1
|
Yang W, Liu C, Li Z, Cui M. Exploring new drug treatment targets for immune related bone diseases using a multi omics joint analysis strategy. Sci Rep 2025; 15:10618. [PMID: 40148470 PMCID: PMC11950375 DOI: 10.1038/s41598-025-94053-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
In the field of treatment and prevention of immune-related bone diseases, significant challenges persist, necessitating the urgent exploration of new and effective treatment methods. However, most existing Mendelian randomization (MR) studies are confined to a single analytical approach, which limits the comprehensive understanding of the pathogenesis and potential therapeutic targets of these diseases. In light of this, we propose the hypothesis that genetic variations in specific plasma proteins have a causal relationship with immune-related bone diseases through the MR mechanism, and that key therapeutic targets can be accurately identified using an integrated multi-omic analysis approach. This study comprehensively applied a variety of analytical methods. Firstly, the protein quantitative trait locus (pQTLs) data from two large plasma protein databases and the Genome-Wide Association Study (GWAS) data of nine immune-related bone diseases were used for Mendelian randomization (MR) analysis. At the same time, we employed the Summary-based Mendelian Randomization (SMR) method, combined with the Bayesian colocalization analysis method of coding genes, as well as the Linkage Disequilibrium Score Regression (LDSC) analysis method based on genetic correlation analysis, as methods to verify the genetic association between genes and complex diseases, thus comprehensively obtaining positive results. In addition, a Phenome-wide Association Study (PheWAS) was conducted on significantly positive genes, and their expression patterns in different tissues were also explored. Subsequently, we integrated Protein-Protein Interaction (PPI) network analysis, Gene Ontology (GO) analysis. Finally, based on the above analytical methods, drug prediction and molecular docking studies were carried out with the aim of accurately identifying key therapeutic targets. Through a comprehensive analysis using four methods, namely the Mendelian randomization (MR) analysis study, Summary-based Mendelian Randomization (SMR) analysis study, Bayesian colocalization analysis study, and Linkage Disequilibrium Score Regression (LDSC) analysis study. We found that through MR, SMR, and combined with Bayesian colocalization analysis, an association was found between rheumatoid arthritis (RA) and HDGF. Using the combination of MR and Bayesian colocalization analysis, as well as LDSC analysis, it was concluded that RA was related to CCL19 and TNFRSF14. Based on the methods of MR and Bayesian colocalization, an association was found between GPT and Crohn's disease-related arthritis, and associations were found between BTN1A1, EVI5, OGA, TNFRSF14 and multiple sclerosis (MS), and associations were found between ICAM5, CCDC50, IL17RD, UBLCP1 and psoriatic arthritis (PsA). Specifically, in the MR analysis of RA, HDGF (P_ivw = 0.0338, OR = 1.0373, 95%CI = 1.0028-1.0730), CCL19 (P_ivw = 0.0004, OR = 0.3885, 95%CI = 0.2299-0.6566), TNFRSF14 (P_ivw = 0.0007, OR = 0.6947, 95%CI = 0.5634-0.8566); in the MR analysis of MS, BTN1A1 (P_ivw = 0.0000, OR = 0.6101, 95%CI = 0.4813-0.7733), EVI5 (P_ivw = 0.0000, OR = 0.3032, 95%CI = 0.1981-0.4642), OGA (P_ivw = 0.0005, OR = 0.4599, 95%CI = 0.2966-0.7131), TNFRSF14 (P_ivw = 0.0002, OR = 0.4026, 95%CI = 0.2505-0.6471); in the MR analysis of PsA, ICAM5 (P_ivw = 0.0281, OR = 1.1742, 95%CI = 1.0174-1.3552), CCDC50 (P_ivw = 0.0092, OR = 0.7359, 95%CI = 0.5843-0.9269), IL17RD (P_ivw = 0.0006, OR = 0.7887, 95%CI = 0.6886-0.9034), UBLCP1 (P_ivw = 0.0021, OR = 0.6901, 95%CI = 0.5448-0.8741); in the MR analysis of Crohn's disease-related arthritis, GPT (P_ivw = 0.0006, OR = 0.0057, 95%CI = 0.0003-0.1111). In the Bayesian colocalization analysis of RA, HDGF (H4 = 0.8426), CCL19 (H4 = 0.9762), TNFRSF14 (H4 = 0.8016); in the Bayesian colocalization analysis of MS, BTN1A1 (H4 = 0.7660), EVI5 (H4 = 0.9800), OGA (H4 = 0.8569), TNFRSF14 (H4 = 0.8904); in the Bayesian colocalization analysis of PsA, ICAM5 (H4 = 0.9476), CCDC50 (H4 = 0.9091), IL17RD (H4 = 0.9301), UBLCP1 (H4 = 0.8862); in the Bayesian colocalization analysis of Crohn's disease-related arthritis, GPT (H4 = 0.8126). In the SMR analysis of RA, HDGF (p_SMR = 0.0338, p_HEIDI = 0.0628). In the LDSC analysis of RA, CCL19 (P = 0.0000), TNFRSF14 (P = 0.0258). By comprehensively analyzing plasma proteomic and transcriptomic data, we successfully identified key therapeutic targets for various clinical subtypes of immune-associated bone diseases. Our findings indicate that the significant positive genes associated with RA include HDGF, CCL19, and TNFRSF14; the positive gene linked to Crohn-related arthropathy is GPT; for MS, the positive genes are BTN1A1, EVI5, OGA, and TNFRSF14; and for PsA, the positive genes are ICAM5, CCDC50, IL17RD, and UBLCP1. Through this comprehensive analytical approach, we have screened potential therapeutic targets for different clinical subtypes of immune-related bone diseases. This research not only enhances our understanding of the pathogenesis of these conditions but also provides a solid theoretical foundation for subsequent drug development and clinical treatment, with the potential to yield significant advancements in the management of patients with immune-related bone diseases.
Collapse
Affiliation(s)
- Wei Yang
- School of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Chenglin Liu
- School of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Zhenhua Li
- Affiliated Hospital of Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin, China.
| | - Miao Cui
- Capital Medical University, No.10, Xitoutiao, You'anmenwai, Beijing, 100069, Fengtai District, China.
| |
Collapse
|
2
|
Pehlivan ÖC, Cavuşoğlu K, Yalçin E, Acar A. In silico interactions and deep neural network modeling for toxicity profile of methyl methanesulfonate. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:117952-117969. [PMID: 37874518 DOI: 10.1007/s11356-023-30465-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 10/10/2023] [Indexed: 10/25/2023]
Abstract
In this study, the toxicity induced by the alkylating agent methyl methanesulfonate (MMS) in Allium cepa L. was investigated. For this aim, bulbs were divided into 4 groups as control and application (100, 500 and 4000 µM MMS) and germinated for 72 h at 22-24 °C. At the end of the germination period root tips were collected and made ready for analysis by applying traditional preparation methods. Germination, root elongation, weight, mitotic index (MI) values, micronucleus (MN) and chromosomal abnormality (CAs) numbers, malondialdehyde (MDA) levels, superoxide dismutase (SOD) and catalase (CAT) activities and anatomical structures of bulbs were used as indicators to determine toxicity. Moreover the extent of DNA fragmentation induced by MMS was determined by comet assay. To confirm the DNA fragmentation induced by MMS, the DNA-MMS interaction was examined with molecular docking. Correlation and principal component analyses (PCA) were performed to examine the relationship between all parameters and understand the underlying structure and relationships among these parameters. In the present study, a deep neural network (DNN) with two hidden layers implemented in Matlab has been developed for the comparison of the estimated data with the real data. The effect of MDA levels, SOD and CAT activities at 4 different endpoints resulting from administration of various concentrations of MMS, including MN, MI, CAs and DNA damage, was attempted to be estimated by DNN model. It is assumed that the predicted results are in close agreement with the actual data. The effectiveness of the model was evaluated using 4 different metrics, MAE, MAPE, RMSE and R2, which together show that the model performs commendably. As a result, the highest germination, root elongation, weight gain and MI were measured in the control group. MMS application caused a decrease in all physiological parameters and an increase in cytogenetic (except MI) and biochemical parameters. MMS application caused an increase in antioxidant enzyme levels (SOD and CAT) up to a concentration of 500 µM and a decrease at 4000 µM. MMS application induced different types of CAs and anatomical damages in root meristem cells. The results of the comet assay showed that the severity of DNA fragmentation increased with increasing MMS concentration. Molecular docking analysis showed a strong DNA-MMS interaction. The results of correlation and PCA revealed significant positive and negative interactions between the studied parameters and confirmed the interactions of these parameters with MMS. It has been shown that the DNN model developed in this study is a valuable resource for predicting genotoxicity due to oxidative stress and lipid peroxidation. In addition, this model has the potential to help evaluate the genotoxicity status of various chemical compounds. At the end of the study, it was concluded that MMS strongly supports a versatile toxicity in plant cells and the selected parameters are suitable indicators for determining this toxicity.
Collapse
Affiliation(s)
- Ömer Can Pehlivan
- Department of Biology, Institute of Science, Giresun University, Giresun, Türkiye
| | - Kültiğin Cavuşoğlu
- Department of Biology, Faculty of Science and Art, Giresun University, Giresun, Türkiye.
| | - Emine Yalçin
- Department of Biology, Faculty of Science and Art, Giresun University, Giresun, Türkiye
| | - Ali Acar
- Department of Medical Services and Techniques, Vocational School of Health Services, Giresun University, Giresun, Türkiye
| |
Collapse
|
3
|
Horvath C, Jarabicova I, Kura B, Kalocayova B, Faurobert E, Davidson SM, Adameova A. Novel, non-conventional pathways of necroptosis in the heart and other organs: Molecular mechanisms, regulation and inter-organelle interplay. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119534. [PMID: 37399908 DOI: 10.1016/j.bbamcr.2023.119534] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/28/2023] [Accepted: 06/28/2023] [Indexed: 07/05/2023]
Abstract
Necroptosis, a cell death modality that is defined as a necrosis-like cell death depending on the receptor-interacting protein kinase 3 (RIPK3) and mixed lineage kinase domain-like pseudokinase (MLKL), has been found to underlie the injury of various organs. Nevertheless, the molecular background of this cell loss seems to also involve, at least under certain circumstances, some novel axes, such as RIPK3-PGAM5-Drp1 (mitochondrial protein phosphatase 5-dynamin-related protein 1), RIPK3-CaMKII (Ca2+/calmodulin-dependent protein kinase II) and RIPK3-JNK-BNIP3 (c-Jun N-terminal kinase-BCL2 Interacting Protein 3). In addition, endoplasmic reticulum stress and oxidative stress via the higher production of reactive oxygen species produced by the mitochondrial enzymes and the enzymes of the plasma membrane have been implicated in necroptosis, thereby depicting an inter-organelle interplay in the mechanisms of this cell death. However, the role and relationship between these novel non-conventional signalling and the well-accepted canonical pathway in terms of tissue- and/or disease-specific prioritisation is completely unknown. In this review, we provide current knowledge on some necroptotic pathways being not directly associated with RIPK3-MLKL execution and report studies showing the role of respective microRNAs in the regulation of necroptotic injury in the heart and in some other tissues having a high expression of the pro-necroptotic proteins.
Collapse
Affiliation(s)
- Csaba Horvath
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Comenius University in Bratislava, Bratislava, Slovak Republic.
| | - Izabela Jarabicova
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Comenius University in Bratislava, Bratislava, Slovak Republic.
| | - Branislav Kura
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, Bratislava, Slovak Republic.
| | - Barbora Kalocayova
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, Bratislava, Slovak Republic.
| | - Eva Faurobert
- French National Centre for Scientific Research, Institute for Advanced Biosciences, France.
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, United Kingdom.
| | - Adriana Adameova
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Comenius University in Bratislava, Bratislava, Slovak Republic; Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, Bratislava, Slovak Republic.
| |
Collapse
|
4
|
Kohl Y, William N, Elje E, Backes N, Rothbauer M, Srancikova A, Rundén-Pran E, El Yamani N, Korenstein R, Madi L, Barbul A, Kozics K, Sramkova M, Steenson K, Gabelova A, Ertl P, Dusinska M, Nelson A. Rapid identification of in vitro cell toxicity using an electrochemical membrane screening platform. Bioelectrochemistry 2023; 153:108467. [PMID: 37244203 DOI: 10.1016/j.bioelechem.2023.108467] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/28/2023] [Accepted: 05/15/2023] [Indexed: 05/29/2023]
Abstract
This study compares the performance and output of an electrochemical phospholipid membrane platform against respective in vitro cell-based toxicity testing methods using three toxicants of different biological action (chlorpromazine (CPZ), colchicine (COL) and methyl methanesulphonate (MMS)). Human cell lines from seven different tissues (lung, liver, kidney, placenta, intestine, immune system) were used to validate this physicochemical testing system. For the cell-based systems, the effective concentration at 50 % cell death (EC50) values are calculated. For the membrane sensor, a limit of detection (LoD) value was extracted as a quantitative parameter describing the minimum concentration of toxicant which significantly affects the structure of the phospholipid sensor membrane layer. LoD values were found to align well with the EC50 values when acute cell viability was used as an end-point and showed a similar toxicity ranking of the tested toxicants. Using the colony forming efficiency (CFE) or DNA damage as end-point, a different order of toxicity ranking was observed. The results of this study showed that the electrochemical membrane sensor generates a parameter relating to biomembrane damage, which is the predominant factor in decreasing cell viability when in vitro models are acutely exposed to toxicants. These results lead the way to using electrochemical membrane-based sensors for rapid relevant preliminary toxicity screens.
Collapse
Affiliation(s)
- Yvonne Kohl
- Fraunhofer Institute for Biomedical Engineering IBMT, Joseph-von-Fraunhofer-Weg 1, Sulzbach 66280, Germany.
| | - Nicola William
- School of Chemistry and Faculty of Engineering and Physical Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom.
| | - Elisabeth Elje
- NILU-Norwegian Institute for Air Research, Department for Environmental Chemistry, Health Effects Laboratory, Instituttveien 18, Kjeller 2007, Norway; Faculty of Medicine, Institute of Basic Medical Sciences Department of Molecular Medicine, University of Oslo, Sognsvannsveien 9, Oslo 0372, Norway.
| | - Nadine Backes
- Fraunhofer Institute for Biomedical Engineering IBMT, Joseph-von-Fraunhofer-Weg 1, Sulzbach 66280, Germany
| | - Mario Rothbauer
- Institute of Applied Synthetic Chemistry, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria.
| | - Annamaria Srancikova
- Department of Nanobiology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, Bratislava 84505, Slovakia.
| | - Elise Rundén-Pran
- NILU-Norwegian Institute for Air Research, Department for Environmental Chemistry, Health Effects Laboratory, Instituttveien 18, Kjeller 2007, Norway.
| | - Naouale El Yamani
- NILU-Norwegian Institute for Air Research, Department for Environmental Chemistry, Health Effects Laboratory, Instituttveien 18, Kjeller 2007, Norway
| | - Rafi Korenstein
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel.
| | - Lea Madi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel.
| | - Alexander Barbul
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel.
| | - Katarina Kozics
- Department of Nanobiology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, Bratislava 84505, Slovakia.
| | - Monika Sramkova
- Department of Nanobiology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, Bratislava 84505, Slovakia.
| | - Karen Steenson
- School of Chemistry and Faculty of Engineering and Physical Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom.
| | - Alena Gabelova
- Department of Nanobiology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, Bratislava 84505, Slovakia.
| | - Peter Ertl
- Institute of Applied Synthetic Chemistry, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria; Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria.
| | - Maria Dusinska
- NILU-Norwegian Institute for Air Research, Department for Environmental Chemistry, Health Effects Laboratory, Instituttveien 18, Kjeller 2007, Norway.
| | - Andrew Nelson
- School of Chemistry and Faculty of Engineering and Physical Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom.
| |
Collapse
|
5
|
Hänggi K, Ruffell B. Cell death, therapeutics, and the immune response in cancer. Trends Cancer 2023; 9:381-396. [PMID: 36841748 PMCID: PMC10121860 DOI: 10.1016/j.trecan.2023.02.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/19/2023] [Accepted: 02/03/2023] [Indexed: 02/27/2023]
Abstract
Induction of cell death is inexorably linked with cancer therapy, but this can also initiate wound-healing processes that have been linked to cancer progression and therapeutic resistance. Here we describe the contribution of apoptosis and the lytic cell death pathways in the response to therapy (including chemotherapy and immunotherapy). We also discuss how necroptosis, pyroptosis, and ferroptosis function to promote tumor immunogenicity, along with emerging findings that these same forms of death can paradoxically contribute to immune suppression and tumor progression. Understanding the duality of cell death in cancer may allow for the development of therapeutics that shift the balance towards regression.
Collapse
Affiliation(s)
- Kay Hänggi
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Brian Ruffell
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA; Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| |
Collapse
|
6
|
Chaouhan HS, Vinod C, Mahapatra N, Yu SH, Wang IK, Chen KB, Yu TM, Li CY. Necroptosis: A Pathogenic Negotiator in Human Diseases. Int J Mol Sci 2022; 23:12714. [PMID: 36361505 PMCID: PMC9655262 DOI: 10.3390/ijms232112714] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 11/25/2022] Open
Abstract
Over the past few decades, mechanisms of programmed cell death have attracted the scientific community because they are involved in diverse human diseases. Initially, apoptosis was considered as a crucial mechanistic pathway for programmed cell death; recently, an alternative regulated mode of cell death was identified, mimicking the features of both apoptosis and necrosis. Several lines of evidence have revealed that dysregulation of necroptosis leads to pathological diseases such as cancer, cardiovascular, lung, renal, hepatic, neurodegenerative, and inflammatory diseases. Regulated forms of necrosis are executed by death receptor ligands through the activation of receptor-interacting protein kinase (RIPK)-1/3 and mixed-lineage kinase domain-like (MLKL), resulting in the formation of a necrosome complex. Many papers based on genetic and pharmacological studies have shown that RIPKs and MLKL are the key regulatory effectors during the progression of multiple pathological diseases. This review focused on illuminating the mechanisms underlying necroptosis, the functions of necroptosis-associated proteins, and their influences on disease progression. We also discuss numerous natural and chemical compounds and novel targeted therapies that elicit beneficial roles of necroptotic cell death in malignant cells to bypass apoptosis and drug resistance and to provide suggestions for further research in this field.
Collapse
Affiliation(s)
- Hitesh Singh Chaouhan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
| | - Ch Vinod
- Department of Biological Sciences, School of Applied Sciences, KIIT University, Bhubaneshwar 751024, India
| | - Nikita Mahapatra
- Department of Biological Sciences, School of Applied Sciences, KIIT University, Bhubaneshwar 751024, India
| | - Shao-Hua Yu
- Department of Emergency Medicine, China Medical University Hospital, Taichung 40402, Taiwan
| | - I-Kuan Wang
- School of Medicine, China Medical University, Taichung 40402, Taiwan
- Department of Internal Medicine, China Medical University Hospital, Taichung 40402, Taiwan
| | - Kuen-Bao Chen
- Department of Anesthesiology, China Medical University Hospital, Taichung 40402, Taiwan
| | - Tung-Min Yu
- School of Medicine, China Medical University, Taichung 40402, Taiwan
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung 40402, Taiwan
| | - Chi-Yuan Li
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- School of Medicine, China Medical University, Taichung 40402, Taiwan
- Department of Anesthesiology, China Medical University Hospital, Taichung 40402, Taiwan
| |
Collapse
|
7
|
Yang HM, Hou TZ, Zhang YN, Zhao SD, Wu YL, Zhang H. Blocked metabotropic glutamate receptor 5 enhances chemosensitivity in hepatocellular carcinoma and attenuates chemotoxicity in the normal liver by regulating DNA damage. Cancer Gene Ther 2022; 29:1487-1501. [PMID: 35396501 DOI: 10.1038/s41417-022-00465-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 03/08/2022] [Accepted: 03/21/2022] [Indexed: 12/12/2022]
Abstract
DNA damaging agents are used as chemotherapeutics in many cancers, including hepatocellular carcinoma (HCC). However, they are associated with problems such as low sensitivity to chemotherapy and the induction of liver injury, underscoring the need to identify new therapies. Here, we investigated the differential regulatory effect of metabotropic glutamate receptor 5 (mGlu5) on chemosensitivity in HCC and chemotoxicity to the normal liver. The expression of mGlu5 was higher in HCC than in the normal liver, and correlated with poor prognosis according to The Cancer Genome Atlas database and Integrative Molecular Database of Hepatocellular Carcinoma. Cisplatin, oxaliplatin or methyl methanesulfonate (MMS) caused cell death by decreasing mGlu5 expression in HCC cells and increased mGlu5 expression in hepatic cells. In HCC cells, inhibition of mGlu5 aggravated MMS-induced DNA damage by increasing intracellular Ca2+ overload and mitogen-activated protein kinase (MAPK) activation, thereby promoting cell death, and activation of mGlu5 rescued the effect of MMS. However, in hepatic cells, mGlu5 inhibition alleviated MMS-induced DNA damage by downregulating Ca2+-derived MAPK pathways to advance hepatic cell survival. The opposite effects of mGlu5 overexpression or knockdown on MMS-induced DNA damage supported that cell death is a result of the differential regulation of mGlu5 expression. Inhibition of mGlu5 increased chemosensitivity and decreased chemotoxicity in a rat tumor model. This study suggests that mGlu5 inhibition could act synergistically with HCC chemotherapeutics with minimal side effects, which may improve the treatment of patients with HCC in the future.
Collapse
Affiliation(s)
- Hui-Min Yang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, 100069, Beijing, China
| | - Tian-Zhong Hou
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, 100069, Beijing, China
| | - Ya-Nan Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, 100069, Beijing, China
| | - Shu-Dong Zhao
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, 100069, Beijing, China
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, Beihang University, 100083, Beijing, China
| | - Yong-Le Wu
- Center of Hepatic and Digestive Disease, Beijing YouAn Hospital, Capital Medical University, 100069, Beijing, China
| | - Hong Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, 100069, Beijing, China.
| |
Collapse
|
8
|
Vale Junior EPDO, Ferreira MVR, Fernandes BCS, Silva TTDA, Martins FA, Almeida PMDE. Protective effect of kavain in meristematic cells of Allium cepa L. AN ACAD BRAS CIENC 2022; 94:e20200520. [PMID: 35703688 DOI: 10.1590/0001-3765202220200520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 06/21/2020] [Indexed: 05/10/2025] Open
Abstract
Kavain is one of the main kavalactones of Piper methysticum (Piperaceae) with anxiolytic, analgesic, and antioxidant activities. Therefore, the aim of the study was to evaluate the cytotoxic, mutagenic, and antimutagenic potential of kavain in Allium cepa cells. Roots of A. cepa were transferred to the negative (2% acetone) and positive (10 µg/mL of Methylmethanesulfonate, MMS) controls and to the concentrations of kavain (32, 64 and 128 µg/mL) for 48 h. A total of 5,000 meristematic cells were analyzed under an optical microscope to determine the mitotic index, mean number of chromosomal alterations and percentage of damage reduction. Data were analyzed by Kruskal-Wallis test (p <0.05). All concentrations of kavain were not cytotoxic and did not show significant chromosomal changes when compared to 2% acetone. Kavain showed a cytoprotective effect in the pre (128 μg/mL) and in the post-treatment (32 and 64 μg/mL) and reduced damage against the mutagenic action of MMS in all concentrations of the pre and simultaneous and at the highest of post (128 μg/mL). Kavain promoted a significant reduction in micronuclei, nuclear buds and chromosomal losses in relation to MMS. The observed data indicate the importance of kavain for the inhibition of damage and chemoprevention.
Collapse
Affiliation(s)
- Erasmo P DO Vale Junior
- Universidade Estadual do Piauí, Centro de Ciências da Natureza (CCN), Laboratório de Genética, Rua João Cabral, 2231, 64002-150 Teresina PI, Brazil
| | - Marcos Vitor R Ferreira
- Universidade Estadual do Piauí, Centro de Ciências da Natureza (CCN), Laboratório de Genética, Rua João Cabral, 2231, 64002-150 Teresina PI, Brazil
| | - Bianca Cristina S Fernandes
- Universidade Estadual do Piauí, Centro de Ciências da Natureza (CCN), Laboratório de Genética, Rua João Cabral, 2231, 64002-150 Teresina PI, Brazil
| | - Thais T DA Silva
- Universidade Estadual do Piauí, Centro de Ciências da Natureza (CCN), Laboratório de Genética, Rua João Cabral, 2231, 64002-150 Teresina PI, Brazil
| | - Francielle Alline Martins
- Universidade Estadual do Piauí, Centro de Ciências da Natureza (CCN), Laboratório de Genética, Rua João Cabral, 2231, 64002-150 Teresina PI, Brazil
| | - Pedro Marcos DE Almeida
- Universidade Estadual do Piauí (UESPI/FACIME), Centro de Ciências da Saúde (CCS), Departamento de Genética, Laboratório de Genética. Rua Olavo Bilac, 2335, 64049-570 Teresina PI, Brazil
| |
Collapse
|
9
|
N6-isopentenyladenosine induces cell death through necroptosis in human glioblastoma cells. Cell Death Dis 2022; 8:173. [PMID: 35393392 PMCID: PMC8991250 DOI: 10.1038/s41420-022-00974-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/21/2022] [Accepted: 03/23/2022] [Indexed: 01/17/2023]
Abstract
Targeting necroptosis is considered a promising therapeutic strategy in cancer, including Glioblastoma Multiforme (GBM), one of the most lethal brain tumors. Necroptosis is a mechanism of programmed cell death overcoming the apoptosis resistance mechanism underlying GBM tumorigenesis and malignant progression. N6-isopentenyladenosine (iPA), adenosine modified with isoprenoid derivative, displays antitumor activity in different cancer models. In previous studies, we demonstrated that iPA interferes with EGFR signaling reducing glioma cell viability. Here, we show that iPA induces necroptosis in glioblastoma cell lines and in primary cells established from tumor explants, without affecting the viability of non-cancerous brain cell lines, (Normal Human Astrocyte). The activation of RIP1, RIP3, and MLKL and the upregulation of necrosome formation were increased upon iPA treatment while caspase-3, caspase-8, and PARP were not activated in GBM cells. Co-treatment with specific necroptosis inhibitor necrostatin-1 (Nec-1) or Necrosulfonamide (NSA) prevented cell death caused by iPA treatment while the general caspase inhibitor Z-VAD-fluoromethylketone (z-VAD-fmk) did not elicit any effect, suggesting that this molecule induces caspase-independent necroptosis. These results suggest that iPA treatment can be able to bypass the apoptosis resistance mechanism in glioblastoma thereby offering higher therapeutic efficacy.
Collapse
|
10
|
Onchieku NM, Kumari S, Pandey R, Sharma V, Kumar M, Deshmukh A, Kaur I, Mohmmed A, Gupta D, Kiboi D, Gaur N, Malhotra P. Artemisinin Binds and Inhibits the Activity of Plasmodium falciparum Ddi1, a Retroviral Aspartyl Protease. Pathogens 2021; 10:pathogens10111465. [PMID: 34832620 PMCID: PMC8621276 DOI: 10.3390/pathogens10111465] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/28/2021] [Accepted: 11/01/2021] [Indexed: 01/10/2023] Open
Abstract
Reduced sensitivity of the human malaria parasite, Plasmodium falciparum, to Artemisinin and its derivatives (ARTs) threatens the global efforts towards eliminating malaria. ARTs have been shown to cause ubiquitous cellular and genetic insults, which results in the activation of the unfolded protein response (UPR) pathways. The UPR restores protein homeostasis, which otherwise would be toxic to cellular survival. Here, we interrogated the role of DNA-damage inducible protein 1 (PfDdi1), a unique proteasome-interacting retropepsin in mediating the actions of the ARTs. We demonstrate that PfDdi1 is an active A2 family protease that hydrolyzes ubiquitinated proteasome substrates. Treatment of P. falciparum parasites with ARTs leads to the accumulation of ubiquitinated proteins in the parasites and blocks the destruction of ubiquitinated proteins by inhibiting the PfDdi1 protease activity. Besides, whereas the PfDdi1 is predominantly localized in the cytoplasm, exposure of the parasites to ARTs leads to DNA fragmentation and increased recruitment of the PfDdi1 into the nucleus. Furthermore, we show that Ddi1 knock-out Saccharomycescerevisiae cells are more susceptible to ARTs and the PfDdI1 protein robustly restores the corresponding functions in the knock-out cells. Together, these results show that ARTs act in multiple ways; by inducing DNA and protein damage and might be impairing the damage recovery by inhibiting the activity of PfDdi1, an essential ubiquitin-proteasome retropepsin.
Collapse
Affiliation(s)
- Noah Machuki Onchieku
- Malaria Biology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India or (N.M.O.); (V.S.); (A.D.); (I.K.)
| | - Sonam Kumari
- Yeast Biofuel Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India; (S.K.); (M.K.); (N.G.)
| | - Rajan Pandey
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India; (R.P.); (D.G.)
| | - Vaibhav Sharma
- Malaria Biology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India or (N.M.O.); (V.S.); (A.D.); (I.K.)
| | - Mohit Kumar
- Yeast Biofuel Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India; (S.K.); (M.K.); (N.G.)
| | - Arunaditya Deshmukh
- Malaria Biology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India or (N.M.O.); (V.S.); (A.D.); (I.K.)
| | - Inderjeet Kaur
- Malaria Biology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India or (N.M.O.); (V.S.); (A.D.); (I.K.)
| | - Asif Mohmmed
- Parasite Cell Biology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India;
| | - Dinesh Gupta
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India; (R.P.); (D.G.)
| | - Daniel Kiboi
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology, Nairobi P.O. Box 62000-00200, Kenya;
| | - Naseem Gaur
- Yeast Biofuel Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India; (S.K.); (M.K.); (N.G.)
| | - Pawan Malhotra
- Malaria Biology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India or (N.M.O.); (V.S.); (A.D.); (I.K.)
- Correspondence: or
| |
Collapse
|
11
|
Dias MS, Junior EPV, Santos BCD, Martins FA, Almeida PMDE, Peron AP. Cytogenotoxicity and protective effect of piperine and capsaicin on meristematic cells of Allium cepa L. AN ACAD BRAS CIENC 2021; 93:e20201772. [PMID: 34550203 DOI: 10.1590/0001-3765202120201772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 03/02/2021] [Indexed: 11/22/2022] Open
Abstract
Piperine and capsaicin are important molecules with biological and pharmacological activities. This study aimed to evaluate the cytogenotoxic and protective effect of piperine and capsaicin on Allium cepa cells. A. cepa roots were exposed to negative (2% Dimethylsulfoxide) and positive (Methylmethanesulfonate, MMS, 10 µg/mL) controls, and four concentrations (25-200 µM) of piperine or capsaicin (alone) or associated before, simultaneously or after with the MMS. Only the lowest concentration of piperine (25 µM) showed a protective effect because it was not genotoxic. Piperine and capsaicin were cytotoxic (50, 100 and 200 µM). Piperine (50 to 200 µM) caused a significant increase in the total average of chromosomal alterations of in A. cepa cells. For capsaicin, the genotoxic effect was dose-dependent with a significant increase for all concentrations, highlighting the significant presence of micronuclei and nuclear buds for the two isolates. In general, bioactive compounds reduced the total average of chromosomal alterations against damage caused by MMS, mainly micronuclei and/or nuclear buds. Therefore, the two molecules were cytotoxic and genotoxic at the highest concentrations, and did not have cytoprotective action, and the lowest concentration of piperine demonstrated important chemopreventive activity.
Collapse
Affiliation(s)
- Marcondes S Dias
- Programa de Pós-Graduação em Genética e Melhoramento, Universidade Federal do Piauí /UFPI, Laboratório de Genética, Ininga, 64049-550 Teresina, PI, Brazil
| | - Erasmo P V Junior
- Universidade Estadual do Piauí, Centro de Ciências Naturais (CCN), Laboratório de Genética, Rua João Cabral, 2231, 64002-150 Teresina, PI, Brazil
| | - Bianca C Dos Santos
- Universidade Estadual do Piauí, Centro de Ciências Naturais (CCN), Laboratório de Genética, Rua João Cabral, 2231, 64002-150 Teresina, PI, Brazil
| | - Francielle A Martins
- Universidade Estadual do Piauí, Centro de Ciências Naturais (CCN), Laboratório de Genética, Rua João Cabral, 2231, 64002-150 Teresina, PI, Brazil
| | - Pedro M DE Almeida
- Universidade Estadual do Piauí, Centro de Ciências Naturais (CCN), Laboratório de Genética, Rua João Cabral, 2231, 64002-150 Teresina, PI, Brazil
| | - Ana P Peron
- Universidade Federal de Tecnologia, Paraná /UTFPR, Departamento de Biodiversidade e Conservação da Natureza, Campus Campo Mourão, Via Rosalina Maria dos Santos, 1233, Caixa Postal 271, 87301-899 Campo Mourão, PR, Brazil
| |
Collapse
|
12
|
Barati M, Javidi MA, Darvishi B, Shariatpanahi SP, Mesbah Moosavi ZS, Ghadirian R, Khani T, Sanati H, Simaee H, Shokrollahi Barough M, Farahmand L, Madjid Ansari A. Necroptosis triggered by ROS accumulation and Ca 2+ overload, partly explains the inflammatory responses and anti-cancer effects associated with 1Hz, 100 mT ELF-MF in vivo. Free Radic Biol Med 2021; 169:84-98. [PMID: 33857627 DOI: 10.1016/j.freeradbiomed.2021.04.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 02/06/2021] [Accepted: 04/02/2021] [Indexed: 12/26/2022]
Abstract
Whereas the anti-neoplastic activity of extremely low frequency magnetic fields (ELF-EMF) is well-documented in literature, little is known about its underlying anti-cancer mechanisms and induced types of cell death. Here, for the first time, we reported induction of necroptosis, a specific type of programed necrotic cell death, in MC4-L2 breast cancer cell lines following a 2 h/day exposure to a 100 Hz, 1 mT ELF-EMF for five days. For in vivo assessment, inbred BALB/c mice bearing established MC-4L2 tumors were exposed to 100 mT, 1 Hz ELF-EMF 2 h daily for a period of 28-day, following which tumors were dissected and fixed for evaluation of tumor biomarkers expression and types of cell death induced using TUNEL assay, Immunohistochemistry and H&E staining. Peripheral blood samples were also collected for assessing pro-inflammatory cytokine profile following exposure. An exaggerated proinflammatory response evident form enhancement of IFN-γ (4.8 ± 0.24 folds) and TNF-α (3.1 ± 0.19 folds) and number of tumors infiltrating lymphocytes (TILs), specially CD8+ Th cells (~20 folds), proposed occurrence of necroptosis in vivo. Meanwhile, exposure could effectively suppress tumor growth and expression of Ki-67, CD31, VEGFR2 and MMP-9. In vitro studies on ELF-EMF exposed MC-4L2 cells demonstrated a meaningful increase in phosphorylation of RIPK1/RIPK3/MLKL proteins and cleavage of caspase-9/caspase-3, confirming occurrence of both necroptosis and apoptosis. Complementary in vitro studies by treating ELF-EMF exposed MC-4L2 cells with verapamil (a calcium channel inhibitor), N-acetyl cysteine (a ROS scavenger) or calcium chloride confirmed the role of elevated intracellular calcium and ROS levels in ELF-EMF induced necroptosis.
Collapse
Affiliation(s)
- Mojdeh Barati
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Mohammad Amin Javidi
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Behrad Darvishi
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | | | - Zahra S Mesbah Moosavi
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Reyhane Ghadirian
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Tahereh Khani
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Hassan Sanati
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Hossein Simaee
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | | | - Leila Farahmand
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Alireza Madjid Ansari
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.
| |
Collapse
|
13
|
Pigarev SE, Trashkov AP, Panchenko AV, Yurova MN, Bykov VN, Fedoros EI, Anisimov VN. Evaluation of the genotoxic and antigenotoxic potential of lignin-derivative BP-C2 in the comet assay in vivo. ENVIRONMENTAL RESEARCH 2021; 192:110321. [PMID: 33075358 DOI: 10.1016/j.envres.2020.110321] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 09/29/2020] [Accepted: 10/06/2020] [Indexed: 06/11/2023]
Abstract
The genotoxic and antigenotoxic potential of BP-C2, a novel lignin-derived polyphenolic composition with ammonium molybdate, was investigated as a radioprotector/radiomitigator for civil applications and as a medical countermeasure for radiation emergencies. Using the alkaline comet assay and methyl methanesulfonate (MMS, 40 mg/kg) as the DNA-damaging agent, these effects of BP-C2 on liver, bone marrow cells and blood leukocytes in rats were studied. The DNA damage was estimated by the DNA content in the comet tail (TDNA, %) 1, 6 and 18 h post exposure to MMS. BP-C2 at doses of 20, 200 and 2000 mg/kg did not exert genotoxic activity in the tested tissues in rats. BP-C2 administered at doses of 20, 100 and 200 mg/kg 1 h before MMS significantly (p < 0.01) mitigated MMS-induced DNA damage, showing a strong genoprotective effect in the liver. In blood leukocytes and bone marrow samples of animals treated with BP-C2, the TDNA % was slightly higher than in the negative control (vehicle) but significantly lower than in the positive control (MMS). Thus, BP-C2 exerted a genoprotective effect against MMS-induced DNA damage to a greater extent towards liver cells, requiring further evaluation of this substance as a genoprotective agent.
Collapse
Affiliation(s)
- S E Pigarev
- N.N. Petrov National Medical Research Center of Oncology, Saint Petersburg, Russia; Nobel LTD, Saint-Petersburg, Russia.
| | - A P Trashkov
- Petersburg Nuclear Physics Institute Named By B.P. Konstantinov of NRC "Kurchatov Institute", Gatchina, Russia
| | - A V Panchenko
- N.N. Petrov National Medical Research Center of Oncology, Saint Petersburg, Russia; FSBSI "Research Institute of Medical Primatology", Sochi, Russian
| | - M N Yurova
- N.N. Petrov National Medical Research Center of Oncology, Saint Petersburg, Russia
| | - V N Bykov
- N.N. Petrov National Medical Research Center of Oncology, Saint Petersburg, Russia
| | - E I Fedoros
- N.N. Petrov National Medical Research Center of Oncology, Saint Petersburg, Russia; Nobel LTD, Saint-Petersburg, Russia
| | - V N Anisimov
- N.N. Petrov National Medical Research Center of Oncology, Saint Petersburg, Russia
| |
Collapse
|
14
|
Wu Y, Dong G, Sheng C. Targeting necroptosis in anticancer therapy: mechanisms and modulators. Acta Pharm Sin B 2020; 10:1601-1618. [PMID: 33088682 PMCID: PMC7563021 DOI: 10.1016/j.apsb.2020.01.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 11/19/2019] [Accepted: 12/30/2019] [Indexed: 02/07/2023] Open
Abstract
Necroptosis, a genetically programmed form of necrotic cell death, serves as an important pathway in human diseases. As a critical cell-killing mechanism, necroptosis is associated with cancer progression, metastasis, and immunosurveillance. Targeting necroptosis pathway by small molecule modulators is emerging as an effective approach in cancer therapy, which has the advantage to bypass the apoptosis-resistance and maintain antitumor immunity. Therefore, a better understanding of the mechanism of necroptosis and necroptosis modulators is necessary to develop novel strategies for cancer therapy. This review will summarize recent progress of the mechanisms and detecting methods of necroptosis. In particular, the relationship between necroptosis and cancer therapy and medicinal chemistry of necroptosis modulators will be focused on.
Collapse
|
15
|
Lai KP, Cheung A, Ho CH, Tam NYK, Li JW, Lin X, Chan TF, Lee NPY, Li R. Transcriptomic analysis reveals the oncogenic role of S6K1 in hepatocellular carcinoma. J Cancer 2020; 11:2645-2655. [PMID: 32201535 PMCID: PMC7065997 DOI: 10.7150/jca.40726] [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: 11/22/2019] [Accepted: 01/19/2020] [Indexed: 12/20/2022] Open
Abstract
The p70 ribosomal protein S6 kinase 1 (S6K1), a serine/threonine kinase, is commonly overexpressed in a variety of cancers. However, its expression level and functional roles in hepatocellular carcinoma (HCC), which ranks as the third leading cause of cancer-related death worldwide, is still largely unknown. In the current report, we show the in vivo and in vitro overexpression of S6K1 in HCC. In the functional analysis, we demonstrate that S6K1 is required for the proliferation and colony formation abilities in HCC. By using comparative transcriptomic analysis followed by gene ontology enrichment analysis and Ingenuity Pathway Analysis, we find that the depletion of S6K1 can elevate the expression of a cluster of apoptotic genes, tumor suppressor genes and immune responsive genes. Moreover, the knockdown of S6K1 is predicted to reduce the tumorigenicity of HCC through the regulation of hubs of genes including STAT1, HDAC4, CEBPA and ONECUT1. In conclusion, we demonstrate the oncogenic role of S6K1 in HCC, suggesting the possible use of S6K1 as a therapeutic target for HCC treatment.
Collapse
Affiliation(s)
- Keng Po Lai
- Guanxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, PR China.,Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
| | - Angela Cheung
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
| | - Cheuk Hin Ho
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
| | - Nathan Yi-Kan Tam
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
| | - Jing Woei Li
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
| | - Xiao Lin
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ting Fung Chan
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.,State Key Laboratory of Agrobiotechnology, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Nikki Pui-Yue Lee
- Department of Surgery, University of Hong Kong, Hong Kong SAR, China
| | - Rong Li
- Guanxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, PR China
| |
Collapse
|
16
|
Gao X, Wang J, Li M, Wang J, Lv J, Zhang L, Sun C, Ji J, Yang W, Zhao Z, Mao W. Berberine attenuates XRCC1-mediated base excision repair and sensitizes breast cancer cells to the chemotherapeutic drugs. J Cell Mol Med 2019; 23:6797-6804. [PMID: 31338966 PMCID: PMC6787507 DOI: 10.1111/jcmm.14560] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 06/21/2019] [Accepted: 07/05/2019] [Indexed: 12/21/2022] Open
Abstract
Berberine (BBR) is a natural isoquinoline alkaloid, which is used in traditional medicine for its anti‐microbial, anti‐protozoal, anti‐diarrhoeal activities. Berberine interacts with DNA and displays anti‐cancer activities, yet its effects on cellular DNA repair and on synthetic treatments with chemotherapeutic drugs remain unclear. In this study, we investigated the effects of BBR on DNA repair and on sensitization of breast cancer cells to different types of DNA damage anti‐tumoural drugs. We found BBR arrested cells in the cell cycle S phase and induced DNA breaks. Cell growth analysis showed BBR sensitized MDA‐MB‐231 cells to cisplatin, camptothecin and methyl methanesulfonate; however, BBR had no synergistic effects with hydroxurea and olaparib. These results suggest BBR only affects specific DNA repair pathways. Western blot showed BBR down‐regulated XRCC1 expressions, and the rescued XRCC1 recovered the resistance of cancer cells to BBR. Therefore, we conclude that BBR interferes with XRCC1‐mediated base excision repair to sensitize cancer cells to chemotherapeutic drugs. These finding can contribute to understanding the effects of BBR on cellular DNA repair and the clinical employment of BBR in treatment of breast cancer.
Collapse
Affiliation(s)
- Xingjie Gao
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Jing Wang
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Meiqi Li
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Jia Wang
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Jian Lv
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Lu Zhang
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Caifeng Sun
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Jiamei Ji
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Wenbo Yang
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Zinan Zhao
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Weifeng Mao
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| |
Collapse
|
17
|
Fernandes Veloso Borges F, Ribeiro e Silva C, Moreira Goes W, Ribeiro Godoy F, Craveiro Franco F, Hollanda Véras J, Luiz Cardoso Bailão EF, de Melo e Silva D, Gomes Cardoso C, Divino da Cruz A, Chen-Chen L. Protective Effects of Silymarin and Silibinin against DNA Damage in Human Blood Cells. BIOMED RESEARCH INTERNATIONAL 2018; 2018:6056948. [PMID: 30370304 PMCID: PMC6189666 DOI: 10.1155/2018/6056948] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 07/25/2018] [Accepted: 09/02/2018] [Indexed: 01/15/2023]
Abstract
Silymarin (SM), a standardized extract derived from Silybum marianum (L.) Gaertn, is primarily composed of flavonolignans, with silibinin (SB) as its major active constituent. The present study aimed to evaluate the antigenotoxic activities of SM and SB using the alkaline comet assay in whole blood cells and to assess their effects on the expression of genes associated with carcinogenesis and chemopreventive processes. Different concentrations of SM or SB (1.0, 2.5, 5.0, and 7.5 mg/ml) were used in combination with the DNA damage-inducing agent methyl methanesulfonate (MMS, 800 μM) to evaluate their genoprotective potential. To investigate the role of SM and SB in modulating gene expression, we performed quantitative real-time PCR (qRT-PCR) analysis of five genes that are known to be involved in DNA damage, carcinogenesis, and/or chemopreventive mechanisms. Treatment with SM or SB was found to significantly reduce the genotoxicity of MMS, upregulate the expression of PTEN and BCL2, and downregulate the expression of BAX and ABL1. We observed no significant changes in ETV6 expression levels following treatment with SM or SB. In conclusion, both SM and SB exerted antigenotoxic activities and modulated the expression of genes related to cell protection against DNA damage.
Collapse
Affiliation(s)
- Flávio Fernandes Veloso Borges
- Laboratório de Radiobiologia e Mutagênese, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Campus II, Goiânia, GO, Brazil
| | - Carolina Ribeiro e Silva
- Laboratório de Radiobiologia e Mutagênese, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Campus II, Goiânia, GO, Brazil
| | - Wanessa Moreira Goes
- Laboratório de Mutagênese (LABMUT), Instituto de Ciências Biológicas, Universidade Federal de Goiás, Campus II, Goiânia, GO, Brazil
| | - Fernanda Ribeiro Godoy
- Laboratório de Mutagênese (LABMUT), Instituto de Ciências Biológicas, Universidade Federal de Goiás, Campus II, Goiânia, GO, Brazil
| | - Fernanda Craveiro Franco
- Laboratório de Mutagênese (LABMUT), Instituto de Ciências Biológicas, Universidade Federal de Goiás, Campus II, Goiânia, GO, Brazil
| | - Jefferson Hollanda Véras
- Laboratório de Radiobiologia e Mutagênese, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Campus II, Goiânia, GO, Brazil
| | | | - Daniela de Melo e Silva
- Laboratório de Mutagênese (LABMUT), Instituto de Ciências Biológicas, Universidade Federal de Goiás, Campus II, Goiânia, GO, Brazil
| | - Clever Gomes Cardoso
- Laboratório de Radiobiologia e Mutagênese, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Campus II, Goiânia, GO, Brazil
| | - Aparecido Divino da Cruz
- Núcleo de Pesquisas Replicon, Escola de Ciências Agrárias e Biológicas, Pontifícia Universidade Católica de Goiás, Goiânia, GO, Brazil
| | - Lee Chen-Chen
- Laboratório de Radiobiologia e Mutagênese, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Campus II, Goiânia, GO, Brazil
| |
Collapse
|
18
|
Selenoprotein S silencing triggers mouse hepatoma cells apoptosis and necrosis involving in intracellular calcium imbalance and ROS-mPTP-ATP. Biochim Biophys Acta Gen Subj 2018; 1862:2113-2123. [PMID: 30017912 DOI: 10.1016/j.bbagen.2018.07.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 06/22/2018] [Accepted: 07/04/2018] [Indexed: 12/26/2022]
Abstract
Selenoprotein S (SelenoS) is one of the cellular endoplasmic reticulum (ER) and membrane located selenoproteins, and it has the main functions of anti-oxidation, anti-apoptosis and anti-ER stress. To investigate the effect of SelenoS silencing on mouse hepatoma cell death and the intracellular biological function of SelenoS, we knocked down SelenoS in Hepa1-6 cells, and detected ER stress, intracellular calcium homeostasis, mitochondrial dynamics, apoptosis and necrosis. To further explore whether reactive oxygen species (ROS) has an effect on apoptosis and necrosis under SelenoS silencing, we used NAC (2.5 mM) to pretreat cells, and detected ΔΨm, ATP, and apoptosis and necrosis rates. SelenoS silencing broke the intracellular calcium homeostasis, induced mitochondrial dynamic disorder, ROS accumulation, loss of ΔΨm and ATP, and triggered apoptosis and necrosis in mouse hepatoma cells. The clearance of ROS alleviated the loss of ΔΨm and ATP caused by silencing of SelenoS, reduced cell necrosis and increased apoptosis. However, SelenoS silencing did not cause ER stress in Hepa1-6 cells. These results indicate that SelenoS silencing triggers mouse hepatoma cells apoptosis and necrosis through affecting intracellular calcium homeostasis and ROS-mPTP-ATP participates in cell death transformation from apoptosis to necrosis to rise damage.
Collapse
|
19
|
Meng MB, Wang HH, Cui YL, Wu ZQ, Shi YY, Zaorsky NG, Deng L, Yuan ZY, Lu Y, Wang P. Necroptosis in tumorigenesis, activation of anti-tumor immunity, and cancer therapy. Oncotarget 2018; 7:57391-57413. [PMID: 27429198 PMCID: PMC5302997 DOI: 10.18632/oncotarget.10548] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 06/20/2016] [Indexed: 02/05/2023] Open
Abstract
While the mechanisms underlying apoptosis and autophagy have been well characterized over recent decades, another regulated cell death event, necroptosis, remains poorly understood. Elucidating the signaling networks involved in the regulation of necroptosis may allow this form of regulated cell death to be exploited for diagnosis and treatment of cancer, and will contribute to the understanding of the complex tumor microenvironment. In this review, we have summarized the mechanisms and regulation of necroptosis, the converging and diverging features of necroptosis in tumorigenesis, activation of anti-tumor immunity, and cancer therapy, as well as attempts to exploit this newly gained knowledge to provide therapeutics for cancer.
Collapse
Affiliation(s)
- Mao-Bin Meng
- Department of Radiation Oncology, Tianjin's Clinical Research Center for Cancer and Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Huan-Huan Wang
- Department of Radiation Oncology, Tianjin's Clinical Research Center for Cancer and Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Yao-Li Cui
- Department of Lymphoma, Tianjin's Clinical Research Center for Cancer and Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Zhi-Qiang Wu
- Department of Radiation Oncology, Tianjin's Clinical Research Center for Cancer and Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Yang-Yang Shi
- Stanford University School of Medicine, Stanford, CA, United States of America
| | - Nicholas G Zaorsky
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA, United States of America
| | - Lei Deng
- Department of Thoracic Cancer and Huaxi Student Society of Oncology Research, West China Hospital, West China School of Medicine, Sichuan University, Sichuan Province, China
| | - Zhi-Yong Yuan
- Department of Radiation Oncology, Tianjin's Clinical Research Center for Cancer and Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - You Lu
- Department of Thoracic Cancer and Huaxi Student Society of Oncology Research, West China Hospital, West China School of Medicine, Sichuan University, Sichuan Province, China
| | - Ping Wang
- Department of Radiation Oncology, Tianjin's Clinical Research Center for Cancer and Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
20
|
Dong W, Li Z, Chen Y, Zhang L, Ye Z, Liang H, Li R, Xu L, Zhang B, Liu S, Wang W, Li C, Luo J, Shi W, Liang X. NADPH oxidase inhibitor, diphenyleneiodonium prevents necroptosis in HK-2 cells. Biomed Rep 2017; 7:226-230. [PMID: 28894570 DOI: 10.3892/br.2017.948] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 06/21/2017] [Indexed: 12/31/2022] Open
Abstract
The aim of the present study was to investigate the protective effect of the NADPH oxidase inhibitor, diphenyleneiodonium (DPI) against necroptosis in renal tubular epithelial cells. A necroptosis model of HK-2 cells was established using tumor necrosis factor-α, benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone and antimycin A (collectively termed TZA), as in our previous research. The necroptosis inhibitor, necrostatin-1 (Nec-1) or the NADPH oxidase inhibitor, DPI were administered to the necroptosis model. Production of reactive oxygen species (ROS) was detected by dichlorodihydrofluorescein diacetate in the different groups, and the manner of cell death was identified by flow cytometry. Western blot analysis was used to determine the levels of phosphorylation of receptor-interacting protein kinase 3 (RIP-3) and mixed lineage kinase domain-like (MLKL), which are essential to necroptosis. The results revealed that TZA increased the percentages of propidium iodide-positive HK-2 cells from 1.22±0.69 to 8.98±0.73% (P<0.001), and augmented the phosphorylation of RIP-3 and MLKL. ROS levels were increased in the TZA group compared with the control group (27.74±1.60×104 vs. 18.51±1.10×104, respectively; P<0.001), and could be inhibited by Nec-1 (TZA + Nec-1 group, 22.90±2.22×104 vs. TZA group, 27.74±1.60×104; P=0.01). DPI decreased ROS production (TZA + DPI group, 22.13±1.86×104 vs. TZA group, 27.74±1.60×104; P<0.001) and also reduced the proportions of necrosis in the necroptosis model (TZA + DPI group, 4.40±1.51% vs. TZA group, 8.98±0.73%; P<0.001). Phosphorylated RIP-3 and MLKL were also decreased by DPI treatment. The results indicate that ROS production increases in HK-2 cells undergoing necroptosis, and that the NADPH oxidase inhibitor, DPI may protect HK-2 cells from necroptosis via inhibition of ROS production.
Collapse
Affiliation(s)
- Wei Dong
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Zhilian Li
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Yuanhan Chen
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Li Zhang
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Zhiming Ye
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Huaban Liang
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Ruizhao Li
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Lixia Xu
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Bin Zhang
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Shuangxin Liu
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Weidong Wang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Chunling Li
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Jialun Luo
- Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Wei Shi
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Xinling Liang
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|