006, Jiangxi, China
| | - Yang Lin
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 1519, Dongyue Avenue, Dongxin Township, Nanchang County, Nanchang 330006, Jiangxi, China
| | - Min Hu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 1519, Dongyue Avenue, Dongxin Township, Nanchang County, Nanchang 330006, Jiangxi, China
| | - Mengzhi Wan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 1519, Dongyue Avenue, Dongxin Township, Nanchang County, Nanchang 330006, Jiangxi, China
| | - Xinyu Tan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 1519, Dongyue Avenue, Dongxin Township, Nanchang County, Nanchang 330006, Jiangxi, China
| | - Xinping Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 1519, Dongyue Avenue, Dongxin Township, Nanchang County, Nanchang 330006, Jiangxi, China.
| | - Fei Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 1519, Dongyue Avenue, Dongxin Township, Nanchang County, Nanchang 330006, Jiangxi, China.
| |
Collapse
|
46
|
Wu Z, Han T, Su H, Xuan J, Wang X. Comprehensive analysis of fatty acid and lactate metabolism–related genes for prognosis value, immune infiltration, and therapy in osteosarcoma patients. Front Oncol 2022; 12:934080. [PMID: 36119478 PMCID: PMC9478861 DOI: 10.3389/fonc.2022.934080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/09/2022] [Indexed: 11/13/2022] Open
Abstract
Osteosarcoma is the most frequent bone tumor. Notwithstanding that significant medical progress has been achieved in recent years, the 5-year overall survival of osteosarcoma patients is inferior. Regulation of fatty acids and lactate plays an essential role in cancer metabolism. Therefore, our study aimed to comprehensively assess the fatty acid and lactate metabolism pattern and construct a fatty acid and lactate metabolism–related risk score system to predict prognosis in osteosarcoma patients. Clinical data and RNA expression data were downloaded from the Therapeutically Applicable Research to Generate Effective Treatments (TARGET) and Gene Expression Omnibus (GEO) databases. We used the least absolute shrinkage and selection operator (LASSO) and Cox regression analyses to construct a prognostic risk score model. Relationships between the risk score model and age, gender, tumor microenvironment characteristics, and drug sensitivity were also explored by correlation analysis. We determined the expression levels of prognostic genes in osteosarcoma cells via Western blotting. We developed an unknown fatty acid and lactate metabolism–related risk score system based on three fatty acid and lactate metabolism–related genes (SLC7A7, MYC, and ACSS2). Survival analysis showed that osteosarcoma patients in the low-risk group were likely to have a better survival time than those in the high-risk group. The area under the curve (AUC) value shows that our risk score model performs well in predicting prognosis. Elevated fatty acids and lactate risk scores weaken immune function and the environment of the body, which causes osteosarcoma patients’ poor survival outcomes. In general, the constructed fatty acid and lactate metabolism–related risk score model can offer essential insights into subsequent mechanisms in available research. In addition, our study may provide rational treatment strategies for clinicians based on immune correlation analysis and drug sensitivity in the future.
Collapse
Affiliation(s)
- Zhouwei Wu
- Department of Orthopaedic Surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tao Han
- Department of Orthopaedic Surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Haohan Su
- Department of Orthopaedic Surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiangwei Xuan
- Department of Orthopaedic Surgery, Zhuji Affiliated Hospital of Wenzhou Medical University, Zhuji, China
- *Correspondence: Xinwei Wang, ; Jiangwei Xuan,
| | - Xinwei Wang
- Department of Orthopaedic Surgery, Zhuji Affiliated Hospital of Wenzhou Medical University, Zhuji, China
- *Correspondence: Xinwei Wang, ; Jiangwei Xuan,
| |
Collapse
|
47
|
Liu Y, Guo Q, Yang H, Zhang XW, Feng N, Wang JK, Liu TT, Zeng KW, Tu PF. Allosteric Regulation of IGF2BP1 as a Novel Strategy for the Activation of Tumor Immune Microenvironment. ACS CENTRAL SCIENCE 2022; 8:1102-1115. [PMID: 36032766 PMCID: PMC9413439 DOI: 10.1021/acscentsci.2c00107] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Tumor immune microenvironment (TIME) regulators are promising cancer immunotherapeutic targets. IGF2BP1, as a crucial N 6-methyladenosine (m6A) reader protein, recognizes m6A target transcripts, ultimately leading to cancer development. However, currently, the biological function of IGF2BP1 in regulating the TIME is not well-understood. In this study, we report that IGF2BP1 knockdown induces cancer cell apoptosis, thereby significantly not only activating immune cell infiltration including CD4+, CD8+ T cells, CD56+ NK cells, and F4/80+ macrophage but also decreasing PD-L1 expression in hepatocellular carcinoma (HCC). Then, chemical genetics identifies a small-molecule cucurbitacin B (CuB), which directly targets IGF2BP1 at a unique site (Cys253) in the KH1-2 domains. This leads to a pharmacological allosteric effect to block IGF2BP1 recognition of m6A mRNA targets such as c-MYC, which is highly associated with cell apoptosis and immune response. In vivo, CuB exhibits an obvious anti-HCC effect through inducing apoptosis and subsequently recruits immune cells to tumor microenvironment as well as blocking PD-L1 expression. Collectively, IGF2BP1 may serve as a novel pharmacological allosteric target for anticancer therapeutics via mediating TIME.
Collapse
Affiliation(s)
- Yang Liu
- State
Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical
Sciences, Peking University, Beijing 100191, China
- Center
of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, 49 N. Garden Road, Haidian District, Beijing 100191, China
| | - Qiang Guo
- State
Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical
Sciences, Peking University, Beijing 100191, China
| | - Heng Yang
- State
Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical
Sciences, Peking University, Beijing 100191, China
| | - Xiao-Wen Zhang
- State
Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical
Sciences, Peking University, Beijing 100191, China
| | - Na Feng
- State
Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical
Sciences, Peking University, Beijing 100191, China
| | - Jing-Kang Wang
- State
Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical
Sciences, Peking University, Beijing 100191, China
| | - Ting-Ting Liu
- State
Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical
Sciences, Peking University, Beijing 100191, China
| | - Ke-Wu Zeng
- State
Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical
Sciences, Peking University, Beijing 100191, China
- E-mail: (KW Zeng)
| | - Peng-Fei Tu
- State
Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical
Sciences, Peking University, Beijing 100191, China
- (PF
Tu)
| |
Collapse
|
48
|
Xiang W, Qi W, Li H, Sun J, Dong C, Ou H, Liu B. Palbociclib Induces the Apoptosis of Lung Squamous Cell Carcinoma Cells via RB-Independent STAT3 Phosphorylation. Curr Oncol 2022; 29:5855-5868. [PMID: 36005200 PMCID: PMC9406926 DOI: 10.3390/curroncol29080462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/09/2022] [Accepted: 08/15/2022] [Indexed: 11/16/2022] Open
Abstract
Lung squamous cell carcinoma (LUSC) treatment response is poor and treatment alternatives are limited. Palbociclib, a cyclin-dependent kinase (CDK) 4/6 inhibitor, has recently been approved for hormone receptor-positive breast cancer patients and applied in multiple preclinical models, but its use for LUSC therapy remains elusive. Here, we investigated whether palbociclib induced cell apoptosis and dissected the underlying mechanism in LUSC. We found that palbociclib induced LUSC cell apoptosis through inhibition of Src tyrosine kinase/signal transducers and activators of transcription 3 (STAT3). Interestingly, palbociclib reduced STAT3 signaling in LUSC cells interfered by retinoblastoma tumor-suppressor gene (RB), suggesting that pro-apoptosis effect of palbociclib was independent of classic CDK4/6-RB signaling. Furthermore, palbociclib could suppress IL-1β and IL-6 expression, and therefore blocked Src/STAT3 signaling, which were rescued by either recombinant human IL-1β or IL-6. Moreover, Myc mediated the sensitivity of LUSC cells to palbociclib. Our discoveries demonstrated that palbociclib induces apoptosis of LUSC cells through the Src/STAT3 axis in an RB-independent manner, and provided a reliable experimental basis of clinical studies in LUSC patients.
Collapse
Affiliation(s)
- Wenjing Xiang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Wanchen Qi
- The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510120, China
| | - Huayu Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jia Sun
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Chao Dong
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Haojie Ou
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Bing Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Correspondence:
| |
Collapse
|
49
|
Xiao L, Huang Y, Li Q, Wang S, Ma L, Fan Z, Tang Z, Yuan X, Liu B. Identification of a prognostic classifier based on EMT-related lncRNAs and the function of LINC01138 in tumor progression for lung adenocarcinoma. Front Mol Biosci 2022; 9:976878. [PMID: 36060239 PMCID: PMC9428519 DOI: 10.3389/fmolb.2022.976878] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 07/12/2022] [Indexed: 12/05/2022] Open
Abstract
Purpose: This study aimed to develop a prognostic indicator based on epithelial-mesenchymal transition (EMT)-related long noncoding RNAs (lncRNAs) and explore the function of EMT-related lncRNAs in malignant progression in lung adenocarcinoma (LUAD). Materials and methods: A LUAD dataset was acquired from The Cancer Genome Atlas (TCGA) to identify prognostic EMT-related lncRNAs via differential expression analysis and univariate Cox regression analysis. Least Absolute Shrinkage and Selection Operator (LASSO) Cox regression analysis was utilized for variable selection and model construction. The EMT-related prognostic index (ERPI) was calculated according to the model and served as a classifier to divide LUAD individuals into high-ERPI and low-ERPI groups. A nomogram incorporating ERPI and clinicopathological variables was constructed. TCGA-LUAD, GSE50081, and GSE31210 were used to test the predictive capacity of the ERPI and nomogram. The characteristics of the tumor microenvironment (TME) were evaluated via the ESTIMATE, TIMER, and ssGSEA algorithms. Gene set variation analysis (GSVA) and ssGSEA were used to annotate the functions of the high-ERPI and low-ERPI groups. CCK8, transwell assay, wound-healing assay, and clone formation assay were conducted to clarify the biological functions of prognostic EMT-related lncRNAs. Results: Ninety-seven differentially expressed EMT-related lncRNAs were identified, 15 of which were related to overall survival (OS). A prognostic signature was constructed based on 14 prognostic EMT-related lncRNAs to calculate the ERPI of each patient, and the predictive ability of ERPI was verified in TCGA, GSE50081, and GSE31210. The low-ERPI group survived longer and had a lower percentage of patients in advanced stage than the high-ERPI group. The nomogram had the highest predictive accuracy, followed by ERPI and stage. Patients with low ERPI had higher infiltration degree of immune cells and stronger immune responses than those with high ERPI. A series of in vitro experiments demonstrated that knockdown of LINC01138 dampened variability, proliferation, and motility of A549 and H460 cells. Conclusion: Our study developed a prognostic classifier with robust prognostic performance and clarified the biological functions of LINC01138 in LUAD, aiding in making individual treatments for patients with LUAD and dissecting the mechanism of oncogenesis.
Collapse
Affiliation(s)
- Lingyan Xiao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongbiao Huang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Li
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Ma
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhijie Fan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhe Tang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Zhe Tang, ; Xianglin Yuan, ; Bo Liu,
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Zhe Tang, ; Xianglin Yuan, ; Bo Liu,
| | - Bo Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Zhe Tang, ; Xianglin Yuan, ; Bo Liu,
| |
Collapse
|
50
|
Thege FI, Rupani DN, Barathi BB, Manning SL, Maitra A, Rhim AD, Wörmann SM. A Programmable In Vivo CRISPR Activation Model Elucidates the Oncogenic and Immunosuppressive Functions of MYC in Lung Adenocarcinoma. Cancer Res 2022; 82:2761-2776. [PMID: 35666804 PMCID: PMC9357118 DOI: 10.1158/0008-5472.can-21-4009] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 04/18/2022] [Accepted: 06/01/2022] [Indexed: 02/05/2023]
Abstract
Conventional genetically engineered mouse models (GEMM) are time-consuming, laborious, and offer limited spatiotemporal control. Here, we describe the development of a streamlined platform for in vivo gene activation using CRISPR activation (CRISPRa) technology. Unlike conventional GEMMs, this model system allows for flexible, sustained, and timed activation of one or more target genes using single or pooled lentiviral guides. Myc and Yap1 were used as model oncogenes to demonstrate gene activation in primary pancreatic organoid cultures in vitro and enhanced tumorigenic potential in Myc-activated organoids when transplanted orthotopically in vivo. Implementation of this model as an autochthonous lung cancer model showed that transduction-mediated activation of Myc led to accelerated tumor progression and significantly reduced overall survival relative to nontargeted tumor controls. Furthermore, Myc activation led to the acquisition of an immune suppressive, "cold" tumor microenvironment. Cross-species validation of these results using publicly available RNA/DNA-seq datasets linked MYC to a previously described immunosuppressive molecular subtype in patient tumors, thus identifying a patient cohort that may benefit from combined MYC- and immune-targeted therapies. Overall, this work demonstrates how CRISPRa can be used for rapid functional validation of putative oncogenes and may allow for the identification and evaluation of potential metastatic and oncogenic drivers through competitive screening. SIGNIFICANCE A streamlined platform for programmable CRISPR gene activation enables rapid evaluation and functional validation of putative oncogenes in vivo.
Collapse
Affiliation(s)
- Fredrik I. Thege
- Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA
- CORRESPONDANCE: Fredrik I. Thege, , Sonja M. Wörmann, , MD Anderson Cancer Center, Zayed Building, Z3.2065, 6565 MD Anderson Blvd., Houston, TX 77030, USA
| | - Dhwani N. Rupani
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bhargavi B. Barathi
- Department of Gastroenterology, Hepatology & Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sara L. Manning
- Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Gastroenterology, Hepatology & Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anirban Maitra
- Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA
| | - Andrew D. Rhim
- Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Gastroenterology, Hepatology & Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sonja M. Wörmann
- Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA
- CORRESPONDANCE: Fredrik I. Thege, , Sonja M. Wörmann, , MD Anderson Cancer Center, Zayed Building, Z3.2065, 6565 MD Anderson Blvd., Houston, TX 77030, USA
| |
Collapse
|