1
|
Shokhirev MN, Kramer DJ, Corley J, Cox SR, Cuellar TL, Johnson AA. CheekAge, a next-generation epigenetic buccal clock, is predictive of mortality in human blood. FRONTIERS IN AGING 2024; 5:1460360. [PMID: 39411517 PMCID: PMC11473594 DOI: 10.3389/fragi.2024.1460360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 08/26/2024] [Indexed: 10/19/2024]
Abstract
While earlier first-generation epigenetic aging clocks were trained to estimate chronological age as accurately as possible, more recent next-generation clocks incorporate DNA methylation information more pertinent to health, lifestyle, and/or outcomes. Recently, we produced a non-invasive next-generation epigenetic clock trained using Infinium MethylationEPIC data from more than 8,000 diverse adult buccal samples. While this clock correlated with various health, lifestyle, and disease factors, we did not assess its ability to capture mortality. To address this gap, we applied CheekAge to the longitudinal Lothian Birth Cohorts of 1921 and 1936. Despite missing nearly half of its CpG inputs, CheekAge was significantly associated with mortality in this longitudinal blood dataset. Specifically, a change in one standard deviation corresponded to a hazard ratio (HR) of 1.21 (FDR q = 1.66e-6). CheekAge performed better than all first-generation clocks tested and displayed a comparable HR to the next-generation, blood-trained DNAm PhenoAge clock (HR = 1.23, q = 2.45e-9). To better understand the relative importance of each CheekAge input in blood, we iteratively removed each clock CpG and re-calculated the overall mortality association. The most significant effect came from omitting the CpG cg14386193, which is annotated to the gene ALPK2. Excluding this DNA methylation site increased the FDR value by nearly threefold (to 4.92e-06). We additionally performed enrichment analyses of the top annotated CpGs that impact mortality to better understand their associated biology. Taken together, we provide important validation for CheekAge and highlight novel CpGs that underlie a newly identified mortality association.
Collapse
Affiliation(s)
| | | | - Janie Corley
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, United Kingdom
| | - Simon R. Cox
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, United Kingdom
| | | | | |
Collapse
|
2
|
Zhang MQ, Li JR, Yang L, Peng ZG, Wu S, Zhang JP. ATG10S promotes IFNL1 expression and autophagic degradation of multiple viral proteins mediated by IFNL1. Autophagy 2024; 20:2238-2254. [PMID: 38842055 PMCID: PMC11423677 DOI: 10.1080/15548627.2024.2361580] [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: 10/07/2023] [Revised: 05/21/2024] [Accepted: 05/24/2024] [Indexed: 06/07/2024] Open
Abstract
ATG10S is a newly discovered subtype of the autophagy protein ATG10. It promotes complete macroautophagy/autophagy, degrades multiple viral proteins, and increases the expression of type III interferons. Here, we aimed to investigate the mechanism of ATG10S cooperation with IFNL1 to degrade viral proteins from different viruses. Using western blot, immunoprecipitation (IP), tandem sensor RFP-GFP-LC3B and in situ proximity ligation assays, we showed that exogenous recombinant ATG10S protein (rHsATG10S) could enter into cells through clathrin, and ATG10S combined with ATG7 with IFNL1 assistance to facilitate ATG12-ATG5 conjugation, thereby contributing to the autophagosome formation in multiple cell lines containing different virions or viral proteins. The results of DNA IP and luciferase assays also showed that ATG10S was able to directly bind to a core motif (CAAGGG) within a binding site of transcription factor ZNF460 on the IFNL1 promoter, by which IFNL1 transcription was activated. These results clarified that ATG10S promoted autophagosome formation with the assistance of IFNL1 to ensure autophagy flux and autophagic degradation of multiple viral proteins and that ATG10S could also act as a novel transcription factor to promote IFNL1 gene expression. Importantly, this study further explored the antiviral mechanism of ATG10S interaction with type III interferon and provided a theoretical basis for the development of ATG10S into a new broad-spectrum antiviral protein drug.Abbreviation: ATG: autophagy related; ATG10S: the shorter isoform of autophagy-related 10; CC50: half cytotoxicity concentration; CCV: clathrin-coated transport vesicle; CLTC: clathrin heavy chain; CM: core motif; co-IP: co-immunoprecipitation; CPZ: chlorpromazine; ER: endoplasmic reticulum; HCV: hepatitis C virus; HBV: hepatitis B virus; HsCoV-OC43: Human coronavirus OC43; IFN: interferon; PLA: proximity ligation assay; rHsATG10S: recombinant human ATG10S protein; RLU: relative light unit; SQSTM1: sequestosome 1; ZNF: zinc finger protein.
Collapse
Affiliation(s)
- Miao-Qing Zhang
- Key Laboratory of Biotechnology of Antibiotics, the National Health Commission (NHC), Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian-Rui Li
- Key Laboratory of Biotechnology of Antibiotics, the National Health Commission (NHC), Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lu Yang
- Key Laboratory of Biotechnology of Antibiotics, the National Health Commission (NHC), Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zong-Gen Peng
- Key Laboratory of Biotechnology of Antibiotics, the National Health Commission (NHC), Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuo Wu
- Key Laboratory of Biotechnology of Antibiotics, the National Health Commission (NHC), Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing-Pu Zhang
- Key Laboratory of Biotechnology of Antibiotics, the National Health Commission (NHC), Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
3
|
Akamatsu K, Golzari S, Amariuta T. Powerful mapping of cis-genetic effects on gene expression across diverse populations reveals novel disease-critical genes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.25.24314410. [PMID: 39399015 PMCID: PMC11469471 DOI: 10.1101/2024.09.25.24314410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
While disease-associated variants identified by genome-wide association studies (GWAS) most likely regulate gene expression levels, linking variants to target genes is critical to determining the functional mechanisms of these variants. Genetic effects on gene expression have been extensively characterized by expression quantitative trait loci (eQTL) studies, yet data from non-European populations is limited. This restricts our understanding of disease to genes whose regulatory variants are common in European populations. While previous work has leveraged data from multiple populations to improve GWAS power and polygenic risk score (PRS) accuracy, multi-ancestry data has not yet been used to better estimate cis-genetic effects on gene expression. Here, we present a new method, Multi-Ancestry Gene Expression Prediction Regularized Optimization (MAGEPRO), which constructs robust genetic models of gene expression in understudied populations or cell types by fitting a regularized linear combination of eQTL summary data across diverse cohorts. In simulations, our tool generates more accurate models of gene expression than widely-used LASSO and the state-of-the-art multi-ancestry PRS method, PRS-CSx, adapted to gene expression prediction. We attribute this improvement to MAGEPRO's ability to more accurately estimate causal eQTL effect sizes (p < 3.98 × 10-4, two-sided paired t-test). With real data, we applied MAGEPRO to 8 eQTL cohorts representing 3 ancestries (average n = 355) and consistently outperformed each of 6 competing methods in gene expression prediction tasks. Integration with GWAS summary statistics across 66 complex traits (representing 22 phenotypes and 3 ancestries) resulted in 2,331 new gene-trait associations, many of which replicate across multiple ancestries, including PHTF1 linked to white blood cell count, a gene which is overexpressed in leukemia patients. MAGEPRO also identified biologically plausible novel findings, such as PIGB, an essential component of GPI biosynthesis, associated with heart failure, which has been previously evidenced by clinical outcome data. Overall, MAGEPRO is a powerful tool to enhance inference of gene regulatory effects in underpowered datasets and has improved our understanding of population-specific and shared genetic effects on complex traits.
Collapse
Affiliation(s)
- Kai Akamatsu
- School of Biological Sciences, UC San Diego, La Jolla, CA, USA
- Department of Medicine, Division of Biomedical Informatics, UC San Diego, La Jolla, CA, USA
- Halıcıoğlu Data Science Institute, UC San Diego, La Jolla, CA, USA
| | - Stephen Golzari
- Department of Medicine, Division of Biomedical Informatics, UC San Diego, La Jolla, CA, USA
- Halıcıoğlu Data Science Institute, UC San Diego, La Jolla, CA, USA
- Shu Chien-Gene Lay Department of Bioengineering, UC San Diego, La Jolla, CA, USA
| | - Tiffany Amariuta
- Department of Medicine, Division of Biomedical Informatics, UC San Diego, La Jolla, CA, USA
- Halıcıoğlu Data Science Institute, UC San Diego, La Jolla, CA, USA
| |
Collapse
|
4
|
Chen X, Lin X, Xia X, Xiang X. YAP1-induced RBM24 promotes the tumorigenesis of triple-negative breast cancer through the β-catenin pathway. J Investig Med 2024; 72:403-413. [PMID: 38441112 DOI: 10.1177/10815589241239577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype and refractory to current treatments. RBM24 is an RNA-binding protein and shows the ability to regulate tumor progression in multiple cancer types. However, its role in TNBC is still unclear. In this study, we analyzed publicly available profiling data from TNBC tissues and cells. Loss- and gain-of-function experiments were performed to determine the function of RBM24 in TNBC cells. The mechanism for RBM24 action in TNBC was investigated. RBM24 was deregulated in TNBC tissues and TNBC cells with depletion of SIPA1, YAP1, or ARID1A, three key regulators of TNBC. Compared to MCF10A breast epithelial cells, TNBC cells had higher levels of RBM24. Knockdown of RBM24 inhibited TNBC cell proliferation, colony formation, and tumorigenesis, while overexpression of RBM24 promoted aggressive phenotype in TNBC cells. YAP1 overexpression induced the expression of RBM24 and the RBM24 promoter-driven luciferase reporter. YAP1 was enriched at the promoter region of RBM24. Overexpression of RBM24 increased β-catenin-dependent transcriptional activity. Most importantly, knockdown of CTNNB1 rescued RBM24 aggressive phenotype in TNBC cells. Collectively, the YAP1/RBM24/β-catenin axis plays a critical role in driving TNBC progression. RBM24 may represent a novel therapeutic target for TNBC treatment.
Collapse
Affiliation(s)
- Xiaohua Chen
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiao Lin
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaodong Xia
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiao Xiang
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
5
|
Nguyen TMH, Lai YS, Chen YC, Lin TC, Nguyen NT, Chiu WT. Hypoxia-induced YAP activation and focal adhesion turnover to promote cell migration in mesenchymal TNBC cells. Cancer Med 2023; 12:9723-9737. [PMID: 36757143 PMCID: PMC10166962 DOI: 10.1002/cam4.5680] [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: 12/05/2022] [Revised: 01/18/2023] [Accepted: 01/31/2023] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND Hypoxia is commonly characterized by malignant tumors that promote the aggressiveness and metastatic potential of cancer. Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, with approximately 46% capacity related to distant metastasis. Transcriptional factor yes-associated protein (YAP), a core component of the Hippo pathway, is associated with poor prognosis and outcome in cancer metastasis. Here, we explored the effect of hypoxia-mediated YAP activation and focal adhesions (FAs) turnover in mesenchymal TNBC cell migration. METHODS We characterized the effect of hypoxia on YAP in different breast cancer cell lines using a hypoxia chamber and CoCl2 . RESULTS Hypoxia-induced YAP nuclear translocation is significantly observed in normal breast epithelial cells, non-TNBC cells, mesenchymal TNBC cells, but not in basal-like TNBC cells. Functionally, we demonstrated that YAP activation was required for hypoxia to promote mesenchymal TNBC cell migration. Furthermore, hypoxia induced the localization of FAs at the leading edge of mesenchymal TNBC cells. In contrast, verteporfin (VP), a YAP inhibitor, significantly reduced the migration and the recruitment of nascent FAs at the cell periphery under hypoxia conditions, which only showed in mesenchymal TNBC cells. CONCLUSIONS Our data support the hypothesis that YAP is novel factor and positively responsible for hypoxia-promoting mesenchymal TNBC cell migration. Our findings provide further evidence and outcomes to help prevent the progression of TNBC.
Collapse
Affiliation(s)
- Thi My Hang Nguyen
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Shyun Lai
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Ying-Chi Chen
- Department of Chemistry, National Cheng Kung University, Taiwan, Taiwan
| | - Tzu-Chien Lin
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ngoc Thang Nguyen
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Wen-Tai Chiu
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, Tainan, Taiwan.,Department of Chemistry, National Cheng Kung University, Taiwan, Taiwan.,Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
6
|
Wang D, Li Z, Li X, Yan C, Yang H, Zhuang T, Wang X, Zang Y, Liu Z, Wang T, Jiang R, Su P, Zhu J, Ding Y. DUB1 suppresses Hippo signaling by modulating TAZ protein expression in gastric cancer. J Exp Clin Cancer Res 2022; 41:219. [PMID: 35820928 PMCID: PMC9275142 DOI: 10.1186/s13046-022-02410-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 06/05/2022] [Indexed: 01/02/2023] Open
Abstract
Abstract
Background
The Hippo pathway functions as a tumor suppressor pathway in human cancers, while dysfunction of the Hippo pathway is frequently observed in malignancies. Although YAP/TAZ activity is tightly controlled by the phosphorylation cascade of the MST-LATS-YAP/TAZ axis, it is still unclear why the YAP/TAZ proteins are activated in human cancers despite Hippo pathway activation. Recent studies have suggested that in addition to phosphorylation, several other posttranslational modifications, including ubiquitination, also play critical roles in modulating TAZ function.
Methods
We used several gastric cancer cell lines and performed western blot analysis, real-time PCR, immunoprecipitation assays, and in vitro ubiquitination assays and established a xenograft mouse model.
Results
Here, by screening a DUB (deubiquitinase) siRNA library, we discovered that DUB1 functions as a critical modulator that facilitates gastric cancer stemness and progression by deubiquitinating and activating the TAZ protein. We also found that DUB1 expression was elevated in gastric cancer and that elevated DUB1 expression correlated with TAZ activation and poor survival. DUB1 associates with the TAZ protein and deubiquitinates TAZ at several lysine residues, which subsequently stabilizes TAZ and facilitates its function.
Conclusions
Our study revealed a novel deubiquitinase in the Hippo/TAZ axis and identified one possible therapeutic target for Hippo-driven gastric cancer.
Collapse
|
7
|
Li Z, Su P, Ding Y, Gao H, Yang H, Li X, Yang X, Xia Y, Zhang C, Fu M, Wang D, Zhang Y, Zhuo S, Zhu J, Zhuang T. RBCK1 is an endogenous inhibitor for triple negative breast cancer via hippo/YAP axis. Cell Commun Signal 2022; 20:164. [PMID: 36280829 PMCID: PMC9590148 DOI: 10.1186/s12964-022-00963-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/17/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Triple negative breast cancer (TNBC) is one of the most lethal breast cancer subtypes. Due to a lack of effective therapeutic targets, chemotherapy is still the main medical treatment for TNBC patients. Thus, it is important and necessary to find new therapeutic targets for TNBC. Recent genomic studies implicated the Hippo / Yap signal is over activated in TNBC, manifesting it plays a key role in TNBC carcinogenesis and cancer progression. RBCK1 was firstly identified as an important component for linear ubiquitin assembly complex (LUBAC) and facilitates NFKB signaling in immune response. Further studies showed RBCK1 also facilitated luminal type breast cancer growth and endocrine resistance via trans-activation estrogen receptor alpha. METHODS RBCK1 and YAP protein expression levels were measured by western blotting, while the mRNA levels of YAP target genes were measured by RT-PCR. RNA sequencing data were analyzed by Ingenuity Pathway Analysis. Identification of Hippo signaling activity was accomplished with luciferase assays, RT-PCR and western blotting. Protein stability assays and ubiquitin assays were used to detect YAP protein degradation. Ubiquitin-based immunoprecipitation assays were used to detect the specific ubiquitination modification on the YAP protein. RESULTS In our current study, our data revealed an opposite function for RBCK1 in TNBC progression. RBCK1 over-expression inhibited TNBC cell progression in vitro and in vivo, while RBCK1 depletion promoted TNBC cell invasion. The whole genomic expression profiling showed that RBCK1 depletion activated Hippo/YAP axis. RBCK1 depletion increased YAP protein level and Hippo target gene expression in TNBC. The molecular biology studies confirmed that RBCK1 could bind to YAP protein and enhance the stability of YAP protein by promoting YAP K48-linked poly-ubiquitination at several YAP lysine sites (K76, K204 and K321). CONCLUSION Our study revealed the multi-faced RBCK1 function in different subtypes of breast cancer patients and a promising therapeutic target for TNBC treatment. Video abstract.
Collapse
Affiliation(s)
- Zhongbo Li
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003 Henan Province People’s Republic of China
| | - Peng Su
- Department of Pathology, Shandong University Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong, Shandong Province People’s Republic of China
| | - Yinlu Ding
- Department of General Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Shandong, Shandong Province People’s Republic of China
| | - Honglei Gao
- Department of General Surgery, Weifang People’s Hospital, Shandong, Shandong Province People’s Republic of China
| | - Huijie Yang
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003 Henan Province People’s Republic of China
| | - Xin Li
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003 Henan Province People’s Republic of China
| | - Xiao Yang
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003 Henan Province People’s Republic of China
| | - Yan Xia
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003 Henan Province People’s Republic of China
| | - Chenmiao Zhang
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003 Henan Province People’s Republic of China
| | - Mingxi Fu
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003 Henan Province People’s Republic of China
| | - Dehai Wang
- Department of General Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Shandong, Shandong Province People’s Republic of China
| | - Ye Zhang
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003 Henan Province People’s Republic of China
| | - Shu Zhuo
- Signet Therapeutics Inc., Shenzhen, 518017 People’s Republic of China
| | - Jian Zhu
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003 Henan Province People’s Republic of China
- Department of General Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Shandong, Shandong Province People’s Republic of China
| | - Ting Zhuang
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003 Henan Province People’s Republic of China
| |
Collapse
|
8
|
Pal AK, Sharma P, Zia A, Siwan D, Nandave D, Nandave M, Gautam RK. Metabolomics and EMT Markers of Breast Cancer: A Crosstalk and Future Perspective. PATHOPHYSIOLOGY 2022; 29:200-222. [PMID: 35736645 PMCID: PMC9230911 DOI: 10.3390/pathophysiology29020017] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/17/2022] [Accepted: 05/24/2022] [Indexed: 11/22/2022] Open
Abstract
Cancer cells undergo transient EMT and MET phenomena or vice versa, along with the parallel interplay of various markers, often correlated as the determining factor in decoding metabolic profiling of breast cancers. Moreover, various cancer signaling pathways and metabolic changes occurring in breast cancer cells modulate the expression of such markers to varying extents. The existing research completed so far considers the expression of such markers as determinants regulating the invasiveness and survival of breast cancer cells. Therefore, this manuscript is crosstalk among the expression levels of such markers and their correlation in regulating the aggressiveness and invasiveness of breast cancer. We also attempted to cover the possible EMT-based metabolic targets to retard migration and invasion of breast cancer.
Collapse
Affiliation(s)
- Ajay Kumar Pal
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India; (A.K.P.); (P.S.); (A.Z.); (D.S.)
| | - Prateek Sharma
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India; (A.K.P.); (P.S.); (A.Z.); (D.S.)
| | - Alishan Zia
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India; (A.K.P.); (P.S.); (A.Z.); (D.S.)
| | - Deepali Siwan
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India; (A.K.P.); (P.S.); (A.Z.); (D.S.)
| | - Dipali Nandave
- Department of Dravyaguna, Karmavir V. T. Randhir Ayurved College, Boradi 425428, India;
| | - Mukesh Nandave
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India; (A.K.P.); (P.S.); (A.Z.); (D.S.)
- Correspondence: (M.N.); (R.K.G.)
| | - Rupesh K. Gautam
- Department of Pharmacology, MM School of Pharmacy, Maharishi Markandeshwar University, Ambala 134007, India
- Correspondence: (M.N.); (R.K.G.)
| |
Collapse
|
9
|
Yang L, Wang S, Pan Z, Du X, Li Q. TGFBR2 is a novel substrate and indirect transcription target of deubiquitylase USP9X in granulosa cells. J Cell Physiol 2022; 237:2969-2979. [PMID: 35578792 DOI: 10.1002/jcp.30776] [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: 12/28/2021] [Revised: 03/18/2022] [Accepted: 04/08/2022] [Indexed: 11/11/2022]
Abstract
The ubiquitin-specific peptidase 9 X-linked (USP9X) is one of the highly conserved members belonging to the ubiquitin-specific proteases (USPs) family, which has been reported to control substrates-mediated biological functions through deubiquitinating and stabilizing substrates. Here, we have found that TGFBR2, the type II receptor of the transforming growth factor beta (TGF-β) signaling pathway, is a novel substrate and indirect transcription target of deubiquitylase USP9X in granulosa cells (GCs). Mechanically, USP9X positively influences the expression of TGFBR2 at different levels through two independent ways: (i) directly targets and deubiquitinates TGFBR2, which maintains the protein stability of TGFBR2 through avoiding degradation mediated by ubiquitin-proteasome system; (ii) indirectly maintains TGFBR2 messenger RNA (mRNA) expression via SMAD4/miR-143 axis. Specifically, SMAD4, another substrate of USP9X, acts as a transcription factor and suppresses miR-143 which inhibits the mRNA level of TGFBR2 by directly binding to its 3'-untranslated region. Functionally, the maintenance of TGFBR2 by USP9X activates the TGF-β signaling pathway, which further represses GC apoptosis. Our study highlights a functional micro-regulatory network composed of deubiquitinase (USP9X), small noncoding RNA (miR-143) and the TGF-β signaling pathway, which plays a crucial role in the regulation of GC apoptosis and female fertility.
Collapse
Affiliation(s)
- Liu Yang
- Laboratory of Statistical Genetics and Epigenetics, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Siqi Wang
- Laboratory of Statistical Genetics and Epigenetics, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Zengxiang Pan
- Laboratory of Statistical Genetics and Epigenetics, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Xing Du
- Laboratory of Statistical Genetics and Epigenetics, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Qifa Li
- Laboratory of Statistical Genetics and Epigenetics, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
10
|
Liu Y, Su P, Zhao W, Li X, Yang X, Fan J, Yang H, Yan C, Mao L, Ding Y, Zhu J, Niu Z, Zhuang T. ZNF213 negatively controls triple negative breast cancer progression via Hippo/YAP signaling. Cancer Sci 2021; 112:2714-2727. [PMID: 33939216 PMCID: PMC8253295 DOI: 10.1111/cas.14916] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 12/18/2022] Open
Abstract
Breast cancer is one of the most commonly diagnosed malignancies worldwide, while the triple negative breast cancer (TNBC) is the most aggressive and virulent subtype in breast cancers. Compared with luminal type breast cancers, which could be well controlled by endocrine treatment, TNBC is worse in prognosis and lack of effective targeted therapy. Thus, it would be interesting and meaningful to identify novel therapeutic targets for TNBC treatments. Recent genomic data showed the activation of Hippo/YAP signaling in TNBC, indicating its critical roles in TNBC carcinogenesis and cancer progression. Hippo/YAP signaling could subject to several kinds of protein modifications, including ubiquitination and phosphorylation. Quite a few studies have demonstrated these modifications, which controlled YAP protein stability and turnover, played critical role in Hippo signaling activation In our current study, we identified ZNF213 as a negative modifier for Hippo/YAP axis. ZNF213 depletion promoted TNBC cell migration and invasion, which could be rescued by further YAP silencing. ZNF213 knocking down facilitated YAP protein stability and Hippo target gene expression, including CTGF and CYR61. Further mechanism studies demonstrated that ZNF213 associated with YAP and facilitated YAP K48-linked poly-ubiquitination at several YAP lysine sites (K252, K254, K321 and K497). Besides, the clinical data showed that ZNF213 negatively correlated with YAP protein level and Hippo target gene expression in TNBC samples. ZNF213 expression correlated with good prognosis in TNBC patients. Our data provided novel insights in YAP proteolytic regulation and TNBC progression.
Collapse
Affiliation(s)
- Yun Liu
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision MedicineXinxiang Medical UniversityXinxiang, Henan ProvinceChina
- Henan Key Laboratory of immunology and targeted therapySchool of Laboratory MedicineHenan Collaborative Innovation Center of Molecular Diagnosis and Laboratory MedicineSchool of Laboratory MedicineXinxiang Medical UniversityXinxiang, Henan ProvinceChina
| | - Peng Su
- Department of PathologyQilu HospitalCheeloo College of MedicineShandong UniversityJinanChina
| | - Wuchen Zhao
- School of International EducationXinxiang Medical UniversityXinxiang, Henan ProvinceChina
| | - Xin Li
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision MedicineXinxiang Medical UniversityXinxiang, Henan ProvinceChina
- Henan Key Laboratory of immunology and targeted therapySchool of Laboratory MedicineHenan Collaborative Innovation Center of Molecular Diagnosis and Laboratory MedicineSchool of Laboratory MedicineXinxiang Medical UniversityXinxiang, Henan ProvinceChina
| | - Xiao Yang
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision MedicineXinxiang Medical UniversityXinxiang, Henan ProvinceChina
- Henan Key Laboratory of immunology and targeted therapySchool of Laboratory MedicineHenan Collaborative Innovation Center of Molecular Diagnosis and Laboratory MedicineSchool of Laboratory MedicineXinxiang Medical UniversityXinxiang, Henan ProvinceChina
| | - Jianing Fan
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision MedicineXinxiang Medical UniversityXinxiang, Henan ProvinceChina
- Henan Key Laboratory of immunology and targeted therapySchool of Laboratory MedicineHenan Collaborative Innovation Center of Molecular Diagnosis and Laboratory MedicineSchool of Laboratory MedicineXinxiang Medical UniversityXinxiang, Henan ProvinceChina
| | - Huijie Yang
- Department of PharmacologySchool of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Cheng Yan
- School of MedicineXinxiang UniversityXinxiangChina
| | - Lanzhi Mao
- Henan Key Laboratory of immunology and targeted therapySchool of Laboratory MedicineHenan Collaborative Innovation Center of Molecular Diagnosis and Laboratory MedicineSchool of Laboratory MedicineXinxiang Medical UniversityXinxiang, Henan ProvinceChina
| | - Yinlu Ding
- Department of General SurgeryThe Second HospitalCheeloo College of MedicineShandong UniversityShandong ProvinceChina
| | - Jian Zhu
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision MedicineXinxiang Medical UniversityXinxiang, Henan ProvinceChina
- Henan Key Laboratory of immunology and targeted therapySchool of Laboratory MedicineHenan Collaborative Innovation Center of Molecular Diagnosis and Laboratory MedicineSchool of Laboratory MedicineXinxiang Medical UniversityXinxiang, Henan ProvinceChina
- Department of General SurgeryThe Second HospitalCheeloo College of MedicineShandong UniversityShandong ProvinceChina
| | - Zhiguo Niu
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision MedicineXinxiang Medical UniversityXinxiang, Henan ProvinceChina
- Henan Key Laboratory of immunology and targeted therapySchool of Laboratory MedicineHenan Collaborative Innovation Center of Molecular Diagnosis and Laboratory MedicineSchool of Laboratory MedicineXinxiang Medical UniversityXinxiang, Henan ProvinceChina
| | - Ting Zhuang
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision MedicineXinxiang Medical UniversityXinxiang, Henan ProvinceChina
- Henan Key Laboratory of immunology and targeted therapySchool of Laboratory MedicineHenan Collaborative Innovation Center of Molecular Diagnosis and Laboratory MedicineSchool of Laboratory MedicineXinxiang Medical UniversityXinxiang, Henan ProvinceChina
| |
Collapse
|