1
|
Xing P, Chen Z, Zhu W, Lin B, Quan M. NRF3 suppresses the malignant progression of TNBC by promoting M1 polarization of macrophages via ROS/HMGB1 axis. Cancer Biol Ther 2024; 25:2416221. [PMID: 39443820 PMCID: PMC11509002 DOI: 10.1080/15384047.2024.2416221] [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: 03/08/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is a highly aggressive form of breast cancer. Due to its lack of targeted therapy options, TNBC remains a significant clinical challenge. In this study, we investigated the role of nuclear respiratory factor 3 (NRF3) and high-mobility group box 1 (HMGB1) in the progression of TNBC. METHODS The study analyzed NRF3's clinical expression, differentially expressed genes (DEGs), and immune infiltration in TNBC using the TCGA database and bioinformatics tools. Cellular functions of MDA-MB-468 and Hs578t cells were evaluated through MTT, colony formation, transwell, flow cytometry, and western blotting. The regulatory function of NRF3 in TNBC cell lines was assessed using Immunofluorescence, Immunohistochemistry, qRT-PCR, CHIP, luciferase assay, and ELISA. Moreover, a xenograft model was established to investigate the role of NRF3 in TNBC in vivo. RESULTS Low expression of NRF3 in TNBC tumors was associated with unfavorable prognosis and transcripts from tumors with higher NRF3 levels were enriched in oxidative stress and immune-related pathways. The subsequent gain- and loss-functional experiments indicated that NRF3 overexpression significantly suppressed malignant phenotypes, MAPK/ERK signaling pathways, and epithelial-mesenchymal transition (EMT), whereas it promoted reactive oxygen species (ROS) levels in TNBC. Further mechanistic exploration showed that NRF3 inhibited TNBC cell function by regulating oxidative stress-related genes to inhibit the MAPK/ERK signaling pathway by promoting the release of HMGB1 via ROS, thereby promoting M1 macrophage polarization. CONCLUSION NRF3 promotes M1 macrophage polarization through the ROS/HMGB1 axis, thereby inhibiting the malignant progression of TNBC. It is expected to become a therapeutic biomarker for TNBC.
Collapse
Affiliation(s)
- Ping Xing
- Department of Surgical Oncology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
- Department of Surgical Oncology, Enze Hospital, Taizhou Enze Medical Center, Taizhou, Zhejiang, China
| | - Zhenzhen Chen
- Department of Ultrasound, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Wenbo Zhu
- Department of Surgical Oncology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
- Department of Surgical Oncology, Enze Hospital, Taizhou Enze Medical Center, Taizhou, Zhejiang, China
| | - Bangyi Lin
- Department of Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Mingming Quan
- Department of Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| |
Collapse
|
2
|
Xiong G, Li J, Yao F, Yang F, Xiang Y. New insight into the CNC-bZIP member, NFE2L3, in human diseases. Front Cell Dev Biol 2024; 12:1430486. [PMID: 39149514 PMCID: PMC11325725 DOI: 10.3389/fcell.2024.1430486] [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: 05/10/2024] [Accepted: 07/08/2024] [Indexed: 08/17/2024] Open
Abstract
Nuclear factor erythroid 2 (NF-E2)-related factor 3 (NFE2L3), a member of the CNC-bZIP subfamily and widely found in a variety of tissues, is an endoplasmic reticulum (ER) membrane-anchored transcription factor that can be released from the ER and moved into the nucleus to bind the promoter region to regulate a series of target genes involved in antioxidant, inflammatory responses, and cell cycle regulation in response to extracellular or intracellular stress. Recent research, particularly in the past 5 years, has shed light on NFE2L3's participation in diverse biological processes, including cell differentiation, inflammatory responses, lipid homeostasis, immune responses, and tumor growth. Notably, NFE2L3 has been identified as a key player in the development and prognosis of multiple cancers including colorectal cancer, thyroid cancer, breast cancer, hepatocellular carcinoma, gastric cancer, renal cancer, bladder cancer, esophageal squamous cell carcinoma, T cell lymphoblastic lymphoma, pancreatic cancer, and squamous cell carcinoma. Furthermore, research has linked NFE2L3 to other cancers such as lung adenocarcinoma, malignant pleural mesothelioma, ovarian cancer, glioblastoma multiforme, and laryngeal carcinoma, indicating its potential as a target for innovative cancer treatment approaches. Therefore, to gain a better understanding of the role of NFE2L3 in disease, this review offers insights into the discovery, structure, function, and recent advancements in the study of NFE2L3 to lay the groundwork for the development of NFE2L3-targeted cancer therapies.
Collapse
Affiliation(s)
- Guanghui Xiong
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
- Department of Children Rehabilitation, Maternal and Child Health Hospital of Jintang County, Chendu, Sichuan, China
| | - Jie Li
- Department of Anaesthesia, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Fuli Yao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Fang Yang
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
- Department of Pathophysiology, College of High Altitude Military Medicine, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yuancai Xiang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
3
|
Cai BQ, Chen WM, Chen MW, Chen YH, Tang JC. Nrf3 alleviates oxidative stress and promotes the survival of colon cancer cells by activating AKT/BCL-2 signal pathway. Open Life Sci 2023; 18:20220790. [PMID: 38027228 PMCID: PMC10668112 DOI: 10.1515/biol-2022-0790] [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: 06/10/2023] [Revised: 10/24/2023] [Accepted: 11/02/2023] [Indexed: 12/01/2023] Open
Abstract
Oxidative stress is closely linked to tumor initiation and development, conferring a survival advantage to cancer cells. Therefore, understanding cancer cells' antioxidant molecular mechanisms is crucial to cancer therapy. In this study, we discovered that H2O2-induced oxidative stress increased Nrf3 expression in colon cancer cells. Overexpression of Nrf3 decreased H2O2-mediated cytotoxicity and apoptosis. Furthermore, Nrf3 reduced reactive oxygen species levels and malondialdehyde concentrations after H2O2 treatment. Mechanistically, H2O2-mediated cell apoptosis involves multiple signaling proteins, including Akt, bcl-2, JNK, and p38. An increase in Nrf3 expression in colon cancer cells treated with H2O2 partly reversed Akt/Bcl-2 inhibition, whereas it decreased activation of p38 and JNK. In addition, we found that increasing Nrf3 decreased stress-associated chemical-induced cell death, resulting in drug resistance. According to these results, Nrf3 is critical for drug resistance and oxidant adaptation.
Collapse
Affiliation(s)
- Bi-Qing Cai
- Institute of Basic Medicine and Forensics Medicine, North Sichuan Medical College, Fu Jiang Road 234, Shunqing District, Nanchong, Sichuan, 637000, China
| | - Wan-Meng Chen
- Institute of Basic Medicine and Forensics Medicine, North Sichuan Medical College, Fu Jiang Road 234, Shunqing District, Nanchong, Sichuan, 637000, China
| | - Meng-Wei Chen
- Institute of Basic Medicine and Forensics Medicine, North Sichuan Medical College, Fu Jiang Road 234, Shunqing District, Nanchong, Sichuan, 637000, China
| | - Ya-Hui Chen
- Institute of Basic Medicine and Forensics Medicine, North Sichuan Medical College, Fu Jiang Road 234, Shunqing District, Nanchong, Sichuan, 637000, China
| | - Jian-Cai Tang
- Institute of Basic Medicine and Forensics Medicine, North Sichuan Medical College, Fu Jiang Road 234, Shunqing District, Nanchong, Sichuan, 637000, China
- Key Laboratory of Metabolic Drugs and Biological Products, Nanchong, China
| |
Collapse
|
4
|
Gurri S, Siegenthaler B, Cangkrama M, Restivo G, Huber M, Saliba J, Dummer R, Blank V, Hohl D, Werner S. NRF3 suppresses squamous carcinogenesis, involving the unfolded protein response regulator HSPA5. EMBO Mol Med 2023; 15:e17761. [PMID: 37807968 PMCID: PMC10630885 DOI: 10.15252/emmm.202317761] [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: 03/28/2023] [Revised: 09/05/2023] [Accepted: 09/15/2023] [Indexed: 10/10/2023] Open
Abstract
Epithelial skin cancers are extremely common, but the mechanisms underlying their malignant progression are still poorly defined. Here, we identify the NRF3 transcription factor as a tumor suppressor in the skin. NRF3 protein expression is strongly downregulated or even absent in invasively growing cancer cells of patients with basal and squamous cell carcinomas (BCC and SCC). NRF3 deficiency promoted malignant conversion of chemically induced skin tumors in immunocompetent mice, clonogenic growth and migration of human SCC cells, their invasiveness in 3D cultures, and xenograft tumor formation. Mechanistically, the tumor-suppressive effect of NRF3 involves HSPA5, a key regulator of the unfolded protein response, which we identified as a potential NRF3 interactor. HSPA5 levels increased in the absence of NRF3, thereby promoting cancer cell survival and migration. Pharmacological inhibition or knock-down of HSPA5 rescued the malignant features of NRF3-deficient SCC cells in vitro and in preclinical mouse models. Together with the strong expression of HSPA5 in NRF3-deficient cancer cells of SCC patients, these results suggest HSPA5 inhibition as a treatment strategy for these malignancies in stratified cancer patients.
Collapse
Affiliation(s)
- Selina Gurri
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Beat Siegenthaler
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Michael Cangkrama
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Gaetana Restivo
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Marcel Huber
- Service of Dermatology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - James Saliba
- Lady Davis Institute for Medical Research, McGill University, Montreal, Canada
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Volker Blank
- Lady Davis Institute for Medical Research, McGill University, Montreal, Canada
| | - Daniel Hohl
- Service of Dermatology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Sabine Werner
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
5
|
Chen WM, Hu QY, Hou W, Chen MW, Chen YH, Tang JC. Nrf3 promotes the proliferation and migration of triple‑negative breast cancer by activating PI3K/AKT/mTOR and epithelial‑mesenchymal transition. Oncol Lett 2023; 26:443. [PMID: 37720674 PMCID: PMC10502936 DOI: 10.3892/ol.2023.14030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/31/2023] [Indexed: 09/19/2023] Open
Abstract
Nuclear factor erythroid 2-related factor 3 (Nrf3) is increasingly implicated in multiple types of cancer; however, its function in triple-negative breast cancer (TNBC) remains unclear. This study aimed to examine the role of Nrf3 in TNBC. Compared with adjacent normal tissues, TNBC tissues expressed higher levels of Nrf3, and its expression was negatively correlated with survival time. Additionally, Nrf3 knockdown reduced the proliferation and migration of TNBC cells, whereas overexpression of Nrf3 had the opposite effects in vitro and in vivo. Moreover, functional enrichment of TNBC cells overexpressing Nrf3 allowed for the identification of numerous genes and pathways that were altered following Nrf3 overexpression. Further study showed that overexpression of Nrf3 activated the PI3K/AKT/mTOR signaling pathway and regulated the expression of proteins associated with epithelial-mesenchymal transition. Nrf3 was found to directly bind to p110α promoter regions, as evidenced by luciferase reporter and chromatin immunoprecipitation assays. Furthermore, PI3K inhibitors partially decreased the proliferation and migration of the Nrf3 overexpressing TNBC cells. In conclusion, Nrf3 enhances cellular proliferation and migration by activating PI3K/AKT/mTOR signaling pathways, highlighting a novel therapeutic target for TNBC.
Collapse
Affiliation(s)
- Wan-Meng Chen
- Department of Biochemistry, Institute of Basic Medicine and Forensics Medicine, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Qing-Yong Hu
- Department of Biochemistry, Institute of Basic Medicine and Forensics Medicine, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Wei Hou
- Department of Biochemistry, Institute of Basic Medicine and Forensics Medicine, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Meng-Wei Chen
- Department of Biochemistry, Institute of Basic Medicine and Forensics Medicine, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Ya-Hui Chen
- Department of Biochemistry, Institute of Basic Medicine and Forensics Medicine, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Jian-Cai Tang
- Department of Biochemistry, Institute of Basic Medicine and Forensics Medicine, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| |
Collapse
|
6
|
Huang P, Zhou X, Zheng M, Yu Y, Jin G, Zhang S. Regulatory T cells are associated with the tumor immune microenvironment and immunotherapy response in triple-negative breast cancer. Front Immunol 2023; 14:1263537. [PMID: 37767092 PMCID: PMC10521732 DOI: 10.3389/fimmu.2023.1263537] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Introduction Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer with a high risk of distant metastasis, an extremely poor prognosis, and a high risk of death. Regulatory T cells (Tregs) contribute to the formation of a tumor immunosuppressive microenvironment, which plays an important role in the progression and treatment resistance of TNBC. Methods A public single-cell sequencing dataset demonstrated increased infiltration of Tregs in TNBC tissues relative to normal breast tissue. Weighted gene co-expression network analysis was used to identify Treg infiltration-related modules for METABRIC TNBC samples. Subsequently, we obtained two Treg infiltration-associated clusters of TNBC by applying consensus clustering and further constructed a prognostic model based on this Treg infiltration-associated gene module. The ability of the selected gene in the prognostic model, thymidine kinase-1 (TK1), to promote the progression of TNBC was evaluated in vitro. Results We concluded that two Treg infiltration-associated clusters had different prognoses and sensitivities to drugs commonly used in breast cancer treatment, and multi-omics analysis revealed that the two clusters had different copy number variations of key tumor progression genes. The 7-gene risk score based on TNBC Treg infiltration was a reliable prognostic indicator both in the training and validation cohorts. Moreover, patients with TNBC with high Treg infiltration-related scores lacked the activation of immune activation pathways and exhibited resistance to anti-PD1 immunotherapy. Knocking down TK1 led to impaired proliferation, migration, and invasion of TNBC cells in vitro. In addition, specimens from patients with TNBC with high TK1 expression showed significantly higher Treg infiltration in tumors. Results of spatial transcriptome analysis showed that TK1 positive cells mainly localize in tumor area, and Treg cell infiltration in TNBC tissues was associated with high expression of TK1. Pan-cancer analysis also demonstrated that TK1 is associated with poor prognosis and activation of proliferation pathways in multiple cancers. Discussion We established a prognostic model related to Treg infiltration and this model can be used to establish a clinically relevant classification of TNBC progression. Additionally, our work revealed the underestimable potential of TK1 as a tumor biomarker and immunotherapeutic target.
Collapse
Affiliation(s)
- Pengfei Huang
- Department of Surgical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Xinyue Zhou
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Minying Zheng
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
| | - Yongjun Yu
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
| | - Gongsheng Jin
- Department of Surgical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
| |
Collapse
|
7
|
Ren Y, Yang J, Ding Z, Zheng M, Qiu L, Tang A, Huang D. NFE2L3 drives hepatocellular carcinoma cell proliferation by regulating the proteasome-dependent degradation of ISGylated p53. Cancer Sci 2023; 114:3523-3536. [PMID: 37350063 PMCID: PMC10475773 DOI: 10.1111/cas.15887] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/11/2023] [Accepted: 05/25/2023] [Indexed: 06/24/2023] Open
Abstract
Nuclear factor erythroid 2-like 3 (NFE2L3) is a member of the cap 'n' collar basic-region leucine zipper (CNC-bZIP) transcription factor family that plays a vital role in modulating oxidation-reduction steady-state and proteolysis. Accumulating evidence suggests that NFE2L3 participates in cancer development; however, little is known about the mechanism by which NFE2L3 regulates hepatocellular carcinoma (HCC) cell growth. Here, we confirmed that NFE2L3 promotes HCC cell proliferation by acting as a transcription factor, which directly induces the expression of proteasome and interferon-stimulated gene 15 (ISG15) to enhance the proteasome-dependent degradation of ISGylated p53. Post-translational ISGylation abated the stability of p53 and facilitated HCC cell growth. In summary, we uncovered the pivotal role of NFE2L3 in promoting HCC cell proliferation during proteostasis. This finding may provide a new target for the clinical treatment of HCC.
Collapse
Affiliation(s)
- Yonggang Ren
- Institute of Basic Medicine and Forensic MedicineNorth Sichuan Medical CollegeNanchongChina
- Research Center of Clinical Medical SciencesAffiliated Hospital of North Sichuan Medical CollegeNanchongChina
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Institute of Translational Medicine, Shenzhen Second People's HospitalThe First Affiliated Hospital of Shenzhen UniversityShenzhenChina
| | - Jing Yang
- Institute of Basic Medicine and Forensic MedicineNorth Sichuan Medical CollegeNanchongChina
| | - Zhiran Ding
- Institute of Basic Medicine and Forensic MedicineNorth Sichuan Medical CollegeNanchongChina
| | - Menghua Zheng
- Institute of Basic Medicine and Forensic MedicineNorth Sichuan Medical CollegeNanchongChina
| | - Lu Qiu
- School of Pharmaceutical Sciences (Shenzhen)Shenzhen Campus of Sun Yat‐Sen UniversityShenzhenChina
| | - Aifa Tang
- Shenzhen Luohu Hospital GroupThe Third Affiliated Hospital of Shenzhen UniversityShenzhenChina
| | - Dandan Huang
- Institute of Basic Medicine and Forensic MedicineNorth Sichuan Medical CollegeNanchongChina
| |
Collapse
|
8
|
Papierniak-Wyglądała A, Lamch W, Jurewicz E, Nałęcz KA. The activity and surface presence of organic cation/carnitine transporter OCTN2 (SLC22A5) in breast cancer cells depends on AKT kinase. Arch Biochem Biophys 2023; 742:109616. [PMID: 37187422 DOI: 10.1016/j.abb.2023.109616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/06/2023] [Accepted: 04/26/2023] [Indexed: 05/17/2023]
Abstract
l-carnitine is indispensable for transfer of fatty acids to mitochondria for the process of β-oxidation, a process, whose significance in cancer has drawn attention in recent years. In humans majority of carnitine is delivered by diet and enters the cell due to activity of solute carriers (SLCs), mainly by ubiquitously expressed organic cation/carnitine transporter (OCTN2/SLC22A5). In control and cancer human breast epithelial cell lines the major fraction of OCTN2 is present as a not matured non-glycosylated form. Studies on overexpressed OCTN2 demonstrated an exclusive interaction with SEC24C, as the cargo-recognizing subunit of coatomer II in transporter exit from endoplasmic reticulum. Co-transfection with SEC24C dominant negative mutant completely abolished presence of the mature form of OCTN2, pointing to a possibility of trafficking regulation. SEC24C was previously shown to be phosphorylated by serine/threonine kinase AKT, known to be activated in cancer. Further studies on breast cell lines showed that inhibition of AKT with MK-2206 in control and cancer lines decreased level of OCTN2 mature form. Proximity ligation assay showed that phosphorylation of OCTN2 on threonine was significantly abolished by AKT inhibition with MK-2206. Carnitine transport was positively correlated with the level of OCTN2 phosphorylated by AKT on threonine moiety. The observed regulation of OCTN2 by AKT places this kinase in the center of metabolic control. This points to both proteins, AKT and OCTN2, as druggable targets, in particular in a combination therapy of breast cancer.
Collapse
Affiliation(s)
- Anna Papierniak-Wyglądała
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland.
| | - Weronika Lamch
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland.
| | - Ewelina Jurewicz
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland.
| | - Katarzyna A Nałęcz
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland.
| |
Collapse
|
9
|
Chen T, Xu B, Chen H, Sun Y, Song J, Sun X, Zhang X, Hua W. Transcription factor NFE2L3 promotes the proliferation of esophageal squamous cell carcinoma cells and causes radiotherapy resistance by regulating IL-6. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2022; 226:107102. [PMID: 36108571 DOI: 10.1016/j.cmpb.2022.107102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/23/2022] [Accepted: 08/29/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVE To scrutinize the impact of overexpression and interference of NFE2L3 on radiosensitivity of esophageal squamous cell carcinoma cells (ESCC) and its downstream mechanism and to assess whether NFE2L3 expression alters in vivo radiosensitivity of ESCC by developing a subcutaneous tumor model in mice. METHODS Through RNA-Seq, we compared the differentially expressed genes between the ECA-109R cell line and its parent ECA-109 cell line. The differentially expressed genes were selected and verified by qRT-PCR. Transfection of ESCC cell lines with NFE2L3 inhibitor or mimic lentivirus constructs was done to study the activity of NFE2L3. To assess the effect of NFE2L3 on cellular growth and proliferation, clonogenic survival assay, EdU incorporation assay, and CCK-8 assay were done after irradiation. To probe how many irradiated DNA double-strand breaks were produced, the corresponding intensity of γ-H2AX foci were detected by immunofluorescence. Apoptotic cells were assayed by flow cytometry assay after irradiation; To investigate the downstream genes of NFE2L3, we knocked NFE2L3, and RNA-Seq was used to find out the downstream genes. qRT-PCR and western blot ensued to score associated protein profiles. The in vivo ESCC cell radiosensitivity was scrutinized by nude mouse xenograft models. RESULTS The differential genes between ECA-109R cells and its parent ECA-109 cells were compared by qRT-PCR to unveil a significant increase in NFE2L3 expression. Functional analysis indicated that NFE2L3 increased radioresistance in ESCC cells. Then, through high-throughput sequencing and bioinformatics analysis, IL-6 was found to be a hub gene that played a role downstream of NFE2L3 and was verified by qRT-PCR, western blot, and double luciferase reporter gene experiment. NFE2L3 could regulate ESCC cell radiosensitivity via the IL-6-STAT3 signaling pathway, and downregulation of IL-6 expression could reverse the effects of highly expressed NFE2L3. In vivo tumor xenograft experiments confirmed that NFE2L3 affects the sensitivity to radiation therapy. CONCLUSION NFE2L3 can affect the radiosensitivity of ESCC cells through IL-6 transcription and IL-6/STAT3 signaling pathway. This makes NFE2L3 a putative target to regulate ESCC cell radiosensitivity.
Collapse
Affiliation(s)
- Tingting Chen
- Department of Oncology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu Province, PR China
| | - Bing Xu
- Department of Oncology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu Province, PR China
| | - Hui Chen
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, PR China
| | - Yuanyuan Sun
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, PR China
| | - Jiahang Song
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, PR China
| | - Xinchen Sun
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, PR China.
| | - Xizhi Zhang
- Department of Oncology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu Province, PR China.
| | - Wei Hua
- Department of Oncology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu Province, PR China.
| |
Collapse
|
10
|
Santonja Á, Moya-García AA, Ribelles N, Jiménez-Rodríguez B, Pajares B, Fernández-De Sousa CE, Pérez-Ruiz E, Del Monte-Millán M, Ruiz-Borrego M, de la Haba J, Sánchez-Rovira P, Romero A, González-Neira A, Lluch A, Alba E. Role of germline variants in the metastasis of breast carcinomas. Oncotarget 2022; 13:843-862. [PMID: 35782051 PMCID: PMC9245581 DOI: 10.18632/oncotarget.28250] [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: 03/04/2022] [Accepted: 06/20/2022] [Indexed: 11/25/2022] Open
Abstract
Most cancer-related deaths in breast cancer patients are associated with metastasis, a multistep, intricate process that requires the cooperation of tumour cells, tumour microenvironment and metastasis target tissues. It is accepted that metastasis does not depend on the tumour characteristics but the host’s genetic makeup. However, there has been limited success in determining the germline genetic variants that influence metastasis development, mainly because of the limitations of traditional genome-wide association studies to detect the relevant genetic polymorphisms underlying complex phenotypes. In this work, we leveraged the extreme discordant phenotypes approach and the epistasis networks to analyse the genotypes of 97 breast cancer patients. We found that the host’s genetic makeup facilitates metastases by the dysregulation of gene expression that can promote the dispersion of metastatic seeds and help establish the metastatic niche—providing a congenial soil for the metastatic seeds.
Collapse
Affiliation(s)
- Ángela Santonja
- Instituto de Investigación Biomédica de Málaga (IBIMA), Hospitales Universitarios Regional y Virgen de la Victoria de Málaga, Spain.,Laboratorio de Biología Molecular del Cáncer, Centro de Investigaciones Médico-Sanitarias (CIMES), Universidad de Málaga, Málaga, Spain.,These authors contributed equally to this work
| | - Aurelio A Moya-García
- Laboratorio de Biología Molecular del Cáncer, Centro de Investigaciones Médico-Sanitarias (CIMES), Universidad de Málaga, Málaga, Spain.,Departmento de Biología Molecular y Bioquímica, Universidad de Málaga, Málaga, Spain.,These authors contributed equally to this work
| | - Nuria Ribelles
- Unidad de Gestión Clínica Intercentro de Oncología, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospitales Universitarios Regional y Virgen de la Victoria de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red de Oncología, CIBERONC-ISCIII, Madrid, Spain
| | - Begoña Jiménez-Rodríguez
- Unidad de Gestión Clínica Intercentro de Oncología, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospitales Universitarios Regional y Virgen de la Victoria de Málaga, Málaga, Spain
| | - Bella Pajares
- Unidad de Gestión Clínica Intercentro de Oncología, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospitales Universitarios Regional y Virgen de la Victoria de Málaga, Málaga, Spain
| | - Cristina E Fernández-De Sousa
- Instituto de Investigación Biomédica de Málaga (IBIMA), Hospitales Universitarios Regional y Virgen de la Victoria de Málaga, Spain.,Laboratorio de Biología Molecular del Cáncer, Centro de Investigaciones Médico-Sanitarias (CIMES), Universidad de Málaga, Málaga, Spain
| | | | - María Del Monte-Millán
- Centro de Investigación Biomédica en Red de Oncología, CIBERONC-ISCIII, Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain
| | | | - Juan de la Haba
- Centro de Investigación Biomédica en Red de Oncología, CIBERONC-ISCIII, Madrid, Spain.,Biomedical Research Institute, Complejo Hospitalario Reina Sofía, Córdoba, Spain
| | | | - Atocha Romero
- Molecular Oncology Laboratory, Hospital Clínico San Carlos, IdISSC, Madrid, Spain
| | - Anna González-Neira
- Human Genotyping-CEGEN Unit, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Ana Lluch
- Centro de Investigación Biomédica en Red de Oncología, CIBERONC-ISCIII, Madrid, Spain.,Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain.,INCLIVA Biomedical Research Institute, Universidad de Valencia, Valencia, Spain
| | - Emilio Alba
- Laboratorio de Biología Molecular del Cáncer, Centro de Investigaciones Médico-Sanitarias (CIMES), Universidad de Málaga, Málaga, Spain.,Unidad de Gestión Clínica Intercentro de Oncología, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospitales Universitarios Regional y Virgen de la Victoria de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red de Oncología, CIBERONC-ISCIII, Madrid, Spain
| |
Collapse
|
11
|
Zhang Y, Hu X, Li H, Yao J, Yang P, Lan Y, Xia H. Circadian Period 2 (Per2) downregulate inhibitor of differentiation 3 (Id3) expression via PTEN/AKT/Smad5 axis to inhibits glioma cell proliferation. Bioengineered 2022; 13:12350-12364. [PMID: 35599595 PMCID: PMC9275974 DOI: 10.1080/21655979.2022.2074107] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
In this study, we employed multiple laboratory techniques to acknowledge the biological activities and processes of Per2 and Id3 in glioma. We analyzed TCGA and CGGA databases for seeking association among Per2, Id3, and clinical features in glioma. Immunohistochemistry and Western blot were used to detect protein expression levels. CCK-8 assay, colony formation assay, Transwell assay, the wound healing assay, flow cytometric, and Xenograft nude mice were used to acknowledge the impact of Per2 and Id3 on biological behavior of glioma. The results showed that the Per2 mRNA expression was negatively correlated with the WHO grade, while the Id3 mRNA expression was positively correlated with the WHO grade in patients with glioma in TCGA and CGGA databases. Per2 and Id3 maintained separate prognostic abilities and had a negative connection in human glioma. In the clinical sample study, Per2 and Id3 were validated at the protein level with the same results compared to the mRNA expression level in TCGA and CGGA. By using a wide range of functional examples, overexpression of Per2 restrains malignant biological behaviors in glioma cells by many ways, while Id3 promotes malignant biological behaviors in glioma cells. Furthermore, overexpression of Per2 can inhibit Id3 expression via regulating PTEN/AKT/Smad5 signaling pathway and thereby abolish malignant biological behaviors that are caused by Id3 overexpression. These results suggested that Per2 inhibits glioma cell proliferation through regulating PTEN/AKT/Smad5/Id3 signaling pathway, which may be a viable therapeutic target for glioma.
Collapse
Affiliation(s)
- Yifan Zhang
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
- Ningxia Human Stem Cell Institute, General Hospital of Ningxia Medical University, Yinchuan, China
- Department of Neurosurgery, Ningxia Human Stem Cell Institute, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Xvlei Hu
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
- Department of Neurosurgery, Ningxia Human Stem Cell Institute, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Hailiang Li
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Jian Yao
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
- Ningxia Human Stem Cell Institute, General Hospital of Ningxia Medical University, Yinchuan, China
- Department of Neurosurgery, Ningxia Human Stem Cell Institute, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Ping Yang
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
- Ningxia Human Stem Cell Institute, General Hospital of Ningxia Medical University, Yinchuan, China
- Department of Neurosurgery, Ningxia Human Stem Cell Institute, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yuanxiang Lan
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
- Ningxia Human Stem Cell Institute, General Hospital of Ningxia Medical University, Yinchuan, China
- Department of Neurosurgery, Ningxia Human Stem Cell Institute, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Hechun Xia
- Ningxia Human Stem Cell Institute, General Hospital of Ningxia Medical University, Yinchuan, China
- Department of Neurosurgery, Ningxia Human Stem Cell Institute, General Hospital of Ningxia Medical University, Yinchuan, China
| |
Collapse
|
12
|
NRF3 Decreases during Melanoma Carcinogenesis and Is an Independent Prognostic Marker in Melanoma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2240223. [PMID: 35378827 PMCID: PMC8976671 DOI: 10.1155/2022/2240223] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 02/28/2022] [Accepted: 03/04/2022] [Indexed: 11/17/2022]
Abstract
The prognostic significance of the major redox regulator, nuclear factor erythroid-2-related factor 2 (NRF2), is recognized in many cancers, but the role of NRF3 is not studied. Analysis from the Gene Expression Omnibus datasets showed that NRF3 mRNA levels increased from benign to dysplastic naevi (p = 0.04). We characterized the immunohistochemical expression of NRF3 in 81 naevi, 67 primary skin melanomas, and 51 lymph node metastases. The immunohistochemical expression of cytoplasmic NRF3 decreased from benign to dysplastic naevi (p < 0.001) and further to primary melanomas (p < 0.001). High cytoplasmic NRF3 protein expression in pigment cells of the primary melanomas associated with worse melanoma-specific survival in multivariate analysis, specifically in the subgroup of patients with the lymph node metastases at the time of diagnosis (hazard ratio 3.179; 95% confidence interval 1.065-9.493; p = 0.038). Intriguingly, we did not observe associations between NRF3 and the traditional prognostic factors such as Breslow thickness, ulceration, or stage. Together, this data represents the primary description about the role of NRF3 in pigment tumours that is worthy of further explorations.
Collapse
|
13
|
Bai S, Chen L, Yan Y, Wang X, Jiang A, Li R, Kang H, Feng Z, Li G, Ma W, Zhang J, Ren J. Identification of Hypoxia-Immune-Related Gene Signatures and Construction of a Prognostic Model in Kidney Renal Clear Cell Carcinoma. Front Cell Dev Biol 2022; 9:796156. [PMID: 35211477 PMCID: PMC8860910 DOI: 10.3389/fcell.2021.796156] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 12/23/2021] [Indexed: 12/24/2022] Open
Abstract
Introduction: Kidney renal clear cell carcinoma (KIRC), a kind of malignant disease, is a severe threat to public health. Tracking the information of tumor progression and conducting a related dynamic prognosis model are necessary for KIRC. It is crucial to identify hypoxia-immune-related genes and construct a prognostic model due to immune interaction and the influence of hypoxia in the prognosis of patients with KIRC. Methods: The hypoxia and immune status of KIRC patients were identified by utilizing t-SNE and ImmuCellAI for gene expression data. COX and Lasso regression were used to identify some hypoxia-immune-related signature genes and further construct a prognostic risk model based on these genes. Internal and external validations were also conducted to construct a prognostic model. Finally, some potentially effective drugs were screened by the CMap dataset. Results: We found that high-hypoxia and low-immune status tend to induce poor overall survival (OS). Six genes, including PLAUR, UCN, PABPC1L, SLC16A12, NFE2L3, and KCNAB1, were identified and involved in our hypoxia-immune-related prognostic risk model. Internal verification showed that the area under the curve (AUC) for the constructed models for 1-, 3-, 4-, and 5-year OS were 0.768, 0.754, 0.775, and 0.792, respectively. For the external verification, the AUC for 1-, 3-, 4-, and 5-year OS were 0.768, 0.739, 0.763, and 0.643 respectively. Furthermore, the decision curve analysis findings demonstrated excellent clinical effectiveness. Finally, we found that four drugs (including vorinostat, fludroxycortide, oxolinic acid, and flutamide) might be effective and efficient in alleviating or reversing the status of severe hypoxia and poor infiltration of immune cells. Conclusion: Our constructed prognostic model, based on hypoxia-immune-related genes, has excellent effectiveness and clinical application value. Moreover, some small-molecule drugs are screened to alleviate severe hypoxia and poor infiltration of immune cells.
Collapse
Affiliation(s)
- Shuheng Bai
- Department of Radiotherapy, Oncology Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ling Chen
- Department of Chemotherapy, Oncology Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yanli Yan
- Department of Radiotherapy, Oncology Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xuan Wang
- Department of Radiotherapy, Oncology Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Aimin Jiang
- Department of Chemotherapy, Oncology Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Rong Li
- Department of Radiotherapy, Oncology Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Haojing Kang
- Department of Radiotherapy, Oncology Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhaode Feng
- Department of Radiotherapy, Oncology Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Guangzu Li
- Department of Radiotherapy, Oncology Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wen Ma
- Medical School, Xi'an Jiaotong University Xi'an, Xi'an, China
| | - Jiangzhou Zhang
- Medical School, Xi'an Jiaotong University Xi'an, Xi'an, China
| | - Juan Ren
- Department of Radiotherapy, Oncology Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
14
|
Saliba J, Coutaud B, Makhani K, Epstein Roth N, Jackson J, Park JY, Gagnon N, Costa P, Jeyakumar T, Bury M, Beauchemin N, Mann KK, Blank V. Loss of NFE2L3 protects against inflammation-induced colorectal cancer through modulation of the tumor microenvironment. Oncogene 2022; 41:1563-1575. [PMID: 35091681 PMCID: PMC8913363 DOI: 10.1038/s41388-022-02192-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 12/16/2021] [Accepted: 01/13/2022] [Indexed: 02/07/2023]
Abstract
We investigated the role of the NFE2L3 transcription factor in inflammation-induced colorectal cancer. Our studies revealed that Nfe2l3−/− mice exhibit significantly less inflammation in the colon, reduced tumor size and numbers, and skewed localization of tumors with a more pronounced decrease of tumors in the distal colon. CIBERSORT analysis of RNA-seq data from normal and tumor tissue predicted a reduction in mast cells in Nfe2l3−/− animals, which was confirmed by toluidine blue staining. Concomitantly, the transcript levels of Il33 and Rab27a, both important regulators of mast cells, were reduced and increased, respectively, in the colorectal tumors of Nfe2l3−/− mice. Furthermore, we validated NFE2L3 binding to the regulatory sequences of the IL33 and RAB27A loci in human colorectal carcinoma cells. Using digital spatial profiling, we found that Nfe2l3−/− mice presented elevated FOXP3 and immune checkpoint markers CTLA4, TIM3, and LAG3, suggesting an increase in Treg counts. Staining for CD3 and FOXP3 confirmed a significant increase in immunosuppressive Tregs in the colon of Nfe2l3−/− animals. Also, Human Microbiome Project (HMP2) data showed that NFE2L3 transcript levels are higher in the rectum of ulcerative colitis patients. The observed changes in the tumor microenvironment provide new insights into the molecular differences regarding colon cancer sidedness. This may be exploited for the treatment of early-onset colorectal cancer as this emerging subtype primarily displays distal/left-sided tumors.
Collapse
Affiliation(s)
- James Saliba
- Lady Davis Institute for Medical Research, Montreal, Canada.,Department of Medicine, McGill University, Montreal, Quebec, Canada
| | | | - Kiran Makhani
- Lady Davis Institute for Medical Research, Montreal, Canada.,Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Noam Epstein Roth
- Lady Davis Institute for Medical Research, Montreal, Canada.,Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Jennie Jackson
- Lady Davis Institute for Medical Research, Montreal, Canada.,Life Sciences Institute and Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Joo Yeoun Park
- Lady Davis Institute for Medical Research, Montreal, Canada
| | | | - Paolo Costa
- Lady Davis Institute for Medical Research, Montreal, Canada
| | - Thiviya Jeyakumar
- Goodman Cancer Institute and Departments of Oncology, Biochemistry and Medicine, McGill University, Montreal, Quebec, Canada
| | - Marina Bury
- Lady Davis Institute for Medical Research, Montreal, Canada.,De Duve Institute, UCLouvain, Brussels, Belgium
| | - Nicole Beauchemin
- Goodman Cancer Institute and Departments of Oncology, Biochemistry and Medicine, McGill University, Montreal, Quebec, Canada
| | - Koren K Mann
- Lady Davis Institute for Medical Research, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Volker Blank
- Lady Davis Institute for Medical Research, Montreal, Canada. .,Department of Medicine, McGill University, Montreal, Quebec, Canada. .,Department of Physiology, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
15
|
Qian J, Huang C, Zhu Z, He Y, Wang Y, Feng N, He S, Li X, Zhou L, Zhang C, Gong Y. NFE2L3 promotes tumor progression and predicts a poor prognosis of bladder cancer. Carcinogenesis 2022; 43:457-468. [PMID: 35022660 DOI: 10.1093/carcin/bgac006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/03/2022] [Accepted: 01/11/2022] [Indexed: 11/14/2022] Open
Abstract
The high incidence and vulnerability to recurrence of bladder urothelial carcinoma (BLCA) is a challenge in the clinical. Recent studies have revealed that NFE2L3 plays a vital role in the carcinogenesis and progression of different human tumors. However, the role of NFE2L3 in bladder cancer has not been elucidated. In this study, NFE2L3 expression was significantly increased in bladder cancer samples. Its high expression was associated with advanced clinicopathological characteristics and was an independent prognostic factor for overall survival (OS) and metastasis-free survival (MFS) in 106 patients with BLCA. In vitro and in vivo experiments demonstrated that NFE2L3 knockdown inhibited bladder cancer cells proliferation by inducing the cell cycle arrest and cell apoptosis. Meanwhile, NFE2L3 overexpression promotes BLCA cell migration and invasion in vitro cell lines and in vivo xenografts. Moreover, we identified many genes and pathway alterations associated with tumor progression and metastasis by performing RNA-Seq analysis and functional enrichment of NFE2L3 overexpressing BLCA cells. Mechanistic investigation reveals that overexpression of NFE2L3 promoted epithelial-mesenchymal transition (EMT) in bladder cancer cells with decreased expression of gap junction-associated protein ZO-1 and epithelial marker E-cadherin with the elevation of transcription factors Snail1 and Snail2. Finally, we performed a comprehensive proteomics analysis to explore more potential molecular mechanisms. Our findings revealed that NFE2L3 might serve as a valuable clinical prognostic biomarker and therapeutic target in BLCA.
Collapse
Affiliation(s)
- Jinqin Qian
- Department of Urology, Peking University First Hospital, Beijing,100034, China.,Institute of Urology, Peking University, Beijing, 100034, China.,National Urological Cancer Center of China, Beijing, 100034, China
| | - Cong Huang
- Department of Urology, Peking University First Hospital, Beijing,100034, China.,Institute of Urology, Peking University, Beijing, 100034, China.,National Urological Cancer Center of China, Beijing, 100034, China
| | - Zhenpeng Zhu
- Department of Urology, Peking University First Hospital, Beijing,100034, China.,Institute of Urology, Peking University, Beijing, 100034, China.,National Urological Cancer Center of China, Beijing, 100034, China
| | - Yuhui He
- Department of Urology, Peking University First Hospital, Beijing,100034, China.,Institute of Urology, Peking University, Beijing, 100034, China.,National Urological Cancer Center of China, Beijing, 100034, China
| | - Yang Wang
- Department of Urology, Wuxi People's Hospital Affiliated Nanjing Medical University, Wuxi, Jiangsu, 214000, China
| | - Ninghan Feng
- Department of Urology, Wuxi People's Hospital Affiliated Nanjing Medical University, Wuxi, Jiangsu, 214000, China
| | - Shiming He
- Department of Urology, Peking University First Hospital, Beijing,100034, China.,Institute of Urology, Peking University, Beijing, 100034, China.,National Urological Cancer Center of China, Beijing, 100034, China
| | - Xuesong Li
- Department of Urology, Peking University First Hospital, Beijing,100034, China.,Institute of Urology, Peking University, Beijing, 100034, China.,National Urological Cancer Center of China, Beijing, 100034, China
| | - Liqun Zhou
- Department of Urology, Peking University First Hospital, Beijing,100034, China.,Institute of Urology, Peking University, Beijing, 100034, China.,National Urological Cancer Center of China, Beijing, 100034, China
| | - Cuijian Zhang
- Department of Urology, Peking University First Hospital, Beijing,100034, China.,Institute of Urology, Peking University, Beijing, 100034, China.,National Urological Cancer Center of China, Beijing, 100034, China
| | - Yanqing Gong
- Department of Urology, Peking University First Hospital, Beijing,100034, China.,Institute of Urology, Peking University, Beijing, 100034, China.,National Urological Cancer Center of China, Beijing, 100034, China
| |
Collapse
|
16
|
Alam MM, Chakma K, Mahmud S, Hossain MN, Karim MR, Amin MA. Multiomics analysis of altered NRF3 expression reveals poor prognosis in cancer. INFORMATICS IN MEDICINE UNLOCKED 2022. [DOI: 10.1016/j.imu.2022.100892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
17
|
Wang X, Li Y, Fang Z, Li Y. Elevated expression of NFE2L3 promotes the development of gastric cancer through epithelial-mesenchymal transformation. Bioengineered 2021; 12:12204-12214. [PMID: 34783304 PMCID: PMC8810066 DOI: 10.1080/21655979.2021.2005915] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Gastric cancer (GC) is a malignant tumor with high mortality, but research on its molecular mechanisms remain limited. This study is the first to explore the biological role of nuclear factor NFE2L3 (nuclear factor, erythroid 2 like 3) in GC. We used Western blot and RT–qPCR to detect gene expression at the protein or mRNA level. Short hairpin RNA (shRNA) transfection was used to inhibit NFE2L3 expression. CCK-8 and colony formation assays were used to detect cell proliferation. Cell migration, invasion, cell cycle and apoptosis were detected by Transwell assays and flow cytometry. The results showed that NFE2L3 was highly expressed in gastric cancer tissues and promoted gastric cancer cell proliferation and metastasis. Inhibiting NFE2L3 expression blocks the cell cycle and increases the proportion of apoptotic cells, whereas NFE2L3 expression promotes the epithelial-mesenchymal transformation (EMT) process. In summary, NFE2L3 is highly expressed in gastric cancer and promotes gastric cancer cell proliferation and metastasis and the EMT process.
Collapse
Affiliation(s)
- Xiaodong Wang
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, People's Republic of China
| | - Yaxian Li
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, People's Republic of China
| | - Ziqing Fang
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, People's Republic of China
| | - Yongxiang Li
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, People's Republic of China
| |
Collapse
|
18
|
Establishment of a prognostic model of ten transcription factors in gastric cancer. Genomics 2021; 113:4075-4087. [PMID: 34688795 DOI: 10.1016/j.ygeno.2021.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 09/23/2021] [Accepted: 10/12/2021] [Indexed: 02/07/2023]
Abstract
Transcription factors (TFs) play an important role in tumors. We integrated and analyzed 13 GPL570 platform gastric cancer (GC) microarrays, identified 10 independent prognostic TFs, and constructed a GC prognostic model. Using GSE26942 as the verification set, the Kaplan-Meier curve showed that the signature distinguish the survival rate of GC patients (P < 0.01), and the AUC values are 0.746 and 0.630, respectively. Compared with the clinicopathological characteristics, the signature is an independent prognostic factor (P < 0.05). A nomogram was established based on the model, and the five-year calibration curve verified that the prediction of the nomogram was almost consistent with the actual survival rate, C-index of 0.747 indicated a moderate prognostic ability. The analysis of target genes of 10 TFs showed that they are closely related to the progression of GC. External database and rt-PCR showed that the RNA and protein expression of TFs are consistent with our analysis.
Collapse
|
19
|
Apolipoprotein M inhibits proliferation and migration of larynx carcinoma cells. Sci Rep 2020; 10:19424. [PMID: 33173129 PMCID: PMC7655836 DOI: 10.1038/s41598-020-76480-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 10/26/2020] [Indexed: 11/14/2022] Open
Abstract
Prior studies have shown that apolipoprotein M (APOM) is involved in the development of some cancers. Here we investigated the effects of APOM on larynx cancer (LC). 20 patients with vocal cord polyps and 18 patients with LC were included in this study. The protein and mRNA levels of the samples were analysed using the Wes-ProteinSimple system (or traditional Western blot) and PCR technology, respectively. APOM protein level in cancer tissues was lower than that in paracarcinomatous (P = 0.0003) and polyp tissues (P < 0.0001). APOM overexpression significantly inhibited TU686 cell proliferation (P < 0.0001) and migration (P < 0.01), and increased expression of vitamin D receptor (VDR, P < 0.0001) as well as nuclear factor erythroid 2-like 3 (NFE2L3, P = 0.0215). In addition, matrix metalloproteinase-10 (MMP-10) mRNA level was significantly reduced in the APOM overexpression group (P = 0.0077). However, Western blot analysis showed that APOM overexpression did not change VDR, NFE2L3 and MMP-10 protein levels (P > 0.05). In summary, APOM inhibits the proliferation and migration of LC cells, but may not be related to VDR, NFE2L3 and MMP-10, which needs further study.
Collapse
|
20
|
NFE2L3 Controls Colon Cancer Cell Growth through Regulation of DUX4, a CDK1 Inhibitor. Cell Rep 2020; 29:1469-1481.e9. [PMID: 31693889 DOI: 10.1016/j.celrep.2019.09.087] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 06/26/2019] [Accepted: 09/27/2019] [Indexed: 12/13/2022] Open
Abstract
Constitutive nuclear factor κB (NF-κB) activation is a hallmark of colon tumor growth. Cyclin-dependent kinases (CDKs) are critical cell-cycle regulators, and inhibition of CDK activity has been used successfully as anticancer therapy. Here, we show that the NFE2L3 transcription factor functions as a key regulator in a pathway that links NF-κB signaling to the control of CDK1 activity, thereby driving colon cancer cell proliferation. We found that NFE2L3 expression is regulated by the RELA subunit of NF-κB and that NFE2L3 levels are elevated in patients with colon adenocarcinoma when compared with normal adjacent tissue. Silencing of NFE2L3 significantly decreases colon cancer cell proliferation in vitro and tumor growth in vivo. NFE2L3 knockdown results in increased levels of double homeobox factor 4 (DUX4), which functions as a direct inhibitor of CDK1. The discovered oncogenic pathway governing cell-cycle progression may open up unique avenues for precision cancer therapy.
Collapse
|
21
|
Kobayashi A. Roles of NRF3 in the Hallmarks of Cancer: Proteasomal Inactivation of Tumor Suppressors. Cancers (Basel) 2020; 12:cancers12092681. [PMID: 32962187 PMCID: PMC7563620 DOI: 10.3390/cancers12092681] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/15/2020] [Accepted: 09/17/2020] [Indexed: 01/18/2023] Open
Abstract
Simple Summary This review summarizes recent advances in our understanding of the physiological roles of the NFE2-related factor 2 (NRF2)-related transcription factor NRF3 in cancer. NRF3 confers cells with six so-called “hallmarks of cancer” through upregulating gene expression of specific target genes, leading to tumorigenesis and cancer malignancy. These driver gene-like functions of NRF3 in cancer are distinct from those of NRF2. Abstract The physiological roles of the NRF2-related transcription factor NRF3 (NFE2L3) have remained unknown for decades. The remarkable development of human cancer genome databases has led to strong suggestions that NRF3 has functional significance in cancer; specifically, high NRF3 mRNA levels are induced in many cancer types, such as colorectal cancer and pancreatic adenocarcinoma, and are associated with poor prognosis. On the basis of this information, the involvement of NRF3 in tumorigenesis and cancer malignancy has been recently proposed. NRF3 confers cancer cells with selective growth advantages by enhancing 20S proteasome assembly through induction of the chaperone gene proteasome maturation protein (POMP) and consequently promoting degradation of the tumor suppressors p53 and retinoblastoma (Rb) in a ubiquitin-independent manner. This new finding offers insight into the proteasomal but not the genetic inactivation mechanism of tumor suppressors. Moreover, NRF3 promotes cancer malignancy-related processes, including metastasis and angiogenesis. Finally, the molecular mechanisms underlying NRF3 activation have been elucidated, and this knowledge is expected to provide many insights that are useful for the development of anticancer drugs that attenuate NRF3 transcriptional activity. Collectively, the evidence indicates that NRF3 confers cells with six so-called “hallmarks of cancer”, implying that it exhibits cancer driver gene-like function. This review describes recent research advances regarding the newly discovered addiction of cancer cells to NRF3 compared to NRF2.
Collapse
Affiliation(s)
- Akira Kobayashi
- Laboratory for Genetic Code, Graduate School of Life and Medical Sciences, Doshisha University, Kyotanabe, Kyoto 610-0394, Japan; ; Tel.: +81-774-65-6273
- Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Kyoto 610-0394, Japan
| |
Collapse
|
22
|
Ren Y, Wang Y, Hao S, Yang Y, Xiong W, Qiu L, Tao J, Tang A. NFE2L3 promotes malignant behavior and EMT of human hepatocellular carcinoma (HepG2) cells via Wnt/β‑catenin pathway. J Cancer 2020; 11:6939-6949. [PMID: 33123284 PMCID: PMC7591994 DOI: 10.7150/jca.48100] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 09/14/2020] [Indexed: 12/12/2022] Open
Abstract
Objective: NFE2L3 is a member of the cap 'n' collar basic-region leucine zipper family. NFE2L3 has turned out to be associated with oxidative stress, but the relevance of NFE2L3 in hepatocellular carcinoma (HCC) has remained elusive. This study aimed to investigate the role of NFE2L3 in HCC and explore underlying mechanisms. Methods: Quantitative real-time PCR, western blot and immunohistochemistry were used to detect the mRNA and protein expression of NFE2L3, the expression of epithelial-mesenchymal transition (EMT) markers and Wnt/β-catenin signaling pathway-related proteins. In loss-function experiments, HepG2 cells were transfected with lentiviral vector containing NFE2L3 short hairpin RNA or scramble control. Cell proliferation and migration were measured by Cell Counting Kit-8, Colony formation, EdU incorporation and Transwell assays respectively. Flow cytometry was used to analyze cell cycle and apoptosis. HepG2 cells were subcutaneously injected into nude mice and tumor size was measured once every other day. Results: The results revealed that high expression of NFE2L3 was positively associated with malignant behavior and EMT in HCC. Knockdown of NFE2L3 inhibited cell proliferation and migration, led to cell cycle G0/G1 arrest and induction of cell apoptosis, increased expression of E-cadherin and decreased expression of N‑cadherin, Vimentin, MMP2, CDK2 and PCNA. In addition, tumor growth was inhibited by silencing of NFE2L3 in vivo. Expression of β-catenin and Wnt target genes cyclin D1 and TCF4 was reduced in HepG2-shNFE2L3 cells. Conclusions: NFE2L3 promotes cell proliferation, metastasis, and induces EMT of hepatocellular carcinoma (HepG2) cells via activation of Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Yonggang Ren
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Institute of Translational Medicine, First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
- Department of Biochemistry, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
- ✉ Corresponding authors: Aifa Tang, Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Institute of Translational Medicine, First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital. No. 3002 Sungang Xi Road, Futian District, Shenzhen, Guangdong, 518035, P.R. China; Tel: (86) 755-8300-3435; Fax: (86) 755-8300-3435; E-mail: ; Yonggang Ren, Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Institute of Translational Medicine, First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital. No. 3002 Sungang Xi Road, Futian District, Shenzhen, Guangdong, 518035, P.R. China; Department of Biochemistry, North Sichuan Medical College. No. 55 Dongshun Road, Gaoping District, Nanchong, Sichuan, 637000, P.R. China; Tel: (86) 755-8300-3435; Fax: (86) 755-8300-3435; E-mail:
| | - Yujiao Wang
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Shuai Hao
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Yuhan Yang
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Wendong Xiong
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Lu Qiu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, P.R. China
| | - Jia Tao
- Department of Pathology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Aifa Tang
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Institute of Translational Medicine, First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
- ✉ Corresponding authors: Aifa Tang, Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Institute of Translational Medicine, First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital. No. 3002 Sungang Xi Road, Futian District, Shenzhen, Guangdong, 518035, P.R. China; Tel: (86) 755-8300-3435; Fax: (86) 755-8300-3435; E-mail: ; Yonggang Ren, Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Institute of Translational Medicine, First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital. No. 3002 Sungang Xi Road, Futian District, Shenzhen, Guangdong, 518035, P.R. China; Department of Biochemistry, North Sichuan Medical College. No. 55 Dongshun Road, Gaoping District, Nanchong, Sichuan, 637000, P.R. China; Tel: (86) 755-8300-3435; Fax: (86) 755-8300-3435; E-mail:
| |
Collapse
|