1
|
Li C, Zhu D, Cao X, Li Y, Hao X. Knockdown of S100A2 inhibits the aggressiveness of endometrial cancer by activating STING pathway. J OBSTET GYNAECOL 2024; 44:2361849. [PMID: 38920019 DOI: 10.1080/01443615.2024.2361849] [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: 12/06/2023] [Accepted: 05/25/2024] [Indexed: 06/27/2024]
Abstract
BACKGROUND Endometrial cancer is a kind of gynaecological cancer. S100A2 is a newfound biomarker to diagnose endometrial cancer. This study was to investigate the role of S100A2 on regulating migration and invasion of endometrial cancer. METHODS The mRNA and protein levels of S100A2 were obtained by quantitative real-time polymerase chain reaction, immunohistochemistry and western blot methods. Cell viability was measured by the Cell Counting Kit-8 assay. Cell migration and invasion were quantified using transwell assays. Western blot assay was conducted to quantify protein expressions of epithelial to mesenchymal transition-related proteins (N-cadherin and E-cadherin). Furthermore, in vivo tumour formation experiments were performed to evaluate the role of S100A2 on tumour xenografts. RESULTS S100A2 was significantly up-regulated in endometrial cancer tissues. Knockdown of S100A2 inhibited cell viability, migration and invasion of endometrial cancer cells. Meanwhile, STING pathway was activated by the inhibited S100A2. STING inhibitor C-176 significantly reversed the effects of S100A2 knockdown on aggressive behaviours of endometrial cancer cells. Inhibition of S100A2 dramatically suppresses the tumour growth in vivo. CONCLUSIONS S100A2 functions as an oncogene in endometrial cancer. Targeting S100A2 may be a promising therapeutic method to treat endometrial carcinoma.
Collapse
Affiliation(s)
- Chengcheng Li
- Gynaecology and Obstetrics, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Dandan Zhu
- Gynaecology and Obstetrics, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xun Cao
- Rehabilitation Medicine Department, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Ying Li
- Gynaecology and Obstetrics, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiaoyuan Hao
- Gynaecology and Obstetrics, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
2
|
Yang X, Zheng F, Yan P, Liu X, Chen X, Du X, Zhang Y, Wang P, Chen C, Lu H, Bai Y. S100A2 activation promotes interstitial fibrosis in kidneys by FoxO1-mediated epithelial-mesenchymal transition. Cell Biol Toxicol 2024; 40:86. [PMID: 39382800 PMCID: PMC11464619 DOI: 10.1007/s10565-024-09929-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 10/02/2024] [Indexed: 10/10/2024]
Abstract
BACKGROUND Renal interstitial fibrosis (RIF) is a common feature of chronic kidney diseases (CKD), with epithelial-mesenchymal transition (EMT) being one of its important mechanisms. S100A2 is a protein associated with cell proliferation and differentiation, but its specific functions and molecular mechanisms in RIF remain to be determined. METHODS S100A2 levels were evaluated in three mouse models, including unilateral ureteral obstruction (UUO), ischemia-reperfusion injury (IRI), and aristolochic acid nephropathy (AAN), as well as in TGF-β1- treated HK-2 cells and in kidney tissue samples. Furthermore, the role of S100A2 and its interaction with FoxO1 was investigated using RT-qPCR, immunoblotting, immunofluorescence staining, co-immunoprecipitation (Co-IP), transcriptome sequencing, and gain- or loss-of-function approaches in vitro. RESULTS Elevated expression levels of S100A2 were observed in three mouse models and TGF-β1-treated HK2 cells, as well as in kidney tissue samples. Following siRNA silencing of S100A2, exposure to TGF-β1 in cultured HK-2 cells suppressed EMT process and extracellular matrix (ECM) accumulation. Conversely, Overexpression of S100A2 induced EMT and ECM deposition. Notably, we identified that S100A2-mediated EMT depends on FoxO1. Immunofluorescence staining indicated that S100A2 and FoxO1 colocalized in the nucleus and cytoplasm, and their interaction was verified in Co-IP assay. S100A2 knockdown decreased TGF-β1-induced phosphorylation of FoxO1 and increased its protein expression, whereas S100A2 overexpression hampered FoxO1 activation. Furthermore, pharmacological blockade of FoxO1 rescued the induction of TGF-β1 on EMT and ECM deposition in S100A2 siRNA-treated cells. CONCLUSION S100A2 activation exacerbates interstitial fibrosis in kidneys by facilitating FoxO1-mediated EMT.
Collapse
Affiliation(s)
- Xuejia Yang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Fan Zheng
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Penghua Yan
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Xueting Liu
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Xuanwen Chen
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Xinyu Du
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Yin Zhang
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Peilei Wang
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Chaosheng Chen
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
- Institute of Chronic Nephropathy, Wenzhou Medical University, Wenzhou, 325035, China.
| | - Hong Lu
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
| | - Yongheng Bai
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
- Institute of Chronic Nephropathy, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
3
|
Qi M, Yi X, Yue B, Huang M, Zhou S, Xiong J. S100A6 inhibits MDM2 to suppress breast cancer growth and enhance sensitivity to chemotherapy. Breast Cancer Res 2023; 25:55. [PMID: 37217945 DOI: 10.1186/s13058-023-01657-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 05/10/2023] [Indexed: 05/24/2023] Open
Abstract
BACKGROUND S100A6 and murine double minute 2 (MDM2) are important cancer-related molecules. A previous study identified an interaction between S100A6 and MDM2 by size exclusion chromatography and surface plasmon resonance experiments. The present study investigated whether S100A6 could bind to MDM2 in vivo and further explored its functional implication. METHODS Co-immunoprecipitation, glutathione-S-transferase pull-down assay, and immunofluorescence were performed to determine the in vivo interaction between S100A6 and MDM2. Cycloheximide pulse-chase assay and ubiquitination assay were performed to clarify the mechanism by which S100A6 downregulated MDM2. In addition, clonogenic assay, WST-1 assay, and flow cytometry of apoptosis and the cell cycle were performed and a xenograft model was established to evaluate the effects of the S100A6/MDM2 interaction on growth and paclitaxel-induced chemosensitivity of breast cancer. The expressions of S100A6 and MDM2 in patients with invasive breast cancer were analyzed by immunohistochemistry. In addition, the correlation between the expression of S100A6 and the response to neoadjuvant chemotherapy was statistically analyzed. RESULTS S100A6 promoted the MDM2 translocation from nucleus to cytoplasm, in which the S100A6 bound to the binding site of the herpesvirus-associated ubiquitin-specific protease (HAUSP) in MDM2, disrupted the MDM2-HAUSP-DAXX interactions, and induced the MDM2 self-ubiquitination and degradation. Furthermore, the S100A6-mediated MDM2 degradation suppressed the growth of breast cancer and enhanced its sensitivity to paclitaxel both in vitro and in vivo. For patients with invasive breast cancer who received epirubicin and cyclophosphamide followed by docetaxel (EC-T), expressions of S100A6 and MDM2 were negatively correlated, and high expression of S100A6 suggested a higher rate of pathologic complete response (pCR). Univariate and multivariate analyses showed that the high expression of S100A6 was an independent predictor of pCR. CONCLUSION These results reveal a novel function for S100A6 in downregulating MDM2, which directly enhances sensitivity to chemotherapy.
Collapse
Affiliation(s)
- Mengxin Qi
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xianglan Yi
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Baohui Yue
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Mingxiang Huang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Sheng Zhou
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Jing Xiong
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
4
|
S100A6 Protein-Expression and Function in Norm and Pathology. Int J Mol Sci 2023; 24:ijms24021341. [PMID: 36674873 PMCID: PMC9866648 DOI: 10.3390/ijms24021341] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 01/13/2023] Open
Abstract
S100A6, also known as calcyclin, is a calcium-binding protein belonging to the S100 protein family. It was first identified and purified more than 30 years ago. Initial structural studies, focused mostly on the mode and affinity of Ca2+ binding and resolution of the resultant conformational changes, were soon complemented by research on its expression, localization and identification of binding partners. With time, the use of biophysical methods helped to resolve the structure and versatility of S100A6 complexes with some of its ligands. Meanwhile, it became clear that S100A6 expression was altered in various pathological states and correlated with the stage/progression of many diseases, including cancers, indicative of its important, and possibly causative, role in some of these diseases. This, in turn, prompted researchers to look for the mechanism of S100A6 action and to identify the intermediary signaling pathways and effectors. After all these years, our knowledge on various aspects of S100A6 biology is robust but still incomplete. The list of S100A6 ligands is growing all the time, as is our understanding of the physiological importance of these interactions. The present review summarizes available data concerning S100A6 expression/localization, interaction with intracellular and extracellular targets, involvement in Ca2+-dependent cellular processes and association with various pathologies.
Collapse
|
5
|
Delangre E, Oppliger E, Berkcan S, Gjorgjieva M, Correia de Sousa M, Foti M. S100 Proteins in Fatty Liver Disease and Hepatocellular Carcinoma. Int J Mol Sci 2022; 23:ijms231911030. [PMID: 36232334 PMCID: PMC9570375 DOI: 10.3390/ijms231911030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 01/27/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a highly prevalent and slow progressing hepatic pathology characterized by different stages of increasing severity which can ultimately give rise to the development of hepatocellular carcinoma (HCC). Besides drastic lifestyle changes, few drugs are effective to some extent alleviate NAFLD and HCC remains a poorly curable cancer. Among the deregulated molecular mechanisms promoting NAFLD and HCC, several members of the S100 proteins family appear to play an important role in the development of hepatic steatosis, non-alcoholic steatohepatitis (NASH) and HCC. Specific members of this Ca2+-binding protein family are indeed significantly overexpressed in either parenchymal or non-parenchymal liver cells, where they exert pleiotropic pathological functions driving NAFLD/NASH to severe stages and/or cancer development. The aberrant activity of S100 specific isoforms has also been reported to drive malignancy in liver cancers. Herein, we discuss the implication of several key members of this family, e.g., S100A4, S100A6, S100A8, S100A9 and S100A11, in NAFLD and HCC, with a particular focus on their intracellular versus extracellular functions in different hepatic cell types. Their clinical relevance as non-invasive diagnostic/prognostic biomarkers for the different stages of NAFLD and HCC, or their pharmacological targeting for therapeutic purpose, is further debated.
Collapse
|
6
|
Liu H, Dehestani M, Blauwendraat C, Makarious MB, Leonard H, Kim JJ, Schulte C, Noyce A, Jacobs BM, Foote I, Sharma M, Nalls M, Singleton A, Gasser T, Bandres‐Ciga S. Polygenic Resilience Modulates the Penetrance of Parkinson Disease Genetic Risk Factors. Ann Neurol 2022; 92:270-278. [PMID: 35599344 PMCID: PMC9329258 DOI: 10.1002/ana.26416] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/06/2022] [Accepted: 05/09/2022] [Indexed: 11/10/2022]
Abstract
OBJECTIVE The aim of the current study is to understand why some individuals avoid developing Parkinson disease (PD) despite being at relatively high genetic risk, using the largest datasets of individual-level genetic data available. METHODS We calculated polygenic risk score to identify controls and matched PD cases with the highest burden of genetic risk for PD in the discovery cohort (International Parkinson's Disease Genomics Consortium, 7,204 PD cases and 9,412 controls) and validation cohorts (Comprehensive Unbiased Risk Factor Assessment for Genetics and Environment in Parkinson's Disease, 8,968 cases and 7,598 controls; UK Biobank, 2,639 PD cases and 14,301 controls; Accelerating Medicines Partnership-Parkinson's Disease Initiative, 2,248 cases and 2,817 controls). A genome-wide association study meta-analysis was performed on these individuals to understand genetic variation associated with resistance to disease. We further constructed a polygenic resilience score, and performed multimarker analysis of genomic annotation (MAGMA) gene-based analyses and functional enrichment analyses. RESULTS A higher polygenic resilience score was associated with a lower risk for PD (β = -0.054, standard error [SE] = 0.022, p = 0.013). Although no single locus reached genome-wide significance, MAGMA gene-based analyses nominated TBCA as a putative gene. Furthermore, we estimated the narrow-sense heritability associated with resilience to PD (h2 = 0.081, SE = 0.035, p = 0.0003). Subsequent functional enrichment analysis highlighted histone methylation as a potential pathway harboring resilience alleles that could mitigate the effects of PD risk loci. INTERPRETATION The present study represents a novel and comprehensive assessment of heritable genetic variation contributing to PD resistance. We show that a genetic resilience score can modify the penetrance of PD genetic risk factors and therefore protect individuals carrying a high-risk genetic burden from developing PD. ANN NEUROL 2022;92:270-278.
Collapse
Affiliation(s)
- Hui Liu
- Department for Neurodegenerative Diseases, Hertie Institute for Clinical Brain ResearchUniversity of Tübingen and German Center of Neurodegenerative DiseasesTübingenGermany
| | - Mohammad Dehestani
- Department for Neurodegenerative Diseases, Hertie Institute for Clinical Brain ResearchUniversity of Tübingen and German Center of Neurodegenerative DiseasesTübingenGermany
| | - Cornelis Blauwendraat
- Laboratory of Neurogenetics, Molecular Genetics Section, National Institute on AgingNational Institutes of HealthBethesdaMDUSA
- Center for Alzheimer's and Related DementiasNational Institutes of HealthBethesdaMDUSA
| | - Mary B. Makarious
- Laboratory of Neurogenetics, Molecular Genetics Section, National Institute on AgingNational Institutes of HealthBethesdaMDUSA
- Center for Alzheimer's and Related DementiasNational Institutes of HealthBethesdaMDUSA
- Data Tecnica InternationalGlen EchoMDUSA
- Department of Clinical and Movement NeurosciencesUCL Queen Square Institute of NeurologyLondonUK
| | - Hampton Leonard
- Data Tecnica InternationalGlen EchoMDUSA
- Department of Clinical and Movement NeurosciencesUCL Queen Square Institute of NeurologyLondonUK
| | - Jonggeol J. Kim
- Laboratory of Neurogenetics, Molecular Genetics Section, National Institute on AgingNational Institutes of HealthBethesdaMDUSA
- Preventive Neurology Unit, Wolfson Institute of Population HealthQueen Mary University of LondonLondonUK
| | - Claudia Schulte
- Department for Neurodegenerative Diseases, Hertie Institute for Clinical Brain ResearchUniversity of Tübingen and German Center of Neurodegenerative DiseasesTübingenGermany
| | - Alastair Noyce
- Preventive Neurology Unit, Wolfson Institute of Population HealthQueen Mary University of LondonLondonUK
| | - Benjamin M. Jacobs
- Preventive Neurology Unit, Wolfson Institute of Population HealthQueen Mary University of LondonLondonUK
| | - Isabelle Foote
- Preventive Neurology Unit, Wolfson Institute of Population HealthQueen Mary University of LondonLondonUK
| | - Manu Sharma
- Department for Neurodegenerative Diseases, Hertie Institute for Clinical Brain ResearchUniversity of Tübingen and German Center of Neurodegenerative DiseasesTübingenGermany
- Center for Genetic Epidemiology, Institute for Clinical Epidemiology and Functional BiometryUniversity of TübingenTübingenGermany
| | - Mike Nalls
- Center for Alzheimer's and Related DementiasNational Institutes of HealthBethesdaMDUSA
- Data Tecnica InternationalGlen EchoMDUSA
| | - Andrew Singleton
- Laboratory of Neurogenetics, Molecular Genetics Section, National Institute on AgingNational Institutes of HealthBethesdaMDUSA
- Center for Alzheimer's and Related DementiasNational Institutes of HealthBethesdaMDUSA
| | - Thomas Gasser
- Department for Neurodegenerative Diseases, Hertie Institute for Clinical Brain ResearchUniversity of Tübingen and German Center of Neurodegenerative DiseasesTübingenGermany
| | - Sara Bandres‐Ciga
- Laboratory of Neurogenetics, Molecular Genetics Section, National Institute on AgingNational Institutes of HealthBethesdaMDUSA
- Center for Alzheimer's and Related DementiasNational Institutes of HealthBethesdaMDUSA
| |
Collapse
|
7
|
Design, Synthesis, Molecular Docking, and Biological Evaluation of Pyrazole Hybrid Chalcone Conjugates as Potential Anticancer Agents and Tubulin Polymerization Inhibitors. Pharmaceuticals (Basel) 2022; 15:ph15030280. [PMID: 35337078 PMCID: PMC8954831 DOI: 10.3390/ph15030280] [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: 12/29/2021] [Revised: 02/10/2022] [Accepted: 02/11/2022] [Indexed: 12/02/2022] Open
Abstract
Some (E)-3-(3-(4-(benzyloxy)phenyl)-1-phenyl-1H-pyrazol-4-yl)-1-phenylprop-2-en-1-one conjugates 5a–r were designed; synthesized; characterized by 1H, 13C NMR, and ESI-MS; and evaluated for tubulin polymerization inhibitory activity and in vitro cytotoxicity against breast (MCF-7), cervical (SiHa), and prostate (PC-3) cancer cell lines, as well as a normal cell line (HEK-293T). The compounds were also tested to determine their binding modes at the colchicine-binding site of tubulin protein (PDB ID-3E22), for in silico ADME prediction, for bioactivity study, and for PASS prediction studies. Among all the synthesized conjugates, compound 5o exhibited excellent cytotoxicity with an IC50 value of 2.13 ± 0.80 µM (MCF-7), 4.34 ± 0.98 µM (SiHa), and 4.46 ± 0.53 µM (PC-3) against cancer cell lines. The compound did not exhibit significant toxicity to the HEK cells. Results of the in silico prediction revealed that the majority of the conjugates possessed drug-like properties.
Collapse
|
8
|
Tomar A, Polygalov D, McHugh TJ. Differential Impact of Acute and Chronic Stress on CA1 Spatial Coding and Gamma Oscillations. Front Behav Neurosci 2021; 15:710725. [PMID: 34354574 PMCID: PMC8329706 DOI: 10.3389/fnbeh.2021.710725] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 06/28/2021] [Indexed: 11/13/2022] Open
Abstract
Chronic and acute stress differentially affect behavior as well as the structural integrity of the hippocampus, a key brain region involved in cognition and memory. However, it remains unclear if and how the facilitatory effects of acute stress on hippocampal information coding are disrupted as the stress becomes chronic. To examine this, we compared the impact of acute and chronic stress on neural activity in the CA1 subregion of male mice subjected to a chronic immobilization stress (CIS) paradigm. We observed that following first exposure to stress (acute stress), the spatial information encoded in the hippocampus sharpened, and the neurons became increasingly tuned to the underlying theta oscillations in the local field potential (LFP). However, following repeated exposure to the same stress (chronic stress), spatial tuning was poorer and the power of both the slow-gamma (30–50 Hz) and fast-gamma (55–90 Hz) oscillations, which correlate with excitatory inputs into the region, decreased. These results support the idea that acute and chronic stress differentially affect neural computations carried out by hippocampal circuits and suggest that acute stress may improve cognitive processing.
Collapse
Affiliation(s)
- Anupratap Tomar
- Laboratory for Circuit and Behavioral Physiology, RIKEN Center for Brain Science, Saitama, Japan
| | - Denis Polygalov
- Laboratory for Circuit and Behavioral Physiology, RIKEN Center for Brain Science, Saitama, Japan
| | - Thomas J McHugh
- Laboratory for Circuit and Behavioral Physiology, RIKEN Center for Brain Science, Saitama, Japan
| |
Collapse
|