1
|
Yu Z, Yang Y. METTL3 as a potential therapeutic target in gastric cancer. Front Oncol 2024; 14:1483435. [PMID: 39678510 PMCID: PMC11638058 DOI: 10.3389/fonc.2024.1483435] [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: 08/20/2024] [Accepted: 10/28/2024] [Indexed: 12/17/2024] Open
Abstract
Gastric cancer (GC) is one of the leading causes of cancer-related death worldwide. N6-methyladenosine (m6A) modification is the most prominent epigenetic modification of eukaryotic mRNAs, and methyltransferase-like 3 (METTL3), a core component of the methyltransferase complex, catalyzes m6A modification. The results of previous studies indicate that the expression level of METTL3 is significantly elevated in gastric cancer tissues and cells. In addition, fluctuations in m6A levels induced by METTL3 are closely associated with the malignant progression of tumors as well as the poor prognosis of patients with gastric cancer. In this review, we focus on the potential mechanism of METTL3 in gastric cancer, and through our analysis, we suggest that targeting METTL3 could be a new therapeutic tool for treating GC.
Collapse
Affiliation(s)
| | - Yang Yang
- The First Affiliated Hospital of Guangxi University Of Chinese Medicine,
Nanning, Guangxi, China
| |
Collapse
|
2
|
Cao L, Weng K, Li L, Lin G, Zhao Y, Gao Y, Huang X, Chen Q, Wang J, Zheng C, Huang C, Xie J, Li P. BATF2 inhibits the stem cell-like properties and chemoresistance of gastric cancer cells through PTEN/AKT/β-catenin pathway. Theranostics 2024; 14:7007-7022. [PMID: 39629124 PMCID: PMC11610130 DOI: 10.7150/thno.98389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 09/23/2024] [Indexed: 12/06/2024] Open
Abstract
Background: Gastric cancer (GC) ranks as the fifth leading cause of cancer mortality, with cancer stem cells (CSCs) playing a critical role in tumor progression and resistance to chemotherapy. Conventional chemotherapy often fails to effectively target these stem cells. BATF2, a tumor suppressor, is known for its role in gastric cancer, but its influence on cancer stem cell-like properties and chemotherapy response remains unclear. Methods: Single-cell RNA sequencing (scRNA-seq) analysis was performed on 9 gastric cancer samples to evaluate the expression and regulatory function of BATF2. In vitro experiments involving cell cultures, tumor cell spheroids, and organoids were conducted to assess BATF2's impact on 5-Fu sensitivity and its interaction with drug transporters and signaling pathways. In vivo studies, including subcutaneous tumor formation assays, immunohistochemistry, and immunoblotting, were used to validate findings. Results: BATF2 was confirmed as a tumor suppressor in gastric cancer through scRNA-seq analysis. Elevated BATF2 expression correlated with improved outcomes from postoperative chemotherapy in GC patients and increased sensitivity to 5-Fu. BATF2 enhanced 5-Fu responsiveness by inhibiting the ABCG2 drug transporter and promoting PTEN stability, which suppressed AKT phosphorylation. This led to reduced nuclear β-catenin levels and decreased expression of stem cell markers CD44, SOX2, and NANOG, ultimately reducing chemoresistance and stem-like properties in GC cells. Conclusions: BATF2 plays a pivotal role in regulating stem-like characteristics and chemoresistance in gastric cancer through the BATF2/PTEN/AKT/ABCG2 pathway. These findings suggest a novel therapeutic strategy targeting BATF2 to enhance chemotherapy effectiveness in gastric cancer treatment.
Collapse
Affiliation(s)
- Longlong Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P. R. China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, Fujian 350001, P. R. China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian 350001, P. R. China
| | - Kai Weng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P. R. China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, Fujian 350001, P. R. China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian 350001, P. R. China
| | - Lujie Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P. R. China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, Fujian 350001, P. R. China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian 350001, P. R. China
| | - Guangtan Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P. R. China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, Fujian 350001, P. R. China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian 350001, P. R. China
| | - Yuxuan Zhao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P. R. China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, Fujian 350001, P. R. China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian 350001, P. R. China
| | - Youxin Gao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P. R. China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, Fujian 350001, P. R. China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian 350001, P. R. China
| | - Xiaobo Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P. R. China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, Fujian 350001, P. R. China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian 350001, P. R. China
| | - Qiyue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P. R. China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, Fujian 350001, P. R. China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian 350001, P. R. China
| | - Jiabin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P. R. China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, Fujian 350001, P. R. China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian 350001, P. R. China
| | - Chaohui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P. R. China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, Fujian 350001, P. R. China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian 350001, P. R. China
| | - Changming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P. R. China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, Fujian 350001, P. R. China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian 350001, P. R. China
| | - Jianwei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P. R. China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, Fujian 350001, P. R. China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian 350001, P. R. China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P. R. China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, Fujian 350001, P. R. China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian 350001, P. R. China
| |
Collapse
|
3
|
Zhao N, Lai C, Wang Y, Dai S, Gu H. Understanding the role of DNA methylation in colorectal cancer: Mechanisms, detection, and clinical significance. Biochim Biophys Acta Rev Cancer 2024; 1879:189096. [PMID: 38499079 DOI: 10.1016/j.bbcan.2024.189096] [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: 10/05/2023] [Revised: 02/18/2024] [Accepted: 03/13/2024] [Indexed: 03/20/2024]
Abstract
Colorectal cancer (CRC) is one of the deadliest malignancies worldwide, ranking third in incidence and second in mortality. Remarkably, early stage localized CRC has a 5-year survival rate of over 90%; in stark contrast, the corresponding 5-year survival rate for metastatic CRC (mCRC) is only 14%. Compounding this problem is the staggering lack of effective therapeutic strategies. Beyond genetic mutations, which have been identified as critical instigators of CRC initiation and progression, the importance of epigenetic modifications, particularly DNA methylation (DNAm), cannot be underestimated, given that DNAm can be used for diagnosis, treatment monitoring and prognostic evaluation. This review addresses the intricate mechanisms governing aberrant DNAm in CRC and its profound impact on critical oncogenic pathways. In addition, a comprehensive review of the various techniques used to detect DNAm alterations in CRC is provided, along with an exploration of the clinical utility of cancer-specific DNAm alterations.
Collapse
Affiliation(s)
- Ningning Zhao
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China; Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, China
| | - Chuanxi Lai
- Division of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
| | - Yunfei Wang
- Zhejiang ShengTing Biotech. Ltd, Hangzhou 310000, China
| | - Sheng Dai
- Division of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China.
| | - Hongcang Gu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China; Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, China.
| |
Collapse
|
4
|
Xu W, Huang Y, Lei Z, Zhou J. miR-939-3p induces sarcoma proliferation and poor prognosis via suppressing BATF2. Front Oncol 2024; 14:1346531. [PMID: 38420020 PMCID: PMC10899471 DOI: 10.3389/fonc.2024.1346531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 01/31/2024] [Indexed: 03/02/2024] Open
Abstract
Background Sarcoma is a rare and aggressive malignancy with poor prognosis, in which oncogene activation and tumor suppressor inactivation are involved. Accumulated studies suggested basic leucine zipper transcription factor ATF-like 2 (BATF2) as a candidate tumor suppressor, but its specific role and mechanism in sarcoma remain unclear. Methods The expression levels of BATF2 and miR-939-3p were evaluated by using human sarcoma samples, cell lines and xenograft mouse models. Bioinformatics analysis, qPCR, Western blot, cell proliferation assay, overexpression plasmid construction, point mutation and dual luciferase reporter assay were utilized to investigate the role and mechanism of miR-939-3p in sarcoma. Results In this study, we demonstrated that the expression of BATF2 was downregulated in human sarcoma tissues and cell lines. The downregulation of BATF2 was negatively associated with the prognosis of sarcoma patients. Subsequent bioinformatic prediction and experimental validations showed that BATF2 expression was reduced by microRNA (miR)-939-3p mimic and increased by miR-939-3p inhibitor. Additionally, miR-939-3p was upregulated in sarcoma tissues and cells, correlating with a poor prognosis of sarcoma patients. Moreover, miR-939-3p overexpression suppressed sarcoma cell proliferation, which was significantly attenuated by the restoration of BATF2, while siRNA-mediated knockdown of BATF2 aggravated the miR-939-3p-induced promotion of sarcoma cell proliferation. Further computational algorithms and dual-luciferase reporter assays demonstrated that miR-939-3p repressed BATF2 expression via directly binding to its 3' untranslated region (3' UTR). Conclusion Collectively, these findings identified miR-939-3p as a novel regulator of BATF2, as well as a prognostic biomarker in sarcoma, and revealed that suppressing miR-939-3p or inducing BATF2 expression may serve as a promising therapeutic strategy against sarcoma.
Collapse
Affiliation(s)
- Wanwen Xu
- Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei, China
| | - Yinghui Huang
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Zengjie Lei
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jie Zhou
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
5
|
Zhang Z, Li B, Wang Z, Yang L, Peng J, Wang H, Wang Y, Hong L. Novel LncRNA LINC02936 Suppresses Ferroptosis and Promotes Tumor Progression by Interacting with SIX1/CP Axis in Endometrial Cancer. Int J Biol Sci 2024; 20:1356-1374. [PMID: 38385087 PMCID: PMC10878161 DOI: 10.7150/ijbs.86256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 01/19/2024] [Indexed: 02/23/2024] Open
Abstract
Endometrial cancer (EC) is a prevalent gynecological malignancy, and metabolic disorders are among its most significant risk factors. Abnormal iron metabolism is associated with the progression of cancer malignancy. Nevertheless, the involvement of iron metabolism in the EC remains uncertain. Ceruloplasmin (CP) functions as a multicopper oxidase and ferroxidase, playing a crucial role in maintaining the metabolic balance between copper and iron. Prior research has demonstrated that the dysregulated expression of CP has important clinical implications in EC. However, the specific underlying molecular mechanisms remains uncertain. This research examined the impact of CP on the malignant advancement of EC by suppressing ferroptosis. Next, we explored the possibility that Long non-coding RNA (lncRNA) LINC02936/SIX1/CP axis may be a key pathway for inhibiting ferroptosis and promoting cancer progression in EC. Mechanistically, SIX1 modulates the expression of CP, whereas LINC02936 interacts with SIX1 and recruits SIX1 to the CP promoter, leading to upregulation of CP, inhibition of ferroptosis, and promotion of EC progression. Administration of a small peptide cloud block the LINC02936-SIX1 interaction, thereby inhibits EC progression by promoting ferroptosis. Altogether, this is the first report on the lncRNA regulation of ferroptosis in EC. Our research enhances the knowledge of the lncRNA-mediated regulation of ferroptosis in EC progression and indicates the potential therapeutic significance of the LINC02936/SIX1/CP axis in treating EC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Li Hong
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province People's Republic of China
| |
Collapse
|
6
|
Ezzeldin S, Osama A, Anwar AM, Mahgoub S, Ahmed EA, Farid N, Zamzam M, El Ghoneimy A, Magdeldin S. Detection of early prognostic biomarkers for metastasis of Ewing's sarcoma in pediatric patients. Life Sci 2023; 334:122237. [PMID: 37926299 DOI: 10.1016/j.lfs.2023.122237] [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: 08/31/2023] [Revised: 10/25/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
AIMS Ewing's Sarcoma is an extremely aggressive tumor in children. The disease is associated with highly metastatic rate, especially at the time of diagnosis, contributing to a lower survival rate and poor prognosis. The study aimed to identify predictive biomarkers for metastatic Ewing's sarcoma through in-depth analysis of the plasma proteome profile of pediatric Ewing's sarcoma patients. MAIN METHODS Plasma samples from Ewing's sarcoma patients and control individuals were profiled using both shotgun and dimethyl-labeled proteomics analysis. Subsequently, Ewing's sarcoma patients were further stratified according to their metastatic state and chemotherapy response. Western blot was used for validation. Univariate and multivariate analyses were performed to determine proteome metastasis predictors. Receiver operating characteristic (ROC) analysis was done to assess the diagnostic significance of the potential plasma Ewing's sarcoma biomarkers. KEY FINDINGS Our results revealed a set of proteins significantly associated with the metastatic Ewing's sarcoma disease profile. These proteins include ceruloplasmin and several immunoglobulins. Additionally, our study disclosed significant differentially expressed proteins in pediatric Ewing's sarcoma, including CD5 antigen-like, clusterin, and dermcidin. Stable isotope dimethyl labeling and western blot further confirmed our results, strengthening the impact of such proteins in disease development. Furthermore, an unbiased ROC curve evaluated and confirmed the predictive power of these biomarker candidates. SIGNIFICANCE This study presented potential empirical predictive circulating biomarkers for determining the disease status of pediatric Ewing's sarcoma, which is vital for early prediction.
Collapse
Affiliation(s)
- Shahd Ezzeldin
- Proteomics and Metabolomics Research Program, Department of Basic Research, Children's Cancer Hospital 57357 Egypt, 11617 Cairo, Egypt
| | - Aya Osama
- Proteomics and Metabolomics Research Program, Department of Basic Research, Children's Cancer Hospital 57357 Egypt, 11617 Cairo, Egypt
| | - Ali Mostafa Anwar
- Proteomics and Metabolomics Research Program, Department of Basic Research, Children's Cancer Hospital 57357 Egypt, 11617 Cairo, Egypt
| | - Sebaey Mahgoub
- Proteomics and Metabolomics Research Program, Department of Basic Research, Children's Cancer Hospital 57357 Egypt, 11617 Cairo, Egypt
| | - Eman A Ahmed
- Proteomics and Metabolomics Research Program, Department of Basic Research, Children's Cancer Hospital 57357 Egypt, 11617 Cairo, Egypt; Department of Pharmacology, Faculty of Veterinary Medicine, Suez Canal University, 41522 Ismailia, Egypt
| | - Nesma Farid
- Clinical Research Program, Department of Basic Research, Children's Cancer Hospital 57357 Egypt, 11617 Cairo, Egypt
| | - Manal Zamzam
- Department of Pediatric Oncology, Children's Cancer Hospital 57357 Egypt, 11617 Cairo, Egypt
| | - Ahmed El Ghoneimy
- Musculoskeletal Tumor Surgery Unit, Children's Cancer Hospital 57357 Egypt, 11617 Cairo, Egypt; Department of Orthopedic Surgery, Faculty of Medicine, Cairo University, 12613 Giza, Egypt
| | - Sameh Magdeldin
- Proteomics and Metabolomics Research Program, Department of Basic Research, Children's Cancer Hospital 57357 Egypt, 11617 Cairo, Egypt; Department of Physiology, Faculty of Veterinary Medicine, Suez Canal University, 41522 Ismailia, Egypt.
| |
Collapse
|
7
|
Zhu L, Guo G, Jin Y, Hu A, Liu Y. IKBKE regulates angiogenesis by modulating VEGF expression and secretion in glioblastoma. Tissue Cell 2023; 84:102180. [PMID: 37573607 DOI: 10.1016/j.tice.2023.102180] [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: 05/13/2023] [Revised: 06/11/2023] [Accepted: 07/20/2023] [Indexed: 08/15/2023]
Abstract
PURPOSE As a noncanonical inflammatory kinase, IKBKE is frequently overexpressed and activated and has been identified as an oncogenic protein in glioblastoma. However, the potential function and underlying mechanism of IKBKE contributing to tumor angiogenesis remain elusive. METHODS First, we analyzed the correlation between IKBKE and VEGF expression in glioma samples by immunohistochemistry (IHC). Second, HUVEC-related assays and Western blot were used to detect the regulatory effect of IKBKE on angiogenesis by modulating VEGF expression. Third, IKBKE depletion could alleviate the influence of VEGF expression on IHC of intracranial glioma model. RESULTS We demonstrate that depletion of IKBKE markedly inhibits tumor growth and angiogenesis in glioblastoma. Mechanistically, IKBKE induces VEGF expression and secretion by regulating AKT/FOXO3a in glioblastoma. CONCLUSIONS This study reveals that IKBKE is a novel oncogenic molecule that induces angiogenesis through the promotion of VEGF expression and highlights the potential of targeting IKBKE for glioblastoma therapy.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Pathology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou 450003, China
| | - Gaochao Guo
- Department of Neurosurgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou 450003, China
| | - Yuwei Jin
- Department of Pathology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou 450003, China
| | - Aixia Hu
- Department of Pathology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou 450003, China.
| | - Yang Liu
- Department of Neurosurgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou 450003, China.
| |
Collapse
|
8
|
Zhou J, Lei Z, Chen J, Liao S, Chen Y, Liu C, Huang S, Li L, Zhang Y, Wang P, Huang Y, Li J, Liang H. Nuclear export of BATF2 enhances colorectal cancer proliferation through binding to CRM1. Clin Transl Med 2023; 13:e1260. [PMID: 37151195 PMCID: PMC10165233 DOI: 10.1002/ctm2.1260] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 04/21/2023] [Accepted: 04/26/2023] [Indexed: 05/09/2023] Open
Abstract
BACKGROUND During the tumourigenesis and development of colorectal cancer (CRC), the inactivation of tumour suppressor genes is closely involved, although detailed molecular mechanisms remain elusive. Accumulating studies, including ours, have demonstrated that basic leucine zipper transcription factor ATF (activating transcription factor)-like 2 (BATF2) is a capable tumour suppressor that localises in the nucleus. However, its different subcellular localisation, potential functions and underlying mechanisms are unclear. METHODS The translocation of BATF2 and its clinical relevance were detected using CRC samples, cell lines and xenograft nude mice. Candidate BATF2-binding proteins were screened using co-immunoprecipitation, quantitative label-free liquid chromatography-tandem mass spectrometry proteomic analysis, Western blotting and immunofluorescence. Recombinant plasmids, point mutations and siRNAs were applied to clarify the binding sites between BATF2 and chromosome region maintenance 1 (CRM1). RESULTS The present study found that BATF2 was mainly localised in the cytoplasm, rather than nucleus, of CRC cells in vitro and in vivo, while cytoplasmic BATF2 expression was inversely correlated with the prognosis of CRC patients. Furthermore, we identified the nuclear export and subsequent ubiquitin-mediated degradation of BATF2 in CRC cells. Mechanistically, a functional nuclear export sequence (any amino acid) was characterised in BATF2 protein, through which BATF2 bound to CRM1 and translocated out of nucleus, ultimately enhancing CRC growth via inducing activator protein 1 (AP-1)/cyclin D1/phosphorylated retinoblastoma protein (pRb) signalling pathway. Additionally, nuclear export of BATF2 can be retarded by the mutation of NES in BATF2 or the knockdown of CRM1, whereas CRM1 expression was negatively associated with nuclear BATF2 expression and the prognosis of CRC patients. CONCLUSION These findings revealed the biological effects and underlying mechanisms of cytoplasmic localisation of BATF2. Furthermore, suppressing nuclear export of BATF2 via mutating its NES region or inhibiting CRM1 expression may serve as a promising therapeutic strategy against CRC.
Collapse
Affiliation(s)
- Jie Zhou
- Department of Oncology and Southwest Cancer CenterSouthwest HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Zengjie Lei
- Department of Medical OncologyAffiliated Jinling HospitalMedical School of Nanjing UniversityNanjingChina
| | - Jianfang Chen
- Department of Oncology and Southwest Cancer CenterSouthwest HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Shengbo Liao
- Department of OtolaryngologyPeople's Hospital of Xishui CountyGuizhouChina
| | - Yanrong Chen
- Department of Oncology and Southwest Cancer CenterSouthwest HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Chengxiang Liu
- Department of Oncology and Southwest Cancer CenterSouthwest HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Shuo Huang
- Department of Oncology and Southwest Cancer CenterSouthwest HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Liuli Li
- Department of Oncology and Southwest Cancer CenterSouthwest HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Yan Zhang
- Department of Oncology and Southwest Cancer CenterSouthwest HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Pei Wang
- Department of Oncology and Southwest Cancer CenterSouthwest HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Yinghui Huang
- Department of NephrologyKey Laboratory for the Prevention and Treatment of Chronic Kidney Disease of ChongqingChongqing Clinical Research Center of Kidney and Urology DiseasesXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Jianjun Li
- Department of Oncology and Southwest Cancer CenterSouthwest HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Houjie Liang
- Department of Oncology and Southwest Cancer CenterSouthwest HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| |
Collapse
|
9
|
Zhang Z, Shen S, Ma J, Qi T, Gao C, Hu X, Han D, Huang Y. Sequential multi-parametric MRI in assessment of the histological subtype and features in the malignant pleural mesothelioma xenografts. Heliyon 2023; 9:e15237. [PMID: 37123972 PMCID: PMC10130770 DOI: 10.1016/j.heliyon.2023.e15237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/05/2023] [Accepted: 03/30/2023] [Indexed: 05/02/2023] Open
Abstract
Objective It is still a challenge to find a noninvasive technique to distinguish the histological subtypes of malignant pleural mesothelioma (MPM) and characterize the development of related histological features. We investigated the potential value of multiparametric MRI in the assessment of the histological subtype and development of histologic features in the MPM xenograft model. Methods MPM xenograft models were developed by injecting tumour cells into the right axillary space of nude mice. The T1, T2, R2*, T2*, apparent diffusion coefficient (ADC), true diffusion coefficient (D), pseudo diffusion coefficient (D*), and perfusion fraction (f) at 14 d, 28 d, and 42 d were measured and compared between the epithelial and biphasic MPM. Correlations between multiparametric MRI parameters and histologic features, including necrotic fraction (NF) and microvessel density (MVD), were analysed. Results This study found that T2, T2* and IVIM-DWI parameters can reflect the spatial and temporal heterogeneity of MPM. Compared to the epithelial MPM, T2 and T2* were higher and ADC, D, D*, and f were lower in the biphasic MPM (P < 0.05). MRI parameters were different in different stages of epithelial and biphasic MPM. Moderate correlations were found between ADC and tumor volume and NF in the epithelial MPM, and there was a correlation between f and tumor volume and NF and MVD in the two groups. Conclusion MRI parameters changed with tumor progression in a xenograft model of MPM. MRI parameters may provide useful biomarkers for evaluating the histological subtype and histological features development of MPM.
Collapse
Affiliation(s)
- Zhenghua Zhang
- Medical Imaging Department, First Affiliated Hospital of Kunming Medical University, Kunming, 650000, China
| | - Shasha Shen
- Medical Imaging Department, First Affiliated Hospital of Kunming Medical University, Kunming, 650000, China
| | - Jiyao Ma
- Medical Imaging Department, First Affiliated Hospital of Kunming Medical University, Kunming, 650000, China
| | - Tianfu Qi
- Medical Imaging Department, First Affiliated Hospital of Kunming Medical University, Kunming, 650000, China
| | - Chao Gao
- Medical Imaging Department, First Affiliated Hospital of Kunming Medical University, Kunming, 650000, China
| | - Xiong Hu
- Pathology Department, First Affiliated Hospital of Kunming Medical University, Kunming, 650000, China
| | - Dan Han
- Medical Imaging Department, First Affiliated Hospital of Kunming Medical University, Kunming, 650000, China
- Corresponding author.
| | - Yilong Huang
- Medical Imaging Department, First Affiliated Hospital of Kunming Medical University, Kunming, 650000, China
| |
Collapse
|
10
|
Yang H, Bao Y, Jin F, Jiang C, Wei Z, Liu Z, Xu Y. Ceruloplasmin inhibits the proliferation, migration and invasion of nasopharyngeal carcinoma cells and is negatively regulated by miR-543. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2022; 41:474-488. [PMID: 35306965 DOI: 10.1080/15257770.2022.2052314] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND Ceruloplasmin (CP), recognized as a member of multicopper oxidase family, is related to the progression of diverse cancers in human beings. This study is designed to clarify the expression characteristics, biological function and related mechanism of CP in nasopharyngeal carcinoma (NPC). METHODS CP expression in NPC tissues and cells was probed by quantitative real-time polymerase chain reaction (qRT-PCR), immunohistochemistry (IHC) and Western blot. After gain-of-function and loss-of-function models were established, cell counting kit-8 (CCK-8), Transwell and BrdU assays were employed to measure cell viability, migration and invasion. The targeting relationship between microRNA-543 (miR-543) and CP was verified by dual-luciferase reporter gene assay. RESULTS As against normal nasopharyngeal epithelial tissues, CP expression was significantly lower in NPC tissues, which was associated with higher clinical stage and the short overall survival time. Compared with the control group, CP overexpression markedly restrained the growth, migration and invasion of NPC cells; knocking down CP had the opposite effect. MiR-543 directly targeted CP and negatively modulated its expression. CONCLUSION CP restrains the growth, migration and invasion of NPC cells and is negatively regulated by miR-543.
Collapse
Affiliation(s)
- Hang Yang
- Department of Otolarynglogy, Jiangshan People's Hospital, Jiangshan, Zhejiang, China
| | - Yangyang Bao
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Fenfen Jin
- Department of Otolarynglogy, Jiangshan People's Hospital, Jiangshan, Zhejiang, China
| | - Chonghan Jiang
- Department of Otolarynglogy, Jiangshan People's Hospital, Jiangshan, Zhejiang, China
| | - Zhanhui Wei
- Department of Otolaryngology, Traditional Chinese Medicine Hospital of Chunan County, Hangzhou, Zhejiang, China
| | - Zhenli Liu
- Department of Neurology, Traditional Chinese Medicine Hospital of Chunan County, Hangzhou, Zhejiang, China
| | - Yaping Xu
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
11
|
Programmable CRISPR-Cas9 microneedle patch for long-term capture and real-time monitoring of universal cell-free DNA. Nat Commun 2022; 13:3999. [PMID: 35810160 PMCID: PMC9271037 DOI: 10.1038/s41467-022-31740-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 06/29/2022] [Indexed: 12/15/2022] Open
Abstract
Recent advances in biointerfaces have led to the development of wearable devices that can provide insights into personal health. As wearable modules, microneedles can extract analytes of interest from interstitial fluid in a minimally invasive fashion. However, some microneedles are limited by their ability to perform highly effective extraction and real-time monitoring for macromolecule biomarkers simultaneously. Here we show the synergetic effect of CRISPR-activated graphene biointerfaces, and report an on-line wearable microneedle patch for extraction and in vivo long-term monitoring of universal cell-free DNA. In this study, this wearable system enables real-time monitoring of Epstein-Barr virus, sepsis, and kidney transplantation cell-free DNA, with anti-interference ability of 60% fetal bovine serum, and has satisfactory stable sensitivity for 10 days in vivo. The experimental results of immunodeficient mouse models shows the feasibility and practicability of this proposed method. This wearable patch holds great promise for long-term in vivo monitoring of cell-free DNA and could potentially be used for early disease screening and prognosis. Real-time sensing of biomarkers via the use of wearable devices is a major aim of personalised medicine. Here, authors demonstrate an on-line wearable microneedle patch for real-time capture and monitoring of universal cell-free DNA.
Collapse
|
12
|
Exploring the Interplay between Metabolism and Tumor Microenvironment Based on Four Major Metabolism Pathways in Colon Adenocarcinoma. JOURNAL OF ONCOLOGY 2022; 2022:2159794. [PMID: 35747126 PMCID: PMC9213191 DOI: 10.1155/2022/2159794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/11/2022] [Accepted: 05/14/2022] [Indexed: 11/17/2022]
Abstract
Tumor metabolism plays a critical role in tumor progression. However, the interaction between metabolism and tumor microenvironment (TME) has not been comprehensively revealed in colon adenocarcinoma (COAD). We used unsupervised consensus clustering to establish three molecular subtypes (clusters) based on the enrichment score of four major metabolism pathways in TCGA-COAD dataset. GSE17536 was used as a validation dataset. Single-cell RNA sequencing data (GSE161277) was employed to further verify the reliability of subtyping and characterize the correlation between metabolism and TME. Three clusters were identified and they performed distinct prognosis and molecular features. Clust3 had the worst overall survival and the highest enrichment score of glycolysis. 86 differentially expressed genes (DEGs) were identified, in which 11 DEGs were associated with favorable prognosis and 75 DEGs were associated with poor prognosis. Striking correlations were observed between hypoxia and glycolysis, clust3 and hypoxia, and clust3 and angiogenesis (P < 0.001).We constructed a molecular subtyping system which was effective and reliable for predicting COAD prognosis. The 86 identified key DEGs may be greatly involved in COAD progression, and they provide new perspectives and directions for further understanding the mechanism of metabolism in promoting aggressive phenotype by interacting with TME.
Collapse
|
13
|
Su R, Jin C, Bu H, Xiang J, Zhou L, Jin C. Development and Validation of an Immune-Related Prognostic Signature in Cervical Cancer. Front Oncol 2022; 12:861392. [PMID: 35651784 PMCID: PMC9148954 DOI: 10.3389/fonc.2022.861392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/08/2022] [Indexed: 11/13/2022] Open
Abstract
Background Cervical cancer is the fourth most frequent gynecological malignancy across the world. Immunotherapies have proved to improve prognosis of cervical cancer. However, few studies on immune-related prognostic signature had been reported in cervical cancer. Methods Raw data and clinical information of cervical cancer samples were downloaded from TCGA and UCSC Xena website. Immunophenoscore of immune infiltration cells in cervical cancer samples was calculated through the ssGSEA method using GSVA package. WGCNA, Cox regression analysis, LASSO analysis, and GSEA analysis were performed to classify cervical cancer prognosis and explore the biological signaling pathway. Results There were eight immune infiltration cells associated with prognosis of cervical cancer. Through WGCNA, 153 genes from 402 immune-related genes were significantly correlated with prognosis of cervical cancer. A 15-gene signature demonstrated powerful predictive ability in prognosis of cervical cancer. GSEA analysis showed multiple signaling pathways containing Programmed cell death ligand-1 (PD-L1) expression and PD-1 checkpoint pathway differences between high-risk and low-risk groups. Furthermore, the 15-gene signature was associated with multiple immune cells and immune infiltration in tumor microenvironment. Conclusion The 15-gene signature is an effective potential prognostic classifier in the immunotherapies and surveillance of cervical cancer.
Collapse
Affiliation(s)
- Rongjia Su
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Gynecologic Oncology, International Peace Maternity and Child Health Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Chengwen Jin
- Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hualei Bu
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, China
| | - Jiangdong Xiang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lina Zhou
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- *Correspondence: Chengjuan Jin, ; Lina Zhou,
| | - Chengjuan Jin
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- *Correspondence: Chengjuan Jin, ; Lina Zhou,
| |
Collapse
|
14
|
Orouji E, Raman AT, Singh AK, Sorokin A, Arslan E, Ghosh AK, Schulz J, Terranova CJ, Jiang S, Tang M, Maitituoheti M, Barrodia P, Jiang Y, Callahan SC, Tomczak KJ, Jiang Z, Davis JS, Ghosh S, Lee HM, Reyes-Uribe L, Chang K, Liu Y, Chen H, Azhdarnia A, Morris JS, Vilar E, Carmon KS, Kopetz S, Rai K. Chromatin state dynamics confers specific therapeutic strategies in enhancer subtypes of colorectal cancer. Gut 2022; 71:938-949. [PMID: 34059508 PMCID: PMC8745382 DOI: 10.1136/gutjnl-2020-322835] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 05/14/2021] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Enhancer aberrations are beginning to emerge as a key epigenetic feature of colorectal cancers (CRC), however, a comprehensive knowledge of chromatin state patterns in tumour progression, heterogeneity of these patterns and imparted therapeutic opportunities remain poorly described. DESIGN We performed comprehensive epigenomic characterisation by mapping 222 chromatin profiles from 69 samples (33 colorectal adenocarcinomas, 4 adenomas, 21 matched normal tissues and 11 colon cancer cell lines) for six histone modification marks: H3K4me3 for Pol II-bound and CpG-rich promoters, H3K4me1 for poised enhancers, H3K27ac for enhancers and transcriptionally active promoters, H3K79me2 for transcribed regions, H3K27me3 for polycomb repressed regions and H3K9me3 for heterochromatin. RESULTS We demonstrate that H3K27ac-marked active enhancer state could distinguish between different stages of CRC progression. By epigenomic editing, we present evidence that gains of tumour-specific enhancers for crucial oncogenes, such as ASCL2 and FZD10, was required for excessive proliferation. Consistently, combination of MEK plus bromodomain inhibition was found to have synergistic effects in CRC patient-derived xenograft models. Probing intertumour heterogeneity, we identified four distinct enhancer subtypes (EPIgenome-based Classification, EpiC), three of which correlate well with previously defined transcriptomic subtypes (consensus molecular subtypes, CMSs). Importantly, CMS2 can be divided into two EpiC subgroups with significant survival differences. Leveraging such correlation, we devised a combinatorial therapeutic strategy of enhancer-blocking bromodomain inhibitors with pathway-specific inhibitors (PARPi, EGFRi, TGFβi, mTORi and SRCi) for EpiC groups. CONCLUSION Our data suggest that the dynamics of active enhancer underlies CRC progression and the patient-specific enhancer patterns can be leveraged for precision combination therapy.
Collapse
Affiliation(s)
- Elias Orouji
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA,Present address: Epigenetics Initiative, Princess Margaret Genomics Centre, Toronto, ON, Canada
| | - Ayush T. Raman
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA,Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX, USA,Present address: Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Anand K. Singh
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alexey Sorokin
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer center, Houston, TX, USA
| | - Emre Arslan
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Archit K. Ghosh
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jonathan Schulz
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher J. Terranova
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shan Jiang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ming Tang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mayinuer Maitituoheti
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Praveen Barrodia
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yingda Jiang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - S. Carson Callahan
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Katarzyna J. Tomczak
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhiqin Jiang
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer center, Houston, TX, USA
| | - Jennifer S. Davis
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sukhen Ghosh
- Center for Translational Cancer Research, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Hey Min Lee
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer center, Houston, TX, USA
| | - Laura Reyes-Uribe
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kyle Chang
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yusha Liu
- Department of Bioinformatics and Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Huiqin Chen
- Department of Bioinformatics and Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ali Azhdarnia
- Center for Translational Cancer Research, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jeffrey S. Morris
- Department of Bioinformatics and Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA,Present address: Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Eduardo Vilar
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kendra S. Carmon
- Center for Translational Cancer Research, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer center, Houston, TX, USA
| | - Kunal Rai
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA .,Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, Texas, USA.,MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
15
|
Li H, Cai E, Cheng H, Ye X, Ma R, Zhu H, Chang X. FGA Controls VEGFA Secretion to Promote Angiogenesis by Activating the VEGFR2-FAK Signalling Pathway. Front Endocrinol (Lausanne) 2022; 13:791860. [PMID: 35498401 PMCID: PMC9043601 DOI: 10.3389/fendo.2022.791860] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 03/15/2022] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Our previous work revealed the high expression of fibrinogen alpha chain (FGA) in patients with endometriosis (EM) and that it could promote the migration and invasion of endometrial stromal cells. Angiogenesis is the key condition for the development of EM. This study was aimed to elucidate the role of FGA in endometrial stromal cells involved in angiogenesis in EM. METHODS Immunohistochemistry was used to detect the microvessel density (MVD) and VEGF expression in the eutopic endometrium samples from EM and non-EM. The conditioned medium (CM) of human primary eutopic endometrial stromal cells (EuESC) and immortalized endometrial stromal cell line hEM15A with FGA knockdown were collected and used to treat human umbilical vein endothelial cells (HUVECs). Then, tube formation assay, EdU assay, wound assay, transwell assay and flow cytometry assays were performed to assess the function of HUEVCs in vitro. The angiogenic capability of HUVECs was further measured using a matrigel plug assay with BALB/c nude mice in vivo. Immunofluorescence was used to detect the expression of F-actin and VE-cadherin. RT-PCR and western blotting were used to detect the expression of angiogenesis-related factors in endometrial stromal cells and downstream signalling pathways in HUVECs. RESULTS MVD and VEGF expression in the eutopic endometrium of EM patients were significantly higher than those in the normal endometrium of non-EM patients, and the increased MVD in EM indicates an increased risk of recurrence. Functionally, we found that CM of endometrial stromal cells with FGA knockdown could inhibit HUEVCs migration and tube formation in vitro and in vivo, while having no significant effect on HUVECs proliferation, apoptosis and cell cycle. Mechanically, the expression of VEGFA, PDGF, FGF-B, VEGF, MMP-2 and MMP-9 was reduced in hEM15A cells with FGA knockdown. CM of hEM15A cells with FGA knockdown reduced the number of microfilaments and pseudopodia, as well as the expression of VE-cadherin, and inhibited the activity of VEGFR2 and the FAK signalling pathway in HUVECs. CONCLUSION Our study demonstrated FGA could enhance the interaction between endometrial stromal cells and HUVECs via the potential VEGA-VEGFR-FAK signalling axis and promote EM angiogenesis, revealing a promising therapeutic approach for EM.
Collapse
Affiliation(s)
- Hui Li
- Department of Obstetrics and Gynaecology, Peking University People’s Hospital, Beijing, China
- Center of Gynaecological Oncology, Peking University People’s Hospital, Beijing, China
| | - E. Cai
- Department of Obstetrics and Gynaecology, Peking University People’s Hospital, Beijing, China
- Center of Gynaecological Oncology, Peking University People’s Hospital, Beijing, China
| | - Hongyan Cheng
- Department of Obstetrics and Gynaecology, Peking University People’s Hospital, Beijing, China
- Center of Gynaecological Oncology, Peking University People’s Hospital, Beijing, China
| | - Xue Ye
- Department of Obstetrics and Gynaecology, Peking University People’s Hospital, Beijing, China
- Center of Gynaecological Oncology, Peking University People’s Hospital, Beijing, China
| | - Ruiqiong Ma
- Department of Obstetrics and Gynaecology, Peking University People’s Hospital, Beijing, China
- Center of Gynaecological Oncology, Peking University People’s Hospital, Beijing, China
| | - Honglan Zhu
- Department of Obstetrics and Gynaecology, Peking University People’s Hospital, Beijing, China
- *Correspondence: Xiaohong Chang, ; Honglan Zhu,
| | - Xiaohong Chang
- Department of Obstetrics and Gynaecology, Peking University People’s Hospital, Beijing, China
- Center of Gynaecological Oncology, Peking University People’s Hospital, Beijing, China
- *Correspondence: Xiaohong Chang, ; Honglan Zhu,
| |
Collapse
|
16
|
SARI inhibits growth and reduces survival of oral squamous cell carcinomas (OSCC) by inducing endoplasmic reticulum stress. Life Sci 2021; 287:120141. [PMID: 34774620 DOI: 10.1016/j.lfs.2021.120141] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 01/14/2023]
Abstract
AIMS SARI (suppressor of activator protein (AP)-1, regulated by interferon (IFN) was identified as a novel tumor suppressor by applying subtraction hybridization to terminally differentiating human melanoma cells. The anti-tumor activity of SARI and the correlation between expression and cancer aggression and metastasis has been examined in multiple cancers, but its potential role in oral squamous cell carcinomas (OSCC) has not been explored. METHODS SARI expression was monitored in tumor tissues of OSCC patients by performing immunohistochemistry. Ectopic expression of SARI was achieved using a replication defective adenovirus expressing SARI (Ad.SARI). A nude mouse xenograft model was used to evaluate the in vivo efficacy of SARI. Endoplasmic reticulum (ER) stress was monitored in SARI infected OSCC cells by confocal microscopy. KEY FINDING In this study, we demonstrate that SARI expression is significantly lower in OSCC tumor tissue as compared to normal adjacent tissue. Ectopic expression of SARI induces cancer-specific cell death in human OSCC cell lines and in a paclitaxel plus cisplatin non-responder OSCC patient-derived (PDC1) cell line. Mechanistically, SARI inhibits zinc finger protein GLI1 expression through induction of endoplasmic reticulum (ER) stress. Using a nude mouse xenograft model, we show that intratumoral injections of Ad.SARI significantly reduce PDC1 tumor burden, whereas treatment with an ER stress inhibitor efficiently rescues tumors from growth inhibition. SIGNIFICANCE Overall, our data provides a link between induction of ER stress and inhibition of the GLI1/Hedgehog signaling pathway and the tumor suppressive activity of SARI in the context of OSCC.
Collapse
|
17
|
Liu P, Huang H, Qi X, Bian C, Cheng M, Liu L, Xue L, Zhao X, Yi T, Quan Y. Hypoxia-Induced LncRNA-MIR210HG Promotes Cancer Progression By Inhibiting HIF-1α Degradation in Ovarian Cancer. Front Oncol 2021; 11:701488. [PMID: 34900667 PMCID: PMC8655245 DOI: 10.3389/fonc.2021.701488] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 11/08/2021] [Indexed: 01/16/2023] Open
Abstract
LncRNA-MIR210HG plays crucial roles in the progression of diverse cancers. However, the expression and function of MIR210HG in ovarian cancer remains unclear. In the present study, we aimed to determine the expression and function of lncRNA-MIR210HG in ovarian cancer under hypoxic conditions. MIR210HG expression in ovarian cancer cells under hypoxic conditions was determined by qPCR analysis, and the distribution was determined by FISH and qPCR analysis based on cell nucleus and cytosol RNA extraction. Epithelial-Mesenchymal Transition (EMT) assay and human umbilical vein endothelial cell-based tube formation and migration assays were employed to determine the potential function of MIR210HG in vitro, followed by establishment of a subcutaneous tumor model in mice. The direct target of MIR210HG was determined by RNA pull-down and western blotting. Furthermore, the expression and clinical correlation of MIR210HG was determined based on malignant tissues from ovarian cancer patients. Our results indicated that MIR210HG was induced by hypoxia, which is HIF-1α dependent and mainly located in the cytosol of ovarian cancer cells. Knockdown of MIR210HG significantly inhibited EMT and tumor angiogenesis in vitro and impaired tumor growth in mice. Molecular investigations indicated that MIR210HG directly targets HIF-1α protein and inhibits VHL-dependent HIF-1α protein degradation in ovarian cancer. Further results demonstrated that MIR210HG was upregulated in ovarian cancer tissues and correlated with tumor progression and poor prognosis of ovarian cancer patients. Our study suggests that hypoxia-induced MIR210HG promotes cancer progression by inhibiting HIF-1α degradation in ovarian cancer, which could be a therapeutic target for ovarian cancer.
Collapse
Affiliation(s)
- Ping Liu
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Huiqiong Huang
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Xiaorong Qi
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Ce Bian
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Meng Cheng
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Lili Liu
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Luqi Xue
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Tao Yi
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Yi Quan
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Luo Y, Yin J, Fang R, Liu J, Wang L, Zhang H, Zhang M, Lei Z, Liang S, Cui W, Zhang Z, Wu K, Hui X. The tumour neovasculature-homing dimeric peptide GX1 demonstrates antiangiogenic activity in the retinal neovasculature. Eur J Pharmacol 2021; 912:174574. [PMID: 34662566 DOI: 10.1016/j.ejphar.2021.174574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 12/13/2022]
Abstract
Identification of molecules specific to the retinal neovasculature will promote antiangiogenic therapy with enhanced targeting ability. The specificity of phage-displayed peptide GX1 (a cyclic 7-mer peptide motif CGNSNPKSC) to gastric cancer neovasculature has been extensively confirmed both in vitro and in vivo. To investigate the potential application of GX1 in antiangiogenic therapy targeting retinal angiogenesis-related diseases, we performed immunohistochemistry and immunofluorescence analyses. GX1 demonstrated positive staining in the retinal neovasculature in an oxygen-induced mouse model of retinopathy (OIR) as well as in rat retinal microvasculature endothelial cells (RMECs), confirming the major role of the GX1 receptor during retinal angiogenesis. Dimeric GX1 was synthesized to increase the binding affinity to the GX1 receptor, and the antiangiogenic effects were examined in RMECs in vitro and the retinal neovasculature in the OIR in vivo. Cell proliferation was evaluated using a Cell Counting Kit-8 (CCK-8) assay, revealing that compared with the GX1 monomer, dimeric GX1 significantly inhibited RMEC proliferation (P < 0.05). This finding may be attributed to the enhanced (P < 0.05) apoptosis induced by dimeric GX1 in RMECs based on results obtained from TUNEL, flow cytometric and cell cycle analyses. In RMECs, in vitro cell migration and tube formation were significantly inhibited following exposure to dimeric GX1. Intravitreal administration of dimeric GX1 resulted in a greater reduction in the retinal neovascularization in vivo than administration of the GX1 monomer (P < 0.05). In conclusion, dimeric GX1 showed greater inhibition of angiogenesis than monomeric GX1 and could be a promising agent for antiangiogenic therapy in retinal angiogenesis-related diseases.
Collapse
Affiliation(s)
- Yingying Luo
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Shaanxi Xi'an, 710061, China
| | - Jipeng Yin
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Shaanxi Xi'an, 710032, China
| | - Rutang Fang
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Shaanxi Xi'an, 710032, China; Department of Gastroenterology, Affiliated No. 986 Hospital of Xijing Hospital, Fourth Military Medical University, Shaanxi Xi'an, 710032, China
| | - Jingtao Liu
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Shaanxi Xi'an, 710061, China; Department of Nuclear Medicine, Affiliated No. 986 Hospital of Xijing Hospital, Fourth Military Medical University, Shaanxi Xi'an, 710032, China
| | - Lu Wang
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Shaanxi Xi'an, 710061, China
| | - Haiping Zhang
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Shaanxi Xi'an, 710061, China
| | - Ming Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, Shaanxi Xi'an, 710061, China
| | - Zhijie Lei
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Shaanxi Xi'an, 710032, China
| | - Shuhui Liang
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Shaanxi Xi'an, 710032, China
| | - Wei Cui
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Shaanxi Xi'an, 710061, China
| | - Zhiyong Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Shaanxi Xi'an, 710061, China.
| | - Kaichun Wu
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Shaanxi Xi'an, 710032, China.
| | - Xiaoli Hui
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Shaanxi Xi'an, 710061, China.
| |
Collapse
|
19
|
Yin Y, Peng J, Zhou J, Chen H, Peng D, Li D, Gan Y, Yin G, Tang Y. Tetrathiomolybdate Partially Alleviates Erectile Dysfunction of Type 1 Diabetic Rats Through Affecting Ceruloplasmin/eNOS and Inhibiting Corporal Fibrosis and Systemic Inflammation. Sex Med 2021; 10:100455. [PMID: 34818604 PMCID: PMC8847815 DOI: 10.1016/j.esxm.2021.100455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 11/28/2022] Open
Abstract
INTRODUCTION Patients with erectile dysfunction induced by diabetes mellitus (DMED) show a poor effect rate for oral phosphodiesterase type 5 inhibitors (PDE5is). Therefore, the new therapeutic strategy is necessary in patients with DMED. AIM To investigate whether Tetrathiomolybdate (TM) supplementation could ameliorate DMED by activation of eNOS. METHODS Twenty-four diabetic rats were induced by intraperitoneal injection of streptozotocin (STZ) and the other 6 normal rats constituted the control group. Eight weeks later, the erectile function of rats was assessed with an apomorphine test. Only some rats with DMED were treated with TM orally every day for 4 weeks; the other rats remained in the same condition for 4 weeks. After 1 week washout, the erectile function of rats in each group was evaluated. Then, the serum concentration of IL-6 and histologic changes of corpus cavernosum were measured. MAIN OUTCOME MEASURE Erectile function was measured after DMED rats treated with TM. The cavernosum level of Ceruloplasmin (Cp), eNOS, endothelial cell content, corporal fibrosis, apoptosis rate and the serum level of IL-6 were also assayed. RESULTS Erectile function in the DMED group was significantly impaired compared with the control group and was partly, but significantly, improved in the DMED+TM group. The DMED group showed upregulation of Cp and inhibition of eNOS, but the inhibition was partly reversed in the DMED+TM group. The DMED group showed serious corporal fibrosis. However, TM supplementation partly increased the ratio of smooth muscle to collagen, decreased the ratio of apoptosis. What's more, gavage administration of TM profoundly decreased the serum level of IL-6 in DMED rats. CONCLUSION TM supplementation inhibits endothelial dysfunction, corporal fibrosis, and systemic inflammation, ultimately leading to partial improvement of DMED in rats. Yin Y, Peng J, Zhou J, et al., Tetrathiomolybdate Partially Alleviates Erectile Dysfunction of Type 1 Diabetic Rats Through Affecting Ceruloplasmin/eNOS and Inhibiting Corporal Fibrosis and Systemic Inflammation. Sex Med 2021;XX:XXXXXX.
Collapse
Affiliation(s)
- Yinghao Yin
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jingxuan Peng
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jun Zhou
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Hanfei Chen
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Dongyi Peng
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Dongjie Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China; Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, China; Xiangya International Medical Center, Department of Geriatric Urology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Yu Gan
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Guangming Yin
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, China.
| | - Yuxin Tang
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China; Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.
| |
Collapse
|
20
|
Fang K, Zhan Y, Zhu R, Wang Y, Wu C, Sun M, Qiu Y, Yuan Z, Liang X, Yin P, Xu K. Bufalin suppresses tumour microenvironment-mediated angiogenesis by inhibiting the STAT3 signalling pathway. J Transl Med 2021; 19:383. [PMID: 34496870 PMCID: PMC8424978 DOI: 10.1186/s12967-021-03058-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/26/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Antiangiogenic therapy has increasingly become an important strategy for the treatment of colorectal cancer. Recent studies have shown that the tumour microenvironment (TME) promotes tumour angiogenesis. Bufalin is an active antitumour compound whose efficacy has been indicated by previous studies. However, there are very few studies on the antiangiogenic effects of bufalin. METHODS Herein, human umbilical vein endothelial cell (HUVEC) tube formation, migration and adhesion tests were used to assess angiogenesis in vitro. Western blotting and quantitative PCR were used to detect relevant protein levels and mRNA expression levels. A subcutaneous xenograft tumour model and a hepatic metastasis model were established in mice to investigate the influence of bufalin on angiogenesis mediated by the TME in vivo. RESULTS We found that angiogenesis mediated by cells in the TME was significantly inhibited in the presence of bufalin. The results demonstrated that the proangiogenic genes in HUVECs, such as VEGF, PDGFA, E-selectin and P-selectin, were downregulated by bufalin and that this downregulation was mediated by inhibition of the STAT3 pathway. Overexpression of STAT3 reversed the inhibitory effects of bufalin on angiogenesis. Furthermore, there was little reduction in angiogenesis when bufalin directly acted on the cells in the tumour microenvironment. CONCLUSION Our findings demonstrate that bufalin suppresses tumour microenvironment-mediated angiogenesis by inhibiting the STAT3 signalling pathway in vascular endothelial cells, revealing that bufalin may be used as a new antiangiogenic adjuvant therapy medicine to treat colorectal cancer.
Collapse
Affiliation(s)
- Kai Fang
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444 China
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Rd, Shanghai, 200237 China
| | - Yueping Zhan
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
| | - Ruiqiu Zhu
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
- Shanghai Putuo Central School of Clinical Medicine, Anhui Medical University, Hefei, 230032 China
| | - Yuqian Wang
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
| | - Chengqi Wu
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444 China
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Rd, Shanghai, 200237 China
| | - Min Sun
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
| | - Yanyan Qiu
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
| | - Zeting Yuan
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
- Shanghai Putuo Central School of Clinical Medicine, Anhui Medical University, Hefei, 230032 China
| | - Xin Liang
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Rd, Shanghai, 200237 China
| | - Peihao Yin
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
- Shanghai Putuo Central School of Clinical Medicine, Anhui Medical University, Hefei, 230032 China
| | - Ke Xu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444 China
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
- Shanghai Putuo Central School of Clinical Medicine, Anhui Medical University, Hefei, 230032 China
- Wenzhou Institute of Shanghai University, Wenzhou, 325000 China
| |
Collapse
|
21
|
Chen F, Han B, Meng Y, Han Y, Liu B, Zhang B, Chang Y, Cao P, Fan Y, Tan K. Ceruloplasmin correlates with immune infiltration and serves as a prognostic biomarker in breast cancer. Aging (Albany NY) 2021; 13:20438-20467. [PMID: 34413268 PMCID: PMC8436892 DOI: 10.18632/aging.203427] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 08/02/2021] [Indexed: 12/29/2022]
Abstract
Breast-invasive carcinoma (BRCA) is the most frequent and malignant tumor in females. Ceruloplasmin (CP) is a multifunctional molecule involved in iron metabolism, but its expression profile, prognostic potential and relationship with immune cell infiltration in BRCA are unknown. Ceruloplasmin mRNA and protein expression was significantly decreased in BRCA patients according to the Oncomine, UALCAN, GEPIA and TCGA databases. Ceruloplasmin expression was strongly correlated with various clinicopathological features of BRCA patients. BRCA patients with high ceruloplasmin expression exhibited shorter survival times than those with low ceruloplasmin expression based on the Kaplan-Meier plotter and PrognoScan databases. GO and KEGG analyses and GSEA revealed a strong correlation between ceruloplasmin and various immune-related pathways. Ceruloplasmin expression was significantly associated with the infiltration of immune cells into tumor sites by analyzing the TIMER and CIBERSORT. Additionally, ceruloplasmin was positively correlated with immune checkpoints in BRCA. These findings suggest that low ceruloplasmin expression correlates with a favorable prognosis and tumor immune cell infiltration in BRCA patients. Ceruloplasmin may serve as a therapeutic target and predict the efficacy of immunotherapy for BRCA.
Collapse
Affiliation(s)
- Fei Chen
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei, China
| | - Bihui Han
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei, China
| | - Yanxiu Meng
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei, China
| | - Yu Han
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei, China
| | - Bing Liu
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei, China
| | - Bo Zhang
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei, China
| | - Yanzhong Chang
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei, China
| | - Pengxiu Cao
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei, China
| | - Yumei Fan
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei, China
| | - Ke Tan
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei, China
| |
Collapse
|
22
|
miR-23a-3p is involved in drug resistance by directly targeting the influx drug transporter organic anion-transporting polypeptide 2. Childs Nerv Syst 2021; 37:2545-2555. [PMID: 33779805 DOI: 10.1007/s00381-021-05146-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/23/2021] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Drug transporters are involved in the drug resistance of individuals with drug-resistant epilepsy by influencing the intracerebral transport of antiepileptic drugs (AEDs). The expression of drug transporters is associated with microRNAs. We previously revealed that miR-23a-3p levels were elevated in the blood of patients with intractable epilepsy. Additionally, the influx drug transporter organic anion-transporting polypeptide 2 (Oatp2) is involved in the intracerebral transport of valproic acid (VPA), the most commonly used AED; repeated seizures lead to decreased expression of Oatp2. However, the role of miR-23a-3p in the expression of Oatp2 and in the development of drug resistance has not been established. Herein, we aimed to determine the potential role of miR-23a-3p in VPA-resistant epilepsy through in vivo and in vitro experiments. METHODS Epilepsy was elicited after status epilepticus (SE) was induced by lithium-pilocarpine in adult Sprague-Dawley rats, followed by VPA treatment to select rats with VPA resistance. The expression of miR-23a-3p was detected by immunohistochemistry and reverse transcription-polymerase chain reaction (RT-PCR). A miR-23a-3p inhibitor was intracerebrally injected into VPA-resistant rats, and histological staining and Morris water maze tests were performed to evaluate brain damage and learning/memory functions in these rats. Subsequently, a dual-luciferase reporter assay and a VPA uptake assay were performed in brain microvascular endothelial cells (BMECs) to investigate the underlying mechanism of action of miR-23a-3p. RESULTS Our results indicated that compared to that in control rats, miR-23a-3p was elevated in VPA-resistant rats. Intracerebral injection of a miR-23a-3p inhibitor reduced brain damage and the associated deficits in learning and memory functions in rats with VPA resistance. Further investigation indicated that Oatp2 was the direct target of miR-23a-3p, and it was negatively regulated by miR-23a-3p in the brain and BMECs. Furthermore, we demonstrated that miR-23a-3p reduced VPA uptake in BMECs by regulating Oatp2 expression. CONCLUSIONS miR-23a-3p is involved in VPA resistance in epilepsy by directly targeting the influx drug transporter Oatp2, indicating that miR-23a-3p could be a potential therapeutic target for intractable epilepsy.
Collapse
|
23
|
Fang C, Dai L, Wang C, Fan C, Yu Y, Yang L, Deng H, Zhou Z. Secretogranin II impairs tumor growth and angiogenesis by promoting degradation of hypoxia-inducible factor-1α in colorectal cancer. Mol Oncol 2021; 15:3513-3526. [PMID: 34160138 PMCID: PMC8637574 DOI: 10.1002/1878-0261.13044] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 05/15/2021] [Accepted: 05/28/2021] [Indexed: 02/05/2023] Open
Abstract
Distant metastasis is a major cause of death in patients with colorectal cancer (CRC) but the management of advanced and metastatic CRC still remains problematic due to the distinct molecular alterations during tumor progression. Tumor angiogenesis is a key step in tumor growth, invasion and metastasis. However, the signaling pathways involved in angiogenesis are poorly understood. The results of the present study showed that secretogranin II (SCG2) was significantly downregulated in malignant CRC tissues, and higher expression of SCG2 was correlated with longer disease‐free survival and overall survival of CRC patients. The results of an animal study showed that ectopic expression of SCG2 significantly inhibited CRC tumor growth by disrupting angiogenesis. Furthermore, the inhibition of expression of vascular endothelial growth factor (VEGF) by SCG2 and rescue of VEGF effectively blocked SCG2‐induced inhibition of angiogenesis. Investigations into the underlying mechanism suggested that SCG2 promoted degradation of hypoxia‐inducible factor (HIF)‐1α by interacting with the von Hippel–Lindau tumor suppressor in CRC cells. Blocking of degradation of HIF‐1α effectively attenuated the SCG2‐mediated decrease in expression of VEGF in CRC cells. Collectively, these results demonstrated that treatment with SCG2 effectively inhibited CRC tumor growth by disrupting the activities of HIF‐1α/VEGF, thereby clarifying the anti‐tumor and anti‐angiogenesis roles of SCG2 in CRC, while providing a novel therapeutic target and a potential prognostic marker of disease progression.
Collapse
Affiliation(s)
- Chao Fang
- Institute of Digestive Surgery, West China Hospital, Sichuan University, Chengdu, China.,Department of Gastrointestinal Surgery, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Lei Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Cun Wang
- Institute of Digestive Surgery, West China Hospital, Sichuan University, Chengdu, China.,Department of Gastrointestinal Surgery, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Chuanwen Fan
- Institute of Digestive Surgery, West China Hospital, Sichuan University, Chengdu, China.,Department of Gastrointestinal Surgery, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Yongyang Yu
- Institute of Digestive Surgery, West China Hospital, Sichuan University, Chengdu, China.,Department of Gastrointestinal Surgery, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Lie Yang
- Institute of Digestive Surgery, West China Hospital, Sichuan University, Chengdu, China.,Department of Gastrointestinal Surgery, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Hongxin Deng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Zongguang Zhou
- Institute of Digestive Surgery, West China Hospital, Sichuan University, Chengdu, China.,Department of Gastrointestinal Surgery, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
24
|
Hu C, Wu Z, Huang Z, Hao X, Wang S, Deng J, Yin Y, Tan C. Nox2 impairs VEGF-A-induced angiogenesis in placenta via mitochondrial ROS-STAT3 pathway. Redox Biol 2021; 45:102051. [PMID: 34217063 PMCID: PMC8258686 DOI: 10.1016/j.redox.2021.102051] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/04/2021] [Accepted: 06/16/2021] [Indexed: 02/06/2023] Open
Abstract
Aberrant placental angiogenesis is associated with fetal intrauterine growth restriction (IUGR), but the mechanism underlying abnormal placental angiogenesis remains largely unknown. Here, lower vessel density and higher expression of NADPH oxidases 2 (Nox2) were observed in the placentae for low birth weight (LBW) fetuses versus normal birth weight (NBW) fetuses, with a negative correlation between Nox2 and placental vessel density. Moreover, it was revealed for the first time that Nox2 deficiency facilitates angiogenesis in vitro and in vivo, and vascular endothelial growth factor-A (VEGF-A) has an essential role in Nox2-controlled inhibition of angiogenesis in porcine vascular endothelial cells (PVECs). Mechanistically, Nox2 inhibited phospho-signal transducer and activator of transcription 3 (p-STAT3) in the nucleus by inducing the production of mitochondrial reactive oxygen species (ROS). Dual-luciferase assay confirmed that knockdown of Nox2 reduces the expression of VEGF-A in an STAT3 dependent manner. Our results indicate that Nox2 is a potential target for therapy by increasing VEGF-A expression to promote angiogenesis and serves as a prognostic indicator for fetus with IUGR.
Collapse
Affiliation(s)
- Chengjun Hu
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China; Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571101, China
| | - Zifang Wu
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Zihao Huang
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Xiangyu Hao
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Shuqi Wang
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Jinping Deng
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Yulong Yin
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, 410125, China
| | - Chengquan Tan
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China.
| |
Collapse
|
25
|
Zhang Z, Deng M, Huang J, Wu J, Li Z, Xing M, Wang J, Guo Q, Zou W. Microglial annexin A3 downregulation alleviates bone cancer-induced pain through inhibiting the Hif-1α/vascular endothelial growth factor signaling pathway. Pain 2021; 161:2750-2762. [PMID: 32569086 DOI: 10.1097/j.pain.0000000000001962] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Bone cancer-induced pain (BCP) is a challenging clinical problem because traditional therapies are often only partially effective. Annexin A3 (ANXA3) is highly expressed in microglia in the spinal cord, and its expression is upregulated during BCP. However, the roles of microglial ANXA3 in the development and maintenance of BCP and the underlying molecular mechanisms remain unclear. This study was performed on male mice using a metastatic lung BCP model. Adeno-associated virus shANXA3 (AAV-shANXA3) was injected intrathecally 14 days before and 7 days after bone cancer induction, and relevant pain behaviors were assessed by measuring the paw withdrawal mechanical threshold, paw withdrawal thermal latency, and spontaneous hind limb lifting. ANXA3 protein expression was downregulated in microglial N9 cells by lentiviral transfection (LV-shANXA3). ANXA3, hypoxia-inducible factor-1α (Hif-1α), vascular endothelial growth factor (VEGF) expression levels, and Hif-1α transactivation activity regulated by ANXA3 were measured. As a result, ANXA3 was expressed in microglia, and its expression significantly increased during BCP. ANXA3 knockdown reversed pain behaviors but did not prevent pain development. Moreover, ANXA3 knockdown significantly reduced Hif-1α and VEGF expression levels in vitro and in vivo. And overexpression of Hif-1α or VEGF blocked the effects of AAV-shANXA3 on BCP. ANXA3 knockdown in N9 cells significantly decreased the p-PKC protein expression in the cocultured neurons. Finally, ANXA3 overexpression significantly increased Hif-1α transactivation activity in 293T cells. Therefore, microglial ANXA3 downregulation alleviates BCP by inhibiting the Hif-1α/VEGF signaling pathway, which indicates that ANXA3 may be a potential target for the treatment of BCP.
Collapse
Affiliation(s)
- Zengli Zhang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Meiling Deng
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Jiangju Huang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Jing Wu
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhengyiqi Li
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Manyu Xing
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Jian Wang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Qulian Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Wangyuan Zou
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
26
|
Li M, Huang T, Li X, Shi Z, Sheng Y, Hu M, Song K. GDC-0575, a CHK1 Inhibitor, Impairs the Development of Colitis and Colitis-Associated Cancer by Inhibiting CCR2 + Macrophage Infiltration in Mice. Onco Targets Ther 2021; 14:2661-2672. [PMID: 33897258 PMCID: PMC8058335 DOI: 10.2147/ott.s297132] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 03/26/2021] [Indexed: 01/15/2023] Open
Abstract
Background Checkpoint kinase 1 (CHK1) plays an important role in DNA damage response and cell cycle progression. Thus, targeting CHK1 is an efficient strategy for cancer therapy. Purpose The present study aimed to investigate the potential therapeutic effects of GDC-0575, a CHK1-specific inhibitor, in colitis-associated cancer (CAC) and colitis. Methods We established a DSS-induced acute colitis model and an azoxymethane/dextran sodium sulfate (DSS)-induced CAC model using mice and tested the effect of GDC-0575 on them. Flow cytometry and immunofluorescence were employed to investigate the infiltration of immune cells, and inflammatory cytokine expression in the colon of mice with CAC or colitis was investigated using ELISA and qPCR. We also investigated the correlation between CHK1 and CCL2/CCR2 in human colorectal cancer (CRC) tissues. Results Administration of GDC-0575 significantly inhibited CHK1 expression in the colon and dramatically impaired the development of CAC and colitis in mice. Moreover, the inhibition of CHK1 expression resulted in efficient inhibition of infiltration by iNOS-positive macrophages, but had no significant effect on CD4 T cells, CD8 T cells, and myeloid-derived suppressor cells (MDSCs). Significant downregulation of TNF-α, IL-6, and IL-1β and dramatic upregulation of IL-10 were observed in the colons of both mice with CAC and colitis treated with GDC-0575. CCL2 expression was also downregulated by GDC-0575 in both mice with CAC and colitis; this was followed by the inhibition of CCR2+ macrophage infiltration in the colon. Furthermore, we report a positive correlation between CHK1 expression and CCL2/CCR2 expression in the malignant tissues of patients with CRC. Conclusion Taken together, we infer that GDC-0575 impairs the development of CAC and colitis by regulating cytokine expression and inhibiting CCR2+ macrophage infiltration in mice colon.
Collapse
Affiliation(s)
- Min Li
- Department of Pharmacy, The First Affiliated Hospital of Jishou University, Jishou, Hunan, 416000, People's Republic of China
| | - Tianqing Huang
- Department of Neurology, The First Affiliated Hospital of Jishou University, Jishou, Hunan, 416000, People's Republic of China
| | - Xiaolan Li
- Department of Hematology, The First Affiliated Hospital of Jishou University, Jishou, Hunan, 416000, People's Republic of China
| | - Zhiwei Shi
- Department of Hematology, The First Affiliated Hospital of Jishou University, Jishou, Hunan, 416000, People's Republic of China
| | - Yue Sheng
- Department of Pediatrics, The First Affiliated Hospital of Jishou University, Jishou, Hunan, 416000, People's Republic of China
| | - Mimi Hu
- Department of Hematology, The First Affiliated Hospital of Jishou University, Jishou, Hunan, 416000, People's Republic of China
| | - Kui Song
- Department of Hematology, The First Affiliated Hospital of Jishou University, Jishou, Hunan, 416000, People's Republic of China
| |
Collapse
|
27
|
Tang RZ, Li ZZ, Hu D, Kanwal F, Yuan CB, Mustaqeem M, Batool AI, Rehman MFU. Sanjie Yiliu Formula Inhibits Colorectal Cancer Growth by Suppression of Proliferation and Induction of Apoptosis. ACS OMEGA 2021; 6:7761-7770. [PMID: 33778287 PMCID: PMC7992181 DOI: 10.1021/acsomega.0c05565] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/24/2021] [Indexed: 05/04/2023]
Abstract
Colorectal cancer (CRC) is one of the most common malignancies worldwide. As current therapies toward CRC, including chemotherapy and radiotherapy, pose limitations, such as multidrug resistance (MDR) as well as the intrinsic and potential cytotoxic effects, necessitating to find more effective treatment options with fewer side effects, traditional Chinese medicine (TCM) has an advantage in complementary therapies. In the present study, 3-(4,5-dimethylthiozol-2-yl)-2,5-diphenyltetrazolium bromide (MTT assays), trypan blue staining, colony formation, 4,6-diamidino-2-phenylindole dihydrochloride (DAPI) staining, cell cycle determination, and Annexin V-FITC/PI staining were used to examine the efficacy of Sanjie Yiliu Formula (SJYLF) against CRC proliferation and to investigate its underlying molecular mechanisms through protein expression of various proapoptotic factors by quantitative polymerase chain reaction (q-PCR) and Western blotting. This four-herb-TCM SJYLF can be suggested as one of the decoctions clinically effective in late-stage cancer treatment. Our results suggest that SJYLF robustly decreased the viability of only CRC cell lines (HCT-8, SW-480, HT-29, and DLD-1) and not the normal human kidney cells (HK-2). Moreover, SJYLF significantly suppressed proliferation and induced apoptosis in HCT-8 and downregulated cyclin D1, CDK4, and BCL-2, while Bax expression was upregulated at both mRNA and protein expression levels.
Collapse
Affiliation(s)
- Rong Zhu Tang
- Department
of Gastroenterology, Seventh People’s
Hospital of Shanghai University of Traditional Chinese Medicine, NO.358, Datong Road, Pudong New
Area, Shanghai 200137, P. R. China
| | - Zhang Zhi Li
- Department
of Hematology, Taihe Hospital Affiliated
to the Hubei University of Medicine, Shiyan, China
| | - Dan Hu
- Department
of Neurology, The Central Hospital of Xiaogan, Xiaogan, Hubei 432100, P. R. China
| | - Fariha Kanwal
- Med-X
Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 201620, P. R.
China
| | - Cheng Bin Yuan
- Department
of Critical Care Medicine, Shanghai General
Hospital, Shanghai 200080, P. R. China
- School
of Medicine, Shanghai Jiao Tong University, Shanghai 200025, P. R. China
| | - Muhammad Mustaqeem
- Department
of Chemistry, University of Sargodha, Sub-Campus Bhakkar, Bhakkar 30000, Pakistan
| | - Aima Iram Batool
- Department
of Zoology, University of Sargodha, Sargodha 40100, Pakistan
| | | |
Collapse
|
28
|
Chen Y, Wang B, Yuan X, Lu Y, Hu J, Gao J, Lin J, Liang J, Hou S, Chen S. Vitexin prevents colitis-associated carcinogenesis in mice through regulating macrophage polarization. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 83:153489. [PMID: 33571919 DOI: 10.1016/j.phymed.2021.153489] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 01/22/2021] [Accepted: 01/28/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Patients with inflammatory bowel disease are at increased risks of developing ulcerative colitis-associated colorectal cancer (CAC). Vitexin can suppress the proliferation of colorectal carcinoma cells in vitro orin vivo. However, different from colorectal carcinoma, CAC is more consistent with the transformation from inflammation to cancer in clinical chronic IBD patients. Therefore, we aim to investigated that vitexin whether possess benefic effects on CAC mice. PURPOSE We aimed to determine the beneficial effects of vitexin on CAC mice and reveal its underlying mechanism. METHODS The mouse CAC model was induced by Azoxymethane and dextran sodium sulfate (AOM/DSS) and CAC mice were treated with vitexin. At the end of this study, inflammatory cytokines of IL-1β, IL-6, TNF-α, IL-10 as well as nitric oxide (NO) were detected by kits after long-term treatment of vitexin. Pathological changes and macrophage polarization were determined by H&E and immunofluorescence in adjacent noncancerous tissue and carcinomatous tissue respectively of CAC mice. RESULTS Our results showed that oral administration of vitexin could significantly improve the clinical signs and symptoms of chronic colitis, relieve colon damage, regulate colonic inflammatory cytokines, as well as suppress tumor incidence and tumor burden. Interesting, vitexin caused a significant increase in serum level of NO and a higher content of NO in tumor tissue. In addition, vitexin significantly decreased M1 phenotype macrophages in the adjacent noncancerous tissue, while markedly up-regulated M1 macrophage polarization in the tumor tissue in the colon of CAC mice. CONCLUSION Vitexin can attenuate chronic colitis-associated carcinogenesis induced by AOM/DSS in mice and its protective effects are partly associated with its alternations in macrophage polarization in the inflammatory and tumor microenvironment .
Collapse
Affiliation(s)
- Yonger Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China
| | - Bingxin Wang
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510000, PR China; School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China
| | - Xin Yuan
- National Engineering Research Center for Modernization of Traditional Chinese Medicine (Guangzhou Branch), Guangzhou, Guangdong, 510006, PR China
| | - Yingyu Lu
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510000, PR China; School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China
| | - Jiliang Hu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China
| | - Jie Gao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China
| | - Jizong Lin
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510000, PR China
| | - Jian Liang
- National Engineering Research Center for Modernization of Traditional Chinese Medicine (Guangzhou Branch), Guangzhou, Guangdong, 510006, PR China
| | - Shaozhen Hou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China.
| | - Shuxian Chen
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510000, PR China.
| |
Collapse
|
29
|
BATF2 prevents glioblastoma multiforme progression by inhibiting recruitment of myeloid-derived suppressor cells. Oncogene 2021; 40:1516-1530. [PMID: 33452462 PMCID: PMC7906906 DOI: 10.1038/s41388-020-01627-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 11/22/2020] [Accepted: 12/15/2020] [Indexed: 02/05/2023]
Abstract
The basic leucine zipper ATF-like transcription factor 2 (BATF2) has been implicated in inflammatory responses and anti-tumour effects. Little, however, is known regarding its extracellular role in maintaining a non-supportive cancer microenvironment. Here, we show that BATF2 inhibits glioma growth and myeloid-derived suppressor cells (MDSCs) recruitment. Interestingly, extracellular vesicles (EVs) from BATF2-overexpressing glioma cell lines (BATF2-EVs) inhibited MDSCs chemotaxis in vitro. Moreover, BATF2 inhibited intracellular SDF-1α and contributes to decreased SDF-1α in EVs. In addition, BATF2 downregulation-induced MDSCs recruitment were reversed by blocking SDF-1α/CXCR4 signalling upon AMD3100 treatment. Specifically, detection of EVs in 24 pairs of gliomas and healthy donors at different stages revealed that the abundance of BATF2-positive EVs in plasma (BATF2+ plEVs) can distinguish stage III-IV glioma from stage I-II glioma and healthy donors. Taken together, our study identified novel regulatory functions of BATF2 in regulating MDSCs recruitment, providing a prognostic value in terms of the number of BATF2+ plEVs in glioma stage.
Collapse
|
30
|
Lin Y, Cheng L, Liu Y, Wang Y, Wang Q, Wang HL, Shi G, Li JS, Wang QN, Yang QM, Chen S, Su XL, Yang Y, Jiang M, Hu X, Fan P, Fang C, Zhou ZG, Dai L, Deng HX. Intestinal epithelium-derived BATF3 promotes colitis-associated colon cancer through facilitating CXCL5-mediated neutrophils recruitment. Mucosal Immunol 2021; 14:187-198. [PMID: 32467604 DOI: 10.1038/s41385-020-0297-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/24/2020] [Accepted: 05/01/2020] [Indexed: 02/05/2023]
Abstract
Inflammation is a critical player in the development and progression of colon cancer. Basic leucine zipper transcription factor ATF-like 3 (BATF3) plays an important role in infection and tumor immunity through regulating the development of conventional type 1 dendritic cells (cDC1s). However, the function of BATF3 in colitis and colitis-associated colon cancer (CAC) remains unclear. Here, BATF3 wild-type and knockout mice were used to construct an AOM/DSS-induced CAC model. In addition, DSS-induced chronic colitis, bone marrow cross-transfusion (BMT), neutrophil knockout, and other animal models were used for in-depth research. We found that BATF3 deficiency in intestinal epithelial cells rather than in cDC1s inhibited CAC, which was depended on inflammatory stimulation. Mechanistically, BATF3 directly promoted transcription of CXCL5 by forming a heterodimer with JunD, and accelerated the recruitment of neutrophils through the CXCL5-CXCR2 axis, ultimately increasing the occurrence and development of CAC. Tissue microarray and TCGA data also indicated that high expression of BATF3 was positively correlated with poor prognosis of colorectal cancer and other inflammation-related tumors. In summary, our results demonstrate that intestinal epithelial-derived BATF3 relies on inflammatory stimulation to promote CAC, and BATF3 is expected to be a novel diagnostic indicator for colitis and CAC.
Collapse
Affiliation(s)
- Y Lin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, Sichuan, China
| | - L Cheng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, Sichuan, China
| | - Y Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, Sichuan, China
| | - Y Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, Sichuan, China
| | - Q Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, Sichuan, China
| | - H L Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, Sichuan, China
| | - G Shi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, Sichuan, China
| | - J S Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, Sichuan, China
| | - Q N Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, Sichuan, China
| | - Q M Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, Sichuan, China
| | - S Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, Sichuan, China
| | - X L Su
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, Sichuan, China
| | - Y Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, Sichuan, China
| | - M Jiang
- Department of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, China.
| | - X Hu
- West China Biobanks, Department of Clinical Research Management and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - P Fan
- West China Biobanks, Department of Clinical Research Management and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - C Fang
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Z G Zhou
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - L Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, Sichuan, China.
| | - H X Deng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
31
|
Li T, Jia L, Deng Y, Wang B, Quan S. TIPE1 impairs ovarian tumor growth by promoting caspase-dependent apoptosis. Oncol Lett 2020; 20:365. [PMID: 33133265 PMCID: PMC7590423 DOI: 10.3892/ol.2020.12227] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 09/14/2020] [Indexed: 12/14/2022] Open
Abstract
Tumor necrosis factor-α-induced protein 8-like 1 (TIPE1) functions as a tumor suppressor in several types of cancer, including lung and breast cancer. The present study aimed to determine the level of expression and the function of TIPE1 in ovarian cancer. TIPE1 expression was determined in tissue microarrays and ovarian cancer cells, and these data were analyzed to assess the association between TIPE1 expression and prognosis in patients with ovarian cancer. The potential antitumor effects of TIPE1 were investigated in vitro and in a xenograft mouse model. Furthermore, the underlying molecular mechanism by which TIPE1 regulates ovarian cancer growth was determined via flow cytometric analysis, western blotting and rescue experiments. The results of the present study indicated that TIPE1 levels were markedly decreased in ovarian cancer tissues, and its level of expression was associated with a favorable prognosis of patients with ovarian cancer. In addition, ectopic TIPE1 expression significantly impaired A2780 and SKOV3 cell proliferation and colony formation in vitro, which was accompanied by efficient inhibition of xenograft tumor growth in mice. Investigations into the underlying molecular mechanism demonstrated that TIPE1 induced ovarian cancer cell apoptosis by promoting caspase protein expression. Inhibition of caspase-dependent apoptosis by z-VAD blocked TIPE1-mediated inhibition of the proliferation and induction of apoptosis in ovarian cancer cells. Collectively, the results of the present study suggest that TIPE1 may be a potential prognostic predictor and therapeutic target for patients with ovarian cancer.
Collapse
Affiliation(s)
- Tingting Li
- Department of Gynecology and Obstetrics, Center for Reproductive Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China.,Reproductive Medicine Research Center, Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Lei Jia
- Reproductive Medicine Research Center, Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510655, P.R. China.,Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yu Deng
- Assisted Reproductive Medical Center, Guangzhou Women and Children's Hospital, Guangzhou, Guangdong 510623, P.R. China
| | - Bo Wang
- Reproductive Medicine Research Center, Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Song Quan
- Department of Gynecology and Obstetrics, Center for Reproductive Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
32
|
Ye T, Yang B, Wang C, Su C, Luo J, Yang X, Yu H, Yuan Z, Meng Z, Xia J. TIPE1 impairs stemness maintenance in colorectal cancer through directly targeting β-catenin. Carcinogenesis 2020; 41:25-35. [PMID: 31111874 DOI: 10.1093/carcin/bgz079] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 04/16/2019] [Accepted: 04/28/2019] [Indexed: 12/30/2022] Open
Abstract
TIPE1 (tumor necrosis factor-α-induced protein 8-like 1) contributes to cell death in diverse cancers. However, the expression and biological functions of TIPE1 in colon cancer remain unclear. In the present study, we report that TIPE1 was downregulated in colon cancer tissues and positively correlates with prognosis of colon cancer patients. TIPE1 overexpression significantly inhibits colon cancer cell growth both in vitro and in vivo through impairing stemness, accompanied with downregulation of the stemness-related markers, ALDH, CD133, CD44 and SOX-9. Mechanically, TIPE1 directly targets β-catenin and promotes β-catenin degradation in a protease-dependent manner, and Wnt/β-catenin signaling plays a crucial role during TIPE1-mediated stemness inhibition in colon cancer. These findings reveal that TIPE1 exerts anti-tumor effects in colon cancer and suggest that TIPE1 would be a therapeutic target for cancers.
Collapse
Affiliation(s)
- Tao Ye
- Minhang Branch, Zhongshan Hospital, Fudan University; Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Biwei Yang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chen Wang
- Minhang Branch, Zhongshan Hospital, Fudan University; Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Chang Su
- Minhang Branch, Zhongshan Hospital, Fudan University; Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Jing Luo
- Minhang Branch, Zhongshan Hospital, Fudan University; Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Xiaodi Yang
- Minhang Branch, Zhongshan Hospital, Fudan University; Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Hongmei Yu
- Minhang Branch, Zhongshan Hospital, Fudan University; Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Zhen Yuan
- Minhang Branch, Zhongshan Hospital, Fudan University; Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Zhefeng Meng
- Minhang Branch, Zhongshan Hospital, Fudan University; Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Jinglin Xia
- Minhang Branch, Zhongshan Hospital, Fudan University; Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China.,Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
33
|
Khandagale A, Åberg M, Wikström G, Bergström Lind S, Shevchenko G, Björklund E, Siegbahn A, Christersson C. Role of Extracellular Vesicles in Pulmonary Arterial Hypertension: Modulation of Pulmonary Endothelial Function and Angiogenesis. Arterioscler Thromb Vasc Biol 2020; 40:2293-2309. [PMID: 32757648 DOI: 10.1161/atvbaha.120.314152] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Extracellular vesicles (EVs) have the potential to act as intercellular communicators. The aims were to characterize circulating EVs in patients with pulmonary arterial hypertension (PAH) and to explore whether these EVs contribute to endothelial activation and angiogenesis. Approach and Results: Patients with PAH (n=70) and healthy controls (HC; n=20) were included in this cross-sectional study. EVs were characterized and human pulmonary endothelial cells (hPAECs) were incubated with purified EVs. Endothelial cell activity and proangiogenic markers were analyzed. Tube formation analysis was performed for hPAECs, and the involvement of PSGL-1 (P-selectin glycoprotein ligand 1) was evaluated. The numbers of CD62P+, CD144+, and CD235a EVs were higher in blood from PAH compared with HC. Thirteen proteins were differently expressed in PAH and HC EVs, where complement fragment C1q was the most significantly elevated protein (P=0.0009) in PAH EVs. Upon EVs-internalization in hPAECs, more PAH compared with HC EVs evaded lysosomes (P<0.01). As oppose to HC, PAH EVs stimulated hPAEC activation and induced transcription and translation of VEGF-A (vascular endothelial growth factor A; P<0.05) and FGF (fibroblast growth factor; P<0.005) which were released in the cell supernatant. These proangiogenic proteins were higher in patient with PAH plasma compered with HC. PAH EVs induced a complex network of angiotubes in vitro, which was abolished by inhibitory PSGL-1antibody. Anti-PSGL-1 also inhibited EV-induced endothelial cell activation and PAH EV dependent increase of VEGF-A. CONCLUSIONS Patients with PAH have higher levels of EVs harboring increased amounts of angiogenic proteins, which induce activation of hPAECs and in vitro angiogenesis. These effects were partly because of platelet-derived EVs evasion of lysosomes upon internalization within hPAEC and through possible involvement of P-selectin-PSGL-1 pathway.
Collapse
Affiliation(s)
- Avinash Khandagale
- From the Department of Medical Sciences, Cardiology and Clinical Chemistry (A.K.), Uppsala University, Sweden
| | - Mikael Åberg
- Department of Medical Sciences, Clinical Chemistry (M.Å., A.S.), Uppsala University, Sweden
| | - Gerhard Wikström
- Department of Medical Sciences, Cardiology and Internal Medicine (G.W.), Uppsala University, Sweden
| | - Sara Bergström Lind
- Department of Chemistry - BMC, Analytical Chemistry (S.B.L., G.S.), Uppsala University, Sweden
| | - Ganna Shevchenko
- Department of Chemistry - BMC, Analytical Chemistry (S.B.L., G.S.), Uppsala University, Sweden
| | - Erik Björklund
- Department of Medical Sciences, Cardiology (E.B., C.C.), Uppsala University, Sweden
| | - Agneta Siegbahn
- Department of Medical Sciences, Clinical Chemistry (M.Å., A.S.), Uppsala University, Sweden
| | | |
Collapse
|
34
|
Prognostic Value of DNA Methylation-Driven Genes in Clear Cell Renal Cell Carcinoma: A Study Based on Methylation and Transcriptome Analyses. DISEASE MARKERS 2020; 2020:8817652. [PMID: 32733620 PMCID: PMC7369658 DOI: 10.1155/2020/8817652] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/15/2020] [Accepted: 06/25/2020] [Indexed: 12/20/2022]
Abstract
Background Few previous studies have comprehensively explored the level of DNA methylation and gene expression in ccRCC. The purpose of this study was to identify the key clear cell renal cell carcinoma- (ccRCC-) related DNA methylation-driven genes (MDG) and to build a prognostic model based on the level of DNA methylation. Methods RNA-seq transcriptome data and DNA methylation data were obtained from The Cancer Genome Atlas. Based on the MethylMix algorithm, we obtain ccRCC-related MDG. The univariate and multivariate Cox regression analyses were employed to investigate the correlation between patient overall survival and the methylation level of each MDG. Finally, a prognosis risk score was established based on a linear combination of the regression coefficient derived from the multivariate Cox regression model (β) multiplied with the methylation level of the gene. Results 19 ccRCC-related MDG were identified. Three MDG (NCKAP1L, EVI2A, and BATF) were further screened and integrated into a prognostic risk score model, risk score = (3.710∗methylation level of NCKAP1L) + (-3.892∗methylation level of EVI2A) + (-3.907∗methylation level of BATF). The risk model was independent from conventional clinical characteristics as a prognostic factor for ccRCC (HR = 1.221, 95% confidence interval: 1.063-1.402, and P = 0.005). The joint survival analysis showed that the gene expression and methylation levels of the prognostic genes EVI2A and BATF were significantly related with prognosis. Conclusion This study provided an important bioinformatics foundation for in-depth studies of ccRCC DNA methylation.
Collapse
|
35
|
Xie JW, Huang XB, Chen QY, Ma YB, Zhao YJ, Liu LC, Wang JB, Lin JX, Lu J, Cao LL, Lin M, Tu RH, Zheng CH, Huang CM, Li P. m 6A modification-mediated BATF2 acts as a tumor suppressor in gastric cancer through inhibition of ERK signaling. Mol Cancer 2020; 19:114. [PMID: 32650804 PMCID: PMC7350710 DOI: 10.1186/s12943-020-01223-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 05/28/2020] [Indexed: 02/07/2023] Open
Abstract
Background BATF2, also known as SARI, has been implicated in tumor progression. However, its role, underlying mechanisms, and prognostic significance in human gastric cancer (GC) are elusive. Methods We obtained GC tissues and corresponding normal tissues from 8 patients and identified BATF2 as a downregulated gene via RNA-seq. qRT-PCR and western blotting were applied to examine BATF2 levels in normal and GC tissues. The prognostic value of BATF2 was elucidated using tissue microarray and IHC analyses in two independent GC cohorts. The functional roles and mechanistic insights of BATF2 in GC growth and metastasis were evaluated in vitro and in vivo. Results BATF2 expression was significantly decreased in GC tissues at both the mRNA and protein level. Multivariate Cox regression analysis revealed that BATF2 was an independent prognostic factor and effective predictor in patients with GC. Low BATF2 expression was remarkably associated with peritoneal recurrence after curative gastrectomy. Moreover, elevated BATF2 expression effectively suppressed GC growth and metastasis in vitro and in vivo. Mechanistically, BATF2 binds to p53 and enhances its protein stability, thereby inhibiting the phosphorylation of ERK. Tissue microarray results indicated that the prognostic value of BATF2 was dependent on ERK activity. In addition, the N6-methyladenosine (m6A) modification of BATF2 mRNA by METTL3 repressed its expression in GC. Conclusions Collectively, our findings indicate the pivotal role of BATF2 in GC and highlight the regulatory function of the METTL3/BATF2/p53/ERK axis in modulating GC progression, which provides potential prognostic and therapeutic targets for GC treatment.
Collapse
Affiliation(s)
- Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou, 350001, Fujian Province, China.,Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Xiao-Bo Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou, 350001, Fujian Province, China.,Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou, 350001, Fujian Province, China.,Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Yu-Bin Ma
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qinghai University, Xining, China
| | - Ya-Jun Zhao
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Li-Chao Liu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou, 350001, Fujian Province, China.,Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Jia-Bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou, 350001, Fujian Province, China.,Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou, 350001, Fujian Province, China.,Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou, 350001, Fujian Province, China.,Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Long-Long Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou, 350001, Fujian Province, China.,Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Mi Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou, 350001, Fujian Province, China.,Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Ru-Hong Tu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou, 350001, Fujian Province, China.,Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou, 350001, Fujian Province, China. .,Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China. .,Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China.
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou, 350001, Fujian Province, China. .,Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China. .,Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China.
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou, 350001, Fujian Province, China. .,Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China. .,Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
36
|
Smith LR, Meyer GA. Skeletal muscle explants: ex-vivo models to study cellular behavior in a complex tissue environment. Connect Tissue Res 2020; 61:248-261. [PMID: 31492079 PMCID: PMC8837600 DOI: 10.1080/03008207.2019.1662409] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 08/22/2019] [Indexed: 02/03/2023]
Abstract
Purpose/Aim: Skeletal muscle tissue explants have been cultured and studied for nearly 100 years. These cultures, which retain complex tissue structure in an environment suited to precision manipulation and measurement, have led to seminal discoveries of the extrinsic and intrinsic mechanisms regulating contractility, metabolism and regeneration. This review discusses the two primary models of muscle explant: isolated myofiber and intact muscle.Materials and Methods: Relevant literature was reviewed and synthesized with a focus on the unique challenges and capabilities of each explant model.Results: Impactful past, current and future novel applications are discussed.Conclusions: Experiments using skeletal muscle explants have been integral to our understanding of the fundamentals of muscle physiology. As they are refined and adapted, they are poised to continue to inform the field for years to come.
Collapse
Affiliation(s)
- Lucas R Smith
- Departments of Neurobiology, Physiology and Behavior and Physical Medicine and Rehabilitation, University of California, Davis, CA, USA
| | - Gretchen A Meyer
- Program in Physical Therapy and Departments of Neurology, Biomedical Engineering and Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
37
|
Li H, Wang J, Wu C, Wang L, Chen ZS, Cui W. The combination of disulfiram and copper for cancer treatment. Drug Discov Today 2020; 25:1099-1108. [PMID: 32320854 DOI: 10.1016/j.drudis.2020.04.003] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/31/2020] [Accepted: 04/09/2020] [Indexed: 12/20/2022]
Abstract
Disulfiram (DSF) is a thiuram derivative that was developed to treat alcoholism but was also found to have antitumor activity. Copper (Cu), as a trace metal, has important roles in the body. Numerous studies have shown that the combination of DSF and copper (DSF/Cu) greatly enhances its antitumor efficacy. Given that the efficacy of DSF is well established and its safety profile is understood, repurposing DSF as a new anticancer drug is a promising strategy. Here, we summarize the pharmacological effects of DSF and the role of Cu in cancer, and focus on the antitumor effect of DSF/Cu, especially the mechanisms involved in enhancing drug sensibility by targeting specific molecules. We also provide rational strategies for using DSF as a cancer therapy.
Collapse
Affiliation(s)
- Hong Li
- Department of Pharmacology, Shenyang Pharmaceutical University, 110016, Shenyang, PR China
| | - Jingyu Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, 110016, Shenyang, PR China
| | - Chunfu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, 110016, Shenyang, PR China
| | - Lihui Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, 110016, Shenyang, PR China.
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, NY 11439, USA.
| | - Wei Cui
- Department of Pharmacology, Shenyang Pharmaceutical University, 110016, Shenyang, PR China.
| |
Collapse
|
38
|
Kayama H, Tani H, Kitada S, Opasawatchai A, Okumura R, Motooka D, Nakamura S, Takeda K. BATF2 prevents T-cell-mediated intestinal inflammation through regulation of the IL-23/IL-17 pathway. Int Immunol 2020; 31:371-383. [PMID: 30753547 PMCID: PMC6528702 DOI: 10.1093/intimm/dxz014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 02/19/2019] [Indexed: 12/15/2022] Open
Abstract
Inappropriate activation of the IL-23 signaling pathway causes chronic inflammation through the induction of immunopathological Th17 cells in several tissues including the intestine, whereas adequate Th17 responses are essential for host defense against harmful organisms. In the intestinal lamina propria, IL-23 is primarily produced by innate myeloid cells including dendritic cells (DCs) and macrophages (Mϕs). However, the molecular mechanisms underlying the regulation of IL-23 production by these cells remains poorly understood. In this study, we demonstrated that BATF2 regulates intestinal homeostasis by inhibiting IL-23-driven T-cell responses. Batf2 was highly expressed in intestinal innate myeloid subsets, such as monocytes, CD11b+ CD64+ Mϕs and CD103+ DCs. Batf2-/- mice spontaneously developed colitis and ileitis with altered microbiota composition. In this context, IL-23, but not TNF-α and IL-10, was produced in high quantities by intestinal CD11b+ CD64+ Mϕs from Batf2-/- mice compared with wild-type mice. Moreover, increased numbers of IFN-γ+, IL-17+ and IFN-γ+ IL-17+ CD4+ T cells, but not IL-10+ CD4+ T cells, accumulated in the colons and small intestines of Batf2-/- mice. In addition, RORγt-expressing innate lymphoid cells were increased in Batf2-/- mice. Batf2-/-Rag2-/- mice showed a reduction in intestinal inflammation present in Batf2-/- mice. Furthermore, the high numbers of intestinal IL-17+ and IFN-γ+ IL-17+ CD4+ T cells were markedly reduced in Batf2-/- mice when introducing Il23a deficiency, which was associated with the abrogation of intestinal inflammation. These results indicated that BATF2 in innate myeloid cells is a key molecule for the suppression of IL-23/IL-17 pathway-mediated adaptive intestinal pathology.
Collapse
Affiliation(s)
- Hisako Kayama
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan.,WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Core Research for Evolutional Science and Technology, Japan Agency for Medical Research and Development, Tokyo, Japan
| | - Haruka Tani
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan.,WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Core Research for Evolutional Science and Technology, Japan Agency for Medical Research and Development, Tokyo, Japan
| | - Shoko Kitada
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan.,WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Core Research for Evolutional Science and Technology, Japan Agency for Medical Research and Development, Tokyo, Japan
| | - Anunya Opasawatchai
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan.,Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | - Ryu Okumura
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan.,WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Core Research for Evolutional Science and Technology, Japan Agency for Medical Research and Development, Tokyo, Japan
| | - Daisuke Motooka
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Shota Nakamura
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
| | - Kiyoshi Takeda
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan.,WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Core Research for Evolutional Science and Technology, Japan Agency for Medical Research and Development, Tokyo, Japan.,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
| |
Collapse
|
39
|
Wang H, Li Y, Shi G, Wang Y, Lin Y, Wang Q, Zhang Y, Yang Q, Dai L, Cheng L, Su X, Yang Y, Zhang S, Li Z, Li J, Wei Y, Yu D, Deng H. A Novel Antitumor Strategy: Simultaneously Inhibiting Angiogenesis and Complement by Targeting VEGFA/PIGF and C3b/C4b. Mol Ther Oncolytics 2020; 16:20-29. [PMID: 31909182 PMCID: PMC6940616 DOI: 10.1016/j.omto.2019.12.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 12/09/2019] [Indexed: 02/05/2023] Open
Abstract
Therapeutic antibodies targeting vascular endothelial growth factor (VEGF) have become a critical regimen for tumor therapy, but the efficacy of monotherapy is usually limited by drug resistance and multiple angiogenic mechanisms. Complement proteins are becoming potential candidates for cancer-targeted therapy based on their role in promoting cancer progression and angiogenesis. However, the antitumor abilities of simultaneous VEGF and complement blockade were unknown. We generated a humanized soluble VEGFR-Fc fusion protein (VID) binding VEGFA/PIGF and a CR1-Fc fusion protein (CID) targeting C3b/C4b. Both VID and CID had good affinities to their ligands and showed effective bioactivities. In vitro, angiogenesis effects induced by VEGF and hemolysis induced by complement were inhibited by VID and CID, respectively. Further, VID and CID confer a synergetic therapeutic effect in a colitis-associated colorectal cancer (CAC) model and an orthotopic 4T1 breast cancer model. Mechanically, combination therapy inhibited tumor angiogenesis, cell proliferation, and MDSC infiltration in the tumor microenvironment and promoted tumor cell apoptosis. Our study offers a novel therapeutic strategy for anti-VEGF-resistant tumors and chronic-inflammation-associated tumors.
Collapse
Affiliation(s)
- Huiling Wang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yiming Li
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Innovent Biologics (Suzhou) Co., Ltd., Suzhou, Jiangsu 215000, China
| | - Gang Shi
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yuan Wang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yi Lin
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Qin Wang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yujing Zhang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Qianmei Yang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Lei Dai
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Lin Cheng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xiaolan Su
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yang Yang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Shuang Zhang
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Zhi Li
- Innovent Biologics (Suzhou) Co., Ltd., Suzhou, Jiangsu 215000, China
| | - Jia Li
- Innovent Biologics (Suzhou) Co., Ltd., Suzhou, Jiangsu 215000, China
| | - Yuquan Wei
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Dechao Yu
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Hongxin Deng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
40
|
Zhang W, Dai L, Li X, Li Y, Hung Yap MK, Liu L, Deng H. SARI prevents ocular angiogenesis and inflammation in mice. J Cell Mol Med 2020; 24:4341-4349. [PMID: 32119762 PMCID: PMC7171405 DOI: 10.1111/jcmm.15096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/22/2019] [Accepted: 11/26/2019] [Indexed: 02/05/2023] Open
Abstract
SARI (Suppressor of AP‐1, regulated by IFN‐β) is known to play an important role in some systemic disease processes such an inflammatory conditions and cancer. We hypothesize that SARI may also play a role in ocular diseases involving inflammation and neovascularization. To explore our hypothesis, further, we investigated an endotoxin‐induced uveitis (EIU) and experimental argon laser‐induced choroidal neovascularization (CNV) model in SARI wild‐type (SARIWT) and SARI‐deficient (SARI−/−) mice. Through imaging, morphological and immunohistochemical (IHC) studies, we found that SARI deficiency exacerbated the growth of CNV. More VEGF‐positive cells were presented in the retina of SARI−/− mice with CNV. Compared to SARIWT mice, more inflammatory cells infiltrated the ocular anterior segment and posterior segments in SARI−/− mice with EIU. Collectively, the results point to a potential dual functional role of SARI in inflammatory ocular diseases, suggesting that SARI could be a potential therapy target for ocular inflammation and neovascularization.
Collapse
Affiliation(s)
- Wenqiu Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China.,Research Laboratory of Ophthalmology and Vision Sciences, West China Hospital, Sichuan University, Chengdu, China.,Department of Optometry and Visual Science, West China Hospital, Sichuan University, Chengdu, China
| | - Lei Dai
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xun Li
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China.,Research Laboratory of Ophthalmology and Vision Sciences, West China Hospital, Sichuan University, Chengdu, China
| | - Yiming Li
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | | | - Longqian Liu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China.,Department of Optometry and Visual Science, West China Hospital, Sichuan University, Chengdu, China
| | - Hongxin Deng
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
41
|
An N, Zhao Y, Lan H, Zhang M, Yin Y, Yi C. SEZ6L2 knockdown impairs tumour growth by promoting caspase-dependent apoptosis in colorectal cancer. J Cell Mol Med 2020; 24:4223-4232. [PMID: 32105413 PMCID: PMC7171412 DOI: 10.1111/jcmm.15082] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/12/2020] [Accepted: 02/08/2020] [Indexed: 02/05/2023] Open
Abstract
Seizure‐related 6 homolog (mouse)‐like 2 (SEZ6L2) was shown to be involved in transcription of a type 1 transmembrane protein for regulating cell fate. Until now, the expression and function of SEZ6L2 in various cancers, including colorectal cancer (CRC), were unclear. In the present study, we determined the expression of SEZ6L2 in a tissue microarray from patients with CRC and then, analysed the correlation between SEZ6L2 expression and the prognosis of the patients. Furthermore, the potential function of SEZ6L2 in CRC was determined using cell counting kit, colony formation assay and xenograft model in vitro and in vivo. Flow cytometry, Western blotting, immunohistochemical staining and a blocking experiment were employed to investigate the underlying mechanism of SEZ6L2 regulating CRC growth. Our results indicated that SEZ6L2 was significantly up‐regulated in tumour tissues of patients with CRC compared with adjacent normal tissues. Up‐regulation of SEZ6L2 was correlated with a poor prognosis in patients with CRC. In vitro experiments suggested that the knockdown of SEZ6L2 inhibits CRC cell growth and colony formation, but it has no significant impact on the invasion. The antitumour effects of shSEZ6L2 were also confirmed by a xenograft model. Investigations of the mechanisms indicated that the knockdown of SEZ6L2 impairs the growth of the CRC cells by inducing caspase‐dependent apoptosis, which was mediated by mitochondria‐related proteins. Furthermore, SEZ6L2 expression was inversely correlated with the expression of cytochrome C in malignant tissues in patients with CRC. Collectively, the present study indicates that SEZ6L2 is a potential prognosis biomarker and therapy target for CRC.
Collapse
Affiliation(s)
- Ning An
- Department of Abdominal Cancer, West China Hospital, West China Clinical Medical School, Sichuan University, Chengdu, China.,Cancer Center, Academy of Medical Sciences and Sichuan Provincial People's Hospital, Affiliated Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Yaqin Zhao
- Department of Abdominal Cancer, West China Hospital, West China Clinical Medical School, Sichuan University, Chengdu, China
| | - Haitao Lan
- Cancer Center, Academy of Medical Sciences and Sichuan Provincial People's Hospital, Affiliated Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Ming Zhang
- Cancer Center, Academy of Medical Sciences and Sichuan Provincial People's Hospital, Affiliated Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Yuan Yin
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Cheng Yi
- Department of Abdominal Cancer, West China Hospital, West China Clinical Medical School, Sichuan University, Chengdu, China
| |
Collapse
|
42
|
Zhou J, Chen P, Li Z, Zuo Q. Gene delivery of TIPE2 attenuates collagen-induced arthritis by modulating inflammation. Int Immunopharmacol 2020; 79:106044. [PMID: 31863922 DOI: 10.1016/j.intimp.2019.106044] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 11/05/2019] [Accepted: 11/10/2019] [Indexed: 12/28/2022]
|
43
|
Feng Y, Pan L, Zhang B, Huang H, Ma H. BATF acts as an oncogene in non-small cell lung cancer. Oncol Lett 2019; 19:205-210. [PMID: 31897131 PMCID: PMC6924102 DOI: 10.3892/ol.2019.11075] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/27/2019] [Indexed: 12/26/2022] Open
Abstract
One of the main causes of cancer incidence and mortality worldwide is lung cancer. This study focused on the function of basic leucine zipper ATF-like transcription factor (BATF) in non-small cell lung cancer (NSCLC). Using NSCLC patient data from The Cancer Genome Atlas, the present study demonstrated that BATF expression in NSCLC tissues was significantly higher compared with that in adjacent non-tumor tissues (P=6.56×10−6). Lentivirus-mediated short hairpin RNA (shRNA) was used to knock down BATF expression in the human A549 NSCLC cell line and assessed by reverse transcription-quantitative PCR and western blotting. Cell proliferation was evaluated by MTT assay and Celigo imaging cytometry. Apoptosis was detected by fluorescence-activated cell sorting and caspase 3/7 activity analysis. The results revealed that knockdown of BATF inhibited the proliferation of A549 cells. Compared with that of the control group, the apoptosis rate of the BATF-shRNA group was significantly higher. In summary, knockdown of BATF inhibited the proliferation of A549 cells and promoted apoptosis. These results provide important information about the underlying mechanism of the pathogenesis of NSCLC.
Collapse
Affiliation(s)
- Yu Feng
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Liangbin Pan
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Biao Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Haitao Huang
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Haitao Ma
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| |
Collapse
|
44
|
Rosquete MR, Worden N, Drakakaki G. AtTRAPPC11 is involved in TRAPPIII mediated control of post-Golgi protein trafficking. PLANT SIGNALING & BEHAVIOR 2019; 14:1676631. [PMID: 31610744 PMCID: PMC6867184 DOI: 10.1080/15592324.2019.1676631] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 09/28/2019] [Accepted: 10/01/2019] [Indexed: 06/10/2023]
Abstract
The plant trans-Golgi Network/Early Endosome (TGN/EE), as an organizer of vesicle trafficking, fulfills a crucial role for plant development and adaptation. Because it coordinates the transport of cell material along different routes, it is expected that a number of TGN/EE associated factors function in the rapid organization of post-Golgi trafficking to ensure that proteins reach their destination. The roles of Transport Protein Particle (TRAPP) complexes in the regulation of plant post-Golgi trafficking start to emerge. We previously demonstrated that the plant TRAPPIII complex is involved in maintenance of TGN organization and function and has a role in endocytic trafficking mediated by the SYP61 TGN/EE compartment. Here we show that attrappc11 mutants display accumulation of the plasma membrane resident proteins CESA6, BRI1 and PIP1;4 in aberrant intracellular compartments. This adds further insights into the functions of TRAPPIII as a regulators of post-Golgi/endosomal traffic.
Collapse
Affiliation(s)
| | - Natasha Worden
- Department of Plant Sciences, University of California, Davis, CA, USA
| | - Georgia Drakakaki
- Department of Plant Sciences, University of California, Davis, CA, USA
| |
Collapse
|
45
|
Dai L, Liu Y, Yin Y, Li J, Dong Z, Chen N, Cheng L, Wang H, Fang C, Lin Y, Shi G, Zhang H, Fan P, Su X, Zhang S, Yang Y, Yang L, Huang W, Zhou Z, Yu D, Deng H. SARI suppresses colitis-associated cancer development by maintaining MCP-1-mediated tumour-associated macrophage recruitment. J Cell Mol Med 2019; 24:189-201. [PMID: 31578820 PMCID: PMC6933368 DOI: 10.1111/jcmm.14699] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/28/2019] [Accepted: 09/06/2019] [Indexed: 02/05/2023] Open
Abstract
SARI (suppressor of AP‐1, regulated by IFN) impaired tumour growth by promoting apoptosis and inhibiting cell proliferation and tumour angiogenesis in various cancers. However, the role of SARI in regulating tumour‐associated inflammation microenvironment is still elusive. In our study, the colitis‐dependent and ‐independent primary model were established in SARI deficiency mice and immuno‐reconstructive mice to investigate the functional role of SARI in regulating tumour‐associated inflammation microenvironment and primary colon cancer formation. The results have shown that SARI deficiency promotes colitis‐associated cancer (CAC) development only in the presence of colon inflammation. SARI inhibited tumour‐associated macrophages (TAM) infiltration in colon tissues, and SARI deficiency in bone marrow cells has no observed role in the promotion of intestinal tumorigenesis. Mechanism investigations indicated that SARI down‐regulates p‐STAT1 and STAT1 expression in colon cancer cells, following inhibition of MCP‐1/CCR2 axis activation during CAC development. Inverse correlations between SARI expression and macrophage infiltration, MCP‐1 expression and p‐STAT1 expression were also demonstrated in colon malignant tissues. Collectively, our results prove the inhibition role of SARI in colon cancer formation through regulating TAM infiltration.
Collapse
Affiliation(s)
- Lei Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Yi Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Yuan Yin
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Junshu Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Zhexu Dong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Na Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Lin Cheng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Huiling Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Chao Fang
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Yi Lin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Gang Shi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Hantao Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Ping Fan
- Department of Clinical Research Management, West China-Liverpool Biomedical Research Center, West China Hospital, West China Biobanks, Sichuan University, Chengdu, China
| | - Xiaolan Su
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Shuang Zhang
- Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Lie Yang
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Wei Huang
- Department of Clinical Research Management, West China-Liverpool Biomedical Research Center, West China Hospital, West China Biobanks, Sichuan University, Chengdu, China
| | - Zongguang Zhou
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Dechao Yu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Hongxin Deng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| |
Collapse
|
46
|
BATF2 inhibits chemotherapy resistance by suppressing AP-1 in vincristine-resistant gastric cancer cells. Cancer Chemother Pharmacol 2019; 84:1279-1288. [DOI: 10.1007/s00280-019-03958-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 09/04/2019] [Indexed: 01/08/2023]
|
47
|
Lin X, Yang F, Qi X, Li Q, Wang D, Yi T, Yin R, Zhao X, Zhong X, Bian C. LncRNA DANCR promotes tumor growth and angiogenesis in ovarian cancer through direct targeting of miR-145. Mol Carcinog 2019; 58:2286-2296. [PMID: 31545000 DOI: 10.1002/mc.23117] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 09/03/2019] [Accepted: 09/04/2019] [Indexed: 12/17/2022]
Abstract
Differentiation antagonizing non-protein coding RNA (DANCR) is a newly identified oncogenic long noncoding RNA found in various cancers. However, the functional role of DANCR in tumor angiogenesis and the underlying mechanisms are still unclear. The expression of DANCR was determined in ovarian malignant tissues and cell lines. The functional role of DANCR in tumor angiogenesis was revealed by the following methods: CD31 staining of ovarian tumor tissues, matrigel-plug assay tissues, HUVEC-related tube formation assay, and invasion assay. Enzyme-linked immunosorbent assay, Western blotting, luciferase assay, and rescue experiments were used to investigate the underlying mechanisms of DANCR-regulating angiogenesis. DANCR was upregulated in ovarian malignant tissues and ovarian cancer cells. Knockdown of DANCR efficiently impaired ovarian tumor growth through inhibition of tumor angiogenesis. Furthermore, the conditional culture medium from DANCR-knockdown ovarian cells significantly inhibited tube formation and invasion of HUVEC in vitro. Mechanistic investigation indicated that vascular endothelial growth factor A (VEGF-A, VEGF) plays a crucial role during DANCR inhibition of tumor angiogenesis in ovarian cancer. Further results demonstrated that miR-145 is the direct binding target of DANCR during regulation of VEGF expression and tumor angiogenesis in ovarian cancer cells. Collectively, DANCR plays a promotional role in tumor angiogenesis in ovarian cancer through regulation of miR-145/VEGF axis. Therefore, DANCR may be a novel therapy target for ovarian cancer.
Collapse
Affiliation(s)
- Xiaojuan Lin
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Fan Yang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xiaorong Qi
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Qingli Li
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Danqing Wang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Tao Yi
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Rutie Yin
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xia Zhao
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xiaomin Zhong
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Center for Stem Cell Biology and Tissue Engineering, Department of Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Ce Bian
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
48
|
Meijers WC, Maglione M, Bakker SJL, Oberhuber R, Kieneker LM, de Jong S, Haubner BJ, Nagengast WB, Lyon AR, van der Vegt B, van Veldhuisen DJ, Westenbrink BD, van der Meer P, Silljé HHW, de Boer RA. Heart Failure Stimulates Tumor Growth by Circulating Factors. Circulation 2019; 138:678-691. [PMID: 29459363 DOI: 10.1161/circulationaha.117.030816] [Citation(s) in RCA: 251] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Heart failure (HF) survival has improved, and nowadays, many patients with HF die of noncardiac causes, including cancer. Our aim was to investigate whether a causal relationship exists between HF and the development of cancer. METHODS HF was induced by inflicting large anterior myocardial infarction in APCmin mice, which are prone to developing precancerous intestinal tumors, and tumor growth was measured. In addition, to rule out hemodynamic impairment, a heterotopic heart transplantation model was used in which an infarcted or sham-operated heart was transplanted into a recipient mouse while the native heart was left in situ. After 6 weeks, tumor number, volume, and proliferation were quantified. Candidate secreted proteins were selected because they were previously associated both with (colon) tumor growth and with myocardial production in post-myocardial infarction proteomic studies. Myocardial gene expression levels of these selected candidates were analyzed, as well as their proliferative effects on HT-29 (colon cancer) cells. We validated these candidates by measuring them in plasma of healthy subjects and patients with HF. Finally, we associated the relation between cardiac specific and inflammatory biomarkers and new-onset cancer in a large, prospective general population cohort. RESULTS The presence of failing hearts, both native and heterotopically transplanted, resulted in significantly increased intestinal tumor load of 2.4-fold in APCmin mice (all P<0.0001). The severity of left ventricular dysfunction and fibrotic scar strongly correlated with tumor growth ( P=0.002 and P=0.016, respectively). We identified several proteins (including serpinA3 and A1, fibronectin, ceruloplasmin, and paraoxonase 1) that were elevated in human patients with chronic HF (n=101) compared with healthy subjects (n=180; P<0.001). Functionally, serpinA3 resulted in marked proliferation effects in human colon cancer (HT-29) cells, associated with Akt-S6 phosphorylation. Finally, elevated cardiac and inflammation biomarkers in apparently healthy humans (n=8319) were predictive of new-onset cancer (n=1124) independently of risk factors for cancer (age, smoking status, and body mass index). CONCLUSIONS We demonstrate that the presence of HF is associated with enhanced tumor growth and that this is independent of hemodynamic impairment and could be caused by cardiac excreted factors. A diagnosis of HF may therefore be considered a risk factor for incident cancer.
Collapse
Affiliation(s)
- Wouter C Meijers
- Department of Cardiology (W.C.M., D.J.v.V., B.D.W., P.v.d.M., H.H.W.S., R.A.d.B.)
| | - Manuel Maglione
- Centre of Operative Medicine, Department of Visceral, Transplant and Thoracic Surgery (M.M., R.O.)
| | - Stephan J L Bakker
- Department of Internal Medicine, Division of Nephrology (S.J.L.B., L.M.K.), University Medical Center Groningen, University of Groningen, The Netherlands
| | - Rupert Oberhuber
- Centre of Operative Medicine, Department of Visceral, Transplant and Thoracic Surgery (M.M., R.O.)
| | - Lyanne M Kieneker
- Department of Internal Medicine, Division of Nephrology (S.J.L.B., L.M.K.), University Medical Center Groningen, University of Groningen, The Netherlands
| | | | - Bernhard J Haubner
- Department of Internal Medicine III (Cardiology and Angiology) (B.J.H.), Medical University of Innsbruck, Austria
| | | | - Alexander R Lyon
- National Heart and Lung Institute, Imperial College London and Royal Brompton Hospital, United Kingdom (A.R.L.)
| | | | | | - B Daan Westenbrink
- Department of Cardiology (W.C.M., D.J.v.V., B.D.W., P.v.d.M., H.H.W.S., R.A.d.B.)
| | - Peter van der Meer
- Department of Cardiology (W.C.M., D.J.v.V., B.D.W., P.v.d.M., H.H.W.S., R.A.d.B.)
| | - Herman H W Silljé
- Department of Cardiology (W.C.M., D.J.v.V., B.D.W., P.v.d.M., H.H.W.S., R.A.d.B.)
| | - Rudolf A de Boer
- Department of Cardiology (W.C.M., D.J.v.V., B.D.W., P.v.d.M., H.H.W.S., R.A.d.B.)
| |
Collapse
|
49
|
SARI attenuates colon inflammation by promoting STAT1 degradation in intestinal epithelial cells. Mucosal Immunol 2019; 12:1130-1140. [PMID: 31182817 DOI: 10.1038/s41385-019-0178-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/30/2019] [Accepted: 05/26/2019] [Indexed: 02/05/2023]
Abstract
SARI functions as a suppressor of colon cancer and predicts survival of colon cancer patients, but its role in regulating colitis has not been characterized. Here we show that SARI-/- mice were highly susceptible to colitis, which was associated with enhanced macrophage infiltration and inflammatory cytokine production. Bone marrow reconstitution experiments demonstrated that disease susceptibility was not dependent on the deficiency of SARI in the immune compartment but on the protective role of SARI in the intestinal epithelial cells (IECs). Furthermore, SARI deficiency enhanced Chemokine (C-C motif) Ligand 2 (CCL2) production and knockout of CCR2 blocks the promoting role of SARI deficiency on colitis. Mechanistically, SARI directly targets and promotes signal transducer and activator of transcription 1 (STAT1) degradation in IECs, followed by persistent inactivation of the STAT1/CCL2 transcription complex. In summary, SARI attenuated colitis in mice by impairing colitis-dependent STAT1/CCL2 transcriptional activation in IECs and macrophages recruitment in colon tissue.
Collapse
|
50
|
Xiong W, Qin J, Cai X, Xiong W, Liu Q, Li C, Ju Y, Wang Q, Li Y, Yang Y. Overexpression LINC01082 suppresses the proliferation, migration and invasion of colon cancer. Mol Cell Biochem 2019; 462:33-40. [PMID: 31432387 DOI: 10.1007/s11010-019-03607-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/10/2019] [Indexed: 02/08/2023]
Abstract
Long non-coding RNAs (lncRNAs) are emerging as pivotal regulators in human cancer. LINC01082 was expressed as decreased in colon cancer by previous lncRNA-seq result and TCGA database, however, the role and function of LINC0182 is not clear in colon cancer. Here, we aimed to explore the role of LINC01082 in colon cancer for exploring the etiopathogenesis of colon cancer. RT-qPCR for LINC01082 expression in tissues (colon cancer vs. their matched adjacent non-cancerous tissues, ANT, n = 39) and cells (colon cancer cells vs. normal colon cells, n = 4) were performed. CCK-8 assay for proliferation of colon cancer, Transwell assay for migration and invasion were carried out in sw480 and sw620 cells. The results revealed that LINC01082 was significantly decreased in tissues and cell lines of colon cancer. Overexpressed LINC01082 significantly suppressed the proliferation ability of colon cancer cells. The migration and invasion of colon cancer cells were also suppressed after LINC01082 overexpression. These findings demonstrated that LINC01082 may act in suppressing the incidence and development of colon cancer via suppressing cell proliferation, migration and invasion, indicating that LINC01082 may act as a new tumor suppressor and may be a promising therapy target for colon cancer.
Collapse
Affiliation(s)
- Wei Xiong
- Department of Radiation Oncology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, 519 Kunzhou Road, Xishan District, Kunming, 650100, Yunnan, People's Republic of China
| | - Jiyong Qin
- Department of Radiation Oncology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, 519 Kunzhou Road, Xishan District, Kunming, 650100, Yunnan, People's Republic of China
| | - Xinyi Cai
- Department of Colorectal Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, 519 Kunzhou Road, Xishan District, Kunming, 650100, Yunnan, People's Republic of China
| | - Wei Xiong
- Department of Colorectal Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, 519 Kunzhou Road, Xishan District, Kunming, 650100, Yunnan, People's Republic of China
| | - Qiuyan Liu
- Department of Radiation Oncology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, 519 Kunzhou Road, Xishan District, Kunming, 650100, Yunnan, People's Republic of China
| | - Cheng Li
- Department of Radiation Oncology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, 519 Kunzhou Road, Xishan District, Kunming, 650100, Yunnan, People's Republic of China
| | - Yunhe Ju
- Department of Radiation Oncology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, 519 Kunzhou Road, Xishan District, Kunming, 650100, Yunnan, People's Republic of China
| | - Qiaoli Wang
- Department of Radiation Oncology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, 519 Kunzhou Road, Xishan District, Kunming, 650100, Yunnan, People's Republic of China
| | - Yunfeng Li
- Department of Colorectal Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, 519 Kunzhou Road, Xishan District, Kunming, 650100, Yunnan, People's Republic of China.
| | - Yi Yang
- Department of Radiation Oncology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, 519 Kunzhou Road, Xishan District, Kunming, 650100, Yunnan, People's Republic of China.
| |
Collapse
|