1
|
Tung MC, Oner M, Soong SW, Cheng PT, Li YH, Chen MC, Chou CK, Kang HY, Lin FCF, Tsai SCS, Lin H. CDK5 targets p21 CIP1 to regulate thyroid cancer cell proliferation and malignancy in patients. Mol Med Rep 2025; 32:182. [PMID: 40280108 DOI: 10.3892/mmr.2025.13547] [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: 10/21/2024] [Accepted: 03/12/2025] [Indexed: 04/29/2025] Open
Abstract
Cyclin‑dependent kinase 5 (CDK5), known for its role in neuronal function, has emerged as a key player in cancer biology, particularly in thyroid cancer. The present study explored the interaction between CDK5 and the cyclin‑dependent kinase inhibitor p21CIP1 in thyroid cancer (TC). Bioinformatic tools and immunoprecipitation assays were used to confirm that CDK5 targets p21 for ubiquitin‑mediated degradation, reducing its stability and tumor‑suppressive effects. Data from The Cancer Genome Atlas revealed a significant inverse correlation between CDK5 and p21 expression, with higher CDK5 levels linked to increased tumor malignancy and worse survival outcomes; conversely, higher p21 expression was correlated with an improved prognosis. Immunohistochemistry analysis of TC samples further confirmed that increased CDK5 and reduced p21 expression were associated with more advanced tumor stages and aggressive phenotypes. These findings suggested that CDK5‑mediated degradation of p21 contributes to TC progression and malignancy, highlighting the potential of targeting the CDK5‑p21 axis as a therapeutic strategy for management of TC.
Collapse
Affiliation(s)
- Min-Che Tung
- Department of Surgery, Tungs' Taichung MetroHarbor Hospital, Taichung 43503, Taiwan, R.O.C
| | - Muhammet Oner
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan, R.O.C
| | - Shiuan-Woei Soong
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan, R.O.C
| | - Pang-Ting Cheng
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan, R.O.C
| | - Yu-Hsuan Li
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan, R.O.C
| | - Mei-Chih Chen
- Translational Cell Therapy Center, Department of Medical Research, China Medical University Hospital, Taichung 40447, Taiwan, R.O.C
| | - Chen-Kai Chou
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kaohsiung 833, Taiwan, R.O.C
| | - Hong-Yo Kang
- Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Taoyuan 83301, Taiwan, R.O.C
| | - Frank Cheau-Feng Lin
- School of Medicine, Chung Shan Medical University, Taichung 402367, Taiwan, R.O.C
| | - Stella Chin-Shaw Tsai
- Department of Otolaryngology, Tungs' Taichung MetroHarbor Hospital, Taichung 43503, Taiwan, R.O.C
| | - Ho Lin
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan, R.O.C
| |
Collapse
|
2
|
Wang J, Zhang C, Jiang T, He Y, Wu Y, Zhou D, Yan J, Zhou Y. CDK5: Insights into its roles in diseases. Mol Biol Rep 2025; 52:145. [PMID: 39836243 DOI: 10.1007/s11033-025-10253-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
Cyclin-dependent kinase 5 (CDK5), a unique member of the CDK family, is a proline-directed serine/threonine protein kinase with critical roles in various physiological and pathological processes. Widely expressed in the central nervous system, CDK5 is strongly implicated in neurological diseases. Beyond its neurological roles, CDK5 is involved in metabolic disorders, psychiatric conditions, and tumor progression, contributing to processes such as proliferation, migration, immune evasion, genomic stability, and angiogenesis. This review explores the structure and biological functions of CDK5, highlighting its regulatory roles in disease development through the phosphorylation of diverse substrate proteins. Additionally, we examine the therapeutic potential of CDK5 inhibition, offering novel perspectives for disease diagnosis and treatment.
Collapse
Affiliation(s)
- Jiahui Wang
- Institute of Pathogenic Biology, Guilin Medical University, Guilin, 541199, China
| | - Chong Zhang
- Department of Neurology, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, China
- Guangxi Medical and Health Key Cultivation Discipline Construction Project, Guilin, 541199, China
| | - Tingting Jiang
- Institute of Pathogenic Biology, Guilin Medical University, Guilin, 541199, China
| | - Yi He
- Institute of Pathogenic Biology, Guilin Medical University, Guilin, 541199, China
| | - Yongli Wu
- Institute of Pathogenic Biology, Guilin Medical University, Guilin, 541199, China
| | - Dongsheng Zhou
- Guangxi Medical and Health Key Cultivation Discipline Construction Project, Guilin, 541199, China
- Department of Infectious Diseases, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, China
| | - Jianguo Yan
- Guangxi Medical and Health Key Cultivation Discipline Construction Project, Guilin, 541199, China.
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, 541199, China.
- Faculty of Basic Medical Sciences, Guilin Medical University, Guilin, 541199, China.
| | - Yali Zhou
- Institute of Pathogenic Biology, Guilin Medical University, Guilin, 541199, China.
- Guangxi Medical and Health Key Cultivation Discipline Construction Project, Guilin, 541199, China.
- Faculty of Basic Medical Sciences, Guilin Medical University, Guilin, 541199, China.
| |
Collapse
|
3
|
Mehrabadi S, Izadi FS, Pasha S, Pourali R, Khazaei M, Hassanian SM, Ferns GA, Avan A. The Potential Therapeutic Applications of CRISPR/Cas9 in the Treatment of Gastrointestinal Cancers. Curr Mol Med 2025; 25:278-288. [PMID: 38243923 DOI: 10.2174/0115665240243076231116080113] [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: 05/22/2023] [Revised: 09/10/2023] [Accepted: 09/12/2023] [Indexed: 01/22/2024]
Abstract
Gastrointestinal (GI) cancer is one the most prevalent types of cancer. Despite current chemotherapy's success, patients with GI cancer continue to have a dismal outcome. The onset and progression of cancer are caused by alterations and the abnormal expression of several families of genes, like tumor-suppressor genes, oncogenes, and chemotherapy-resistant genes. The final purpose of tumor therapy is to inhibit cellular development by modifying mutations and editing the irregular expression of genes It has been reported that CDH1, TP53, KRAS, ARID1A, PTEN, and HLA-B are the commonly mutated genes in GI cancer. Gene editing has become one potential approach for cases with advanced or recurrent CRC, who are nonresponsive to conventional treatments and a variety of driver mutations along with progression cause GI progression. CRISPR/Cas9 technique is a reliable tool to edit the genome and understand the functions of mutations driving GI cancer development. CRISPR/Cas9 can be applied to genome therapy for GI cancers, particularly with reference to molecular-targeted medicines and suppressors. Moreover, it can be used as a therapeutic approach by knocking in/out multiple genes. The use of CRISPR/ Cas9 gene editing method for GI cancer therapy has therefore resulted in some improvements. There are several research works on the role of CRISPR/Cas9 in cancer treatment that are summarized in the following separate sections. Here, the use of CRISPR/Cas9-based genome editing in GI and the use of CRISPR/Cas9 is discussed in terms of Targeting Chemotherapy Resistance-related Genes like; KRAS, TP53, PTEN, and ARID1A.
Collapse
Affiliation(s)
- Shima Mehrabadi
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Faezeh Salmani Izadi
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shiva Pasha
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Roozbeh Pourali
- Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex, BN1 9PH, UK
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- College of Medicine, University of Warith Al-Anbiyaa, Karbala, Iraq
| |
Collapse
|
4
|
Liu L, Chen J, Ye F, Chu F, Rao C, Wang Y, Yan Y, Wu J. Prognostic value of oxidative phosphorylation-related genes in hepatocellular carcinoma. Discov Oncol 2024; 15:258. [PMID: 38960931 PMCID: PMC11222354 DOI: 10.1007/s12672-024-01129-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 06/27/2024] [Indexed: 07/05/2024] Open
Abstract
PURPOSE Hepatocellular carcinoma (HCC) is the most prevalent malignancies worldwide. Recently, oxidative phosphorylation (OXPHOS) has received extensive concern as an emerging target in antitumor therapy. However, the OXPHOS-involved underlying genes and clinical utilization in HCC remain worth exploring. The present research aimed to create an OXPHOS-relevant signature in HCC. PATIENTS AND METHODS In this study, the prognostic signature genes linked with OXPHOS were identified, and prognostic models were built using least absolute shrinkage and selection operator (LASSO) cox regression analysis. Furthermore, the combination study of immune microenvironment and signature genes looked into the involvement of immune cells in signature-based genes in HCC. Following that, chemotherapeutic drug sensitivity and immunotherapy analysis was implemented to predict clinical efficacy in HCC patients. Finally, clinical samples were collected to measure the expression of OXPHOS-related signature genes. RESULTS Following a series of screens, six prognostic signature genes related with OXPHOS were identified: MRPS23, MPV17, MAPK3, IGF2BP2, CDK5, and IDH2, on which a risk model was built. The findings revealed a significant drop in the survival rate of HCC patients as their risk score increased. Meanwhile, independent prognostic study demonstrated that the risk score could accurately identify HCC patients. Immuno-microenvironmental correlation research suggested that the prognostic characteristics could serve as a reference index for both immunotherapy and chemotherapy. Finally, RT-qPCR exhibited a trend in signature gene expression that was consistent with the results. CONCLUSION In this study, a total of six prognostic genes associated with OXPHOS were selected and a prognostic model was constructed, providing an essential reference for the study of OXPHOS in HCC.
Collapse
Affiliation(s)
- Luzheng Liu
- Department of Interventional Radiology and Vascular Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570100, Hainan Province, China
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan Province, China
| | - Jiacheng Chen
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan Province, China
| | - Fei Ye
- Department of Blood Cell Therapy, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570100, Hainan Province, China
| | - Fengran Chu
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan Province, China
| | - Chaoluan Rao
- Department of Nursing, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570100, Hainan Province, China
| | - Yong Wang
- Department of Interventional Radiology and Vascular Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570100, Hainan Province, China
| | - Yanggang Yan
- Department of Interventional Radiology and Vascular Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570100, Hainan Province, China.
| | - Jincai Wu
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan Province, China.
| |
Collapse
|
5
|
Zhang HY, Zhu JJ, Liu ZM, Zhang YX, Chen JJ, Chen KD. A prognostic four-gene signature and a therapeutic strategy for hepatocellular carcinoma: Construction and analysis of a circRNA-mediated competing endogenous RNA network. Hepatobiliary Pancreat Dis Int 2024; 23:272-287. [PMID: 37407412 DOI: 10.1016/j.hbpd.2023.06.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 06/13/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) has a poor long-term prognosis. The competition of circular RNAs (circRNAs) with endogenous RNA is a novel tool for predicting HCC prognosis. Based on the alterations of circRNA regulatory networks, the analysis of gene modules related to HCC is feasible. METHODS Multiple expression datasets and RNA element targeting prediction tools were used to construct a circRNA-microRNA-mRNA network in HCC. Gene function, pathway, and protein interaction analyses were performed for the differentially expressed genes (DEGs) in this regulatory network. In the protein-protein interaction network, hub genes were identified and subjected to regression analysis, producing an optimized four-gene signature for prognostic risk stratification in HCC patients. Anti-HCC drugs were excavated by assessing the DEGs between the low- and high-risk groups. A circRNA-microRNA-hub gene subnetwork was constructed, in which three hallmark genes, KIF4A, CCNA2, and PBK, were subjected to functional enrichment analysis. RESULTS A four-gene signature (KIF4A, CCNA2, PBK, and ZWINT) that effectively estimated the overall survival and aided in prognostic risk assessment in the The Cancer Genome Atlas (TCGA) cohort and International Cancer Genome Consortium (ICGC) cohort was developed. CDK inhibitors, PI3K inhibitors, HDAC inhibitors, and EGFR inhibitors were predicted as four potential mechanisms of drug action (MOA) in high-risk HCC patients. Subsequent analysis has revealed that PBK, CCNA2, and KIF4A play a crucial role in regulating the tumor microenvironment by promoting immune cell invasion, regulating microsatellite instability (MSI), and exerting an impact on HCC progression. CONCLUSIONS The present study highlights the role of the circRNA-related regulatory network, identifies a four-gene prognostic signature and biomarkers, and further identifies novel therapy for HCC.
Collapse
Affiliation(s)
- Hai-Yan Zhang
- Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jia-Jie Zhu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China
| | - Zong-Ming Liu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China
| | - Yu-Xuan Zhang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China
| | - Jia-Jia Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Ke-Da Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China.
| |
Collapse
|
6
|
Palaz F, Ozsoz M, Zarrinpar A, Sahin I. CRISPR in Targeted Therapy and Adoptive T Cell Immunotherapy for Hepatocellular Carcinoma. J Hepatocell Carcinoma 2024; 11:975-995. [PMID: 38832119 PMCID: PMC11146628 DOI: 10.2147/jhc.s456683] [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: 12/26/2023] [Accepted: 05/21/2024] [Indexed: 06/05/2024] Open
Abstract
Despite recent therapeutic advancements, outcomes for advanced hepatocellular carcinoma (HCC) remain unsatisfactory, highlighting the need for novel treatments. The CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) gene-editing technology offers innovative treatment approaches, involving genetic manipulation of either cancer cells or adoptive T cells to combat HCC. This review comprehensively assesses the applications of CRISPR systems in HCC treatment, focusing on in vivo targeting of cancer cells and the development of chimeric antigen receptor (CAR) T cells and T cell receptor (TCR)-engineered T cells. We explore potential synergies between CRISPR-based cancer therapeutics and existing treatment options, discussing ongoing clinical trials and the role of CRISPR technology in improving HCC treatment outcomes with advanced safety measures. In summary, this review provides insights into the promising prospects and current challenges of using CRISPR technology in HCC treatment, with the ultimate goal of improving patient outcomes and revolutionizing the landscape of HCC therapeutics.
Collapse
Affiliation(s)
- Fahreddin Palaz
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Mehmet Ozsoz
- Department of Biomedical Engineering, Near East University, Nicosia, Turkey
| | - Ali Zarrinpar
- Department of Surgery, College of Medicine, University of Florida, Gainesville, FL, USA
- University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Ilyas Sahin
- University of Florida Health Cancer Center, Gainesville, FL, USA
- Division of Hematology and Oncology, Department of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
7
|
Wang C, Dong R, Yang F, Zheng L, Liu Y, Yan Y, Zhang M, Ni B, Li J. LARP4B promotes hepatocellular carcinoma progression and impairs sorafenib efficacy by activating SPINK1-mediated EGFR pathway. Cell Death Discov 2024; 10:208. [PMID: 38693111 PMCID: PMC11063073 DOI: 10.1038/s41420-024-01985-6] [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: 09/15/2023] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/03/2024] Open
Abstract
La-related proteins (LARPs) regulate gene expression by binding to RNAs and exhibit critical effects on disease progression, including tumors. However, the role of LARP4B and its underlying mechanisms in the progression of hepatocellular carcinoma (HCC) remain largely unclear. In this study, we found that LARP4B expression is upregulated and correlates with poor prognosis in patients with HCC. Gain- and loss-of-function assays showed that LARP4B promotes stemness, proliferation, metastasis, and angiogenesis in vitro and in vivo. Furthermore, LARP4B inhibition enhances the antitumor effects of sorafenib and blocks the metastasis-enhancing effects of low sorafenib concentrations in HCC. Mechanistically, LARP4B expression is upregulated by METTL3-mediated N6-methyladenosine (m6A)-IGF2BP3-dependent modification in HCC. RNA- and RNA immunoprecipitation (RIP)- sequencing uncovered that LARP4B upregulates SPINK1 by binding to SPINK1 mRNA via the La motif and maintaining mRNA stability. LARP4B activates the SPINK1-mediated EGFR signaling pathway, which supports stemness, progression and sorafenib resistance in HCC. Additionally, a positive feedback loop with the LARP4B/SPINK1/p-AKT/C/EBP-β axis is responsible for the sorafenib-therapeutic benefit of LARP4B depletion. Overall, this study demonstrated that LARP4B facilitates HCC progression, and LARP4B inhibition provides benefits to sorafenib treatment in HCC, suggesting that LARP4B might be a potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Chuanxu Wang
- Department of Hepatobiliary Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Rui Dong
- Department of Pathophysiology, Third Military Medical University (Army Medical University), Chongqing, China
- Chongqing International Institute for Immunology, Chongqing, China
| | - Feicheng Yang
- Department of Pathology, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, China
| | - Lu Zheng
- Department of Hepatobiliary Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yingling Liu
- Department of Hepatobiliary Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yue Yan
- Department of Hepatobiliary Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Mengjie Zhang
- Department of Pathophysiology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Bing Ni
- Department of Pathophysiology, Third Military Medical University (Army Medical University), Chongqing, China.
| | - Jing Li
- Department of Hepatobiliary Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
8
|
Li Y, Zheng Z, Xiao L, Chen Y, Liu X, Long D, Chai L, Li Y, Tan C. Dinaciclib exerts a tumor-suppressing effect via β-catenin/YAP axis in pancreatic ductal adenocarcinoma. Anticancer Drugs 2024; 35:140-154. [PMID: 37694833 DOI: 10.1097/cad.0000000000001545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Dinaciclib, a cyclin-dependent kinase-5 (CDK5) inhibitor, has significant anti-tumor properties. However, the precise mechanism of dinaciclib requires further investigation. Herein, we investigated the anti-tumor functions and molecular basis of dinaciclib in pancreatic ductal adenocarcinoma (PDAC). PDAC and matched para-carcinoma specimens were collected from the patients who underwent radical resection. Immunohistochemistry was performed to assess CDK5 expression. Cell proliferation ability, migration, and invasion were measured using Cell Counting Kit-8, wound healing, and transwell assay, respectively. The cell cycle and apoptosis were assessed using flow cytometry. Gene expression was examined using RNA-seq and quantitative real-time PCR. Protein expression of proteins was measured by western blot analysis and immunofluorescence microscopy. Tumor-bearing mice were intraperitoneally injected with dinaciclib. CDK5 is highly expressed in PDAC. The expression level of CDK5 was significantly related to tumor size, T stage, and the American Joint Committee on Cancer stage. High CDK5 expression can predict poor survival in PDAC patients. In addition, the expression level of CDK5 might be an independent prognostic factor for PDAC patients. Dinaciclib inhibits the growth and motility of PDAC cells and induces apoptosis and cell cycle arrest in the G2/M phase. Mechanistically, dinaciclib down-regulated yes-associated protein (YAP) mRNA and protein expression by reducing β-catenin expression. Moreover, dinaciclib significantly inhibited PDAC cell growth in vivo . Our findings reveal a novel anti-tumor mechanism of dinaciclib in which it decreases YAP expression by down-regulating β-catenin at the transcriptional level rather than by activating Hippo pathway-mediated phosphorylation-dependent degradation.
Collapse
Affiliation(s)
- Yichen Li
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University
| | - Zhenjiang Zheng
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University
| | - Li Xiao
- Department of Traditional Chinese Medicine, Chengdu Third People's Hospital
| | - Yonghua Chen
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University
| | - Xubao Liu
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University
| | - Dan Long
- Key Laboratory of Transplant Engineering and Immunology, National Clinical Research Center for Geriatrics, Frontiers Science Center for Diseaserelated Molecular Network, West China Hospital, Sichuan University
| | - Li Chai
- Research Core Facility of West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yi Li
- Research Core Facility of West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Chunlu Tan
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University
| |
Collapse
|
9
|
Huang L, Xia L, Nie T, Cui B, Lu J, Lu F, Fan F, Ren D, Lu Y, Gao G, Yang Q. Maintaining Drosha expression with Cdk5 inhibitors as a potential therapeutic strategy for early intervention after TBI. Exp Mol Med 2024; 56:210-219. [PMID: 38200156 PMCID: PMC10834983 DOI: 10.1038/s12276-023-01152-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 09/23/2023] [Accepted: 10/26/2023] [Indexed: 01/12/2024] Open
Abstract
Traumatic brain injury (TBI) is a major cause of death and disability in adults. The pathological process of TBI involves a multifactorial cascade in which kinases have been proven contribute to interactions between relevant factors and amplification of signaling cascades. Cyclin-dependent kinase 5 (Cdk5) is a promising kinase that has been implicated in various brain disorders, including TBI. However, the mechanism by which Cdk5 induces neuronal damage remains unclear. Here, we show for the first time that Drosha, a key enzyme in microRNA biogenesis, is a pivotal substrate of abnormally activated Cdk5. Cdk5-mediated phosphorylation decreases Drosha expression and exacerbates nerve injury in TBI. We proved that maintaining Drosha expression via the administration of repurposed Cdk5 inhibitors that were previously studied in clinical trials is a promising approach for the early treatment of TBI. Together, our work identifies Drosha as a novel target for neuroprotective strategies after TBI and suggests Cdk5-mediated regulation of Drosha expression as a potential therapeutic strategy for early TBI intervention.
Collapse
Affiliation(s)
- Lu Huang
- Department of Experimental Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
- Department of Anesthesiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Li Xia
- Department of Experimental Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Tiejian Nie
- Department of Experimental Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Bozhou Cui
- Department of Experimental Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Jianjun Lu
- Department of Experimental Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Fangfang Lu
- Department of Experimental Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Feiyan Fan
- Department of Experimental Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Dongni Ren
- Department of Experimental Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Yuan Lu
- Department of Experimental Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Guodong Gao
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Qian Yang
- Department of Experimental Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi, China.
| |
Collapse
|
10
|
Jiang Y, Xu Y, Zhu C, Xu G, Xu L, Rao Z, Zhou L, Jiang P, Malik S, Fang J, Lin H, Zhang M. STAT3 palmitoylation initiates a positive feedback loop that promotes the malignancy of hepatocellular carcinoma cells in mice. Sci Signal 2023; 16:eadd2282. [PMID: 38051779 PMCID: PMC10907978 DOI: 10.1126/scisignal.add2282] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 11/14/2023] [Indexed: 12/07/2023]
Abstract
Constitutive activation of the transcription factor STAT3 (signal transducer and activator of transcription 3) contributes to the malignancy of many cancers such as hepatocellular carcinoma (HCC) and is associated with poor prognosis. STAT3 activity is increased by the reversible palmitoylation of Cys108 by the palmitoyltransferase DHHC7 (encoded by ZDHHC7). Here, we investigated the consequences of S-palmitoylation of STAT3 in HCC. Increased ZDHHC7 abundance in HCC cases was associated with poor prognosis, as revealed by bioinformatics analysis of patient data. In HepG2 cells in vitro, DHHC7-mediated palmitoylation enhanced the expression of STAT3 target genes, including HIF1A, which encodes the hypoxia-inducible transcription factor HIF1α. Inhibiting DHHC7 decreased the S-palmitoylation of STAT3 and decreased HIF1α abundance. Furthermore, stabilization of HIF1α by cyclin-dependent kinase 5 (CDK5) enabled it to promote the expression of ZDHHC7, which generated a positive feedback loop between DHHC7, STAT3, and HIF1α. Perturbing this loop reduced the growth of HCC cells in vivo. Moreover, DHHC7, STAT3, and HIF1α were all abundant in human HCC tissues. Our study identifies a pathway connecting these proteins that is initiated by S-palmitoylation, which may be broadly applicable to understanding the role of this modification in cancer.
Collapse
Affiliation(s)
- Yi Jiang
- Division of Gastroenterology and Hepatology; Shanghai Institute of Digestive Disease; NHC Key Laboratory of Digestive Diseases; State Key Laboratory for Oncogenes and Related Genes; Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China
| | - Yuejie Xu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210008, China
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Chengliang Zhu
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Center for Drug Safety Evaluation and Research of Zhejiang University, Zhejiang University, Hangzhou 310058, China
| | - Guifang Xu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210008, China
| | - Lei Xu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210008, China
| | - Zijian Rao
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lixing Zhou
- The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ping Jiang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210008, China
| | - Sara Malik
- Northwestern University Feinberg School of Medicine, Chicago, 60611, IL, United States
| | - Jingyuan Fang
- Division of Gastroenterology and Hepatology; Shanghai Institute of Digestive Disease; NHC Key Laboratory of Digestive Diseases; State Key Laboratory for Oncogenes and Related Genes; Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China
| | - Hening Lin
- Howard Hughes Medical Institute; Department of Chemistry and Chemical Biology, Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, United States
| | - Mingming Zhang
- Division of Gastroenterology and Hepatology; Shanghai Institute of Digestive Disease; NHC Key Laboratory of Digestive Diseases; State Key Laboratory for Oncogenes and Related Genes; Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China
- Howard Hughes Medical Institute; Department of Chemistry and Chemical Biology, Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, United States
| |
Collapse
|
11
|
Yi K, Kong H, Zheng C, Zhuo C, Jin Y, Zhong Q, Mintz RL, Ju E, Wang H, Lv S, Lao YH, Tao Y, Li M. A LIGHTFUL nanomedicine overcomes EGFR-mediated drug resistance for enhanced tyrosine-kinase-inhibitor-based hepatocellular carcinoma therapy. Biomaterials 2023; 302:122349. [PMID: 37844429 DOI: 10.1016/j.biomaterials.2023.122349] [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: 07/02/2023] [Revised: 09/18/2023] [Accepted: 10/06/2023] [Indexed: 10/18/2023]
Abstract
Targeting the activated epidermal growth factor receptor (EGFR) via clustered regularly interspaced short palindromic repeat (CRISPR) technology is appealing to overcome the drug resistance of hepatocellular carcinoma (HCC) towards tyrosine kinase inhibitor (TKI) therapy. However, combining these two distinct drugs using traditional liposomes results in a suboptimal synergistic anti-HCC effect due to the limited CRISPR/Cas9 delivery efficiency caused by lysosomal entrapment after endocytosis. Herein, we developed a liver-targeting gene-hybridizing-TKI fusogenic liposome (LIGHTFUL) that can achieve high CRISPR/Cas9 expression to reverse the EGFR-mediated drug resistance for enhanced TKI-based HCC therapy efficiently. Coated with a galactose-modified membrane-fusogenic lipid layer, LIGHTFUL reached the targeting liver site to fuse with HCC tumor cells, directly and efficiently transporting interior CDK5- and PLK1-targeting CRISPR/Cas9 plasmids (pXG333-CPs) into the HCC cell cytoplasm and then the cell nucleus for efficient expression. Such membrane-fusion-mediated pXG333-CP delivery resulted in effective downregulation of both CDK5 and PLK1, sufficiently inactivating EGFR to improve the anti-HCC effects of the co-delivered TKI, lenvatinib. This membrane-fusion-participant codelivery strategy optimized the synergetic effect of CRISPR/Cas9 and TKI combinational therapy as indicated by the 0.35 combination index in vitro and the dramatic reduction of subcutaneous and orthotopic TKI-insensitive HCC tumor growth in mice. Therefore, the established LIGHTFUL provides a unique co-delivery platform to combine gene editing and TKI therapies for enhanced synergetic therapy.
Collapse
Affiliation(s)
- Ke Yi
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Huimin Kong
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Chunxiong Zheng
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Chenya Zhuo
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Yuanyuan Jin
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Qingguo Zhong
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Rachel L Mintz
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Enguo Ju
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Haixia Wang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Shixian Lv
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Yeh-Hsing Lao
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China; Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China; Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China.
| |
Collapse
|
12
|
Xiang J, Wu X, Liu W, Wei H, Zhu Z, Liu S, Song C, Gu Q, Wei S, Zhang Y. Bioinformatic analyzes and validation of cystathionine gamma-lyase as a prognostic biomarker and related to immune infiltrates in hepatocellular carcinoma. Heliyon 2023; 9:e16152. [PMID: 37251842 PMCID: PMC10209420 DOI: 10.1016/j.heliyon.2023.e16152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 05/05/2023] [Accepted: 05/07/2023] [Indexed: 05/31/2023] Open
Abstract
Background The role of cystathionine γ-lyase (CTH) in the prognosis and immune invasion of hepatocellular carcinoma (HCC) remains poorly understood. Methods In this study, the clinical data of patients with HCC were analyzed, and the expression level of CTH was compared between HCC and normal tissues using the R package and various databases. Results We found that CTH expression was significantly decreased in HCC compared with normal tissues, and its expression was associated with various clinicopathological factors, including tumor stage, gender, tumor status, residual tumor, histologic stage, race, alpha-fetoprotein (AFP), albumin, drinking, and smoking. Our results suggest that CTH might be a protective factor for the survival of patients with HCC. Further functional analysis revealed that high CTH expression was enriched in the Reactome signaling by interleukins and the Reactome neutrophil degranulation. Moreover, CTH expression was closely correlated with a variety of immune cells, including a negative correlation with the CD56 (bright) NK cells and follicular helper T cell (TFH), while a positive correlation with Th17 cells and central memory T cell (Tcm). High expression of CTH in immune cells predicted a better prognosis of HCC. Our findings further indicated Pyridoxal phosphate, l-cysteine, Carboxymethylthio-3-(3-chlorophenyl)-1,2,4-oxadiazol, 2-[(3-Hydroxy-2-Methyl-5-Phosphonooxymethyl-Pyridin-4-Ylmethyl)-Imino]-5-phosphono-pent-3-enoic acid and L-2-amino-3-butynoic acid as potential target candidate medications for HCC treatment based on CTH. Conclusion Our study suggests that CTH can serve as a biomarker to predict the prognosis and immune infiltration of HCC.
Collapse
Affiliation(s)
- Jianfeng Xiang
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Xinrui Wu
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Wangrui Liu
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Huagen Wei
- Dental Materials Science, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Zhu Zhu
- Medical School of Nantong University, China
| | - Shifan Liu
- Medical School of Nantong University, China
| | | | - Qiang Gu
- Affiliated Maternity and Child Health Care Hospital of Nantong University, China
| | - Shiyin Wei
- Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, China
| | - Yichi Zhang
- Department of Transplantation, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
Chiu CH. CRISPR/Cas9 genetic screens in hepatocellular carcinoma gene discovery. CURRENT RESEARCH IN BIOTECHNOLOGY 2023; 5:100127. [DOI: 10.1016/j.crbiot.2023.100127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
|
14
|
Shang B, Wang R, Qiao H, Zhao X, Wang L, Sui S. Multi-omics analysis of pyroptosis regulation patterns and characterization of tumor microenvironment in patients with hepatocellular carcinoma. PeerJ 2023; 11:e15340. [PMID: 37193028 PMCID: PMC10183172 DOI: 10.7717/peerj.15340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 04/12/2023] [Indexed: 05/18/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a primary malignant tumor of the liver, and pyroptosis has been identified as a novel cellular program that plays a role in numerous diseases including cancer. However, the functional role of pyroptosis in HCC remains unclear. The purpose of this study is to explore the relationship between the two found hub genes and provide targets for clinical treatment. Methods The Cancer Genome Atlas (TCGA) database was used to collect the gene data and clinically-related information of patients with HCC. After the differentially expressed genes (DEGs) were identified, they were intersected with the genes related to pyroptosis, and a risk prediction model was established to predict the overall survival (OS). Subsequently, drug sensitivity analysis, Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Set Enrichment Analysis (GSEA), and Gene Set Variation Analysis (GSVA) was used to analyze the biological characteristics of the DEGs. Different immune cell infiltration and related pathways were analyzed, and hub genes were identified by protein-protein interaction (PPI). Finally, the expression of hub genes was verified by real-time quantitative PCR (qRT-PCR) and immunohistochemistry. Results We conducted a comprehensive bioinformatics analysis to investigate the molecular mechanisms of pyroptosis in hepatocellular carcinoma (HCC). A total of 8,958 differentially expressed genes were identified, and 37 differentially expressed genes were associated with pyroptosis through intersection. Moreover, we developed an OS model with excellent predictive ability and discovered the differences in biological function, drug sensitivity, and immune microenvironment between high-risk and low-risk groups. Through enrichment analysis, we found that the differentially expressed genes are related to various biological processes. Then, 10 hub genes were identified from protein-protein interaction networks. Finally, midkine (MDK) was screened from the 10 hub genes and further verified by PCR and immunohistochemistry, which revealed its high expression in HCC. Conclusion We have developed a reliable and consistent predictive model based on the identification of potential hub genes, which can be used to accurately forecast the prognosis of patients, thus providing direction for further clinical research and treatment.
Collapse
Affiliation(s)
- Bingbing Shang
- Emergency Department, The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Ruohan Wang
- Emergency Department, The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Haiyan Qiao
- Research and Teaching Department of Comparative Medicine, Dalian Medical University, Dalian, China
| | - Xixi Zhao
- Emergency Department, The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Liang Wang
- Research and Teaching Department of Comparative Medicine, Dalian Medical University, Dalian, China
| | - Shaoguang Sui
- Emergency Department, The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
15
|
CRISPR/Cas9 system: a reliable and facile genome editing tool in modern biology. Mol Biol Rep 2022; 49:12133-12150. [PMID: 36030476 PMCID: PMC9420241 DOI: 10.1007/s11033-022-07880-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 08/17/2022] [Indexed: 11/10/2022]
Abstract
Genome engineering has always been a versatile technique in biological research and medicine, with several applications. In the last several years, the discovery of Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-Cas9 technology has swept the scientific community and revolutionised the speed of modern biology, heralding a new era of disease detection and rapid biotechnology discoveries. It enables successful gene editing by producing targeted double-strand breaks in virtually any organism or cell type. So, this review presents a comprehensive knowledge about the mechanism and structure of Cas9-mediated RNA-guided DNA targeting and cleavage. In addition, genome editing via CRISPR-Cas9 technology in various animals which are being used as models in scientific research including Non-Human Primates Pigs, Dogs, Zebra, fish and Drosophila has been discussed in this review. This review also aims to understand the applications, serious concerns and future perspective of CRISPR/Cas9-mediated genome editing.
Collapse
|
16
|
Vishnoi K, Ke R, Viswakarma N, Srivastava P, Kumar S, Das S, Singh SK, Principe DR, Rana A, Rana B. Ets1 mediates sorafenib resistance by regulating mitochondrial ROS pathway in hepatocellular carcinoma. Cell Death Dis 2022; 13:581. [PMID: 35789155 PMCID: PMC9253325 DOI: 10.1038/s41419-022-05022-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 01/21/2023]
Abstract
The incidence and mortality of hepatocellular carcinoma (HCC) are on a rise in the Western countries including US, attributed mostly to late detection. Sorafenib has been the first-line FDA-approved drug for advanced unresectable HCC for almost a decade, but with limited efficacy due to the development of resistance. More recently, several other multi-kinase inhibitors (lenvatinib, cabozantinib, regorafenib), human monoclonal antibody (ramucirumab), and immune checkpoint inhibitors (nivolumab, pembrolizumab) have been approved as systemic therapies. Despite this, the median survival of patients is not significantly increased. Understanding of the molecular mechanism(s) that govern HCC resistance is critically needed to increase efficacy of current drugs and to develop more efficacious ones in the future. Our studies with sorafenib-resistant (soraR) HCC cells using transcription factor RT2 Profiler PCR Arrays revealed an increase in E26 transformation-specific-1 (Ets-1) transcription factor in all soraR cells. HCC TMA studies showed an increase in Ets-1 expression in advanced HCC compared to the normal livers. Overexpression or knocking down Ets-1 modulated sorafenib resistance-related epithelial-mesenchymal transition (EMT), migration, and cell survival. In addition, the soraR cells showed a significant reduction of mitochondrial damage and mitochondrial reactive oxygen species (mROS) generation, which were antagonized by knocking down Ets-1 expression. More in-depth analysis identified GPX-2 as a downstream mediator of Ets-1-induced sorafenib resistance, which was down-regulated by Ets-1 knockdown while other antioxidant pathway genes were not affected. Interestingly, knocking down GPX2 expression significantly increased sorafenib sensitivity in the soraR cells. Our studies indicate the activation of a novel Ets-1-GPX2 signaling axis in soraR cells, targeting which might successfully antagonize resistance and increase efficacy.
Collapse
Affiliation(s)
- Kanchan Vishnoi
- grid.185648.60000 0001 2175 0319Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Rong Ke
- grid.185648.60000 0001 2175 0319Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Navin Viswakarma
- grid.185648.60000 0001 2175 0319Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Piush Srivastava
- grid.185648.60000 0001 2175 0319Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Sandeep Kumar
- grid.185648.60000 0001 2175 0319Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Subhasis Das
- grid.185648.60000 0001 2175 0319Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612 USA ,grid.185648.60000 0001 2175 0319University of Illinois Hospital and Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Sunil Kumar Singh
- grid.185648.60000 0001 2175 0319Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Daniel R. Principe
- grid.185648.60000 0001 2175 0319Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Ajay Rana
- grid.185648.60000 0001 2175 0319Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612 USA ,grid.185648.60000 0001 2175 0319University of Illinois Hospital and Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL 60612 USA ,grid.280892.90000 0004 0419 4711Jesse Brown VA Medical Center, Chicago, IL 60612 USA
| | - Basabi Rana
- grid.185648.60000 0001 2175 0319Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612 USA ,grid.185648.60000 0001 2175 0319University of Illinois Hospital and Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL 60612 USA ,grid.280892.90000 0004 0419 4711Jesse Brown VA Medical Center, Chicago, IL 60612 USA
| |
Collapse
|
17
|
Vaghari-Tabari M, Hassanpour P, Sadeghsoltani F, Malakoti F, Alemi F, Qujeq D, Asemi Z, Yousefi B. CRISPR/Cas9 gene editing: a new approach for overcoming drug resistance in cancer. Cell Mol Biol Lett 2022; 27:49. [PMID: 35715750 PMCID: PMC9204876 DOI: 10.1186/s11658-022-00348-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 05/24/2022] [Indexed: 12/18/2022] Open
Abstract
The CRISPR/Cas9 system is an RNA-based adaptive immune system in bacteria and archaea. Various studies have shown that it is possible to target a wide range of human genes and treat some human diseases, including cancers, by the CRISPR/Cas9 system. In fact, CRISPR/Cas9 gene editing is one of the most efficient genome manipulation techniques. Studies have shown that CRISPR/Cas9 technology, in addition to having the potential to be used as a new therapeutic approach in the treatment of cancers, can also be used to enhance the effectiveness of existing treatments. Undoubtedly, the issue of drug resistance is one of the main obstacles in the treatment of cancers. Cancer cells resist anticancer drugs by a variety of mechanisms, such as enhancing anticancer drugs efflux, enhancing DNA repair, enhancing stemness, and attenuating apoptosis. Mutations in some proteins of different cellular signaling pathways are associated with these events and drug resistance. Recent studies have shown that the CRISPR/Cas9 technique can be used to target important genes involved in these mechanisms, thereby increasing the effectiveness of anticancer drugs. In this review article, studies related to the applications of this technique in overcoming drug resistance in cancer cells will be reviewed. In addition, we will give a brief overview of the limitations of the CRISP/Cas9 gene-editing technique.
Collapse
Affiliation(s)
- Mostafa Vaghari-Tabari
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parisa Hassanpour
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Sadeghsoltani
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faezeh Malakoti
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Forough Alemi
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Durdi Qujeq
- Cellular and Molecular Biology Research Center (CMBRC), Health Research Institute, Babol University of Medical Sciences, Babol, Iran.,Department of Clinical Biochemistry, Babol University of Medical Sciences, Babol, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran.
| | - Bahman Yousefi
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran. .,Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
18
|
Zhou S, Ma Y, Xu R, Tang X. Nanoparticles Loaded with GSK1059615 Combined with Sorafenib Inhibited Programmed Cell Death 1 Ligand 1 Expression by Negatively Regulating the PI3K/Akt/NF- κB Pathway, Thereby Reversing the Drug Resistance of Hepatocellular Carcinoma to Sorafenib. J Biomed Nanotechnol 2022; 18:693-704. [PMID: 35715918 DOI: 10.1166/jbn.2022.3279] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Activation of the cellular signaling pathways can induce sorafenib-resistant hepatocellular carcinoma (HCCR). In this work, the PI3K/mTOR inhibitor GSK1059615 inhibited the proliferation and invasion of HCCR cells. PLGA-PEG-mal diblock copolymer was used to load GSK1059615 and sorafenib, and the vector was further modified with GPC3 antibody (hGC33) to obtain hGC33-modified GSK1059615 and sorafenib-loaded nanoparticles (Ab-G/S-NP). Ab-G/S-NP regulated the activation of cellular signaling pathways in HCCR cells by inhibiting the expression and activation of NF-κB and downregulating the level of programmed cell death 1 ligand 1(PD-L1) to reverse drug resistance of HCCR cells to sorafenib. These findings deserve further study in the combined treatment of HCCR cells with GSK1059615 in vivo to develop a more effective treatment of sorafenib-resistant cancers.
Collapse
Affiliation(s)
- Shuping Zhou
- First Affiliated Hospital, Anhui University of Science & Technology, Huainan, 232001, China
| | - Yongfang Ma
- First Affiliated Hospital, Anhui University of Science & Technology, Huainan, 232001, China
| | - Ruyue Xu
- First Affiliated Hospital, Anhui University of Science & Technology, Huainan, 232001, China
| | - Xiaolong Tang
- First Affiliated Hospital, Anhui University of Science & Technology, Huainan, 232001, China
| |
Collapse
|
19
|
Zhang Y, Zhang Y, Shi XJ, Li JX, Wang LH, Xie CE, Wang YL. Chenodeoxycholic Acid Enhances the Effect of Sorafenib in Inhibiting HepG2 Cell Growth Through EGFR/Stat3 Pathway. Front Oncol 2022; 12:836333. [PMID: 35252007 PMCID: PMC8891169 DOI: 10.3389/fonc.2022.836333] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/28/2022] [Indexed: 01/15/2023] Open
Abstract
BackgroundHepatocellular carcinoma (HCC) is a highly invasive disease with a high mortality rate. Our previous study found that Chenodeoxycholic acid (CDCA) as an endogenous metabolite can enhance the anti-tumor effect. Sorafenib has limited overall efficacy as a first-line agent in HCC, and combined with CDCA may improve its efficacy.MethodsHepG2 cells and Balb/c nude mice were used respectively for in vitro and in vivo experiments. Flow cytometry, Western blotting, HE and immunohistochemical staining and immunofluorescence were used to study the effects of CDCA combined with sorafenib on HepG2 cell growth and apoptosis-related proteins. Magnetic bead coupling, protein profiling and magnetic bead immunoprecipitation were used to find the targets of CDCA action. The effect of CDCA on EGFR/Stat3 signaling pathway was further verified by knocking down Stat3 and EGFR. Finally, fluorescence confocal, and molecular docking were used to study the binding site of CDCA to EGFR.ResultsIn this study, we found that CDCA enhanced the effect of sorafenib in inhibiting the proliferation, migration and invasion of HepG2 cells. Magnetic bead immunoprecipitation and protein profiling revealed that CDCA may enhance the effect of sorafenib by affecting the EGFR/Stat3 signaling pathway. Further results from in vitro and in vivo gene knockdown experiments, confocal experiments and molecular docking showed that CDCA enhances the efficacy of sorafenib by binding to the extracellular structural domain of EGFR.ConclusionThis study reveals the mechanism that CDCA enhances the inhibitory effect of sorafenib on HepG2 cell growth in vitro and in vivo, providing a potential new combination strategy for the treatment of HCC.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Gastroenterology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Yan Zhang
- Department of Gastroenterology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Xiao-Jun Shi
- Department of Gastroenterology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Jun-Xiang Li
- Department of Gastroenterology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Lin-Heng Wang
- Department of Gastroenterology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Chun-E Xie
- Department of Gastroenterology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yun-Liang Wang
- Department of Gastroenterology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Yun-Liang Wang,
| |
Collapse
|
20
|
Jefremow A, Neurath MF, Waldner MJ. CRISPR/Cas9 in Gastrointestinal Malignancies. Front Cell Dev Biol 2021; 9:727217. [PMID: 34912798 PMCID: PMC8667614 DOI: 10.3389/fcell.2021.727217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 10/28/2021] [Indexed: 12/27/2022] Open
Abstract
Gastrointestinal (GI) cancers such as colorectal cancer (CRC), gastric cancer (GC), esophageal cancer (EG), pancreatic duct adenocarcinoma (PDAC) or hepatocellular cancer (HCC) belong to the most commonly diagnosed types of cancer and are among the most frequent causes of cancer related death worldwide. Most types of GI cancer develop in a stepwise fashion with the occurrence of various driver mutations during tumor progression. Understanding the precise function of mutations driving GI cancer development has been regarded as a prerequisite for an improved clinical management of GI malignancies. During recent years, CRISPR/Cas9 has developed into a powerful tool for genome editing in cancer research by knocking in and knocking out even multiple genes at the same time. Within this review, we discuss recent applications for CRISPR/Cas9-based genome editing in GI cancer research including CRC, GC, EG, PDAC and HCC. These applications include functional studies of candidate genes in cancer cell lines or organoids in vitro as well as in murine cancer models in vivo, library screening for the identification of previously unknown driver mutations and even gene therapy of GI cancers.
Collapse
Affiliation(s)
- André Jefremow
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Maximilian J Waldner
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
21
|
Kong H, Ju E, Yi K, Xu W, Lao Y, Cheng D, Zhang Q, Tao Y, Li M, Ding J. Advanced Nanotheranostics of CRISPR/Cas for Viral Hepatitis and Hepatocellular Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2102051. [PMID: 34665528 PMCID: PMC8693080 DOI: 10.1002/advs.202102051] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/25/2021] [Indexed: 05/08/2023]
Abstract
Liver disease, particularly viral hepatitis and hepatocellular carcinoma (HCC), is a global healthcare burden and leads to more than 2 million deaths per year worldwide. Despite some success in diagnosis and vaccine development, there are still unmet needs to improve diagnostics and therapeutics for viral hepatitis and HCC. The emerging clustered regularly interspaced short palindromic repeat/associated proteins (CRISPR/Cas) technology may open up a unique avenue to tackle these two diseases at the genetic level in a precise manner. Especially, liver is a more accessible organ over others from the delivery point of view, and many advanced strategies applied for nanotheranostics can be adapted in CRISPR-mediated diagnostics or liver gene editing. In this review, the focus is on these two aspects of viral hepatitis and HCC applications. An overview on CRISPR editor development and current progress in clinical trials is first given, followed by highlighting the recent advances integrating the merits of gene editing and nanotheranostics. The promising systems that are used in other applications but may hold potentials in liver gene editing are also discussed. This review concludes with the perspectives on rationally designing the next-generation CRISPR approaches and improving the editing performance.
Collapse
Affiliation(s)
- Huimin Kong
- Laboratory of Biomaterials and Translational MedicineCenter for Nanomedicine and Biotherapy CenterThe Third Affiliated HospitalSun Yat‐sen University600 Tianhe RoadGuangzhou510630P. R. China
| | - Enguo Ju
- Laboratory of Biomaterials and Translational MedicineCenter for Nanomedicine and Biotherapy CenterThe Third Affiliated HospitalSun Yat‐sen University600 Tianhe RoadGuangzhou510630P. R. China
| | - Ke Yi
- Laboratory of Biomaterials and Translational MedicineCenter for Nanomedicine and Biotherapy CenterThe Third Affiliated HospitalSun Yat‐sen University600 Tianhe RoadGuangzhou510630P. R. China
| | - Weiguo Xu
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Yeh‐Hsing Lao
- Department of Biomedical EngineeringColumbia University3960 Broadway Lasker Room 450New YorkNY10032USA
| | - Du Cheng
- PCFM Lab of Ministry of EducationSchool of Materials Science and EngineeringSun Yat‐sen University135 Xingangxi RoadGuangzhou510275P. R. China
| | - Qi Zhang
- Laboratory of Biomaterials and Translational MedicineCenter for Nanomedicine and Biotherapy CenterThe Third Affiliated HospitalSun Yat‐sen University600 Tianhe RoadGuangzhou510630P. R. China
- Guangdong Provincial Key Laboratory of Liver Disease Research600 Tianhe RoadGuangzhou510630P. R. China
| | - Yu Tao
- Laboratory of Biomaterials and Translational MedicineCenter for Nanomedicine and Biotherapy CenterThe Third Affiliated HospitalSun Yat‐sen University600 Tianhe RoadGuangzhou510630P. R. China
- Guangdong Provincial Key Laboratory of Liver Disease Research600 Tianhe RoadGuangzhou510630P. R. China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational MedicineCenter for Nanomedicine and Biotherapy CenterThe Third Affiliated HospitalSun Yat‐sen University600 Tianhe RoadGuangzhou510630P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| |
Collapse
|
22
|
Fornari F, Giovannini C, Piscaglia F, Gramantieri L. Elucidating the Molecular Basis of Sorafenib Resistance in HCC: Current Findings and Future Directions. J Hepatocell Carcinoma 2021; 8:741-757. [PMID: 34239844 PMCID: PMC8260177 DOI: 10.2147/jhc.s285726] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 06/11/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related mortality worldwide. Sorafenib is the first multi-tyrosine kinase inhibitor approved for HCC and it has represented the standard of care for advanced HCC for almost 10 years, offering a survival benefit when compared to placebo. However, this benefit is limited, showing rare objective responses and a disease control rate approaching 50–60%, with most patients experiencing disease progression at 6 months. These scant results dictate the urgent need for strategies to overcome both primary and acquired resistance. Herein we report several mechanisms supporting resistance to sorafenib in HCC patients, including activation of oncogenic pathways. Among these, the AKT/mTOR pathway plays a crucial role being at the crossroad of multiple driving events. Autophagy, multidrug-resistant phenotype, hypoxia-related mechanisms and endoplasmic reticulum stress are gaining more and more relevance as crucial events driving the response to anticancer drugs, including sorafenib. Several HCC-specific miRNAs take part to the regulation of these cellular processes. Remarkably, molecularly targeted strategies able to overcome resistance in these settings have also been reported. So far, the vast majority of data has been derived from laboratory studies, which means the need for an extensive validation. Indeed, most of the possible drug associations displaying promising effects in improving sorafenib efficacy herein described derive from preclinical explorations. Notably, data obtained in animal models can be inconsistent with regard to the human disease for efficacy, safety, side effects, best formulation and pharmacokinetics. However, they represent the necessary preliminary step to improve the management of advanced HCC.
Collapse
Affiliation(s)
- Francesca Fornari
- Centre for Applied Biomedical Research - CRBA, University of Bologna, St. Orsola Hospital, Bologna, Italy.,Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - Catia Giovannini
- Centre for Applied Biomedical Research - CRBA, University of Bologna, St. Orsola Hospital, Bologna, Italy.,Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, 40138, Italy
| | - Fabio Piscaglia
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Laura Gramantieri
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
23
|
Chu H, Wu C, Zhao Q, Sun R, Yang K, Zhao B, Liu Y, Liang Z, Zhong S, Zhang L, Zhang Y. Quantitative proteomics identifies FOLR1 to drive sorafenib resistance via activating autophagy in hepatocellular carcinoma cells. Carcinogenesis 2021; 42:753-761. [PMID: 33677528 DOI: 10.1093/carcin/bgab019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/03/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022] Open
Abstract
Sorafenib is commonly used to treat advanced human hepatocellular carcinoma (HCC). However, clinical efficacy has been limited by drug resistance. In this study, we used label-free quantitative proteomic analysis to systematically investigate the underlying mechanisms of sorafenib resistance in HCC cells. A total of 1709 proteins were confidently quantified. Among them, 89 were differentially expressed and highly enriched in the processes of cell-cell adhesion, negative regulation of apoptosis, response to drug and metabolic processes involving in sorafenib resistance. Notably, folate receptor α (FOLR1) was found to be significantly upregulated in resistant HCC cells. In addition, in vitro studies showed that overexpression of FOLR1 decreased the sensitivity of HCC cells to sorafenib, whereas siRNA-directed knockdown of FOLR1 increased the sensitivity of HCC cells to sorafenib. Immunoprecipitation-mass spectrometry analysis suggested a strong link between FOLR1 and autophagy-related proteins. Further biological experiments found that FOLR1-related sorafenib resistance was accompanied by the activation of autophagy, whereas inhibition of autophagy significantly reduced FOLR1-induced cell resistance. These results suggest the driving role of FOLR1 in HCC resistance to sorafenib, which may be exerted through FOLR1-induced autophagy. Therefore, this study may provide new insights into understanding the mechanism of sorafenib resistance.
Collapse
Affiliation(s)
- Hongwei Chu
- Zhang Dayu School of Chemistry, Dalian University of Technology, Dalian, China.,CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, China
| | - Changqing Wu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, China.,Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai, China
| | - Qun Zhao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, China
| | - Rui Sun
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, China
| | - Kuo Yang
- BNRIST/Department of Automation, Tsinghua University, Beijing, China
| | - Baofeng Zhao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, China
| | - Yang Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, China.,Innovative Research Center for Integrated Cancer Omics, the Second Hospital Affiliated to China Medical University, Shenyang, China
| | - Zhen Liang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, China
| | - Shijun Zhong
- School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Lihua Zhang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, China
| | - Yukui Zhang
- Zhang Dayu School of Chemistry, Dalian University of Technology, Dalian, China.,CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, China
| |
Collapse
|
24
|
Müller M, Gerndt S, Chao YK, Zisis T, Nguyen ONP, Gerwien A, Urban N, Müller C, Gegenfurtner FA, Geisslinger F, Ortler C, Chen CC, Zahler S, Biel M, Schaefer M, Grimm C, Bracher F, Vollmar AM, Bartel K. Gene editing and synthetically accessible inhibitors reveal role for TPC2 in HCC cell proliferation and tumor growth. Cell Chem Biol 2021; 28:1119-1131.e27. [PMID: 33626324 DOI: 10.1016/j.chembiol.2021.01.023] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 01/08/2021] [Accepted: 01/28/2021] [Indexed: 01/02/2023]
Abstract
The role of two-pore channel 2 (TPC2), one of the few cation channels localized on endolysosomal membranes, in cancer remains poorly understood. Here, we report that TPC2 knockout reduces proliferation of cancer cells in vitro, affects their energy metabolism, and successfully abrogates tumor growth in vivo. Concurrently, we have developed simplified analogs of the alkaloid tetrandrine as potent TPC2 inhibitors by screening a library of synthesized benzyltetrahydroisoquinoline derivatives. Removal of dispensable substructures of the lead molecule tetrandrine increases antiproliferative properties against cancer cells and impairs proangiogenic signaling of endothelial cells to a greater extent than tetrandrine. Simultaneously, toxic effects on non-cancerous cells are reduced, allowing in vivo administration and revealing a TPC2 inhibitor with antitumor efficacy in mice. Hence, our study unveils TPC2 as valid target for cancer therapy and provides easily accessible tetrandrine analogs as a promising option for effective pharmacological interference.
Collapse
Affiliation(s)
- Martin Müller
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University Munich, 81377 Munich, Germany
| | - Susanne Gerndt
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-University Munich, 81377 Munich, Germany
| | - Yu-Kai Chao
- Walther-Straub-Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University Munich, 80336 Munich, Germany
| | - Themistoklis Zisis
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University Munich, 81377 Munich, Germany
| | - Ong Nam Phuong Nguyen
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University Munich, 81377 Munich, Germany
| | - Aaron Gerwien
- Department of Chemistry and Munich Center for Integrated Protein Science (CIPSM), Ludwig-Maximilians-University Munich, 81377 Munich, Germany
| | - Nicole Urban
- Rudolf-Boehm-Institute of Pharmacology and Toxicology, Leipzig University, 04107 Leipzig, Germany
| | - Christoph Müller
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-University Munich, 81377 Munich, Germany
| | - Florian A Gegenfurtner
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University Munich, 81377 Munich, Germany
| | - Franz Geisslinger
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University Munich, 81377 Munich, Germany
| | - Carina Ortler
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University Munich, 81377 Munich, Germany
| | - Cheng-Chang Chen
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, 100 Taipei, Taiwan
| | - Stefan Zahler
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University Munich, 81377 Munich, Germany
| | - Martin Biel
- Department of Pharmacy, Pharmacology, Ludwig-Maximilians-University Munich, 81377 Munich, Germany
| | - Michael Schaefer
- Rudolf-Boehm-Institute of Pharmacology and Toxicology, Leipzig University, 04107 Leipzig, Germany
| | - Christian Grimm
- Walther-Straub-Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University Munich, 80336 Munich, Germany.
| | - Franz Bracher
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-University Munich, 81377 Munich, Germany.
| | - Angelika M Vollmar
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University Munich, 81377 Munich, Germany.
| | - Karin Bartel
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University Munich, 81377 Munich, Germany.
| |
Collapse
|
25
|
Elevated CDK5R1 predicts worse prognosis in hepatocellular carcinoma based on TCGA data. Biosci Rep 2021; 41:227408. [PMID: 33346796 PMCID: PMC7791553 DOI: 10.1042/bsr20203594] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/15/2020] [Accepted: 12/15/2020] [Indexed: 12/13/2022] Open
Abstract
Background: Hepatocellular carcinoma (HCC) is a malignant tumor with rapid progression, high recurrence rate and poor prognosis. The objective of our investigation was to explore the prognostic value of CDK5R1 in HCC. Methods: The raw data of HCC raw data were downloaded from The Cancer Genome Atlas (TCGA) database. The Wilcoxon signed-rank test, Kruskal–Wallis test and logistic regression were applied to investigate the relevance between the CDK5R1 expression and clinicopathologic characteristics in HCC. Kaplan–Meier and Cox regression analysis were employed to examine the association between clinicopathologic features and survival. Gene set enrichment analysis (GSEA) was applied to annotate the biological function of CDK5R1. Results: CDK5R1 was highly expressed in HCC tissues. The high expression of CDK5R1 in HCC tissues was significantly associated with tumor status (P=0.00), new tumor event (P=0.00), clinical stage (P=0.00) and topography (P=0.00). Elevated CDK5R1 had significant correlation with worse overall survival (OS; P=7.414e−04), disease-specific survival (DSS; P=5.642e−04), disease-free interval (DFI; P=1.785e−05) and progression-free interval (PFI; P=2.512e−06). Besides, univariate and multivariate Cox regression analysis uncovered that increased CDK5R1 can independently predict adverse OS (P=0.037, hazard ratio [HR]= 1.7 (95% CI [1.0–2.7])), DFI (P=0.007, hazard ratio [HR]= 3.0 (95% CI [1.4–6.7])), PFI (P=0.007, hazard ratio [HR]= 2.8 (95% CI [1.3–5.9])). GSEA disclosed that notch signaling pathway and non-small cell lung cancer were prominently enriched in CDK5R1 high expression phenotype. Conclusions: Increased CDK5R1 may act as a promising independent prognostic factor of poor survival in HCC.
Collapse
|
26
|
Wu H, Wang T, Liu Y, Li X, Xu S, Wu C, Zou H, Cao M, Jin G, Lang J, Wang B, Liu B, Luo X, Xu C. Mitophagy promotes sorafenib resistance through hypoxia-inducible ATAD3A dependent Axis. J Exp Clin Cancer Res 2020; 39:274. [PMID: 33280610 PMCID: PMC7720487 DOI: 10.1186/s13046-020-01768-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 11/05/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The identification of novel targets for recovering sorafenib resistance is pivotal for Hepatocellular carcinoma (HCC) patients. Mitophagy is the programmed degradation of mitochondria, and is likely involved in drug resistance of cancer cells. Here, we identified hyperactivated mitophagy is essential for sorafenib resistance, and the mitophagy core regulator gene ATAD3A (ATPase family AAA domain containing 3A) was down regulated in hypoxia induced resistant HCC cells. Blocking mitophagy may restore the sorafenib sensitivity of these cells and provide a new treatment strategy for HCC patients. METHODS Hypoxia induced sorafenib resistant cancer cells were established by culturing under 1% O2 with increasing drug treatment. RNA sequencing was conducted in transfecting LM3 cells with sh-ATAD3A lentivirus. Subsequent mechanistic studies were performed in HCC cell lines by manipulating ATAD3A expression isogenically where we evaluated drug sensitivity, molecular signaling events. In vivo study, we investigated the combined treatment effect of sorafenib and miR-210-5P antagomir. RESULTS We found a hyperactivated mitophagy regulating by ATAD3A-PINK1/PARKIN axis in hypoxia induced sorafenib resistant HCC cells. Gain- and loss- of ATAD3A were related to hypoxia-induced mitophagy and sorafenib resistance. In addition, ATAD3A is a functional target of miR-210-5p and its oncogenic functions are likely mediated by increased miR-210-5P expression. miR-210-5P was upregulated under hypoxia and participated in regulating sorafenib resistance. In vivo xenograft assay showed that miR-210-5P antagomir combined with sorafenib abrogated the tumorigenic effect of ATAD3A down-regulation in mice. CONCLUSIONS Loss of ATAD3A hyperactivates mitophagy which is a core event in hypoxia induced sorafenib resistance in HCC cells. Targeting miR-210-5P-ATAD3A axis is a novel therapeutic target for sorafenib-resistant HCC.
Collapse
Affiliation(s)
- Hong Wu
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, 518055, Shenzhen, China
- Integrative Cancer Center&Cancer Clinical Research Center, Sichuan Cancer Hospital & Institute Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610000, P. R. China
- Department of Experimental Research, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 510000, P. R. China
| | - Tao Wang
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, P. R. China
| | - Yiqiang Liu
- Integrative Cancer Center&Cancer Clinical Research Center, Sichuan Cancer Hospital & Institute Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610000, P. R. China
- Department of Experimental Research, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 510000, P. R. China
| | - Xin Li
- Department of Experimental Research, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 510000, P. R. China
| | - Senlin Xu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital and Key Laboratory of Tumor Immunopathology, Army Medical University (Third Military Medical University), Chongqing, 400042, P. R. China
| | - Changtao Wu
- Integrative Cancer Center&Cancer Clinical Research Center, Sichuan Cancer Hospital & Institute Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610000, P. R. China
- Department of Experimental Research, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 510000, P. R. China
| | - Hongbo Zou
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital and Key Laboratory of Tumor Immunopathology, Army Medical University (Third Military Medical University), Chongqing, 400042, P. R. China
| | - Mianfu Cao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital and Key Laboratory of Tumor Immunopathology, Army Medical University (Third Military Medical University), Chongqing, 400042, P. R. China
| | - Guoxiang Jin
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital and Key Laboratory of Tumor Immunopathology, Army Medical University (Third Military Medical University), Chongqing, 400042, P. R. China
| | - Jinyi Lang
- Integrative Cancer Center&Cancer Clinical Research Center, Sichuan Cancer Hospital & Institute Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610000, P. R. China
| | - Bin Wang
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, P. R. China
| | - Baohua Liu
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, 518055, Shenzhen, China.
| | - Xiaolin Luo
- Department of Experimental Research, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 510000, P. R. China.
| | - Chuan Xu
- Integrative Cancer Center&Cancer Clinical Research Center, Sichuan Cancer Hospital & Institute Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610000, P. R. China.
| |
Collapse
|
27
|
Zeng Y, Liu Q, Wang Y, Tian C, Yang Q, Zhao Y, Liu L, Wu G, Xu S. CDK5 Activates Hippo Signaling to Confer Resistance to Radiation Therapy Via Upregulating TAZ in Lung Cancer. Int J Radiat Oncol Biol Phys 2020; 108:758-769. [PMID: 32407930 DOI: 10.1016/j.ijrobp.2020.05.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/17/2020] [Accepted: 05/01/2020] [Indexed: 12/15/2022]
Abstract
PURPOSE Tumor resistance to radiation therapy is a therapeutic challenge in the treatment of patients with non-small cell lung cancer. Cyclin-dependent kinase 5 (CDK5) has been proposed to participate in cell proliferation, migration and invasion, drug resistance, and immune evasion. However, the functions and regulatory mechanisms of CDK5 in lung cancer radioresistance have not been investigated. METHODS AND MATERIALS DNA damage response and repair were measured by neutral comet assay and γ-H2AX and Rad51 foci staining. The biological functions of CDK5 in lung cancer radioresistance were investigated with clonogenic survival assays and xenograft tumor models. Small interfering RNAs and short hairpin RNAs were used to knock down CDK5 in A549 and H1299 cells. The effects of CDK5 depletion on the tumorigenic behaviors of lung cancer cells were evaluated in vitro and in vivo. Gene expression was examined by RNA-seq and quantitative real-time polymerase chain reaction. RESULTS We report that CDK5 depletion impairs lung cancer progression and radioresistance in vitro and in vivo. Mechanistically, we identify TAZ, a component of the Hippo pathway, as a critical downstream effector of CDK5. Loss of CDK5 downregulates TAZ expression and attenuates Hippo signaling activation. Importantly, we provide evidence that TAZ is the major effector mediating the biological functions of CDK5 in lung cancer. CONCLUSIONS These results illustrate that CDK5 activates Hippo signaling via TAZ to participate in tumorigenesis and radioresistance, suggesting that CDK5 may be a promising radiosensitization target for the treatment of lung cancer.
Collapse
Affiliation(s)
- Yulan Zeng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Quan Liu
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ye Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Tian
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qifan Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ye Zhao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Shuangbing Xu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
28
|
Li M, Wei L, Liu PY, Zhang XM, Liu F, Yang F, Hu XS, Mo ZC. Lnc-ATG9B-4 aggravates progress of hepatocellular carcinoma through cell proliferation and migration by upregulating CDK5. Exp Biol Med (Maywood) 2020; 246:177-186. [PMID: 33023330 DOI: 10.1177/1535370220963197] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Long noncoding RNAs play an important role in the occurrence, invasion, as well as metastasis of various human cancers, including hepatocellular carcinoma. Long noncoding RNAs can affect the biological functions of hepatocellular carcinoma cells by regulating various genes; however, only a small fraction of molecular mechanisms of long noncoding RNAs have been elucidated. In the present study, lnc AC010973.1 (lnc-ATG9B-4) was first identified by microarray analysis from 8 patients with hepatocellular carcinoma and confirmed by quantitative PCR in 176 patients with hepatocellular carcinoma. We demonstrated that lnc-ATG9B-4 was tightly relative to the tumorous size, TNM stages, portal vein tumor thrombus (PVTT), the tumor capsule, metastasis, degree of differentiation, and poor prognosis of hepatocellular carcinoma according to long-term follow-up data. In hepatocellular carcinoma cells, overexpression of lnc-ATG9B-4 promoted proliferation, invasion, as well as migration, while inhibiting lnc-ATG9B-4 by siRNA significantly attenuated the proliferation, invasion, as well as migration. Interestingly, lnc-ATG9B-4 increased the expression of cyclin-dependent kinase 5 (CDK5), which was closely related to the development and chemotherapy sensitivity of hepatocellular carcinoma. In summary, our results revealed that lnc-ATG9B-4 suggests an unfavorable prognosis of hepatocellular carcinoma and facilitates the proliferation, invasion, as well as migration of hepatocellular carcinoma cells by upregulating CDK5. This research suggests that lnc-ATG9B-4 may be a new biomarker for predicting the prognosis of hepatocellular carcinoma; meanwhile, targeting lnc-ATG9B-4 might serve as a potential strategy for the treatment hepatocellular carcinoma.
Collapse
Affiliation(s)
- Ming Li
- Department of Histology and Embryology, Hunan Province Key Laboratory for Antibody-based Drug and Intelligent Delivery System, Hunan University of Medicine, Hunan 418000, China.,Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Guilin Medical University, Guilin 541001, China
| | - Le Wei
- Department of Histology and Embryology, Hunan Province Key Laboratory for Antibody-based Drug and Intelligent Delivery System, Hunan University of Medicine, Hunan 418000, China
| | - Pin-Yue Liu
- Department of Histology and Embryology, Hunan Province Key Laboratory for Antibody-based Drug and Intelligent Delivery System, Hunan University of Medicine, Hunan 418000, China
| | - Xue-Mei Zhang
- Department of Histology and Embryology, Hunan Province Key Laboratory for Antibody-based Drug and Intelligent Delivery System, Hunan University of Medicine, Hunan 418000, China
| | - Fang Liu
- Department of Histology and Embryology, Hunan Province Key Laboratory for Antibody-based Drug and Intelligent Delivery System, Hunan University of Medicine, Hunan 418000, China
| | - Fen Yang
- The First People's Hospital of Huaihua, Huaihua 418000, China
| | - Xiang-Shang Hu
- Department of Human Anatomy, Hunan University of Medicine, Huaihua418000, China
| | - Zhong-Cheng Mo
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Guilin Medical University, Guilin 541001, China
| |
Collapse
|
29
|
Pavlovic K, Tristán-Manzano M, Maldonado-Pérez N, Cortijo-Gutierrez M, Sánchez-Hernández S, Justicia-Lirio P, Carmona MD, Herrera C, Martin F, Benabdellah K. Using Gene Editing Approaches to Fine-Tune the Immune System. Front Immunol 2020; 11:570672. [PMID: 33117361 PMCID: PMC7553077 DOI: 10.3389/fimmu.2020.570672] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 08/20/2020] [Indexed: 12/26/2022] Open
Abstract
Genome editing technologies not only provide unprecedented opportunities to study basic cellular system functionality but also improve the outcomes of several clinical applications. In this review, we analyze various gene editing techniques used to fine-tune immune systems from a basic research and clinical perspective. We discuss recent advances in the development of programmable nucleases, such as zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and clustered regularly interspaced short palindromic repeat (CRISPR)-Cas-associated nucleases. We also discuss the use of programmable nucleases and their derivative reagents such as base editing tools to engineer immune cells via gene disruption, insertion, and rewriting of T cells and other immune components, such natural killers (NKs) and hematopoietic stem and progenitor cells (HSPCs). In addition, with regard to chimeric antigen receptors (CARs), we describe how different gene editing tools enable healthy donor cells to be used in CAR T therapy instead of autologous cells without risking graft-versus-host disease or rejection, leading to reduced adoptive cell therapy costs and instant treatment availability for patients. We pay particular attention to the delivery of therapeutic transgenes, such as CARs, to endogenous loci which prevents collateral damage and increases therapeutic effectiveness. Finally, we review creative innovations, including immune system repurposing, that facilitate safe and efficient genome surgery within the framework of clinical cancer immunotherapies.
Collapse
Affiliation(s)
- Kristina Pavlovic
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada (Andalusian Regional Government), Health Sciences Technology Park, Granada, Spain
- Maimonides Institute of Biomedical Research in Cordoba (IMIBIC), Cellular Therapy Unit, Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - María Tristán-Manzano
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada (Andalusian Regional Government), Health Sciences Technology Park, Granada, Spain
| | - Noelia Maldonado-Pérez
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada (Andalusian Regional Government), Health Sciences Technology Park, Granada, Spain
| | - Marina Cortijo-Gutierrez
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada (Andalusian Regional Government), Health Sciences Technology Park, Granada, Spain
| | - Sabina Sánchez-Hernández
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada (Andalusian Regional Government), Health Sciences Technology Park, Granada, Spain
| | - Pedro Justicia-Lirio
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada (Andalusian Regional Government), Health Sciences Technology Park, Granada, Spain
- LentiStem Biotech, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada (Andalusian Regional Government), Health Sciences Technology Park, Granada, Spain
| | - M. Dolores Carmona
- Maimonides Institute of Biomedical Research in Cordoba (IMIBIC), Cellular Therapy Unit, Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - Concha Herrera
- Maimonides Institute of Biomedical Research in Cordoba (IMIBIC), Cellular Therapy Unit, Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
- Department of Hematology, Reina Sofía University Hospital, Córdoba, Spain
| | - Francisco Martin
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada (Andalusian Regional Government), Health Sciences Technology Park, Granada, Spain
| | - Karim Benabdellah
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada (Andalusian Regional Government), Health Sciences Technology Park, Granada, Spain
| |
Collapse
|
30
|
Schütz R, Müller M, Geisslinger F, Vollmar A, Bartel K, Bracher F. Synthesis, biological evaluation and toxicity of novel tetrandrine analogues. Eur J Med Chem 2020; 207:112810. [PMID: 32942071 PMCID: PMC7473156 DOI: 10.1016/j.ejmech.2020.112810] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/22/2020] [Accepted: 08/31/2020] [Indexed: 11/16/2022]
Abstract
In this work, we present the design and synthesis of novel fully synthetic analogues of the bisbenzylisoquinoline tetrandrine, a molecule with numerous pharmacological properties and the potential to treat life-threatening diseases, such as viral infections and cancer. Its toxicity to liver and lungs and the underlying mechanisms, however, are controversially discussed. Along this line, novel tetrandrine analogues were synthesized and biologically evaluated for their hepatotoxicity, as well as their antiproliferative and chemoresistance reversing activity on cancer cells. Previous studies suggesting CYP-mediated toxification of tetrandrine prompted us to amend/replace the suspected metabolically instable 12-methoxy group. Of note, employing several in vitro models showed that the proposed CYP3A4-driven metabolism of tetrandrine and analogues is not the major cause of hepatotoxicity. Biological characterization revealed that some of the novel tetrandrine analogues sensitized drug-resistant leukemia cells by inhibition of the P-glycoprotein. Interestingly, direct anticancer effects improved in comparison to tetrandrine, as several compounds displayed a markedly enhanced ability to reduce proliferation of drug-resistant leukemia cells and to induce cell death of liver cancer cells. Those enhanced anticancer properties were linked to influences on activation of the kinase Akt and mitochondrial events. In sum, our study clarifies the role of CYP3A4-mediated toxicity of the bisbenzylisoquinoline alkaloid tetrandrine and provides the basis for the exploitation of novel synthetic analogues for their antitumoral potential.
Collapse
Affiliation(s)
- Ramona Schütz
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-University of Munich, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Martin Müller
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-University of Munich, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Franz Geisslinger
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-University of Munich, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Angelika Vollmar
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-University of Munich, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Karin Bartel
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-University of Munich, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Franz Bracher
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-University of Munich, Butenandtstr. 5-13, 81377, Munich, Germany.
| |
Collapse
|
31
|
Peng X, Yang L, Ma Y, Li Y, Li H. Focus on the morphogenesis, fate and the role in tumor progression of multivesicular bodies. Cell Commun Signal 2020; 18:122. [PMID: 32771015 PMCID: PMC7414566 DOI: 10.1186/s12964-020-00619-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/27/2020] [Indexed: 12/11/2022] Open
Abstract
Multivesicular bodies (MVBs) are endosome organelles that are gradually attracting research attention. Initially, MVBs were considered as important components of the endosomal-lysosomal degradation pathway. In recent years, with an increase in extracellular vesicle (EV) research, the biogenesis, fate, and pathological effects of MVBs have been increasingly studied. However, the mechanisms by which MVBs are sorted to the lysosome and plasma membrane remain unclear. In addition, whether the trafficking of MVBs can determine whether exosomes are released from cells, the factors are involved in cargo loading and regulating the fate of MVBs, and the roles that MVBs play in the development of disease are unknown. Consequently, this review focuses on the mechanism of MVB biogenesis, intraluminal vesicle formation, sorting of different cargoes, and regulation of their fate. We also discuss the mechanisms of emerging amphisome-dependent secretion and degradation. In addition, we highlight the contributions of MVBs to the heterogeneity of EVs, and their important roles in cancer. Thus, we attempt to unravel the various functions of MVBs in the cell and their multiple roles in tumor progression. Video Abstract
Collapse
Affiliation(s)
- Xueqiang Peng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Liang Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Yingbo Ma
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Yan Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Hangyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
| |
Collapse
|
32
|
Shao QP, Wei C, Yang J, Zhang WZ. miR-3609 Decelerates the Clearance of Sorafenib in Hepatocellular Carcinoma Cells by Targeting EPAS-1 and Reducing the Activation of the Pregnane X Receptor Pathway. Onco Targets Ther 2020; 13:7213-7227. [PMID: 32801751 PMCID: PMC7394586 DOI: 10.2147/ott.s246471] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 07/02/2020] [Indexed: 12/11/2022] Open
Abstract
Background The pregnane X receptor (PXR) not only plays an important role in cellular metabolism processes but also induces the resistance of hepatocellular carcinoma (HCC) cells to molecularly targeted drugs by mediating their metabolism and clearance by these cells. Endothelial PAS domain-containing protein 1 (EPAS-1) acts as a coactivator to regulate the transcription factor activity of PXR. In the present study, a microRNA that potentially targets EPAS-1, namely miR-3609, was identified using the miRDB tool. Methods The expression of miR-3609 and EPAS-1 was examined by qPCR. Lentiviral particles containing the full-length sequences of miR-3609 (pri-miR-3609) were prepared. The antitumor effect of antitumor agents was examined by the in vitro and in vivo assays. Results The expression of miR-3609 was negatively correlated with that of EPAS-1 in both HCC clinical specimens and paired non-tumor specimens, and the effect of miR-3609 on the expression of EPAS-1 was confirmed by Western blot experiments. Overexpression of miR-3609 decreased the expression of EPAS-1 and, in turn, repressed the activation of the PXR pathway. miR-3609 decreased the transcription factor activation of PXR, repressed its recruitment to its target gene promoter regions, and decreased the expression of its target genes CYP3A4 and P-GP. In addition, miR-3609 decelerated the metabolism and clearance of sorafenib in HCC cells and enhanced the antitumor effect of sorafenib in HCC cells. Conclusion Therefore, the results indicate that miR-3609 decreases the expression of EPAS-1 and enhances the sensitivity of HCC cells to sorafenib.
Collapse
Affiliation(s)
- Qing-Ping Shao
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan Province 450008, People's Republic of China
| | - Chen Wei
- Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450052, People's Republic of China
| | - Jie Yang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450052, People's Republic of China
| | - Wen-Zhou Zhang
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan Province 450008, People's Republic of China
| |
Collapse
|
33
|
Lin Z, Ni X, Dai S, Chen H, Chen J, Wu B, Ao J, Shi K, Sun H. Screening and verification of long noncoding RNA promoter methylation sites in hepatocellular carcinoma. Cancer Cell Int 2020; 20:311. [PMID: 32684848 PMCID: PMC7362420 DOI: 10.1186/s12935-020-01407-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 07/07/2020] [Indexed: 12/20/2022] Open
Abstract
Background Long noncoding ribonucleic acid (lncRNA) promoter methylation is closely related to the occurrence and development of hepatocellular carcinoma (HCC). Thus, we aim to screen and verify the lncRNA promoter methylation sites associated with overall survival (OS), vascular invasion, pathological grade, and clinical stage in HCC. Methods Methylation-related data including clinical characteristic, transcriptome, methylation, and messenger RNA (mRNA) expression were taken from the Cancer Genome Atlas (TCGA) database. The OS, vascular invasion, pathological grade, and clinical stage-related lncRNA promoter methylation models were developed by the least absolute shrinkage and selection operator (LASSO) algorithm based on the lncRNA promoter methylation sites screened via R software. The Kaplan-Meier analysis, the area under the receiver operating characteristic (ROC) curve (AUC), the calibration curve (C-index) were performed to evaluate the performance of these models. Finally, the methylation-specific polymerase chain reaction (MS-PCR) was performed to verify the accuracy of these models based on 146 HCC tissues from our hospital. Results A total of 10 methylation sites were included in the OS-related lncRNA promoter methylation model that could effectively divide HCC patients into high-risk and low-risk groups (P < 0.0001) via survival analysis. COX univariable and multivariable regression analysis found that the OS-related model (P < 0.001, 95% CI 1.378-2.942) and T stage (P < 0.001, 95% CI 1.490-3.418) were independent risk factors affecting OS in HCC patients. The vascular invasion-related model contained 8 methylation sites with its AUC value of 0.657; the pathological grade-related model contained 22 methylation sites with its AUC value of 0.797; the clinical stage-related model contained 13 methylation sites with its AUC of 0.724. Target genes corresponded to vascular invasion-related lncRNA promoter methylation sites were involved in many kinds of biological processes in HCC such as PI3K-Akt signaling pathway. The accuracy of the vascular invasion-related model was consistent with our bioinformatics conclusion after being verified via MS-PCR. Conclusion The lncRNA promoter methylation sites are closely correlated with the process of HCC and can be utilized to improve the therapy and prognosis of HCC.
Collapse
Affiliation(s)
- Zhuo Lin
- Department of Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province People's Republic of China
| | - Xiaofeng Ni
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province People's Republic of China
| | - Shengjie Dai
- Department of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province People's Republic of China
| | - Hao Chen
- Department of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province People's Republic of China
| | - Jianhui Chen
- Department of Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province People's Republic of China.,Chinese Academy of Sciences Shanghai Branch, Shanghai, People's Republic of China
| | - Boda Wu
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province People's Republic of China
| | - Jianyang Ao
- Department of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province People's Republic of China
| | - Keqing Shi
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province People's Republic of China.,Laboratory of Precision Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province People's Republic of China
| | - Hongwei Sun
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province People's Republic of China.,Department of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province People's Republic of China
| |
Collapse
|
34
|
Saber A, Liu B, Ebrahimi P, Haisma HJ. CRISPR/Cas9 for overcoming drug resistance in solid tumors. Daru 2020; 28:295-304. [PMID: 30666557 PMCID: PMC7214581 DOI: 10.1007/s40199-019-00240-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/04/2019] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVES In this review, we focus on the application of clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR associated nuclease 9 (Cas9), as a powerful genome editing system, in the identification of resistance mechanisms and in overcoming drug resistance in the most frequent solid tumors. DATA ACQUISITION Data were collected by conducting systematic searching of scientific English literature using specific keywords such as "cancer", "CRISPR" and related combinations. RESULTS The review findings revealed the importance of CRISPR/Cas9 system in understanding drug resistance mechanisms and identification of resistance-related genes such as PBRM1, SLFN11 and ATPE1 in different cancers. We also provided an overview of genes, including RSF1, CDK5, and SGOL1, whose disruption can synergize with the currently available drugs such as paclitaxel and sorafenib. CONCLUSION The data suggest CRISPR/Cas9 system as a useful tool in elucidating the molecular basis of drug resistance and improving clinical outcomes. Graphical abstract The mechanisms of CRISPR/Cas9-mediated genome editing and double-strand breaks (DSBs) repair.
Collapse
Affiliation(s)
- Ali Saber
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Bin Liu
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Pirooz Ebrahimi
- Universal Scientific Education and Research Network, Tehran, Iran
- Parseh Medical Genetics Clinic, Tehran, Iran
| | - Hidde J Haisma
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
35
|
Lin Z, Niu Y, Wan A, Chen D, Liang H, Chen X, Sun L, Zhan S, Chen L, Cheng C, Zhang X, Bu X, He W, Wan G. RNA m 6 A methylation regulates sorafenib resistance in liver cancer through FOXO3-mediated autophagy. EMBO J 2020; 39:e103181. [PMID: 32368828 PMCID: PMC7298296 DOI: 10.15252/embj.2019103181] [Citation(s) in RCA: 324] [Impact Index Per Article: 64.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 03/25/2020] [Accepted: 04/03/2020] [Indexed: 12/31/2022] Open
Abstract
N6‐methyladenosine (m6A) is an abundant nucleotide modification in mRNA, known to regulate mRNA stability, splicing, and translation, but it is unclear whether it is also has a physiological role in the intratumoral microenvironment and cancer drug resistance. Here, we find that METTL3, a primary m6A methyltransferase, is significantly down‐regulated in human sorafenib‐resistant hepatocellular carcinoma (HCC). Depletion of METTL3 under hypoxia promotes sorafenib resistance and expression of angiogenesis genes in cultured HCC cells and activates autophagy‐associated pathways. Mechanistically, we have identified FOXO3 as a key downstream target of METTL3, with m6A modification of the FOXO3 mRNA 3′‐untranslated region increasing its stability through a YTHDF1‐dependent mechanism. Analysis of clinical samples furthermore showed that METTL3 and FOXO3 levels are tightly correlated in HCC patients. In mouse xenograft models, METTL3 depletion significantly enhances sorafenib resistance of HCC by abolishing the identified METTL3‐mediated FOXO3 mRNA stabilization, and overexpression of FOXO3 restores m6A‐dependent sorafenib sensitivity. Collectively, our work reveals a critical function for METTL3‐mediated m6A modification in the hypoxic tumor microenvironment and identifies FOXO3 as an important target of m6A modification in the resistance of HCC to sorafenib therapy.
Collapse
Affiliation(s)
- Ziyou Lin
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.,National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, National Engineering Research Center for New Drug and Druggability (cultivation), Guangdong Province Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Yi Niu
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, National Engineering Research Center for New Drug and Druggability (cultivation), Guangdong Province Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Arabella Wan
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Dongshi Chen
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Heng Liang
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, National Engineering Research Center for New Drug and Druggability (cultivation), Guangdong Province Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Xijun Chen
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Lei Sun
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, National Engineering Research Center for New Drug and Druggability (cultivation), Guangdong Province Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Siyue Zhan
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, National Engineering Research Center for New Drug and Druggability (cultivation), Guangdong Province Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Liutao Chen
- School of Life Science, Sun Yat-Sen University, Guangzhou, China
| | - Chao Cheng
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaolei Zhang
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, National Engineering Research Center for New Drug and Druggability (cultivation), Guangdong Province Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Xianzhang Bu
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, National Engineering Research Center for New Drug and Druggability (cultivation), Guangdong Province Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Weiling He
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.,National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, National Engineering Research Center for New Drug and Druggability (cultivation), Guangdong Province Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China.,Center for Precision Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Guohui Wan
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.,National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, National Engineering Research Center for New Drug and Druggability (cultivation), Guangdong Province Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
36
|
Cheng X, Fan S, Wen C, Du X. CRISPR/Cas9 for cancer treatment: technology, clinical applications and challenges. Brief Funct Genomics 2020; 19:209-214. [PMID: 32052006 DOI: 10.1093/bfgp/elaa001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/08/2019] [Accepted: 01/06/2020] [Indexed: 12/25/2022] Open
Abstract
AbstractClustered regularly interspaced short palindromic repeats (CRISPR) is described as RNA mediated adaptive immune system defense, which is naturally found in bacteria and archaea. CRISPR-Cas9 has shown great promise for cancer treatment in cancer immunotherapy, manipulation of cancer genome and epigenome and elimination or inactivation of carcinogenic viral infections. However, many challenges remain to be addressed to increase its efficacy, including off-target effects, editing efficiency, fitness of edited cells, immune response and delivery methods. Here, we explain CRISPR-Cas classification and its general function mechanism for gene editing. Then, we summarize these preclinical CRISPR-Cas9-based therapeutic strategies against cancer. Moreover, the challenges and improvements of CRISPR-Cas9 clinical applications will be discussed.
Collapse
Affiliation(s)
- Xing Cheng
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Shaoyi Fan
- Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine,Guangzhou, China
| | - Chengcai Wen
- Department of Rehabilitation, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| | - Xianfa Du
- Department of Orthopaedics, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
37
|
Cervello M, Emma MR, Augello G, Cusimano A, Giannitrapani L, Soresi M, Akula SM, Abrams SL, Steelman LS, Gulino A, Belmonte B, Montalto G, McCubrey JA. New landscapes and horizons in hepatocellular carcinoma therapy. Aging (Albany NY) 2020; 12:3053-3094. [PMID: 32018226 PMCID: PMC7041742 DOI: 10.18632/aging.102777] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 01/12/2020] [Indexed: 04/12/2023]
Abstract
Hepatocellular carcinoma (HCC), is the sixth most frequent form of cancer and leads to the fourth highest number of deaths each year. HCC results from a combination of environmental factors and aging as there are driver mutations at oncogenes which occur during aging. Most of HCCs are diagnosed at advanced stage preventing curative therapies. Treatment in advanced stage is a challenging and pressing problem, and novel and well-tolerated therapies are urgently needed. We will discuss further advances beyond sorafenib that target additional signaling pathways and immune checkpoint proteins. The scenario of possible systemic therapies for patients with advanced HCC has changed dramatically in recent years. Personalized genomics and various other omics approaches may identify actionable biochemical targets, which are activated in individual patients, which may enhance therapeutic outcomes. Further studies are needed to identify predictive biomarkers and aberrantly activated signaling pathways capable of guiding the clinician in choosing the most appropriate therapy for the individual patient.
Collapse
Affiliation(s)
- Melchiorre Cervello
- Institute for Biomedical Research and Innovation, National Research Council (CNR), Palermo, Italy
| | - Maria R. Emma
- Institute for Biomedical Research and Innovation, National Research Council (CNR), Palermo, Italy
| | - Giuseppa Augello
- Institute for Biomedical Research and Innovation, National Research Council (CNR), Palermo, Italy
| | - Antonella Cusimano
- Institute for Biomedical Research and Innovation, National Research Council (CNR), Palermo, Italy
| | - Lydia Giannitrapani
- Institute for Biomedical Research and Innovation, National Research Council (CNR), Palermo, Italy
- Department of Health Promotion Sciences Maternal and Infantile Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Maurizio Soresi
- Department of Health Promotion Sciences Maternal and Infantile Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Shaw M. Akula
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC 27858, USA
| | - Stephen L. Abrams
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC 27858, USA
| | - Linda S. Steelman
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC 27858, USA
| | - Alessandro Gulino
- Tumour Immunology Unit, Human Pathology Section, Department of Health Science, University of Palermo, Palermo, Italy
| | - Beatrice Belmonte
- Tumour Immunology Unit, Human Pathology Section, Department of Health Science, University of Palermo, Palermo, Italy
| | - Giuseppe Montalto
- Institute for Biomedical Research and Innovation, National Research Council (CNR), Palermo, Italy
- Department of Health Promotion Sciences Maternal and Infantile Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - James A. McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC 27858, USA
| |
Collapse
|
38
|
Sharma S, Sicinski P. A kinase of many talents: non-neuronal functions of CDK5 in development and disease. Open Biol 2020; 10:190287. [PMID: 31910742 PMCID: PMC7014686 DOI: 10.1098/rsob.190287] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The cyclin-dependent kinase 5 (CDK5) represents an unusual member of the family of cyclin-dependent kinases, which is activated upon binding to non-cyclin p35 and p39 proteins. The role of CDK5 in the nervous system has been very well established. In addition, there is growing evidence that CDK5 is also active in non-neuronal tissues, where it has been postulated to affect a variety of functions such as the immune response, angiogenesis, myogenesis, melanogenesis and regulation of insulin levels. Moreover, high levels of CDK5 have been observed in different tumour types, and CDK5 was proposed to play various roles in the tumorigenic process. In this review, we discuss these various CDK5 functions in normal physiology and disease, and highlight the therapeutic potential of targeting CDK5.
Collapse
Affiliation(s)
- Samanta Sharma
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Piotr Sicinski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
39
|
PES1 promotes BET inhibitors resistance and cells proliferation through increasing c-Myc expression in pancreatic cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:463. [PMID: 31718704 PMCID: PMC6852745 DOI: 10.1186/s13046-019-1466-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 10/22/2019] [Indexed: 12/20/2022]
Abstract
Background Overexpressed PES1 promotes carcinogenesis in various types of malignant tumors. However, the biological role and clinical significance of PES1 in pancreatic cancer are still unexplored. Methods The expression level of PES1 in pancreatic cancer cell lines and pancreatic cancer patient samples was determined using Western Blotting analysis, RT-qPCR analysis, immunohistochemical (IHC) analysis of tissue microarray, and the GEPIA web tool. MTS assay, colony formation assay, and xenograft tumor assay were used to evaluate the tumor growth ability of pancreatic cancer cells. Results We established that the expression of PES1 was abnormally increased in pancreatic cancer tissues and led to poor prognosis of pancreatic cancer patients. We also found that PES1 was responsible for promoting cell growth and contributed to bromodomain and cancer cell resistance to extra-terminal (BET) inhibitors in pancreatic cancer. Furthermore, we showed that PES1 interacted with BRD4 to enhance c-Myc expression, which is the primary cause of cancer cell resistance to BET inhibitors in pancreatic cancer. Finally, CDK5 inhibitors were proven to destabilize PES1 and overcome cancer cell resistance to BET inhibitors in pancreatic cancer cells. Conclusions We have shown that PES1 could be one of the promoting factors of tumor growth and a prognosis-related protein of pancreatic cancer. Targeting PES1 with CDK5 inhibitors might help overcome cancer cell resistance to BET inhibitors in pancreatic cancer cells.
Collapse
|
40
|
Shao YY, Li YS, Hsu HW, Lin H, Wang HY, Wo RR, Cheng AL, Hsu CH. Potent Activity of Composite Cyclin Dependent Kinase Inhibition against Hepatocellular Carcinoma. Cancers (Basel) 2019; 11:cancers11101433. [PMID: 31561409 PMCID: PMC6827105 DOI: 10.3390/cancers11101433] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 09/19/2019] [Indexed: 12/18/2022] Open
Abstract
Alterations in cell cycle regulators are common in hepatocellular carcinoma (HCC). We tested the efficacy of composite inhibition of CDKs 1, 2, 5, and 9 through dinaciclib on HCC. In vitro, dinaciclib exhibited potent antiproliferative activities in HCC cell lines regardless of Rb or c-myc expression levels. Dinaciclib significantly downregulated the phosphorylation of Rb (target of CDKs 1 and 2), ataxia telangiectasia mutated kinase (target of CDK5), and RNA polymerase II (target of CDK9) in the HCC cells. In xenograft studies, mice receiving dinaciclib tolerated the treatment well without significant body weight changes and exhibited a significantly slower tumor growth rate than the mice receiving vehicles. RNA interference (RNAi) of CDKs 1 and 9 was more effective in inhibiting the cell proliferation of HCC cells than RNAi of CDKs 2 and 5. Overexpression of CDK9 significantly reduced the efficacy of dinaciclib in HCC cells, but overexpression of CDK1 did not. In conclusion, composite inhibition of CDKs 1, 2, 5, and 9 through dinaciclib exhibited potent in vitro and in vivo activity against HCC. CDK9 inhibition might be the crucial mechanism.
Collapse
Affiliation(s)
- Yu-Yun Shao
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei 10051, Taiwan.
- National Taiwan University Cancer Center, National Taiwan University College of Medicine, Taipei 10051, Taiwan.
- Department of Oncology, National Taiwan University Hospital, Taipei 10002, Taiwan.
| | - Yong-Shi Li
- Department of Oncology, National Taiwan University Hospital, Taipei 10002, Taiwan.
| | - Hung-Wei Hsu
- Department of Oncology, National Taiwan University Hospital, Taipei 10002, Taiwan.
| | - Hang Lin
- Department of Oncology, National Taiwan University Hospital, Taipei 10002, Taiwan.
| | - Han-Yu Wang
- Department of Oncology, National Taiwan University Hospital, Taipei 10002, Taiwan.
| | - Rita Robin Wo
- Department of Oncology, National Taiwan University Hospital, Taipei 10002, Taiwan.
| | - Ann-Lii Cheng
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei 10051, Taiwan.
- National Taiwan University Cancer Center, National Taiwan University College of Medicine, Taipei 10051, Taiwan.
- Department of Oncology, National Taiwan University Hospital, Taipei 10002, Taiwan.
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei 10051, Taiwan.
| | - Chih-Hung Hsu
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei 10051, Taiwan.
- National Taiwan University Cancer Center, National Taiwan University College of Medicine, Taipei 10051, Taiwan.
- Department of Oncology, National Taiwan University Hospital, Taipei 10002, Taiwan.
| |
Collapse
|
41
|
Fu Y, Liu S, Zeng S, Shen H. From bench to bed: the tumor immune microenvironment and current immunotherapeutic strategies for hepatocellular carcinoma. J Exp Clin Cancer Res 2019; 38:396. [PMID: 31500650 PMCID: PMC6734524 DOI: 10.1186/s13046-019-1396-4] [Citation(s) in RCA: 291] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/27/2019] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) ranks the most common primary liver malignancy and the third leading cause of tumor-related mortality worldwide. Unfortunately, despite advances in HCC treatment, less than 40% of HCC patients are eligible for potentially curative therapies. Recently, cancer immunotherapy has emerged as one of the most promising approaches for cancer treatment. It has been proven therapeutically effective in many types of solid tumors, such as non-small cell lung cancer and melanoma. As an inflammation-associated tumor, it's well-evidenced that the immunosuppressive microenvironment of HCC can promote immune tolerance and evasion by various mechanisms. Triggering more vigorous HCC-specific immune response represents a novel strategy for its management. Pre-clinical and clinical investigations have revealed that various immunotherapies might extend current options for needed HCC treatment. In this review, we provide the recent progress on HCC immunology from both basic and clinical perspectives, and discuss potential advances and challenges of immunotherapy in HCC.
Collapse
MESH Headings
- Adaptive Immunity
- Animals
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- Biomarkers, Tumor
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/therapy
- Clinical Trials as Topic
- Combined Modality Therapy/methods
- Humans
- Immunity, Innate
- Immunotherapy/adverse effects
- Immunotherapy/methods
- Liver Neoplasms/immunology
- Liver Neoplasms/pathology
- Liver Neoplasms/therapy
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/pathology
- Translational Research, Biomedical
- Treatment Outcome
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Yaojie Fu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| | - Shanshan Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| | - Hong Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| |
Collapse
|
42
|
Zhang X, Wang J, Jia Y, Liu T, Wang M, Lv W, Zhang R, Shi J, Liu L. CDK5 neutralizes the tumor suppressing effect of BIN1 via mediating phosphorylation of c-MYC at Ser-62 site in NSCLC. Cancer Cell Int 2019; 19:226. [PMID: 31496920 PMCID: PMC6720419 DOI: 10.1186/s12935-019-0952-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 08/27/2019] [Indexed: 02/07/2023] Open
Abstract
Background Bridging integrator 1 (BIN1) has showed outstanding tumor-suppressive potential via inhibiting c-MYC-mediated tumorigenesis. However, a frequent phosphorylation of c-MYC at Ser-62 site could block the BIN1/c-MYC interaction and limits the tumor-suppressive effect of BIN1. Cyclin-dependent kinase 5 (CDK5), a generally dysregulated protein in various carcinomas, can mediate c-MYC phosphorylation at Ser-62 site. However, whether the existence of CDK5 could block the BIN1/c-MYC interaction remains unclear. Materials and methods The expression of CDK5 and BIN1 in non-small cell lung cancer (NSCLC) cell lines were measured. CDK5 was knocked down and overexpressed in H460 and PC9 cells, respectively. CCK-8, wound healing and transwell were used to detect the proliferation, migration and invasion ability of NSCLC cells. Tumor-bearing nude mouse model was built with H460 cells. Dinaciclib was added to realize the effect of CDK5 inhibition in vivo. NSCLC and matched para-carcinoma specimens were collected from 153 patients who underwent radical operation. IHC was performed to determine the expression of CDK5 in the specimens. Kaplan–Meier analysis was used to analyze the correlation between the postoperative survival and CDK5 expression. Results CDK5 was highly expressed in H460 cells, and knockdown of CDK5 could restore the BIN1/c-MYC interaction. Meanwhile, low expression of CDK5 was observed in PC9 cells, and overexpression of CDK5 blocked the BIN1/c-MYC interaction. Consequently, the growth, migration, invasion and epithelial mesenchymal transition (EMT) ability of H460 and PC9 cells could be facilitated by CDK5. The addition of CDK5 inhibitor Dinaciclib significantly suppressed the tumorigenesis ability of NSCLC cells in tumor-bearing mouse model. Furthermore, high expression of CDK5, along with low expression of BIN1, could predict poor postoperative prognosis of NSCLC patients. The patients with high expression of CDK5 and low expression of BIN1 showed similar prognosis, indicating that CDK5 could neutralize the tumor suppressing effect of BIN1 in clinical situation. Conclusions CDK5 blocked the interaction of BIN1 and c-MYC via promoting phosphorylation of c-MYC at ser-62 site, ultimately facilitated the progression of NSCLC.
Collapse
Affiliation(s)
- Xiangyu Zhang
- 1Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Tianshan Street 169, Shijiazhuang, China
| | - Jiali Wang
- 1Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Tianshan Street 169, Shijiazhuang, China
| | - Yunlong Jia
- 1Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Tianshan Street 169, Shijiazhuang, China
| | - Tianxu Liu
- 1Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Tianshan Street 169, Shijiazhuang, China
| | - Mengjie Wang
- 1Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Tianshan Street 169, Shijiazhuang, China
| | - Wei Lv
- 1Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Tianshan Street 169, Shijiazhuang, China
| | - Rong Zhang
- 2Department of Toxicology, Hebei Medical University, Shijiazhuang, China
| | - Juan Shi
- 3State Key Laboratory of Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lihua Liu
- 1Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Tianshan Street 169, Shijiazhuang, China
| |
Collapse
|
43
|
Vögerl K, Ong N, Senger J, Herp D, Schmidtkunz K, Marek M, Müller M, Bartel K, Shaik TB, Porter NJ, Robaa D, Christianson DW, Romier C, Sippl W, Jung M, Bracher F. Synthesis and Biological Investigation of Phenothiazine-Based Benzhydroxamic Acids as Selective Histone Deacetylase 6 Inhibitors. J Med Chem 2019; 62:1138-1166. [PMID: 30645113 DOI: 10.1021/acs.jmedchem.8b01090] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The phenothiazine system was identified as a favorable cap group for potent and selective histone deacetylase 6 (HDAC6) inhibitors. Here, we report the preparation and systematic variation of phenothiazines and their analogues containing a benzhydroxamic acid moiety as the zinc-binding group. We evaluated their ability to selectively inhibit HDAC6 by a recombinant HDAC enzyme assay, by determining the protein acetylation levels in cells by western blotting (tubulin vs histone acetylation), and by assessing their effects on various cancer cell lines. Structure-activity relationship studies revealed that incorporation of a nitrogen atom into the phenothiazine framework results in increased potency and selectivity for HDAC6 (more than 500-fold selectivity relative to the inhibition of HDAC1, HDAC4, and HDAC8), as rationalized by molecular modeling and docking studies. The binding mode was confirmed by co-crystallization of the potent azaphenothiazine inhibitor with catalytic domain 2 from Danio rerio HDAC6.
Collapse
Affiliation(s)
- Katharina Vögerl
- Department of Pharmacy-Center for Drug Research , Ludwig-Maximilians University Munich , Butenandtstr. 5-13 , 81377 Munich , Germany
| | - Nghia Ong
- Department of Pharmacy-Center for Drug Research , Ludwig-Maximilians University Munich , Butenandtstr. 5-13 , 81377 Munich , Germany
| | - Johanna Senger
- Institute of Pharmaceutical Sciences , University of Freiburg , Albertstraße 25 , 79104 Freiburg , Germany
| | - Daniel Herp
- Institute of Pharmaceutical Sciences , University of Freiburg , Albertstraße 25 , 79104 Freiburg , Germany
| | - Karin Schmidtkunz
- Institute of Pharmaceutical Sciences , University of Freiburg , Albertstraße 25 , 79104 Freiburg , Germany
| | - Martin Marek
- Département de Biologie Structurale Intégrative, Institut de Génétique et Biologie Moléculaire et Cellulaire (IGBMC) , Université de Strasbourg (UDS), CNRS, INSERM , 67404 Illkirch Cedex , France
| | - Martin Müller
- Department of Pharmacy-Center for Drug Research , Ludwig-Maximilians University Munich , Butenandtstr. 5-13 , 81377 Munich , Germany
| | - Karin Bartel
- Department of Pharmacy-Center for Drug Research , Ludwig-Maximilians University Munich , Butenandtstr. 5-13 , 81377 Munich , Germany
| | - Tajith B Shaik
- Département de Biologie Structurale Intégrative, Institut de Génétique et Biologie Moléculaire et Cellulaire (IGBMC) , Université de Strasbourg (UDS), CNRS, INSERM , 67404 Illkirch Cedex , France
| | - Nicholas J Porter
- Department of Chemistry , University of Pennsylvania , 231 South 34th Street , Philadelphia , Pennsylvania 19104-6323 , United States
| | - Dina Robaa
- Institute of Pharmacy , Martin-Luther University of Halle-Wittenberg , 06120 Halle/Saale , Germany
| | - David W Christianson
- Department of Chemistry , University of Pennsylvania , 231 South 34th Street , Philadelphia , Pennsylvania 19104-6323 , United States
| | - Christophe Romier
- Département de Biologie Structurale Intégrative, Institut de Génétique et Biologie Moléculaire et Cellulaire (IGBMC) , Université de Strasbourg (UDS), CNRS, INSERM , 67404 Illkirch Cedex , France
| | - Wolfgang Sippl
- Institute of Pharmacy , Martin-Luther University of Halle-Wittenberg , 06120 Halle/Saale , Germany
| | - Manfred Jung
- Institute of Pharmaceutical Sciences , University of Freiburg , Albertstraße 25 , 79104 Freiburg , Germany
| | - Franz Bracher
- Department of Pharmacy-Center for Drug Research , Ludwig-Maximilians University Munich , Butenandtstr. 5-13 , 81377 Munich , Germany
| |
Collapse
|