1
|
Liu N, Liu S, Zhang X, Tian W, Jia H, Ye X, Yan X, Yu C, Yu H. Zinc finger domain of p62/SQSTM1 is involved in the necroptosis of human cisplatin‑resistant ovarian cancer cells treated with sulfasalazine. Oncol Lett 2024; 28:529. [PMID: 39290957 PMCID: PMC11406577 DOI: 10.3892/ol.2024.14662] [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: 02/19/2024] [Accepted: 08/08/2024] [Indexed: 09/19/2024] Open
Abstract
Cisplatin resistance in ovarian cancer cells is mainly apoptosis resistant. Although other types of programmed cell death are highly involved in chemoresistance, which type can overcome cisplatin resistance remains unclear. The present study observed that cisplatin-sensitive SKOV3 cells and cisplatin-resistant SKOV3/DDP cells had different levels of sensitivity to sulfasalazine (SAS). The present study aimed to investigate the effect of SAS on necroptosis under the same inhibition rate in these two types of cells. Necroptosis inhibitor Necrostatin-1 (Nec-1) attenuated SAS-induced SKOV3/DDP cytotoxicity. SAS decreased SKOV3/DDP cells survival rate, accompanied by decreased cell adhesion and spreading. SAS treatment activated necrosome formation in SKOV3/DDP cells, suggesting the possibility of necroptosis. p62/sequestosome-1 (SQSTM1) protein expression levels were also increased over the same time period. The transfection of small interfering (si)-p62 could decrease the ratios of phosphorylated (p)-receptor-interacting serine/threonine kinase 1 (RIP1)/RIP1, p-receptor-interacting serine/threonine kinase 3 (RIP3)/RIP3 and p-mixed lineage kinase domain-like protein (MLKL)/MLKL proteins in SKOV3/DDP cells. Under the si-p62 condition, there was no increase in the rate of cell survival in Nec-1 and SAS combination group compared with SAS. The zinc finger domain deletion of p62/SQSTM1 effectively decreased the expression levels of necroptosis-related p-proteins. Collectively, certain drugs were able to induce necroptosis in SKOV3/DDP, while p62/RIP1/RIP3/MLKL was associated with the induction of necroptosis and with increasing the sensitivity of cisplatin-resistant ovarian cancer cells, which provided evidence for potential as a therapeutic target for overcoming resistance.
Collapse
Affiliation(s)
- Nannan Liu
- Department of Pathology, College of Basic Medicine, Beihua University, Jilin, Jilin 132013, P.R. China
| | - Shanshan Liu
- Department of Pathology, College of Basic Medicine, Beihua University, Jilin, Jilin 132013, P.R. China
| | - Xueshuang Zhang
- Department of Transplantation, Hebei Yanda Ludaopei Hospital, Langfang, Hebei 065200, P.R. China
| | - Wenzhu Tian
- Department of Hematology, Changchun Central Hospital, Changchun, Jilin 130041, P.R. China
| | - Heqiang Jia
- Department of Pathology, College of Basic Medicine, Beihua University, Jilin, Jilin 132013, P.R. China
| | - Xin Ye
- Department of Pathology, College of Basic Medicine, Beihua University, Jilin, Jilin 132013, P.R. China
| | - Xiaoyu Yan
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Chunyan Yu
- Department of Pathology, College of Basic Medicine, Beihua University, Jilin, Jilin 132013, P.R. China
| | - Huimei Yu
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
2
|
Liu Y, Teng X, Yan Y, Zhao S, Wang G. Dexmedetomidine promotes necroptosis by upregulating PARP1 in non-small cell lung cancer. Biotechnol Genet Eng Rev 2024; 40:1281-1301. [PMID: 37066722 DOI: 10.1080/02648725.2023.2193469] [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: 02/09/2023] [Accepted: 03/16/2023] [Indexed: 04/18/2023]
Abstract
The score and prognostic value of necroptosis were analyzed in the TCGA and GSE120622 datasets. Necroptosis has the highest correlation with the immune microenvironment, and the high score in NSCLC correlates with poor prognosis. Differentially expressed genes between non-small cell lung cancer (NSCLC) and controls in both datasets were identified and subjected to construct co-expression networks, respectively. Black and blue modules were selected because of high correction with necroptosis. The intersected two module genes were mainly involved in immune and inflammatory response, cell cycle process and DNA replication. Nine marker genes of necroptosis were identified in these modules and considered as candidate genes. Based on candidate genes, we identified two clusters utilizing concordance clustering, additionally dividing NSCLC samples into high- and low-risk groups. There were significant differences in overall survival between two clusters and between high- and low-risk groups. Furthermore, PARP1 was found among the candidate genes to be the target gene of dexmedetomidine acting on necroptosis. Molecular experimental results found that PARP1 was highly expressed in the dexmedetomidine treated NSCLC compared with the NSCLC. Candidate genes associated with necroptosis may provide a powerful prognostic tool for precision oncology. Dexmedetomidine may target PARP1 to promote necroptosis and then affect NSCLC.
Collapse
Affiliation(s)
- Yang Liu
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xiaodan Teng
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yubo Yan
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Su Zhao
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Guonian Wang
- Department of Anesthesiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
3
|
Zhang Z, Zhu J, Wang J, Chen J, Pang Y, Wu C. A novel bakuchiol aminoguanidine derivative induces apoptosis in human triple-negative breast cancer cells. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:509-518. [PMID: 39183056 PMCID: PMC11375495 DOI: 10.3724/zdxbyxb-2024-0070] [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] [Indexed: 08/27/2024]
Abstract
OBJECTIVES To synthesize new bakuchiol aminoguanidine derivatives and test their effect on viability and apoptosis of human triple-negative breast cancer (TNBC) cells. METHODS Two bakuchiol derivatives 1 and 2 were obtained by formylation and Shiff base reaction of bakuchol. The structures of derivatives 1 and 2 were identified by 1H-NMR, 13C-NMR, and high-resolution electrospray ionization mass spectrometry (HR-ESI-MS) analysis. Human TNBC MDA-MB-231 cells were treated with bakuchiol and its derivatives and cell viability was measured by MTT assay. Apoptosis was detected by fluorescence microscopy and flow cytometry with Annexin V-FITC/PI staining. The expressions of apoptosis-related proteins were analyzed with Western blotting. The JC-1 and reactive oxygen species (ROS) assay kits were used to determine the effect of new bakuchiol derivatives on mitochondrial function. RESULTS Based on spectroscopic analysis, a new bakuchiol schiff base derivative was elucidated as 2-{(E)-5-[(S, E)-3, 7-dimethyl-3-vinylocta-1, 6-dien-1-yl]-2-hydroxylbenzylidene} hydrazine-1-carboximidamide (derivative 2). Bakuchiol and its derivatives 1 and 2 all showed cytotoxic activity against the MDA-MB-231 cells. Derivative 2 exhibited the most potent cytotoxic activity to MDA-MB-231 cell with IC50 of (13.11±1.09), (6.91±1.78), and (2.23±1.32) μmol/L after 24, 48, and 72 h. It had low toxicity to normal mouse liver (AML-12) cells with IC50 of (31.23±1.58) μmol/L at 72 h. Fluorescence microscopy and flow cytometry demonstrated apoptosis in breast cancer cells after treating with derivative 2 in a concentration dependent manner. Western blotting showed that after derivative 2 treatment, the expression of apoptosis-related proteins cytochrome C, cleaving caspase-3 and Bax/Bcl-2 radio in MDA-MB-231 cells increased; in addition, apoptosis was associated with the decreased mitochondrial membrane potential and increased reactive oxygen species accumulation. CONCLUSIONS The novel bakuchiol aminoguanidine derivative (derivative 2) is capable of inducing apoptosis in MDA-MB-231 cells, but has low toxicity to normal liver cells, suggesting that it may be used as a lead compound for an anti-TNBC agent.
Collapse
Affiliation(s)
- Zhenhai Zhang
- Department of Emergency Surgery, the First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, Anhui Province, China.
| | - Jing Zhu
- School of Pharmacy, Bengbu Medical University, Bengbu 233030, Anhui Province, China
| | - Jian'an Wang
- School of Pharmacy, Bengbu Medical University, Bengbu 233030, Anhui Province, China
| | - Jie Chen
- School of Pharmacy, Bengbu Medical University, Bengbu 233030, Anhui Province, China
| | - Yingying Pang
- Department of Respiratory Disease, the First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, Anhui Province, China.
| | - Chengzhu Wu
- School of Pharmacy, Bengbu Medical University, Bengbu 233030, Anhui Province, China.
- Anhui Provincial Biochemical Pharmaceutical Technology Research Center, Bengbu 233030, Anhui Province, China.
| |
Collapse
|
4
|
Chakraborty S, Wei D, Tran M, Lang FF, Newman RA, Yang P. PBI-05204, a supercritical CO 2 extract of Nerium oleander, suppresses glioblastoma stem cells by inhibiting GRP78 and inducing programmed necroptotic cell death. Neoplasia 2024; 54:101008. [PMID: 38823209 PMCID: PMC11177059 DOI: 10.1016/j.neo.2024.101008] [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: 12/19/2023] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 06/03/2024]
Abstract
Successful treatment of glioblastoma multiforme (GBM), an aggressive form of primary brain neoplasm, mandates the need to develop new therapeutic strategies. In this study, we investigated the potential of PBI-05204 in targeting GBM stem cells (GSCs) and the underlying mechanisms. Treatment with PBI-05204 significantly reduced both the number and size of tumor spheres derived from patient-derived GSCs (GBM9, GSC28 and TS543), and suppressed the tumorigenesis of GBM9 xenografts. Moreover, PBI-05204 treatment led to a significant decrease in the expression of CD44 and NANOG, crucial markers of progenitor stem cells, in GBM9 and GSC28 GSCs. This treatment also down-regulated GRP78 expression in both GSC types. Knocking down GRP78 expression through GRP78 siRNA transfection in GBM9 and GSC28 GSCs also resulted in reduced spheroid size and CD44 expression. Combining PBI-05204 with GRP78 siRNA further decreased spheroid numbers compared to GRP78 siRNA treatment alone. PBI-05204 treatment led to increased expression of pRIP1K and pRIP3K, along with enhanced binding of RIPK1/RIPK3 in GBM9 and GSC28 cells, resembling the effects observed in GRP78-silenced GSCs, suggesting that PBI-05204 induced necroptosis in these cells. Furthermore, oleandrin, a principle active cardiac glycoside component of PBI-05204, showed the ability to inhibit the self-renewal capacity in GSCs. These findings highlight the potential of PBI-05204 as a promising candidate for the development of novel therapies that target GBM stem cells.
Collapse
Affiliation(s)
- Sharmistha Chakraborty
- Department of Palliative, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Daoyan Wei
- Department of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Megan Tran
- Department of Palliative, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Frederick F Lang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Robert A Newman
- Phoenix Biotechnology, San Antonio, Texas 78217, United States
| | - Peiying Yang
- Department of Palliative, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States.
| |
Collapse
|
5
|
Chen Y, Luo X, Xu B, Bao X, Jia H, Yu B. Oxidative Stress-Mediated Programmed Cell Death: a Potential Therapy Target for Atherosclerosis. Cardiovasc Drugs Ther 2024; 38:819-832. [PMID: 36522550 DOI: 10.1007/s10557-022-07414-z] [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] [Accepted: 12/04/2022] [Indexed: 12/23/2022]
Abstract
Nowadays, as a type of orderly and active death determined by genes, programmed cell death (PCD), including apoptosis, pyroptosis, ferroptosis, and necroptosis, has attracted much attention owing to its participation in numerous chronic cardiovascular diseases, especially atherosclerosis (AS), a canonical chronic inflammatory disease featured by lipid metabolism disturbance. Abundant researches have reported that PCD under distinct internal conditions fulfills different roles of atherosclerotic pathological processes, including lipid core expansion, leukocyte adhesion, and infiltration. Noteworthy, emerging evidence recently has also suggested that oxidative stress (OS), an imbalance of antioxidants and oxygen free radicals, has the potential to mediate PCD occurrence via multiple ways, including oxidization and deubiquitination. Interestingly, more recently, several studies have proposed that the mediating mechanisms could effect on the atherosclerotic initiation and progression significantly from variable aspects, so it is of great clinical importance to clarify how OS-mediated PCD and AS interact. Herein, with the aim of summarizing potential and sufficient atherosclerotic therapy targets, we seek to provide extensive analysis of the specific regulatory mechanisms of PCD mediated by OS and their multifaceted effects on the entire pathological atherosclerotic progression.
Collapse
Affiliation(s)
- Yuwu Chen
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Xing Luo
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Biyi Xu
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Xiaoyi Bao
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Haibo Jia
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China.
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, People's Republic of China.
| | - Bo Yu
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, People's Republic of China
| |
Collapse
|
6
|
Vo TTT, Peng TY, Nguyen TH, Bui TNH, Wang CS, Lee WJ, Chen YL, Wu YC, Lee IT. The crosstalk between copper-induced oxidative stress and cuproptosis: a novel potential anticancer paradigm. Cell Commun Signal 2024; 22:353. [PMID: 38970072 PMCID: PMC11225285 DOI: 10.1186/s12964-024-01726-3] [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: 04/11/2024] [Accepted: 06/25/2024] [Indexed: 07/07/2024] Open
Abstract
Copper is a crucial trace element that plays a role in various pathophysiological processes in the human body. Copper also acts as a transition metal involved in redox reactions, contributing to the generation of reactive oxygen species (ROS). Under prolonged and increased ROS levels, oxidative stress occurs, which has been implicated in different types of regulated cell death. The recent discovery of cuproptosis, a copper-dependent regulated cell death pathway that is distinct from other known regulated cell death forms, has raised interest to researchers in the field of cancer therapy. Herein, the present work aims to outline the current understanding of cuproptosis, with an emphasis on its anticancer activities through the interplay with copper-induced oxidative stress, thereby providing new ideas for therapeutic approaches targeting modes of cell death in the future.
Collapse
Affiliation(s)
- Thi Thuy Tien Vo
- Faculty of Dentistry, Nguyen Tat Thanh University, Ho Chi Minh City, 700000, Vietnam
| | - Tzu-Yu Peng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan
| | - Thi Hong Nguyen
- Faculty of Dentistry, Nguyen Tat Thanh University, Ho Chi Minh City, 700000, Vietnam
| | - Trang Ngoc Huyen Bui
- Faculty of Dentistry, Nguyen Tat Thanh University, Ho Chi Minh City, 700000, Vietnam
| | - Ching-Shuen Wang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan
| | - Wei-Ju Lee
- School of Food Safety, College of Nutrition, Taipei Medical University, Taipei, 110301, Taiwan
| | - Yuh-Lien Chen
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, 100233, Taiwan
| | - Yang-Che Wu
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan
| | - I-Ta Lee
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan.
| |
Collapse
|
7
|
Gu SC, Xie ZG, Gu MJ, Wang CD, Xu LM, Gao C, Yuan XL, Wu Y, Hu YQ, Cao Y, Ye Q. Myricetin mitigates motor disturbance and decreases neuronal ferroptosis in a rat model of Parkinson's disease. Sci Rep 2024; 14:15107. [PMID: 38956066 PMCID: PMC11219851 DOI: 10.1038/s41598-024-62910-6] [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: 02/19/2024] [Accepted: 05/22/2024] [Indexed: 07/04/2024] Open
Abstract
Ferroptosis is an iron-dependent cell death form characterized by reactive oxygen species (ROS) overgeneration and lipid peroxidation. Myricetin, a flavonoid that exists in numerous plants, exhibits potent antioxidant capacity. Given that iron accumulation and ROS-provoked dopaminergic neuron death are the two main pathological hallmarks of Parkinson's disease (PD), we aimed to investigate whether myricetin decreases neuronal death through suppressing ferroptosis. The PD models were established by intraperitoneally injecting 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) into rats and by treating SH-SY5Y cells with 1-methyl-4-phenylpyridinium (MPP+), respectively. Ferroptosis was identified by assessing the levels of Fe2+, ROS, malondialdehyde (MDA), and glutathione (GSH). The results demonstrated that myricetin treatment effectively mitigated MPTP-triggered motor impairment, dopamine neuronal death, and α-synuclein (α-Syn) accumulation in PD models. Myricetin also alleviated MPTP-induced ferroptosis, as evidenced by decreased levels of Fe2+, ROS, and MDA and increased levels of GSH in the substantia nigra (SN) and serum in PD models. All these changes were reversed by erastin, a ferroptosis activator. In vitro, myricetin treatment restored SH-SY5Y cell viability and alleviated MPP+-induced SH-SY5Y cell ferroptosis. Mechanistically, myricetin accelerated nuclear translocation of nuclear factor E2-related factor 2 (Nrf2) and subsequent glutathione peroxidase 4 (Gpx4) expression in MPP+-treated SH-SY5Y cells, two critical inhibitors of ferroptosis. Collectively, these data demonstrate that myricetin may be a potential agent for decreasing dopaminergic neuron death by inhibiting ferroptosis in PD.
Collapse
Affiliation(s)
- Si-Chun Gu
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China
| | - Zhi-Guo Xie
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China
| | - Min-Jue Gu
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China
| | - Chang-De Wang
- Department of Gynecology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Li-Min Xu
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China
| | - Chen Gao
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China
| | - Xiao-Lei Yuan
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China
| | - You Wu
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China
| | - Yu-Qing Hu
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China
| | - Yang Cao
- Department of Gynecology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, 230 Baoding Road, Shanghai, 200082, China.
| | - Qing Ye
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China.
| |
Collapse
|
8
|
Sciaccotta R, Gangemi S, Penna G, Giordano L, Pioggia G, Allegra A. Potential New Therapies "ROS-Based" in CLL: An Innovative Paradigm in the Induction of Tumor Cell Apoptosis. Antioxidants (Basel) 2024; 13:475. [PMID: 38671922 PMCID: PMC11047475 DOI: 10.3390/antiox13040475] [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: 03/18/2024] [Revised: 04/09/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024] Open
Abstract
Chronic lymphocytic leukemia, in spite of recent advancements, is still an incurable disease; the majority of patients eventually acquire resistance to treatment through relapses. In all subtypes of chronic lymphocytic leukemia, the disruption of normal B-cell homeostasis is thought to be mostly caused by the absence of apoptosis. Consequently, apoptosis induction is crucial to the management of this illness. Damaged biological components can accumulate as a result of the oxidation of intracellular lipids, proteins, and DNA by reactive oxygen species. It is possible that cancer cells are more susceptible to apoptosis because of their increased production of reactive oxygen species. An excess of reactive oxygen species can lead to oxidative stress, which can harm biological elements like DNA and trigger apoptotic pathways that cause planned cell death. In order to upset the balance of oxidative stress in cells, recent therapeutic treatments in chronic lymphocytic leukemia have focused on either producing reactive oxygen species or inhibiting it. Examples include targets created in the field of nanomedicine, natural extracts and nutraceuticals, tailored therapy using biomarkers, and metabolic targets. Current developments in the complex connection between apoptosis, particularly ferroptosis and its involvement in epigenomics and alterations, have created a new paradigm.
Collapse
Affiliation(s)
- Raffaele Sciaccotta
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (R.S.); (G.P.); (L.G.)
| | - Sebastiano Gangemi
- Allergy and Clinical Immunology Unit, Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy;
| | - Giuseppa Penna
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (R.S.); (G.P.); (L.G.)
| | - Laura Giordano
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (R.S.); (G.P.); (L.G.)
| | - Giovanni Pioggia
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 98164 Messina, Italy;
| | - Alessandro Allegra
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (R.S.); (G.P.); (L.G.)
| |
Collapse
|
9
|
Sayed AEDH, Emeish WFA, Bakry KA, Al-Amgad Z, Lee JS, Mansour S. Polystyrene nanoplastic and engine oil synergistically intensify toxicity in Nile tilapia, Oreochromis niloticus : Polystyrene nanoplastic and engine oil toxicity in Nile tilapia. BMC Vet Res 2024; 20:143. [PMID: 38622626 PMCID: PMC11020678 DOI: 10.1186/s12917-024-03987-z] [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: 01/28/2024] [Accepted: 03/23/2024] [Indexed: 04/17/2024] Open
Abstract
Polystyrene nanoplastic (PS-NPs) and Engine oil (EO) pose multiple ecotoxic effects with increasing threat to fish ecosystems. The current study investigated the toxicity of 15 days exposure to PS-NPs and / or EO to explore their combined synergistic effects on Nile tilapia, Oreochromis niloticus (O. niloticus). Hematobiochemical parameters, proinflammatory cytokines, and oxidative stress biomarkers as well as histological alterations were evaluated. The experimental design contained 120 acclimated Nile tilapia distributed into four groups, control, PS-NPs (5 mg/L), EO (1%) and their combination (PS-NPs + EO). After 15-days of exposure, blood and tissue samples were collected from all fish experimental groups. Results indicated that Nile tilapia exposed to PS-NPs and / or EO revealed a significant decrease in almost all the measured hematological parameters in comparison to the control, whereas WBCs and lymphocyte counts were significantly increased in the combined group only. Results clarified that the combined PS-NPs + EO group showed the maximum decrease in RBCs, Hb, MCH and MCHC, and showed the maximum significant rise in interleukin-1β (IL-1β), and interleukin-6 (IL-6) in comparison to all other exposed groups. Meanwhile, total antioxidant capacity (TAC) showed a significant (p < 0.05) decline only in the combination group, whereas reduced glutathione (GSH) showed a significant decline in all exposed groups in comparison to the control. Both malondialdehyde (MDA) and aspartate aminotransferase (AST) showed a significant elevation only in the combination group. Uric acid showed the maximum elevation in the combination group than all other groups, whereas creatinine showed significant elevation in the EO and combination group when compared to the control. Furthermore, the present experiment proved that exposure to these toxicants either individually or in combination is accompanied by pronounced histomorpholgical damage characterized by severe necrosis and hemorrhage of the vital organs of Nile tilapia, additionally extensively inflammatory conditions with leucocytes infiltration. We concluded that combination exposure to both PS-NPs and EO caused severe anemia, extreme inflammatory response, oxidative stress, and lipid peroxidation effects, thus they can synergize with each other to intensify toxicity in fish.
Collapse
Affiliation(s)
- Alaa El-Din H Sayed
- Department of Zoology, Assiut University, Assiut, 71516, Egypt.
- Department of Biotechnology, Molecular Biology Research & Studies Institute, Assiut University, Assiut, 71516, Egypt.
| | | | - Karima A Bakry
- Fish Diseases Department, South Valley University, Qena, Egypt
| | - Zeinab Al-Amgad
- General Authority for Veterinary Services, Qena Veterinary Directorate, Qena, Egypt
| | - Jae-Seong Lee
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon, 16419, South Korea
| | - Salwa Mansour
- Zoology Department, South Valley University, Qena, Egypt
| |
Collapse
|
10
|
Voronina MV, Frolova AS, Kolesova EP, Kuldyushev NA, Parodi A, Zamyatnin AA. The Intricate Balance between Life and Death: ROS, Cathepsins, and Their Interplay in Cell Death and Autophagy. Int J Mol Sci 2024; 25:4087. [PMID: 38612897 PMCID: PMC11012956 DOI: 10.3390/ijms25074087] [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: 02/06/2024] [Revised: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
Cellular survival hinges on a delicate balance between accumulating damages and repair mechanisms. In this intricate equilibrium, oxidants, currently considered physiological molecules, can compromise vital cellular components, ultimately triggering cell death. On the other hand, cells possess countermeasures, such as autophagy, which degrades and recycles damaged molecules and organelles, restoring homeostasis. Lysosomes and their enzymatic arsenal, including cathepsins, play critical roles in this balance, influencing the cell's fate toward either apoptosis and other mechanisms of regulated cell death or autophagy. However, the interplay between reactive oxygen species (ROS) and cathepsins in these life-or-death pathways transcends a simple cause-and-effect relationship. These elements directly and indirectly influence each other's activities, creating a complex web of interactions. This review delves into the inner workings of regulated cell death and autophagy, highlighting the pivotal role of ROS and cathepsins in these pathways and their intricate interplay.
Collapse
Affiliation(s)
- Maya V. Voronina
- Research Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (M.V.V.); (A.S.F.); (E.P.K.); (N.A.K.); (A.P.)
| | - Anastasia S. Frolova
- Research Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (M.V.V.); (A.S.F.); (E.P.K.); (N.A.K.); (A.P.)
- Institute of Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Ekaterina P. Kolesova
- Research Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (M.V.V.); (A.S.F.); (E.P.K.); (N.A.K.); (A.P.)
| | - Nikita A. Kuldyushev
- Research Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (M.V.V.); (A.S.F.); (E.P.K.); (N.A.K.); (A.P.)
| | - Alessandro Parodi
- Research Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (M.V.V.); (A.S.F.); (E.P.K.); (N.A.K.); (A.P.)
| | - Andrey A. Zamyatnin
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- Department of Biological Chemistry, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| |
Collapse
|
11
|
Syed RU, Afsar S, Aboshouk NAM, Salem Alanzi S, Abdalla RAH, Khalifa AAS, Enrera JA, Elafandy NM, Abdalla RAH, Ali OHH, Satheesh Kumar G, Alshammari MD. LncRNAs in necroptosis: Deciphering their role in cancer pathogenesis and therapy. Pathol Res Pract 2024; 256:155252. [PMID: 38479121 DOI: 10.1016/j.prp.2024.155252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 03/02/2024] [Accepted: 03/04/2024] [Indexed: 04/14/2024]
Abstract
Necroptosis, a controlled type of cell death that is different from apoptosis, has become a key figure in the aetiology of cancer and offers a possible target for treatment. A growing number of biological activities, including necroptosis, have been linked to long noncoding RNAs (lncRNAs), a varied family of RNA molecules with limited capacity to code for proteins. The complex interactions between LncRNAs and important molecular effectors of necroptosis, including mixed lineage kinase domain-like pseudokinase (MLKL) and receptor-interacting protein kinase 3 (RIPK3), will be investigated. We will explore the many methods that LncRNAs use to affect necroptosis, including protein-protein interactions, transcriptional control, and post-transcriptional modification. Additionally, the deregulation of certain LncRNAs in different forms of cancer will be discussed, highlighting their dual function in influencing necroptotic processes as tumour suppressors and oncogenes. The goal of this study is to thoroughly examine the complex role that LncRNAs play in controlling necroptotic pathways and how that regulation affects the onset and spread of cancer. In the necroptosis for cancer treatment, this review will also provide insight into the possible therapeutic uses of targeting LncRNAs. Techniques utilising LncRNA-based medicines show promise in controlling necroptotic pathways to prevent cancer from spreading and improve the effectiveness of treatment.
Collapse
Affiliation(s)
- Rahamat Unissa Syed
- Department of Pharmaceutics, College of Pharmacy, University of Ha'il, Hail 81442, Saudi Arabia.
| | - S Afsar
- Department of Virology, Sri Venkateswara University, Tirupathi, Andhra Pradesh 517502, India.
| | - Nayla Ahmed Mohammed Aboshouk
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | | | | | - Amna Abakar Suleiman Khalifa
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - Jerlyn Apatan Enrera
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - Nancy Mohammad Elafandy
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - Randa Abdeen Husien Abdalla
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - Omar Hafiz Haj Ali
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - G Satheesh Kumar
- Department of Pharmaceutical Chemistry, College of Pharmacy, Seven Hills College of Pharmacy, Venkataramapuram, Tirupati, India
| | - Maali D Alshammari
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, Hail 81442, Saudi Arabia
| |
Collapse
|
12
|
Li W, Li T, Pan Y, Li S, Xu G, Zhang Z, Liang H, Yang F. Designing a Mitochondria-Targeted Theranostic Cyclometalated Iridium(III) Complex: Overcoming Cisplatin Resistance and Inhibiting Tumor Metastasis through Necroptosis and Immune Response. J Med Chem 2024; 67:3843-3859. [PMID: 38442035 DOI: 10.1021/acs.jmedchem.3c02227] [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: 03/07/2024]
Abstract
To develop a potential theranostic metal agent to reverse the resistance of cancer cells to cisplatin and effectively inhibit tumor growth and metastasis, we proposed to design a cyclometalated iridium (Ir) complex based on the properties of the tumor environment (TME). To the end, we designed and synthesized a series of Ir(III) 2-hydroxy-1-naphthaldehyde thiosemicarbazone complexes by modifying the hydrogen atom(s) of the N-3 position of 2-hydroxy-1-naphthaldehyde thiosemicarbazone compounds and the structure of cyclometalated Ir(III) dimers and then investigated their structure-activity and structure-fluorescence relationships to obtain an Ir(III) complex (Ir5) with remarkable fluorescence and cytotoxicity to cancer cells. Ir5 not only possesses mitochondria-targeted properties but also overcomes cisplatin resistance and effectively inhibits tumor growth and metastasis in vivo. Besides, we confirmed the anticancer mechanisms of Ir5 acting on different components in the TME: directly killing liver cancer cells by inducing necroptosis and activating the necroptosis-related immune response.
Collapse
Affiliation(s)
- Wenjuan Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Ting Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Ying Pan
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Shanhe Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Gang Xu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Zhenlei Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| |
Collapse
|
13
|
Hsieh MY, Hsu SK, Liu TY, Wu CY, Chiu CC. Melanoma biology and treatment: a review of novel regulated cell death-based approaches. Cancer Cell Int 2024; 24:63. [PMID: 38336727 PMCID: PMC10858604 DOI: 10.1186/s12935-024-03220-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 01/09/2024] [Indexed: 02/12/2024] Open
Abstract
The incidence of melanoma, the most lethal form of skin cancer, has increased due to ultraviolet exposure. The treatment of advanced melanoma, particularly metastatic cases, remains challenging with poor outcomes. Targeted therapies involving BRAF/MEK inhibitors and immunotherapy based on anti-PD1/anti-CTLA4 antibodies have achieved long-term survival rates of approximately 50% for patients with advanced melanoma. However, therapy resistance and inadequate treatment response continue to hinder further breakthroughs in treatments that increase survival rates. This review provides an introduction to the molecular-level pathogenesis of melanoma and offers an overview of current treatment options and their limitations. Cells can die by either accidental or regulated cell death (RCD). RCD is an orderly cell death controlled by a variety of macromolecules to maintain the stability of the internal environment. Since the uncontrolled proliferation of tumor cells requires evasion of RCD programs, inducing the RCD of melanoma cells may be a treatment strategy. This review summarizes studies on various types of nonapoptotic RCDs, such as autophagy-dependent cell death, necroptosis, ferroptosis, pyroptosis, and the recently discovered cuproptosis, in the context of melanoma. The relationships between these RCDs and melanoma are examined, and the interplay between these RCDs and immunotherapy or targeted therapy in patients with melanoma is discussed. Given the findings demonstrating melanoma cell death in response to different stimuli associated with these RCDs, the induction of RCD shows promise as an integral component of treatment strategies for melanoma.
Collapse
Affiliation(s)
- Ming-Yun Hsieh
- Department of Pediatrics, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan
| | - Sheng-Kai Hsu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Tzu-Yu Liu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Chang-Yi Wu
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan.
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
| | - Chien-Chih Chiu
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan.
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan.
| |
Collapse
|
14
|
Ma Q, Wu J, Li H, Ma X, Yin R, Bai L, Tang H, Liu N. The role of TRPV4 in programmed cell deaths. Mol Biol Rep 2024; 51:248. [PMID: 38300413 DOI: 10.1007/s11033-023-09199-2] [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: 10/01/2023] [Accepted: 12/30/2023] [Indexed: 02/02/2024]
Abstract
Programmed cell death is a major life activity of both normal development and disease. Necroptosis is early recognized as a caspase-independent form of programmed cell death followed obviously inflammation. Apoptosis is a gradually recognized mode of cell death that is characterized by a special morphological changes and unique caspase-dependent biological process. Ferroptosis, pyroptosis and autophagy are recently identified non-apoptotic regulated cell death that each has its own characteristics. The transient receptor potential vanilloid 4 (TRPV4) is a kind of nonselective calcium-permeable cation channel, which is received more and more attention in biology studies. It is widely expressed in human tissues and mainly located on the membrane of cells. Several researchers have identified that the influx Ca2+ from TRPV4 acts as a key role in the loss of cells by apoptosis, ferroptosis, necroptosis, pyroptosis and autophagy via mediating endoplasmic reticulum (ER) stress, oxidative stress and inflammation. This effect is bad for the normal function of organs on the one hand, on the other hand, it is benefit for anticancer activities. In this review, we will summarize the current discovery on the role and impact of TRPV4 in these programmed cell death pathological mechanisms to provide a new prospect of gene therapeutic target of related diseases.
Collapse
Affiliation(s)
- Qingjie Ma
- Department of Anesthesiology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China
| | - Jilin Wu
- Department of Anesthesiology, Kunming Children's Hospital, Kunming, 650034, China
| | - Huixian Li
- Department of Anesthesiology, The People's Hospital of Wenshan Zhuang and Miao Minority Autonomous Prefecture, Wenshan, 663099, China
| | - Xiaoshu Ma
- The Second Clinical Medical College of Binzhou Medical College, Binzhou, 256699, China
| | - Renwan Yin
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Liping Bai
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Heng Tang
- Department of Anesthesiology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China
| | - Na Liu
- Department of Anesthesiology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China.
| |
Collapse
|
15
|
Zeng X, Han Z, Chen K, Zeng P, Tang Y, Li L. Single-Cell Analyses Reveal Necroptosis's Potential Role in Neuron Degeneration and Show Enhanced Neuron-Immune Cell Interaction in Parkinson's Disease Progression. PARKINSON'S DISEASE 2023; 2023:5057778. [PMID: 38149092 PMCID: PMC10751163 DOI: 10.1155/2023/5057778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/09/2023] [Accepted: 12/13/2023] [Indexed: 12/28/2023]
Abstract
Parkinson's disease (PD) is a common neuron degenerative disease among the old, characterized by uncontrollable movements and an impaired posture. Although widely investigated on its pathology and treatment, the disease remains incompletely understood. Single-cell RNA sequencing (scRNA-seq) has been applied to the area of PD, providing valuable data for related research. However, few works have taken deeper insights into the causes of neuron death and cell-cell interaction between the cell types in the brain. Our bioinformatics analyses revealed necroptosis-related genes (NRGs) enrichment in neuron degeneration and selecting the cells by NRGs levels showed two subtypes within the main degenerative cell types in the midbrain. NRG-low subtype was largely replaced by NRG-high subtype in the patients, indicating the striking change of cell state related to necroptosis in PD progression. Moreover, we carried out cell-cell interaction analyses between cell types and found that microglia (MG)'s interaction strength with glutamatergic neuron (GLU), GABAergic neuron (GABA), and dopaminergic neuron (DA) was significantly upregulated in PD. Also, MG show much stronger interaction with NRG-high subtypes and a stronger cell killing function in PD samples. Additionally, we identified CLDN11 as a novel interaction pattern specific to necroptosis neurons and MG. We also found LEF1 and TCF4 as key transcriptional regulators in neuron degeneration. These findings suggest that MG were significantly overactivated in PD patients to clear abnormal neurons, especially the NRG-high cells, explaining the neuron inflammation in PD. Our analyses provide insights into the causes of neuron death and inflammation in PD from single-cell resolution, which could be seriously considered in clinical trials.
Collapse
Affiliation(s)
- Xiaomei Zeng
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Sichuan University, Chengdu, China
| | - Zhifen Han
- Department of Ultrasound, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Sichuan, Chengdu, China
| | - Kehan Chen
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Sichuan University, Chengdu, China
| | - Peng Zeng
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Sichuan University, Chengdu, China
| | - Yidan Tang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Sichuan University, Chengdu, China
| | - Lijuan Li
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Sichuan University, Chengdu, China
| |
Collapse
|
16
|
Xie Y, Zhao G, Lei X, Cui N, Wang H. Advances in the regulatory mechanisms of mTOR in necroptosis. Front Immunol 2023; 14:1297408. [PMID: 38164133 PMCID: PMC10757967 DOI: 10.3389/fimmu.2023.1297408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/01/2023] [Indexed: 01/03/2024] Open
Abstract
The mammalian target of rapamycin (mTOR), an evolutionarily highly conserved serine/threonine protein kinase, plays a prominent role in controlling gene expression, metabolism, and cell death. Programmed cell death (PCD) is indispensable for maintaining homeostasis by removing senescent, defective, or malignant cells. Necroptosis, a type of PCD, relies on the interplay between receptor-interacting serine-threonine kinases (RIPKs) and the membrane perforation by mixed lineage kinase domain-like protein (MLKL), which is distinguished from apoptosis. With the development of necroptosis-regulating mechanisms, the importance of mTOR in the complex network of intersecting signaling pathways that govern the process has become more evident. mTOR is directly responsible for the regulation of RIPKs. Autophagy is an indirect mechanism by which mTOR regulates the removal and interaction of RIPKs. Another necroptosis trigger is reactive oxygen species (ROS) produced by oxidative stress; mTOR regulates necroptosis by exploiting ROS. Considering the intricacy of the signal network, it is reasonable to assume that mTOR exerts a bifacial effect on necroptosis. However, additional research is necessary to elucidate the underlying mechanisms. In this review, we summarized the mechanisms underlying mTOR activation and necroptosis and highlighted the signaling pathway through which mTOR regulates necroptosis. The development of therapeutic targets for various diseases has been greatly advanced by the expanding knowledge of how mTOR regulates necroptosis.
Collapse
Affiliation(s)
- Yawen Xie
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Guoyu Zhao
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xianli Lei
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Na Cui
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Hao Wang
- Department of Critical Care Medicine, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
17
|
Krishnan RP, Pandiar D, Ramani P, Jayaraman S. Necroptosis in human cancers with special emphasis on oral squamous cell carcinoma. JOURNAL OF STOMATOLOGY, ORAL AND MAXILLOFACIAL SURGERY 2023; 124:101565. [PMID: 37459966 DOI: 10.1016/j.jormas.2023.101565] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 07/12/2023] [Indexed: 11/06/2023]
Abstract
Necroptosis is a type of caspase independent 'programmed or regulated' necrotic cell death that has a morphological resemblance to necrosis and mechanistic analogy to apoptosis. This type of cell death requires RIPK1, RIPK3, MLKL, death receptors, toll like receptors, interferons, and various other proteins. Necroptosis is implicated in plethora of diseases like rheumatoid arthritis, Alzheimer's disease, Crohn's disease, and head and neck cancers including oral squamous cell carcinoma. Oral carcinomas show dysregulation or mutation of necroptotic proteins, mediate antitumoral immunity, activate immune response and control tumor progression. Necroptosis is known to play a dual role (pro tumorigenic and anti-tumorigenic) in cancer progression and targeting this pathway could be an effective approach in cancer therapy. Necroptosis based chemotherapy has been proposed in malignancies, highlighting the importance of necroptotic pathway to overcome apoptosis resistance and serve as a "fail-safe" pathway to modulate cancer initiation, progression, and metastasis. However, there is dearth of information regarding the use of necroptotic cell death mechanism in the treatment of oral squamous cell carcinoma. In this review, we summarise molecular mechanism of necroptosis, and its protumorigenic and antitumorigenic role in cancers to shed light on the possible therapeutic significance of necroptosis in oral squamous cell carcinoma.
Collapse
Affiliation(s)
| | - Deepak Pandiar
- Department of Oral Pathology and Microbiology, Saveetha Dental College and Hospitals, Chennai, Tamil Nadu.
| | - Pratibha Ramani
- Department of Oral Pathology and Microbiology, Saveetha Dental College and Hospitals, Chennai, Tamil Nadu.
| | - Selvaraj Jayaraman
- Department of Biochemistry, Saveetha Dental College and Hospitals, Chennai, Tamil Nadu.
| |
Collapse
|
18
|
Fang W, Lin H, Chen J, Guo W. A novel necroptosis-related gene signature in acute myeloid leukemia. Hematology 2023; 28:2227491. [PMID: 37367830 DOI: 10.1080/16078454.2023.2227491] [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: 04/08/2023] [Accepted: 06/15/2023] [Indexed: 06/28/2023] Open
Abstract
OBJECTIVE Necroptosis has been reported to play an important role in different cancers, including leukemia. However, biomarkers of necroptosis-related genes (NRGs) that help predict the prognosis of AML are still lacking. Our research aims to build a novel signature of NRGs that could enhance our understanding of the molecular heterogeneity in leukemia. METHOD Gene expression profiles as well as clinical features were downloaded from TCGA and GEO databases. Data analysis were executed using R software version 4.2.1 and GraphPad Prism version 9.0.0. RESULT Univariate cox regression and lasso regression were applied to identify survival-specific genes. Four genes including FADD, PLA2G4A, PYCARD and ZBP1 were considered as independent risk factors that affect the prognosis of patients. Risk scores were calculated according to the coefficient of four genes. Then clinical characteristics and risk scores were enrolled to construct a nomogram. CellMiner was also used to screen potential drugs and analyze the correlations between genes and drug sensitivity. CONCLUSION In general, we established a signature of four genes related to necroptosis that could be helpful for future risk stratification in patients with AML.
Collapse
Affiliation(s)
- Weiyue Fang
- Department of Hematology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Hongdou Lin
- Department of Hematology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Junyi Chen
- Department of Hematology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Wenjian Guo
- Medical and Radiation Oncology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
- Zhejiang Engineering Research Center for Innovation and Application of Intelligent Radiotherapy Technology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| |
Collapse
|
19
|
Kim N, Park CJ, Kim Y, Ryu S, Cho H, Nam Y, Han M, Shin JS, Sim T. Identification of Pyrido[3,4-d]pyrimidine derivatives as RIPK3-Mediated necroptosis inhibitors. Eur J Med Chem 2023; 259:115635. [PMID: 37494773 DOI: 10.1016/j.ejmech.2023.115635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/01/2023] [Accepted: 07/10/2023] [Indexed: 07/28/2023]
Abstract
Necroptosis executed by RIPK3-mediated phosphorylation of MLKL is a programmed necrotic cell death and implicated with various diseases such as sterile inflammation. We designed and synthesized pyrido[3,4-d]pyrimidine derivatives as novel necroptosis inhibitors capable of suppressing the phosphorylation of MLKL. Our SAR studies reveal that 20 possesses comparable inhibitory activity against RIPK3-mediated pMLKL in HT-29 cells relative to GSK872 (2), a representative selective RIPK3 inhibitor. Based on biochemical kinase assay results, 20 is comparable to GSK872 (2) with regard to activity against RIPK3 and less potent against RIPK1 than GSK872, indicating selectivity of 20 towards RIPK3 over RIPK1 is higher than that of GSK872. In HT-29 cells, 20 inhibits necroptosis via MLKL oligomerization impediment. Moreover, 20 suppresses migration and invasion of AsPC-1 cells by necroptosis induced- CXCL5 secretion downregulation. Significantly, 20 could relieve the TNFα-induced systemic inflammatory response syndrome in vivo. Taken together, this study would provide a useful insight into the design of novel necroptosis inhibitors possessing RIPK3-mediated pMLKL inhibitory activity.
Collapse
Affiliation(s)
- Namkyoung Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Severance Biomedical Science Institute, Graduate School of Medicinal Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Chan-Jung Park
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Severance Biomedical Science Institute, Graduate School of Medicinal Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Younghoon Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Severance Biomedical Science Institute, Graduate School of Medicinal Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - SeongShick Ryu
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Severance Biomedical Science Institute, Graduate School of Medicinal Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hanna Cho
- Severance Biomedical Science Institute, Graduate School of Medicinal Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Yunju Nam
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Severance Biomedical Science Institute, Graduate School of Medicinal Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Myeonggil Han
- Department of Microbiology, Yonsei University College of Medicine, Seoul, South Korea
| | - Jeon-Soo Shin
- Department of Microbiology, Yonsei University College of Medicine, Seoul, South Korea
| | - Taebo Sim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Severance Biomedical Science Institute, Graduate School of Medicinal Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
20
|
Ma D, Wang X, Liu J, Cui Y, Luo S, Wang F. The development of necroptosis: what we can learn. Cell Stress Chaperones 2023; 28:969-987. [PMID: 37995025 PMCID: PMC10746674 DOI: 10.1007/s12192-023-01390-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 08/03/2023] [Accepted: 10/17/2023] [Indexed: 11/24/2023] Open
Abstract
Necroptosis is a new type of programmed cell death discovered in recent years, playing an important role in various diseases. Since it was conceptualized in 2005, research on necroptosis has developed rapidly. However, few bibliometric analyses have provided a comprehensive overview of the field. This study aimed to employ a bibliometric analysis to assess necroptosis research's current status and hotspot, highlight landmark findings, and orientate future research. A total of 3993 publications from the WoSCC were collected for this study. Multiple tools were used for bibliometric analysis and data visualization, including an online website, VOSviewer, CiteSpace, and HistCite. Publications related to necroptosis have increased significantly annually, especially in the last 5 years. Globally, the USA and Harvard University are the most outstanding countries and institutions in this field, respectively. The academic groups managed by Peter Vandenabeele and Junying Yuan both have permanent and intensive research on necroptosis. Cell Death and Differentiation is the most vital journal in this field. The molecular mechanisms of necroptosis and its role in disease are the focus of current research, while the crosstalk between programmed cell death is an emerging direction in the field. The "reactive oxygen species", "innate immunity", and "programmed cell death" may be potential research hotspots. Our results present a comprehensive knowledge map and explore research trends. Researchers and funding agencies on necroptosis can obtain helpful references from our study.
Collapse
Affiliation(s)
- Dongbin Ma
- Department of Neurosurgery, Chengdu Fifth People's Hospital, Chengdu, China
| | - Xuan Wang
- Department of Obstetrics, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, China
| | - Jia Liu
- Department of Neurosurgery, Chengdu Fifth People's Hospital, Chengdu, China
| | - Yang Cui
- Department of Neurosurgery, Hebei Yanda Hospital, Langfang, China
| | - Shuang Luo
- Department of Neurosurgery, Chengdu Fifth People's Hospital, Chengdu, China
| | - Fanchen Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
- Department of Graduate School, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
21
|
Li S, Zhang W, Hu X. Comprehensive analysis of necroptosis-related genes in renal ischemia-reperfusion injury. Front Immunol 2023; 14:1279603. [PMID: 37965311 PMCID: PMC10641517 DOI: 10.3389/fimmu.2023.1279603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/16/2023] [Indexed: 11/16/2023] Open
Abstract
Background Oxidative stress is the primary cause of ischemia-reperfusion injury (IRI) in kidney transplantation, leading to delayed graft function (DGF) and implications on patient health. Necroptosis is believed to play a role in renal IRI. This research presents a comprehensive analysis of necroptosis-related genes and their functional implications in the context of IRI in renal transplantation. Methods The necroptosis-related differentially expressed genes (NR-DEGs) were identified using gene expression data from pre- and post-reperfusion renal biopsies, and consensus clustering analysis was performed to distinguish necroptosis-related clusters. A predictive model for DGF was developed based on the NR-DEGs and patients were divided into high- and low-risk groups. We investigated the differences in functional enrichment and immune infiltration between different clusters and risk groups and further validated them in single-cell RNA-sequencing (scRNA-seq) data. Finally, we verified the expression changes of NR-DEGs in an IRI mouse model. Results Five NR-DEGs were identified and were involved in various biological processes. The renal samples were further stratified into two necroptosis-related clusters (C1 and C2) showing different occurrences of DGF. The predictive model had a reliable performance in identifying patients at higher risk of DGF with the area under the curve as 0.798. Additionally, immune infiltration analysis indicated more abundant proinflammatory cells in the high-risk group, which was also found in C2 cluster with more DGF patients. Validation of NR-DEG in scRNA-seq data further supported their involvement in immune cells. Lastly, the mouse model validated the up-regulation of NR-DEGs after IR and indicated the correlations with kidney function markers. Conclusions Our research provides valuable insights into the identification and functional characterization of NR-DEGs in the context of renal transplantation and sheds light on their involvement in immune responses and the progression of IRI and DGF.
Collapse
Affiliation(s)
- Shuai Li
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Weixun Zhang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Xiaopeng Hu
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| |
Collapse
|
22
|
Barar E, Shi J. Genome, Metabolism, or Immunity: Which Is the Primary Decider of Pancreatic Cancer Fate through Non-Apoptotic Cell Death? Biomedicines 2023; 11:2792. [PMID: 37893166 PMCID: PMC10603981 DOI: 10.3390/biomedicines11102792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a solid tumor characterized by poor prognosis and resistance to treatment. Resistance to apoptosis, a cell death process, and anti-apoptotic mechanisms, are some of the hallmarks of cancer. Exploring non-apoptotic cell death mechanisms provides an opportunity to overcome apoptosis resistance in PDAC. Several recent studies evaluated ferroptosis, necroptosis, and pyroptosis as the non-apoptotic cell death processes in PDAC that play a crucial role in the prognosis and treatment of this disease. Ferroptosis, necroptosis, and pyroptosis play a crucial role in PDAC development via several signaling pathways, gene expression, and immunity regulation. This review summarizes the current understanding of how ferroptosis, necroptosis, and pyroptosis interact with signaling pathways, the genome, the immune system, the metabolism, and other factors in the prognosis and treatment of PDAC.
Collapse
Affiliation(s)
- Erfaneh Barar
- Liver and Pancreatobiliary Diseases Research Center, Digestive Disease Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran 1416753955, Iran
| | - Jiaqi Shi
- Department of Pathology & Clinical Labs, Rogel Cancer Center, Center for RNA Biomedicine, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
23
|
He J, Zheng Z, Li S, Liao C, Li Y. Identification and assessment of differentially expressed necroptosis long non-coding RNAs associated with periodontitis in human. BMC Oral Health 2023; 23:632. [PMID: 37667236 PMCID: PMC10478209 DOI: 10.1186/s12903-023-03308-0] [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: 04/02/2023] [Accepted: 08/13/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND Periodontitis is the most common oral disease and is closely related to immune infiltration in the periodontal microenvironment and its poor prognosis is related to the complex immune response. The progression of periodontitis is closely related to necroptosis, but there is still no systematic study of long non-coding RNA (lncRNA) associated with necroptosis for diagnosis and treatment of periodontitis. MATERIAL AND METHODS Transcriptome data and clinical data of periodontitis and healthy populations were obtained from the Gene Expression Omnibus (GEO) database, and necroptosis-related genes were obtained from previously published literature. FactoMineR package in R was used to perform principal component analysis (PCA) for obtaining the necroptosis-related lncRNAs. The core necroptosis-related lncRNAs were screened by the Linear Models for Microarray Data (limma) package in R, PCA principal component analysis and lasso algorithm. These lncRNAs were then used to construct a classifier for periodontitis with logistic regression. The receiver operating characteristic (ROC) curve was used to evaluate the sensitivity and specificity of the model. The CIBERSORT method and ssGSEA algorithm were used to estimate the immune infiltration and immune pathway activation of periodontitis. Spearman's correlation analysis was used to further verify the correlation between core genes and periodontitis immune microenvironment. The expression level of core genes in human periodontal ligament cells (hPDLCs) was detected by RT-qPCR. RESULTS A total of 10 core necroptosis-related lncRNAs (10-lncRNAs) were identified, including EPB41L4A-AS1, FAM30A, LINC01004, MALAT1, MIAT, OSER1-DT, PCOLCE-AS1, RNF144A-AS1, CARMN, and LINC00582. The classifier for periodontitis was successfully constructed. The Area Under the Curve (AUC) was 0.952, which suggested that the model had good predictive performance. The correlation analysis of 10-lncRNAs and periodontitis immune microenvironment showed that 10-lncRNAs had an impact on the immune infiltration of periodontitis. Notably, the RT-qPCR results showed that the expression level of the 10-lncRNAs obtained was consistent with the chip analysis results. CONCLUSIONS The 10-lncRNAs identified from the GEO dataset had a significant impact on the immune infiltration of periodontitis and the classifier based on 10-lncRNAs had good detection efficiency for periodontitis, which provided a new target for diagnosis and treatment of periodontitis.
Collapse
Affiliation(s)
- Jiangfeng He
- Department of Orthodontics, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, 200072, China
| | - Zhanglong Zheng
- Department of Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, 200072, China
| | - Sijin Li
- Department of Orthodontics, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, 200072, China
| | - Chongshan Liao
- Department of Orthodontics, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, 200072, China.
| | - Yongming Li
- Department of Orthodontics, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, 200072, China.
| |
Collapse
|
24
|
Lin W, Mo CQ, Kong LJ, Chen L, Wu KL, Wu X. FTO-mediated epigenetic upregulation of LINC01559 confers cell resistance to docetaxel in breast carcinoma by suppressing miR-1343-3p. Kaohsiung J Med Sci 2023; 39:873-882. [PMID: 37584416 DOI: 10.1002/kjm2.12728] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/27/2023] [Accepted: 06/04/2023] [Indexed: 08/17/2023] Open
Abstract
This study was to explore the regulatory effect of long non-coding RNA LINC01559 on Docetaxel resistance in breast carcinoma (BCa) and its underlying mechanism. In the present study, we found that LINC01559 expression was elevated and LINC01559 overexpression facilitated docetaxel resistance in BCa cells. Moreover, it was revealed that the upregulation of LINC01559 in BCa cells was induced by FTO-mediated demethylation in an m6A-YTHDF2-dependent manner. Additionally, Dual-luciferase reporter assay confirmed the binding ability between LINC01559 and miR-1343-3p, and Pearson correlation analysis showed a negative correlation between them. Particularly, miR-1343-3p inhibition partly abolished the suppression on docetaxel resistance in BCa cells caused by LINC01559 knockdown. To sum up, FTO-mediated epigenetic upregulation of LINC01559 promoted cell resistance to Docetaxel in BCa by negatively regulating miR-1343-3p.
Collapse
Affiliation(s)
- Wei Lin
- Department of Thyroid and Breast Surgery, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Cai-Qin Mo
- Department of Thyroid and Breast Surgery, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Ling-Jun Kong
- Department of Thyroid and Breast Surgery, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Ling Chen
- Department of Thyroid and Breast Surgery, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Kun-Lin Wu
- Department of Thyroid and Breast Surgery, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xian Wu
- Department of Thyroid and Breast Surgery, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
25
|
Orfanoudaki M, Smith EA, Hill NT, Garman KA, Brownell I, Copp BR, Grkovic T, Henrich CJ. An Investigation of Structure-Activity Relationships and Cell Death Mechanisms of the Marine Alkaloids Discorhabdins in Merkel Cell Carcinoma Cells. Mar Drugs 2023; 21:474. [PMID: 37755087 PMCID: PMC10532587 DOI: 10.3390/md21090474] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/28/2023] Open
Abstract
A library of naturally occurring and semi-synthetic discorhabdins was assessed for their effects on Merkel cell carcinoma (MCC) cell viability. The set included five new natural products and semi-synthetic compounds whose structures were elucidated with NMR, HRMS, and ECD techniques. Several discorhabdins averaged sub-micromolar potency against the MCC cell lines tested and most of the active compounds showed selectivity towards virus-positive MCC cell lines. An investigation of structure-activity relationships resulted in an expanded understanding of the crucial structural features of the discorhabdin scaffold. Mechanistic cell death assays suggested that discorhabdins, unlike many other MCC-active small molecules, do not induce apoptosis, as shown by the lack of caspase activation, annexin V staining, and response to caspase inhibition. Similarly, discorhabdin treatment failed to increase MCC intracellular calcium and ROS levels. In contrast, the rapid loss of cellular reducing potential and mitochondrial membrane potential suggested that discorhabdins induce mitochondrial dysfunction leading to non-apoptotic cell death.
Collapse
Affiliation(s)
- Maria Orfanoudaki
- Molecular Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA; (M.O.); (E.A.S.)
| | - Emily A. Smith
- Molecular Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA; (M.O.); (E.A.S.)
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Natasha T. Hill
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, MD 20891, USA; (N.T.H.); (K.A.G.); (I.B.)
| | - Khalid A. Garman
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, MD 20891, USA; (N.T.H.); (K.A.G.); (I.B.)
| | - Isaac Brownell
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, MD 20891, USA; (N.T.H.); (K.A.G.); (I.B.)
| | - Brent R. Copp
- School of Chemical Sciences, University of Auckland, Auckland 1142, New Zealand;
| | - Tanja Grkovic
- Molecular Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA; (M.O.); (E.A.S.)
- Natural Products Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, MD 21702, USA
| | - Curtis J. Henrich
- Molecular Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA; (M.O.); (E.A.S.)
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| |
Collapse
|
26
|
Bow YD, Ko CC, Chang WT, Chou SY, Hung CT, Huang JL, Tseng CH, Chen YL, Li RN, Chiu CC. A novel quinoline derivative, DFIQ, sensitizes NSCLC cells to ferroptosis by promoting oxidative stress accompanied by autophagic dysfunction and mitochondrial damage. Cancer Cell Int 2023; 23:171. [PMID: 37587444 PMCID: PMC10433610 DOI: 10.1186/s12935-023-02984-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 07/01/2023] [Indexed: 08/18/2023] Open
Abstract
BACKGROUND The development of nonapoptotic programmed cell death inducers as anticancer agents has emerged as a cancer therapy field. Ferroptosis, ferrous ion-driven programmed cell death that is induced by redox imbalance and dysfunctional reactive oxygen species (ROS) clearance, is triggered during sorafenib and PD-1/PD-L1 immunotherapy. DFIQ, a quinoline derivative, promotes apoptosis by disrupting autophagic flux and promoting ROS accumulation. Our pilot experiments suggest that DFIQ participates in ferroptosis sensitization. Thus, in this study, we aimed to reveal the mechanisms of DFIQ in ferroptosis sensitization and evaluate the clinical potential of DFIQ. METHODS We treated the non-small cell lung cancer (NSCLC) cell lines H1299, A549, and H460 with the ferroptosis inducer (FI) DFIQ and analyzed viability, protein expression, ROS generation, and fluorescence staining at different time points. Colocalization analysis was performed with ImageJ. RESULTS DFIQ sensitized cells to FIs such as erastin and RSL3, resulting in a decrease in IC50 of at least 0.5-fold. Measurement of ROS accumulation to explore the underlying mechanism indicated that DFIQ and FIs treatment promoted ROS accumulation and SOD1/SOD2 switching. Mitochondria, known ROS sources, produced high ROS levels during DFIQ/FI treatment. RSL3 treatment promoted mitochondrial damage and mitophagy, an autophagy-associated mitochondrial recycling system, and cotreatment with DFIQ induced accumulation of mitochondrial proteins, which indicated disruption of mitophagic flux. Thus, autophagic flux was measured in cells cotreated with DFIQ. DFIQ treatment was found to disrupt autophagic flux, leading to accumulation of damaged mitochondria and eventually inducing ferroptosis. Furthermore, the influence of DFIQ on the effects of clinical FIs, such as sorafenib, was evaluated, and DFIQ was discovered to sensitize NSCLC cells to sorafenib and promote ferroptosis. CONCLUSIONS This study indicates that DFIQ not only promotes NSCLC apoptosis but also sensitizes cells to ferroptosis by disrupting autophagic flux, leading to accumulation of dysfunctional mitochondria and thus to ferroptosis. Ferroptosis is a novel therapeutic target in cancer therapy. DFIQ shows the potential to enhance the effects of FIs in NSCLC and act as a potential therapeutic adjuvant in ferroptosis-mediated therapy.
Collapse
Affiliation(s)
- Yung-Ding Bow
- PhD Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Ching-Chung Ko
- Department of Medical Imaging, Chi Mei Medical Center, Tainan, 71004, Taiwan
- Department of Health and Nutrition, Chia Nan University of Pharmacy and Science, Tainan, 71710, Taiwan
| | - Wen-Tsan Chang
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Sih-Yan Chou
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Chun-Tzu Hung
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Jau-Ling Huang
- Department of Bioscience Technology, College of Health Science, Chang Jung Christian University, Tainan, 71101, Taiwan
| | - Chih-Hua Tseng
- School of Pharmacy, College of Pharmacy, Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Yeh-Long Chen
- Department of Medicinal and Applied Chemistry, Drug Development and Value Creation Research Center, Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| | - Ruei-Nian Li
- Department of Biomedical Science and Environment Biology, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan.
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan.
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, 11571, Taiwan.
| |
Collapse
|
27
|
Xie Y, Lei X, Zhao G, Guo R, Cui N. mTOR in programmed cell death and its therapeutic implications. Cytokine Growth Factor Rev 2023; 71-72:66-81. [PMID: 37380596 DOI: 10.1016/j.cytogfr.2023.06.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/12/2023] [Accepted: 06/19/2023] [Indexed: 06/30/2023]
Abstract
Mechanistic target of rapamycin (mTOR), a highly conserved serine/threonine kinase, is involved in cellular metabolism, protein synthesis, and cell death. Programmed cell death (PCD) assists in eliminating aging, damaged, or neoplastic cells, and is indispensable for sustaining normal growth, fighting pathogenic microorganisms, and maintaining body homeostasis. mTOR has crucial functions in the intricate signaling pathway network of multiple forms of PCD. mTOR can inhibit autophagy, which is part of PCD regulation. Cell survival is affected by mTOR through autophagy to control reactive oxygen species production and the degradation of pertinent proteins. Additionally, mTOR can regulate PCD in an autophagy-independent manner by affecting the expression levels of related genes and phosphorylating proteins. Therefore, mTOR acts through both autophagy-dependent and -independent pathways to regulate PCD. It is conceivable that mTOR exerts bidirectional regulation of PCD, such as ferroptosis, according to the complexity of signaling pathway networks, but the underlying mechanisms have not been fully explained. This review summarizes the recent advances in understanding mTOR-mediated regulatory mechanisms in PCD. Rigorous investigations into PCD-related signaling pathways have provided prospective therapeutic targets that may be clinically beneficial for treating various diseases.
Collapse
Affiliation(s)
- Yawen Xie
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xianli Lei
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Guoyu Zhao
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Ran Guo
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Na Cui
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.
| |
Collapse
|
28
|
Li J, Yi X, Liu L, Wang X, Ai J. Advances in tumor nanotechnology: theragnostic implications in tumors via targeting regulated cell death. Apoptosis 2023:10.1007/s10495-023-01851-3. [PMID: 37184582 DOI: 10.1007/s10495-023-01851-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2023] [Indexed: 05/16/2023]
Abstract
Cell death constitutes an indispensable part of the organismal balance in the human body. Generally, cell death includes regulated cell death (RCD) and accidental cell death (ACD), reflecting the intricately molecule-dependent process and the uncontrolled response, respectively. Furthermore, diverse RCD pathways correlate with multiple diseases, such as tumors and neurodegenerative diseases. Meanwhile, with the development of precision medicine, novel nano-based materials have gradually been applied in the clinical diagnosis and treatment of tumor patients. As the carrier, organic, inorganic, and biomimetic nanomaterials could facilitate the distribution, improve solubility and bioavailability, enhance biocompatibility and decrease the toxicity of drugs in the body, therefore, benefiting tumor patients with better survival outcomes and quality of life. In terms of the most studied cell death pathways, such as apoptosis, necroptosis, and pyroptosis, plenty of studies have explored specific types of nanomaterials targeting the molecules and signals in these pathways. However, no attempt was made to display diverse nanomaterials targeting different RCD pathways comprehensively. In this review, we elaborate on the potential mechanisms of RCD, including intrinsic and extrinsic apoptosis, necroptosis, ferroptosis, pyroptosis, autophagy-dependent cell death, and other cell death pathways together with corresponding nanomaterials. The thorough presentation of RCD pathways and diverse nano-based materials may provide a wider cellular and molecular landscape of tumor diagnosis and treatments.
Collapse
Affiliation(s)
- Jin Li
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Xianyanling Yi
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Liangren Liu
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, China.
| | - Xiaohui Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China.
| | - Jianzhong Ai
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
29
|
Chettri A, Yang T, Cole HD, Shi G, Cameron CG, McFarland SA, Dietzek-Ivanšić B. Using Biological Photophysics to Map the Excited-State Topology of Molecular Photosensitizers for Photodynamic Therapy. Angew Chem Int Ed Engl 2023; 62:e202301452. [PMID: 36827484 PMCID: PMC10079593 DOI: 10.1002/anie.202301452] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 02/26/2023]
Abstract
This study employs TLD1433, a RuII -based photodynamic therapy (PDT) agent in human clinical trials, as a benchmark to establish protocols for studying the excited-state dynamics of photosensitizers (PSs) in cellulo, in the local environment provided by human cancer cells. Very little is known about the excited-state properties of any PS in live cells, and for TLD1433, it is terra incognita. This contribution targets a general problem in phototherapy, which is how to interrogate the light-triggered, function-determining processes of the PSs in the relevant biological environment, and establishes methodological advances to study the ultrafast photoinduced processes for TLD1433 when taken up by MCF7 cells. We generalize the methodological developments and results in terms of molecular physics by applying them to TLD1433's analogue TLD1633, making this study a benchmark to investigate the excited-state dynamics of phototoxic compounds in the complex biological environment.
Collapse
Affiliation(s)
- Avinash Chettri
- Department Functional Interfaces, Leibniz-Institute of Photonic Technology Jena, Albert-Einstein-Straße 9, 07745, Jena, Germany
- Institute of Physical Chemistry, Friedrich-Schiller University Jena, Helmholtzweg 4, 07743, Jena, Germany
| | - Tingxiang Yang
- Department Functional Interfaces, Leibniz-Institute of Photonic Technology Jena, Albert-Einstein-Straße 9, 07745, Jena, Germany
- Institute of Physical Chemistry, Friedrich-Schiller University Jena, Helmholtzweg 4, 07743, Jena, Germany
| | - Houston D Cole
- Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX-76019, USA
| | - Ge Shi
- Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX-76019, USA
| | - Colin G Cameron
- Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX-76019, USA
| | - Sherri A McFarland
- Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX-76019, USA
| | - Benjamin Dietzek-Ivanšić
- Department Functional Interfaces, Leibniz-Institute of Photonic Technology Jena, Albert-Einstein-Straße 9, 07745, Jena, Germany
- Institute of Physical Chemistry, Friedrich-Schiller University Jena, Helmholtzweg 4, 07743, Jena, Germany
| |
Collapse
|
30
|
Gupta R, Kumari S, Tripathi R, Ambasta RK, Kumar P. Unwinding the modalities of necrosome activation and necroptosis machinery in neurological diseases. Ageing Res Rev 2023; 86:101855. [PMID: 36681250 DOI: 10.1016/j.arr.2023.101855] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/09/2022] [Accepted: 01/15/2023] [Indexed: 01/20/2023]
Abstract
Necroptosis, a regulated form of cell death, is involved in the genesis and development of various life-threatening diseases, including cancer, neurological disorders, cardiac myopathy, and diabetes. Necroptosis initiates with the formation and activation of a necrosome complex, which consists of RIPK1, RIPK2, RIPK3, and MLKL. Emerging studies has demonstrated the regulation of the necroptosis cell death pathway through the implication of numerous post-translational modifications, namely ubiquitination, acetylation, methylation, SUMOylation, hydroxylation, and others. In addition, the negative regulation of the necroptosis pathway has been shown to interfere with brain homeostasis through the regulation of axonal degeneration, mitochondrial dynamics, lysosomal defects, and inflammatory response. Necroptosis is controlled by the activity and expression of signaling molecules, namely VEGF/VEGFR, PI3K/Akt/GSK-3β, c-Jun N-terminal kinases (JNK), ERK/MAPK, and Wnt/β-catenin. Herein, we briefly discussed the implication and potential of necrosome activation in the pathogenesis and progression of neurological manifestations, such as Alzheimer's disease, Parkinson's disease, multiple sclerosis, traumatic brain injury, and others. Further, we present a detailed picture of natural compounds, micro-RNAs, and chemical compounds as therapeutic agents for treating neurological manifestations.
Collapse
Affiliation(s)
- Rohan Gupta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly Delhi College of Engineering), India
| | - Smita Kumari
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly Delhi College of Engineering), India
| | - Rahul Tripathi
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly Delhi College of Engineering), India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly Delhi College of Engineering), India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly Delhi College of Engineering), India.
| |
Collapse
|
31
|
Pei Z, Lei H, Cheng L. Bioactive inorganic nanomaterials for cancer theranostics. Chem Soc Rev 2023; 52:2031-2081. [PMID: 36633202 DOI: 10.1039/d2cs00352j] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Bioactive materials are a special class of biomaterials that can react in vivo to induce a biological response or regulate biological functions, thus achieving a better curative effect than traditional inert biomaterials. For cancer theranostics, compared with organic or polymer nanomaterials, inorganic nanomaterials possess unique physical and chemical properties, have stronger mechanical stability on the basis of maintaining certain bioactivity, and are easy to be compounded with various carriers (polymer carriers, biological carriers, etc.), so as to achieve specific antitumor efficacy. After entering the nanoscale, due to the nano-size effect, high specific surface area and special nanostructures, inorganic nanomaterials exhibit unique biological effects, which significantly influence the interaction with biological organisms. Therefore, the research and applications of bioactive inorganic nanomaterials in cancer theranostics have attracted wide attention. In this review, we mainly summarize the recent progress of bioactive inorganic nanomaterials in cancer theranostics, and also introduce the definition, synthesis and modification strategies of bioactive inorganic nanomaterials. Thereafter, the applications of bioactive inorganic nanomaterials in tumor imaging and antitumor therapy, including tumor microenvironment (TME) regulation, catalytic therapy, gas therapy, regulatory cell death and immunotherapy, are discussed. Finally, the biosafety and challenges of bioactive inorganic nanomaterials are also mentioned, and their future development opportunities are prospected. This review highlights the bioapplication of bioactive inorganic nanomaterials.
Collapse
Affiliation(s)
- Zifan Pei
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China.
| | - Huali Lei
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China.
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China.
| |
Collapse
|
32
|
Yang P, Zhou X, Xie Y. Cytotoxic Effects of the Benzophenanthridine Alkaloids Isolated from Eomecon chionantha on MCF-7 Cells and Its Potential Mechanism. Chem Biodivers 2023; 20:e202200871. [PMID: 36529680 DOI: 10.1002/cbdv.202200871] [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: 09/16/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Seven benzophenanthridine alkaloids (1-7) were obtained from the 75 % EtOH extract of Eomecon chionantha, and exhibited moderate biological activity against MCF-7 cells. 8,12-dimethoxysanguinarine (1, DSG) strongly decreased the cell viability of MCF-7 cell lines with an IC50 value of 7.12 μΜ. Based on RNA-sequencing measure and KEGG pathway enrichment analysis results, the significant differentially expressed genes (DEGs) were associated with Pathways in Cancer and PI3 K-AKT signaling pathways in DSG treated group. The potential molecular regulatory mechanisms underlying the effect of DSG induces necroptosis in MCF-7 cells via molecular docking, TEM analysis, and ROS measurement. Besides, DEGs of bone metastasis-related genes such as PI3 K, IGF1R, Notch, and Wnt mRNA were significantly downregulated in the DSG-treated group on MCF-7 cells. DSG might be selected as a bone metastasis proteins inhibitor of IL-1β, IL-6, IκBα, IGF1R, Notch, NF-κB, PTHrp, PI3 K, PKB/AKT, PTEN, TNF-α, and Wnt via molecular docking. DSG suppressed bone metastasis by regulating the expression levels of IL-1β, IL-6, PTH, CROSS, TP1NP, and OSTEOC on MCF-7 cells using ELISA measurement. Thus, our findings reveal that DSG could be a lead compound for suppressing tumor cells to bone metastasis in breast cancer cells.
Collapse
Affiliation(s)
- Peng Yang
- Hunan Provincial Key Laboratory for Synthetic Biology of Traditional Chinese Medicine, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua, 418000, P. R. China
| | - Xi Zhou
- School of Life Sciences, Central South University, Changsha, 410008, P. R. China
| | - Yang Xie
- Xiangya Hospital, Central South University, Changsha, 410008, P. R. China
| |
Collapse
|
33
|
Tkachenko A, Havránek O. Redox Status of Erythrocytes as an Important Factor in Eryptosis and Erythronecroptosis. Folia Biol (Praha) 2023; 69:116-126. [PMID: 38410969 DOI: 10.14712/fb2023069040116] [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] [Indexed: 02/28/2024]
Abstract
Overall, reactive oxygen species (ROS) signalling significantly contributes to initiation and mo-dulation of multiple regulated cell death (RCD) pathways. Lately, more information has become available about RCD modalities of erythrocytes, including the role of ROS. ROS accumulation has therefore been increasingly recognized as a critical factor involved in eryptosis (apoptosis of erythrocytes) and erythro-necroptosis (necroptosis of erythrocytes). Eryptosis is a Ca2+-dependent apoptosis-like RCD of erythrocytes that occurs in response to oxidative stress, hyperosmolarity, ATP depletion, and a wide range of xenobiotics. Moreover, eryptosis seems to be involved in the pathogenesis of multiple human diseases and pathological processes. Several studies have reported that erythrocytes can also undergo necroptosis, a lytic RIPK1/RIPK3/MLKL-mediated RCD. As an example, erythronecroptosis can occur in response to CD59-specific pore-forming toxins. We have systematically summarized available studies regarding the involvement of ROS and oxidative stress in these two distinct RCDs of erythrocytes. We have focused specifically on cellular signalling pathways involved in ROS-mediated cell death decisions in erythrocytes. Furthermore, we have summarized dysregulation of related erythrocytic antioxidant defence systems. The general concept of the ROS role in eryptotic and necroptotic cell death pathways in erythrocytes seems to be established. However, further studies are required to uncover the complex role of ROS in the crosstalk and interplay between the survival and RCDs of erythrocytes.
Collapse
Affiliation(s)
- Anton Tkachenko
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Ondřej Havránek
- 1st Department of Medicine - Department of Haematology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic.
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic.
| |
Collapse
|
34
|
Li Y, Zhang X, Wang Z, Li B, Zhu H. Modulation of redox homeostasis: A strategy to overcome cancer drug resistance. Front Pharmacol 2023; 14:1156538. [PMID: 37033606 PMCID: PMC10073466 DOI: 10.3389/fphar.2023.1156538] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/13/2023] [Indexed: 04/11/2023] Open
Abstract
Cancer treatment is hampered by resistance to conventional therapeutic strategies, including chemotherapy, immunotherapy, and targeted therapy. Redox homeostasis manipulation is one of the most effective innovative treatment techniques for overcoming drug resistance. Reactive oxygen species (ROS), previously considered intracellular byproducts of aerobic metabolism, are now known to regulate multiple signaling pathways as second messengers. Cancer cells cope with elevated amounts of ROS during therapy by upregulating the antioxidant system, enabling tumor therapeutic resistance via a variety of mechanisms. In this review, we aim to shed light on redox modification and signaling pathways that may contribute to therapeutic resistance. We summarized the molecular mechanisms by which redox signaling-regulated drug resistance, including altered drug efflux, action targets and metabolism, enhanced DNA damage repair, maintained stemness, and reshaped tumor microenvironment. A comprehensive understanding of these interrelationships should improve treatment efficacy from a fundamental and clinical research point of view.
Collapse
Affiliation(s)
- Yang Li
- State Key Laboratory of Biotherapy and Cancer Center, West China School of Basic Medical Sciences and Forensic Medicine, West China Hospital, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Xiaoyue Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China School of Basic Medical Sciences and Forensic Medicine, West China Hospital, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Zhihan Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China School of Basic Medical Sciences and Forensic Medicine, West China Hospital, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Bowen Li
- State Key Laboratory of Biotherapy and Cancer Center, West China School of Basic Medical Sciences and Forensic Medicine, West China Hospital, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Huili Zhu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, Department of Reproductive Medicine, West China Second University Hospital of Sichuan University, Chengdu, China
- *Correspondence: Huili Zhu,
| |
Collapse
|
35
|
Marji SM, Bayan MF, Jaradat A. Facile Fabrication of Methyl Gallate Encapsulated Folate ZIF-L Nanoframeworks as a pH Responsive Drug Delivery System for Anti-Biofilm and Anticancer Therapy. Biomimetics (Basel) 2022; 7:biomimetics7040242. [PMID: 36546942 PMCID: PMC9775553 DOI: 10.3390/biomimetics7040242] [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: 11/19/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Zeolitic imidazole frameworks are emerging materials and have been considered an efficient platform for biomedical applications. The present study highlights the simple fabrication of methyl gallate encapsulated folate-ZIF-L nanoframeworks (MG@Folate ZIF-L) by a simple synthesis. The nanoframeworks were characterized by different sophisticated instruments. In addition, the drug-releasing mechanism was evidenced by in vitro releasing kinetics at various pH conditions. The anti-biofilm potential confirmed by the biofilm architectural deformations against human infectious pathogens MRSA and N7 clinical strains. Furthermore, anticancer efficacy assessed against A549 lung cancer cells. The result reveals that the MG@Folate ZIF-L exposed a superior cytotoxic effect due to the pH-responsive and receptor-based drug-releasing mechanism. Based on the unique physicochemical and biological characteristics of nanoframeworks, it has overcome the problems of undesired side effects and uncontrolled drug release of existing drug delivery systems. Finally, the in vitro toxicity effect of MG@Folate ZIF-L was tested against the Artemia salina (A. salina) model organism, and the results show enhanced biocompatibility. Overall, the study suggested that the novel MG@Folate ZIF-L nanoframeworks is a suitable material for biomedical applications. It will be very helpful to the future design for targeted drug delivery systems.
Collapse
Affiliation(s)
- Saeed M. Marji
- Faculty of Pharmacy, Philadelphia University, P.O. Box 1, Amman 19392, Jordan
- Correspondence: (S.M.M.); (M.F.B.)
| | - Mohammad F. Bayan
- Faculty of Pharmacy, Philadelphia University, P.O. Box 1, Amman 19392, Jordan
- Correspondence: (S.M.M.); (M.F.B.)
| | - Abdolelah Jaradat
- Faculty of Pharmacy, Isra University, P.O. Box 33, Amman 11622, Jordan
| |
Collapse
|
36
|
Magnolia officinalis Bark Extract Prevents Enterocyte Death in a Colitis Mouse Model by Inhibiting ROS-Mediated Necroptosis. Antioxidants (Basel) 2022; 11:antiox11122435. [PMID: 36552643 PMCID: PMC9774795 DOI: 10.3390/antiox11122435] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Necroptosis is a form of programmed cell death with features of necrosis and apoptosis that occurs in the intestinal epithelium of patients with inflammatory bowel disease (IBD), including ulcerative colitis and Crohn's disease. In addition, necroptosis has also been observed in enterocytes in animal models of dextran sulfate sodium (DSS)-induced colitis. Thus, the discovery of natural products for regulating necroptosis may represent an important therapeutic strategy for improving IBD. We found that Magnolia officinalis bark extract (MBE) prevented weight loss and suppressed the activation of the proinflammatory cytokine IL6 in DSS-induced colitis. Furthermore, MBE restored the length of the damaged colon and decreased the expression of necroptosis markers in mice with DSS-induced colitis. In vitro, necroptosis-induced reactive oxygen species (ROS) production was reduced by MBE, and the expression of COX2, a target protein of ROS, was simultaneously suppressed. Both magnolol and honokiol, the two major bioactive compounds in MBE, inhibited necroptosis in human primary intestinal epithelial cells and colorectal adenocarcinoma cells. Our findings highlight the effectiveness of MBE in modulating enterocyte necroptosis and suggest that MBE may be developed as a natural, disease-targeting drug for the treatment of colitis.
Collapse
|
37
|
Scabertopin Derived from Elephantopus scaber L. Mediates Necroptosis by Inducing Reactive Oxygen Species Production in Bladder Cancer In Vitro. Cancers (Basel) 2022; 14:cancers14235976. [PMID: 36497458 PMCID: PMC9738305 DOI: 10.3390/cancers14235976] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/15/2022] [Accepted: 12/01/2022] [Indexed: 12/11/2022] Open
Abstract
Bladder cancer remains one of the most common malignant tumors that threatens human health worldwide. It imposes a heavy burden on patients and society due to the high medical costs associated with its easy metastasis and recurrence. Although several treatment options for bladder cancer are available, their clinical efficacy remains unsatisfactory. Therefore, actively exploring new drugs and their mechanisms of action for the clinical treatment of bladder cancer is very important. Scabertopin is one of the major sesquiterpene lactones found in Elephantopus scaber L. Sesquiterpene lactones are thought to have fairly strong anti-cancer efficacy. However, the anticancer effect of sesquiterpenoid scabertopin on bladder cancer and its mechanism are still unclear. The aim of this study is to evaluate the antitumor activity of scabertopin in bladder cancer and its potential molecular mechanism in vitro. Our results suggest that scabertopin can induce RIP1/RIP3-dependent necroptosis in bladder cancer cells by promoting the production of mitochondrial reactive oxygen species (ROS), inhibit the expression of MMP-9 by inhibiting the FAK/PI3K/Akt signaling pathway, and ultimately inhibit the migration and invasion ability of bladder cancer cells. At the same time, we also demonstrated that the half-inhibition concentration (IC50) of scabertopin on various bladder cancer cell lines (J82, T24, RT4 and 5637) is much lower than that on human ureteral epithelial immortalized cells (SV-HUC-1). The above observations indicate that scabertopin is a potential therapeutic agent for bladder cancer that acts by inducing necroptosis and inhibiting metastasis.
Collapse
|
38
|
Li Y, Chen G. Upconversion Nanoparticles for Cancer Therapy. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Yang Li
- School of Chemistry and Chemical Engineering Harbin Institute of Technology Harbin 150001 China
| | - Guanying Chen
- School of Chemistry and Chemical Engineering Harbin Institute of Technology Harbin 150001 China
| |
Collapse
|
39
|
Tang X, Fan X, Xu T, He Y, Chi Q, Li Z, Li S. Polystyrene nanoplastics exacerbated lipopolysaccharide-induced necroptosis and inflammation via the ROS/MAPK pathway in mice spleen. ENVIRONMENTAL TOXICOLOGY 2022; 37:2552-2565. [PMID: 35833596 DOI: 10.1002/tox.23618] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/30/2022] [Accepted: 07/03/2022] [Indexed: 06/15/2023]
Abstract
Plastics are novel environmental pollutants with potential threats to the ecosystem. At least 5.25 trillion plastic particles in the environment, of which nanoplastics are <100 nm in diameter. Polystyrene nanoplastics (PS-NPs) exposure damaged the spleen's immune function. Lipopolysaccharide (LPS) induced other toxicants to damage cells and organs, triggering inflammation. However, the mechanism of PS-NPs aggravated LPS-induced spleen injury remains unclear. In this study, the PS-NPs or/and LPS mice exposure model was replicated by intraperitoneal injection of PS-NPs or/and LPS, and PS-NPs or/and LPS were exposed to RAW264.7 cells. The histopathological and ultrastructural changes of the mice spleen were observed by H&E staining and transmission electron microscope. Western Blot, qRT-PCR, and fluorescent probes staining were used to detect reactive oxygen species (ROS), oxidative stress indicators, inflammatory factors, and necroptosis-related indicators in mice spleen and RAW264.7 cells. The results showed that PS-NPs or LPS induced oxidative stress, activated the MAPK pathway, and eventually caused necroptosis and inflammation in mice spleen and RAW264.7 cells. Compared with the single treatment group, the changes in PS-NPs + LPS group were more obvious. Furthermore, ROS inhibitor N-Acetyl-L-cysteine (NAC) significantly inhibited the activation of the mitogen-activated protein kinase (MAPK) signaling pathway caused by co-treatment of PS-NPs and LPS, reducing necroptosis and inflammation. The results demonstrated that PS-NPs promoted LPS-induced spleen necroptosis and inflammation in mice through the ROS/MAPK pathway. This study increases the data on the damage of PS-NPs to the organism and expands the research ideas and clues.
Collapse
Affiliation(s)
- Xinyu Tang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Xue Fan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Tong Xu
- College of Chemistry, Jilin University, Changchun, People's Republic of China
| | - Yujiao He
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Qianru Chi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Zhe Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Shu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| |
Collapse
|
40
|
Wu L, Lu H, Pan Y, Liu C, Wang J, Chen B, Wang Y. The role of pyroptosis and its crosstalk with immune therapy in breast cancer. Front Immunol 2022; 13:973935. [PMID: 36119049 PMCID: PMC9477010 DOI: 10.3389/fimmu.2022.973935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022] Open
Abstract
Pyroptosis is a brand-new category of programmed cell death (PCD) that is brought on by multitudinous inflammasomes, which can recognize several stimuli to pilot the cleavage of and activate inflammatory cytokines like IL-18 and IL-1β is believed to have dual effects on the development of multiple cancers including breast cancer. However, pyroptosis has different effects on cancers depending on the type of tissues and their distinct heredity. Recently, the association between pyroptosis and breast cancer has received more and more attention, and it is thought that inducing pyroptosis could be used as a cancer treatment option. In addition, a great deal of evidence accumulating over the past decades has evinced the crosstalk between pyroptosis and tumor immunological therapy. Thus, a comprehensive summary combining the function of pyroptosis in breast cancer and antitumor immunity is imperative. We portray the prevalent knowledge of the multidimensional roles of pyroptosis in cancer and summarize the pyroptosis in breast cancer principally. Moreover, we elucidate the influence of inflammasomes and pyroptosis-produced cytokines on the tumor microenvironment (TME) of breast cancer. Taken together, we aim to provide a clue to harness pyroptosis rationally and apply it to augment immunotherapy efficiency for breast cancer.
Collapse
Affiliation(s)
- Ling Wu
- Department of Clinical Laboratory Medicine, Taizhou Central Hospital (Taizhou Univesity Hospital), Taizhou, China
- Medical College, Yangzhou University, Yangzhou, China
| | - Hongsheng Lu
- Department of Clinical Laboratory Medicine, Taizhou Central Hospital (Taizhou Univesity Hospital), Taizhou, China
| | - Yin Pan
- Department of Clinical Laboratory Medicine, Taizhou Central Hospital (Taizhou Univesity Hospital), Taizhou, China
| | - Chen Liu
- Department of Clinical Laboratory Medicine, Taizhou Central Hospital (Taizhou Univesity Hospital), Taizhou, China
| | - Jinyan Wang
- Department of Clinical Laboratory Medicine, Taizhou Central Hospital (Taizhou Univesity Hospital), Taizhou, China
| | - Baofu Chen
- Department of Clinical Laboratory Medicine, Taizhou Central Hospital (Taizhou Univesity Hospital), Taizhou, China
| | - Yichao Wang
- Department of Clinical Laboratory Medicine, Taizhou Central Hospital (Taizhou Univesity Hospital), Taizhou, China
| |
Collapse
|
41
|
Zhu L, Li HD, Xu JJ, Li JJ, Cheng M, Meng XM, Huang C, Li J. Advancements in the Alcohol-Associated Liver Disease Model. Biomolecules 2022; 12:biom12081035. [PMID: 36008929 PMCID: PMC9406170 DOI: 10.3390/biom12081035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 02/06/2023] Open
Abstract
Alcohol-associated liver disease (ALD) is an intricate disease that results in a broad spectrum of liver damage. The presentation of ALD can include simple steatosis, steatohepatitis, liver fibrosis, cirrhosis, and even hepatocellular carcinoma (HCC). Effective prevention and treatment strategies are urgently required for ALD patients. In previous decades, numerous rodent models were established to investigate the mechanisms of alcohol-associated liver disease and explore therapeutic targets. This review provides a summary of the latest developments in rodent models, including those that involve EtOH administration, which will help us to understand the characteristics and causes of ALD at different stages. In addition, we discuss the pathogenesis of ALD and summarize the existing in vitro models. We analyse the pros and cons of these models and their translational relevance and summarize the insights that have been gained regarding the mechanisms of alcoholic liver injury.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiao-Ming Meng
- Correspondence: (X.-M.M.); (C.H.); (J.L.); Tel.: +86-551-65161001 (J.L.); Fax: +86-551-65161001 (J.L.)
| | - Cheng Huang
- Correspondence: (X.-M.M.); (C.H.); (J.L.); Tel.: +86-551-65161001 (J.L.); Fax: +86-551-65161001 (J.L.)
| | - Jun Li
- Correspondence: (X.-M.M.); (C.H.); (J.L.); Tel.: +86-551-65161001 (J.L.); Fax: +86-551-65161001 (J.L.)
| |
Collapse
|
42
|
Osthole Induces Apoptosis and Caspase-3/GSDME-Dependent Pyroptosis via NQO1-Mediated ROS Generation in HeLa Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8585598. [PMID: 35720178 PMCID: PMC9200556 DOI: 10.1155/2022/8585598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 04/06/2022] [Accepted: 05/16/2022] [Indexed: 12/24/2022]
Abstract
Osthole is a natural coumarin which has been proved to inhibit growth of cancer cells by inducing cell death, while its mechanism was considered to be just caused by apoptosis. In our study, we found that osthole activated not just apoptosis, but also pyroptosis which is a form of regulated cell death accompanied by loss of cell membrane integrity and lactate dehydrogenase (LDH) release. Caspase-3 is a key protein of apoptosis as well as pyroptosis. The apoptosis and pyroptosis induced by osthole were all inhibited by irreversible caspase-3 inhibitor Z-DEVD-FMK. Meanwhile, knockdown of gasdermin E (GSDME) only reduced the osthole-induced pyroptosis but did not affect the occurrence of apoptosis. Our proteomic analysis revealed that the expression of NAD(P)H: quinone oxidoreductase 1 (NQO1) was decreased in osthole-treated cells. Moreover, NQO1 inhibition by osthole induced the overproduction of reactive oxygen species (ROS), as well as apoptosis and pyroptosis. ROS inhibitor N-Acetyl-L-cysteine (NAC) not only reduced osthole-induced apoptosis but also reversed its effect on the pyroptosis. Meanwhile, knockdown of NQO1 by si-NQO1 or its inhibitor dicoumarol (DIC) not only enhanced ROS generation but also strengthened the GSDME-mediated pyroptosis. Finally, we demonstrated that osthole inhibited tumor growth and the expression of NQO1 in a HeLa xenograft mode. Similar to the results in vitro, osthole stimulated the activation of caspase-3, PARP, and GSDME in vivo. Taken together, all these data suggested that osthole induced apoptosis and caspase-3/GSDME-mediated pyroptosis via NQO1-mediated ROS accumulation.
Collapse
|
43
|
Hsu SK, Chu YH, Syue WJ, Lin HYH, Chang WT, Chen JYF, Wu CY, Yen CH, Cheng KC, Chiu CC. The Role of Nonapoptotic Programmed Cell Death — Ferroptosis, Necroptosis, and Pyroptosis — in Pancreatic Ductal Adenocarcinoma Treatment. Front Oncol 2022; 12:872883. [PMID: 35664778 PMCID: PMC9160188 DOI: 10.3389/fonc.2022.872883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 04/19/2022] [Indexed: 11/26/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most lethal cancer, with a dismal 5-year survival rate of less than 10%. It is estimated that approximately 80% of pancreatic ductal carcinoma (PDAC) patients are diagnosed at an advanced or metastatic stage. Hence, most patients are not appropriate candidates for surgical resection and therefore require systemic chemotherapy. However, it has been reported that most patients develop chemoresistance within several months, partly because of antiapoptotic mechanisms. Hence, inducing alternative programmed cell death (PCD), including ferroptosis, necroptosis or pyroptosis, seems to be a promising strategy to overcome antiapoptosis-mediated chemoresistance. In this review, we shed light on the molecular mechanisms of ferroptosis, necroptosis and pyroptosis and suggest several potential strategies (e.g., compounds and nanoparticles [NPs]) that are capable of triggering nonapoptotic PCD to suppress PDAC progression. In conclusion, these strategies might serve as adjuvants in combination with clinical first-line chemotherapies to improve patient survival rates.
Collapse
Affiliation(s)
- Sheng-Kai Hsu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Hsuan Chu
- Department of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wun-Jyun Syue
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hugo You-Hsien Lin
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wen-Tsan Chang
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Jeff Yi-Fu Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chang-Yi Wu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Chia-Hung Yen
- The Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Kai-Chun Cheng
- Department of Ophthalmology, Kaohsiung Municipal Siaogang Hospital, Kaohsiung, Taiwan
- Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Ophthalmology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- *Correspondence: Kai-Chun Cheng, ; Chien-Chih Chiu,
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- The Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- *Correspondence: Kai-Chun Cheng, ; Chien-Chih Chiu,
| |
Collapse
|
44
|
Maurici CE, Colenbier R, Wylleman B, Brancato L, van Zwol E, Van den Bossche J, Timmermans JP, Giovannetti E, Mori da Cunha MGMC, Bogers J. Hyperthermia Enhances Efficacy of Chemotherapeutic Agents in Pancreatic Cancer Cell Lines. Biomolecules 2022; 12:651. [PMID: 35625581 PMCID: PMC9138677 DOI: 10.3390/biom12050651] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 02/01/2023] Open
Abstract
Chemotherapy (CT) is the standard care for advanced pancreatic ductal adenocarcinoma (PDAC); however, with limited efficacy. Hyperthermia (HT) treatment has been suggested as a sensitizer to improve outcomes. However, the direct effect of the HT and CT combination is not fully understood. Therefore, we aim to assess the direct cytotoxic effect of HT in PDAC cells as monotherapy or in combination with chemotherapeutics. Different temperatures (37-, 40.5-, 41-, and 41.5 °C) and durations (6-, 12-, and 24 h) were tested in PDAC cell lines (BxPC-3, Capan-1, Capan-2, PANC-1, and MIA-PaCa-2). Different concentrations of gemcitabine, 5-fluorouracil, and cisplatin were also tested in these conditions. The impact on cell metabolic activity was determined by an MTS assay. Enhancement of chemosensitivity was assessed by a reduction in half-maximal inhibitory concentration (IC50). HT and chemotherapeutics interactions were classified as antagonistic, additive, or synergistic using the combination index. HT inhibited cell proliferation in a cell type, temperature, and duration-dependent manner. The induction of apoptosis was seen after 6 h of HT treatment, eventually followed by secondary necrosis. The HT and CT combination led to an IC50 reduction of the tested CT. At 12 h of HT, this effect was between 25 to 90% and reached a 95% reduction at 24 h. The additive or synergistic effect was demonstrated in all cell lines and chemotherapeutics, although, again, this depended on cell type, duration, and temperature. HT is cytotoxic and enhances the therapeutic effectiveness of gemcitabine, 5-fluorouracil, and cisplatin on PDAC cells. This result was further confirmed by the decrease in the expression of RRM2, TS, and ERCC1 in BxPC-3 and Capan-2 cells. These observations warrant further study in specific subsets of PDAC patients to improve their clinical outcomes.
Collapse
Affiliation(s)
- Costanza E. Maurici
- ElmediX NV, 2800 Mechelen, Belgium; (C.E.M.); (B.W.); (L.B.); (E.v.Z.); (J.V.d.B.); (M.G.M.C.M.d.C.)
| | - Robin Colenbier
- Laboratory of Cell Biology and Histology, Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium; (R.C.); (J.-P.T.)
- Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam UMC, 1006 Amsterdam, The Netherlands;
| | - Britta Wylleman
- ElmediX NV, 2800 Mechelen, Belgium; (C.E.M.); (B.W.); (L.B.); (E.v.Z.); (J.V.d.B.); (M.G.M.C.M.d.C.)
| | - Luigi Brancato
- ElmediX NV, 2800 Mechelen, Belgium; (C.E.M.); (B.W.); (L.B.); (E.v.Z.); (J.V.d.B.); (M.G.M.C.M.d.C.)
| | - Eke van Zwol
- ElmediX NV, 2800 Mechelen, Belgium; (C.E.M.); (B.W.); (L.B.); (E.v.Z.); (J.V.d.B.); (M.G.M.C.M.d.C.)
| | - Johan Van den Bossche
- ElmediX NV, 2800 Mechelen, Belgium; (C.E.M.); (B.W.); (L.B.); (E.v.Z.); (J.V.d.B.); (M.G.M.C.M.d.C.)
| | - Jean-Pierre Timmermans
- Laboratory of Cell Biology and Histology, Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium; (R.C.); (J.-P.T.)
| | - Elisa Giovannetti
- Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam UMC, 1006 Amsterdam, The Netherlands;
- Cancer Pharmacology Lab, Fondazione Pisana per la Scienza, 56017 Pisa, Italy
| | | | - Johannes Bogers
- ElmediX NV, 2800 Mechelen, Belgium; (C.E.M.); (B.W.); (L.B.); (E.v.Z.); (J.V.d.B.); (M.G.M.C.M.d.C.)
- Laboratory of Cell Biology and Histology, Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium; (R.C.); (J.-P.T.)
- Applied Molecular Biology Research Group (AMBIOR), University of Antwerp, 2610 Antwerp, Belgium
| |
Collapse
|
45
|
Deciphering a Novel Necroptosis-Related miRNA Signature for Predicting the Prognosis of Clear Cell Renal Carcinoma. Anal Cell Pathol (Amst) 2022; 2022:2721005. [PMID: 35509814 PMCID: PMC9061065 DOI: 10.1155/2022/2721005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/05/2022] [Indexed: 11/28/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common histological and devastating subtype of renal cell carcinoma. Necroptosis is a form of programmed cell death that causes prominent inflammatory responses. miRNAs play a significant role in cancer progression through necroptosis. However, the prognostic value of necroptosis-related miRNAs remains ambiguous. In this study, 39 necroptosis-related miRNAs (NRMs) were extracted and 17 differentially expressed NRMs between normal and tumor samples were identified using data form The Cancer Genome Atlas (TCGA). After applying univariate Cox proportional hazard regression analysis and LASSO Cox regression model, six necroptosis-related miRNA signatures were identified in the training cohort and their expression levels were verified by qRT-PCR. Using the expression levels of these miRNAs, all patients were divided into the high- and low-risk groups. Patients in the high-risk group showed poor overall survival (P < 0.0001). Time-dependent ROC curves confirmed the good performance of our signature. The results were verified in the testing cohort and the entire TCGA cohort. Univariate and multivariate Cox regression models demonstrated that the risk score was an independent prognostic factor. Additionally, a predictive nomogram with good performance was constructed to enhance the implementation of the constructed signature in a clinical setting. We then employed miRBD, miRTarBase, and TargetScan to predict the target genes of six necroptosis-related miRNAs. Gene ontology and Kyoto Encyclopedia of Genes and Genomes analyses indicated that 392 potential target genes were enriched in cell proliferation-related biological processes. Six miRNAs and 59 differentially expressed target genes were used to construct an miRNA–mRNA interaction network, and 11 hub genes were selected for survival and tumor infiltration analysis. Drug sensitivity analysis revealed potential drugs that may contribute to cancer management. Hence, necroptosis-related genes play an important role in cancer biology. We developed, for the first time, a necroptosis-related miRNA signature to predict ccRCC prognosis.
Collapse
|
46
|
Zhao W, Song Y, Wang QQ, Han S, Li XX, Cui Y, Gao H, Yuan R, Yang S. Cryptotanshinone Induces Necroptosis through Ca2+ Release and ROS Production in vitro and in vivo. Curr Mol Pharmacol 2022; 15:1009-1023. [PMID: 35086466 DOI: 10.2174/1874467215666220127112201] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/15/2021] [Accepted: 11/01/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Necroptosis is a type of programmed necrosis mediated by receptor-interacting protein kinases 1 and 3 (RIP1 and RIP3), which is morphologically characterized by enlarged organelles, ruptured plasma membrane, and subsequent loss of intracellular contents. Cryptotanshinone (CPT), a diterpene quinone compound extracted from the root of Salvia miltiorrhiza Bunge, has been reported to have significant anticancer activities. However, the detailed mechanism of CPT has not been clearly illustrated. OBJECTIVE The present study aimed to explore the cell death type and mechanisms of CPT-induced in non-small cell lung cancer (NSCLC) cells. METHODS The cytotoxicity of CPT on A549 cells was assessed by MTS assay. Ca2+ release and reactive oxygen species (ROS) generation were detected by flow cytometry. The changes in mitochondrial membrane potential (MMP) were observed through JC-1 staining. The expressions of p-RIP1, p-RIP3, p-MLKL, and MAPKs pathway proteins were analyzed by western blotting analysis. The efficacy of CPT in vivo was evaluated by the Lewis lung carcinoma (LLC) xenograft mice model. Blood samples were collected for hematology analysis. ELISA investigated the effects of CPT on tumor necrosis factor α (TNF-α). Hematoxylin and eosin staining (HE) was used to determine the tumor tissues. Proteins' expression of tumor tissues was quantified by western blotting. RESULTS CPT inhibited the cell viability of A549 cells in a time- and concentration-dependent manner, which was reversed by Necrostatin-1 (Nec-1). In addition, CPT treatment increased the expression of p-RIP1, p-RIP3, p-MLKL, the release of Ca2+, ROS generation, and the MAPKs pathway activated in A549 cells. Moreover, animal experiment results showed that intraperitoneal injection of CPT (15 mg/kg and 30 mg/kg) significantly inhibited tumor growth in C57BL/6 mice without affecting the bodyweight and injuring the organs. CONCLUSION Our findings suggested that CPT-induced necroptosis via RIP1/RIP3/MLKL signaling pathway both in vitro and in vivo, indicating that CPT may be a promising agent in the treatment of NSCLC.
Collapse
Affiliation(s)
- Wentong Zhao
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530000, China
- Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Yuanbo Song
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine
| | - Qin-Qin Wang
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning 530200, China
| | - Shan Han
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530000, China
- Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Xin-Xing Li
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530000, China
- Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Yushun Cui
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning 530200, China
| | - Hongwei Gao
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530000, China
- Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Renyikun Yuan
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530000, China
- Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Shilin Yang
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530000, China
- Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| |
Collapse
|
47
|
Zhou Y, Wu R, Wang X, Bao X, Lu C. Roles of necroptosis in alcoholic liver disease and hepatic pathogenesis. Cell Prolif 2022; 55:e13193. [PMID: 35083817 PMCID: PMC8891559 DOI: 10.1111/cpr.13193] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 12/30/2021] [Accepted: 01/06/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic alcohol consumption can cause alcoholic liver disease (ALD), leading to morbidity and mortality worldwide. Complex disease progression of ALD varies from alcoholic fatty liver to alcoholic steatohepatitis, eventually contributing to fibrosis and cirrhosis. Accumulating evidence revealed that necroptosis, a way of programmed cell death different from apoptosis and traditional necrosis, is involved in the underlying pathogenic molecular mechanism of ALD. Receptor‐interacting protein kinase 1 (RIPK1), RIPK3 and mixed‐lineage kinase domain‐like pseudokinase have been implicated as key mediators to execute necroptosis. Also, necroptosis has gained increasing attention due to its potential association with primary pathological hallmarks of ALD, including oxidative stress, hepatic steatosis and inflammation. This review summarizes the recent progress on the roles and mechanisms of necroptosis and focuses on the crosstalk between necroptosis and the other pathogenesis of ALD, providing a theoretical basis for targeting necroptosis as a novel treatment for ALD.
Collapse
Affiliation(s)
- Ying Zhou
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Ruoman Wu
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Xinqi Wang
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Xiaofeng Bao
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Chunfeng Lu
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
48
|
Liu YP, Lei J, Yin MM, Chen Y. Organoantimony (III) Derivative induces necroptosis in human breast cancer MDA-MB-231 cells. Anticancer Agents Med Chem 2022; 22:2448-2457. [PMID: 35040419 DOI: 10.2174/1871520622666220118093643] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 11/01/2021] [Accepted: 12/09/2021] [Indexed: 11/22/2022]
Abstract
PURPOSE This study aimed to investigate the anticancer effect and the underlying mechanisms of organoantimony (III) fluoride on MDA-MB-231 human breast cancer cells. METHODS Five cancer and one normal cell line were treated with an organoantimony (III) compound 6-cyclohexyl-12-fluoro-5,6,7,12-tetrahydrodibenzo[c,f][1,5]azastibocine (denoted as C4). The cell viability was detected by MTT assay. Induction of cell death was determined by Hoechst 33342/PI staining and Annexin-V/PI staining. The effect of C4 on the necroptotic relative protein was determined by Western blot analysis. RESULTS Among the five cancer cell lines, C4 decreased the viability of MDA-MB-231, MCF-7 and A2780/cisR, and showed less toxicity to normal human embryonic kidney cells. In breast cancer cell line MDA-MB-231, the C4 treatment induced the percentage of necrotic cell death as well as LDH releasing in a time- and dose-dependent manner. Moreover, C4 could increase the expression of phosphorylated RIPK3 and MLKL proteins. Overall, the C4 treatment resulted in reduction of mitochondrial transmembrane potential and accumulation ROS in MDA-MB-231 cells. CONCLUSION C4-induced necroptosis could be ascribed to glutathione depletion and ROS elevation in MDA-MB-231 cells. Our findings illustrate that C4 is a potential necroptosis inducer for breast cancer treatment.
Collapse
Affiliation(s)
- Yong-Ping Liu
- Department of Physiology, School of Medicine, Hunan University of Chinese Medicine, Changsha, Hunan,410208, PR China
| | - Jian Lei
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan, 410082, PR China
| | - Ming-Ming Yin
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Hunan,410208, PR China
| | - Yi Chen
- Department of Physiology, School of Medicine, Hunan University of Chinese Medicine, Changsha, Hunan,410208, PR China
| |
Collapse
|
49
|
Gamero-Quijano A, Bhattacharya S, Cazade PA, Molina-Osorio AF, Beecher C, Djeghader A, Soulimane T, Dossot M, Thompson D, Herzog G, Scanlon MD. Modulating the pro-apoptotic activity of cytochrome c at a biomimetic electrified interface. SCIENCE ADVANCES 2021; 7:eabg4119. [PMID: 34739310 PMCID: PMC8570605 DOI: 10.1126/sciadv.abg4119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 09/17/2021] [Indexed: 06/13/2023]
Abstract
Programmed cell death via apoptosis is a natural defence against excessive cell division, crucial for fetal development to maintenance of homeostasis and elimination of precancerous and senescent cells. Here, we demonstrate an electrified liquid biointerface that replicates the molecular machinery of the inner mitochondrial membrane at the onset of apoptosis. By mimicking in vivo cytochrome c (Cyt c) interactions with cell membranes, our platform allows us to modulate the conformational plasticity of the protein by simply varying the electrochemical environment at an aqueous-organic interface. We observe interfacial electron transfer between an organic electron donor decamethylferrocene and O2, electrocatalyzed by Cyt c. This interfacial reaction requires partial Cyt c unfolding, mimicking Cyt c in vivo peroxidase activity. As proof of concept, we use our electrified liquid biointerface to identify drug molecules, such as bifonazole, that can potentially down-regulate Cyt c and protect against uncontrolled neuronal cell death in neurodegenerative disorders.
Collapse
Affiliation(s)
- Alonso Gamero-Quijano
- The Bernal Institute, University of Limerick (UL), Limerick V94 T9PX, Ireland
- Department of Chemical Sciences, School of Natural Sciences, University of Limerick (UL), Limerick V94 T9PX, Ireland
| | - Shayon Bhattacharya
- The Bernal Institute, University of Limerick (UL), Limerick V94 T9PX, Ireland
- Department of Physics, School of Natural Sciences, University of Limerick (UL), Limerick V94 T9PX, Ireland
| | - Pierre-André Cazade
- The Bernal Institute, University of Limerick (UL), Limerick V94 T9PX, Ireland
- Department of Physics, School of Natural Sciences, University of Limerick (UL), Limerick V94 T9PX, Ireland
| | - Andrés F. Molina-Osorio
- The Bernal Institute, University of Limerick (UL), Limerick V94 T9PX, Ireland
- Department of Chemical Sciences, School of Natural Sciences, University of Limerick (UL), Limerick V94 T9PX, Ireland
| | - Cillian Beecher
- The Bernal Institute, University of Limerick (UL), Limerick V94 T9PX, Ireland
- Department of Chemical Sciences, School of Natural Sciences, University of Limerick (UL), Limerick V94 T9PX, Ireland
| | - Ahmed Djeghader
- Department of Chemical Sciences, School of Natural Sciences, University of Limerick (UL), Limerick V94 T9PX, Ireland
| | - Tewfik Soulimane
- Department of Chemical Sciences, School of Natural Sciences, University of Limerick (UL), Limerick V94 T9PX, Ireland
| | - Manuel Dossot
- Laboratoire de Chimie Physique et Microbiologie pour les Matériaux et l’Environnement, Université de Lorraine, CNRS, LCPME, F-54000 Nancy, France
| | - Damien Thompson
- The Bernal Institute, University of Limerick (UL), Limerick V94 T9PX, Ireland
- Department of Physics, School of Natural Sciences, University of Limerick (UL), Limerick V94 T9PX, Ireland
| | - Grégoire Herzog
- Laboratoire de Chimie Physique et Microbiologie pour les Matériaux et l’Environnement, Université de Lorraine, CNRS, LCPME, F-54000 Nancy, France
| | - Micheál D. Scanlon
- The Bernal Institute, University of Limerick (UL), Limerick V94 T9PX, Ireland
- Department of Chemical Sciences, School of Natural Sciences, University of Limerick (UL), Limerick V94 T9PX, Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, Dublin, Ireland
| |
Collapse
|
50
|
Ishikawa C, Mori N. The antipsychotic drug pimozide is effective against human T-cell leukemia virus type 1-infected T cells. Eur J Pharmacol 2021; 908:174373. [PMID: 34303663 DOI: 10.1016/j.ejphar.2021.174373] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 06/28/2021] [Accepted: 07/21/2021] [Indexed: 11/22/2022]
Abstract
Patients with adult T-cell leukemia (ATL), caused by the human T-cell leukemia virus type 1 (HTLV-1), exhibit poor prognosis owing to drug resistance. Pimozide is a dopamine D2 receptor antagonist and antipsychotic shown to exhibit anticancer activity. Herein, we investigated whether pimozide exerts anti-ATL effects and explored the mechanisms underlying these effects. Pimozide inhibited cell growth and survival in HTLV-1-infected T cells but not in the uninfected T cells. The dopamine D2 receptor subfamily mRNA expression levels in HTLV-1-infected T cells were high. Pimozide induced G1 cell cycle arrest concomitant with the upregulation of p21/p27/p53, and suppression of cyclin D2/E, cyclin-dependent kinase 2/4/6 and c-Myc expression, and pRb phosphorylation. Pimozide also induced apoptosis by activating caspases, upregulating pro-apoptotic proteins and downregulating anti-apoptotic proteins. Additionally, it promoted reactive oxygen species (ROS) generation and increased the expression of the endoplasmic reticulum stress marker activating transcription factor 4 and the DNA damage-inducible protein GADD45α and the phosphorylation of the DNA damage marker H2AX. Furthermore, pimozide-induced cytotoxicity was partially inhibited by a ROS scavenger, and pan-caspase and necroptosis inhibitors, indicating the involvement of caspase-dependent and -independent lethal pathways. The activities of the nuclear factor-κB, Akt, STAT3/5 and AP-1 signaling pathways were inhibited via the dephosphorylation of IκBα, IκB kinase α/β, Akt and STAT3/5, in addition to reduced JunB and JunD expression in HTLV-1-infected T cells. Pimozide also exhibited potent anti-ATL activity in the xenograft mouse model. These findings demonstrated the efficacy of pimozide as a potential therapeutic agent for ATL.
Collapse
Affiliation(s)
- Chie Ishikawa
- Department of Microbiology and Oncology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Okinawa, 903-0215, Japan; Division of Health Sciences, Transdisciplinary Research Organization for Subtropics and Island Studies, University of the Ryukyus, Nishihara, Okinawa, 903-0213, Japan
| | - Naoki Mori
- Department of Microbiology and Oncology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Okinawa, 903-0215, Japan.
| |
Collapse
|