1
|
Antov GG, Gospodinova ZI, Novakovic M, Tesevic V, Krasteva NA, Pavlov DV, Valcheva-Kuzmanova SV. Molecular mechanisms of the anticancer action of fustin isolated from Cotinus coggygria Scop. in MDA-MB-231 triple-negative breast cancer cell line. Z NATURFORSCH C 2025; 80:233-250. [PMID: 39331583 DOI: 10.1515/znc-2024-0140] [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: 06/12/2024] [Accepted: 09/10/2024] [Indexed: 09/29/2024]
Abstract
The aim of the present work was to investigate some of the molecular mechanisms and targets of the anticancer action of the bioflavonoid fustin isolated from the heartwood of Cotinus coggygria Scop. in the triple-negative breast cancer cell line MDA-MB-231. For this purpose, we applied fluorescence microscopy analysis to evaluate apoptosis, necrosis, and mitochondrial integrity, wound healing assay to study fustin antimigratory potential and quantitative reverse transcription-polymerase chain reaction to analyze the expression of genes associated with cell cycle control, programmed cell death, metastasis, and epigenetic alterations. A complex network-based bioinformatic analysis was also employed for protein-protein network construction, hub genes identification, and functional enrichment. The results revealed a significant induction of early and late apoptotic and necrotic events, a slight alteration of the mitochondria-related fluorescence, and marked antimotility effect after fustin treatment. Of 34 analyzed genes, seven fustin targets were identified, of which CDKN1A, ATM, and MYC were significantly enriched in pathways such as cell cycle, intrinsic apoptotic signaling pathway in response to DNA damage and generic transcription pathway. Our findings outline some molecular mechanisms of the anticancer action of fustin pointing it out as a potential oncotherapeutic agent and provide directions for future in vivo research.
Collapse
Affiliation(s)
- Georgi G Antov
- Laboratory of Genome Dynamics and Stability, Institute of Plant Physiology and Genetics, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Zlatina I Gospodinova
- Laboratory of Genome Dynamics and Stability, Institute of Plant Physiology and Genetics, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Miroslav Novakovic
- Department of Chemistry, University of Belgrade - Institute of Chemistry, Technology and Metallurgy, National Institute of the Republic of Serbia, Belgrade, Serbia
| | - Vele Tesevic
- University of Belgrade - Faculty of Chemistry, Belgrade, Serbia
| | - Natalia A Krasteva
- Department of Electroinduced and Adhesive Properties, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Danail V Pavlov
- Department of Biochemistry, Molecular Medicine and Nutrigenomics with Laboratory of Nutrigenomics, Functional Foods and Nutraceuticals, Faculty of Pharmacy, Medical University "Prof. Dr. Paraskev Stoyanov", Varna, Bulgaria
| | - Stefka V Valcheva-Kuzmanova
- Department of Pharmacology and Clinical Pharmacology and Therapeutics, Faculty of Medicine, Medical University "Prof. Dr. Paraskev Stoyanov", Varna, Bulgaria
| |
Collapse
|
2
|
Mussulini BHM, Maruszczak KK, Draczkowski P, Borrero-Landazabal MA, Ayyamperumal S, Wnorowski A, Wasilewski M, Chacinska A. MIA40 suppresses cell death induced by apoptosis-inducing factor 1. EMBO Rep 2025; 26:1835-1862. [PMID: 40055465 PMCID: PMC11976965 DOI: 10.1038/s44319-025-00406-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 01/08/2025] [Accepted: 01/24/2025] [Indexed: 04/09/2025] Open
Abstract
Mitochondria harbor respiratory complexes that perform oxidative phosphorylation. Complex I is the first enzyme of the respiratory chain that oxidizes NADH. A dysfunction in complex I can result in higher cellular levels of NADH, which in turn strengthens the interaction between apoptosis-inducing factor 1 (AIFM1) and Mitochondrial intermembrane space import and assembly protein 40 (MIA40) in the mitochondrial intermembrane space. We investigated whether MIA40 modulates the activity of AIFM1 upon increased NADH/NAD+ balance. We found that in model cells characterized by an increase in NADH the AIFM1-MIA40 interaction is strengthened and these cells demonstrate resistance to AIFM1-induced cell death. Either silencing of MIA40, rescue of complex I, or depletion of NADH through the expression of yeast NADH-ubiquinone oxidoreductase-2 sensitized NDUFA13-KO cells to AIFM1-induced cell death. These findings indicate that the complex of MIA40 and AIFM1 suppresses AIFM1-induced cell death in a NADH-dependent manner. This study identifies an effector complex involved in regulating the programmed cell death that accommodates the metabolic changes in the cell and provides a molecular explanation for AIFM1-mediated chemoresistance of cancer cells.
Collapse
Affiliation(s)
- Ben Hur Marins Mussulini
- ReMedy International Research Agenda Unit, IMol Polish Academy of Sciences, Warsaw, Poland
- IMol Polish Academy of Sciences, Warsaw, Poland
| | | | - Piotr Draczkowski
- National Bioinformatics Infrastructure Sweden, SciLifeLab, Solna, Sweden
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances, Medical University of Lublin, Lublin, Poland
| | | | | | - Artur Wnorowski
- Department of Biopharmacy, Medical University of Lublin, Lublin, Poland
| | - Michal Wasilewski
- ReMedy International Research Agenda Unit, IMol Polish Academy of Sciences, Warsaw, Poland
- IMol Polish Academy of Sciences, Warsaw, Poland
| | - Agnieszka Chacinska
- ReMedy International Research Agenda Unit, IMol Polish Academy of Sciences, Warsaw, Poland.
- IMol Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
3
|
Zhang J, Yu Q, Zhu W, Sun X. Recent advances in the role of circRNA in cisplatin resistance in tumors. Cancer Gene Ther 2025:10.1038/s41417-025-00899-4. [PMID: 40148680 DOI: 10.1038/s41417-025-00899-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 03/08/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
Cancer remains a major threat to human health, with chemotherapy serving as one of the main treatment strategies to alleviate patient suffering. However, prolonged chemotherapy often leads to the development of drug resistance, complicating treatment outcomes. Cisplatin, a commonly utilized chemotherapeutic agent, demonstrates efficacy against a range of cancers but frequently encounters resistance, posing a significant challenge in tumor management and prognosis. Drug resistance not only facilitates tumor progression but also reduces survival rates, highlighting the urgent need for innovative strategies to overcome this issue. In recent years, non-coding RNAs, particularly circular RNAs (circRNAs), have gained attention in cancer therapy due to their stability and specificity. Moreover, an increasing number of studies have reported that circRNAs are involved in cisplatin resistance across various types of cancer. This paper primarily reviews the mechanisms and roles of circRNA in mediating cisplatin resistance over the past 3 years. These findings highlight circRNAs as promising therapeutic targets for overcoming cancer drug resistance.
Collapse
Affiliation(s)
- Jiawen Zhang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Qiwen Yu
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Weijin Zhu
- Department of Clinical Laboratory Medicine, Hospital of Traditional Chinese Medicine, Changzhou, Jiangsu, China
| | - Xiaochun Sun
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China.
| |
Collapse
|
4
|
Dong TR, Li YJ, Jin SY, Yang FL, Xiong RX, Dai YQ, Song XZ, Guan CP. Progress on mitochondria and hair follicle development in androgenetic alopecia: relationships and therapeutic perspectives. Stem Cell Res Ther 2025; 16:44. [PMID: 39901201 PMCID: PMC11792644 DOI: 10.1186/s13287-025-04182-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: 11/13/2024] [Accepted: 01/23/2025] [Indexed: 02/05/2025] Open
Abstract
Hair loss has long been a significant concern for many individuals. Recent studies have indicated that mitochondria play a more crucial role in hair loss than previously recognized. This review summarizes the connection between mitochondrial dysfunction and hair follicle development, outlines the links between diseases related to mitochondrial disorders and hair issues, and highlights the influence of mitochondrial dysfunction on androgenetic alopecia. We discuss the cellular and signaling mechanisms associated with hair loss and examine how mitochondrial dysfunction, such as insufficient energy supply, signaling irregularities, protein/gene abnormalities, and programmed cell death, can hinder the normal proliferation, differentiation, and growth of hair follicle cells. Furthermore, we discuss current treatment approaches and potential innovative therapies, including mitochondrion-targeting drugs and advanced techniques that directly target hair follicle cells, providing fresh insights into the crucial role of mitochondria in maintaining hair follicle health and managing hair disorders. Furthermore, this review explores future therapeutic strategies and proposes that mitochondrial research could lead to groundbreaking treatments for hair loss, thus providing optimism and new avenues for the treatment of individuals experiencing hair loss. This review not only underscores the central importance of mitochondria in hair health but also emphasizes the importance of advancing research and treatment in this field.
Collapse
Affiliation(s)
- Ting-Ru Dong
- Department of Dermatology, Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310009, China
| | - Yu-Jie Li
- Department of Dermatology, Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310009, China
| | - Shi-Yu Jin
- Department of Dermatology, Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310009, China
| | - Feng-Lan Yang
- Department of Dermatology, Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310009, China
| | - Ren-Xue Xiong
- Department of Dermatology, Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310009, China
- Department of Dermatology, Hangzhou Third People's Hospital, No 38 Xihu Rd, Hangzhou, 310009, China
| | - Ye-Qin Dai
- Department of Dermatology, Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310009, China
- Department of Dermatology, Hangzhou Third People's Hospital, No 38 Xihu Rd, Hangzhou, 310009, China
| | - Xiu-Zu Song
- Department of Dermatology, Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310009, China
- Department of Dermatology, Hangzhou Third People's Hospital, No 38 Xihu Rd, Hangzhou, 310009, China
| | - Cui-Ping Guan
- Department of Dermatology, Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310009, China.
- Department of Dermatology, Hangzhou Third People's Hospital, No 38 Xihu Rd, Hangzhou, 310009, China.
| |
Collapse
|
5
|
Sun B, Wang Y, Chen H, Huang Q, An C, Zhan Q, Wang X, Chen T. Disulfiram/copper induces BAK-mediated caspase-independent apoptosis in MCF-7 cells. Int J Biochem Cell Biol 2025; 179:106731. [PMID: 39719221 DOI: 10.1016/j.biocel.2024.106731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/08/2024] [Accepted: 12/19/2024] [Indexed: 12/26/2024]
Abstract
Disulfiram (DSF) and copper (Cu2 +) in combination exhibit powerful anti-cancer effect on a variety of cancer cell lines. Here, we found that DSF/Cu2+ facilitated the accumulation of intracellular reactive oxygen species (ROS), and induced ROS-dependent apoptosis accompanied by chromatin condensation and phosphatidylserine externalization in MCF-7 cells. DSF/Cu2+ caused caspase-independent apoptosis by promoting the AIF translocation from mitochondria to nucleus. Most importantly, the cytotoxicity of DSF/Cu2+ was markedly inhibited by knocking out AIF, suggesting the indispensability of AIF in DSF/Cu2+-induced apoptosis. The pro-apoptotic protein BAK instead of BAX was upregulated and activated upon DSF/Cu2+ treatment, and the BAK knockout cells exhibited high resistance to DSF/Cu2+, indicating the importance of BAK in DSF/Cu2+-induced apoptosis. Additionally, both co-immunoprecipitation and live-cell quantitative fluorescence resonance energy transfer (FRET) analysis revealed that DSF/Cu2+ unlocked the binding of MCL-1 to BAK, which resulted in subsequent BAK homo-oligomerization. Overall, our data demonstrate for the first time that DSF/Cu2+ unlocks the binding of MCL-1 to BAK, thus leading BAK oligomerization and subsequent AIF nucleus translocation to mediate caspase-independent apoptosis in MCF-7 cells.
Collapse
Affiliation(s)
- Beini Sun
- MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510631, China
| | - Yu Wang
- MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510631, China
| | - Hongce Chen
- MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510631, China
| | - Qialing Huang
- MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510631, China
| | - Chunchun An
- MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510631, China
| | - Qiuqiang Zhan
- Centre for Optical and Electromagnetic Research, Guangdong Engineering Research Centre of Optoelectronic Intelligent Information Perception, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, China
| | - Xiaoping Wang
- Department of Pain Management, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China.
| | - Tongsheng Chen
- MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510631, China.
| |
Collapse
|
6
|
Chen L, Tang J, Tan H. Penehyclidine hydrochloride activates PARK2 and modulates ubiquitination of AIFM1 to rescue renal tubular injury in diabetic kidney disease. J Pharmacol Sci 2025; 157:45-56. [PMID: 39828393 DOI: 10.1016/j.jphs.2024.12.001] [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: 07/17/2024] [Revised: 11/03/2024] [Accepted: 12/04/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Renal tubular injury (RTI) is one of the key characteristics of diabetic nephropathy (DN). Penehyclidine hydrochloride (PHC) was an anticholinergic drug with renoprotective effects, but its specific mechanism in the treatment of DN was still unclear. METHODS We treated different diabetic mouse models and high glucose-induced RTI models by PHC. Histological analyses were performed using flow cytometry and staining, and ELISA evaluated the ROS, apoptosis, and related markers under different treatments. The molecular interactions were analyzed by ChIP, dual-luciferase reporter, and CoIP. RESULTS PHC alleviated RTI by activating mitophagy and inhibiting apoptosis, and the protective effect could be rescued by PARK2 knockdown. Nrf2 bound to the promoter region of PARK2 and promoted its expression. PHC reduced the level of apoptosis by reducing the degree of nuclear translocation of AIFM1, which was rescued by PARK2 knockdown. PARK2 knockdown reduced the non-degradative ubiquitination of AIFM1, thus promoting its nuclear translocation and ultimately facilitating renal tubular cells (RTCs) apoptosis. The over-expression of AIFM1 rescued the RTCs apoptosis antagonized by PHC. CONCLUSIONS PHC activated Nrf2 to up-regulate PARK2 transcription to induce mitophagy and inhibit apoptosis mediated by nuclear translocation of AIFM1 through promoting non-degradative ubiquitination of AIFM1, ultimately rescuing RTI in DN.
Collapse
Affiliation(s)
- Li Chen
- Brain Hospital of Hunan Province (The Second People's Hospital of Hunan Province), Department of Nephrology, 410007, Changsha, Hunan Province, China
| | - Jing Tang
- Brain Hospital of Hunan Province (The Second People's Hospital of Hunan Province), Department of Nephrology, 410007, Changsha, Hunan Province, China
| | - HongBao Tan
- The Fourth Hospital of Changsha, Department of Anesthesiology, 410006, Changsha, Hunan Province, China.
| |
Collapse
|
7
|
Tábara LC, Segawa M, Prudent J. Molecular mechanisms of mitochondrial dynamics. Nat Rev Mol Cell Biol 2025; 26:123-146. [PMID: 39420231 DOI: 10.1038/s41580-024-00785-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2024] [Indexed: 10/19/2024]
Abstract
Mitochondria not only synthesize energy required for cellular functions but are also involved in numerous cellular pathways including apoptosis, calcium homoeostasis, inflammation and immunity. Mitochondria are dynamic organelles that undergo cycles of fission and fusion, and these transitions between fragmented and hyperfused networks ensure mitochondrial function, enabling adaptations to metabolic changes or cellular stress. Defects in mitochondrial morphology have been associated with numerous diseases, highlighting the importance of elucidating the molecular mechanisms regulating mitochondrial morphology. Here, we discuss recent structural insights into the assembly and mechanism of action of the core mitochondrial dynamics proteins, such as the dynamin-related protein 1 (DRP1) that controls division, and the mitofusins (MFN1 and MFN2) and optic atrophy 1 (OPA1) driving membrane fusion. Furthermore, we provide an updated view of the complex interplay between different proteins, lipids and organelles during the processes of mitochondrial membrane fusion and fission. Overall, we aim to present a valuable framework reflecting current perspectives on how mitochondrial membrane remodelling is regulated.
Collapse
Affiliation(s)
- Luis-Carlos Tábara
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Mayuko Segawa
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Julien Prudent
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK.
| |
Collapse
|
8
|
Khaliulin I, Hamoudi W, Amal H. The multifaceted role of mitochondria in autism spectrum disorder. Mol Psychiatry 2025; 30:629-650. [PMID: 39223276 PMCID: PMC11753362 DOI: 10.1038/s41380-024-02725-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] [Received: 03/10/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Normal brain functioning relies on high aerobic energy production provided by mitochondria. Failure to supply a sufficient amount of energy, seen in different brain disorders, including autism spectrum disorder (ASD), may have a significant negative impact on brain development and support of different brain functions. Mitochondrial dysfunction, manifested in the abnormal activities of the electron transport chain and impaired energy metabolism, greatly contributes to ASD. The aberrant functioning of this organelle is of such high importance that ASD has been proposed as a mitochondrial disease. It should be noted that aerobic energy production is not the only function of the mitochondria. In particular, these organelles are involved in the regulation of Ca2+ homeostasis, different mechanisms of programmed cell death, autophagy, and reactive oxygen and nitrogen species (ROS and RNS) production. Several syndromes originated from mitochondria-related mutations display ASD phenotype. Abnormalities in Ca2+ handling and ATP production in the brain mitochondria affect synaptic transmission, plasticity, and synaptic development, contributing to ASD. ROS and Ca2+ regulate the activity of the mitochondrial permeability transition pore (mPTP). The prolonged opening of this pore affects the redox state of the mitochondria, impairs oxidative phosphorylation, and activates apoptosis, ultimately leading to cell death. A dysregulation between the enhanced mitochondria-related processes of apoptosis and the inhibited autophagy leads to the accumulation of toxic products in the brains of individuals with ASD. Although many mitochondria-related mechanisms still have to be investigated, and whether they are the cause or consequence of this disorder is still unknown, the accumulating data show that the breakdown of any of the mitochondrial functions may contribute to abnormal brain development leading to ASD. In this review, we discuss the multifaceted role of mitochondria in ASD from the various aspects of neuroscience.
Collapse
Affiliation(s)
- Igor Khaliulin
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wajeha Hamoudi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
9
|
Liang H, Zhang Y, Sun W, Xiao X, Zhao X, Tan B, Zhang J, Song X, He Z, Ye L. Identification of a Novel Alkaloid Zj6-11 as a Potent Inhibitor of Influenza Virus Infection via Repression of Virus-Induced Mitochondria-Dependent Apoptosis. J Med Virol 2025; 97:e70230. [PMID: 39921608 DOI: 10.1002/jmv.70230] [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: 09/12/2024] [Revised: 01/23/2025] [Accepted: 01/29/2025] [Indexed: 02/10/2025]
Abstract
Influenza A virus (IAV) remains a major global public health threat, especially with the emergence of antiviral resistance, highlighting the urgent need for novel therapeutics. Alkaloids are known for their antiviral properties, and chemical synthesis has become a key strategy in developing new alkaloid compounds. In this study, we synthesized a series of novel alkaloids using the Ugi reaction and assessed their antiviral potential and mechanisms. Through screening and validation, Zj6-11 was identified as a promising compound that effectively inhibits IAV infection in vitro. Molecular docking and binding affinity assays showed that Zj6-11 binds with high affinity to IAV nucleoprotein (NP) and inhibits its interaction with nucleic acids. Further, in vitro nuclear translocation assays confirmed that Zj6-11 suppresses NP nuclear import. Mechanistically, Zj6-11 significantly inhibits IAV-induced apoptosis and mitigates mitochondrial membrane potential dysfunction. Zj6-11 also inhibits cytochrome c release, reduces the expression of cleaved Caspase-9 and Caspase-3, and suppresses IAV-induced apoptosis-inducing factor (Aif) expression, suppressing IAV-induced mitochondrial apoptosis. More importantly, Zj6-11 plays a crucial role in protecting mice from IAV infection and reducing IAV pathogenicity. Our study provides mechanistic insights into Zj6-11's control of IAV infection in vitro and in vivo, offering new perspectives for antiviral therapy development.
Collapse
Affiliation(s)
- Hanbai Liang
- Department of Immunology, International Cancer Center, College of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Ying Zhang
- Department of Immunology, International Cancer Center, College of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Wenhao Sun
- Department of Immunology, International Cancer Center, College of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Xiaomei Xiao
- Department of Immunology, International Cancer Center, College of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Xiwen Zhao
- Department of Immunology, International Cancer Center, College of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Bin Tan
- Shenzhen Grubbs Institute and Department of Chemistry, Southern University of Science and Technology, Shenzhen, China
| | - Jian Zhang
- Shenzhen Grubbs Institute and Department of Chemistry, Southern University of Science and Technology, Shenzhen, China
| | - Xun Song
- College of Pharmacy, Shenzhen Technology University, Shenzhen, China
| | - Zhengdan He
- College of Pharmacy, Shenzhen Technology University, Shenzhen, China
| | - Liang Ye
- Department of Immunology, International Cancer Center, College of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| |
Collapse
|
10
|
Xu W, Yan J, Travis ZD, Lenahan C, Gao L, Wu H, Zheng J, Zhang J, Shao A, Yu J. Apelin/APJ system: a novel promising target for anti-oxidative stress in stroke. Front Pharmacol 2025; 15:1352927. [PMID: 39881878 PMCID: PMC11775478 DOI: 10.3389/fphar.2024.1352927] [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: 12/09/2023] [Accepted: 12/10/2024] [Indexed: 01/31/2025] Open
Abstract
The apelin/APJ system has garnered increasing attention in recent years. In this review, we comprehensively discuss the physiological and pathological mechanisms of the apelin/APJ system in stroke. The apelin/APJ system is widely expressed in the central nervous system (CNS). However, the distribution of the apelin/APJ system varies across different regions and subcellular organelles of the brain. Additionally, the neuroprotective effects of the apelin/APJ system have been reported to inhibit oxidative and nitrative stresses via various signaling pathways. Despite this, the clinical application of the apelin/APJ system remains distant, as apelin has numerous active forms and signaling pathways. The development of a range of drugs targeting the apelin/APJ system holds promise for treating stroke.
Collapse
Affiliation(s)
- Weilin Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Jun Yan
- Department of Neurosurgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Zachary D. Travis
- Department of Medical Science Education, College of Health Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Cameron Lenahan
- Burrell College of Osteopathic Medicine, New Mexico State University, Las Cruces, NM, United States
| | - Liansheng Gao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Haijian Wu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Jingwei Zheng
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Anwen Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Jun Yu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| |
Collapse
|
11
|
Stawikowska A, Dziembowska M, Kuzniewska B. Metabolic Phenotyping of Synaptic Mitochondria Using MitoPlates™ and Synaptoneurosomes. Methods Mol Biol 2025; 2878:67-74. [PMID: 39546257 DOI: 10.1007/978-1-0716-4264-1_4] [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: 11/17/2024]
Abstract
Mitochondrial functional assays using MitoPlates™ S-1 allow us to characterize mitochondria in terms of substrate metabolism. MitoPlates™ are 96-well microplates pre-coated with a diverse set of substrates. The electron flow from NADH and FADH2 producing mitochondrial substrates is measured based on the reduction of redox dye, that acts as a terminal electron acceptor. Here, we describe the application of MitoPlates™ to characterize the metabolism of synaptic mitochondria enclosed in isolated pre- and postsynaptic terminals (synaptoneurosomes).
Collapse
Affiliation(s)
- Aleksandra Stawikowska
- Department of Animal Physiology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Magdalena Dziembowska
- Department of Animal Physiology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Bozena Kuzniewska
- Department of Animal Physiology, Faculty of Biology, University of Warsaw, Warsaw, Poland.
| |
Collapse
|
12
|
Zhang B, Hou S, Tang J. Riboflavin Deficiency and Apoptosis: A Review. J Nutr 2025; 155:27-36. [PMID: 39510506 DOI: 10.1016/j.tjnut.2024.10.053] [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: 09/06/2024] [Revised: 10/25/2024] [Accepted: 10/31/2024] [Indexed: 11/15/2024] Open
Abstract
Riboflavin, commonly known as vitamin B2, is an essential micronutrient critical for the function of flavoproteins, which utilize flavin mononucleotide and flavin adenine dinucleotide as cofactors in energy metabolism, lipid metabolism, redox regulation, and protein folding. Nutritional riboflavin deficiency (RD) has previously been observed in humans and animals, leading to adverse outcomes such as growth retardation, increased mortality, and liver damage, which may be attributed to apoptosis. Although such deficiencies are now uncommon because of improved living standards, certain high-risk groups (e.g. those with chronic diseases, the elderly, and pregnant) have increased riboflavin demands, making them vulnerable to physiological RD associated with apoptosis. Understanding the pathways through which RD induces apoptosis, including mitochondrial dysfunction, endoplasmic reticulum stress, and reactive oxygen species, is essential for grasping its broader health impacts. Additionally, this deficiency disrupts fatty acid metabolism, potentially resulting in lipotoxic apoptosis. Despite its significance, RD-induced apoptosis remains underexplored in the literature. Therefore, this review will discuss the roles of redox imbalance, mitochondrial dysfunction, endoplasmic reticulum stress, and lipotoxicity in apoptosis regulation because of RD, aiming to highlight its importance for improving riboflavin nutrition and overall health.
Collapse
Affiliation(s)
- Bo Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China; Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China.
| | - Shuisheng Hou
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China.
| | - Jing Tang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China.
| |
Collapse
|
13
|
Kritskaya KA, Stelmashchuk OA, Abramov AY. Point of No Return-What Is the Threshold of Mitochondria With Permeability Transition in Cells to Trigger Cell Death. J Cell Physiol 2025; 240:e31521. [PMID: 39760157 DOI: 10.1002/jcp.31521] [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: 09/25/2024] [Revised: 11/21/2024] [Accepted: 12/21/2024] [Indexed: 01/07/2025]
Abstract
Programmed cell death (apoptosis) is essential part of the process of tissue regeneration that also plays role in the mechanism of pathology. The phenomenon of fast and transient permeability of mitochondrial membranes by various triggers, known as permeability transition pore (mPTP) leads to the release of proapoptotic proteins and acts as an initial step in initiation of apoptosis. However, a role for mPTP was also suggested for physiology and it is unclear if there is a threshold in number of mitochondria with mPTP which induces cell death and how this mechanism is regulated in different tissues. Using simultaneous measurements of mitochondrial membrane potential and a fluorescent marker for caspase-3 activation we studied the number of mitochondria with calcium-induced mPTP opening necessary for induction of apoptosis in rat primary cortical neurons, astrocytes, fibroblasts, and cancer (BT-474) cells. The induction of apoptosis was correlated with 80%-90% mitochondrial signal loss in neural cells but only 35% in fibroblasts, and in BT-474 cancer cells over 90% of mitochondria opens mPTP before apoptosis becomes obvious. The number of mitochondria with mPTP which induce cell death did not correlate with total expression levels of proapoptotic proteins but was consistent with the Bax/Bcl-2 ratio in these cells. Calcium-induced mPTP opening increased levels of necrosis which was higher in fibroblasts compared to neurons, astrocytes and BT-474 cells. Thus, different tissues require specific numbers of mitochondria with PTP opening to induce apoptosis and it correlates to the proapoptotic/antiapoptotic proteins expression ratio in them.
Collapse
Affiliation(s)
- Kristina A Kritskaya
- Institute of Cell Biophysics of the Russian Academy of Sciences, Puschino, Russia
| | | | - Andrey Y Abramov
- Orel State University named after I.S. Turgenev, Orel, Russia
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London, UK
| |
Collapse
|
14
|
de Lemos AC, Teixeira J, Cunha-Oliveira T. Characterization of the Mitochondria Function and Metabolism in Skin Fibroblasts Using the Biolog MitoPlate S-1. Methods Mol Biol 2025; 2878:75-98. [PMID: 39546258 DOI: 10.1007/978-1-0716-4264-1_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] [Indexed: 11/17/2024]
Abstract
S-1 MitoPlates™ from Biolog enable the characterization of mitochondria's function in live cells by measuring the rates of electron flow into and through the electron transport chain from different NADH or FADH2 producing metabolic substrates. This technology uses 96-well microplates pre-coated with triplicate repeats of a set of 31 substrates. Those 31 metabolic substrates have different routes of entry into the mitochondria, use different transporters, and are also oxidated by different dehydrogenases, producing reducing equivalents in the form of NADH or FADH2. The electrons produced upon oxidation of NADH or FADH2 at complex I or II, respectively, then travel to cytochrome c, where a tetrazolium redox dye (MC) can act as terminal acceptor, turning purple and absorbing at 590 nm. This mechanism allows the evaluation of cellular substrate preference by following the kinetics of MC reduction in the presence of selected substrates.In this chapter, we describe the step-by-step protocol to prepare an experiment using MitoPlate S-1 array and the OmniLog instrument to assess the metabolism of human dermal fibroblasts. We also give detailed information on how to analyze the raw data generated by the Biolog Data Analysis software to extract meaningful information and produce useful data visualizations, using reproducible methods based on a single structured dataset.
Collapse
Affiliation(s)
- Alexandra C de Lemos
- CNC-Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - José Teixeira
- CNC-Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.
- CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.
| | - Teresa Cunha-Oliveira
- CNC-Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
15
|
Zhong F, Xiong L, Wu J, Chen Y, Song J, Yu W, Lü Y. The KEAP1/PGAM5/AIFM1-Mediated oxeiptosis pathway in Alzheimer's disease. Brain Res 2024; 1845:149173. [PMID: 39168265 DOI: 10.1016/j.brainres.2024.149173] [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: 05/14/2024] [Revised: 08/09/2024] [Accepted: 08/17/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND Alzheimer's Disease (AD) is a neurodegenerative disease with mitochondrial dysfunction and oxidative stress. Oxeiptosis is a cell death pathway sensitive to reactive oxygen species (ROS). This study investigates the role of oxeiptosis pathway and mitochondrial damage in AD. METHODS An AD model was developed in C57BL/6 mice by injecting Aβ1-42 oligomers into the brain. Cognitive function was tested using the Morris water maze. Exposure of HT22 mouse hippocampal neurons to H2O2 induces oxidative stress. Protein levels of KEAP1, PGAM5 and AIFM1 were analyzed by western blot, and mitochondrial damage was observed with electron microscopy. Cell survival rates were using the CCK8 assay and flow cytometry after knocking down KEAP1, PGAM5 and AIFM1. RESULTS The protein concentrations of KEAP1, PGAM5 and AIFM1 were found to be elevated in the hippocampal tissues of AD mice compared to control group, accompanied by mitochondrial damage in the hippocampal neurons of the AD group. Similarly, in the HT22 oxidative stress model, there was an increase in the protein levels of KEAP1, PGAM5 and AIFM1, along with observed mitochondrial damage. Following individual and combined knockdown of KEAP1, PGAM5 and AIFM1, cell survival rates under oxidative stress conditions were higher compared to H2O2 group, with no significant difference in cell survival rates among the knockdown groups. CONCLUSION This research underscores the critical role of the KEAP1/PGAM5/AIFM1-mediated oxeiptosis pathway in neuronal cell death, offering insights into potential therapeutic targets for mitigating neurodegeneration in AD.
Collapse
Affiliation(s)
- Fuxin Zhong
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Lei Xiong
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Jiani Wu
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Yingxi Chen
- Institute of Neuroscience, Department of Human Anatomy, Chongqing Medical University, Chongqing 400016, PR China
| | - Jiaqi Song
- Institute of Neuroscience, Department of Human Anatomy, Chongqing Medical University, Chongqing 400016, PR China
| | - Weihua Yu
- Institute of Neuroscience, Department of Human Anatomy, Chongqing Medical University, Chongqing 400016, PR China.
| | - Yang Lü
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
16
|
Gao L, Gao B, Ge W, Li S, Wang F. Stimulated Emission Depletion Imaging Reveals Mitochondrial Phenotypic Heterogeneity under Apoptosis Stimuli across Living Glioma Models. NANO LETTERS 2024; 24:15904-15911. [PMID: 39587402 DOI: 10.1021/acs.nanolett.4c04986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
The mitochondrial phenotypes contribute to the understanding of disease mechanisms and treatments, which are typically characterized through the omics methods. However, the high dynamics and phenotypic heterogeneity of mitochondria require high-resolution characterization within individual living cells. Therefore, we introduce a fluorescence analysis method, based on two-color and fluorescence lifetime stimulated emission depletion (STED) super-resolution imaging, to explore mitochondrial phenotypic heterogeneity in human (U87) and mouse (GL261) glioma models. Furthermore, we used rotenone and etoposide to simulate the effects of antitumor drugs, inducing apoptosis through mitochondrial dysfunction, respectively. The two-color labeling introduces intracellular parameters to qualitatively visualize changes in mitochondrial morphology, while fluorescence lifetime reflects the status of mitochondria and their microenvironment from the perspective of probe characteristics. This method reveals mitochondria phenotypic heterogeneity induced by the apoptotic stimuli in human and mouse glioma models from a morphological perspective.
Collapse
Affiliation(s)
- Lu Gao
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Beibei Gao
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Wei Ge
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shuxian Li
- College of Chemistry and Chemical Engineering, Xinjiang Normal University, Urumqi 830054, China
| | - Fu Wang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
17
|
Schultz A, Albertos-Arranz H, Sáez XS, Morgan J, Darland DC, Gonzalez-Duarte A, Kaufmann H, Mendoza-Santiesteban CE, Cuenca N, Lefcort F. Neuronal and glial cell alterations involved in the retinal degeneration of the familial dysautonomia optic neuropathy. Glia 2024; 72:2268-2294. [PMID: 39228100 DOI: 10.1002/glia.24612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 07/27/2024] [Accepted: 08/19/2024] [Indexed: 09/05/2024]
Abstract
Familial dysautonomia (FD) is a rare genetic neurodevelopmental and neurodegenerative disorder. In addition to the autonomic and peripheral sensory neuropathies that challenge patient survival, one of the most debilitating symptoms affecting patients' quality of life is progressive blindness resulting from the steady loss of retinal ganglion cells (RGCs). Within the FD community, there is a concerted effort to develop treatments to prevent the loss of RGCs. However, the mechanisms underlying the death of RGCs are not well understood. To study the mechanisms underlying RGC death, Pax6-cre;Elp1loxp/loxp male and female mice and postmortem retinal tissue from an FD patient were used to explore the neuronal and non-neuronal cellular pathology associated with the FD optic neuropathy. Neurons, astrocytes, microglia, Müller glia, and endothelial cells were investigated using a combination of histological analyses. We identified a novel disruption of cellular homeostasis and gliosis in the FD retina. Beginning shortly after birth and progressing with age, the FD retina is marked by astrogliosis and perturbations in microglia, which coincide with vascular remodeling. These changes begin before the onset of RGC death, suggesting alterations in the retinal neurovascular unit may contribute to and exacerbate RGC death. We reveal for the first time that the FD retina pathology includes reactive gliosis, increased microglial recruitment to the ganglion cell layer (GCL), disruptions in the deep and superficial vascular plexuses, and alterations in signaling pathways. These studies implicate the neurovascular unit as a disease-modifying target for therapeutic interventions in FD.
Collapse
Affiliation(s)
- Anastasia Schultz
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, USA
| | - Henar Albertos-Arranz
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Xavier Sánchez Sáez
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Jamie Morgan
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, USA
| | - Diane C Darland
- Department of Biology, University of North Dakota, Grand Forks, North Dakota, USA
| | | | - Horacio Kaufmann
- Department of Neurology, NYU Langone Health, New York, New York, USA
| | - Carlos E Mendoza-Santiesteban
- Department of Neurology, NYU Langone Health, New York, New York, USA
- Bascom Palmer Eye Institute, University of Miami, Miami, Florida, USA
| | - Nicolás Cuenca
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Frances Lefcort
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, USA
| |
Collapse
|
18
|
Zhang N, Han G, Li C, Huang L, Liu Q, Lin M, Xu B, Xu J. Cnaphalocrocis medinalis granulovirus regulates apoptosis by targeting AIF1 and ASPP1 through tca-miR-3885-5p and tca-miR-3897-3p to promote infection. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 206:106196. [PMID: 39672625 DOI: 10.1016/j.pestbp.2024.106196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/10/2024] [Accepted: 10/26/2024] [Indexed: 12/15/2024]
Abstract
Cnaphalocrocis medinalis granulovirus (CnmeGV) is a potential biocontrol agent for C. medinalis which is a major rice pest. However, its insecticidal efficacy is slow due to cell apoptosis. This study investigated the role of miRNAs in CnmeGV-mediated apoptosis. Small RNA sequencing and qRT-PCR identified miRNAs tca-miR-3885-5p and tca-miR-3897-3p, which initially increased and then decreased post-infection, but remained higher than controls. This trend was opposite to the changes in midgut apoptosis levels detected using terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and DNA ladder assays. Compared to the group treated with CnmeGV alone, agomirs increased the CnmeGV-induced larval mortality, reduced midgut apoptosis, whereas antagomirs had the opposite effects. We found that the upregulation of CnmeGV replication induced by agomirs initially increased and then decreased, while the apoptosis inducer PAC-1 compensated for the weakening trend of CnmeGV replication upregulation induced by agomirs in the later stages of infection. Results indicated the virus hijacks these miRNAs to inhibit early apoptosis, later requiring apoptosis for systemic infection from the midgut. Agomirs treatment and dual-luciferase assays showed these miRNAs functioned via apoptosis-inducing factor 1 (AIF1) and apoptosis-stimulating protein of p53 1 (ASPP1) mRNA expression. This study highlights the role of these miRNAs in infection and provides insights for developing viral insecticide enhancers.
Collapse
Affiliation(s)
- Nan Zhang
- Jiangsu Lixiahe District Institute of Agricultural Sciences, Yangzhou, China; National Agricultural Experimental Station for Agricultural Microbiology in Yangzhou, Yangzhou, China
| | - Guangjie Han
- Jiangsu Lixiahe District Institute of Agricultural Sciences, Yangzhou, China; National Agricultural Experimental Station for Agricultural Microbiology in Yangzhou, Yangzhou, China
| | - Chuanming Li
- Jiangsu Lixiahe District Institute of Agricultural Sciences, Yangzhou, China; National Agricultural Experimental Station for Agricultural Microbiology in Yangzhou, Yangzhou, China
| | - Lixin Huang
- Jiangsu Lixiahe District Institute of Agricultural Sciences, Yangzhou, China; National Agricultural Experimental Station for Agricultural Microbiology in Yangzhou, Yangzhou, China
| | - Qin Liu
- Jiangsu Lixiahe District Institute of Agricultural Sciences, Yangzhou, China; National Agricultural Experimental Station for Agricultural Microbiology in Yangzhou, Yangzhou, China
| | - Manman Lin
- Jiangsu Lixiahe District Institute of Agricultural Sciences, Yangzhou, China; National Agricultural Experimental Station for Agricultural Microbiology in Yangzhou, Yangzhou, China
| | - Bin Xu
- Jiangsu Lixiahe District Institute of Agricultural Sciences, Yangzhou, China
| | - Jian Xu
- Jiangsu Lixiahe District Institute of Agricultural Sciences, Yangzhou, China; National Agricultural Experimental Station for Agricultural Microbiology in Yangzhou, Yangzhou, China.
| |
Collapse
|
19
|
Zheng Y, Sun J, Luo Z, Li Y, Huang Y. Emerging mechanisms of lipid peroxidation in regulated cell death and its physiological implications. Cell Death Dis 2024; 15:859. [PMID: 39587094 PMCID: PMC11589755 DOI: 10.1038/s41419-024-07244-x] [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: 08/09/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 11/27/2024]
Abstract
Regulated cell death (RCD) refers to the form of cell death that can be regulated by various biomacromolecules. Each cell death modalities have their distinct morphological changes and molecular mechanisms. However, intense evidences suggest that lipid peroxidation can be the common feature that initiates and propagates the cell death. Excessive lipid peroxidation alters the property of membrane and further damage the proteins and nucleic acids, which is implicated in various human pathologies. Here, we firstly review the classical chain process of lipid peroxidation, and further clarify the current understanding of the myriad roles and molecular mechanisms of lipid peroxidation in various RCD types. We also discuss how lipid peroxidation involves in diseases and how such intimate association between lipid peroxidation-driven cell death and diseases can be leveraged to develop rational therapeutic strategies.
Collapse
Affiliation(s)
- Yongxin Zheng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Respiratory Health Guangzhou, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou, China
| | - Junlu Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Respiratory Health Guangzhou, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Zhiting Luo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Respiratory Health Guangzhou, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Yimin Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou Institute of Respiratory Health Guangzhou, Guangzhou, China.
- State Key Laboratory of Respiratory Diseases, Guangzhou, China.
- Guangzhou National Laboratory, Guangzhou, China.
| | - Yongbo Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou Institute of Respiratory Health Guangzhou, Guangzhou, China.
- State Key Laboratory of Respiratory Diseases, Guangzhou, China.
| |
Collapse
|
20
|
Li XM, Chen X, Zhao DG. Overexpression of the Eucommia ulmoides chitinase EuCHIT73.88 gene improves tobacco disease resistance. Gene 2024; 927:148619. [PMID: 38821325 DOI: 10.1016/j.gene.2024.148619] [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: 02/06/2024] [Revised: 05/17/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
Black shank disease is the main disease affecting tobacco crops worldwide, and the main impacted by the disease are the stem base and root. At present, transgenic technology is an effective method to improve plant disease resistance through transgenic technology. In this study, the EuCHIT73.88 gene was cloned from Eucommia ulmoides Oliver (E. ulmoides) by using RT-PCR. The full length of the gene was 897 bp, encoding 298 amino acid residues. An overexpression vector of from the EuCHIT73.88 gene driven by the 35S promoter was constructed and transferred into tobacco plants via transgenic technology. After inoculation with the black shank pathogen, the number of visible lesions on the stems and leaves of the transgenic tobacco variety EuCHIT73.88 was significantly shorter than that on the stems and leaves of the of wild type (WT) and empty vector (EV) plants, and the lesion area was significantly smaller than on the stems and leaves of the WT and EV plants. With increasing inoculation time, introduction of the WT and EV vectors was obviously lethal, whereas transgenic tobacco only exhibited wilted characteristics, and the stems were black, which indicated that the EuCHIT73.88 gene could improve the resistance of tobacco to black shank disease. Furthermore, the activity of protective enzymes and the gene expression of resistance-related proteins were measured. The results showed that compared with those of the WT and EV plants, the CAT and POD activities of the TP tobacco plants were greater, peaking at 72 h at concentrations of 446.87 U/g and 4562.24 U/g, which were 1.63 and 1.61 times greater than those of the WT and EV plants, respectively. This indicated that CAT and POD may be involved in the process of disease resistance of in the transgenic plants. The MDA content of the transgenic tobacco plants was significantly lower than that of the WT and EV plants with increasing EuCHIT73.88 expression, thus indicating that the overexpression of the transgenic EuCHIT73.88 gene could alleviate the levels of lipid peroxidation and reduce the damage to plant cell membranes. The expression of disease-related protein genes (PR2, PR5, PR1a, PDF1.2 and MLP423) was significantly greater in the EuCHIT73.88 ransgenic tobacco than in the WT and EV-transgenic tobacco. and these findings consistently showed that EuCHIT73.88 could improve the resistance to black shank.
Collapse
Affiliation(s)
- Xiao-Man Li
- Key Laboratory of Plant Resources Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), Guizhou University, College of Life Sciences/Institute of Agro-bioengineering, Guiyang 550025, China
| | - Xi Chen
- Plant Conservation Technology Center, Guizhou Key Laboratory of Agricultural Biotechnology, Guizhou Academy of Agricultural Sciences, Guiyang 550006, China
| | - De-Gang Zhao
- Key Laboratory of Plant Resources Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), Guizhou University, College of Life Sciences/Institute of Agro-bioengineering, Guiyang 550025, China; Plant Conservation Technology Center, Guizhou Key Laboratory of Agricultural Biotechnology, Guizhou Academy of Agricultural Sciences, Guiyang 550006, China.
| |
Collapse
|
21
|
Kaur S, Verma H, Dhiman M, Mantha AK. Activation of multifunctional DNA repair APE1/Ref-1 enzyme by the dietary phytochemical Ferulic acid protects human neuroblastoma SH-SY5Y cells against Aβ(25-35)-induced oxidative stress and inflammatory responses. Mitochondrion 2024; 79:101947. [PMID: 39151817 DOI: 10.1016/j.mito.2024.101947] [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: 05/16/2024] [Revised: 07/24/2024] [Accepted: 08/12/2024] [Indexed: 08/19/2024]
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disorder associated with the amyloid beta (Aβ) and tau hallmarks. The molecular insights into how neuroinflammation is initially triggered and how it affects neuronal cells are yet at the age of infancy. In this study, SH-SY5Y cells were used as a model for neurons by differentiating and were co-cultured with differentiated THP1 cells (microglia model) as well as treated with Aβ(25-35) and with antioxidant FA to study inflammatory, oxidative stress responses and their effects on co-cultured neurons. Neurons co-cultured with microglial cells showed pronounced increase in ROS levels, NOS expression, truncated N-terminal form (34 kDa) of APE1 expression and AIF's translocation in the nucleus. The pre-treatment of FA, on the other hand reversed these effects. It was further evaluated how FA/Aβ treatment altered microglial phenotype that in turn affected the neurons. Microglial cells showed M1 phenotype upon Aβ(25-35) stress, while FA induced M2 phenotype against Aβ stress, suggesting that FA alleviated Aβ induced phenotype and its associated effects in the co-cultured neurons by altering the phenotype of microglial cells and induced expression of full length (37 kDa) APE1 enzyme and inhibiting AIF's nuclear translocation, thus inhibiting apoptosis. This is the first study that revealed Aβ induced cleavage of APE1 enzyme in differentiated neurons, suggesting that APE1 may be the potential early target of Aβ that loses its function and exacerbates AD pathology. FA activated a fully functional form of APE1 against Aβ stress. The impaired function of APE1 could be the initial mechanism by which Aβ induces oxidative and inflammatory responses and dietary phytochemical FA can be a potential therapeutic strategy in managing the disease by activating APE1 that not only repairs oxidative DNA base damage but also maintains mitochondrial function and alleviates neuroinflammatory responses.
Collapse
Affiliation(s)
- Sharanjot Kaur
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Ghudda Village, Bathinda, Punjab, India
| | - Harkomal Verma
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda Village, Bathinda, Punjab, India
| | - Monisha Dhiman
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Ghudda Village, Bathinda, Punjab, India.
| | - Anil Kumar Mantha
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda Village, Bathinda, Punjab, India.
| |
Collapse
|
22
|
Li S, Redweik G, Lin JLJ, Chen YN, Yuan HS, Xue D. Probing the importance of AIF interaction with endonuclease G in mitochondrial inheritance and neurodegeneration. Cell Discov 2024; 10:107. [PMID: 39438436 PMCID: PMC11496498 DOI: 10.1038/s41421-024-00736-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 09/16/2024] [Indexed: 10/25/2024] Open
Affiliation(s)
- Shanshan Li
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Graham Redweik
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, USA
| | - Jason L J Lin
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, China
| | - Yi-Ning Chen
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, China
| | - Hanna S Yuan
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, China
| | - Ding Xue
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, USA.
| |
Collapse
|
23
|
Fogarasi M, Dima S. Immunomodulatory Functions of TNF-Related Apoptosis-Inducing Ligand in Type 1 Diabetes. Cells 2024; 13:1676. [PMID: 39451194 PMCID: PMC11506310 DOI: 10.3390/cells13201676] [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: 09/02/2024] [Revised: 10/01/2024] [Accepted: 10/09/2024] [Indexed: 10/26/2024] Open
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF protein superfamily and was initially identified as a protein capable of inducing apoptosis in cancer cells. In addition, TRAIL can promote pro-survival and proliferation signaling in various cell types. Subsequent studies have demonstrated that TRAIL plays several important roles in immunoregulation, immunosuppression, and immune effector functions. Type 1 diabetes (T1D) is an autoimmune disease characterized by hyperglycemia due to the loss of insulin-producing β-cells, primarily driven by T-cell-mediated pancreatic islet inflammation. Various genetic, epigenetic, and environmental factors, in conjunction with the immune system, contribute to the initiation, development, and progression of T1D. Recent reports have highlighted TRAIL as an important immunomodulatory molecule with protective effects on pancreatic islets. Experimental data suggest that TRAIL protects against T1D by reducing the proliferation of diabetogenic T cells and pancreatic islet inflammation and restoring normoglycemia in animal models. In this review, we aimed to summarize the consequences of TRAIL action in T1D, focusing on and discussing its signaling mechanisms, role in the immune system, and protective effects in T1D.
Collapse
Affiliation(s)
- Marton Fogarasi
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Simona Dima
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, 022328 Bucharest, Romania
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
24
|
Wang J, Wang ZQ, Zong W. ADP-ribose hydrolases: biological functions and potential therapeutic targets. Expert Rev Mol Med 2024; 26:e21. [PMID: 39375922 PMCID: PMC11488344 DOI: 10.1017/erm.2024.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/19/2024] [Accepted: 05/15/2024] [Indexed: 10/09/2024]
Abstract
ADP-ribosylation (ADPRylation), which encompasses poly(ADP-ribosyl)ation and mono(ADP-ribosyl)ation, is an important post-translational modification catalysed by the poly(ADP-ribose) polymerase (PARP) enzyme superfamily. The process involves writers (PARPs) and erasers (ADP-ribose hydrolases), which work together to precisely regulate diverse cellular and molecular responses. Although the PARP-mediated synthesis of ADP-ribose (ADPr) has been well studied, ADPr degradation by degrading enzymes deserves further investigation. Nonetheless, recent studies have provided important new insights into the biology and functions of ADPr hydrolases. Notably, research has illuminated the significance of the poly(ADP-ribose) degradation pathway and its activation by the coordinated actions of poly(ADP-ribose) glycohydrolase and other ADPr hydrolases, which have been identified as key components of ADPRylation signalling networks. The degradation pathway has been proposed to play crucial roles in key cellular processes, such as DNA damage repair, chromatin dynamics, transcriptional regulation and cell death. A deep understanding of these ADPr erasing enzymes provides insights into the biological roles of ADPRylation in human health and disease aetiology and paves the road for the development of novel therapeutic strategies. This review article provides a summary of current knowledge about the biochemical and molecular functions of ADPr erasers and their physiological implications in human pathology.
Collapse
Affiliation(s)
- Jingpeng Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, P. R. China
| | - Zhao-Qi Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, P. R. China
- Faculty of Biological Sciences, Friedrich-Schiller University of Jena, Jena 07743, Germany
| | - Wen Zong
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, P. R. China
| |
Collapse
|
25
|
Zhu Y, Liu H, Zheng L, Luo Y, Zhou G, Li J, Hou Y, Fu X. Vitrification of Mammalian Oocytes: Recent Studies on Mitochondrial Dysfunction. Biopreserv Biobank 2024; 22:428-440. [PMID: 38227396 DOI: 10.1089/bio.2023.0062] [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: 01/17/2024] Open
Abstract
Vitrification of reproductive cells is definitely essential and integral in animal breeding, as well as in assisted reproduction. However, issues accompanied with this technology such as decreased oocyte competency and relatively low embryo survival rates appear to be a tough conundrum that has long perplexed us. As significant organelles in cell metabolism, mitochondria play pivotal roles in numerous pathways. Nonetheless, extensive evidence has demonstrated that vitrification can seriously impair mitochondrial function in mammalian oocytes. Thus, in this article, we summarize the current progress in oocyte vitrification and particularly outline the common mitochondrial abnormalities alongside subsequent injury cascades seen in mammalian oocytes following vitrification. Based on existing literature, we tentatively come up with the potential mechanisms related to mitochondrial dysfunction and generalize efficacious ways which have been recommended to restore mitochondrial function.
Collapse
Affiliation(s)
- Yixiao Zhu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the MARA, National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory for Animal Genetic Improvement, State Key Laboratory of Animal Biotech Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Hongyu Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the MARA, National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory for Animal Genetic Improvement, State Key Laboratory of Animal Biotech Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Lv Zheng
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the MARA, National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory for Animal Genetic Improvement, State Key Laboratory of Animal Biotech Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Yuwen Luo
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Guizhen Zhou
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the MARA, National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory for Animal Genetic Improvement, State Key Laboratory of Animal Biotech Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Jun Li
- Department of Reproductive Medicine, Reproductive Medical Center, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yunpeng Hou
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xiangwei Fu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the MARA, National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory for Animal Genetic Improvement, State Key Laboratory of Animal Biotech Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| |
Collapse
|
26
|
Yang L, Guttman L, Dawson VL, Dawson TM. Parthanatos: Mechanisms, modulation, and therapeutic prospects in neurodegenerative disease and stroke. Biochem Pharmacol 2024; 228:116174. [PMID: 38552851 PMCID: PMC11410548 DOI: 10.1016/j.bcp.2024.116174] [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: 01/21/2024] [Revised: 03/16/2024] [Accepted: 03/26/2024] [Indexed: 04/06/2024]
Abstract
Parthanatos is a cell death signaling pathway that has emerged as a compelling target for pharmaceutical intervention. It plays a pivotal role in the neuron loss and neuroinflammation that occurs in Parkinson's Disease (PD), Alzheimer's Disease (AD), Huntington's Disease (HD), Amyotrophic Lateral Sclerosis (ALS), and stroke. There are currently no treatments available to humans to prevent cell death in any of these diseases. This review provides an in-depth examination of the current understanding of the Parthanatos mechanism, with a particular focus on its implications in neuroinflammation and various diseases discussed herein. Furthermore, we thoroughly review potential intervention targets within the Parthanatos pathway. We dissect recent progress in inhibitory strategies, complimented by a detailed structural analysis of key Parthanatos executioners, PARP-1, AIF, and MIF, along with an assessment of their established inhibitors. We hope to introduce a new perspective on the feasibility of targeting components within the Parthanatos pathway, emphasizing its potential to bring about transformative outcomes in therapeutic interventions. By delineating therapeutic opportunities and known targets, we seek to emphasize the imperative of blocking Parthanatos as a precursor to developing disease-modifying treatments. This comprehensive exploration aims to catalyze a paradigm shift in our understanding of potential neurodegenerative disease therapeutics, advocating for the pursuit of effective interventions centered around Parthanatos inhibition.
Collapse
Affiliation(s)
- Liu Yang
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Lauren Guttman
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
27
|
Song X, Hao X, Zhu BT. Role of mitochondrial reactive oxygen species in chemically-induced ferroptosis. Free Radic Biol Med 2024; 223:473-492. [PMID: 38992393 DOI: 10.1016/j.freeradbiomed.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 07/02/2024] [Accepted: 07/05/2024] [Indexed: 07/13/2024]
Abstract
Ferroptosis is a form of iron-dependent regulated cell death which is different from apoptosis. Chemically-induced ferroptosis is characterized by an accumulation of lipid reactive oxygen species (ROS) in the cells. A number of earlier studies have suggested the involvement of mitochondrial ROS in ferroptosis, and the present study seeks to further investigate the role of mitochondrial ROS in the induction of chemically-induced ferroptotic cell death. We find that during erastin-induced, glutathione depletion-associated ferroptosis, mitochondrial ROS accumulation is an important late event, which likely is involved in the final execution of ferroptotic cell death. The mitochondrion-originated ROS is found to accumulate in large quantities inside the nuclei during the late phases of erastin-induced ferroptosis. Completion of the late-phase accumulation of mitochondrion-produced ROS inside the nucleus of a cell likely marks an irreversible point in the cell death process. Similarly, accumulation of large amounts of mitochondrion-produced ROS inside the nucleus is also observed in the late phases of RSL3-induced ferroptosis. The results of this study indicate that the mitochondrial ROS play an important role in the final steps of both erastin- and RSL3-induced ferroptotic cell death.
Collapse
Affiliation(s)
- Xiuhan Song
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Xiangyu Hao
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Bao Ting Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China; Shenzhen Bay Laboratory, Shenzhen, 518055, China.
| |
Collapse
|
28
|
Brogyanyi T, Kejík Z, Veselá K, Dytrych P, Hoskovec D, Masařik M, Babula P, Kaplánek R, Přibyl T, Zelenka J, Ruml T, Vokurka M, Martásek P, Jakubek M. Iron chelators as mitophagy agents: Potential and limitations. Biomed Pharmacother 2024; 179:117407. [PMID: 39265234 DOI: 10.1016/j.biopha.2024.117407] [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: 06/14/2024] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/14/2024] Open
Abstract
Mitochondrial autophagy (mitophagy) is very important process for the maintenance of cellular homeostasis, functionality and survival. Its dysregulation is associated with high risk and progression numerous serious diseases (e.g., oncological, neurodegenerative and cardiovascular ones). Therefore, targeting mitophagy mechanisms is very hot topic in the biological and medicinal research. The interrelationships between the regulation of mitophagy and iron homeostasis are now becoming apparent. In short, mitochondria are central point for the regulation of iron homeostasis, but change in intracellular cheatable iron level can induce/repress mitophagy. In this review, relationships between iron homeostasis and mitophagy are thoroughly discussed and described. Also, therapeutic applicability of mitophagy chelators in the context of individual diseases is comprehensively and critically evaluated.
Collapse
Affiliation(s)
- Tereza Brogyanyi
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic; Institute of Pathological Physiology, First Faculty of Medicine, Charles University in Prague, U Nemocnice 5, 1, Prague 28 53, Czech Republic
| | - Zdeněk Kejík
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic
| | - Kateřina Veselá
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic
| | - Petr Dytrych
- 1st Department of Surgery-Department of Abdominal, Thoracic Surgery and Traumatology, First Faculty of Medicine, Charles University and General University Hospital, U Nemocnice 2, Prague 121 08, Czech Republic
| | - David Hoskovec
- 1st Department of Surgery-Department of Abdominal, Thoracic Surgery and Traumatology, First Faculty of Medicine, Charles University and General University Hospital, U Nemocnice 2, Prague 121 08, Czech Republic
| | - Michal Masařik
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic; Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno CZ-625 00, Czech Republic; Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
| | - Petr Babula
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno CZ-625 00, Czech Republic
| | - Robert Kaplánek
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic
| | - Tomáš Přibyl
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Prague 166 28, Czech Republic
| | - Jaroslav Zelenka
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Prague 166 28, Czech Republic
| | - Tomáš Ruml
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Prague 166 28, Czech Republic
| | - Martin Vokurka
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University in Prague, U Nemocnice 5, 1, Prague 28 53, Czech Republic
| | - Pavel Martásek
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic
| | - Milan Jakubek
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic.
| |
Collapse
|
29
|
Tang Y, Guo S, Yu N, Li H. ZIP4: a promising early diagnostic and therapeutic targets for pancreatic cancer. Am J Cancer Res 2024; 14:4652-4664. [PMID: 39417191 PMCID: PMC11477812 DOI: 10.62347/avym3477] [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: 05/11/2024] [Accepted: 09/15/2024] [Indexed: 10/19/2024] Open
Abstract
Pancreatic cancer is an aggressive and metastatic tumor that lacks effective early detection and treatment methods. There is an urgent need to further understand its underlying molecular mechanisms and identify new biomarkers for early detection. Zinc, a critical trace element and catalytic cofactor, is tightly regulated within cells. ZIP4, a zinc transporter protein significantly overexpressed in human pancreatic cancer, appears to play a pivotal role in tumor development by modulating intracellular zinc concentration. This review highlights the role of ZIP4 in tumorigenesis, including its impact on pancreatic cancer growth, proliferation, migration, and drug resistance. ZIP4 exerts its effects by regulating zinc dependent transcriptional factors like CREB, STAT3, and ZEB1, resulting in upregulation of Cyclin D1, TP53INP1, ITGA3, CD44, ENT1 proteins, and miR-373. Moreover, ZIP4 mediates the miR373-PHLPP2-AKT signaling axis, which increases TGF-β expression. Coupled with CREB-activated macrophage catabolism-related genes SDC1 and DNM2, ZIP4 promotes cancer cachexia and supports amino acids to tumor cells under metabolic stress. Furthermore, ZIP4 facilitates bone resorption by osteoclasts via the RANKL-activated NF-κB pathway. A deeper understanding of these mechanisms may unveil potential targets for early diagnosis, prognosis assessment, and dietary recommendations for pancreatic cancer. These findings hold clinical significance not only for pancreatic cancer but also for other malignancies exhibiting heightened ZIP4 expression.
Collapse
Affiliation(s)
- Yunpeng Tang
- Department of Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai, PR China
| | - Sheng Guo
- Department of Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai, PR China
| | - Nianhui Yu
- Department of Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai, PR China
| | - Hui Li
- Department of Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai, PR China
| |
Collapse
|
30
|
Hou W, Shen L, Zhu Y, Wang X, Du T, Yang F, Zhu Y. Fullerene Derivatives for Tumor Treatment: Mechanisms and Application. Int J Nanomedicine 2024; 19:9771-9797. [PMID: 39345909 PMCID: PMC11430870 DOI: 10.2147/ijn.s476601] [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: 05/02/2024] [Accepted: 08/28/2024] [Indexed: 10/01/2024] Open
Abstract
Fullerenes hold tremendous potential as alternatives to conventional chemotherapy or radiotherapy for tumor treatment due to their abilities to photodynamically kill tumor cells, destroy the tumor vasculature, inhibit tumor metastasis and activate anti-tumor immune responses, while protecting normal tissue through antioxidative effects. The symmetrical hollow molecular structures of fullerenes with abundant C=C bonds allow versatile chemical modification with diverse functional groups, metal clusters and biomacromolecules to synthesize a wide range of fullerene derivatives with increased water solubility, improved biocompatibility, enhanced photodynamic properties and stronger targeting abilities. This review introduces the anti-tumor mechanisms of fullerenes and summarizes the most recent works on the functionalization of fullerenes and the application of fullerene derivatives in tumor treatment. This review aims to serve as a valuable reference for further development and clinical application of anti-tumor fullerene derivatives.
Collapse
Affiliation(s)
- Wenjia Hou
- Health Science Center, Ningbo University, Ningbo, 315211, People's Republic of China
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200438, People's Republic of China
| | - Lan Shen
- Health Science Center, Ningbo University, Ningbo, 315211, People's Republic of China
| | - Yimin Zhu
- Health Science Center, Ningbo University, Ningbo, 315211, People's Republic of China
| | - Xuanjia Wang
- Health Science Center, Ningbo University, Ningbo, 315211, People's Republic of China
| | - Tianyu Du
- Health Science Center, Ningbo University, Ningbo, 315211, People's Republic of China
| | - Fang Yang
- Health Science Center, Ningbo University, Ningbo, 315211, People's Republic of China
| | - Yabin Zhu
- Health Science Center, Ningbo University, Ningbo, 315211, People's Republic of China
| |
Collapse
|
31
|
Lu H, Xu L, Steriopoulos J, McLeod P, Huang X, Min J, Peng T, Jevnikar AM, Zhang ZX. An acidic pH environment converts necroptosis to apoptosis. Biochem Biophys Res Commun 2024; 725:150215. [PMID: 38870845 DOI: 10.1016/j.bbrc.2024.150215] [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: 05/21/2024] [Revised: 05/24/2024] [Accepted: 06/02/2024] [Indexed: 06/15/2024]
Abstract
Cardiac ischemia results in anaerobic metabolism and lactic acid accumulation and with time, intracellular and extracellular acidosis. Ischemia and subsequent reperfusion injury (IRI) lead to various forms of programmed cell death. Necroptosis is a major form of programmed necrosis that worsens cardiac function directly and also promotes inflammation by the release of cellular contents. Potential effects of increasing acidosis on programmed cell death and their specific components have not been well studied. While apoptosis is caspase-dependent, in contrast, necroptosis is mediated by the receptor-interacting protein kinases 1 and 3 (RIPK1/3). In our study, we observed that at physiological pH = 7.4, caspase-8 inhibition did not prevent TNFα-induced cell death in mouse cardiac vascular endothelial cells (MVECs) but promoted necroptotic cell death. As expected, necroptosis was blocked by RIPK1 inhibition. However, at pH = 6.5, TNFα induced an apoptosis-like pattern which was inhibited by caspase-8 inhibition. Interestingly phosphorylation of necroptotic molecules RIPK1, RIPK3, and mixed lineage kinase domain-like protein (MLKL) was enhanced in an acidic pH environment. However, RIPK3 and MLKL phosphorylation was self-limited which may have limited their participation in necroptosis. In addition, an acidic pH promoted apoptosis-inducing factor (AIF) cleavage and nuclear translocation. AIF RNA silencing inhibited cell death, supporting the role of AIF in this cell death. In summary, our study demonstrated that the pH of the micro-environment during inflammation can bias cell death pathways by altering the function of necroptosis-related molecules and promoting AIF-mediated cell death. Further insights into the mechanisms by which an acidic cellular micro-environment influences these and perhaps other forms of regulated cell death, may lead to therapeutic strategies to attenuate IRI.
Collapse
Affiliation(s)
- Haitao Lu
- Matthew Mailing Centre for Translational Transplantation Studies. Lawson Health Research Institute, London, Canada; Department of Pathology, Western University, London, Canada
| | - Laura Xu
- Matthew Mailing Centre for Translational Transplantation Studies. Lawson Health Research Institute, London, Canada; Department of Pathology, Western University, London, Canada
| | - Julia Steriopoulos
- Matthew Mailing Centre for Translational Transplantation Studies. Lawson Health Research Institute, London, Canada; Department of Pathology, Western University, London, Canada
| | - Patrick McLeod
- Matthew Mailing Centre for Translational Transplantation Studies. Lawson Health Research Institute, London, Canada; Multi-Organ Transplant Program, London Health Sciences Centre. London, Canada
| | - Xuyan Huang
- Matthew Mailing Centre for Translational Transplantation Studies. Lawson Health Research Institute, London, Canada
| | - Jeffery Min
- Matthew Mailing Centre for Translational Transplantation Studies. Lawson Health Research Institute, London, Canada
| | - Tianging Peng
- Department of Pathology, Western University, London, Canada; Division of Nephrology, Department of Medicine, Western University. London, Canada
| | - Anthony M Jevnikar
- Matthew Mailing Centre for Translational Transplantation Studies. Lawson Health Research Institute, London, Canada; Multi-Organ Transplant Program, London Health Sciences Centre. London, Canada; Division of Nephrology, Department of Medicine, Western University. London, Canada
| | - Zhu-Xu Zhang
- Matthew Mailing Centre for Translational Transplantation Studies. Lawson Health Research Institute, London, Canada; Department of Pathology, Western University, London, Canada; Multi-Organ Transplant Program, London Health Sciences Centre. London, Canada; Division of Nephrology, Department of Medicine, Western University. London, Canada.
| |
Collapse
|
32
|
Guo D, Liu Z, Zhou J, Ke C, Li D. Significance of Programmed Cell Death Pathways in Neurodegenerative Diseases. Int J Mol Sci 2024; 25:9947. [PMID: 39337436 PMCID: PMC11432010 DOI: 10.3390/ijms25189947] [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: 07/27/2024] [Revised: 09/07/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Programmed cell death (PCD) is a form of cell death distinct from accidental cell death (ACD) and is also referred to as regulated cell death (RCD). Typically, PCD signaling events are precisely regulated by various biomolecules in both spatial and temporal contexts to promote neuronal development, establish neural architecture, and shape the central nervous system (CNS), although the role of PCD extends beyond the CNS. Abnormalities in PCD signaling cascades contribute to the irreversible loss of neuronal cells and function, leading to the onset and progression of neurodegenerative diseases. In this review, we summarize the molecular processes and features of different modalities of PCD, including apoptosis, necroptosis, pyroptosis, ferroptosis, cuproptosis, and other novel forms of PCD, and their effects on the pathogenesis of neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS), spinal muscular atrophy (SMA), multiple sclerosis (MS), traumatic brain injury (TBI), and stroke. Additionally, we examine the key factors involved in these PCD signaling pathways and discuss the potential for their development as therapeutic targets and strategies. Therefore, therapeutic strategies targeting the inhibition or facilitation of PCD signaling pathways offer a promising approach for clinical applications in treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Dong Guo
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou 350117, China
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou 350117, China
| | - Zhihao Liu
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou 350117, China
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou 350117, China
| | - Jinglin Zhou
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou 350117, China
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou 350117, China
| | - Chongrong Ke
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou 350117, China
| | - Daliang Li
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou 350117, China
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou 350117, China
| |
Collapse
|
33
|
Gupta KB, Taylor TL, Panda SS, Thangaraju M, Lokeshwar BL. Curcumin-Dichloroacetate Hybrid Molecule as an Antitumor Oral Drug against Multidrug-Resistant Advanced Bladder Cancers. Cancers (Basel) 2024; 16:3108. [PMID: 39272966 PMCID: PMC11394085 DOI: 10.3390/cancers16173108] [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: 08/26/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
Tumor cells produce excessive reactive oxygen species (ROS) but cannot detoxify ROS if they are due to an external agent. An agent that produces toxic levels of ROS, specifically in tumor cells, could be an effective anticancer drug. CMC-2 is a molecular hybrid of the bioactive polyphenol curcumin conjugated to dichloroacetate (DCA) via a glycine bridge. The CMC-2 was tested for its cytotoxic antitumor activities and killed both naïve and multidrug-resistant (MDR) bladder cancer (BCa) cells with equal potency (<1.0 µM); CMC-2 was about 10-15 folds more potent than curcumin or DCA. Growth of human BCa xenograft in mice was reduced by >50% by oral gavage of 50 mg/kg of CMC-2 without recognizable systemic toxicity. Doses that used curcumin or DCA showed minimum antitumor effects. In vitro, the toxicity of CMC-2 in both naïve and MDR cells depended on increased intracellular ROS in tumor cells but not in normal cells at comparable doses. Increased ROS caused the permeabilization of mitochondria and induced apoptosis. Further, adding N-Acetyl cysteine (NAC), a hydroxyl radical scavenger, abolished excessive ROS production and CMC-2's cytotoxicity. The lack of systemic toxicity, equal potency against chemotherapy -naïve and resistant tumors, and oral bioavailability establish the potential of CMC-2 as a potent drug against bladder cancers.
Collapse
Affiliation(s)
| | - Truett L Taylor
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
| | - Siva S Panda
- Department of Chemistry and Biochemistry, College of Science and Mathematics, Augusta University, Augusta, GA 30912, USA
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Muthusamy Thangaraju
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Bal L Lokeshwar
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
34
|
Emamnejad R, Pagnin M, Petratos S. The iron maiden: Oligodendroglial metabolic dysfunction in multiple sclerosis and mitochondrial signaling. Neurosci Biobehav Rev 2024; 164:105788. [PMID: 38950685 DOI: 10.1016/j.neubiorev.2024.105788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/06/2024] [Accepted: 06/24/2024] [Indexed: 07/03/2024]
Abstract
Multiple sclerosis (MS) is an autoimmune disease, governed by oligodendrocyte (OL) dystrophy and central nervous system (CNS) demyelination manifesting variable neurological impairments. Mitochondrial mechanisms may drive myelin biogenesis maintaining the axo-glial unit according to dynamic requisite demands imposed by the axons they ensheath. The promotion of OL maturation and myelination by actively transporting thyroid hormone (TH) into the CNS and thereby facilitating key transcriptional and metabolic pathways that regulate myelin biogenesis is fundamental to sustain the profound energy demands at each axo-glial interface. Deficits in regulatory functions exerted through TH for these physiological roles to be orchestrated by mature OLs, can occur in genetic and acquired myelin disorders, whereby mitochondrial efficiency and eventual dysfunction can lead to profound oligodendrocytopathy, demyelination and neurodegenerative sequelae. TH-dependent transcriptional and metabolic pathways can be dysregulated during acute and chronic MS lesion activity depriving OLs from critical acetyl-CoA biochemical mechanisms governing myelin lipid biosynthesis and at the same time altering the generation of iron metabolism that may drive ferroptotic mechanisms, leading to advancing neurodegeneration.
Collapse
Affiliation(s)
- Rahimeh Emamnejad
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, Victoria 3004, Australia.
| | - Maurice Pagnin
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, Victoria 3004, Australia.
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, Victoria 3004, Australia.
| |
Collapse
|
35
|
Moura RDD, Mattos PDD, Valente PF, Hoch NC. Molecular mechanisms of cell death by parthanatos: More questions than answers. Genet Mol Biol 2024; 47Suppl 1:e20230357. [PMID: 39356140 PMCID: PMC11445734 DOI: 10.1590/1678-4685-gmb-2023-0357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 06/16/2024] [Indexed: 10/03/2024] Open
Abstract
Regulated cell death by a non-apoptotic pathway known as parthanatos is increasingly recognised as a central player in pathological processes, including ischaemic tissue damage and neurodegenerative diseases. Parthanatos is activated under conditions that induce high levels of DNA damage, leading to hyperactivation of the DNA damage sensor PARP1. While this strict dependence on PARP1 activation is a defining feature of parthanatos that distinguishes it from other forms of cell death, the molecular events downstream of PARP1 activation remain poorly understood. In this mini-review, we highlight a number of important questions that remain to be answered about this enigmatic form of cell death.
Collapse
Affiliation(s)
- Rafael Dias de Moura
- Universidade de São Paulo, Instituto de Química, Departamento de Bioquímica, São Paulo, SP, Brasil
| | | | | | - Nícolas Carlos Hoch
- Universidade de São Paulo, Instituto de Química, Departamento de Bioquímica, São Paulo, SP, Brasil
| |
Collapse
|
36
|
Liu H, Cai G, Yuan S, Zhou X, Gui R, Huang R. Platelet Membrane-Camouflaged Silver Metal-Organic Framework Biomimetic Nanoparticles for the Treatment of Triple-Negative Breast Cancer. Mol Pharm 2024; 21:3577-3590. [PMID: 38857525 DOI: 10.1021/acs.molpharmaceut.4c00307] [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: 06/12/2024]
Abstract
Triple-negative breast cancer (TNBC) is characterized by high malignancy and limited treatment options. Given the pressing need for more effective treatments for TNBC, this study aimed to develop platelet membrane (PM)-camouflaged silver metal-organic framework nanoparticles (PM@MOF-Ag NPs), a biomimetic nanodrug. PM@MOF-Ag NP construction involved the utilization of 2-methylimidazole and silver nitrate to prepare silver metal-organic framework (MOF-Ag) NPs. The PM@MOF-Ag NPs, due to their camouflage, possess excellent blood compatibility, immune escape ability, and a strong affinity for 4T1 tumor cells. This enhances their circulation time in vivo and promotes the aggregation of PM@MOF-Ag NPs at the 4T1 tumor site. Importantly, PM@MOF-Ag NPs demonstrated promising antitumor activity in vitro and in vivo. We further revealed that PM@MOF-Ag NPs induced tumor cell death by overproducing reactive oxygen species and promoting cell apoptosis. Moreover, PM@MOF-Ag NPs enhanced apoptosis by upregulating the ratios of Bax/Bcl-2 and cleaved caspase3/pro-caspase3. Notably, PM@MOF-Ag NPs exhibited no significant organ toxicity, whereas the administration of MOF-Ag NPs resulted in liver inflammation compared to the control group.
Collapse
Affiliation(s)
- Haiting Liu
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P. R. China
| | - Guangqing Cai
- Department of Orthopedics, Changsha Hospital of Traditional Chinese Medicine, Changsha Eighth Hospital, Changsha, Hunan 410013, P. R. China
| | - Shuai Yuan
- Guangzhou Customs District Technology Center, Guangzhou 510700, China
| | - Xionghui Zhou
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P. R. China
| | - Rong Gui
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P. R. China
| | - Rong Huang
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P. R. China
| |
Collapse
|
37
|
Masanta S, Wiesyk A, Panja C, Pilch S, Ciesla J, Sipko M, De A, Enkhbaatar T, Maslanka R, Skoneczna A, Kucharczyk R. Fmp40 ampylase regulates cell survival upon oxidative stress by controlling Prx1 and Trx3 oxidation. Redox Biol 2024; 73:103201. [PMID: 38795545 PMCID: PMC11140801 DOI: 10.1016/j.redox.2024.103201] [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: 04/21/2024] [Revised: 05/16/2024] [Accepted: 05/19/2024] [Indexed: 05/28/2024] Open
Abstract
Reactive oxygen species (ROS), play important roles in cellular signaling, nonetheless are toxic at higher concentrations. Cells have many interconnected, overlapped or backup systems to neutralize ROS, but their regulatory mechanisms remain poorly understood. Here, we reveal an essential role for mitochondrial AMPylase Fmp40 from budding yeast in regulating the redox states of the mitochondrial 1-Cys peroxiredoxin Prx1, which is the only protein shown to neutralize H2O2 with the oxidation of the mitochondrial glutathione and the thioredoxin Trx3, directly involved in the reduction of Prx1. Deletion of FMP40 impacts a cellular response to H2O2 treatment that leads to programmed cell death (PCD) induction and an adaptive response involving up or down regulation of genes encoding, among others the catalase Cta1, PCD inducing factor Aif1, and mitochondrial redoxins Trx3 and Grx2. This ultimately perturbs the reduced glutathione and NADPH cellular pools. We further demonstrated that Fmp40 AMPylates Prx1, Trx3, and Grx2 in vitro and interacts with Trx3 in vivo. AMPylation of the threonine residue 66 in Trx3 is essential for this protein's proper endogenous level and its precursor forms' maturation under oxidative stress conditions. Additionally, we showed the Grx2 involvement in the reduction of Trx3 in vivo. Taken together, Fmp40, through control of the reduction of mitochondrial redoxins, regulates the hydrogen peroxide, GSH and NADPH signaling influencing the yeast cell survival.
Collapse
Affiliation(s)
- Suchismita Masanta
- Institute of Biochemistry and Biophysics PAS, Warsaw, 02-106, Pawinskiego 5A, Poland
| | - Aneta Wiesyk
- Institute of Biochemistry and Biophysics PAS, Warsaw, 02-106, Pawinskiego 5A, Poland
| | - Chiranjit Panja
- Institute of Biochemistry and Biophysics PAS, Warsaw, 02-106, Pawinskiego 5A, Poland
| | - Sylwia Pilch
- Institute of Biochemistry and Biophysics PAS, Warsaw, 02-106, Pawinskiego 5A, Poland
| | - Jaroslaw Ciesla
- Institute of Biochemistry and Biophysics PAS, Warsaw, 02-106, Pawinskiego 5A, Poland
| | - Marta Sipko
- Institute of Biochemistry and Biophysics PAS, Warsaw, 02-106, Pawinskiego 5A, Poland
| | - Abhipsita De
- Institute of Biochemistry and Biophysics PAS, Warsaw, 02-106, Pawinskiego 5A, Poland
| | - Tuguldur Enkhbaatar
- Institute of Biochemistry and Biophysics PAS, Warsaw, 02-106, Pawinskiego 5A, Poland
| | - Roman Maslanka
- Institute of Biology, College of Natural Sciences, University of Rzeszow, Rzeszow, Poland
| | - Adrianna Skoneczna
- Institute of Biochemistry and Biophysics PAS, Warsaw, 02-106, Pawinskiego 5A, Poland
| | - Roza Kucharczyk
- Institute of Biochemistry and Biophysics PAS, Warsaw, 02-106, Pawinskiego 5A, Poland.
| |
Collapse
|
38
|
Huang M, Li Y, Li Y, Liu S. C-Terminal Binding Protein: Regulator between Viral Infection and Tumorigenesis. Viruses 2024; 16:988. [PMID: 38932279 PMCID: PMC11209466 DOI: 10.3390/v16060988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
C-terminal binding protein (CtBP), a transcriptional co-repressor, significantly influences cellular signaling, impacting various biological processes including cell proliferation, differentiation, apoptosis, and immune responses. The CtBP family comprises two highly conserved proteins, CtBP1 and CtBP2, which have been shown to play critical roles in both tumorigenesis and the regulation of viral infections. Elevated CtBP expression is noted in various tumor tissues, promoting tumorigenesis, invasiveness, and metastasis through multiple pathways. Additionally, CtBP's role in viral infections varies, exhibiting differing or even opposing effects depending on the virus. This review synthesizes the advances in CtBP's function research in viral infections and virus-associated tumorigenesis, offering new insights into potential antiviral and anticancer strategies.
Collapse
Affiliation(s)
- Meihui Huang
- Xiangya School of Medicine, Central South University, Changsha 410013, China; (M.H.); (Y.L.); (Y.L.)
| | - Yucong Li
- Xiangya School of Medicine, Central South University, Changsha 410013, China; (M.H.); (Y.L.); (Y.L.)
| | - Yuxiao Li
- Xiangya School of Medicine, Central South University, Changsha 410013, China; (M.H.); (Y.L.); (Y.L.)
| | - Shuiping Liu
- Xiangya School of Medicine, Central South University, Changsha 410013, China; (M.H.); (Y.L.); (Y.L.)
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410013, China
| |
Collapse
|
39
|
Vu A, Glassman I, Campbell G, Yeganyan S, Nguyen J, Shin A, Venketaraman V. Host Cell Death and Modulation of Immune Response against Mycobacterium tuberculosis Infection. Int J Mol Sci 2024; 25:6255. [PMID: 38892443 PMCID: PMC11172987 DOI: 10.3390/ijms25116255] [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: 05/01/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
Mycobacterium tuberculosis (Mtb) is the causative agent of tuberculosis (TB), a prevalent infectious disease affecting populations worldwide. A classic trait of TB pathology is the formation of granulomas, which wall off the pathogen, via the innate and adaptive immune systems. Some key players involved include tumor necrosis factor-alpha (TNF-α), foamy macrophages, type I interferons (IFNs), and reactive oxygen species, which may also show overlap with cell death pathways. Additionally, host cell death is a primary method for combating and controlling Mtb within the body, a process which is influenced by both host and bacterial factors. These cell death modalities have distinct molecular mechanisms and pathways. Programmed cell death (PCD), encompassing apoptosis and autophagy, typically confers a protective response against Mtb by containing the bacteria within dead macrophages, facilitating their phagocytosis by uninfected or neighboring cells, whereas necrotic cell death benefits the pathogen, leading to the release of bacteria extracellularly. Apoptosis is triggered via intrinsic and extrinsic caspase-dependent pathways as well as caspase-independent pathways. Necrosis is induced via various pathways, including necroptosis, pyroptosis, and ferroptosis. Given the pivotal role of host cell death pathways in host defense against Mtb, therapeutic agents targeting cell death signaling have been investigated for TB treatment. This review provides an overview of the diverse mechanisms underlying Mtb-induced host cell death, examining their implications for host immunity. Furthermore, it discusses the potential of targeting host cell death pathways as therapeutic and preventive strategies against Mtb infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (G.C.); (A.S.)
| |
Collapse
|
40
|
Cheng WH, Chang PL, Wu YC, Wang SA, Chen CL, Hsu FL, Neoh MM, Lin LY, Yuliani FS, Lin CH, Chen BC. Neutralization of CX3CL1 Attenuates TGF-β-Induced Fibroblast Differentiation Through NF-κB Activation and Mitochondrial Dysfunction in Airway Fibrosis. Lung 2024; 202:343-356. [PMID: 38678499 DOI: 10.1007/s00408-024-00701-6] [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: 01/25/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Severe asthma, characterized by inflammation and airway remodeling, involves fibroblast differentiation into myofibroblasts expressing α-SMA. This process leads to the production of fibronectin and connective tissue growth factor (CTGF), driven by factors such as transforming growth factor (TGF)-β. Furthermore, the persistent presence of myofibroblasts is associated with resistance to apoptosis and mitochondrial dysfunction. The chemokine (C-X3-C motif) ligand 1 (CX3CL1) plays a role in tissue fibrosis. However, it is currently unknown whether neutralization of CX3CL1 decreases TGF-β-induced fibroblast differentiation and mitochondrial dysfunction in normal human lung fibroblasts (NHLFs). METHODS CX3CL1/C-X3-C motif chemokine receptor 1 (CX3CR1), CX3CL1 was analyzed by immunofluorescence (IF) or immunohistochemical (IHC) staining of ovalbumin-challenged mice. CX3CL1 release was detected by ELISA. TGF-β-induced CTGF, fibronectin, and α-SMA expression were evaluated in NHLFs following neutralization of CX3CL1 (TP213) treatment for the indicated times by Western blotting or IF staining. Mitochondrion function was detected by a JC-1 assay and seahorse assay. Cell apoptosis was observed by a terminal uridine nick-end labeling (TUNEL) assay. RESULTS An increase in CX3CL1 expression was observed in lung tissues from mice with ovalbumin-induced asthma by IF staining. CX3CR1 was increased in the subepithelial layer of the airway by IHC staining. Moreover, CX3CR1 small interfering (si)RNA downregulated TGF-β-induced CTGF and fibronectin expression in NHLFs. CX3CL1 induced CTGF and fibronectin expression in NHLFs. TGF-β-induced CX3CL1 secretion from NHLFs. Furthermore, TP213 decreased TGF-β-induced CTGF, fibronectin, and α-SMA expression in NHLFs. Mitochondrion-related differentially expressed genes (DEGs) were examined after CX3CL1 neutralization in TGF-β-treated NHLFs. TP213 alleviated TGF-β-induced mitochondrial dysfunction and apoptosis resistance in NHLFs. CX3CL1 induced p65, IκBα, and IKKα phosphorylation in a time-dependent manner. Furthermore, CX3CL1-induced fibronectin expression and JC-1 monomer were decreased by p65 siRNA. TP213 reduced TGF-β-induced p65 and α-SMA expression in NHLFs. CONCLUSIONS These findings suggest that neutralizing CX3CL1 attenuates lung fibroblast activation and mitochondrial dysfunction. Understanding the impacts of CX3CL1 neutralization on fibroblast mitochondrial function could contribute to the development of therapeutic strategies for managing airway remodeling in severe asthma.
Collapse
Affiliation(s)
- Wun-Hao Cheng
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031, Taiwan
- Respiratory Therapy, Division of Pulmonary Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Division of Thoracic Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Pao-Lung Chang
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Chih Wu
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031, Taiwan
- Division of Thoracic Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shao-An Wang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031, Taiwan
- Division of Thoracic Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chia-Ling Chen
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031, Taiwan
- Division of Thoracic Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Feng-Lin Hsu
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031, Taiwan
| | - Mei-May Neoh
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031, Taiwan
| | - Lee-Yuan Lin
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Fara Silvia Yuliani
- Department of Pharmacology and Therapy, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Chien-Huang Lin
- Division of Thoracic Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031, Taiwan.
| | - Bing-Chang Chen
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031, Taiwan.
- Division of Thoracic Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
41
|
Liu Q, Wang D, Cui M, Li M, Zhang XE. A genetically encoded fluorescent protein sensor for mitochondrial membrane damage detection. Biochem Biophys Res Commun 2024; 709:149836. [PMID: 38564937 DOI: 10.1016/j.bbrc.2024.149836] [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: 12/20/2023] [Revised: 03/18/2024] [Accepted: 03/25/2024] [Indexed: 04/04/2024]
Abstract
Mitochondria are essential cellular organelles; detecting mitochondrial damage is crucial in cellular biology and toxicology. Compared with existing chemical probe detection methods, genetically encoded fluorescent protein sensors can directly indicate cellular and molecular events without involving exogenous reagents. In this study, we introduced a molecular sensor system, MMD-Sensor, for monitoring mitochondrial membrane damage. The sensor consists of two molecular modules. Module I is a fusion structure of the mitochondrial localization sequence (MLS), AIF cleavage site sequence (CSS), nuclear localization sequence (NLS), N-terminus of mNeonGreen and mCherry. Module II is a fusion structure of the C-terminus of mNeonGreen, NLS sequence, and mtagBFP2. Under normal condition, Module I is constrained in the inner mitochondrial membrane anchored by MLS, while Module II is restricted to the nucleus by its NLS fusion component. If the mitochondrial membrane is damaged, CSS is cut from the inner membrane, causing Module I to shift into the nucleus guided by the NLS fusion component. After Module I enters the nucleus, the N- and C-terminus of mNeonGreen meet each other and rebuild its intact 3D structure through fragment complementation and thus generates green fluorescence in the nucleus. Dynamic migration of red fluorescence from mitochondria to the nucleus and generation of green fluorescence in the nucleus indicate mitochondrial membrane damage. Using the MMD-Sensor, mitochondrial membrane damage induced by various reagents, such as uncoupling agents, ATP synthase inhibitors, monovalent cationic carriers, and ROS, in HeLa and 293T cells are directly observed and evaluated.
Collapse
Affiliation(s)
- Qian Liu
- Faculty of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Dianbing Wang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Mengmeng Cui
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Min Li
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xian-En Zhang
- Faculty of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
42
|
Wang X, Qiu W, Hu G, Diao X, Li Y, Li Y, Li P, Liu Y, Feng Y, Xue C, Cao Y, Xu Z. NS7a of SADS-CoV promotes viral infection via inducing apoptosis to suppress type III interferon production. J Virol 2024; 98:e0031724. [PMID: 38624231 PMCID: PMC11092342 DOI: 10.1128/jvi.00317-24] [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: 02/20/2024] [Accepted: 03/10/2024] [Indexed: 04/17/2024] Open
Abstract
Swine acute diarrhea syndrome coronavirus (SADS-CoV) is a newly discovered swine coronavirus with potential cross-species transmission risk. Although SADS-CoV-induced host cell apoptosis and innate immunity antagonization has been revealed, underlying signaling pathways remain obscure. Here, we demonstrated that infection of SADS-CoV induced apoptosis in vivo and in vitro, and that viral protein NS7a is mainly responsible for SADS-CoV-induced apoptosis in host cells. Furthermore, we found that NS7a interacted with apoptosis-inducing factor mitochondria associated 1 (AIFM1) to activate caspase-3 via caspase-6 in SADS-CoV-infected cells, and enhanced SADS-CoV replication. Importantly, NS7a suppressed poly(I:C)-induced expression of type III interferon (IFN-λ) via activating caspase-3 to cleave interferon regulatory factor 3 (IRF3), and caspase-3 inhibitor protects piglets against SADS-CoV infection in vivo. These findings reveal how SADS-CoV induced apoptosis to inhibit innate immunity and provide a valuable clue to the development of effective drugs for the clinical control of SADS-CoV infection.IMPORTANCEOver the last 20 years, multiple animal-originated coronaviruses, including severe acute respiratory syndrome coronavirus (SARS-CoV), middle east respiratory syndrome coronavirus (MERS-CoV), and SARS-CoV-2, have caused millions of deaths, seriously jeopardized human health, and hindered social development, indicating that the study of animal-originated coronaviruses with potential for cross-species transmission is particularly important. Bat-originated swine acute diarrhea syndrome coronavirus (SADS-CoV), discovered in 2017, can not only cause fatal diarrhea in piglets, but also infect multiple human cells, with a potential risk of cross-species transmission, but its pathogenesis is unclear. In this study, we demonstrated that NS7a of SADS-CoV suppresses IFN-λ production via apoptosis-inducing factor mitochondria associated 1 (AIFM1)-caspase-6-caspase-3-interferon regulatory factor 3 (IRF3) pathway, and caspase-3 inhibitor (Z-DEVD-FMK) can effectively inhibit SADS-CoV replication and protect infected piglets. Our findings in this study contribute to a better understanding of SADS-CoV-host interactions as a part of the coronaviruses pathogenesis and using apoptosis-inhibitor as a drug as potential therapeutic approaches for prevention and control of SADS-CoV infection.
Collapse
Affiliation(s)
- Xiaowei Wang
- State Key Laboratory of Biocontrol, School of Life Science, Sun Yat-sen University, Guangzhou, China
| | - Wenjing Qiu
- State Key Laboratory of Biocontrol, School of Life Science, Sun Yat-sen University, Guangzhou, China
| | - Guangli Hu
- State Key Laboratory of Biocontrol, School of Life Science, Sun Yat-sen University, Guangzhou, China
| | - Xiaoyuan Diao
- State Key Laboratory of Biocontrol, School of Life Science, Sun Yat-sen University, Guangzhou, China
| | - Yunfei Li
- State Key Laboratory of Biocontrol, School of Life Science, Sun Yat-sen University, Guangzhou, China
| | - Yue Li
- State Key Laboratory of Biocontrol, School of Life Science, Sun Yat-sen University, Guangzhou, China
| | - Peng Li
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, USA
| | - Yufang Liu
- State Key Laboratory of Biocontrol, School of Life Science, Sun Yat-sen University, Guangzhou, China
| | - Yongtong Feng
- State Key Laboratory of Biocontrol, School of Life Science, Sun Yat-sen University, Guangzhou, China
| | - Chunyi Xue
- State Key Laboratory of Biocontrol, School of Life Science, Sun Yat-sen University, Guangzhou, China
| | - Yongchang Cao
- State Key Laboratory of Biocontrol, School of Life Science, Sun Yat-sen University, Guangzhou, China
| | - Zhichao Xu
- State Key Laboratory of Biocontrol, School of Life Science, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
43
|
Singh A, Tiwari S, Singh S. Pirh2 modulates the mitochondrial function and cytochrome c-mediated neuronal death during Alzheimer's disease. Cell Death Dis 2024; 15:331. [PMID: 38740775 PMCID: PMC11091053 DOI: 10.1038/s41419-024-06662-1] [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: 08/29/2022] [Revised: 03/26/2024] [Accepted: 04/09/2024] [Indexed: 05/16/2024]
Abstract
Pirh2 is an E3 ubiquitin ligase known to regulate the DNA damage responses through ubiquitylation of various participating signaling factors. DNA damage is a key pathological contributor to Alzheimer's disease (AD), therefore, the role of Pirh2 was investigated in streptozotocin and oligomer Aβ1-42 induced rodent experimental model of AD. Pirh2 protein abundance increased during AD conditions, and transient silencing of Pirh2 inhibited the disease-specific pathological markers like level of p-Tau, βamyloid, acetylcholinesterase activity, and neuronal death. Biochemically, Pirh2 silencing significantly attenuated the oxidative stress, depleted mitochondrial membrane potential, cytochrome c translocation from mitochondria to cytosol, and depleted mitochondrial complex-I activity, and ATP level. Pirh2 silencing also inhibited the altered level of VDAC1, hsp75, hexokinase1, t-Bid, caspase-9, and altered level of apoptotic proteins (Bcl-2, Bax). MALDI-TOF/TOF, co-immunoprecipitation, and UbcH13-linked ubiquitylation assay confirmed the interaction of Pirh2 with cytochrome c and the role of Pirh2 in ubiquitylation of cytochrome c, along with Pirh2-dependent altered proteasome activity. Additionally, Pirh2 silencing further inhibited the translocation of mitochondrion-specific endonuclease G and apoptosis-inducing factors to the nucleus and DNA damage. In conclusion, findings suggested the significant implication of Pirh2 in disease pathogenesis, particularly through impaired mitochondrial function, including biochemical alterations, translocation of cytochrome c, endonuclease G and apoptosis-inducing factor, DNA damage, and neuronal apoptosis.
Collapse
Affiliation(s)
- Abhishek Singh
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shubhangini Tiwari
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Sarika Singh
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, 226031, India.
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
44
|
Shi T, Chen Z, Li J, Wang H, Wang Q. AIF translocation into nucleus caused by Aifm1 R450Q mutation: generation and characterization of a mouse model for AUNX1. Hum Mol Genet 2024; 33:905-918. [PMID: 38449065 PMCID: PMC11070138 DOI: 10.1093/hmg/ddae010] [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: 09/06/2023] [Revised: 10/30/2023] [Accepted: 01/05/2024] [Indexed: 03/08/2024] Open
Abstract
Mutations in AIFM1, encoding for apoptosis-inducing factor (AIF), cause AUNX1, an X-linked neurologic disorder with late-onset auditory neuropathy (AN) and peripheral neuropathy. Despite significant research on AIF, there are limited animal models with the disrupted AIFM1 representing the corresponding phenotype of human AUNX1, characterized by late-onset hearing loss and impaired auditory pathways. Here, we generated an Aifm1 p.R450Q knock-in mouse model (KI) based on the human AIFM1 p.R451Q mutation. Hemizygote KI male mice exhibited progressive hearing loss from P30 onward, with greater severity at P60 and stabilization until P210. Additionally, muscle atrophy was observed at P210. These phenotypic changes were accompanied by a gradual reduction in the number of spiral ganglion neuron cells (SGNs) at P30 and ribbons at P60, which coincided with the translocation of AIF into the nucleus starting from P21 and P30, respectively. The SGNs of KI mice at P210 displayed loss of cytomembrane integrity, abnormal nuclear morphology, and dendritic and axonal demyelination. Furthermore, the inner hair cells and myelin sheath displayed abnormal mitochondrial morphology, while fibroblasts from KI mice showed impaired mitochondrial function. In conclusion, we successfully generated a mouse model recapitulating AUNX1. Our findings indicate that disruption of Aifm1 induced the nuclear translocation of AIF, resulting in the impairment in the auditory pathway.
Collapse
Affiliation(s)
- Tao Shi
- Senior Department of Otolaryngology-Head and Neck Surgery, the Sixth Medical Center of PLA General Hospital, Medical School of Chinese PLA, 6 Fucheng Road, Beijing 100048, P. R. China
- National Clinical Research Center for Otolaryngologic Diseases, Chinese PLA General Hospital, 6 Fucheng Road, Beijing 100048, P. R. China
| | - Ziyi Chen
- Senior Department of Otolaryngology-Head and Neck Surgery, the Sixth Medical Center of PLA General Hospital, Medical School of Chinese PLA, 6 Fucheng Road, Beijing 100048, P. R. China
- National Clinical Research Center for Otolaryngologic Diseases, Chinese PLA General Hospital, 6 Fucheng Road, Beijing 100048, P. R. China
| | - Jin Li
- Senior Department of Otolaryngology-Head and Neck Surgery, the Sixth Medical Center of PLA General Hospital, Medical School of Chinese PLA, 6 Fucheng Road, Beijing 100048, P. R. China
- National Clinical Research Center for Otolaryngologic Diseases, Chinese PLA General Hospital, 6 Fucheng Road, Beijing 100048, P. R. China
| | - Hongyang Wang
- Senior Department of Otolaryngology-Head and Neck Surgery, the Sixth Medical Center of PLA General Hospital, Medical School of Chinese PLA, 6 Fucheng Road, Beijing 100048, P. R. China
- National Clinical Research Center for Otolaryngologic Diseases, Chinese PLA General Hospital, 6 Fucheng Road, Beijing 100048, P. R. China
| | - Qiuju Wang
- Senior Department of Otolaryngology-Head and Neck Surgery, the Sixth Medical Center of PLA General Hospital, Medical School of Chinese PLA, 6 Fucheng Road, Beijing 100048, P. R. China
- National Clinical Research Center for Otolaryngologic Diseases, Chinese PLA General Hospital, 6 Fucheng Road, Beijing 100048, P. R. China
| |
Collapse
|
45
|
Fagnani E, Cocomazzi P, Pellegrino S, Tedeschi G, Scalvini FG, Cossu F, Da Vela S, Aliverti A, Mastrangelo E, Milani M. CHCHD4 binding affects the active site of apoptosis inducing factor (AIF): Structural determinants for allosteric regulation. Structure 2024; 32:594-602.e4. [PMID: 38460521 DOI: 10.1016/j.str.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/08/2024] [Accepted: 02/13/2024] [Indexed: 03/11/2024]
Abstract
Apoptosis-inducing factor (AIF), which is confined to mitochondria of normal healthy cells, is the first identified caspase-independent cell death effector. Moreover, AIF is required for the optimal functioning of the respiratory chain machinery. Recent findings have revealed that AIF fulfills its pro-survival function by interacting with CHCHD4, a soluble mitochondrial protein which promotes the entrance and the oxidative folding of different proteins in the inner membrane space. Here, we report the crystal structure of the ternary complex involving the N-terminal 27-mer peptide of CHCHD4, NAD+, and AIF harboring its FAD (flavin adenine dinucleotide) prosthetic group in oxidized form. Combining this information with biophysical and biochemical data on the CHCHD4/AIF complex, we provide a detailed structural description of the interaction between the two proteins, validated by both chemical cross-linking mass spectrometry analysis and site-directed mutagenesis.
Collapse
Affiliation(s)
- Elisa Fagnani
- Biophysics Institute, CNR-IBF, Via Corti 12, 20133 Milan, Italy; Department of Bioscience, Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy
| | - Paolo Cocomazzi
- Biophysics Institute, CNR-IBF, Via Corti 12, 20133 Milan, Italy; Department of Bioscience, Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy
| | - Sara Pellegrino
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Via Golgi 19, 20133 Milan, Italy
| | - Gabriella Tedeschi
- Department of Veterinary Medicine and Animal Science (DIVAS), Università degli Studi di Milano, Via dell'Università 6, 26900 Lodi, Italy; Cimaina, Università degli Studi di Milano, Milan, Italy
| | - Francesca Grassi Scalvini
- Department of Veterinary Medicine and Animal Science (DIVAS), Università degli Studi di Milano, Via dell'Università 6, 26900 Lodi, Italy
| | - Federica Cossu
- Biophysics Institute, CNR-IBF, Via Corti 12, 20133 Milan, Italy; Department of Bioscience, Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy
| | - Stefano Da Vela
- Hochschule Bremerhaven, Karlstadt 8, 27568 Bremerhaven, Germany
| | - Alessandro Aliverti
- Department of Bioscience, Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy.
| | - Eloise Mastrangelo
- Biophysics Institute, CNR-IBF, Via Corti 12, 20133 Milan, Italy; Department of Bioscience, Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy.
| | - Mario Milani
- Biophysics Institute, CNR-IBF, Via Corti 12, 20133 Milan, Italy; Department of Bioscience, Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy.
| |
Collapse
|
46
|
Qian Z, Chen K, Yang L, Li C. Apoptosis-inducing factor 1 mediates Vibrio splendidus-induced coelomocyte apoptosis via importin β dependent nuclear translocation in Apostichopus japonicus. FISH & SHELLFISH IMMUNOLOGY 2024; 148:109491. [PMID: 38490346 DOI: 10.1016/j.fsi.2024.109491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 02/23/2024] [Accepted: 03/07/2024] [Indexed: 03/17/2024]
Abstract
As is well known, apoptosis is an important form of immune response and immune regulation, particularly playing a crucial role in combating microbial infections. Apoptosis-inducing factor 1 (AIF-1) is essential for apoptosis to induce chromatin condensation and DNA fragmentation via a caspase-independent pathway. The nuclear translocation of AIF-1 is a key step in apoptosis but the molecular mechanism is still unclear. In this study, the homologous gene of AIF-1, named AjAIF-1, was cloned and identified in Apostichopus japonicus. The mRNA expression of AjAIF-1 was significantly increased by 46.63-fold after Vibrio splendidus challenge. Silencing of AjAIF-1 was found to significantly inhibit coelomocyte apoptosis because the apoptosis rate of coelomocyte decreased by 0.62-fold lower compared with the control group. AjAIF-1 was able to promote coelomocyte apoptosis through nuclear translocation under the V. splendidus challenge. Moreover, AjAIF-1 and Ajimportin β were mainly co-localized around the nucleus in vivo and silencing Ajimportin β significantly inhibited the nuclear translocation of AjAIF-1 and suppressed coelomocyte apoptosis by 0.64-fold compared with control. In summary, nuclear translocation of AjAIF-1 will likely mediate coelomocyte apoptosis through an importin β-dependent pathway in sea cucumber.
Collapse
Affiliation(s)
- Zepeng Qian
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Kaiyu Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Lei Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China.
| | - Chenghua Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, PR China.
| |
Collapse
|
47
|
Wang Y, Feng H, Li X, Ruan Y, Guo Y, Cui X, Zhang P, Li Y, Wang X, Wang X, Wei L, Yi Y, Zhang L, Yang X, Liu H. Dampening of ISGylation of RIG-I by ADAP regulates type I interferon response of macrophages to RNA virus infection. PLoS Pathog 2024; 20:e1012230. [PMID: 38776321 PMCID: PMC11111093 DOI: 10.1371/journal.ppat.1012230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 04/29/2024] [Indexed: 05/24/2024] Open
Abstract
While macrophage is one of the major type I interferon (IFN-I) producers in multiple tissues during viral infections, it also serves as an important target cell for many RNA viruses. However, the regulatory mechanism for the IFN-I response of macrophages to respond to a viral challenge is not fully understood. Here we report ADAP, an immune adaptor protein, is indispensable for the induction of the IFN-I response of macrophages to RNA virus infections via an inhibition of the conjugation of ubiquitin-like ISG15 (ISGylation) to RIG-I. Loss of ADAP increases RNA virus replication in macrophages, accompanied with a decrease in LPS-induced IFN-β and ISG15 mRNA expression and an impairment in the RNA virus-induced phosphorylation of IRF3 and TBK1. Moreover, using Adap-/- mice, we show ADAP deficiency strongly increases the susceptibility of macrophages to RNA-virus infection in vivo. Mechanically, ADAP selectively interacts and functionally cooperates with RIG-I but not MDA5 in the activation of IFN-β transcription. Loss of ADAP results in an enhancement of ISGylation of RIG-I, whereas overexpression of ADAP exhibits the opposite effect in vitro, indicating ADAP is detrimental to the RNA virus-induced ISGylation of RIG-I. Together, our data demonstrate a novel antagonistic activity of ADAP in the cell-intrinsic control of RIG-I ISGylation, which is indispensable for initiating and sustaining the IFN-I response of macrophages to RNA virus infections and replication.
Collapse
Affiliation(s)
- Yan Wang
- Institutes of Biology and Medical Sciences (IBMS), Soochow University, Suzhou, Jiangsu Province, China
- Department of Biological Sciences, Xi’an Jiaotong-Liverpool University, Suzhou, Jiangsu Province, China
| | - Haixia Feng
- Institutes of Biology and Medical Sciences (IBMS), Soochow University, Suzhou, Jiangsu Province, China
| | - Xiao Li
- Institutes of Biology and Medical Sciences (IBMS), Soochow University, Suzhou, Jiangsu Province, China
| | - Yina Ruan
- Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Department of Veterinary Medicine, Zhejiang A&F University, Hangzhou, Zhejiang Province, China
| | - Yueping Guo
- Department of Veterinary Medicine, Zhejiang A&F University, Hangzhou, Zhejiang Province, China
| | - Xinxing Cui
- Institutes of Biology and Medical Sciences (IBMS), Soochow University, Suzhou, Jiangsu Province, China
| | - Pengchao Zhang
- Institutes of Biology and Medical Sciences (IBMS), Soochow University, Suzhou, Jiangsu Province, China
| | - Yanli Li
- Institutes of Biology and Medical Sciences (IBMS), Soochow University, Suzhou, Jiangsu Province, China
| | - Xinning Wang
- Institutes of Biology and Medical Sciences (IBMS), Soochow University, Suzhou, Jiangsu Province, China
| | - Xingran Wang
- Institutes of Biology and Medical Sciences (IBMS), Soochow University, Suzhou, Jiangsu Province, China
| | - Luxin Wei
- Department of General Surgery, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Yulan Yi
- Department of Biological Sciences, Xi’an Jiaotong-Liverpool University, Suzhou, Jiangsu Province, China
| | - Lifeng Zhang
- Department of General Surgery, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaodong Yang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Hebin Liu
- Institutes of Biology and Medical Sciences (IBMS), Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
48
|
Feng F, He S, Li X, He J, Luo L. Mitochondria-mediated Ferroptosis in Diseases Therapy: From Molecular Mechanisms to Implications. Aging Dis 2024; 15:714-738. [PMID: 37548939 PMCID: PMC10917537 DOI: 10.14336/ad.2023.0717] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/17/2023] [Indexed: 08/08/2023] Open
Abstract
Ferroptosis, a type of cell death involving iron and lipid peroxidation, has been found to be closely associated with the development of many diseases. Mitochondria are vital components of eukaryotic cells, serving important functions in energy production, cellular metabolism, and apoptosis regulation. Presently, the precise relationship between mitochondria and ferroptosis remains unclear. In this study, we aim to systematically elucidate the mechanisms via which mitochondria regulate ferroptosis from multiple perspectives to provide novel insights into mitochondrial functions in ferroptosis. Additionally, we present a comprehensive overview of how mitochondria contribute to ferroptosis in different conditions, including cancer, cardiovascular disease, inflammatory disease, mitochondrial DNA depletion syndrome, and novel coronavirus pneumonia. Gaining a comprehensive understanding of the involvement of mitochondria in ferroptosis could lead to more effective approaches for both basic cell biology studies and medical treatments.
Collapse
Affiliation(s)
- Fuhai Feng
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, China.
| | - Shasha He
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| | - Xiaoling Li
- Animal Experiment Center, Guangdong Medical University, Zhanjiang, China.
| | - Jiake He
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, China.
| | - Lianxiang Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong, China.
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong, China.
| |
Collapse
|
49
|
Kim S, Lee M, Kim NY, Kwon YS, Nam GS, Lee K, Kwon KM, Kim DK, Hwang IH. Oxidative tryptamine dimers from Corynebacterium durum directly target survivin to induce AIF-mediated apoptosis in cancer cells. Biomed Pharmacother 2024; 173:116335. [PMID: 38422661 DOI: 10.1016/j.biopha.2024.116335] [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: 01/03/2024] [Revised: 02/18/2024] [Accepted: 02/22/2024] [Indexed: 03/02/2024] Open
Abstract
Accumulating evidence indicates that microbial communities in the human body crucially affect health through the production of chemical messengers. However, the relationship between human microbiota and cancer has been underexplored. As a result of a biochemical investigation of the commensal oral microbe, Corynebacterium durum, we identified the non-enzymatic transformation of tryptamine into an anticancer compound, durumamide A (1). The structure of 1 was determined using LC-MS and NMR data analysis as bis(indolyl)glyoxylamide, which was confirmed using one-pot synthesis and X-ray crystallographic analysis, suggesting that 1 is an oxidative dimer of tryptamine. Compound 1 displayed cytotoxic activity against various cancer cell lines with IC50 values ranging from 25 to 35 μM. A drug affinity responsive target stability assay revealed that survivin is the direct target protein responsible for the anticancer effect of 1, which subsequently induces apoptosis-inducing factor (AIF)-mediated apoptosis. Inspired by the chemical structure and bioactivity of 1, a new derivative, durumamide B (2), was synthesized using another indole-based neurotransmitter, serotonin. The anticancer properties of 2 were similar to those of 1; however, it was less active. These findings reinforce the notion of human microbiota-host interplay by showing that 1 is naturally produced from the human microbial metabolite, tryptamine, which protects the host against cancer.
Collapse
Affiliation(s)
- Soyoung Kim
- Department of Pharmacology, School of Medicine, Dongguk University, Gyeongju, Gyeongsangbuk-do 38066, Republic of Korea
| | - Munseon Lee
- Department of Pharmacy, Woosuk University, Wanju, Jeonbuk 55338, Republic of Korea
| | - Nam-Yi Kim
- Department of Pharmacology, School of Medicine, Dongguk University, Gyeongju, Gyeongsangbuk-do 38066, Republic of Korea
| | - Yun-Suk Kwon
- Research Institute of Climate Change and Agriculture, National Institute of Horticultural and Herbal Science, Jeju, Jeju-do 63240, Republic of Korea
| | - Gi Suk Nam
- Department of Biomedical Laboratory Science, Honam University, 120, Honamdae-gil, Gwangsan-gu, Gwangju 62399, Republic of Korea
| | - Kyounghoon Lee
- Department of Chemical Education and Research Institute of Natural Sciences, Gyeongsang National University, Gyeongsangnam-do 52828, Republic of Korea
| | - Kang Mu Kwon
- Department of Pharmacy, Woosuk University, Wanju, Jeonbuk 55338, Republic of Korea
| | - Dae Keun Kim
- Department of Pharmacy, Woosuk University, Wanju, Jeonbuk 55338, Republic of Korea; Research Institute of Pharmaceutical Sciences, Woosuk University, Wanju 55338, Republic of Korea
| | - In Hyun Hwang
- Department of Pharmacy, Woosuk University, Wanju, Jeonbuk 55338, Republic of Korea; Research Institute of Pharmaceutical Sciences, Woosuk University, Wanju 55338, Republic of Korea.
| |
Collapse
|
50
|
Patel J, Dawson VL, Dawson TM. Blocking the Self-Destruct Program of Dopamine Neurons through Macrophage Migration Inhibitory Factor Nuclease Inhibition. Mov Disord 2024; 39:644-650. [PMID: 38396375 PMCID: PMC11160583 DOI: 10.1002/mds.29748] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/10/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative condition that pathognomonically involves the death of dopaminergic neurons in the substantia nigra pars compacta, resulting in a myriad of motor and non-motor symptoms. Given the insurmountable burden of this disease on the population and healthcare system, significant efforts have been put forth toward generating disease modifying therapies. This class of treatments characteristically alters disease course, as opposed to current strategies that focus on managing symptoms. Previous literature has implicated the cell death pathway known as parthanatos in PD progression. Inhibition of this pathway by targeting poly (ADP)-ribose polymerase 1 (PARP1) prevents neurodegeneration in a model of idiopathic PD. However, PARP1 has a vast repertoire of functions within the body, increasing the probability of side effects with the long-term treatment likely necessary for clinically significant neuroprotection. Recent work culminated in the development of a novel agent targeting the macrophage migration inhibitory factor (MIF) nuclease domain, also named parthanatos-associated apoptosis-inducing factor nuclease (PAAN). This nuclease activity specifically executes the terminal step in parthanatos. Parthanatos-associated apoptosis-inducing factor nuclease inhibitor-1 was neuroprotective in multiple preclinical mouse models of PD. This piece will focus on contextualizing this discovery, emphasizing its significance, and discussing its potential implications for parthanatos-directed treatment. © 2024 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Jaimin Patel
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Valina L. Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ted M. Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|