1
|
Qiu C, Xu H. A Six-Gene Signature Related to Liquid-Liquid Phase Separation for Diagnosis of Alzheimer's Disease. ACTAS ESPANOLAS DE PSIQUIATRIA 2024; 52:759-768. [PMID: 39665604 PMCID: PMC11636544 DOI: 10.62641/aep.v52i6.1762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
BACKGROUND Liquid-liquid phase separation (LLPS) has been increasingly recognized as a crucial mechanism in the pathogenesis of various neurodegenerative disorders, including Alzheimer's disease (AD). There remains a paucity of effective diagnostic biomarkers for this condition. This study aims to develop and validate a novel LLPS-related molecular signature to enhance the diagnostic accuracy and early detection of AD. METHODS LLPS-related genes were identified from online databases and subjected to bioinformatic analyses, including protein-protein interaction (PPI) network analysis and least absolute shrinkage and selection operator (LASSO) regression. Based on the optimal LLPS-related genes, a diagnosis risk model was constructed, and the diagnostic ability was evaluated using a receiver operator characteristic (ROC) curve. To elucidate the biological functions of the identified LLPS-related genes, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were conducted. RESULTS A total of 149 LLPS-related genes were screened, which were found to be involved in functions related to oxidative stress, apoptosis, and cancer progression. The 149 genes were refined to six optimal candidates through PPI network analysis and LASSO regression: Activator of HSP90 ATPase Activity 1 (AHSA1), Eukaryotic Translation Initiation Factor 2 Alpha Kinase 2 (EIF2AK2), Heat Shock Protein Family A (Hsp70) Member 4 (HSPA4), Notch Receptor 1 (NOTCH1), Superoxide Dismutase 1 (SOD1), and Thioredoxin (TXN). Based on the six optimal genes, a diagnostic risk model was constructed, and the diagnostic ability was verified to be promising in AD both in training, internal validation, and two external validation datasets, with area under ROC curve (AUC) above 0.8. Furthermore, significant correlations were observed between the expression of these genes and tumor immune cell infiltration. CONCLUSIONS A six-gene diagnosis model was constructed and verified to exhibit robust diagnostic ability in AD.
Collapse
Affiliation(s)
- Chao Qiu
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), 310006 Hangzhou, Zhejiang, China
| | - Hui Xu
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), 310006 Hangzhou, Zhejiang, China
| |
Collapse
|
2
|
He SJ, Li J, Zhou JC, Yang ZY, Liu X, Ge YW. Chemical proteomics accelerates the target discovery of natural products. Biochem Pharmacol 2024; 230:116609. [PMID: 39510194 DOI: 10.1016/j.bcp.2024.116609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/15/2024]
Abstract
More than half of the global novel drugs are directly or indirectly derived from natural products (NPs) because of their better selectivity towards proteins. Traditional medicines perform multiple bioactivities through various NPs binding to drug targets, which highlights the opportunities of target discovery for drug development. However, detecting the binding relationship between NPs and targets remains challenging. Chemical proteomics, an interdisciplinary field of chemistry, proteomics, biology, and bioinformatics, has emerged as a potential approach for uncovering drug-target interactions. This review summarizes the principles and characteristics of the current widely applied chemical proteomic technologies, while delving into their latest applications in the target discovery of natural medicine. These endeavours demonstrate the potential of chemical proteomics for target discovery to supply dependable methodologies for the target elucidation of NPs.
Collapse
Affiliation(s)
- Shu-Jie He
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Innovative Team of Research on Effective Substances of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jun Li
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Innovative Team of Research on Effective Substances of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jie-Chun Zhou
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Innovative Team of Research on Effective Substances of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhi-You Yang
- College of Food Science and Technology, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Institute of Nutrition and Marine Drugs, Guangdong Ocean University, Zhanjiang, China
| | - Xi Liu
- School of Medical Information Engineering, Guangdong Pharmaceutical University, Guangzhou, China.
| | - Yue-Wei Ge
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Innovative Team of Research on Effective Substances of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China.
| |
Collapse
|
3
|
Pan X, Jiang S, Zhang X, Wang Z, Wang X, Cao L, Xiao W. Recent strategies in target identification of natural products: Exploring applications in chronic inflammation and beyond. Br J Pharmacol 2024. [PMID: 39428703 DOI: 10.1111/bph.17356] [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: 04/07/2024] [Revised: 08/01/2024] [Accepted: 08/25/2024] [Indexed: 10/22/2024] Open
Abstract
Natural products are a treasure trove for drug discovery, especially in the areas of infection, inflammation and cancer, due to their diverse bioactivities and complex, and varied structures. Chronic inflammation is closely related to many diseases, including complex diseases such as cancer and neurodegeneration. Improving target identification for natural products contributes to elucidating their mechanism of action and clinical progress. It also facilitates the discovery of novel druggable targets and the elimination of undesirable ones, thereby significantly enhancing the productivity of drug discovery and development. Moreover, the rise of polypharmacological strategies, considered promising for the treatment of complex diseases, will further increase the demand for target deconvolution. This review underscores strategies for identifying natural product targets (NPs) in the context of chronic inflammation over the past 5 years. These strategies encompass computational methodologies for early target discovery and the anticipation of compound binding sites, proteomics-driven approaches for target delineation and experimental biology techniques for target validation and comprehensive mechanistic exploration.
Collapse
Affiliation(s)
- Xian Pan
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
- Jiangsu Kanion Pharmaceutical Co Ltd, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
| | - Shan Jiang
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
- Jiangsu Kanion Pharmaceutical Co Ltd, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
| | - Xinzhuang Zhang
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
- Jiangsu Kanion Pharmaceutical Co Ltd, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
| | - Zhenzhong Wang
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
- Jiangsu Kanion Pharmaceutical Co Ltd, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
| | - Xin Wang
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Liang Cao
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
- Jiangsu Kanion Pharmaceutical Co Ltd, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Wei Xiao
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
- Jiangsu Kanion Pharmaceutical Co Ltd, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
| |
Collapse
|
4
|
Gao Y, Li Y, Liu Z, Dong Y, Yang S, Wu B, Xiao M, Chen C, Wen Y, Chen L, Jiang H, Yao Y. AHSA1 Regulates Hepatocellular Carcinoma Progression via the TGF-β/Akt-Cyclin D1/CDK6 Pathway. J Hepatocell Carcinoma 2023; 10:2021-2036. [PMID: 38022728 PMCID: PMC10640837 DOI: 10.2147/jhc.s407680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Background Activator of heat shock protein 90 (HSP90) ATPase Activity 1 (AHSA1) regulates proliferation, apoptosis, migration, and invasion of osteosarcoma and hepatocellular carcinoma (HCC). However, the novel mechanism of AHSA1 in the tumor biology of hepatocellular carcinoma (HCC) remains unclear. Methods We analyzed AHSA1 expression in 85 pairs of clinical samples of HCC and the Cancer Genome Atlas database. The role of AHSA1 in HCC was proved by cell proliferation, colony formation, migration, cell cycle analysis in vitro, xenograft models and tumor metastasis assay in vivo, and bioinformatics. Results High AHSA1 expression was demonstrated in HCC and associated with invasive depth, clinical stage, and poor overall survival of patients. Univariate Cox analysis confirmed that AHSA1 was an independent prognostic factor for patients with HCC. Meanwhile, AHSA1 upregulation promoted cell proliferation, colony formation, and cell migration in vitro and tumor cell proliferation and metastasis of HCC cells in vivo. AHSA1 upregulation increased the cell cycle transition from G1 to S phase by increasing the expression of cyclinD1, cyclinD3, and cyclin-dependent kinase 6(CD). Transforming growth factor beta 1 (TGF-β1)-induced protein kinase B (Akt) signaling regulated the expression of downstream targets, including cyclinD1. AHSA1 expression was closely correlated with the expression of TGF-β, Akt, cyclinD1, cyclinD3, and CDK6 using the Gene Expression Profiling Interactive Analysis database. AHSA1 upregulation participated in HCC progression by regulating TGF-β/Akt-cyclinD1/CDK6 signaling. Conclusion AHSA1 might serve as a biomarker for predicting the clinical outcome of patients with HCC. It is vital in tumor metastasis and disease progression of HCC and may facilitate the development of clinical intervention strategies against HCC.
Collapse
Affiliation(s)
- Yanjun Gao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
- Hubei Provincial Research Center for Precision Medicine of Cancer, Wuhan, 430200, People’s Republic of China
| | - Yingge Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
- Hubei Provincial Research Center for Precision Medicine of Cancer, Wuhan, 430200, People’s Republic of China
| | - Zheming Liu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
- Hubei Provincial Research Center for Precision Medicine of Cancer, Wuhan, 430200, People’s Republic of China
| | - Yi Dong
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
- Hubei Provincial Research Center for Precision Medicine of Cancer, Wuhan, 430200, People’s Republic of China
| | - Siqi Yang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
- Hubei Provincial Research Center for Precision Medicine of Cancer, Wuhan, 430200, People’s Republic of China
| | - Bin Wu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
- Department of Oncology, Huang-Gang Central Hospital, Huanggang, 438000, People’s Republic of China
| | - Mengxia Xiao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
- Department of Oncology, Yichun People’s Hospital, Yichun, 336000, People’s Republic of China
| | - Chen Chen
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
| | - Yingmei Wen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
- Hubei Provincial Research Center for Precision Medicine of Cancer, Wuhan, 430200, People’s Republic of China
| | - Lei Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
| | - Haijuan Jiang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, People’s Republic of China
| | - Yi Yao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
- Hubei Provincial Research Center for Precision Medicine of Cancer, Wuhan, 430200, People’s Republic of China
| |
Collapse
|
5
|
Li Z, Yin P. Tumor microenvironment diversity and plasticity in cancer multidrug resistance. Biochim Biophys Acta Rev Cancer 2023; 1878:188997. [PMID: 37832894 DOI: 10.1016/j.bbcan.2023.188997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/22/2023] [Accepted: 10/08/2023] [Indexed: 10/15/2023]
Abstract
Multidrug resistance (MDR) poses a significant obstacle to effective cancer treatment, and the tumor microenvironment (TME) is crucial for MDR development and reversal. The TME plays an active role in promoting MDR through several pathways. However, a promising therapeutic approach for battling MDR involves targeting specific elements within the TME. Therefore, this comprehensive review elaborates on the research developments regarding the dual role of the TME in promoting and reversing MDR in cancer. Understanding the complex role of the TME in promoting and reversing MDR is essential to developing effective cancer therapies. Utilizing the adaptability of the TME by targeting novel TME-specific factors, utilizing combination therapies, and employing innovative treatment strategies can potentially combat MDR and achieve personalized treatment outcomes for patients with cancer.
Collapse
Affiliation(s)
- Zhi Li
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China; Department of General surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China.
| | - Peihao Yin
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China.
| |
Collapse
|
6
|
Zhang Y, Qian J, Jiang M, Yang S, Zhou L, Zhang Q, Lin L, Yang Y. LTe2 induces cell apoptosis in multiple myeloma by suppressing AKT phosphorylation at Thr308 and Ser473. Front Oncol 2023; 13:1269670. [PMID: 37781194 PMCID: PMC10539572 DOI: 10.3389/fonc.2023.1269670] [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: 07/30/2023] [Accepted: 08/30/2023] [Indexed: 10/03/2023] Open
Abstract
Multiple myeloma (MM) is a highly heterogeneous hematological malignancy originating from B lymphocytes, with a high recurrence rate primarily due to drug resistance. 2-((1H-indol-3-yl)methyl)-3-((3-((1H-indol-3-yl)methyl)-1H-indol-2-yl)methyl)-1H-indole (LTe2), a tetrameric indole oligomer, possesses a wide range of anticancer activities through various mechanisms. Here, we aim to explore the anti-tumor efficiency and potential downstream targets of LTe2 in MM. Its bioactivity was assessed by employing MTT assays, flow cytometry, and the 5TMM3VT mouse model. Additionally, transcriptomic RNA-seq analysis and molecular dynamics (MD) experiments were conducted to elucidate the mechanism underlying LTe2 induced MM cell apoptosis. The results demonstrated that LTe2 significantly inhibited MM cell proliferation both in vitro and in vivo, and revealed that LTe2 exerts its effect by inhibiting the phosphorylation of AKT at the Thr308 and Ser473 sites. In summary, our findings highlight the potential of LTe2 as a novel candidate drug for MM treatment and provided a solid foundation for future clinical trials involving LTe2.
Collapse
Affiliation(s)
- Yuanjiao Zhang
- Nanjing Hospital of Chinese Medicine and School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiacheng Qian
- State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Mingmei Jiang
- Nanjing Hospital of Chinese Medicine and School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shu Yang
- Nanjing Hospital of Chinese Medicine and School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lianxin Zhou
- Nanjing Hospital of Chinese Medicine and School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qin Zhang
- Department of Gynecology, Jiangsu Province Hospital Affiliated Hospital of Nanjing Unviersity of Chinese Medicina, Nanjing, China
| | - Liping Lin
- State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ye Yang
- Nanjing Hospital of Chinese Medicine and School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
7
|
Wei R, Cao Y, Wu H, Liu X, Jiang M, Luo X, Deng Z, Wang Z, Ke M, Zhu Y, Chen S, Gu C, Yang Y. Inhibition of VCP modulates NF-κB signaling pathway to suppress multiple myeloma cell proliferation and osteoclast differentiation. Aging (Albany NY) 2023; 15:8220-8236. [PMID: 37606987 PMCID: PMC10497005 DOI: 10.18632/aging.204965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/20/2023] [Indexed: 08/23/2023]
Abstract
Multiple myeloma (MM) is the second most common hematological malignancy, in which the dysfunction of the ubiquitin-proteasome pathway is associated with the pathogenesis. The valosin containing protein (VCP)/p97, a member of the AAA+ ATPase family, possesses multiple functions to regulate the protein quality control including ubiquitin-proteasome system and molecular chaperone. VCP is involved in the occurrence and development of various tumors while still elusive in MM. VCP inhibitors have gradually shown great potential for cancer treatment. This study aims to identify if VCP is a therapeutic target in MM and confirm the effect of a novel inhibitor of VCP (VCP20) on MM. We found that VCP was elevated in MM patients and correlated with shorter survival in clinical TT2 cohort. Silencing VCP using siRNA resulted in decreased MM cell proliferation via NF-κB signaling pathway. VCP20 evidently inhibited MM cell proliferation and osteoclast differentiation. Moreover, exosomes containing VCP derived from MM cells partially alleviated the inhibitory effect of VCP20 on cell proliferation and osteoclast differentiation. Mechanism study revealed that VCP20 inactivated the NF-κB signaling pathway by inhibiting ubiquitination degradation of IκBα. Furthermore, VCP20 suppressed MM cell proliferation, prolonged the survival of MM model mice and improved bone destruction in vivo. Collectively, our findings suggest that VCP is a novel target in MM progression. Targeting VCP with VCP20 suppresses malignancy progression of MM via inhibition of NF-κB signaling pathway.
Collapse
Affiliation(s)
- Rongfang Wei
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuhao Cao
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hongjie Wu
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xin Liu
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Mingmei Jiang
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xian Luo
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhendong Deng
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ze Wang
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Mengying Ke
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yongqiang Zhu
- College of Life Science, Nanjing Normal University, Nanjing, China
| | - Siqing Chen
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Chunyan Gu
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ye Yang
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
8
|
Hammad S, Ogris C, Othman A, Erdoesi P, Schmidt-Heck W, Biermayer I, Helm B, Gao Y, Piorońska W, Holland CH, D'Alessandro LA, de la Torre C, Sticht C, Al Aoua S, Theis FJ, Bantel H, Ebert MP, Klingmüller U, Hengstler JG, Dooley S, Mueller NS. Tolerance of repeated toxic injuries of murine livers is associated with steatosis and inflammation. Cell Death Dis 2023; 14:414. [PMID: 37438332 DOI: 10.1038/s41419-023-05855-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 04/13/2023] [Accepted: 05/05/2023] [Indexed: 07/14/2023]
Abstract
The human liver has a remarkable capacity to regenerate and thus compensate over decades for fibrosis caused by toxic chemicals, drugs, alcohol, or malnutrition. To date, no protective mechanisms have been identified that help the liver tolerate these repeated injuries. In this study, we revealed dysregulation of lipid metabolism and mild inflammation as protective mechanisms by studying longitudinal multi-omic measurements of liver fibrosis induced by repeated CCl4 injections in mice (n = 45). Based on comprehensive proteomics, transcriptomics, blood- and tissue-level profiling, we uncovered three phases of early disease development-initiation, progression, and tolerance. Using novel multi-omic network analysis, we identified multi-level mechanisms that are significantly dysregulated in the injury-tolerant response. Public data analysis shows that these profiles are altered in human liver diseases, including fibrosis and early cirrhosis stages. Our findings mark the beginning of the tolerance phase as the critical switching point in liver response to repetitive toxic doses. After fostering extracellular matrix accumulation as an acute response, we observe a deposition of tiny lipid droplets in hepatocytes only in the Tolerant phase. Our comprehensive study shows that lipid metabolism and mild inflammation may serve as biomarkers and are putative functional requirements to resist further disease progression.
Collapse
Affiliation(s)
- Seddik Hammad
- Molecular Hepatology Section, Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- Department of Forensic Medicine and Veterinary Toxicology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt.
| | - Christoph Ogris
- Institute of Computational Biology, Helmholtz-Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Amnah Othman
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors (IfADo), Dortmund, Germany
| | - Pia Erdoesi
- Molecular Hepatology Section, Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Wolfgang Schmidt-Heck
- Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute, Jena, Germany
| | - Ina Biermayer
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), INF 280, Heidelberg, Germany
| | - Barbara Helm
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), INF 280, Heidelberg, Germany
| | - Yan Gao
- Molecular Hepatology Section, Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Weronika Piorońska
- Molecular Hepatology Section, Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christian H Holland
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors (IfADo), Dortmund, Germany
| | - Lorenza A D'Alessandro
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), INF 280, Heidelberg, Germany
| | - Carolina de la Torre
- Core Facility Next Generation Sequencing, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Carsten Sticht
- Core Facility Next Generation Sequencing, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Sherin Al Aoua
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz-Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Heike Bantel
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover, Germany
| | - Matthias P Ebert
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Clinical Cooperation Unit Healthy Metabolism, Center of Preventive Medicine and Digital Health, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ursula Klingmüller
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), INF 280, Heidelberg, Germany
| | - Jan G Hengstler
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors (IfADo), Dortmund, Germany
| | - Steven Dooley
- Molecular Hepatology Section, Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Nikola S Mueller
- Institute of Computational Biology, Helmholtz-Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
| |
Collapse
|
9
|
Gu C, Wang Y, Zhang L, Qiao L, Sun S, Shao M, Tang X, Ding P, Tang C, Cao Y, Zhou Y, Guo M, Wei R, Li N, Xiao Y, Duan J, Yang Y. Correction: AHSA1 is a promising therapeutic target for cellular proliferation and proteasome inhibitor resistance in multiple myeloma. J Exp Clin Cancer Res 2023; 42:91. [PMID: 37072818 PMCID: PMC10114437 DOI: 10.1186/s13046-023-02672-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023] Open
Affiliation(s)
- Chunyan Gu
- Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, 210023, China
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yajun Wang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lulin Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Li Qiao
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shanliang Sun
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Miaomiao Shao
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xiaozhu Tang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Pinggang Ding
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chao Tang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yuhao Cao
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yanyan Zhou
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Mengjie Guo
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Rongfang Wei
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Nianguang Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yibei Xiao
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| | - Jinao Duan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- State Administration of Traditional Chinese Medicine Key Laboratory of Chinese Medicinal Resources Recycling Utilization, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Ye Yang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
10
|
Yin Z, Lv Y, Deng L, Li G, Ou R, Chen L, Zhu Y, Zhong Q, Liu Z, Huang J, Wu H, Zhang Q, Fei J, Liu S. Targeting ABCB6 with nitidine chloride inhibits PI3K/AKT signaling pathway to promote ferroptosis in multiple myeloma. Free Radic Biol Med 2023; 203:86-101. [PMID: 37044150 DOI: 10.1016/j.freeradbiomed.2023.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/19/2023] [Accepted: 04/04/2023] [Indexed: 04/14/2023]
Abstract
Since multiple myeloma (MM) remains a cureless malignancy of plasma cells to date, it becomes imperative to develop novel drugs and therapeutic targets for MM. We screened a small molecule library comprising 3633 natural product drugs, which demonstrated that Nitidine Chloride (NC), an extract from traditional Chinese medicine Zanthoxylum nitidum. We used Surface Plasmon Resonance-High Performance Liquid Chromatography-Protein Mass Spectrometry (SPR-HPLC-MS), Cellular Thermal Shift Assay (CETSA), molecular docking, and SPR assay to identify the potential targets of NC, in which ABCB6 was the unique target of NC. The effects of ABCB6 on cellular proliferation and drug resistance were determined by CCK8, western blot, flow cytometry, site-mutation cells, transmission electron microscopy, immunohistochemistry staining and xenograft model in vitro and in vivo. NC induced MM cell death by promoting ferroptosis. ABCB6 is the direct target of NC. ABCB6 expression was increased in MM samples compared to normal controls, which was significantly associated with MM relapse and poor outcomes. VGSK was the inferred binding epitope of NC on the ABCB6 protein. In the ABCB6-mutated MM cells, NC did not display cancer resistance, implying the vital role of ABCB6 in NC's bioactivity. Moreover, the silencing of ABCB6 significantly inhibited MM cell growth. Mechanistically, the direct binding of NC to ABCB6 suppressed PI3K/AKT signaling pathway to promote ferroptosis. In conclusion, ABCB6 can be a potential therapeutic target and prognostic biomarker in MM, while NC can be considered a novel drug for MM treatment.
Collapse
Affiliation(s)
- Zhao Yin
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Yiwen Lv
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Li Deng
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Guangchao Li
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Ruiming Ou
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Lizhi Chen
- Department of Science and Education, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Yangmin Zhu
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Qi Zhong
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Zhi Liu
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Jing Huang
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Hong Wu
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Qing Zhang
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Jia Fei
- Department of Biochemistry and Molecular Biology, Medical College of Jinan University, Guangzhou, 510632, China
| | - Shuang Liu
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China.
| |
Collapse
|
11
|
Song Z, Zhao C, Yan J, Jiang D, Jia G. Carbenoxolone disodium suppresses the migration of gastric cancer by targeting HDAC6. Future Med Chem 2023; 15:333-344. [PMID: 36946221 DOI: 10.4155/fmc-2022-0246] [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] [Indexed: 03/23/2023] Open
Abstract
Aim: Because of the severe morbidity and mortality of gastric cancer, discovering new candidate drugs has been an urgent issue. The close association between histone deacetylase 6 (HDAC6) and gastric cancer makes the development of HDAC6-targeted anti-gastric cancer drugs a viable idea. Methods & results: Carbenoxolone disodium was identified as a novel HDAC6 inhibitor. Cellular thermal shift assay, surface plasmon resonance assay and molecular docking confirmed its binding ability to HDAC6. Cell viability, wound healing and transwell assays as well as animal studies have demonstrated that carbenoxolone disodium could block the proliferation and migration of gastric cancer cells MGC-803 in vitro and in vivo. Conclusion: This is the first report to indicate that carbenoxolone disodium could be an HDAC6 inhibitor with potential for treatment of gastric cancer.
Collapse
Affiliation(s)
- Zhiyu Song
- Department of Pharmacy, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| | - Chenglong Zhao
- Department of Pharmacy, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| | - Jingjing Yan
- Department of Pharmacy, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| | - Dandan Jiang
- Department of Pharmacy, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| | - Gang Jia
- Department of Oncology, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| |
Collapse
|
12
|
Bufalin-Mediated Regulation of Cell Signaling Pathways in Different Cancers: Spotlight on JAK/STAT, Wnt/β-Catenin, mTOR, TRAIL/TRAIL-R, and Non-Coding RNAs. Molecules 2023; 28:molecules28052231. [PMID: 36903477 PMCID: PMC10004807 DOI: 10.3390/molecules28052231] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 03/04/2023] Open
Abstract
The renaissance of research into natural products has unequivocally and paradigmatically shifted our knowledge about the significant role of natural products in cancer chemoprevention. Bufalin is a pharmacologically active molecule isolated from the skin of the toad Bufo gargarizans or Bufo melanostictus. Bufalin has characteristically unique properties to regulate multiple molecular targets and can be used to harness multi-targeted therapeutic regimes against different cancers. There is burgeoning evidence related to functional roles of signaling cascades in carcinogenesis and metastasis. Bufalin has been reported to regulate pleiotropically a myriad of signal transduction cascades in various cancers. Importantly, bufalin mechanistically regulated JAK/STAT, Wnt/β-Catenin, mTOR, TRAIL/TRAIL-R, EGFR, and c-MET pathways. Furthermore, bufalin-mediated modulation of non-coding RNAs in different cancers has also started to gain tremendous momentum. Similarly, bufalin-mediated targeting of tumor microenvironments and tumor macrophages is an area of exciting research and we have only started to scratch the surface of the complicated nature of molecular oncology. Cell culture studies and animal models provide proof-of-concept for the impetus role of bufalin in the inhibition of carcinogenesis and metastasis. Bufalin-related clinical studies are insufficient and interdisciplinary researchers require detailed analysis of the existing knowledge gaps.
Collapse
|
13
|
Zhu Y, Ouyang Z, Du H, Wang M, Wang J, Sun H, Kong L, Xu Q, Ma H, Sun Y. New opportunities and challenges of natural products research: When target identification meets single-cell multiomics. Acta Pharm Sin B 2022; 12:4011-4039. [PMID: 36386472 PMCID: PMC9643300 DOI: 10.1016/j.apsb.2022.08.022] [Citation(s) in RCA: 162] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/06/2022] [Accepted: 08/22/2022] [Indexed: 12/12/2022] Open
Abstract
Natural products, and especially the active ingredients found in traditional Chinese medicine (TCM), have a thousand-year-long history of clinical use and a strong theoretical basis in TCM. As such, traditional remedies provide shortcuts for the development of original new drugs in China, and increasing numbers of natural products are showing great therapeutic potential in various diseases. This paper reviews the molecular mechanisms of action of natural products from different sources used in the treatment of inflammatory diseases and cancer, introduces the methods and newly emerging technologies used to identify and validate the targets of natural active ingredients, enumerates the expansive list of TCM used to treat inflammatory diseases and cancer, and summarizes the patterns of action of emerging technologies such as single-cell multiomics, network pharmacology, and artificial intelligence in the pharmacological studies of natural products to provide insights for the development of innovative natural product-based drugs. Our hope is that we can make use of advances in target identification and single-cell multiomics to obtain a deeper understanding of actions of mechanisms of natural products that will allow innovation and revitalization of TCM and its swift industrialization and internationalization.
Collapse
Affiliation(s)
- Yuyu Zhu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zijun Ouyang
- Institute of Marine Biomedicine, School of Food and Drug, Shenzhen Polytechnic, Shenzhen 518055, China
| | - Haojie Du
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing 210023, China
| | - Meijing Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing 210023, China
| | - Jiaojiao Wang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Haiyan Sun
- Institute of Marine Biomedicine, School of Food and Drug, Shenzhen Polytechnic, Shenzhen 518055, China
| | - Lingdong Kong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing 210023, China
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing 210023, China
| | - Hongyue Ma
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing 210023, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| |
Collapse
|
14
|
Feng Y, Ma F, Wu E, Cheng Z, Wang Z, Yang L, Zhang J. Ginsenosides: Allies of gastrointestinal tumor immunotherapy. Front Pharmacol 2022; 13:922029. [PMID: 36386161 PMCID: PMC9659574 DOI: 10.3389/fphar.2022.922029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 09/26/2022] [Indexed: 09/25/2023] Open
Abstract
In the past decade, immunotherapy has been the most promising treatment for gastrointestinal tumors. But the low response rate and drug resistance remain major concerns. It is therefore imperative to develop adjuvant therapies to increase the effectiveness of immunotherapy and prevent drug resistance. Ginseng has been used in Traditional Chinese medicine as a natural immune booster for thousands of years. The active components of ginseng, ginsenosides, have played an essential role in tumor treatment for decades and are candidates for anti-tumor adjuvant therapy. They are hypothesized to cooperate with immunotherapy drugs to improve the curative effect and reduce tumor resistance and adverse reactions. This review summarizes the research into the use of ginsenosides in immunotherapy of gastrointestinal tumors and discusses potential future applications.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jiwei Zhang
- Shanghai Key Laboratory of Compound Chinese Medicines, The MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
15
|
AHSA1 Promotes Proliferation and EMT by Regulating ERK/CALD1 Axis in Hepatocellular Carcinoma. Cancers (Basel) 2022; 14:cancers14194600. [PMID: 36230524 PMCID: PMC9562867 DOI: 10.3390/cancers14194600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/13/2022] [Accepted: 09/17/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Globally, hepatocellular carcinoma is one of the leading causes of cancer-related death. Activator of HSP90 ATPase activity 1(AHSA1) was reported to be involved in regulating the maturation, stability, and degradation of related cancer-promoting proteins. However, the biological function and regulatory mechanism of AHSA1 in hepatocellular carcinoma were unclearly. In this study, we found AHSA1 was upregulated and associated with poor clinical characteristics and prognosis of hepatocellular carcinoma patients. Moreover, AHSA1 promoted proliferation, metastasis, epithelium-mesenchymal transition of hepatocellular carcinoma both in vitro and in vivo by recruiting ERK1/2 and promoting the phosphorylation and inactivation of CALD1. Taken together, our findings provided a mechanistic insight into the role of AHSA1 in hepatocellular carcinoma progression and implied AHSA1 may be a biomarker and therapeutic target for hepatocellular carcinoma patients. Abstract Hepatocellular carcinoma (HCC) is one of the major causes of cancer-related death worldwide. AHSA1 as a chaperone of HSP90 promotes the maturation, stability, and degradation of related cancer-promoting proteins. However, the regulatory mechanism and biological function of AHSA1 in HCC are largely unknown. Actually, we found that AHSA1 was significantly upregulated in HCC tissues and cell lines and was notably correlated with the poor clinical characteristics and prognosis of HCC patients in this study. Furthermore, both in vitro and in vivo, gain- and loss-of-function studies demonstrated that AHSA1 promoted the proliferation, invasion, metastasis, and epithelial-mesenchymal transition (EMT) of HCC. Moreover, the mechanistic study indicated that AHSA1 recruited ERK1/2 and promoted the phosphorylation and inactivation of CALD1, while ERK1/2 phosphorylation inhibitor SCH772984 reversed the role of AHSA1 in the proliferation and EMT of HCC. Furthermore, we demonstrated that the knockdown of CALD1 reversed the inhibition of proliferation and EMT by knocking AHSA1 in HCC. We also illustrated a new molecular mechanism associated with AHSA1 in HCC that was independent of HSP90 and MEK1/2. In summary, AHSA1 may play an oncogenic role in HCC by regulating ERK/CALD1 axis and may serve as a novel therapeutic target for HCC.
Collapse
|