1
|
Shi K, Xiao Y, Qu M, Xie Y, Wang Y, Ke C, Qu L, Liu Y. Atractylodin modulates ASAH3L to improve galactose metabolism and inflammation to alleviate acute lung injury. iScience 2024; 27:110751. [PMID: 39351199 PMCID: PMC11440247 DOI: 10.1016/j.isci.2024.110751] [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: 04/07/2024] [Revised: 05/30/2024] [Accepted: 08/13/2024] [Indexed: 10/04/2024] Open
Abstract
Acute lung injury (ALI) is a lung disease characterized by an excessive inflammatory response and damage to lung epithelial cells. Atractylodin (ATL) has good anti-inflammatory activity and protects the integrity of the epithelial cell barrier. However, the efficacy of ATL in the treatment of ALI and its mechanism is unclear. We investigated the efficacy of ATL in treating ALI and explored its targets and mechanisms. The results showed that ATL significantly reduced the wet-dry ratio of lungs of rats with ALI, improved the pathological changes, and lowered the expression of the inflammatory factors. Combined metabolomic and transcriptomic analyses showed that ATL can reduce inflammation by inhibiting and activating the HIF-1 signaling pathway and modulating ASAH3L to improve galactose metabolism, thereby alleviating ALI. In conclusion, ATL may be a potential drug for the treatment of acute lung injury.
Collapse
Affiliation(s)
- Kun Shi
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
- Center for Hubei TCM Processing Technology Engineering, Wuhan 430065, China
- Hubei Shizhen Laboratory, Wuhan 430065, China
| | - Yangxin Xiao
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
- Center for Hubei TCM Processing Technology Engineering, Wuhan 430065, China
- Hubei Shizhen Laboratory, Wuhan 430065, China
| | - Mumujiang Qu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Ying Xie
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Yan Wang
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Chang Ke
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Linghang Qu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
- Center for Hubei TCM Processing Technology Engineering, Wuhan 430065, China
- Hubei Shizhen Laboratory, Wuhan 430065, China
| | - Yanju Liu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
- Center for Hubei TCM Processing Technology Engineering, Wuhan 430065, China
- Hubei Shizhen Laboratory, Wuhan 430065, China
| |
Collapse
|
2
|
Attar GS, Kumar M, Bhalla V. Targeting sub-cellular organelles for boosting precision photodynamic therapy. Chem Commun (Camb) 2024; 60:11610-11624. [PMID: 39320942 DOI: 10.1039/d4cc02702g] [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: 09/26/2024]
Abstract
Among various cancer treatment methods, photodynamic therapy has received significant attention due to its non-invasiveness and high efficiency in inhibiting tumour growth. Recently, specific organelle targeting photosensitizers have received increasing interest due to their precise accumulation and ability to trigger organelle-mediated cell death signalling pathways, which greatly reduces the drug dosage, minimizes toxicity, avoids multidrug resistance, and prevents recurrence. In this review, recent advances and representative photosensitizers used in targeted photodynamic therapy on organelles, specifically including the endoplasmic reticulum, Golgi apparatus, mitochondria, nucleus, and lysosomes, have been comprehensively reviewed with a focus on organelle structure and organelle-mediated cell death signalling pathways. Furthermore, a perspective on future research and potential challenges in precision photodynamic therapy has been presented at the end.
Collapse
Affiliation(s)
- Gopal Singh Attar
- Department of chemistry UGC Sponsored-Centre for Advanced Studies-I, Guru Nanak Dev University, Amritsar-143005, Punjab, India.
| | - Manoj Kumar
- Department of chemistry UGC Sponsored-Centre for Advanced Studies-I, Guru Nanak Dev University, Amritsar-143005, Punjab, India.
| | - Vandana Bhalla
- Department of chemistry UGC Sponsored-Centre for Advanced Studies-I, Guru Nanak Dev University, Amritsar-143005, Punjab, India.
| |
Collapse
|
3
|
Zhu L, Du Y. A promising new approach to cancer therapy: Manipulate ferroptosis by hijacking endogenous iron. Int J Pharm 2024; 662:124517. [PMID: 39084581 DOI: 10.1016/j.ijpharm.2024.124517] [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: 04/08/2024] [Revised: 07/12/2024] [Accepted: 07/24/2024] [Indexed: 08/02/2024]
Abstract
Ferroptosis, a form of regulated cell death characterized by iron-dependent phospholipid peroxidation, has emerged as a focal point in the field of cancer therapy. Compared with other cell death modes such as apoptosis and necrosis, ferroptosis exhibits many distinct characteristics in the molecular mechanisms and cell morphology, offering a promising avenue for combating cancers that are resistant to conventional therapeutic modalities. In light of the serious side effects associated with current Fenton-modulating ferroptosis therapies utilizing exogenous iron-based inorganic nanomaterials, hijacking endogenous iron could serve as an effective alternative strategy to trigger ferroptosis through targeting cellular iron regulatory mechanisms. A better understanding of the underlying iron regulatory mechanism in the process of ferroptosis has shed light on the current findings of endogenous ferroptosis-based nanomedicine strategies for cancer therapy. Here in this review article, we provide a comprehensive discussion on the regulatory network of iron metabolism and its pivotal role in ferroptosis, and present recent updates on the application of nanoparticles endowed with the ability to hijack endogenous iron for ferroptosis. We envision that the insights in the study may expedite the development and translation of endogenous ferroptosis-based nanomedicines for effective cancer treatment.
Collapse
Affiliation(s)
- Luwen Zhu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yongzhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Jinhua Institute of Zhejiang University, Jinhua, Zhejiang 321299, China.
| |
Collapse
|
4
|
Agrawal HG, Khatun S, Rengan AK, Mishra AK. Tuning the Flavin Core via Donor Appendage for Selective Subcellular Bioimaging and PDT Application. Chemistry 2024; 30:e202401483. [PMID: 38853431 DOI: 10.1002/chem.202401483] [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: 04/16/2024] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 06/11/2024]
Abstract
Herein, we report a novel flavin analogue as singular chemical component for lysosome bioimaging, and inherited photosensitizer capability of the flavin core was demonstrated as a promising candidate for photodynamic therapy (PDT) application. Fine-tuning the flavin core with the incorporation of methoxy naphthyl appendage provides an appropriate chemical design, thereby offering photostability, selectivity, and lysosomal colocalization, along with the aggregation-induced emissive nature, making it suitable for lysosomal bioimaging applications. Additionally, photosensitization capability of the flavin core with photostable nature of the synthesized analogue has shown remarkable capacity for generating reactive oxygen species (ROS) within cells, making it a promising candidate for photodynamic therapy (PDT) application.
Collapse
Affiliation(s)
- Harsha Gopal Agrawal
- Department of Chemistry, Indian Institute of Technology, Hyderabad, Sangareddy, Telangana, 502285, India
| | - Sajmina Khatun
- Department of Biomedical Engineering, Indian Institute of Technology, Hyderabad, Sangareddy, Telangana, 502285, India
| | - Aravind Kumar Rengan
- Department of Biomedical Engineering, Indian Institute of Technology, Hyderabad, Sangareddy, Telangana, 502285, India
| | - Ashutosh Kumar Mishra
- Department of Chemistry, Indian Institute of Technology, Hyderabad, Sangareddy, Telangana, 502285, India
| |
Collapse
|
5
|
Manivannan MS, Yang X, Patel N, Peters A, Johnston JB, Gibson SB. Lysosome-Disrupting Agents in Combination with Venetoclax Increase Apoptotic Response in Primary Chronic Lymphocytic Leukemia (CLL) Cells Mediated by Lysosomal Cathepsin D Release and Inhibition of Autophagy. Cells 2024; 13:1041. [PMID: 38920669 PMCID: PMC11202145 DOI: 10.3390/cells13121041] [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: 03/18/2024] [Revised: 06/10/2024] [Accepted: 06/13/2024] [Indexed: 06/27/2024] Open
Abstract
Venetoclax and obinutuzumab are becoming frontline therapies for chronic lymphocytic leukemia (CLL) patients. Unfortunately, drug resistance still occurs, and the combination could be immunosuppressive. Lysosomes have previously been identified as a target for obinutuzumab cytotoxicity in CLL cells, but the mechanism remains unclear. In addition, studies have shown that lysosomotropic agents can cause synergistic cell death in vitro when combined with the BTK inhibitor, ibrutinib, in primary CLL cells. This indicates that targeting lysosomes could be a treatment strategy for CLL. In this study, we have shown that obinutuzumab induces lysosome membrane permeabilization (LMP) and cathepsin D release in CLL cells. Inhibition of cathepsins reduced obinutuzumab-induced cell death in CLL cells. We further determined that the lysosomotropic agent siramesine in combination with venetoclax increased cell death in primary CLL cells through an increase in reactive oxygen species (ROS) and cathepsin release. Siramesine treatment also induced synergistic cytotoxicity when combined with venetoclax. Microenvironmental factors IL4 and CD40L or incubation with HS-5 stromal cells failed to significantly protect CLL cells from siramesine- and venetoclax-induced apoptosis. We also found that siramesine treatment inhibited autophagy through reduced autolysosomes. Finally, the autophagy inhibitor chloroquine failed to further increase siramesine-induced cell death. Taken together, lysosome-targeting drugs could be an effective strategy in combination with venetoclax to overcome drug resistance in CLL.
Collapse
MESH Headings
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Sulfonamides/pharmacology
- Lysosomes/metabolism
- Lysosomes/drug effects
- Apoptosis/drug effects
- Autophagy/drug effects
- Cathepsin D/metabolism
- Reactive Oxygen Species/metabolism
- Drug Synergism
- Cell Line, Tumor
Collapse
Affiliation(s)
- Madhumita S. Manivannan
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada; (M.S.M.); (X.Y.); (N.P.); (A.P.)
| | - Xiaoyan Yang
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada; (M.S.M.); (X.Y.); (N.P.); (A.P.)
| | - Nirav Patel
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada; (M.S.M.); (X.Y.); (N.P.); (A.P.)
| | - Anthea Peters
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada; (M.S.M.); (X.Y.); (N.P.); (A.P.)
- Cross Cancer Institute, Alberta Health Services, Edmonton, AB T5J 3E4, Canada
| | - James B. Johnston
- CancerCare Manitoba Research Institute, Hematologist/Oncologist, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada;
| | - Spencer B. Gibson
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada; (M.S.M.); (X.Y.); (N.P.); (A.P.)
- Department of Internal Medicine, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Department of Biochemistry and Medical Genetics, Faculty of Medicine, University of Manitoba, Winnipeg, MB R3T 2N2, USA
| |
Collapse
|
6
|
Lieu AS, Pan YC, Lee JH, Hsieh YC, Lin CJ, Hsu YL, Chang KC, Kuo SH, Tseng TT, Tsai HP. Antitumor Efficacy of Arylquin 1 through Dose-Dependent Cytotoxicity, Apoptosis Induction, and Synergy with Radiotherapy in Glioblastoma Models. Biomedicines 2024; 12:907. [PMID: 38672261 PMCID: PMC11048020 DOI: 10.3390/biomedicines12040907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Glioblastoma (GBM), the most aggressive form of brain cancer, is characterized by rapid growth and resistance to conventional therapies. Current treatments offer limited effectiveness, leading to poor survival rates and the need for novel therapeutic strategies. Arylquin 1 has emerged as a potential therapeutic candidate because of its unique mechanism of inducing apoptosis in cancer cells without affecting normal cells. This study investigated the efficacy of Arylquin 1 against GBM using the GBM8401 and A172 cells by assessing its dose-dependent cytotoxicity, apoptosis induction, and synergy with radiotherapy. In vitro assays demonstrated a significant reduction in cell viability and increased apoptosis, particularly at high concentrations of Arylquin 1. Migration and invasion analyses revealed notable inhibition of cellular motility. In vivo experiments on NU/NU nude mice with intracranially implanted GBM cells revealed that Arylquin 1 substantially reduced tumor growth, an effect magnified by concurrent radiotherapy. These findings indicate that by promoting apoptosis and enhancing radiosensitivity, Arylquin 1 is a potent therapeutic option for GBM treatment.
Collapse
Affiliation(s)
- Ann-Shung Lieu
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan; (A.-S.L.); (T.-T.T.)
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
| | - Yu-Chi Pan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80756, Taiwan; (Y.-C.P.); (Y.-L.H.)
| | - Jia-Hau Lee
- National Institute of Cancer Research, National Health Research Institutes, Tainan 70456, Taiwan;
| | - Yuan-Chin Hsieh
- School of Medicine for International Students, I-Shou University, Kaoshiung 82445, Taiwan;
| | - Chien-Ju Lin
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80756, Taiwan;
| | - Ya-Ling Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80756, Taiwan; (Y.-C.P.); (Y.-L.H.)
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
| | - Kung-Chao Chang
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan;
| | - Shih-Hsun Kuo
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan;
| | - Tzu-Ting Tseng
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan; (A.-S.L.); (T.-T.T.)
| | - Hung-Pei Tsai
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan; (A.-S.L.); (T.-T.T.)
| |
Collapse
|
7
|
Liu X, Sun K, Yang H, Zou D, Xia L, Lu K, Meng X, Li Y. Molecular subtype identification and prognosis stratification based on lysosome-related genes in breast cancer. Heliyon 2024; 10:e25643. [PMID: 38420434 PMCID: PMC10900431 DOI: 10.1016/j.heliyon.2024.e25643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 01/13/2024] [Accepted: 01/31/2024] [Indexed: 03/02/2024] Open
Abstract
Background Lysosomes are known to have a significant impact on the development and recurrence of breast cancer. However, the association between lysosome-related genes (LRGs) and breast cancer remains unclear. This study aims to explore the potential role of LRGs in predicting the prognosis and treatment response of breast cancer. Methods Breast cancer gene expression profile data and clinical information were downloaded from TCGA and GEO databases, and prognosis-related LRGs were screened for consensus clustering analysis. Lasso Cox regression analysis was used to construct risk features derived from LRGs, and immune cell infiltration, immune therapy response, drug sensitivity, and clinical pathological feature differences were evaluated for different molecular subtypes and risk groups. A nomogram based on risk features derived from LRGs was constructed and evaluated. Results Our study identified 176 differentially expressed LRGs that are associated with breast cancer prognosis. Based on these genes, we divided breast cancer into two molecular subtypes with significant prognostic differences. We also found significant differences in immune cell infiltration between these subtypes. Furthermore, we constructed a prognostic risk model consisting of 7 LRGs, which effectively divides breast cancer patients into high-risk and low-risk groups. Patients in the low-risk group have better prognostic characteristics, respond better to immunotherapy, and have lower sensitivity to chemotherapy drugs, indicating that the low-risk group is more likely to benefit from immunotherapy and chemotherapy. Additionally, the risk score based on LRGs is significantly correlated with immune cell infiltration, including CD8 T cells and macrophages. This risk score model, along with age, chemotherapy, clinical stage, and N stage, is an independent prognostic factor for breast cancer. Finally, the nomogram composed of these factors has excellent performance in predicting overall survival of breast cancer. Conclusions In conclusion, this study has constructed a novel LRG-derived breast cancer risk feature, which performs well in prognostic prediction when combined with clinical pathological features.
Collapse
Affiliation(s)
- Xiaozhen Liu
- General Surgery, Cancer Center, Department of Breast Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Kewang Sun
- General Surgery, Cancer Center, Department of Breast Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Hongjian Yang
- Department of Breast Surgery, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China
| | - Dehomg Zou
- Department of Breast Surgery, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China
| | - Lingli Xia
- Department of Breast Surgery, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China
| | - Kefeng Lu
- Department of Outpatient Service, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Xuli Meng
- General Surgery, Cancer Center, Department of Breast Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Yongfeng Li
- General Surgery, Cancer Center, Department of Breast Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| |
Collapse
|
8
|
Chen Y, Zhu S, Liao T, Wang C, Han J, Yang Z, Lu X, Hu Z, Hu J, Wang X, Gu M, Gao R, Liu K, Liu X, Ding C, Hu S, Liu X. The HN protein of Newcastle disease virus induces cell apoptosis through the induction of lysosomal membrane permeabilization. PLoS Pathog 2024; 20:e1011981. [PMID: 38354122 PMCID: PMC10866534 DOI: 10.1371/journal.ppat.1011981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 01/17/2024] [Indexed: 02/16/2024] Open
Abstract
Lysosomes are acidic organelles that mediate the degradation and recycling of cellular waste materials. Damage to lysosomes can cause lysosomal membrane permeabilization (LMP) and trigger different types of cell death, including apoptosis. Newcastle disease virus (NDV) can naturally infect most birds. Additionally, it serves as a promising oncolytic virus known for its effective infection of tumor cells and induction of intensive apoptotic responses. However, the involvement of lysosomes in NDV-induced apoptosis remains poorly understood. Here, we demonstrate that NDV infection profoundly triggers LMP, leading to the translocation of cathepsin B and D and subsequent mitochondria-dependent apoptosis in various tumor and avian cells. Notably, the released cathepsin B and D exacerbate NDV-induced LMP by inducing the generation of reactive oxygen species. Additionally, we uncover that the viral Hemagglutinin neuraminidase (HN) protein induces the deglycosylation and degradation of lysosome-associated membrane protein 1 (LAMP1) and LAMP2 dependent on its sialidase activity, which finally contributes to NDV-induced LMP and cellular apoptosis. Overall, our findings elucidate the role of LMP in NDV-induced cell apoptosis and provide novel insights into the function of HN during NDV-induced LMP, which provide innovative approaches for the development of NDV-based oncolytic agents.
Collapse
Affiliation(s)
- Yu Chen
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University; Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
| | - Shanshan Zhu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University; Yangzhou, Jiangsu, China
| | - Tianxing Liao
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University; Yangzhou, Jiangsu, China
| | - Chunxuan Wang
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University; Yangzhou, Jiangsu, China
| | - Jiajun Han
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University; Yangzhou, Jiangsu, China
| | - Zhenyu Yang
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University; Yangzhou, Jiangsu, China
| | - Xiaolong Lu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University; Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
| | - Zenglei Hu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Jiao Hu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University; Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
| | - Xiaoquan Wang
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University; Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
| | - Min Gu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University; Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
| | - Ruyi Gao
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University; Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
| | - Kaituo Liu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Xiaowen Liu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University; Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
| | - Chan Ding
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Shunlin Hu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University; Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
| | - Xiufan Liu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University; Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
| |
Collapse
|
9
|
Halliwell B. Understanding mechanisms of antioxidant action in health and disease. Nat Rev Mol Cell Biol 2024; 25:13-33. [PMID: 37714962 DOI: 10.1038/s41580-023-00645-4] [Citation(s) in RCA: 100] [Impact Index Per Article: 100.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2023] [Indexed: 09/17/2023]
Abstract
Several different reactive oxygen species (ROS) are generated in vivo. They have roles in the development of certain human diseases whilst also performing physiological functions. ROS are counterbalanced by an antioxidant defence network, which functions to modulate ROS levels to allow their physiological roles whilst minimizing the oxidative damage they cause that can contribute to disease development. This Review describes the mechanisms of action of antioxidants synthesized in vivo, antioxidants derived from the human diet and synthetic antioxidants developed as therapeutic agents, with a focus on the gaps in our current knowledge and the approaches needed to close them. The Review also explores the reasons behind the successes and failures of antioxidants in treating or preventing human disease. Antioxidants may have special roles in the gastrointestinal tract, and many lifestyle features known to promote health (especially diet, exercise and the control of blood glucose and cholesterol levels) may be acting, at least in part, by antioxidant mechanisms. Certain reactive sulfur species may be important antioxidants but more accurate determinations of their concentrations in vivo are needed to help assess their contributions.
Collapse
Affiliation(s)
- Barry Halliwell
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Neurobiology Research Programme, Life Sciences Institute, Centre for Life Sciences, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
10
|
Shirbhate E, Singh V, Mishra A, Jahoriya V, Veerasamy R, Tiwari AK, Rajak H. Targeting Lysosomes: A Strategy Against Chemoresistance in Cancer. Mini Rev Med Chem 2024; 24:1449-1468. [PMID: 38343053 DOI: 10.2174/0113895575287242240129120002] [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: 10/13/2023] [Revised: 01/11/2024] [Accepted: 01/19/2024] [Indexed: 07/23/2024]
Abstract
Chemotherapy is still the major method of treatment for many types of cancer. Curative cancer therapy is hampered significantly by medication resistance. Acidic organelles like lysosomes serve as protagonists in cellular digestion. Lysosomes, however, are gaining popularity due to their speeding involvement in cancer progression and resistance. For instance, weak chemotherapeutic drugs of basic nature permeate through the lysosomal membrane and are retained in lysosomes in their cationic state, while extracellular release of lysosomal enzymes induces cancer, cytosolic escape of lysosomal hydrolases causes apoptosis, and so on. Drug availability at the sites of action is decreased due to lysosomal drug sequestration, which also enhances cancer resistance. This review looks at lysosomal drug sequestration mechanisms and how they affect cancer treatment resistance. Using lysosomes as subcellular targets to combat drug resistance and reverse drug sequestration is another method for overcoming drug resistance that is covered in this article. The present review has identified lysosomal drug sequestration as one of the reasons behind chemoresistance. The article delves deeper into specific aspects of lysosomal sequestration, providing nuanced insights, critical evaluations, or novel interpretations of different approaches that target lysosomes to defect cancer.
Collapse
Affiliation(s)
- Ekta Shirbhate
- Department of Pharmacy, Guru Ghasidas University, Bilaspur-495 009, (C.G.), India
| | - Vaibhav Singh
- Department of Pharmacy, Guru Ghasidas University, Bilaspur-495 009, (C.G.), India
| | - Aditya Mishra
- Department of Pharmacy, Guru Ghasidas University, Bilaspur-495 009, (C.G.), India
| | - Varsha Jahoriya
- Department of Pharmacy, Guru Ghasidas University, Bilaspur-495 009, (C.G.), India
| | - Ravichandran Veerasamy
- Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Kedah Darul Aman, Malaysia
| | - Amit K Tiwari
- UAMS College of Pharmacy; UAMS - University of Arkansas for Medical Sciences, (AR) USA
| | - Harish Rajak
- Department of Pharmacy, Guru Ghasidas University, Bilaspur-495 009, (C.G.), India
| |
Collapse
|
11
|
Li H, Li J, Qu X, Dai H, Liu J, Ma M, Wang J, Dong W, Wang W. Establishment and validation of a novel lysosome-related gene signature for predicting prognosis and immune landscape in hepatocellular carcinoma. J Cancer Res Clin Oncol 2023; 149:17543-17557. [PMID: 37903936 DOI: 10.1007/s00432-023-05477-5] [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: 06/12/2023] [Accepted: 10/12/2023] [Indexed: 11/01/2023]
Abstract
BACKGROUND Recent studies have shown that lysosomes not only provide energy for tumor cell growth, but also participate in the occurrence and development of malignant tumors by regulating various ways of tumor cell death. However, the role of lysosome associated genes (LSAGs) in hepatocellular carcinoma (HCC) remains unclear. METHODS Transcriptome data and clinical data of HCC were downloaded from the Cancer Genome Atlas (TCGA) and the International Cancer Genome Consortium (ICGC) databases. We identified differential expression of LSAGs by comparing tumor tissue with normal liver tissue. Subsequently, we used univariate COX analysis and least absolute shrinkage and selection operator (LASSO) COX regression to construct the prognostic feature of LSAGs. Kaplan-Meier survival curve and receiver operating characteristic curve were used to evaluate the predictive ability of LSAGs feature. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were used for functional enrichment analysis of risk differential genes. The relationship between LSAGs score and tumor microenvironment and chemotherapy drug sensitivity was analyzed. Finally, the cellular communication of tumor cells with high and low expression of model LSAGs was explored. RESULTS We identified sixteen prognostic associated LSAGs, four of which were selected to construct prognostic feature of LSAGs. Patients in the low LSAGs group had a better prognosis than those in the high LSAGs group. GO and KEGG analyses showed that risk differential genes were enriched in leukocyte migration, cytokine-cytokine receptor interaction and PI3K-Akt signaling pathway. The group with low LSAGs score had lower immune score. Patients in the high LSAGs group were more sensitive to drugs for chemotherapy. In addition, tumor cells with high expression of model LSAGs showed stronger association with immune cells through the interleukin-2 (IL2), fibroblast growth factor (FGF), adiponectin, and bone morphogenetic proteins (BMP) signaling pathways. CONCLUSION We established a LSAGs signature that had the ability to predict clinical prognosis and immune landscape, proposing potential therapeutic targets for HCC.
Collapse
Affiliation(s)
- Haoling Li
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Anhui, 233030, China
- Department of Clinical Medicine, Bengbu Medical College, Anhui, 233030, China
| | - Jing Li
- Department of Pharmacy, Shanghai Pudong New Area People's Hospital, Shanghai, 201299, China
| | - Xiangyu Qu
- Department of Clinical Medicine, Bengbu Medical College, Anhui, 233030, China
| | - Hengwen Dai
- Department of Clinical Medicine, Bengbu Medical College, Anhui, 233030, China
| | - Junjie Liu
- Department of Clinical Medicine, Bengbu Medical College, Anhui, 233030, China
| | - Mengxi Ma
- Department of Clinical Medicine, Bengbu Medical College, Anhui, 233030, China
| | - Jian Wang
- Department of Pharmacy, Shanghai Pudong New Area People's Hospital, Shanghai, 201299, China.
| | - Wei Dong
- Department of Hepatic Surgery, The Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China.
| | - Wenrui Wang
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Anhui, 233030, China.
- Department of Biotechnology, School of Life Sciences, Bengbu Medical College, Anhui, 233030, China.
| |
Collapse
|
12
|
Duan X, Tong Q, Fu C, Chen L. Lysosome-targeted fluorescent probes: Design mechanism and biological applications. Bioorg Chem 2023; 140:106832. [PMID: 37683542 DOI: 10.1016/j.bioorg.2023.106832] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/22/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023]
Abstract
As an integral organelle in the eukaryote, the lysosome is the degradation center and metabolic signal center in living cells, and partakes in significant physiological processes such as autophagy, cell death and cellular senescence. Fluorescent probe has become a favorite tool for studying organelles and their chemical microenvironments because of its high specificity and non-destructive merits. Over recent years, it has been reported that increasingly new lysosome-targeted probes play a major role in the diagnosis and monitor of diseases, in particular cancer and neurodegenerative diseases. In order to deepen the relevant research on lysosome, it is challenging and inevitability to design novel lysosomal targeting probes. This review first introduces the concepts of lysosome and its closely related biological activities, and then introduces the fluorescent probes for lysosome in detail according to different detection targets, including targeting mechanism, biological imaging, and application in diseases. Finally, we summarize the specific challenges and discuss the future development direction facing the current lysosome-targeted fluorescent probes. We hope that this review can help biologists grasp the application of fluorescent probes and broaden the research ideas of researchers targeting fluorescent probes so as to design more accurate and functional probes for application in diseases.
Collapse
Affiliation(s)
- Xiangning Duan
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, Hunan, China
| | - Qin Tong
- The First Affiliated Hospital, Department of Oncology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Chengxiao Fu
- The First Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Linxi Chen
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, Hunan, China.
| |
Collapse
|
13
|
Meng Q, Ding B, Ma P, Lin J. Interrelation between Programmed Cell Death and Immunogenic Cell Death: Take Antitumor Nanodrug as an Example. SMALL METHODS 2023; 7:e2201406. [PMID: 36707416 DOI: 10.1002/smtd.202201406] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/20/2022] [Indexed: 05/17/2023]
Abstract
Programmed cell death (PCD, mainly including apoptosis, necrosis, ferroptosis, pyroptosis, and autophagy) and immunogenic cell death (ICD), as important cell death mechanisms, are widely reported in cancer therapy, and understanding the relationship between the two is significant for clinical tumor treatments. Considering that vast nanodrugs are developed to induce tumor PCD and ICD simultaneously, in this review, the interrelationship between PCD and ICD is described using nanomedicines as examples. First, an overview of PCD patterns and focus on the morphological differences and interconnections among them are provided. Then the interrelationship between apoptosis and ICD in terms of endoplasmic reticulum stress is described by introducing various cancer treatments and the recent developments of nanomedicines with inducible immunogenicity. Next, the crosstalk between non-apoptotic (including necrosis, ferroptosis, pyroptosis, and autophagy) signaling pathways and ICD is introduced and their relationship through various nanomedicines as examples is further illustrated. Finally, the relationship between PCD and ICD and its application prospects in the development of new ICD nanomaterials are summarized. This review is believed to deepen the understanding of the relationship between PCD and ICD, extend the biomedical applications of various nanodrugs, and promote the progress of clinical tumor therapy.
Collapse
Affiliation(s)
- Qi Meng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Binbin Ding
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Ping'an Ma
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| |
Collapse
|
14
|
Chen Y, Wang S, Ma Q, Wu X, Guo Q, Luo X, Tao L, Shen X. Utilizing endosomal capture for tumor therapy via membrane-lytic mechanism-based Pickering emulsion. J Control Release 2023; 354:523-537. [PMID: 36657600 DOI: 10.1016/j.jconrel.2023.01.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/21/2023]
Abstract
Nanocarriers are easily captured by endosomes, where the abundant hydrolases inevitably destroy the nanocarriers and the drugs they carry, ultimately resulting in a compromised or lost therapeutic efficacy. Herein, we report a membrane-lytic mechanism-based Pickering emulsion that can in turn utilize this seemingly unfavorable endosomal capture behavior for tumor therapy. This Pickering emulsion is constructed as an oil-in-water (O/W) emulsion stabilized by the hybrid nanoparticles (HNPs) composed of two molecules with opposite charges, cetyl trimethylamine bromide (CTAB) and linoleic acid (LA), through electrostatic interaction (defined as HNPs@PE). After HNPs@PE enters the lysosomes through macropinocytosis-mediated endocytosis, LA can be protonated in response to the acidic stimulus, and causing the swelling or disintegration of HNPs due to the disrupted electrostatic interaction. The released CTAB holds strong membrane-lytic activity and can directly damage the lysosomal membranes. Under the acidic condition and the participation of excessive iron ions (II) in lysosomes, LA induces lipid peroxidation and the resulting lipid peroxides (LPO) will oxidize the lysosomal membranes, collectively causing the leakage of lysosome membranes and the release of contents into cytoplasm. Subsequently, the diffused CTAB and LPO will continue to attack the mitochondrial membranes and cell membranes, resulting in the death of different types of tumor cells both in vitro and in vivo due to membrane damage. This Pickering emulsion with membrane-lytic ability represents a potential self-anticancer nanocarrier.
Collapse
Affiliation(s)
- Ying Chen
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China; The Department of Pharmacology of Materia Medical (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China.
| | - Sibu Wang
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China; The Department of Pharmacology of Materia Medical (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China
| | - Qin Ma
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China; The Department of Pharmacology of Materia Medical (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China
| | - Xingjie Wu
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China; The Department of Pharmacology of Materia Medical (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China
| | - Qianqian Guo
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China; The Department of Pharmacology of Materia Medical (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China
| | - Xinghong Luo
- Jiangsu Simcere Pharmaceutical Co, Ltd., State Key Laboratory of Translational Medicine and Innovative Drug, 699-18 Xuanwu Avenue, Nanjing 210042,China
| | - Ling Tao
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China; The Department of Pharmacology of Materia Medical (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China.
| | - Xiangchun Shen
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China; The Department of Pharmacology of Materia Medical (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China.
| |
Collapse
|
15
|
Chen F, Kang R, Tang D, Liu J. Monitoring Lysosome Function in Ferroptosis. Methods Mol Biol 2023; 2712:91-102. [PMID: 37578699 DOI: 10.1007/978-1-0716-3433-2_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Ferroptosis is a type of regulated cell death that occurs due to iron-induced membrane lipid peroxidation. Lysosomes, which are acidic, membrane-bound organelles containing various hydrolases, play a vital role in ferroptosis. They not only aid in the degradation of autophagic substrates, but also serve as signaling hubs in cell death. Specifically, lysosomes are involved in the induction and execution of ferroptosis through autophagy-mediated degradation of anti-ferroptotic proteins, lysosomal membrane permeability-mediated release of cathepsins, and iron-induced lysosomal membrane lipid peroxidation. Therefore, it is essential to have reliable methods for monitoring lysosomal functions, including lysosomal activity, pH, and membrane integrity, as well as iron accumulation and lipid peroxidation, to understand ferroptosis. This chapter introduces several protocols, such as western blotting, immunofluorescence, lysosomal probes, and lipid peroxidation assay kits, for monitoring the process of lysosome-related ferroptosis.
Collapse
Affiliation(s)
- Fangquan Chen
- DAMP Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jiao Liu
- DAMP Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
16
|
Chen Y, Yang Z, Wang S, Ma Q, Li L, Wu X, Guo Q, Tao L, Shen X. Boosting ROS-Mediated Lysosomal Membrane Permeabilization for Cancer Ferroptosis Therapy. Adv Healthc Mater 2023; 12:e2202150. [PMID: 36408929 DOI: 10.1002/adhm.202202150] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/04/2022] [Indexed: 11/22/2022]
Abstract
Due to the deficient catalase, abundant reduced iron and low acidic environment in lysosomes, inducing lysosomal membrane permeabilization (LMP) through Fenton reaction-based reactive oxygen species (ROS) generation recently attracts increasing attention in cancer therapy. However, the lysosomal membranes are protected by highly glycosylated membrane proteins and several endolysosomal damage-response mechanisms can rapidly repair the injured lysosomes. To produce sufficient ROS and cause complete lysosomal membranes rupture, a lysosome-targeted ROS inducer, N-(3-Aminopropyl) morpholine grafted cross-linked lipoic acid vesicles with vitamin C-loading (VC@N3AM cLAVs), is developed. VC@N3AM cLAVs efficiently accumulate in lysosomes and convert into two redox couples LA/DHLA (dihydrolipoic acid, reduced form of LA) and VC/DHA (dehydroascorbic acid, oxidized form of VC) by the lysosomal glutathione, which can not only produce a large amount of H2 O2 by pro-oxidant action but also accelerate iron transformation through the cyclic redox reactions between each other and cause the efficient conversion of the generated H2 O2 into highly toxic •OH. Both in vitro and in vivo experiments demonstrate that VC@N3AM cLAVs can effectively enhance ROS production and boost LMP, finally initiation irreversible death of tumor cells via ferroptosis pathway, thus representing a potential anticancer drug for cancer therapy.
Collapse
Affiliation(s)
- Ying Chen
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang, 550025, China.,The Department of Pharmacology of Materia Medical (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), Guizhou Medical University, University Town, Guian New District, Guiyang, 550025, China
| | - Zengqiu Yang
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang, 550025, China.,The Department of Pharmacology of Materia Medical (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), Guizhou Medical University, University Town, Guian New District, Guiyang, 550025, China
| | - Sibu Wang
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang, 550025, China.,The Department of Pharmacology of Materia Medical (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), Guizhou Medical University, University Town, Guian New District, Guiyang, 550025, China
| | - Qin Ma
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang, 550025, China.,The Department of Pharmacology of Materia Medical (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), Guizhou Medical University, University Town, Guian New District, Guiyang, 550025, China
| | - Lingyan Li
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang, 550025, China.,The Department of Pharmacology of Materia Medical (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), Guizhou Medical University, University Town, Guian New District, Guiyang, 550025, China
| | - Xingjie Wu
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang, 550025, China.,The Department of Pharmacology of Materia Medical (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), Guizhou Medical University, University Town, Guian New District, Guiyang, 550025, China
| | - Qianqian Guo
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang, 550025, China.,The Department of Pharmacology of Materia Medical (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), Guizhou Medical University, University Town, Guian New District, Guiyang, 550025, China
| | - Ling Tao
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang, 550025, China.,The Department of Pharmacology of Materia Medical (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), Guizhou Medical University, University Town, Guian New District, Guiyang, 550025, China
| | - Xiangchun Shen
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang, 550025, China.,The Department of Pharmacology of Materia Medical (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), Guizhou Medical University, University Town, Guian New District, Guiyang, 550025, China
| |
Collapse
|
17
|
Shiralipour A, Khorsand B, Jafari L, Salehi M, Kazemi M, Zahiri J, Jajarmi V, Kazemi B. Identifying Key Lysosome-Related Genes Associated with Drug-Resistant Breast Cancer Using Computational and Systems Biology Approach. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2022; 21:e130342. [PMID: 36915401 PMCID: PMC10007991 DOI: 10.5812/ijpr-130342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/11/2022] [Accepted: 09/18/2022] [Indexed: 11/17/2022]
Abstract
Background Drug resistance in breast cancer is an unsolved problem in treating patients. It has been recently discussed that lysosomes contribute to the invasion and angiogenesis of cancer cells. There is evidence that lysosomes can also cause multi-drug resistance. We analyzed this emerging concept in breast cancer through computational and systems biology approaches. Objectives We aimed to identify the key lysosome-related genes associated with drug-resistant breast cancer. Methods All genes contributing to the structure and function of lysosomes were inquired through the Human Lysosome Gene Database. The prioritized top 51 genes from the provided lists of Endeavour, ToppGene, and GPSy as prioritization tools were selected. All lysosomal genes and 12 breast cancer-related genes aligned to identify the most similar genes to breast cancer-related genes. Different centralities were applied to score each human protein to calculate the most central lysosomal genes in the human protein-protein interaction (PPI) network. Common genes were extracted from the results of the mentioned methods as a selected gene set. For Gene Ontology enrichment, the selected gene set was analyzed by WebGestalt, DAVID, and KOBAS. The PPI network was constructed via the STRING database. The PPI network was analyzed utilizing Cytoscape for topology network interaction and CytoHubba to extract hub genes. Results Based on biological studies, literature reviews, and comparing all mentioned analyzing methods, six genes were introduced as essential in breast cancer. This computational approach to all lysosome-related genes suggested that candidate genes include PRF1, TLR9, CLTC, GJA1, AP3B1, and RPTOR. The analyses of these six genes suggest that they may have a crucial role in breast cancer development, which has rarely been evaluated. These genes have a potential therapeutic implication for new drug discovery for chemo-resistant breast cancer. Conclusions The present work focused on all the functional and structural lysosome-related genes associated with breast cancer. It revealed the top six lysosome hub genes that might serve as therapeutic targets in drug-resistant breast cancer. Since these genes play a pivotal role in the structure and function of lysosomes, targeting them can effectively overcome drug resistance.
Collapse
Affiliation(s)
- Aref Shiralipour
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Babak Khorsand
- Computer Engineering Department, Faculty of Engineering, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Leila Jafari
- Bioinformatics and Computational Omics Lab (BioCOOL), Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University (TMU), Tehran, Iran
| | - Mohammad Salehi
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahsa Kazemi
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Javad Zahiri
- Department of Neurosciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0662, USA
| | - Vahid Jajarmi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Corresponding Author: Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Bahram Kazemi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Corresponding Author: Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
18
|
Yang J, Griffin A, Qiang Z, Ren J. Organelle-targeted therapies: a comprehensive review on system design for enabling precision oncology. Signal Transduct Target Ther 2022; 7:379. [PMID: 36402753 PMCID: PMC9675787 DOI: 10.1038/s41392-022-01243-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/19/2022] [Accepted: 10/25/2022] [Indexed: 11/21/2022] Open
Abstract
Cancer is a major threat to human health. Among various treatment methods, precision therapy has received significant attention since the inception, due to its ability to efficiently inhibit tumor growth, while curtailing common shortcomings from conventional cancer treatment, leading towards enhanced survival rates. Particularly, organelle-targeted strategies enable precise accumulation of therapeutic agents in organelles, locally triggering organelle-mediated cell death signals which can greatly reduce the therapeutic threshold dosage and minimize side-effects. In this review, we comprehensively discuss history and recent advances in targeted therapies on organelles, specifically including nucleus, mitochondria, lysosomes and endoplasmic reticulum, while focusing on organelle structures, organelle-mediated cell death signal pathways, and design guidelines of organelle-targeted nanomedicines based on intervention mechanisms. Furthermore, a perspective on future research and clinical opportunities and potential challenges in precision oncology is presented. Through demonstrating recent developments in organelle-targeted therapies, we believe this article can further stimulate broader interests in multidisciplinary research and technology development for enabling advanced organelle-targeted nanomedicines and their corresponding clinic translations.
Collapse
Affiliation(s)
- Jingjing Yang
- grid.24516.340000000123704535Institute of Nano and Biopolymeric Materials, School of Materials Science and Engineering, Tongji University, 201804 Shanghai, China
| | - Anthony Griffin
- grid.267193.80000 0001 2295 628XSchool of Polymer Science and Engineering, University of Southern Mississippi, Hattiesburg, MS 39406 USA
| | - Zhe Qiang
- grid.267193.80000 0001 2295 628XSchool of Polymer Science and Engineering, University of Southern Mississippi, Hattiesburg, MS 39406 USA
| | - Jie Ren
- grid.24516.340000000123704535Institute of Nano and Biopolymeric Materials, School of Materials Science and Engineering, Tongji University, 201804 Shanghai, China
| |
Collapse
|
19
|
Garcia EA, Bhatti I, Henson ES, Gibson SB. Prostate Cancer Cells Are Sensitive to Lysosomotropic Agent Siramesine through Generation Reactive Oxygen Species and in Combination with Tyrosine Kinase Inhibitors. Cancers (Basel) 2022; 14:cancers14225478. [PMID: 36428570 PMCID: PMC9688505 DOI: 10.3390/cancers14225478] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Prostate cancer is the most common cancer affecting men often resulting in aggressive tumors with poor prognosis. Even with new treatment strategies, drug resistance often occurs in advanced prostate cancers. The use of lysosomotropic agents offers a new treatment possibility since they disrupt lysosomal membranes and can trigger a series of events leading to cell death. In addition, combining lysosomotropic agents with targeted inhibitors can induce increased cell death in different cancer types, but prostate cancer cells have not been investigated. METHODS We treated prostate cancer cells with lysosomotropic agents and determine their cytotoxicity, lysosome membrane permeabilization (LMP), reactive oxygen species (ROS) levels, and mitochondrial dysfunction. In addition, we treated cells with lysosomotropic agent in combination with tyrosine kinase inhibitor, lapatinib, and determined cell death, and the role of ROS in this cell death. RESULTS Herein, we found that siramesine was the most effective lysosomotropic agent at inducing LMP, increasing ROS, and inducing cell death in three different prostate cancer cell lines. Siramesine was also effective at increasing cell death in combination with the tyrosine kinase inhibitor, lapatinib. This increase in cell death was mediated by lysosome membrane permeabilization, an increased in ROS levels, loss of mitochondrial membrane potential and increase in mitochondrial ROS levels. The combination of siramesine and lapatinib induced apoptosis, cleavage of PARP and decreased expression of Bcl-2 family member Mcl-1. Furthermore, lipid peroxidation occurred with siramesine treatment alone or in combination with lapatinib. Treating cells with the lipid peroxidation inhibitor alpha-tocopherol resulted in reduced siramesine induced cell death alone or in combination with lapatinib. The combination of siramesine and lapatinib failed to increase cell death responses in normal prostate epithelial cells. CONCLUSIONS This suggests that lysomotropic agents such as siramesine in combination with tyrosine kinase inhibitors induces cell death mediated by ROS and could be an effective treatment strategy in advanced prostate cancer.
Collapse
Affiliation(s)
- Emily A. Garcia
- Department of Biochemistry and Medical Genetics, University of Manitoba Winnipeg, Winnipeg, MB R3T 2N2, Canada
- CancerCare Manitoba Research Institute, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Ilsa Bhatti
- Department of Oncology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Elizabeth S. Henson
- Department of Biochemistry and Medical Genetics, University of Manitoba Winnipeg, Winnipeg, MB R3T 2N2, Canada
- CancerCare Manitoba Research Institute, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Spencer B. Gibson
- Department of Biochemistry and Medical Genetics, University of Manitoba Winnipeg, Winnipeg, MB R3T 2N2, Canada
- CancerCare Manitoba Research Institute, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
- Department of Oncology, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Spencer Gibson, Department of Oncology, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Correspondence:
| |
Collapse
|
20
|
Zamyatnin AA, Gregory LC, Townsend PA, Soond SM. Beyond basic research: the contribution of cathepsin B to cancer development, diagnosis and therapy. Expert Opin Ther Targets 2022; 26:963-977. [PMID: 36562407 DOI: 10.1080/14728222.2022.2161888] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION In view of other candidate proteins from the cathepsin family of proteases holding great potential in being targeted during cancer therapy, the importance of Cathepsin B (CtsB) stands out as being truly exceptional. Based on its contribution to oncogenesis, its intimate connection with regulating apoptosis and modulating extracellular and intracellular functions through its secretion or compartmentalized subcellular localization, collectively highlight its complex molecular involvement with a myriad of normal and pathological regulatory processes. Despite its complex functional nature, CtsB is emerging as one of the few cathepsin proteases that has been extensively researched to yield tangible outcomes for cancer therapy. AREAS COVERED In this article, we review the scientific literature that has justified or shaped the importance of CtsB expression in cancer progression, from the perspective of highlighting a paradigm that is rapidly changing from basic research toward a broader clinical and translational context. EXPERT OPINION In doing so, we detail its maturation as a diagnostic marker through describing the development of CtsB-specific Activity-Based Probes, the rapid evolution of these toward a new generation of Prodrugs, and the evaluation of these in model systems for their therapeutic potential as anti-cancer agents in the clinic.
Collapse
Affiliation(s)
- Andrey A Zamyatnin
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK.,Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russian Federation.,Department of Biotechnology, Sirius University of Science and Technology, Sochi, Russia.,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Levy C Gregory
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Paul A Townsend
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK.,Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Surinder M Soond
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK.,Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| |
Collapse
|
21
|
Zuniga K, Ghousifam N, Sansalone J, Senecal K, Van Dyke M, Rylander MN. Keratin Promotes Differentiation of Keratinocytes Seeded on Collagen/Keratin Hydrogels. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9100559. [PMID: 36290526 PMCID: PMC9598618 DOI: 10.3390/bioengineering9100559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/07/2022] [Accepted: 10/09/2022] [Indexed: 11/05/2022]
Abstract
Keratinocytes undergo a complex process of differentiation to form the stratified stratum corneum layer of the skin. In most biomimetic skin models, a 3D hydrogel fabricated out of collagen type I is used to mimic human skin. However, native skin also contains keratin, which makes up 90% of the epidermis and is produced by the keratinocytes present. We hypothesized that the addition of keratin (KTN) in our collagen hydrogel may aid in the process of keratinocyte differentiation compared to a pure collagen hydrogel. Keratinocytes were seeded on top of a 100% collagen or 50/50 C/KTN hydrogel cultured in either calcium-free (Ca-free) or calcium+ (Ca+) media. Our study demonstrates that the addition of keratin and calcium in the media increased lysosomal activity by measuring the glucocerebrosidase (GBA) activity and lysosomal distribution length, an indication of greater keratinocyte differentiation. We also found that the presence of KTN in the hydrogel also increased the expression of involucrin, a differentiation marker, compared to a pure collagen hydrogel. We demonstrate that a combination (i.e., containing both collagen and kerateine or “C/KTN”) hydrogel was able to increase keratinocyte differentiation compared to a pure collagen hydrogel, and the addition of calcium further increased the differentiation of keratinocytes. This multi-protein hydrogel shows promise in future models or treatments to increase keratinocyte differentiation into the stratum corneum.
Collapse
Affiliation(s)
- Kameel Zuniga
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
- Correspondence:
| | - Neda Ghousifam
- Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - John Sansalone
- Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Kris Senecal
- Natick Soldier Center, U.S. Army Soldier & Biological Chemical Command, Natick, MA 01760, USA
| | - Mark Van Dyke
- College of Biomedical Engineering, The University of Arizona, Tucson, AZ 85721, USA
| | | |
Collapse
|
22
|
Yip AMH, Lai CKH, Yiu KSM, Lo KKW. Phosphorogenic Iridium(III) bis-Tetrazine Complexes for Bioorthogonal Peptide Stapling, Bioimaging, Photocytotoxic Applications, and the Construction of Nanosized Hydrogels. Angew Chem Int Ed Engl 2022; 61:e202116078. [PMID: 35119163 DOI: 10.1002/anie.202116078] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Indexed: 12/28/2022]
Abstract
The dual functionality of 1,2,4,5-tetrazine as a bioorthogonal reactive unit and a luminescence quencher has shaped tetrazine-based probes as attractive candidates for luminogenic labeling of biomolecules in living systems. In this work, three cyclometalated iridium(III) complexes featuring two tetrazine units were synthesized and characterized. Upon photoexcitation, the complexes were non-emissive but displayed up to 3900-fold emission enhancement upon the inverse electron-demand Diels-Alder (IEDDA) [4+2] cycloaddition with (1R,8S,9s)-bicyclo[6.1.0]non-4-yne (BCN) substrates. The rapid reaction kinetics (k2 up to 1.47×104 M-1 s-1 ) of the complexes toward BCN substrates allowed effective peptide labeling. The complexes were also applied as live cell bioimaging reagents and photocytotoxic agents. One of the complexes was utilized in the preparation of luminescent nanosized hydrogels that exhibited interesting cargo delivery properties.
Collapse
Affiliation(s)
- Alex Man-Hei Yip
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China
| | - Calvin Kin-Ho Lai
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China
| | - Ken Shek-Man Yiu
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China
| | - Kenneth Kam-Wing Lo
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China.,State Key Laboratory of Terahertz and Millimeter Waves, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China.,Center for Functional Photonics, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China
| |
Collapse
|
23
|
Molecular mechanisms of reactive oxygen species in regulated cell deaths: Impact of ferroptosis in cancer therapy. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
24
|
Yip AMH, Lai CKH, Yiu KSM, Lo KKW. Phosphorogenic Iridium(III) bis‐Tetrazine Complexes for Bioorthogonal Peptide Stapling, Bioimaging, Photocytotoxic Applications, and the Construction of Nanosized Hydrogels. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202116078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
| | | | | | - Kenneth Kam-Wing Lo
- City University of Hong Kong Department of Chemistry Tat Chee AvenueKowloon Tong N. A. Hong Kong HONG KONG
| |
Collapse
|
25
|
Roy B, Mengji R, Roy S, Pal B, Jana A, Singh NDP. NIR-Responsive Lysosomotropic Phototrigger: An "AIE + ESIPT" Active Naphthalene-Based Single-Component Photoresponsive Nanocarrier with Two-Photon Uncaging and Real-Time Monitoring Ability. ACS APPLIED MATERIALS & INTERFACES 2022; 14:4862-4870. [PMID: 35049266 DOI: 10.1021/acsami.1c19022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
In recent times, organelle-targeted drug delivery systems have gained tremendous attention due to the site-specific delivery of active drug molecules, resulting in enhanced bioefficacy. In this context, a phototriggered drug delivery system (DDS) for releasing an active molecule is superior, as it provides spatial and temporal control over the release. So far, a near-infrared (NIR) light-responsive organelle-targeted DDS has not yet been developed. Hence, we introduced a two-photon NIR light-responsive lysosome-targeted "AIE + ESIPT" active single-component DDS based on the naphthalene chromophore. The two-photon absorption cross section of our DDS is 142 GM at 850 nm. The DDS was converted into pure organic nanoparticles for biological applications. Our nano-DDS is capable of selective targeting, AIE luminogenic imaging, and drug release within the lysosome. In vitro studies using cancerous cell lines showed that our single-component photoresponsive nanocarrier exhibited enhanced cytotoxicity and real-time monitoring ability of drug release.
Collapse
Affiliation(s)
- Biswajit Roy
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | - Rakesh Mengji
- Department of Organic Synthesis and Process Chemistry, CSIR-Indian Institute of Chemical Technology Hyderabad, Uppal Road, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Samrat Roy
- Department of Physics, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal 741246, India
| | - Bipul Pal
- Department of Physics, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal 741246, India
| | - Avijit Jana
- Department of Organic Synthesis and Process Chemistry, CSIR-Indian Institute of Chemical Technology Hyderabad, Uppal Road, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - N D Pradeep Singh
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| |
Collapse
|
26
|
Gnanaguru G, Mackey A, Choi EY, Arta A, Rossato FA, Gero TW, Urquhart AJ, Scott DA, D'Amore PA, Ng YSE. Discovery of sterically-hindered phenol compounds with potent cytoprotective activities against ox-LDL-induced retinal pigment epithelial cell death as a potential pharmacotherapy. Free Radic Biol Med 2022; 178:360-368. [PMID: 34843917 PMCID: PMC8758799 DOI: 10.1016/j.freeradbiomed.2021.11.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/22/2021] [Accepted: 11/19/2021] [Indexed: 01/03/2023]
Abstract
Late-stage dry age-related macular degeneration (AMD) or geographic atrophy (GA) is an irreversible blinding condition characterized by degeneration of retinal pigment epithelium (RPE) and the associated photoreceptors. Clinical and genetic evidence supports a role for dysfunctional lipid processing and accumulation of harmful oxidized lipids in the pathogenesis of GA. Using an oxidized low-density lipoprotein (ox-LDL)-induced RPE death assay, we screened and identified sterically-hindered phenol compounds with potent protective activities for RPE. The phenol-containing PPARγ agonist, troglitazone, protected against ox-LDL-induced RPE cell death, whereas other more potent PPARγ agonists did not protect RPE cells. Knockdown of PPARγ did not affect the protective activity of troglitazone in RPE, confirming the protective function is not due to the thiazolidine (TZD) group of troglitazone. Prototypical hindered phenol trolox and its analogs potently protected against ox-LDL-induced RPE cell death whereas potent antioxidants without the phenol group failed to protect RPE. Hindered phenols preserved lysosomal integrity against ox-LDL-induced damage and FITC-labeled trolox was localized to the lysosomes in RPE cells. Analogs of trolox inhibited reactive oxygen species (ROS) formation induced by ox-LDL uptake in a dose-dependent fashion and were effective at sub-micromolar concentrations. Treatment with trolox analog 2,2,5,7,8-pentamethyl-6-chromanol (PMC) significantly induced the expression of the lysosomal protein NPC-1 and reduced intracellular cholesterol level upon ox-LDL uptake. Our data indicate that the lysosomal-localized hindered phenols are uniquely potent in protecting the RPE against the toxic effects of ox-LDL, and may represent a novel pharmacotherapy to preserve the vision in patients with GA.
Collapse
Affiliation(s)
- Gopalan Gnanaguru
- Harvard Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA, USA
| | - Ashley Mackey
- Harvard Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA, USA
| | - Eun Young Choi
- Harvard Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA, USA
| | - Anthoula Arta
- Harvard Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA, USA; Department of Health Technology, Institut for Sundhedsteknologi, Lyngby, Denmark
| | - Franco Aparecido Rossato
- Harvard Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA, USA
| | - Thomas W Gero
- Department of Cancer Biology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
| | - Andrew J Urquhart
- Department of Health Technology, Institut for Sundhedsteknologi, Lyngby, Denmark
| | - David A Scott
- Department of Cancer Biology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
| | - Patricia A D'Amore
- Harvard Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA, USA
| | - Yin Shan E Ng
- Harvard Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA, USA.
| |
Collapse
|
27
|
Wu C, Wang C, Zhang T, Gao G, Wei M, Chen Y, Li X, Wang F, Liang G. Lysosome-Targeted and Fluorescence-Turned "On" Cytotoxicity Induced by Alkaline Phosphatase-Triggered Self-Assembly. Adv Healthc Mater 2022; 11:e2101346. [PMID: 34624168 DOI: 10.1002/adhm.202101346] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/04/2021] [Indexed: 01/03/2023]
Abstract
Selectively inducing lysosomal membrane permeabilization (LMP) is a promising strategy for cancer therapy. But integrating alkaline phosphatase (ALP)-instructed self-assembly and lysosome-targeting to induce LMP for selective killing of cancer cells was not reported. Herein, a pyrene-peptide conjugate Py-Phe-Phe-Glu-Tyr(H2 PO3 )-Gly-lyso (Py-Yp-Lyso) is rationally designed and demonstrated for its lysosome-targeting cytotoxicity on cancer cells, together with its pyrene (Py) excimer fluorescence turning "on" at 480 nm. In vitro results showed that, Py-Yp-Lyso is efficiently dephosphorylated by ALP to yield Py-Phe-Phe-Glu-Tyr-Gly-lyso (Py-Y-Lyso) which self-assembles into nanofibers. Cell experiments verified that, after being taken up by HeLa cells, the excimer fluorescence of Py-Yp-Lyso assemblies has turned "on" and the assemblies specifically target the lysosomes, inducing LMP and ultimate cancer cell death. In vivo experiments indicated that Py-Yp-Lyso has the highest inhibition effect on HeLa tumors among the four compounds studied. This is anticipated for applying Py-Yp-Lyso to treat cancers in the clinic in the future.
Collapse
Affiliation(s)
- Chengfan Wu
- Hefei National Laboratory of Physical Sciences at Microscale Department of Chemistry University of Science and Technology of China 96 Jinzhai Road Hefei Anhui 230026 China
| | - Chenchen Wang
- Hefei National Laboratory of Physical Sciences at Microscale Department of Chemistry University of Science and Technology of China 96 Jinzhai Road Hefei Anhui 230026 China
| | - Tong Zhang
- School of Life Sciences University of Science and Technology of China 443 Huangshan Road Hefei Anhui 230027 China
| | - Ge Gao
- State Key Laboratory of Bioelectronics School of Biological Science and Medical Engineering Southeast University 2 Sipailou Road Nanjing 210096 China
| | - Mengxing Wei
- Hefei National Laboratory of Physical Sciences at Microscale Department of Chemistry University of Science and Technology of China 96 Jinzhai Road Hefei Anhui 230026 China
| | - Yinglu Chen
- Hefei National Laboratory of Physical Sciences at Microscale Department of Chemistry University of Science and Technology of China 96 Jinzhai Road Hefei Anhui 230026 China
| | - Xiaoyan Li
- Analysis Center Nanjing Medical University 140 Hanzhong Road Nanjing Jiangsu 210029 China
| | - Fuqiang Wang
- Analysis Center Nanjing Medical University 140 Hanzhong Road Nanjing Jiangsu 210029 China
| | - Gaolin Liang
- Hefei National Laboratory of Physical Sciences at Microscale Department of Chemistry University of Science and Technology of China 96 Jinzhai Road Hefei Anhui 230026 China
- State Key Laboratory of Bioelectronics School of Biological Science and Medical Engineering Southeast University 2 Sipailou Road Nanjing 210096 China
| |
Collapse
|
28
|
Wan B, Wu Z, Zhang X, Huang B. Mefloquine as a dual inhibitor of glioblastoma angiogenesis and glioblastoma via disrupting lysosomal function. Biochem Biophys Res Commun 2021; 580:7-13. [PMID: 34607260 DOI: 10.1016/j.bbrc.2021.09.069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 01/04/2023]
Abstract
Angiogenesis, the formation of new blood vessels from the pre-existing ones, is a hallmark characteristic of glioblastoma, making it an appealing target for treatment development. Given potent anti-cancer efficacy of mefloquine, FDA-approved anti-malarial drug, there is increasing interest in repurposing mefloquine for treatment of cancers, including glioblastoma. In line with these efforts, our work is the first to demonstrate that mefloquine is also an inhibitor of glioblastoma angiogenesis. Using glioblastoma microvascular endothelial cell (GMEC) isolated from glioblastoma patients, we show that mefloquine at clinically achievable concentration inhibits GMEC differentiation, capillary network formation, adhesion to Matrix, growth and survival. Mefloquine also inhibits growth and induces apoptosis in glioblastoma cells regardless of cellular origin and genetic background. We further show that mefloquine significantly inhibits glioblastoma growth but not formation, and this is associated with decreased glioblastoma angiogenesis in mice. Mechanistically, mefloquine disrupted lysosomal integrity and function in GMECs, leading to oxidative stress and lysosomal lipid damage. Rescue studies confirm that mefloquine acts on GMECs in a lysosomal disruption-dependent manner. Our findings demonstrate the anti-angiogenic activity of mefloquine via disrupting lysosomal function. The dual inhibitory role of mefloquine in glioblastoma angiogenesis and glioblastoma displays its advantage over other anti-cancer drugs for glioblastoma treatment. Our work also highlights the essential role of lysosome in both glioblastoma and its angiogenesis.
Collapse
Affiliation(s)
- Bing Wan
- Department of Radiology, China Three Gorges University, Affiliated Renhe Hospital, Yichang, Hubei, China
| | - Zhenghong Wu
- Department of Radiology, Jingzhou Central Hospital, Jingzhou Hospital, Yangtze University, Jingzhou, Hubei, China
| | - Xiulan Zhang
- Department of Radiology, The First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China.
| | - Birun Huang
- Department of Vascular Surgery, The First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China.
| |
Collapse
|
29
|
Halaby R. Natural Products Induce Lysosomal Membrane Permeabilization as an Anticancer Strategy. MEDICINES 2021; 8:medicines8110069. [PMID: 34822366 PMCID: PMC8624533 DOI: 10.3390/medicines8110069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/03/2021] [Accepted: 11/08/2021] [Indexed: 11/26/2022]
Abstract
Cancer is a global health and economic issue. The majority of anticancer therapies become ineffective due to frequent genomic turnover and chemoresistance. Furthermore, chemotherapy and radiation are non-specific, killing all rapidly dividing cells including healthy cells. In this review, we examine the ability of some natural products to induce lysosomal-mediated cell death in neoplastic cells as a way to kill them more specifically than conventional therapies. This list is by no means exhaustive. We postulate mechanisms to explain lysosomal membrane permeabilization and its role in triggering cell death in cancer cells.
Collapse
Affiliation(s)
- Reginald Halaby
- Department of Biology, Montclair State University, Montclair, NJ 07043, USA
| |
Collapse
|
30
|
Tarallo A, Damiano C, Strollo S, Minopoli N, Indrieri A, Polishchuk E, Zappa F, Nusco E, Fecarotta S, Porto C, Coletta M, Iacono R, Moracci M, Polishchuk R, Medina DL, Imbimbo P, Monti DM, De Matteis MA, Parenti G. Correction of oxidative stress enhances enzyme replacement therapy in Pompe disease. EMBO Mol Med 2021; 13:e14434. [PMID: 34606154 PMCID: PMC8573602 DOI: 10.15252/emmm.202114434] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 09/15/2021] [Accepted: 09/20/2021] [Indexed: 02/06/2023] Open
Abstract
Pompe disease is a metabolic myopathy due to acid alpha-glucosidase deficiency. In addition to glycogen storage, secondary dysregulation of cellular functions, such as autophagy and oxidative stress, contributes to the disease pathophysiology. We have tested whether oxidative stress impacts on enzyme replacement therapy with recombinant human alpha-glucosidase (rhGAA), currently the standard of care for Pompe disease patients, and whether correction of oxidative stress may be beneficial for rhGAA therapy. We found elevated oxidative stress levels in tissues from the Pompe disease murine model and in patients' cells. In cells, stress levels inversely correlated with the ability of rhGAA to correct the enzymatic deficiency. Antioxidants (N-acetylcysteine, idebenone, resveratrol, edaravone) improved alpha-glucosidase activity in rhGAA-treated cells, enhanced enzyme processing, and improved mannose-6-phosphate receptor localization. When co-administered with rhGAA, antioxidants improved alpha-glucosidase activity in tissues from the Pompe disease mouse model. These results indicate that oxidative stress impacts on the efficacy of enzyme replacement therapy in Pompe disease and that manipulation of secondary abnormalities may represent a strategy to improve the efficacy of therapies for this disorder.
Collapse
Affiliation(s)
- Antonietta Tarallo
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Department of Translational Medical SciencesFederico II UniversityNaplesItaly
| | - Carla Damiano
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Department of Translational Medical SciencesFederico II UniversityNaplesItaly
| | - Sandra Strollo
- Telethon Institute of Genetics and MedicinePozzuoliItaly
| | - Nadia Minopoli
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Department of Translational Medical SciencesFederico II UniversityNaplesItaly
| | - Alessia Indrieri
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Institute for Genetic and Biomedical Research (IRGB)National Research Council (CNR)MilanItaly
| | | | - Francesca Zappa
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Present address:
Department of Molecular, Cellular, and Developmental BiologyUniversity of CaliforniaSanta BarbaraCAUSA
| | - Edoardo Nusco
- Telethon Institute of Genetics and MedicinePozzuoliItaly
| | - Simona Fecarotta
- Department of Translational Medical SciencesFederico II UniversityNaplesItaly
| | - Caterina Porto
- Department of Translational Medical SciencesFederico II UniversityNaplesItaly
| | - Marcella Coletta
- Department of Translational Medical SciencesFederico II UniversityNaplesItaly
- Present address:
IInd Division of NeurologyMultiple Sclerosis CenterUniversity of Campania "Luigi Vanvitelli"NaplesItaly
| | - Roberta Iacono
- Department of BiologyUniversity of Naples "Federico II", Complesso Universitario di Monte S. AngeloNaplesItaly
- Institute of Biosciences and BioResources ‐ National Research Council of ItalyNaplesItaly
| | - Marco Moracci
- Department of BiologyUniversity of Naples "Federico II", Complesso Universitario di Monte S. AngeloNaplesItaly
- Institute of Biosciences and BioResources ‐ National Research Council of ItalyNaplesItaly
| | | | - Diego Luis Medina
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Department of Translational Medical SciencesFederico II UniversityNaplesItaly
| | - Paola Imbimbo
- Department of Chemical SciencesFederico II UniversityNaplesItaly
| | | | - Maria Antonietta De Matteis
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Department of Molecular Medicine and Medical BiotechnologiesFederico II UniversityNaplesItaly
| | - Giancarlo Parenti
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Department of Translational Medical SciencesFederico II UniversityNaplesItaly
| |
Collapse
|
31
|
Halcrow PW, Lynch ML, Geiger JD, Ohm JE. Role of endolysosome function in iron metabolism and brain carcinogenesis. Semin Cancer Biol 2021; 76:74-85. [PMID: 34139350 PMCID: PMC8627927 DOI: 10.1016/j.semcancer.2021.06.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 02/07/2023]
Abstract
Iron, the most abundant metal in human brain, is an essential microelement that regulates numerous cellular mechanisms. Some key physiological roles of iron include oxidative phosphorylation and ATP production, embryonic neuronal development, formation of iron-sulfur clusters, and the regulation of enzymes involved in DNA synthesis and repair. Because of its physiological and pathological importance, iron homeostasis must be tightly regulated by balancing its uptake, transport, and storage. Endosomes and lysosomes (endolysosomes) are acidic organelles known to contain readily releasable stores of various cations including iron and other metals. Increased levels of ferrous (Fe2+) iron can generate reactive oxygen species (ROS) via Fenton chemistry reactions and these increases can damage mitochondria and genomic DNA as well as promote carcinogenesis. Accumulation of iron in the brain has been linked with aging, diet, disease, and cerebral hemorrhage. Further, deregulation of brain iron metabolism has been implicated in carcinogenesis and may be a contributing factor to the increased incidence of brain tumors around the world. Here, we provide insight into mechanisms by which iron accumulation in endolysosomes is altered by pH and lysosome membrane permeabilization. Such events generate excess ROS resulting in mitochondrial DNA damage, fission, and dysfunction, as well as DNA oxidative damage in the nucleus; all of which promote carcinogenesis. A better understanding of the roles that endolysosome iron plays in carcinogenesis may help better inform the development of strategic therapeutic options for cancer treatment and prevention.
Collapse
Affiliation(s)
- Peter W Halcrow
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| | - Miranda L Lynch
- Hauptman-Woodward Medical Research Institute, Buffalo, NY, United States
| | - Jonathan D Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| | - Joyce E Ohm
- Department of Cancer Genetics and Genomics, Roswell Park Cancer Institute, Buffalo, NY, United States.
| |
Collapse
|
32
|
Liu Q, Tian J, Liu J, Zhu M, Gao Z, Hu X, Midgley AC, Wu J, Wang X, Kong D, Zhuang J, Liu J, Yan X, Huang X. Modular Assembly of Tumor-Penetrating and Oligomeric Nanozyme Based on Intrinsically Self-Assembling Protein Nanocages. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2103128. [PMID: 34350648 DOI: 10.1002/adma.202103128] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/26/2021] [Indexed: 06/13/2023]
Abstract
Biomimetic design of nanomaterials with enzyme-like characteristics has emerged as a promising method for the generation of novel therapeutics. However, synthesis of nanomaterials while maintaining a high degree of control over both geometry and valency poses a prominent challenge. Herein, the authors introduce a nanomaterial-based synthetic biology strategy for accurate and quantitative tailoring of high-ordered nanostructures that uses a "bottom-up" hierarchical incorporation of protein building blocks. The assembled nano-oligomers possessed tunable protein motifs and multivalent binding domains, which facilitated prolonged blood circulation time, accumulation within tumor cells through direct targeting of cell receptors, and deep tumor tissue penetration via a transcytosis mechanism. Using these protein/protein nano-oligomers as scaffolds, the authors created a new series of artificial nano-scaled metalloenzymes (nanozymes) by the in situ incorporation of metal nanoclusters within the cavity of the protein nanocages. Nanozymes were capable of mimicking peroxidase-like activity and generated cytotoxic free radicals. Compared to nanozyme alone, the systemic delivery of oligomeric nanozymes demonstrated significantly enhanced therapeutic and anti-tumor benefits. This study shows a new insight into nanotechnology by taking advantage of synthetic biotechnology.
Collapse
Affiliation(s)
- Qiqi Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Frontiers Science Center for Cell Responses, Nankai University, Tianjin, 300071, China
| | - Jingwei Tian
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Frontiers Science Center for Cell Responses, Nankai University, Tianjin, 300071, China
| | - Jinjian Liu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Mingsheng Zhu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Frontiers Science Center for Cell Responses, Nankai University, Tianjin, 300071, China
| | - Zhanxia Gao
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Xueyan Hu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Frontiers Science Center for Cell Responses, Nankai University, Tianjin, 300071, China
| | - Adam C Midgley
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Frontiers Science Center for Cell Responses, Nankai University, Tianjin, 300071, China
| | - Jin Wu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Frontiers Science Center for Cell Responses, Nankai University, Tianjin, 300071, China
- Joint Laboratory of Nanozymes, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Xinyue Wang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Frontiers Science Center for Cell Responses, Nankai University, Tianjin, 300071, China
| | - Deling Kong
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Frontiers Science Center for Cell Responses, Nankai University, Tianjin, 300071, China
| | - Jie Zhuang
- School of Medicine, Nankai University, Tianjin, 300071, China
- Joint Laboratory of Nanozymes, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jianfeng Liu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Xiyun Yan
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Frontiers Science Center for Cell Responses, Nankai University, Tianjin, 300071, China
- Joint Laboratory of Nanozymes, College of Life Sciences, Nankai University, Tianjin, 300071, China
- CAS Engineering Laboratory for Nanozymes, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xinglu Huang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Frontiers Science Center for Cell Responses, Nankai University, Tianjin, 300071, China
- Joint Laboratory of Nanozymes, College of Life Sciences, Nankai University, Tianjin, 300071, China
| |
Collapse
|
33
|
Hu P, Li H, Sun W, Wang H, Yu X, Qing Y, Wang Z, Zhu M, Xu J, Guo Q, Hui H. Cholesterol-associated lysosomal disorder triggers cell death of hematological malignancy: Dynamic analysis on cytotoxic effects of LW-218. Acta Pharm Sin B 2021; 11:3178-3192. [PMID: 34729308 PMCID: PMC8546890 DOI: 10.1016/j.apsb.2021.02.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/03/2020] [Accepted: 01/07/2021] [Indexed: 12/15/2022] Open
Abstract
The integrity of lysosomes is of vital importance to survival of tumor cells. We demonstrated that LW-218, a synthetic flavonoid, induced rapid lysosomal enlargement accompanied with lysosomal membrane permeabilization in hematological malignancy. LW-218-induced lysosomal damage and lysosome-dependent cell death were mediated by cathepsin D, as the lysosomal damage and cell apoptosis could be suppressed by depletion of cathepsin D or lysosome alkalization agents, which can alter the activity of cathepsins. Lysophagy, was initiated for cell self-rescue after LW-218 treatment and correlated with calcium release and nuclei translocation of transcription factor EB. LW-218 treatment enhanced the expression of autophagy-related genes which could be inhibited by intracellular calcium chelator. Sustained exposure to LW-218 exhausted the lysosomal capacity so as to repress the normal autophagy. LW-218-induced enlargement and damage of lysosomes were triggered by abnormal cholesterol deposition on lysosome membrane which caused by interaction between LW-218 and NPC intracellular cholesterol transporter 1. Moreover, LW-218 inhibited the leukemia cell growth in vivo. Thus, the necessary impact of integral lysosomal function in cell rescue and death were illustrated.
Collapse
Key Words
- AO, acridine orange
- ATG, autophagy related
- BAF A1, bafilomycin A1
- BID, BH3-interacting domain death agonist
- CCK8, Cell Counting Kit
- CTSB, cathepsin B
- CTSD, cathepsin D
- CaN, calcineurin
- Cathepsin D
- Cholesterol
- CsA, cyclosporine A
- DAPI, 4′,6-diamidino-2-phenylindole dihydrochloride
- DCFH-DA, 2,7-dichlorodi-hydrofluorescein diacetate
- Dex, dexamethasone
- EGTA, ethylene glycol-bis(2-aminoethyl ether)-N,N,N′,N′-tetraacetic acid
- FBS, fetal bovine serum
- Hematological malignancies
- K48, lysine 48
- K63, lysine 63
- LAMPs, lysosomal-associated membrane proteins
- LC3, microtubule-associated protein 1 light chain 3
- LCD, lysosome-dependent cell death
- LMP, lysosome membrane permeabilization
- LW-218
- Lysophagy
- Lysosomal damage
- Lysosomal membrane permeabilization
- Lysosome-dependent cell death
- NH4Cl, ammonium chloride
- NPC, Niemann-Pick type disease C
- NPC1, NPC intracellular cholesterol transporter 1
- OD, optical density
- P62/SQSTM1, sequestosome 1
- PBMCs, peripheral blood mononuclear cells
- PBS, phosphate-buffered saline
- RAB7A, RAS-related protein RAB-7a
- ROS, reactive oxygen species
- RT-qPCR, real time quantitative PCR
- TFEB, transcription factor EB
- TRPML1, transient receptor potential mucolipin 1
- shRNA, short hairpin RNA
Collapse
Affiliation(s)
- Po Hu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, China
| | - Hui Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, China
| | - Wenzhuo Sun
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, China
| | - Hongzheng Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaoxuan Yu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacology, School of Medicine & Holostic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Yingjie Qing
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, China
| | - Zhanyu Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, China
| | - Mengyuan Zhu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, China
| | - Jingyan Xu
- Department of Hematology, the Affiliated DrumTower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Qinglong Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, China
| | - Hui Hui
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
34
|
Transformable amyloid-beta mimetic peptide amphiphiles for lysosomal disruption in non-small cell lung cancer. Biomaterials 2021; 277:121078. [PMID: 34461458 PMCID: PMC9969961 DOI: 10.1016/j.biomaterials.2021.121078] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 08/11/2021] [Accepted: 08/18/2021] [Indexed: 12/24/2022]
Abstract
Non-small cell lung cancer (NSCLC) is the largest contributor to cancer mortality in the United States. Traditional chemotherapies are toxic and prone to the development of drug-resistance. Recently, several drug candidates were shown to induce lysosomal membrane permeabilization (LMP) in aggressive cancers. This has led to increased interest in lysosome dysregulation as a therapeutic target. However, approaches are needed to overcome two limitations of current lysosomal inhibitors: low specificity and potency. Here, we report the development of a transformable nanomaterial which is triggered to induce LMP of lysosomes in NSCLC. The nanomaterial consists of peptide amphiphiles, which self-assemble into nanoparticles, colocalize with the lysosome, and change conformation to nanofibrils due to lysosomal pH shift, which leads to the disruption of the lysosome, cell death, and cisplatin sensitization. We have found that this cell-penetrating transformable peptide nanoparticle (CPTNP) was cytotoxic to NSCLC cells in the low-micromolar range and it synergized cisplatin cytotoxicity four-fold. Moreover, we demonstrate CPTNP's promising antitumor effect in mouse xenograft models with limited toxicity when given in combination with low dose cisplatin chemotherapy. This is the first example of enhanced LMP via transformable peptide nanomaterial and offers a promising new strategy for cancer therapy.
Collapse
|
35
|
Mahmud KM, Niloy MS, Shakil MS, Islam MA. Ruthenium Complexes: An Alternative to Platinum Drugs in Colorectal Cancer Treatment. Pharmaceutics 2021; 13:1295. [PMID: 34452256 PMCID: PMC8398452 DOI: 10.3390/pharmaceutics13081295] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/13/2021] [Accepted: 08/16/2021] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is one of the intimidating causes of death around the world. CRC originated from mutations of tumor suppressor genes, proto-oncogenes and DNA repair genes. Though platinum (Pt)-based anticancer drugs have been widely used in the treatment of cancer, their toxicity and CRC cells' resistance to Pt drugs has piqued interest in the search for alternative metal-based drugs. Ruthenium (Ru)-based compounds displayed promising anticancer activity due to their unique chemical properties. Ru-complexes are reported to exert their anticancer activities in CRC cells by regulating different cell signaling pathways that are either directly or indirectly associated with cell growth, division, proliferation, and migration. Additionally, some Ru-based drug candidates showed higher potency compared to commercially available Pt-based anticancer drugs in CRC cell line models. Meanwhile Ru nanoparticles coupled with photosensitizers or anticancer agents have also shown theranostic potential towards CRC. Ru-nanoformulations improve drug efficacy, targeted drug delivery, immune activation, and biocompatibility, and therefore may be capable of overcoming some of the existing chemotherapeutic limitations. Among the potential Ru-based compounds, only Ru (III)-based drug NKP-1339 has undergone phase-Ib clinical trials in CRC treatment.
Collapse
Affiliation(s)
- Kazi Mustafa Mahmud
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh; (K.M.M.); (M.S.N.)
| | - Mahruba Sultana Niloy
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh; (K.M.M.); (M.S.N.)
| | - Md Salman Shakil
- Department of Pharmacology & Toxicology, University of Otago, Dunedin 9016, New Zealand
- Department of Biochemistry, Primeasia University, Banani, Dhaka 1213, Bangladesh
| | - Md Asiful Islam
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
| |
Collapse
|
36
|
Allemailem KS, Almatroudi A, Alrumaihi F, Almatroodi SA, Alkurbi MO, Basfar GT, Rahmani AH, Khan AA. Novel Approaches of Dysregulating Lysosome Functions in Cancer Cells by Specific Drugs and Its Nanoformulations: A Smart Approach of Modern Therapeutics. Int J Nanomedicine 2021; 16:5065-5098. [PMID: 34345172 PMCID: PMC8324981 DOI: 10.2147/ijn.s321343] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 07/08/2021] [Indexed: 01/18/2023] Open
Abstract
The smart strategy of cancer cells to bypass the caspase-dependent apoptotic pathway has led to the discovery of novel anti-cancer approaches including the targeting of lysosomes. Recent discoveries observed that lysosomes perform far beyond just recycling of cellular waste, as these organelles are metabolically very active and mediate several signalling pathways to sense the cellular metabolic status. These organelles also play a significant role in mediating the immune system functions. Thus, direct or indirect lysosome-targeting with different drugs can be considered a novel therapeutic approach in different disease including cancer. Recently, some anticancer lysosomotropic drugs (eg, nortriptyline, siramesine, desipramine) and their nanoformulations have been engineered to specifically accumulate within these organelles. These drugs can enhance lysosome membrane permeabilization (LMP) or disrupt the activity of resident enzymes and protein complexes, like v-ATPase and mTORC1. Other anticancer drugs like doxorubicin, quinacrine, chloroquine and DQ661 have also been used which act through multi-target points. In addition, autophagy inhibitors, ferroptosis inducers and fluorescent probes have also been used as novel theranostic agents. Several lysosome-specific drug nanoformulations like mixed charge and peptide conjugated gold nanoparticles (AuNPs), Au-ZnO hybrid NPs, TPP-PEG-biotin NPs, octadecyl-rhodamine-B and cationic liposomes, etc. have been synthesized by diverse methods. These nanoformulations can target cathepsins, glucose-regulated protein 78, or other lysosome specific proteins in different cancers. The specific targeting of cancer cell lysosomes with drug nanoformulations is quite recent and faces tremendous challenges like toxicity concerns to normal tissues, which may be resolved in future research. The anticancer applications of these nanoformulations have led them up to various stages of clinical trials. Here in this review article, we present the recent updates about the lysosome ultrastructure, its cross-talk with other organelles, and the novel strategies of targeting this organelle in tumor cells as a recent innovative approach of cancer management.
Collapse
Affiliation(s)
- Khaled S Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Saleh A Almatroodi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Mohammad O Alkurbi
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Ghaiyda Talal Basfar
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Amjad Ali Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| |
Collapse
|
37
|
Li Q, Wang L, Ji D, Bao X, Tan G, Liang X, Deng P, Pi H, Lu Y, Chen C, He M, Zhang L, Zhou Z, Yu Z, Deng A. BIX-01294, a G9a inhibitor, suppresses cell proliferation by inhibiting autophagic flux in nasopharyngeal carcinoma cells. Invest New Drugs 2021; 39:686-696. [PMID: 33387131 DOI: 10.1007/s10637-020-01053-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/15/2020] [Indexed: 10/22/2022]
Abstract
G9a, a histone methyltransferase, has been found to be upregulated in a range of tumor tissues, and contributes to tumor growth and metastasis. However, the impact of G9a inhibition as a potential therapeutic target in nasopharyngeal carcinoma (NPC) is unclear. In the present study we aimed to investigate the anti-proliferative effect of G9a inhibition in the NPC cell lines CNE1 and CNE2, and to further elucidate the molecular mechanisms underlying these effects. The expression of G9a in NPC tumor tissues was significantly higher than that in normal nasopharyngeal tissues. The pharmacological inhibition of G9a by BIX-01294 (BIX) inhibited proliferation and induced caspase-independent apoptosis in NPC cells in vitro. Treatment with BIX induced autophagosome accumulation, which exacerbated the cytotoxic activity of BIX in NPC cells. Mechanistic studies have found that BIX impairs autophagosomes by initiating autophagy in a Beclin-1-independent way, and impairs autophagic degradation by inhibiting lysosomal cathepsin D activation, leading to lysosomal dysfunction. BIX was able to suppress tumor growth, possibly by inhibiting autophagic flux; it might therefore constitute a promising candidate for NPC therapy.
Collapse
Affiliation(s)
- Qian Li
- Department of Otolaryngology Head and Neck Surgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Liuqian Wang
- Department of Otolaryngology Head and Neck Surgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Di Ji
- Department of Otolaryngology Head and Neck Surgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiaomin Bao
- Department of Otolaryngology Head and Neck Surgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Guojing Tan
- Department of Otolaryngology Head and Neck Surgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiaojun Liang
- Department of Otolaryngology Head and Neck Surgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Ping Deng
- Department of Occupational Health, Army Medical University (Third Military Medical University), Chongqing, China
| | - Huifeng Pi
- Department of Occupational Health, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yonghui Lu
- Department of Occupational Health, Army Medical University (Third Military Medical University), Chongqing, China
| | - Chunhai Chen
- Department of Occupational Health, Army Medical University (Third Military Medical University), Chongqing, China
| | - Mindi He
- Department of Occupational Health, Army Medical University (Third Military Medical University), Chongqing, China
| | - Lei Zhang
- Department of Occupational Health, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhou Zhou
- Department of Environmental Medicine, Department of Emergency Medicine of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhengping Yu
- Department of Occupational Health, Army Medical University (Third Military Medical University), Chongqing, China
| | - Anchun Deng
- Department of Otolaryngology Head and Neck Surgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China.
| |
Collapse
|
38
|
Chaeichi-Tehrani N, Ferns GA, Hassanian SM, Khazaei M, Avan A. The Therapeutic Potential of Targeting Autophagy in The Treatment of Cancer. Curr Cancer Drug Targets 2021; 21:725-736. [PMID: 34077348 DOI: 10.2174/1568009621666210601113144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 03/02/2021] [Accepted: 03/12/2021] [Indexed: 11/22/2022]
Abstract
Autophagy is a mechanism by which unwanted cellular components are degraded through a pathway that involves the lysosomes and contributes to several pathological conditions such as cancer. Gastrointestinal cancers affect the digestive organs from the esophagus to the anus and are among the most commonly diagnosed cancers globally. The modulation of autophagy using pharmacologic agents potentially offers a great potential for cancer therapy. In this review, some commonly used compounds, together with their molecular target and the mechanism through which they stimulate or block the autophagy pathway as well as their therapeutic benefit in treating patients with gastrointestinal cancers, are summarized.
Collapse
Affiliation(s)
- Negin Chaeichi-Tehrani
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, United Kingdom
| | - Seyed Mahdi Hassanian
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Metabolic syndrome Research centre, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
39
|
Targeting nutrient metabolism with FDA-approved drugs for cancer chemoprevention: Drugs and mechanisms. Cancer Lett 2021; 510:1-12. [PMID: 33857528 DOI: 10.1016/j.canlet.2021.03.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/21/2021] [Accepted: 03/30/2021] [Indexed: 12/14/2022]
Abstract
Proliferating cancer cells exhibit metabolic alterations and specific nutritional needs for adapting to their rapid growth. These changes include using aerobic glycolysis, lipid metabolic disorder, and irregular protein degradation. It may be useful to target metabolic abnormalities for cancer chemoprevention. Epidemiological and mechanism-related studies have indicated that many FDA-approved anti-metabolic drugs decrease tumor risk, inhibit tumor growth, or enhance the effect of chemotherapeutic drugs. Drugs targeting nutrient metabolism have fewer side effects with long-term use compared to chemotherapeutic drugs. The characteristics of these drugs make them promising candidates for cancer chemoprevention. Here, we summarize recent discoveries of the chemo-preventive effects of drugs targeting nutrient metabolic pathways and discuss future applications and challenges. Understanding the effects and mechanisms of anti-metabolic drugs in cancer has important implications for exploring strategies for cancer chemoprevention.
Collapse
|
40
|
Xiong H, Wang C, Wang Z, Lu H, Yao J. Self-assembled nano-activator constructed ferroptosis-immunotherapy through hijacking endogenous iron to intracellular positive feedback loop. J Control Release 2021; 332:539-552. [DOI: 10.1016/j.jconrel.2021.03.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 02/28/2021] [Accepted: 03/03/2021] [Indexed: 12/31/2022]
|
41
|
Wang P, Gong Q, Hu J, Li X, Zhang X. Reactive Oxygen Species (ROS)-Responsive Prodrugs, Probes, and Theranostic Prodrugs: Applications in the ROS-Related Diseases. J Med Chem 2020; 64:298-325. [PMID: 33356214 DOI: 10.1021/acs.jmedchem.0c01704] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Elevated levels of reactive oxygen species (ROS) have commonly been implicated in a variety of diseases, including cancer, inflammation, and neurodegenerative diseases. In light of significant differences in ROS levels between the nonpathogenic and pathological tissues, an increasing number of ROS-responsive prodrugs, probes, and theranostic prodrugs have been developed for the targeted treatment and precise diagnosis of ROS-related diseases. This review will summarize and provide insight into recent advances in ROS-responsive prodrugs, fluorescent probes, and theranostic prodrugs, with applications to different ROS-related diseases and various subcellular organelle-targetable and disease-targetable features. The ROS-responsive moieties, the self-immolative linkers, and the typical activation mechanism for the ROS-responsive release are also summarized and discussed.
Collapse
Affiliation(s)
- Pengfei Wang
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China.,Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Qijie Gong
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Jiabao Hu
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Xiang Li
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaojin Zhang
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
42
|
Hu M, Carraway KL. Repurposing Cationic Amphiphilic Drugs and Derivatives to Engage Lysosomal Cell Death in Cancer Treatment. Front Oncol 2020; 10:605361. [PMID: 33425762 PMCID: PMC7793984 DOI: 10.3389/fonc.2020.605361] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 11/13/2020] [Indexed: 12/22/2022] Open
Abstract
A major confounding issue in the successful treatment of cancer is the existence of tumor cell populations that resist therapeutic agents and regimens. While tremendous effort has gone into understanding the biochemical mechanisms underlying resistance to each traditional and targeted therapeutic, a broader approach to the problem may emerge from the recognition that existing anti-cancer agents elicit their cytotoxic effects almost exclusively through apoptosis. Considering the myriad mechanisms cancer cells employ to subvert apoptotic death, an attractive alternative approach would leverage programmed necrotic mechanisms to side-step therapeutic resistance to apoptosis-inducing agents. Lysosomal cell death (LCD) is a programmed necrotic cell death mechanism that is engaged upon the compromise of the limiting membrane of the lysosome, a process called lysosomal membrane permeabilization (LMP). The release of lysosomal components into the cytosol upon LMP triggers biochemical cascades that lead to plasma membrane rupture and necrotic cell death. Interestingly, the process of cellular transformation appears to render the limiting lysosomal membranes of tumor cells more fragile than non-transformed cells, offering a potential therapeutic window for drug development. Here we outline the concepts of LMP and LCD, and discuss strategies for the development of agents to engage these processes. Importantly, the potential exists for existing cationic amphiphilic drugs such as antidepressants, antibiotics, antiarrhythmics, and diuretics to be repurposed to engage LCD within therapy-resistant tumor cell populations.
Collapse
Affiliation(s)
- Michelle Hu
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, CA, United States
- UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA, United States
| | - Kermit L. Carraway
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, CA, United States
- UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA, United States
| |
Collapse
|
43
|
Shear Stress Increases V-H + -ATPase and Acidic Vesicle Number Density, and p-mTORC2 Activation in Prostate Cancer Cells. Cell Mol Bioeng 2020; 13:591-604. [PMID: 33281989 DOI: 10.1007/s12195-020-00632-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 06/18/2020] [Indexed: 02/06/2023] Open
Abstract
Introduction Cells in the tumor microenvironment experience mechanical stresses, such as compression generated by uncontrolled cell growth within a tissue, increased substrate stiffness due to tumor cell extracellular matrix (ECM) remodeling, and leaky angiogenic vessels which involve low fluid shear stress. With our hypothesis that shear stress increases V-H + -ATPase number density in prostate cancer cells via activation of the mTORC1 and mTORC2 pathways, we demonstrated and quantified such a mechanism in prostate cancer cells. Methods Moderately metastatic DU145 and highly metastatic PC3 prostate cancer cells were subjected to 0.05 dynes cm - 2 wall shear stress for 24 h, followed by immunocytochemistry and fluorescence measurements of β 1 integrin, endosome, lysosome, V-H + -ATPase proton pump, mTORC1, and p-mTORC2 antibodies. Post shear stress migration assays, and the effects of vacuolar proton pump inhibitor Bafilomycin A1 (60 nM, 24 h) as well as shear stress on the ICC fluorescence intensity of the proteins of interest were conducted with DU145 cells. Results Low fluid shear stress increases the fluorescence intensity of β 1 integrin, endosome, lysosome, V-H + -ATPase, mTORC1, and p-mTORC2 antibodies in PC3 and DU145 cells, and also increased cell migration. However, Bafilomycin A1 decreased fluorescence intensity of all of these proteins in DU145 cells exposed to shear stress, revealing that V-H + -ATPase controls the expression of these proteins. Conclusions Prostate cancer cell mechanotransduction increases endosomes, lysosomes, and proton pumps-where increases have been associated with enhanced cancer aggressiveness. We also show that the prostate cancer cell's response to force promotes the cancer drivers mTORC1 and mTORC2.
Collapse
|
44
|
Zhai X, El Hiani Y. Getting Lost in the Cell-Lysosomal Entrapment of Chemotherapeutics. Cancers (Basel) 2020; 12:E3669. [PMID: 33297435 PMCID: PMC7762281 DOI: 10.3390/cancers12123669] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/24/2020] [Accepted: 12/03/2020] [Indexed: 12/15/2022] Open
Abstract
Despite extensive research, resistance to chemotherapy still poses a major obstacle in clinical oncology. An exciting strategy to circumvent chemoresistance involves the identification and subsequent disruption of cellular processes that are aberrantly altered in oncogenic states. Upon chemotherapeutic challenges, lysosomes are deemed to be essential mediators that enable cellular adaptation to stress conditions. Therefore, lysosomes potentially hold the key to disarming the fundamental mechanisms of chemoresistance. This review explores modes of action of classical chemotherapeutic agents, adaptive response of the lysosomes to cell stress, and presents physiological and pharmacological insights pertaining to drug compartmentalization, sequestration, and extracellular clearance through the lens of lysosomes.
Collapse
Affiliation(s)
| | - Yassine El Hiani
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada;
| |
Collapse
|
45
|
Korolenko TA, Johnston TP, Vetvicka V. Lysosomotropic Features and Autophagy Modulators among Medical Drugs: Evaluation of Their Role in Pathologies. Molecules 2020; 25:molecules25215052. [PMID: 33143272 PMCID: PMC7662698 DOI: 10.3390/molecules25215052] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/23/2020] [Accepted: 10/25/2020] [Indexed: 12/25/2022] Open
Abstract
The concept of lysosomotropic agents significantly changed numerous aspects of cellular biochemistry, biochemical pharmacology, and clinical medicine. In the present review, we focused on numerous low-molecular and high-molecular lipophilic basic compounds and on the role of lipophagy and autophagy in experimental and clinical medicine. Attention was primarily focused on the most promising agents acting as autophagy inducers, which offer a new window for treatment and/or prophylaxis of various diseases, including type 2 diabetes mellitus, Parkinson's disease, and atherosclerosis. The present review summarizes current knowledge on the lysosomotropic features of medical drugs, as well as autophagy inducers, and their role in pathological processes.
Collapse
Affiliation(s)
- Tatiana A. Korolenko
- Federal State Budgetary Scientific Institution Scientific Research Institute of Physiology and Basic Medicine, Timakova Str. 4, 630117 Novosibirsk, Russia;
| | - Thomas P. Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO 64108, USA;
| | - Vaclav Vetvicka
- Department of Pathology, University of Louisville, Louisville, KY 40292, USA
- Correspondence:
| |
Collapse
|
46
|
Alu A, Han X, Ma X, Wu M, Wei Y, Wei X. The role of lysosome in regulated necrosis. Acta Pharm Sin B 2020; 10:1880-1903. [PMID: 33163342 PMCID: PMC7606114 DOI: 10.1016/j.apsb.2020.07.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/29/2020] [Accepted: 04/24/2020] [Indexed: 02/06/2023] Open
Abstract
Lysosome is a ubiquitous acidic organelle fundamental for the turnover of unwanted cellular molecules, particles, and organelles. Currently, the pivotal role of lysosome in regulating cell death is drawing great attention. Over the past decades, we largely focused on how lysosome influences apoptosis and autophagic cell death. However, extensive studies showed that lysosome is also prerequisite for the execution of regulated necrosis (RN). Different types of RN have been uncovered, among which, necroptosis, ferroptosis, and pyroptosis are under the most intensive investigation. It becomes a hot topic nowadays to target RN as a therapeutic intervention, since it is important in many patho/physiological settings and contributing to numerous diseases. It is promising to target lysosome to control the occurrence of RN thus altering the outcomes of diseases. Therefore, we aim to give an introduction about the common factors influencing lysosomal stability and then summarize the current knowledge on the role of lysosome in the execution of RN, especially in that of necroptosis, ferroptosis, and pyroptosis.
Collapse
Affiliation(s)
- Aqu Alu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xuejiao Han
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xuelei Ma
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Min Wu
- Department of Biochemistry and Molecular Biology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
47
|
Liu C, Zhang L, Zhu W, Guo R, Sun H, Chen X, Deng N. Barriers and Strategies of Cationic Liposomes for Cancer Gene Therapy. Mol Ther Methods Clin Dev 2020; 18:751-764. [PMID: 32913882 PMCID: PMC7452052 DOI: 10.1016/j.omtm.2020.07.015] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cationic liposomes (CLs) have been regarded as the most promising gene delivery vectors for decades with the advantages of excellent biodegradability, biocompatibility, and high nucleic acid encapsulation efficiency. However, the clinical use of CLs in cancer gene therapy is limited because of many uncertain factors in vivo. Extracellular barriers such as opsonization, rapid clearance by the reticuloendothelial system and poor tumor penetration, and intracellular barriers, including endosomal/lysosomal entrapped network and restricted diffusion to the nucleus, make CLs not the ideal vector for transferring extrinsic genes in the body. However, the obstacles in achieving productive therapeutic effects of nucleic acids can be addressed by tailoring the properties of CLs, which are influenced by lipid compositions and surface modification. This review focuses on the physiological barriers of CLs against cancer gene therapy and the effects of lipid compositions on governing transfection efficiency, and it briefly discusses the impacts of particle size, membrane charge density, and surface modification on the fate of CLs in vivo, which may provide guidance for their preclinical studies.
Collapse
Affiliation(s)
- Chunyan Liu
- Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Department of Biology, Jinan University, Guangzhou 510632, China
| | - Ligang Zhang
- Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Department of Biology, Jinan University, Guangzhou 510632, China
| | - Wenhui Zhu
- Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Department of Biology, Jinan University, Guangzhou 510632, China
| | - Raoqing Guo
- Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Department of Biology, Jinan University, Guangzhou 510632, China
| | - Huamin Sun
- Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Department of Biology, Jinan University, Guangzhou 510632, China
| | - Xi Chen
- Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Department of Biology, Jinan University, Guangzhou 510632, China
| | - Ning Deng
- Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Department of Biology, Jinan University, Guangzhou 510632, China
| |
Collapse
|
48
|
Kattan WE, Hancock JF. RAS Function in cancer cells: translating membrane biology and biochemistry into new therapeutics. Biochem J 2020; 477:2893-2919. [PMID: 32797215 PMCID: PMC7891675 DOI: 10.1042/bcj20190839] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 02/07/2023]
Abstract
The three human RAS proteins are mutated and constitutively activated in ∼20% of cancers leading to cell growth and proliferation. For the past three decades, many attempts have been made to inhibit these proteins with little success. Recently; however, multiple methods have emerged to inhibit KRAS, the most prevalently mutated isoform. These methods and the underlying biology will be discussed in this review with a special focus on KRAS-plasma membrane interactions.
Collapse
Affiliation(s)
- Walaa E. Kattan
- Department of Integrative Biology and Pharmacology, McGovern Medical School University of Texas Health Science Center at Houston, TX 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, TX 77030, USA
| | - John F. Hancock
- Department of Integrative Biology and Pharmacology, McGovern Medical School University of Texas Health Science Center at Houston, TX 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, TX 77030, USA
| |
Collapse
|
49
|
Chanas-Larue A, Villalpando-Rodriguez GE, Henson ES, Johnston JB, Gibson SB. Antihistamines are synergistic with Bruton's tyrosine kinase inhibiter ibrutinib mediated by lysosome disruption in chronic lymphocytic leukemia (CLL) cells. Leuk Res 2020; 96:106423. [PMID: 32712432 DOI: 10.1016/j.leukres.2020.106423] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 06/30/2020] [Accepted: 07/12/2020] [Indexed: 12/14/2022]
Abstract
Lysosomes in chronic lymphocytic leukemia (CLL) cells have previously been identified as a promising target for therapeutic intervention in combination with targeted therapies. Recent studies have shown that antihistamines can induce lysosomal membrane permeabilization (LMP) in a variety of cell lines. Furthermore, our previous data indicates that lysosomotropic agents can cause synergistic cell death in vitro when combined with some tyrosine kinase inhibitors (TKI). In the current study, we have shown that three over-the-counter antihistamines, clemastine, desloratadine, and loratadine, preferentially induce cell death via LMP in CLL cells, as compared to normal lymphocytes. We treated primary CLL cells with antihistamines and found clemastine was the most effective at inducing LMP and cell death. More importantly, the antihistamines induced synergistic cytotoxicity when combined with the tyrosine kinase inhibitor, ibrutinib, but not with chemotherapy. Moreover, the synergy between clemastine and ibrutinib was associated with the induction of reactive oxygen species (ROS), loss of mitochondrial membrane potential and decreased Mcl-1 expression leading to apoptosis. This study proposes a potential novel treatment strategy for CLL, repurposing FDA-approved allergy medications in combination with the targeted therapy ibrutinib to enhance drug efficacy.
Collapse
Affiliation(s)
- Aaron Chanas-Larue
- Research Institute of Oncology and Hematology, CancerCare Manitoba, Winnipeg, Manitoba, Canada; Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Gloria E Villalpando-Rodriguez
- Research Institute of Oncology and Hematology, CancerCare Manitoba, Winnipeg, Manitoba, Canada; Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Elizabeth S Henson
- Research Institute of Oncology and Hematology, CancerCare Manitoba, Winnipeg, Manitoba, Canada; Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - James B Johnston
- Research Institute of Oncology and Hematology, CancerCare Manitoba, Winnipeg, Manitoba, Canada; Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Spencer B Gibson
- Research Institute of Oncology and Hematology, CancerCare Manitoba, Winnipeg, Manitoba, Canada; Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
50
|
Delasoie J, Pavic A, Voutier N, Vojnovic S, Crochet A, Nikodinovic-Runic J, Zobi F. Identification of novel potent and non-toxic anticancer, anti-angiogenic and antimetastatic rhenium complexes against colorectal carcinoma. Eur J Med Chem 2020; 204:112583. [PMID: 32731186 DOI: 10.1016/j.ejmech.2020.112583] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/06/2020] [Accepted: 06/14/2020] [Indexed: 12/26/2022]
Abstract
Combination therapy targeting both tumor growth and vascularization is considered to be a cornerstone for colorectal carcinomas (CRC) treatment. However, the major obstacles of most clinical anticancer drugs are their weak selective activity towards cancer cells and inherent inner organs toxicity, accompanied with fast drug resistance development. In our effort to discover novel selective and non-toxic agents effective against CRC, we designed, synthesized and characterized a series of rhenium(I) tricarbonyl-based complexes with increased lipophilicity. Two of these novel compounds were discovered to possess remarkable anticancer, anti-angiogenic and antimetastatic activity in vivo (zebrafish-human HCT-116 xenograft model), being effective at very low doses (1-3 μM). At doses as high as 250 μM the complexes did not provoke toxicity issues encountered in clinical anticancer drugs (cardio-, hepato-, and myelotoxicity). In vivo assays showed that the two compounds exceed the anti-tumor and anti-angiogenic activity of clinical drugs cisplatin and sunitinib malate, and display a large therapeutic window.
Collapse
Affiliation(s)
- Joachim Delasoie
- Department of Chemistry, Fribourg University, Chemin Du Musée 9, 1700, Fribourg, Switzerland
| | - Aleksandar Pavic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 152, Belgrade, Republic of Serbia.
| | - Noémie Voutier
- Department of Chemistry, Fribourg University, Chemin Du Musée 9, 1700, Fribourg, Switzerland
| | - Sandra Vojnovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 152, Belgrade, Republic of Serbia
| | - Aurelien Crochet
- Department of Chemistry, Fribourg University, Chemin Du Musée 9, 1700, Fribourg, Switzerland
| | - Jasmina Nikodinovic-Runic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 152, Belgrade, Republic of Serbia.
| | - Fabio Zobi
- Department of Chemistry, Fribourg University, Chemin Du Musée 9, 1700, Fribourg, Switzerland.
| |
Collapse
|