1
|
Sahu M, Jain U. Activation, interaction and intimation of Nrf2 pathway and their mutational studies causing Nrf2 associated cancer. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167764. [PMID: 40088576 DOI: 10.1016/j.bbadis.2025.167764] [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: 09/09/2024] [Revised: 02/15/2025] [Accepted: 02/25/2025] [Indexed: 03/17/2025]
Abstract
Responses against infection trigger several signaling pathways that lead to the production of cytokines, these cytokines release ROS and RNS, damaging DNA and proteins turn into various diseases including cancer. To combat these harmful cytokines, the Nrf2 pathway is activated. The gene NFE2L2 encodes Nrf2, which is divided into seven conserved domains (Neh1-7). The DLG and ETGE motifs, conserved sequences of amino acid in the Neh2 domain of Nrf2, bind to the BTB domain of Keap1. BTB domain promotes Keap1's homodimerization resulting in Cul3 recruitment providing scaffold formation to E2 ubiquitin ligase to form ubiquitin complex. Under normal conditions, this complex regularly degrades Nrf2. However, once the cell is exposed to oxidative stress by ROS interaction with Keap1 resulting in conformational changes that stabilize the Nrf2. Nrf2 further concentrates on the nucleus where it binds with the transcriptional factor to perform the desired genes transcription for synthesizing SOD, GSH, CAT, and various other proteins which reduce the ROS levels preventing certain diseases. To prevent cells from oxidative stress, molecular hydrogen activates the Nrf2 pathway. To activate the Nrf2 pathway, molecular hydrogen oxidizes the iron porphyrin which acts as an electrophile and interacts with Keap1's cysteine residue.
Collapse
Affiliation(s)
- Mridul Sahu
- School of Health Sciences and Technology (SoHST), UPES, Bidholi, Dehradun - 248007, India
| | - Utkarsh Jain
- School of Health Sciences and Technology (SoHST), UPES, Bidholi, Dehradun - 248007, India.
| |
Collapse
|
2
|
Jalali P, Shahmoradi A, Samii A, Mazloomnejad R, Hatamnejad MR, Saeed A, Namdar A, Salehi Z. The role of autophagy in cancer: from molecular mechanism to therapeutic window. Front Immunol 2025; 16:1528230. [PMID: 40248706 PMCID: PMC12003146 DOI: 10.3389/fimmu.2025.1528230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/12/2025] [Indexed: 04/19/2025] Open
Abstract
Autophagy is a cellular degradation process that plays a crucial role in maintaining metabolic homeostasis under conditions of stress or nutrient deprivation. This process involves sequestering, breaking down, and recycling intracellular components such as proteins, organelles, and cytoplasmic materials. Autophagy also serves as a mechanism for eliminating pathogens and engulfing apoptotic cells. In the absence of stress, baseline autophagy activity is essential for degrading damaged cellular components and recycling nutrients to maintain cellular vitality. The relationship between autophagy and cancer is well-established; however, the biphasic nature of autophagy, acting as either a tumor growth inhibitor or promoter, has raised concerns regarding the regulation of tumorigenesis without inadvertently activating harmful aspects of autophagy. Consequently, elucidating the mechanisms by which autophagy contributes to cancer pathogenesis and the factors determining its pro- or anti-tumor effects is vital for devising effective therapeutic strategies. Furthermore, precision medicine approaches that tailor interventions to individual patients may enhance the efficacy of autophagy-related cancer treatments. To this end, interventions aimed at modulating the fate of tumor cells by controlling or inducing autophagy substrates necessitate meticulous monitoring of these mediators' functions within the tumor microenvironment to make informed decisions regarding their activation or inactivation. This review provides an updated perspective on the roles of autophagy in cancer, and discusses the potential challenges associated with autophagy-related cancer treatment. The article also highlights currently available strategies and identifies questions that require further investigation in the future.
Collapse
Affiliation(s)
- Pooya Jalali
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arvin Shahmoradi
- Department of Laboratory Medicine, Faculty of Paramedical, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Amir Samii
- Department of Hematology and Blood Transfusion, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Radman Mazloomnejad
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Hatamnejad
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Anwaar Saeed
- Department of Medicine, Division of Hematology and Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Afshin Namdar
- Program in Cell Biology, The Hospital for Sick Children Peter Gilgan Centre for Research and Learning, Toronto, ON, United States
| | - Zahra Salehi
- Department of Hematology, Oncology and Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Liu X, Zheng Z, Xue C, Wang X, Li J, Liu Z, Xin W, Xu X, Zhou D, Yao L, Lu G. LRRK2 Mediates α-Synuclein-Induced Neuroinflammation and Ferroptosis through the p62-Keap1-Nrf2 Pathway in Parkinson's Disease. Inflammation 2025:10.1007/s10753-025-02291-8. [PMID: 40169487 DOI: 10.1007/s10753-025-02291-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 02/23/2025] [Accepted: 03/06/2025] [Indexed: 04/03/2025]
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder worldwide, characterized by the progressive degeneration of dopaminergic neurons in the substantia nigra pars compacta and the abnormal aggregation of α-synuclein (α-syn). Despite extensive research, the mechanisms underlying microglial-mediated neuroinflammation and ferroptosis in PD remain inadequately understood. In particular, the role of leucine-rich repeat kinase 2 (LRRK2) in microglial cells and its modulation of the p62-Keap1-Nrf2 signaling pathway warrant further investigation.In this study, we present novel findings demonstrating that LRRK2 regulates microglial neuroinflammation and ferroptosis through the p62-Keap1-Nrf2 signaling axis in the context of PD. Using α-syn-stimulated BV2 microglial cells, we found that LRRK2 inhibition significantly reduced the production of pro-inflammatory cytokines and enhanced the activation of the p62-Keap1-Nrf2 pathway, thereby mitigating ferroptosis and oxidative stress. Furthermore, conditioned medium from LRRK2-inhibited microglia conferred neuroprotective effects on cultured neurons, highlighting the therapeutic potential of targeting LRRK2 in microglia.Importantly, these in vitro findings were corroborated in the MPTP-induced PD mouse model, where LRRK2 inhibition led to diminished microglial activation, decreased apoptosis of midbrain dopaminergic neurons, and upregulation of the p62-Keap1-Nrf2 pathway.Our study fills a critical gap in understanding the microglial mechanisms mediated by LRRK2 and provides novel insights into the pathogenesis of PD. These findings suggest that targeting LRRK2 in microglia may represent a promising therapeutic strategy for PD.
Collapse
Affiliation(s)
- Xinjie Liu
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Zijian Zheng
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Cheng Xue
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xiangrong Wang
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Jianwei Li
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Zheng Liu
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Wenqiang Xin
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xinping Xu
- Jiangxi Institute of Respiratory Disease, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Dongwei Zhou
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Longping Yao
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
- Department of Neuroanatomy, Group for Regeneration and Reprogramming, Institute for Anatomy and Cell Biology, Medical Faculty, Heidelberg University, Heidelberg, Germany.
| | - Guohui Lu
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
4
|
Fang C, Qian J, Tu BZ, Xia X, Jia CY, Shen CL. MiR-124 Delivered by Extracellular Vesicles from Mesenchymal Stem Cell Exerts Neuroprotective Effects by Stabilizing the p62-Keap1-Nrf2 Pathway after Spinal Cord Injury in Rats. Mol Neurobiol 2025:10.1007/s12035-025-04755-2. [PMID: 39992585 DOI: 10.1007/s12035-025-04755-2] [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: 06/07/2024] [Accepted: 02/06/2025] [Indexed: 02/26/2025]
Abstract
Spinal cord injury (SCI) can cause irreversible trauma to nervous tissue, leading to permanent damage to the patient's motor and sensory functions. Extracellular vesicles derived from mesenchymal stem cells (MSC-EVs) can simulate most of the functions of MSCs and are considered an ideal treatment option for SCI. However, the potential mechanism of MSC-EVs treatment for SCI still needs to be explored. We cultured neurons in vitro to investigate the effect of miR-124 on the p62-Keap1-Nrf2 pathway. Besides, MSC-EVs containing miR-124 were injected into a rat spinal cord injury model to observe their neural repair effect. The accumulation of p62 can be reversed by miR-124, which promotes autophagy and alleviates oxidative stress, thereby exerting neuroprotective effects. Rats who received injection of MSC-EVs overexpressing miR-124 after surgery showed higher BBB scores, lower levels of cell apoptosis, and better spinal cord tissue morphology. Our results indicated that miR-124 can stabilize the p62-Keap1-Nrf2 loop, thereby promoting autophagy and alleviating oxidative stress to exert neuroprotective effects. Our research proposes a novel potential target for treating SCI.
Collapse
Affiliation(s)
- Chao Fang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui Province, China
| | - Jun Qian
- Department of Orthopedics and Spine Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui Province, China
| | - Bi-Zhi Tu
- Department of Orthopedics, The First People's Hospital of Hefei, No. 390 Huaihe Road, Hefei, Anhui Province, China
| | - Xiang Xia
- Department of Orthopedics and Spine Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui Province, China
| | - Chong-Yu Jia
- Department of Orthopedics and Spine Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui Province, China
| | - Cai-Liang Shen
- Department of Orthopedics and Spine Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui Province, China.
- Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, The First Affiliated Hospital of Anhui Medical University, Shushan District of Hefei, No. 218 Jixi Road, Anhui Province, China.
| |
Collapse
|
5
|
Kausar MA, Anwar S, Khan YS, Saleh AA, Ahmed MAA, Kaur S, Iqbal N, Siddiqui WA, Najm MZ. Autophagy and Cancer: Insights into Molecular Mechanisms and Therapeutic Approaches for Chronic Myeloid Leukemia. Biomolecules 2025; 15:215. [PMID: 40001518 PMCID: PMC11853340 DOI: 10.3390/biom15020215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Autophagy is a critical cellular process that maintains homeostasis by recycling damaged or aberrant components. This process is orchestrated by a network of proteins that form autophagosomes, which engulf and degrade intracellular material. In cancer, autophagy plays a dual role: it suppresses tumor initiation in the early stages but supports tumor growth and survival in advanced stages. Chronic myeloid leukemia (CML), a hematological malignancy, is characterized by the Philadelphia chromosome, a chromosomal abnormality resulting from a translocation between chromosomes 9 and 22. Autophagy has emerged as a key factor in CML pathogenesis, promoting cancer cell survival and contributing to resistance against tyrosine kinase inhibitors (TKIs), the primary treatment for CML. Targeting autophagic pathways is being actively explored as a therapeutic approach to overcome drug resistance and enhance cancer cell death. Recent research highlights the intricate interplay between autophagy and CML progression, underscoring its role in disease biology and treatment outcomes. This review aims to provide a comprehensive analysis of the molecular and cellular mechanisms underlying CML, with a focus on the therapeutic potential of targeting autophagy.
Collapse
MESH Headings
- Humans
- Autophagy/drug effects
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Protein Kinase Inhibitors/therapeutic use
- Protein Kinase Inhibitors/pharmacology
- Drug Resistance, Neoplasm/drug effects
- Animals
- Antineoplastic Agents/therapeutic use
- Antineoplastic Agents/pharmacology
Collapse
Affiliation(s)
- Mohd Adnan Kausar
- Department of Biochemistry, College of Medicine, University of Ha’il, Hail 55476, Saudi Arabia;
| | - Sadaf Anwar
- Department of Biochemistry, College of Medicine, University of Ha’il, Hail 55476, Saudi Arabia;
| | - Yusuf Saleem Khan
- Department of Anatomy, College of Medicine, University of Ha’il, Hail 55476, Saudi Arabia;
| | - Ayman A. Saleh
- Department of Pathology, College of Medicine, University of Ha’il, Hail 55476, Saudi Arabia;
| | | | - Simran Kaur
- School of Biosciences, Apeejay Stya University, Sohna, Gurugram 122103, Haryana, India;
| | - Naveed Iqbal
- Department of Obstetrics and Gynecology, College of Medicine, University of Ha’il, Ha’il 55476, Saudi Arabia;
| | - Waseem Ahmad Siddiqui
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202001, Uttar Pradesh, India;
| | - Mohammad Zeeshan Najm
- School of Biosciences, Apeejay Stya University, Sohna, Gurugram 122103, Haryana, India;
| |
Collapse
|
6
|
Liu M, Liu S, Lin Z, Chen X, Jiao Q, Du X, Jiang H. Targeting the Interplay Between Autophagy and the Nrf2 Pathway in Parkinson's Disease with Potential Therapeutic Implications. Biomolecules 2025; 15:149. [PMID: 39858542 PMCID: PMC11764135 DOI: 10.3390/biom15010149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/12/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative disorder marked by the progressive degeneration of midbrain dopaminergic neurons and resultant locomotor dysfunction. Despite over two centuries of recognition as a chronic disease, the exact pathogenesis of PD remains elusive. The onset and progression of PD involve multiple complex pathological processes, with dysfunctional autophagy and elevated oxidative stress serving as critical contributors. Notably, emerging research has underscored the interplay between autophagy and oxidative stress in PD pathogenesis. Given the limited efficacy of therapies targeting either autophagy dysfunction or oxidative stress, it is crucial to elucidate the intricate mechanisms governing their interplay in PD to develop more effective therapeutics. This review overviews the role of autophagy and nuclear factor erythroid 2-related factor 2 (Nrf2), a pivotal transcriptional regulator orchestrating cellular defense mechanisms against oxidative stress, and the complex interplay between these processes. By elucidating the intricate interplay between these key pathological processes in PD, this review will deepen our comprehensive understanding of the multifaceted pathological processes underlying PD and may uncover potential strategies for its prevention and treatment.
Collapse
Affiliation(s)
- Mengru Liu
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao 266000, China; (M.L.); (S.L.)
| | - Siqi Liu
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao 266000, China; (M.L.); (S.L.)
| | - Zihan Lin
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao 266000, China; (M.L.); (S.L.)
| | - Xi Chen
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao 266000, China; (M.L.); (S.L.)
| | - Qian Jiao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao 266000, China; (M.L.); (S.L.)
| | - Xixun Du
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao 266000, China; (M.L.); (S.L.)
| | - Hong Jiang
- Qingdao Key Laboratory of Neurorehabilitation, Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao 266113, China
| |
Collapse
|
7
|
Chen Y, Li S, Hou X, Jia Y. PDE4B abrogation extenuates angiotensin II-induced endothelial dysfunction related to hypertension through up-regulation of AMPK/Sirt1/Nrf2/ARE signaling. Tissue Cell 2024; 91:102637. [PMID: 39591723 DOI: 10.1016/j.tice.2024.102637] [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: 04/11/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 11/28/2024]
Abstract
Endothelial dysfunction is commonly perceived as a precursor in the process of hypertension, a severe cardiovascular disorder. Phosphodiesterase 4B (PDE4B) inactivation has been proposed to exert cardioprotective effects and prevent pulmonary hypertension. However, the role of PDE4B in endothelial dysfunction in hypertension remains inexplicit, which will be investigated in the present work. In angiotensin II (Ang II)-induced human umbilical vein endothelial cells (HUVECs), RT-qPCR and Western blotting were used to analyze PDE4B expression. CCK-8 method was used to detect cell viability. Flow cytometry assay and Caspase 3 assay kit were used to detect cellular apoptotic level. Wound healing and tube formation assays were respectively used to detect cell migration and angiogenesis. Western blotting and corresponding assay kits were respectively used to analyze the expressions and contents of endothelial dysfunction markers. JC-1 assay, RT-qPCR and relevant assay kit were respectively used to detect mitochondrial membrane potential (ΔΨm), quantify mitochondrial DNA (mtDNA) copy number and mitochondrial permeability transition pore (mPTP) opening. Besides, Western blotting was used to analyze the expressions of endoplasmic reticulum stress (ERS) and AMP-activated protein kinase (AMPK)/sirtuin 1 (Sirt1)/nuclear factor-erythroid 2 related factor 2 (Nrf2)/antioxidant response element (ARE) signaling-associated proteins. PDE4B expression was increased in Ang II- induced HUVECs. PDE4B knockdown promoted the viability, migration, angiogenesis while inhibiting the apoptosis, endothelial dysfunction, ERS and mitochondrial damage in Ang II-induced HUVECs. Additionally, PDE4B silence activated AMPK/Sirt1/Nrf2/ARE pathway and AMPK inhibitor Compound C (CC) partially reversed the effects of PDE4B down-regulation on Ang II-induced HUVECs. Conclusively, PDE4B inhibition might protect against Ang II-induced endothelial dysfunction in HUVECs via up-regulating AMPK/Sirt1/Nrf2/ARE pathway, which might be mediated by the suppression of ERS and mitochondrial damage.
Collapse
Affiliation(s)
- Yong Chen
- Cardiovascular Department, Yueqing Second People's Hospital, Hongqiao Town, Yueqing City, Zhejiang Province 325608, China.
| | - Suipeng Li
- Cardiovascular Department, Yueqing Second People's Hospital, Hongqiao Town, Yueqing City, Zhejiang Province 325608, China
| | - Xuqing Hou
- Cardiovascular Department, Yueqing Second People's Hospital, Hongqiao Town, Yueqing City, Zhejiang Province 325608, China
| | - Yinfeng Jia
- Cardiovascular Department, Yueqing Second People's Hospital, Hongqiao Town, Yueqing City, Zhejiang Province 325608, China
| |
Collapse
|
8
|
Fan Q, Xiao K, A R, Gao LP, Wu YZ, Chen DD, Hu C, Jia XX, Liu CM, Liu X, Chen C, Shi Q, Dong XP. Accumulation of Prion Triggers the Enhanced Glycolysis via Activation of AMKP Pathway in Prion-Infected Rodent and Cell Models. Mol Neurobiol 2024; 61:9810-9834. [PMID: 37726499 DOI: 10.1007/s12035-023-03621-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 08/28/2023] [Indexed: 09/21/2023]
Abstract
Mitochondrial dysfunction is one of the hallmarks in the pathophysiology of prion disease and other neurodegenerative diseases. Various metabolic dysfunctions are identified and considered to contribute to the progression of some types of neurodegenerative diseases. In this study, we evaluated the status of glycolysis pathway in prion-infected rodent and cell models. The levels of the key enzymes, hexokinase (HK), phosphofructokinase (PFK), and pyruvate kinase (PK) were significantly increased, accompanying with markedly downregulated mitochondrial complexes. Double-stained IFAs revealed that the increased HK2 and PFK distributed widely in GFAP-, Iba1-, and NeuN-positive cells. We also identified increased levels of AMP-activated protein kinase (AMPK) and the downstream signaling. Changes of AMPK activity in prion-infected cells by the AMPK-specific inhibitor or activator induced the corresponding alterations not only in the downstream signaling, but also the expressions of three key kinases in glycolysis pathway and the mitochondrial complexes. Transient removal or complete clearance of prion propagation in the prion-infected cells partially but significantly reversed the increases of the key enzymes in glycolysis, the upregulation of AMPK signaling pathway, and the decreases of the mitochondrial complexes. Measurements of the cellular oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) showed lower OCR and higher ECAR in prion-infected cell line, which were sufficiently reversed by clearance of prion propagation. Those data indicate a metabolic reprogramming from oxidative phosphorylation to glycolysis in the brains during the progression of prion disease. Accumulation of PrPSc is critical for the switch to glycolysis, largely via activating AMPK pathway.
Collapse
Affiliation(s)
- Qin Fan
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Kang Xiao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ruhan A
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Li-Ping Gao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yue-Zhang Wu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Dong-Dong Chen
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Chao Hu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiao-Xi Jia
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Chu-Mou Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xin Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Cao Chen
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Qi Shi
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.
- China Academy of Chinese Medical Sciences, Beijing, China.
| | - Xiao-Ping Dong
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.
- China Academy of Chinese Medical Sciences, Beijing, China.
- Shanghai Institute of Infectious Disease and Biosafety, Shanghai, China.
| |
Collapse
|
9
|
Fournier LA, Kalantari F, Wells JP, Lee JS, Trigo-Gonzalez G, Moksa MM, Smith T, White J, Shanks A, Wang SL, Su E, Wang Y, Huntsman DG, Hirst M, Stirling PC. Genome-Wide CRISPR Screen Identifies KEAP1 Perturbation as a Vulnerability of ARID1A-Deficient Cells. Cancers (Basel) 2024; 16:2949. [PMID: 39272807 PMCID: PMC11394604 DOI: 10.3390/cancers16172949] [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: 07/22/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/15/2024] Open
Abstract
ARID1A is the core DNA-binding subunit of the BAF chromatin remodeling complex and is mutated in about 8% of all cancers. The frequency of ARID1A loss varies between cancer subtypes, with clear cell ovarian carcinoma (CCOC) presenting the highest incidence at > 50% of cases. Despite a growing understanding of the consequences of ARID1A loss in cancer, there remains limited targeted therapeutic options for ARID1A-deficient cancers. Using a genome-wide CRISPR screening approach, we identify KEAP1 as a genetic dependency of ARID1A in CCOC. Depletion or chemical perturbation of KEAP1 results in selective growth inhibition of ARID1A-KO cell lines and edited primary endometrial epithelial cells. While we confirm that KEAP1-NRF2 signalling is dysregulated in ARID1A-KO cells, we suggest that this synthetic lethality is not due to aberrant NRF2 signalling. Rather, we find that KEAP1 perturbation exacerbates genome instability phenotypes associated with ARID1A deficiency. Together, our findings identify a potentially novel synthetic lethal interaction of ARID1A-deficient cells.
Collapse
Affiliation(s)
- Louis-Alexandre Fournier
- Terry Fox Laboratory, BC Cancer, Vancouver, BC V5L1Z3, Canada
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC V5L1Z3, Canada
| | - Forouh Kalantari
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T1Z4, Canada
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, BC V5L1Z3, Canada
| | - James P Wells
- Terry Fox Laboratory, BC Cancer, Vancouver, BC V5L1Z3, Canada
| | - Joon Seon Lee
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T1Z4, Canada
| | - Genny Trigo-Gonzalez
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, BC V5L1Z3, Canada
| | - Michelle M Moksa
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T1Z4, Canada
| | - Theodore Smith
- Terry Fox Laboratory, BC Cancer, Vancouver, BC V5L1Z3, Canada
| | - Justin White
- Terry Fox Laboratory, BC Cancer, Vancouver, BC V5L1Z3, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T1Z4, Canada
| | - Alynn Shanks
- Terry Fox Laboratory, BC Cancer, Vancouver, BC V5L1Z3, Canada
| | - Siyun L Wang
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T1Z4, Canada
| | - Edmund Su
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T1Z4, Canada
| | - Yemin Wang
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T1Z4, Canada
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, BC V5L1Z3, Canada
| | - David G Huntsman
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T1Z4, Canada
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, BC V5L1Z3, Canada
- Department of Obstetrics and Gynaecology, University of British Columbia, Vancouver, BC V6T1Z4, Canada
| | - Martin Hirst
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T1Z4, Canada
| | - Peter C Stirling
- Terry Fox Laboratory, BC Cancer, Vancouver, BC V5L1Z3, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T1Z4, Canada
| |
Collapse
|
10
|
Christoudia N, Bekas N, Kanata E, Chatziefsthathiou A, Pettas S, Karagianni K, Da Silva Correia SM, Schmitz M, Zerr I, Tsamesidis I, Xanthopoulos K, Dafou D, Sklaviadis T. Αnti-prion effects of anthocyanins. Redox Biol 2024; 72:103133. [PMID: 38565068 PMCID: PMC10990977 DOI: 10.1016/j.redox.2024.103133] [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: 02/26/2024] [Revised: 03/22/2024] [Accepted: 03/24/2024] [Indexed: 04/04/2024] Open
Abstract
Prion diseases, also known as Transmissible Spongiform Encephalopathies (TSEs), are protein-based neurodegenerative disorders (NDs) affecting humans and animals. They are characterized by the conformational conversion of the normal cellular prion protein, PrPC, into the pathogenic isoform, PrPSc. Prion diseases are invariably fatal and despite ongoing research, no effective prophylactic or therapeutic avenues are currently available. Anthocyanins (ACNs) are unique flavonoid compounds and interest in their use as potential neuroprotective and/or therapeutic agents against NDs, has increased significantly in recent years. Therefore, we investigated the potential anti-oxidant and anti-prion effects of Oenin and Myrtillin, two of the most common anthocyanins, using the most accepted in the field overexpressing PrPScin vitro model and a cell free protein aggregation model. Our results, indicate both anthocyanins as strong anti-oxidant compounds, upregulating the expression of genes involved in the anti-oxidant response, and reducing the levels of Reactive Oxygen Species (ROS), produced due to pathogenic prion infection, through the activation of the Keap1-Nrf2 pathway. Importantly, they showcased remarkable anti-prion potential, as they not only caused the clearance of pathogenic PrPSc aggregates, but also completely inhibited the formation of PrPSc fibrils in the Cerebrospinal Fluid (CSF) of patients with Creutzfeldt-Jakob disease (CJD). Therefore, Oenin and Myrtillin possess pleiotropic effects, suggesting their potential use as promising preventive and/or therapeutic agents in prion diseases and possibly in the spectrum of neurodegenerative proteinopathies.
Collapse
Affiliation(s)
- Nikoletta Christoudia
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, 541 24, Thessaloniki, Greece.
| | - Nikolaos Bekas
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, 541 24, Thessaloniki, Greece.
| | - Eirini Kanata
- Neurodegenerative Diseases Research Group, Department of Pharmacy, School of Health Sciences, Aristotle University of Thessaloniki, 541 24, Thessaloniki, Greece.
| | - Athanasia Chatziefsthathiou
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, 541 24, Thessaloniki, Greece.
| | - Spyros Pettas
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, 541 24, Thessaloniki, Greece; Neurodegenerative Diseases Research Group, Department of Pharmacy, School of Health Sciences, Aristotle University of Thessaloniki, 541 24, Thessaloniki, Greece.
| | - Korina Karagianni
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, 541 24, Thessaloniki, Greece.
| | - Susana Margarida Da Silva Correia
- Department of Neurology, German Center for Neurodegenerative Diseases (DZNE), University Medicine Goettingen, 37075, Goettingen, Germany
| | - Matthias Schmitz
- Department of Neurology, German Center for Neurodegenerative Diseases (DZNE), University Medicine Goettingen, 37075, Goettingen, Germany.
| | - Inga Zerr
- Department of Neurology, German Center for Neurodegenerative Diseases (DZNE), University Medicine Goettingen, 37075, Goettingen, Germany.
| | - Ioannis Tsamesidis
- Department of Prosthodontics, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24, Thessaloniki, Greece.
| | - Konstantinos Xanthopoulos
- Neurodegenerative Diseases Research Group, Department of Pharmacy, School of Health Sciences, Aristotle University of Thessaloniki, 541 24, Thessaloniki, Greece.
| | - Dimitra Dafou
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, 541 24, Thessaloniki, Greece.
| | - Theodoros Sklaviadis
- Neurodegenerative Diseases Research Group, Department of Pharmacy, School of Health Sciences, Aristotle University of Thessaloniki, 541 24, Thessaloniki, Greece.
| |
Collapse
|
11
|
Giri RK. Molecular signatures in prion disease: altered death receptor pathways in a mouse model. J Transl Med 2024; 22:503. [PMID: 38802941 PMCID: PMC11129387 DOI: 10.1186/s12967-024-05121-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: 12/02/2023] [Accepted: 03/20/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Prion diseases are transmissible and fatal neurodegenerative diseases characterized by accumulation of misfolded prion protein isoform (PrPSc), astrocytosis, microgliosis, spongiosis, and neurodegeneration. Elevated levels of cell membrane associated PrPSc protein and inflammatory cytokines hint towards the activation of death receptor (DR) pathway/s in prion diseases. Activation of DRs regulate, either cell survival or apoptosis, autophagy and necroptosis based on the adaptors they interact. Very little is known about the DR pathways activation in prion disease. DR3 and DR5 that are expressed in normal mouse brain were never studied in prion disease, so also their ligands and any DR adaptors. This research gap is notable and investigated in the present study. METHODS C57BL/6J mice were infected with Rocky Mountain Laboratory scrapie mouse prion strain. The progression of prion disease was examined by observing morphological and behavioural abnormalities. The levels of PrP isoforms and GFAP were measured as the marker of PrPSc accumulation and astrocytosis respectively using antibody-based techniques that detect proteins on blot and brain section. The levels of DRs, their glycosylation and ectodomain shedding, and associated factors warrant their examination at protein level, hence western blot analysis was employed in this study. RESULTS Prion-infected mice developed motor deficits and neuropathology like PrPSc accumulation and astrocytosis similar to other prion diseases. Results from this research show higher expression of all DR ligands, TNFR1, Fas and p75NTR but decreased levels DR3 and DR5. The levels of DR adaptor proteins like TRADD and TRAF2 (primarily regulate pro-survival pathways) are reduced. FADD, which primarily regulate cell death, its level remains unchanged. RIPK1, which regulate pro-survival, apoptosis and necroptosis, its expression and proteolysis (inhibits necroptosis but activates apoptosis) are increased. CONCLUSIONS The findings from the present study provide evidence towards the involvement of DR3, DR5, DR6, TL1A, TRAIL, TRADD, TRAF2, FADD and RIPK1 for the first time in prion diseases. The knowledge obtained from this research discuss the possible impacts of these 16 differentially expressed DR factors on our understanding towards the multifaceted neuropathology of prion diseases and towards future explorations into potential targeted therapeutic interventions for prion disease specific neuropathology.
Collapse
Affiliation(s)
- Ranjit Kumar Giri
- Molecular and Cellular Neuroscience Division, National Brain Research Centre, Manesar, Gurgaon, Haryana, 122052, India.
| |
Collapse
|
12
|
Li C, Liu Y. Puerarin reduces cell damage from cerebral ischemia-reperfusion by inhibiting ferroptosis. Biochem Biophys Res Commun 2024; 693:149324. [PMID: 38101001 DOI: 10.1016/j.bbrc.2023.149324] [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: 11/17/2023] [Accepted: 11/22/2023] [Indexed: 12/17/2023]
Abstract
This study explores the protective effects of Puerarin, a compound derived from the traditional Chinese herb Pueraria, against cellular damage induced by Oxygen-Glucose Deprivation/Reoxygenation (OGD/R) in PC12 cells. The research focuses on understanding how Puerarin influences the mechanisms of ferroptosis and oxidative stress, key factors in ischemia-reperfusion injury relevant to neurodegenerative diseases. In our in vitro model, we identified the optimal OGD duration to induce significant cell stress and confirmed the non-toxicity of Puerarin up to 100uM. The results reveal that Puerarin substantially mitigates the detrimental effects of OGD/R, including improvements in cell viability, mitochondrial integrity, and reductions in oxidative stress markers like ROS and lipid peroxidation. Notably, Puerarin modulates key proteins in the autophagy process and the Nrf2 pathway, crucial in cellular stress responses. Further, the use of 3-Methyladenine, an autophagy inhibitor, underscores the significance of autophagy in managing OGD/R-induced stress. These findings suggest Puerarin's potential as a therapeutic agent for conditions characterized by ischemic cellular damage, highlighting the need for further clinical exploration.
Collapse
Affiliation(s)
- Changxuan Li
- Department of Neurosurgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, Hunan, China
| | - Yu Liu
- Department of Neurosurgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, Hunan, China.
| |
Collapse
|
13
|
Bhushan B, Singh NK. Role of Astrogliosis in the Pathogenesis of Parkinson's Disease: Insights into Astrocytic Nrf2 Pathway as a Potential Therapeutic Target. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:1015-1029. [PMID: 37817521 DOI: 10.2174/0118715273270473231002104610] [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: 07/04/2023] [Revised: 08/21/2023] [Accepted: 08/31/2023] [Indexed: 10/12/2023]
Abstract
Recently, Parkinson's disease (PD) has become a remarkable burden on families and society with an acceleration of population aging having several pathological hallmarks such as dopaminergic neuronal loss of the substantia nigra pars compacta, α-synucleinopathy, neuroinflammation, autophagy, last but not the least astrogliosis. Astrocyte, star-shaped glial cells perform notable physiological functions in the brain through several molecular and cellular mechanisms including nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway. It has been well established that the downregulation of the astrocytic Nrf2 signaling pathway plays a crucial role in the pathogenesis of PD because it is a master regulator of cellular defense mechanism along with a regulator of numerous detoxifying and antioxidant enzymes gene expression. Fascinatingly, upregulation of the astrocytic Nrf2 signaling pathway attenuates the degeneration of nigrostriatal neurons, restores neuronal proliferation, rejuvenates astrocytic functions, and exhibits neuroprotective effects via numerous cellular and molecular mechanisms in the PD-like brain of the experimental animal. Here, we discuss the numerous in-vitro and in-vivo studies that evaluate the neuroprotective potential of the astrocytic Nrf2 signaling pathway against experimentally-induced PD-like manifestation. In conclusion, based on available preclinical reports, it can be assumed that the astrocytic Nrf2 signaling pathway could be an alternative target in the drug discovery process for the prevention, management, and treatment of PD.
Collapse
Affiliation(s)
- Bharat Bhushan
- Division of Pharmacology, Institute of Pharmaceutical Research, GLA University, NH-19, Mathura-Delhi Road, Chaumuhan, Mathura 281406, U.P. India
| | - Niraj Kumar Singh
- Division of Pharmacology, Institute of Pharmaceutical Research, GLA University, NH-19, Mathura-Delhi Road, Chaumuhan, Mathura 281406, U.P. India
| |
Collapse
|
14
|
Ates I, Yılmaz AD, Buttari B, Arese M, Saso L, Suzen S. A Review of the Potential of Nuclear Factor [Erythroid-Derived 2]-like 2 Activation in Autoimmune Diseases. Brain Sci 2023; 13:1532. [PMID: 38002492 PMCID: PMC10669303 DOI: 10.3390/brainsci13111532] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 10/07/2023] [Accepted: 10/11/2023] [Indexed: 11/26/2023] Open
Abstract
An autoimmune disease is the consequence of the immune system attacking healthy cells, tissues, and organs by mistake instead of protecting them. Inflammation and oxidative stress (OS) are well-recognized processes occurring in association with acute or chronic impairment of cell homeostasis. The transcription factor Nrf2 (nuclear factor [erythroid-derived 2]-like 2) is of major importance as the defense instrument against OS and alters anti-inflammatory activities related to different pathological states. Researchers have described Nrf2 as a significant regulator of innate immunity. Growing indications suggest that the Nrf2 signaling pathway is deregulated in numerous diseases, including autoimmune disorders. The advantageous outcome of the pharmacological activation of Nrf2 is an essential part of Nrf2-based chemoprevention and intervention in other chronic illnesses, such as neurodegeneration, cardiovascular disease, autoimmune diseases, and chronic kidney and liver disease. Nevertheless, a growing number of investigations have indicated that Nrf2 is already elevated in specific cancer and disease steps, suggesting that the pharmacological agents developed to mitigate the potentially destructive or transformative results associated with the protracted activation of Nrf2 should also be evaluated. The activators of Nrf2 have revealed an improvement in the progress of OS-associated diseases, resulting in immunoregulatory and anti-inflammatory activities; by contrast, the depletion of Nrf2 worsens disease progression. These data strengthen the growing attention to the biological properties of Nrf2 and its possible healing power on diseases. The evidence supporting a correlation between Nrf2 signaling and the most common autoimmune diseases is reviewed here. We focus on the aspects related to the possible effect of Nrf2 activation in ameliorating pathologic conditions based on the role of this regulator of antioxidant genes in the control of inflammation and OS, which are processes related to the progression of autoimmune diseases. Finally, the possibility of Nrf2 activation as a new drug development strategy to target pathogenesis is proposed.
Collapse
Affiliation(s)
- Ilker Ates
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Ankara University, Degol Str. No. 4, 06560 Ankara, Turkey
| | - Ayşe Didem Yılmaz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ankara University, Degol Str. No. 4, 06560 Ankara, Turkey; (A.D.Y.); (S.S.)
| | - Brigitta Buttari
- Department of Cardiovascular and Endocrine-Metabolic Diseases and Aging, Italian National Institute of Health, 00161 Rome, Italy;
| | - Marzia Arese
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of Rome, Piazzae Aldo Moro 5, 00185 Rome, Italy;
| | - Luciano Saso
- Department of Physiology and Pharmacology ‘‘Vittorio Erspamer”, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy;
| | - Sibel Suzen
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ankara University, Degol Str. No. 4, 06560 Ankara, Turkey; (A.D.Y.); (S.S.)
| |
Collapse
|
15
|
Al-Harbi NO, Imam F, Al-Harbi MM, Qamar W, Aljerian K, Khalid Anwer M, Alharbi M, Almudimeegh S, Alhamed AS, Alshamrani AA. Effect of Apremilast on LPS-induced immunomodulation and inflammation via activation of Nrf2/HO-1 pathways in rat lungs. Saudi Pharm J 2023; 31:1327-1338. [PMID: 37323920 PMCID: PMC10267521 DOI: 10.1016/j.jsps.2023.05.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/23/2023] [Indexed: 06/17/2023] Open
Abstract
Lipopolysaccharides (LPS), the lipid component of gram-negative bacterial cell wall, is recognized as the key factor in acute lung inflammation and is found to exhibit severe immunologic reactions. Phosphodiesterase-4 (PDE-4) inhibitor: "apremilast (AP)" is an immune suppressant and anti-inflammatory drug which introduced to treat psoriatic arthritis. The contemporary experiment designed to study the protective influences of AP against LPS induced lung injury in rodents. Twenty-four (24) male experimental Wistar rats selected, acclimatized, and administered with normal saline, LPS, or AP + LPS respectively from 1 to 4 groups. The lung tissues were evaluated for biochemical parameters (MPO), Enzyme Linked Immunosorbent Assay (ELISA), flowcytometry assay, gene expressions, proteins expression and histopathological examination. AP ameliorates the lung injuries by attenuating immunomodulation and inflammation. LPS exposure upregulated IL-6, TNF-α, and MPO while downregulating IL-4 which were restored in AP pretreated rats. The changes in immunomodulation markers by LPS were reduced by AP treatment. Furthermore, results from the qPCR analysis represented an upregulation in IL-1β, MPO, TNF-α, and p38 whereas downregulated in IL-10 and p53 gene expressions in disease control animals while AP pretreated rats exhibited significant reversal in these expressions. Western blot analysis suggested an upregulation of MCP-1, and NOS-2, whereas HO-1, and Nrf-2 expression were suppressed in LPS exposed animals, while pretreatment with AP showed down regulation in the expression MCP-1, NOS-2, and upregulation of HO-1, and Nrf-2 expression of the mentioned intracellular proteins. Histological studies further affirmed the toxic influences of LPS on the pulmonary tissues. It is concluded that, LPS exposure causes pulmonary toxicities via up regulation of oxidative stress, inflammatory cytokines and stimulation of IL-1β, MPO, TNF-α, p38, MCP-1, and NOS-2 while downregulation of IL-4, IL-10, p53, HO-1, and Nrf-2 at different expression level. Pretreatment with AP controlled the toxic influences of LPS by modulating these signaling pathways.
Collapse
Affiliation(s)
- Naif O. Al-Harbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Faisal Imam
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Mohammad Matar Al-Harbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Wajhul Qamar
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Khaldoon Aljerian
- Department of Pathology, College of Medicine, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Md. Khalid Anwer
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Mohammed Alharbi
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Sultan Almudimeegh
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Abdullah S. Alhamed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Ali A Alshamrani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| |
Collapse
|
16
|
Huang M, Yan Y, Deng Z, Zhou L, She M, Yang Y, Zhang M, Wang D. Saikosaponin A and D attenuate skeletal muscle atrophy in chronic kidney disease by reducing oxidative stress through activation of PI3K/AKT/Nrf2 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 114:154766. [PMID: 37002971 DOI: 10.1016/j.phymed.2023.154766] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 02/22/2023] [Accepted: 03/12/2023] [Indexed: 06/19/2023]
Abstract
BACKGROUND Skeletal muscle atrophy in chronic kidney disease (CKD) leads to a decline in quality of life and increased risk of morbidity and mortality. We have obtained evidence that oxidative stress is essential in the progression of CKD-related muscle atrophy. Whether Saikosaponin A and D, two emerging antioxidants extracted from Bupleurum chinense DC, alleviate muscle atrophy remains to be further studied. The purpose of this study was to investigate the effects and mechanisms of these two components on CKD complicated with muscle atrophy. METHODS In this research, muscle dystrophy model was established using 5/6 nephrectomized mice in vivo and in vitro with Dexamethasone (Dex)-managed C2C12 myotubes. RESULTS The results of RNA-sequencing showed that exposure to Dex affected the antioxidant activity, catalytic activity and enzyme regulator activity of C2C12 cells. According to KEGG analysis, the largest numbers of differentially expressed genes detected were enriched in the PI3K/AKT pathway. In vivo, Saikosaponin A and D remain renal function, cross-section size, fiber-type composition and anti-inflammatory ability. These two components suppressed the expression of MuRF-1 and enhanced the expression of MyoD and Dystrophin. In addition, Saikosaponin A and D maintained redox balance by increasing the activities of antioxidant enzymes while inhibiting the excessive accumulation of reactive oxygen species. Furthermore, Saikosaponin A and D stimulated PI3K/AKT and its downstream Nrf2 pathway in CKD mice. The effects of Saikosaponin A and D on increasing the inner diameter of C2C12 myotube, reducing oxidative stress and enhancing expression of p-AKT, p-mTOR, p70S6K, Nrf2 and HO-1 proteins were observed in vitro. Importantly, we verified that these protective effects could be significantly reversed by inhibiting PI3K and knocking out Nrf2. CONCLUSIONS In summary, Saikosaponin A and D improve CKD-induced muscle atrophy by reducing oxidative stress through the PI3K/AKT/Nrf2 pathway.
Collapse
Affiliation(s)
- Minna Huang
- Department of Traditional Chinese Medicine, Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510000, China
| | - Yan Yan
- Department of Traditional Chinese Medicine, Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510000, China
| | - Zihao Deng
- The First Clinical Medical College, Southern Medical University, Guangzhou, 510000, China
| | - Lingli Zhou
- The First Clinical Medical College, Southern Medical University, Guangzhou, 510000, China
| | - Meiling She
- Department of Traditional Chinese Medicine, Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510000, China
| | - Yajun Yang
- Department of Pharmacology, Guangdong Key Laboratory for R&D of Natural Drug, Guangdong Medical University, Zhanjiang,524000, China
| | - Meng Zhang
- Department of Traditional Chinese Medicine, Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510000, China
| | - Dongtao Wang
- Department of Traditional Chinese Medicine, Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510000, China.
| |
Collapse
|
17
|
Santarelli S, Londero C, Soldano A, Candelaresi C, Todeschini L, Vernizzi L, Bellosta P. Drosophila melanogaster as a model to study autophagy in neurodegenerative diseases induced by proteinopathies. Front Neurosci 2023; 17:1082047. [PMID: 37274187 PMCID: PMC10232775 DOI: 10.3389/fnins.2023.1082047] [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: 10/27/2022] [Accepted: 04/14/2023] [Indexed: 06/06/2023] Open
Abstract
Proteinopathies are a large group of neurodegenerative diseases caused by both genetic and sporadic mutations in particular genes which can lead to alterations of the protein structure and to the formation of aggregates, especially toxic for neurons. Autophagy is a key mechanism for clearing those aggregates and its function has been strongly associated with the ubiquitin-proteasome system (UPS), hence mutations in both pathways have been associated with the onset of neurodegenerative diseases, particularly those induced by protein misfolding and accumulation of aggregates. Many crucial discoveries regarding the molecular and cellular events underlying the role of autophagy in these diseases have come from studies using Drosophila models. Indeed, despite the physiological and morphological differences between the fly and the human brain, most of the biochemical and molecular aspects regulating protein homeostasis, including autophagy, are conserved between the two species.In this review, we will provide an overview of the most common neurodegenerative proteinopathies, which include PolyQ diseases (Huntington's disease, Spinocerebellar ataxia 1, 2, and 3), Amyotrophic Lateral Sclerosis (C9orf72, SOD1, TDP-43, FUS), Alzheimer's disease (APP, Tau) Parkinson's disease (a-syn, parkin and PINK1, LRRK2) and prion diseases, highlighting the studies using Drosophila that have contributed to understanding the conserved mechanisms and elucidating the role of autophagy in these diseases.
Collapse
Affiliation(s)
- Stefania Santarelli
- Department of Cellular, Computational and Integrative Biology (CiBiO), University of Trento, Trento, Italy
| | - Chiara Londero
- Department of Cellular, Computational and Integrative Biology (CiBiO), University of Trento, Trento, Italy
| | - Alessia Soldano
- Department of Cellular, Computational and Integrative Biology (CiBiO), University of Trento, Trento, Italy
- Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy
| | - Carlotta Candelaresi
- Department of Cellular, Computational and Integrative Biology (CiBiO), University of Trento, Trento, Italy
| | - Leonardo Todeschini
- Department of Cellular, Computational and Integrative Biology (CiBiO), University of Trento, Trento, Italy
| | - Luisa Vernizzi
- Institute of Molecular Life Sciences, University of Zurich, Zürich, Switzerland
| | - Paola Bellosta
- Department of Cellular, Computational and Integrative Biology (CiBiO), University of Trento, Trento, Italy
- Department of Medicine, NYU Langone Medical Center, New York, NY, United States
| |
Collapse
|
18
|
Kim S, Choi J, Kwon J. Thymosin Beta 4 Protects Hippocampal Neuronal Cells against PrP (106-126) via Neurotrophic Factor Signaling. Molecules 2023; 28:molecules28093920. [PMID: 37175330 PMCID: PMC10180446 DOI: 10.3390/molecules28093920] [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: 03/15/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Prion protein peptide (PrP) has demonstrated neurotoxicity in brain cells, resulting in the progression of prion diseases with spongiform degenerative, amyloidogenic, and aggregative properties. Thymosin beta 4 (Tβ4) plays a role in the nervous system and may be related to motility, axonal enlargement, differentiation, neurite outgrowth, and proliferation. However, no studies about the effects of Tβ4 on prion disease have been performed yet. In the present study, we investigated the protective effect of Tβ4 against synthetic PrP (106-126) and considered possible mechanisms. Hippocampal neuronal HT22 cells were treated with Tβ4 and PrP (106-126) for 24 h. Tβ4 significantly reversed cell viability and reactive oxidative species (ROS) affected by PrP (106-126). Apoptotic proteins induced by PrP (106-126) were reduced by Tβ4. Interestingly, a balance of neurotrophic factors (nerve growth factor and brain-derived neurotrophic factor) and receptors (nerve growth factor receptor p75, tropomyosin related kinase A and B) were competitively maintained by Tβ4 through receptors reacting to PrP (106-126). Our results demonstrate that Tβ4 protects neuronal cells against PrP (106-126) neurotoxicity via the interaction of neurotrophic factors/receptors.
Collapse
Affiliation(s)
- Sokho Kim
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Jeonbuk National University, Gobong-ro 79, Iksan 54596, Jeollabuk-do, Republic of Korea
- Knotus Co., Ltd., Incheon 22014, Republic of Korea
| | - Jihye Choi
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Jeonbuk National University, Gobong-ro 79, Iksan 54596, Jeollabuk-do, Republic of Korea
| | - Jungkee Kwon
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Jeonbuk National University, Gobong-ro 79, Iksan 54596, Jeollabuk-do, Republic of Korea
| |
Collapse
|
19
|
Depleted uranium causes renal mitochondrial dysfunction through the ETHE1/Nrf2 pathway. Chem Biol Interact 2023; 372:110356. [PMID: 36681261 DOI: 10.1016/j.cbi.2023.110356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 01/08/2023] [Accepted: 01/18/2023] [Indexed: 01/20/2023]
Abstract
The kidney is the main organ affected by acute depleted uranium (DU) toxicity. The mechanism of nephrotoxicity induced by DU is complex and needs to be further explored. This study aimed to elucidate the function of mitochondrial dysfunction in nephrotoxicity generated by DU and confirm the latent mechanism. We verified that DU (2.5-10 mg/kg) caused mitochondrial dysfunction in male rat kidneys and decreased ATP content and the mitochondrial membrane potential. In addition, melatonin (20 mg/kg), as an antioxidant, alleviated DU-induced oxidative stress and mitochondrial dysfunction in male rats, further reducing kidney damage caused by DU. These results indicate that mitochondrial dysfunction plays a vital role in DU nephrotoxicity. When ethylmalonic encephalopathy 1 (ETHE1) was knocked down, DU-induced oxidative stress and mitochondrial dysfunction were increased, and renal injury was aggravated. When exogenous ETHE1 protein was applied to renal cells, the opposite changes were observed. We also found that ETHE1 knockdown increased the expression of NF-E2-related factor 2 (Nrf2), a vital oxidative stress regulator, and its downstream molecules heme oxygenase-1 (HO-1) and NADPH quinone oxidoreductase 1 (NQO1). Nrf2 knockout also aggravated DU-induced oxidative stress, mitochondrial dysfunction, and kidney damage. In conclusion, DU causes oxidative stress and antioxidant defense imbalance in renal cells through the ETHE1/Nrf2 pathway, further causing mitochondrial dysfunction and ultimately leading to nephrotoxicity.
Collapse
|
20
|
Recent Advances in Cellular Signaling Interplay between Redox Metabolism and Autophagy Modulation in Cancer: An Overview of Molecular Mechanisms and Therapeutic Interventions. Antioxidants (Basel) 2023; 12:antiox12020428. [PMID: 36829987 PMCID: PMC9951923 DOI: 10.3390/antiox12020428] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/12/2023] Open
Abstract
Autophagy is a fundamental homeostatic process in which certain cellular components are ingested by double-membrane autophagosomes and then degraded to create energy or to maintain cellular homeostasis and survival. It is typically observed in nutrient-deprived cells as a survival mechanism. However, it has also been identified as a crucial process in maintaining cellular homeostasis and disease progression. Normal cellular metabolism produces reactive oxygen (ROS) and nitrogen species at low levels. However, increased production causes oxidative stress, which can lead to diabetes, cardiovascular diseases, neurological disorders, and cancer. It was recently shown that maintaining redox equilibrium via autophagy is critical for cellular responses to oxidative stress. However, little is understood about the molecular cancer processes that connect to the control of autophagy. In cancer cells, oncogenic mutations, carcinogens, and metabolic reprogramming cause increased ROS generation and oxidative stress. Recent studies have suggested that increased ROS generation activates survival pathways that promote cancer development and metastasis. Moreover, the relationship between metabolic programming and ROS in cancer cells is involved in redox homeostasis and the malignant phenotype. Currently, while the signaling events governing autophagy and how redox homeostasis affects signaling cascades are well understood, very little is known about molecular events related to autophagy. In this review, we focus on current knowledge about autophagy modulation and the role of redox metabolism to further the knowledge of oxidative stress and disease progression in cancer regulation. Therefore, this review focuses on understanding how oxidation/reduction events fine-tune autophagy to help understand how oxidative stress and autophagy govern cancer, either as processes leading to cell death or as survival strategies for maintaining redox homeostasis in cancer.
Collapse
|
21
|
Yang H, Ping X, Cui Y, Zheng S, Shentu X. Role of Rapamycin and 3-MA in oxidative damage of HLECs caused by two doses of UVB radiation. ADVANCES IN OPHTHALMOLOGY PRACTICE AND RESEARCH 2023; 3:15-22. [PMID: 37846426 PMCID: PMC10577839 DOI: 10.1016/j.aopr.2022.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/15/2022] [Accepted: 09/16/2022] [Indexed: 10/18/2023]
Abstract
Background This study compared the role of autophagy regulators Rapamycin and 3-MA in oxidative damage and apoptosis of human lens epithelial cells (HLECs) caused by two doses of Ultraviolet Radiation B (UVB). Methods HLECs were irradiated with UVB, and two doses of UVB damage models were constructed. After treatment with autophagy regulators, cell damage tests such as CCK-8, LDH activity, and Ros detection were performed. Western blotting was used to detect the levels of autophagy-related proteins and apoptosis-related proteins. Quantitative real-time PCR (RT-qPCR) was used to detect the mRNA leve of secondary antioxidant enzymes.Flow cytometry was used to examine cell viability and apoptosis. Finally, the proportion of autophagy and apoptosis was observed by electron microscope. Results Autophagy inhibitor 3-MA promoted oxidative damage and apoptosis of HLECs at low doses of UVB (5 mJ/cm2), which corresponds to 1.3 h of exposure to sunlight in human eyes. Under the high dose of UVB (50mJ/cm2), which is equivalent to 13 h of exposure to sunlight in human eyes, the autophagy inducer Rapamycin caused more extensive oxidative damage and apoptosis of HLECs. 3-MA was able to reduce this damage, indicating that moderate autophagy is necessary for HLECs to cope with mild oxidative stress. For high dose UVB-induced oxidative stress, the use of 3-MA inhibiting autophagy is more beneficial to reduce cell damage and apoptosis. The mechanisms include degradation of damaged organelles, regulation of the expression of antioxidant enzymes HO-1, NQO1, GCS and regulation of apoptosis-related proteins. Conclusions Autophagy played different roles in HLECs oxidative stress induced by two doses of UVB. It provides new ideas for reducing oxidative damage and apoptosis of HLECs to prevent or delay the progression of age-related cataract (ARC).
Collapse
Affiliation(s)
- Hao Yang
- Department of Ophthalmology, The Second Affiliated Hospital of Zhejiang University School of Medicine; Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, China
| | - Xiyuan Ping
- Department of Ophthalmology, The Second Affiliated Hospital of Zhejiang University School of Medicine; Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, China
| | - Yilei Cui
- Department of Ophthalmology, The Second Affiliated Hospital of Zhejiang University School of Medicine; Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, China
| | - Sifan Zheng
- GKT School of Medical Education, King's College London, London, UK
| | - Xingchao Shentu
- Department of Ophthalmology, The Second Affiliated Hospital of Zhejiang University School of Medicine; Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, China
| |
Collapse
|
22
|
Mitra S, Rauf A, Sutradhar H, Sadaf S, Hossain MJ, Soma MA, Emran TB, Ahmad B, Aljohani ASM, Al Abdulmonem W, Thiruvengadam M. Potential candidates from marine and terrestrial resources targeting mitochondrial inhibition: Insights from the molecular approach. Comp Biochem Physiol C Toxicol Pharmacol 2023; 264:109509. [PMID: 36368509 DOI: 10.1016/j.cbpc.2022.109509] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/21/2022] [Accepted: 11/03/2022] [Indexed: 11/11/2022]
Abstract
Mitochondria are the target sites for multiple disease manifestations, for which it is appealing to researchers' attention for advanced pharmacological interventions. Mitochondrial inhibitors from natural sources are of therapeutic interest due to their promising benefits on physiological complications. Mitochondrial complexes I, II, III, IV, and V are the most common sites for the induction of inhibition by drug candidates, henceforth alleviating the manifestations, prevalence, as well as severity of diseases. Though there are few therapeutic options currently available on the market. However, it is crucial to develop new candidates from natural resources, as mitochondria-targeting abnormalities are rising to a greater extent. Marine and terrestrial sources possess plenty of bioactive compounds that are appeared to be effective in this regard. Ample research investigations have been performed to appraise the potentiality of these compounds in terms of mitochondrial disorders. So, this review outlines the role of terrestrial and marine-derived compounds in mitochondrial inhibition as well as their clinical status too. Additionally, mitochondrial regulation and, therefore, the significance of mitochondrial inhibition by terrestrial and marine-derived compounds in drug discovery are also discussed.
Collapse
Affiliation(s)
- Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar, Swabi 23430, Khyber Pakhtunkhwa (KP), Pakistan.
| | - Hriday Sutradhar
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh
| | - Samia Sadaf
- Department of Genetic Engineering and Biotechnology, University of Chittagong, Chittagong 4331, Bangladesh
| | - Md Jamal Hossain
- Department of Pharmacy, State University of Bangladesh, 77 Satmasjid Road Dhanmondi, Dhaka 1205, Bangladesh
| | - Mahfuza Afroz Soma
- Department of Pharmacy, State University of Bangladesh, 77 Satmasjid Road Dhanmondi, Dhaka 1205, Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh; Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Bashir Ahmad
- Institute of Biotechnology & Microbiology, Bacha Khan University, Charsadda, KP, Pakistan
| | - Abdullah S M Aljohani
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, Qassim University, Buraydah, Saudi Arabia
| | - Waleed Al Abdulmonem
- Department of Pathology, College of Medicine, Qassim University, Buraydah, Saudi Arabia
| | - Muthu Thiruvengadam
- Department of Applied Bioscience, College of Life and Environmental Sciences, Konkuk University, Seoul 05029, Republic of Korea; Saveetha Dental College and Hospital, Saveetha Institute of Medical Technical Sciences, Chennai 600077, Tamil Nadu, India.
| |
Collapse
|
23
|
Zhang W, Geng X, Dong Q, Li X, Ye P, Lin M, Xu B, Jiang H. Crosstalk between autophagy and the Keap1-Nrf2-ARE pathway regulates realgar-induced neurotoxicity. JOURNAL OF ETHNOPHARMACOLOGY 2023; 301:115776. [PMID: 36191662 DOI: 10.1016/j.jep.2022.115776] [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: 07/18/2022] [Revised: 09/22/2022] [Accepted: 09/27/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Realgar, the main component of which is As2S2 or As4S4 (≥90%), is a traditional Chinese natural medicine that has been used to treat carbuncles, furuncles, snake and insect bites, abdominal pain caused by parasitic worms, and epilepsy in China for many years. Because realgar contains arsenic, chronic or excessive use of single-flavor realgar and realgar-containing Chinese patent medicine can lead to drug-induced arsenic poisoning, but the exact mechanism underlying its toxicity to the central nervous system is unclear. AIM OF THE STUDY The aim of this study was to clarify the mechanism of realgar-induced neurotoxicity and to investigate the effects of realgar on autophagy and the Keap1-Nrf2-ARE pathway. MATERIALS AND METHODS We used rats treated with the autophagy inhibitor 3-methyladenine (3-MA) or adeno-associated virus (AAV2/9-r-shRNA-Sqstm1, sh-p62) to investigate realgar-induced neurotoxicity and explore the specific relationship between autophagy and the Keap1-Nrf2-ARE pathway (the Nrf2 pathway) in the cerebral cortex. Molecular docking analysis was used to assess the interactions among the Nrf2, p62 and Keap1 proteins. RESULTS Our results showed that arsenic from realgar accumulated in the brain and blood to cause neuronal and synaptic damage, decrease exploratory behavior and spontaneous movement, and impair memory ability in rats. The mechanism may have involved realgar-mediated autophagy impairment and continuous activation of the Nrf2 pathway via the LC3-p62-Keap1-Nrf2 axis. However, because this activation of the Nrf2 pathway was not sufficient to counteract oxidative damage, apoptosis was aggravated in the cerebral cortex. CONCLUSIONS This study revealed that autophagy, the Nrf2 pathway, and apoptosis are involved in realgar-induced central nervous system toxicity and identified p62 as the hub of the LC3-p62-Keap1-Nrf2 axis in the regulation of autophagy, the Nrf2 pathway, and apoptosis.
Collapse
Affiliation(s)
- Weiwei Zhang
- School of Public Health, China Medical University, Shenyang, Liaoning, 110122, PR China; The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, Liaoning, PR China.
| | - Xu Geng
- School of Public Health, China Medical University, Shenyang, Liaoning, 110122, PR China.
| | - Qing Dong
- School of Public Health, China Medical University, Shenyang, Liaoning, 110122, PR China.
| | - Xiuhan Li
- School of Public Health, China Medical University, Shenyang, Liaoning, 110122, PR China.
| | - Ping Ye
- School of Public Health, China Medical University, Shenyang, Liaoning, 110122, PR China.
| | - Mengyuan Lin
- School of Public Health, China Medical University, Shenyang, Liaoning, 110122, PR China.
| | - Bin Xu
- School of Public Health, China Medical University, Shenyang, Liaoning, 110122, PR China.
| | - Hong Jiang
- School of Public Health, China Medical University, Shenyang, Liaoning, 110122, PR China; The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, Liaoning, PR China.
| |
Collapse
|
24
|
He J, Gao J, Zhu H, Zhao Y, Zhang X, Wang X, Wan S, Cao H, Zhai L, Wang Y, Wang S. Effects of NBP on postoperative cognitive dysfunction in rats via Nrf 2/ARE pathway. Aging (Albany NY) 2023; 15:276-286. [PMID: 36626245 PMCID: PMC9876636 DOI: 10.18632/aging.204481] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/29/2022] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Postoperative cognitive dysfunction (POCD) is a common postoperative disease that threatens patients' quality of life, especially elderly patients. With the popularity of anesthesia/surgery, POCD has received more attention worldwide. The objective of this research is to evaluate 3-n-Butylphthalide (NBP)'s protective effect on postoperative cognitive function in rats and its related mechanisms. METHODS Tibial fracture models of senile rats of POCD were established and divided into blank control group, solvent group, NBP group, Nrf 2 agonist group, and Nrf 2 inhibitor group. The changes in the cognitive abilities of rats were systematically evaluated by the Morris water maze test. After hematoxylin-eosin (HE) staining of the hippocampus, the morphological and structural changes of hippocampal neurons were observed by light microscopy. The expressions of apoptosis-related proteins were analyzed by immunohistochemistry and Western blot was used to detect the expressions of Nrf 2,HO-1,Mfn1,Mfn2,Drp1 proteins. Moreover, the changes in the morphology of mitochondria were observed by transmission electron microscopy. RESULTS Through the water maze test, we observed that the incidence of postoperative cognitive impairment in the NBP, agonist, and inhibitor groups was substantially lower as compared to the blank control group and solvent group (P < 0.05). The expressions of Nrf 2, HO-1, Mfn1, Mfn2, and Drp1 proteins in the NBP group were upregulated in comparison to the blank control group and the solvent group. The expressions of related proteins in the inhibitor group were substantially lower in comparison to the NBP group. CONCLUSIONS NBP can affect the postoperative cognitive function of rats by activating the Nrf 2/ARE signaling pathway.
Collapse
Affiliation(s)
- Jianshuai He
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Junqiong Gao
- Department of Anesthesiology, Weihai Municipal Hospital, Weihai, China
| | - He Zhu
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yang Zhao
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaotian Zhang
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiufang Wang
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shengnan Wan
- Department of Thyroid Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hongying Cao
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lin Zhai
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuanyong Wang
- Department of Thoracic Surgery, Tangdu Hospital of Air Force Military Medical University, Xi’an, China
| | - Shilei Wang
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
25
|
Hydrogen sulfide protects retinal pigment epithelium cells against ferroptosis through the AMPK- and p62-dependent non-canonical NRF2-KEAP1 pathway. Exp Cell Res 2023; 422:113436. [PMID: 36435220 DOI: 10.1016/j.yexcr.2022.113436] [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: 10/19/2022] [Revised: 11/17/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022]
Abstract
Oxidative stress-induced ferroptosis of retinal pigment epithelium (RPE) cells contributes to retinal degenerative diseases. The antioxidant molecule hydrogen sulfide (H2S) regulates oxidative stress response, but its effect on the ferroptosis of RPE cells is unclear. In this study, sodium hydrosulfide (NaHS) was used as an exogenous H2S donor to intervene tert-butyl hydroperoxide (t-BHP)-induced ferroptosis of APRE-19 cells. We found that NaHS pretreatment attenuates t-BHP-induced oxidative stress and ferroptosis. Analysis of mRNA-sequencing coupled with FerrDb database identified nuclear factor erythroid-2-related factor 2 (NRF2) as a primary target for the cytoprotective role of H2S. NRF2 inhibitor ML385 reverses the effects of H2S on ferroptosis. Biochemical analysis revealed that H2S stabilizes NRF2. H2S decreases the interaction between NRF2 and KEAP1, but enhances the interaction between KEAP1 and p62. These results suggest that H2S activates the non-canonical NRF2-KEAP1 pathway. Further study demonstrated that H2S stimulates AMPK to interact and phosphorylate p62. Additionally, inhibiting AMPK or knocking down p62 blocks the effects of H2S. We speculate that targeting the non-canonical NRF2-KEAP1 pathway by H2S-based drug may benefit the treatment of retinal degenerative diseases.
Collapse
|
26
|
Das TK, Ganesh BP. Interlink between the gut microbiota and inflammation in the context of oxidative stress in Alzheimer's disease progression. Gut Microbes 2023; 15:2206504. [PMID: 37127846 PMCID: PMC10153019 DOI: 10.1080/19490976.2023.2206504] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/03/2023] Open
Abstract
The microbiota-gut-brain axis is an important pathway of communication and may dynamically contribute to Alzheimer's disease (AD) pathogenesis. Pathological commensal gut microbiota alterations, termed as dysbiosis, can influence intestinal permeability and break the blood-brain barrier which may trigger AD pathogenesis via redox signaling, neuronal, immune, and metabolic pathways. Dysbiosis increases the oxidative stress. Oxidants affect the innate immune system through recognizing microbial-derived pathogens by Toll-like receptors and initiating the inflammatory process. Most of the gut microbiome research work highlights the relationship between the gut microbiota and AD, but the contributory connection between precise bacteria and brain dysfunction in AD pathology cannot be fully demonstrated. Here, we summarize the current information of the fundamental connections between oxidative stress, inflammation, and gut dysbiosis in AD. This review emphasizes on the involvement of gut microbiota in the regulation of oxidative stress, inflammation, immune responses including central and peripheral cross-talk. It provides insights for novel preventative and therapeutic approaches in AD.
Collapse
Affiliation(s)
- Tushar K Das
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Bhanu P Ganesh
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
27
|
Wei M, Bao G, Li S, Yang Z, Cheng C, Le W. PM2.5 exposure triggers cell death through lysosomal membrane permeabilization and leads to ferroptosis insensitivity via the autophagy dysfunction/p62-KEAP1-NRF2 activation in neuronal cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 248:114333. [PMID: 36446170 DOI: 10.1016/j.ecoenv.2022.114333] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 11/14/2022] [Accepted: 11/22/2022] [Indexed: 06/16/2023]
Abstract
PM2.5 exposure can be associated with the onset of neurodegenerative diseases, with oxidative stress-induced cellular homeostasis disruption and cell death as one of the main mechanisms. However, the exact cellular and molecular processes are still rarely investigated. Autophagy and KEAP1-NRF2 (Kelch-like ECH-Associating protein 1-nuclear factor erythroid 2 related factor 2) signaling pathway are two main cellular defense systems for maintaining cellular homeostasis and resisting oxidative stress. In this study, we primarily investigated the role of autophagy and KEAP1-NRF2 in regulating cell death resulting from PM2.5 exposure in mouse neuroblastoma N2a cells. Our results showed that PM2.5 exposure disrupted autophagic flux by impairing lysosomal function, including lysosomal alkalinization, increased lysosome membrane permeabilization (LMP), and Cathepsin B release. Furthermore, dysregulated autophagy enhances NRF2 activity in a p62-dependent manner, which then initiates the expression of a series of antioxidant genes and increases cellular insensitivity to ferroptosis. Meanwhile, autophagy dysfunction impairs the intracellular degradation of ferroptosis related proteins such as GPX4 and ferritin. As these proteins accumulate, cells also become less sensitive to ferroptosis. LMP-associated cell death may be the main mechanism of PM2.5-induced N2a cytotoxicity. Our results may provide insights into the mechanisms of PM2.5-induced neurotoxicity and predict effective prevention and treatment strategies.
Collapse
Affiliation(s)
- Min Wei
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian 116021, China.
| | - Guangming Bao
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian 116021, China
| | - Song Li
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian 116021, China
| | - Zhaofei Yang
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian 116021, China
| | - Cheng Cheng
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian 116021, China
| | - Weidong Le
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian 116021, China; Institute of Neurology, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, Chengdu 610072, China.
| |
Collapse
|
28
|
Impellizzeri D, D’Amico R, Fusco R, Genovese T, Peritore AF, Gugliandolo E, Crupi R, Interdonato L, Di Paola D, Di Paola R, Cuzzocrea S, Siracusa R, Cordaro M. Açai Berry Mitigates Vascular Dementia-Induced Neuropathological Alterations Modulating Nrf-2/Beclin1 Pathways. Cells 2022; 11:cells11162616. [PMID: 36010690 PMCID: PMC9406985 DOI: 10.3390/cells11162616] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/19/2022] [Accepted: 08/20/2022] [Indexed: 12/14/2022] Open
Abstract
The second-most common cause of dementia is vascular dementia (VaD). The majority of VaD patients experience cognitive impairment, which is brought on by oxidative stress and changes in autophagic function, which ultimately result in neuronal impairment and death. In this study, we examine a novel method for reversing VaD-induced changes brought on by açai berry supplementation in a VaD mouse model. The purpose of this study was to examine the impact of açai berries on the molecular mechanisms underlying VaD in a mouse model of the disease that was created by repeated ischemia-reperfusion (IR) of the whole bilateral carotid artery. Here, we found that açai berry was able to reduce VaD-induced behavioral alteration, as well as hippocampal death, in CA1 and CA3 regions. These effects are probably due to the modulation of nuclear factor erythroid 2-related factor 2 (Nrf-2) and Beclin-1, suggesting a possible crosstalk between these molecular pathways. In conclusion, the protective effects of açai berry could be a good supplementation in the future for the management of vascular dementia.
Collapse
Affiliation(s)
- Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy
| | - Ramona D’Amico
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy
| | - Tiziana Genovese
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy
| | - Alessio Filippo Peritore
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy
| | - Enrico Gugliandolo
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Rosalia Crupi
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Livia Interdonato
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy
| | - Davide Di Paola
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy
| | - Rosanna Di Paola
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
- Correspondence: (R.D.P.); (S.C.)
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, Saint Louis, MO 63104, USA
- Correspondence: (R.D.P.); (S.C.)
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy
| | - Marika Cordaro
- Department of Biomedical, Dental and Morphological and Functional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| |
Collapse
|
29
|
Zhai Z, Huang Y, Zhang Y, Zhao L, Li W. Clinical Research Progress of Small Molecule Compounds Targeting Nrf2 for Treating Inflammation-Related Diseases. Antioxidants (Basel) 2022; 11:1564. [PMID: 36009283 PMCID: PMC9405369 DOI: 10.3390/antiox11081564] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/03/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
Studies have found that inflammation is a symptom of various diseases, such as coronavirus disease 2019 (COVID-19) and rheumatoid arthritis (RA); it is also the source of other diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), lupus erythematosus (LE), and liver damage. Nrf2 (nuclear factor erythroid 2-related factor 2) is an important multifunctional transcription factor in cells and plays a central regulatory role in cellular defense mechanisms. In recent years, several studies have found a strong association between the activation of Nrf2 and the fight against inflammation-related diseases. A number of small molecule compounds targeting Nrf2 have entered clinical research. This article reviews the research status of small molecule compounds that are in clinical trials for the treatment of COVID-19, rheumatoid arthritis, Alzheimer's disease, Parkinson's disease, lupus erythematosus, and liver injury.
Collapse
Affiliation(s)
- Zhenzhen Zhai
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yanxin Huang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yawei Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Lili Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Wen Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education & Key Laboratory of Henan Province for Drug Quality and Evaluation, Zhengzhou 450001, China
| |
Collapse
|
30
|
Dai XY, Zhu SY, Chen J, Li MZ, Zhao Y, Talukder M, Li JL. Lycopene alleviates di(2-ethylhexyl) phthalate-induced splenic injury by activating P62-Keap1-NRF2 signaling. Food Chem Toxicol 2022; 168:113324. [PMID: 35917956 DOI: 10.1016/j.fct.2022.113324] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 07/16/2022] [Accepted: 07/20/2022] [Indexed: 01/15/2023]
Abstract
Di(2-ethylhexyl) phthalate (DEHP) is an omnipresent environmental pollutant. It has been determined that DEHP is involved in multiple health disorders. Lycopene (Lyc) is a natural carotenoid pigment, with anti-inflammatory and antioxidant properties. However, it is not clear whether Lyc can protect the spleen from DEHP-induced oxidative damage. A total of 140 mice were randomly divided into seven groups (n = 20) and continuously gavaged with corn oil, distilled water, DEHP (500 or 1000 mg/kg BW/day) and/or Lyc (5 mg/kg BW/day) for 28 days. Histopathological and ultrastructural results showed a DEHP-induced inflammatory response and mitochondrial injuries. Moreover, DEHP exposure induced redox imbalance, which resulted in the up-regulation of ROS activity and MDA content, and the down-regulation of T-AOC, T-SOD and CAT in the DEHP groups. Simultaneously, our results also demonstrated that DEHP-induced kelch-like ECH-associated protein 1 (Keap1) expression was downregulated, and the expression levels of P62, nuclear factor erythroid 2-related factor (NRF2) and their downstream target genes were up-regulated. However, the supplementary Lyc reverted these changes to normal levels. Together, Lyc prevented DEHP-induced splenic injuries by regulating the P62-Keap1-NRF2 signaling pathway. Hence, the protective effects of Lyc might be a therapeutic strategy to ameliorate DEHP-induced splenic damage.
Collapse
Affiliation(s)
- Xue-Yan Dai
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China; College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, PR China
| | - Shi-Yong Zhu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Jian Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Mu-Zi Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Yi Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Milton Talukder
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China; Department of Physiology and Pharmacology, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Barishal, 8210, Bangladesh
| | - Jin-Long Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, PR China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
31
|
Groth M, Skrzydlewska E, Dobrzyńska M, Pancewicz S, Moniuszko-Malinowska A. Redox Imbalance and Its Metabolic Consequences in Tick-Borne Diseases. Front Cell Infect Microbiol 2022; 12:870398. [PMID: 35937690 PMCID: PMC9353526 DOI: 10.3389/fcimb.2022.870398] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 06/13/2022] [Indexed: 11/21/2022] Open
Abstract
One of the growing global health problems are vector-borne diseases, including tick-borne diseases. The most common tick-borne diseases include Lyme disease, tick-borne encephalitis, human granulocytic anaplasmosis, and babesiosis. Taking into account the metabolic effects in the patient's body, tick-borne diseases are a significant problem from an epidemiological and clinical point of view. Inflammation and oxidative stress are key elements in the pathogenesis of infectious diseases, including tick-borne diseases. In consequence, this leads to oxidative modifications of the structure and function of phospholipids and proteins and results in qualitative and quantitative changes at the level of lipid mediators arising in both reactive oxygen species (ROS) and ROS enzyme-dependent reactions. These types of metabolic modifications affect the functioning of the cells and the host organism. Therefore, links between the severity of the disease state and redox imbalance and the level of phospholipid metabolites are being searched, hoping to find unambiguous diagnostic biomarkers. Assessment of molecular effects of oxidative stress may also enable the monitoring of the disease process and treatment efficacy.
Collapse
Affiliation(s)
- Monika Groth
- Department of Infectious Diseases and Neuroinfections, Medical University of Bialystok, Bialystok, Poland
| | - Elżbieta Skrzydlewska
- Department of Inorganic and Analytical Chemistry, Medical University of Bialystok, Bialystok, Poland
| | - Marta Dobrzyńska
- Department of Inorganic and Analytical Chemistry, Medical University of Bialystok, Bialystok, Poland
| | - Sławomir Pancewicz
- Department of Infectious Diseases and Neuroinfections, Medical University of Bialystok, Bialystok, Poland
| | - Anna Moniuszko-Malinowska
- Department of Infectious Diseases and Neuroinfections, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
32
|
Hussain Y, Khan H, Alsharif KF, Hayat Khan A, Aschner M, Saso L. The Therapeutic Potential of Kaemferol and Other Naturally Occurring Polyphenols Might Be Modulated by Nrf2-ARE Signaling Pathway: Current Status and Future Direction. Molecules 2022; 27:4145. [PMID: 35807387 PMCID: PMC9268049 DOI: 10.3390/molecules27134145] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/13/2022] [Accepted: 06/20/2022] [Indexed: 02/01/2023] Open
Abstract
Kaempferol is a natural flavonoid, which has been widely investigated in the treatment of cancer, cardiovascular diseases, metabolic complications, and neurological disorders. Nrf2 (nuclear factor erythroid 2-related factor 2) is a transcription factor involved in mediating carcinogenesis and other ailments, playing an important role in regulating oxidative stress. The activation of Nrf2 results in the expression of proteins and cytoprotective enzymes, which provide cellular protection against reactive oxygen species. Phytochemicals, either alone or in combination, have been used to modulate Nrf2 in cancer and other ailments. Among them, kaempferol has been recently explored for its anti-cancer and other anti-disease therapeutic efficacy, targeting Nrf2 modulation. In combating cancer, diabetic complications, metabolic disorders, and neurological disorders, kaempferol has been shown to regulate Nrf2 and reduce redox homeostasis. In this context, this review article highlights the current status of the therapeutic potential of kaempferol by targeting Nrf2 modulation in cancer, diabetic complications, neurological disorders, and cardiovascular disorders. In addition, we provide future perspectives on kaempferol targeting Nrf2 modulation as a potential therapeutic approach.
Collapse
Affiliation(s)
- Yaseen Hussain
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China;
- Department of Pharmacy, Bashir Institute of Health Sciences, Islamabad 45400, Pakistan
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan
| | - Khalaf F. Alsharif
- Department of Clinical Laboratory, College of Applied Medical Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
| | - Amjad Hayat Khan
- Department of Allied Health Sciences, Bashir Institute of Health Sciences, Islamabad 45400, Pakistan;
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY 10463, USA;
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University, P.le Aldo Moro 5, 00185 Rome, Italy
| |
Collapse
|
33
|
Zoungrana LI, Krause-Hauch M, Wang H, Fatmi MK, Bates L, Li Z, Kulkarni P, Ren D, Li J. The Interaction of mTOR and Nrf2 in Neurogenesis and Its Implication in Neurodegenerative Diseases. Cells 2022; 11:cells11132048. [PMID: 35805130 PMCID: PMC9265429 DOI: 10.3390/cells11132048] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/23/2022] [Accepted: 06/26/2022] [Indexed: 12/24/2022] Open
Abstract
Neurogenesis occurs in the brain during embryonic development and throughout adulthood. Neurogenesis occurs in the hippocampus and under normal conditions and persists in two regions of the brain—the subgranular zone (SGZ) in the dentate gyrus of the hippocampus and the subventricular zone (SVZ) of the lateral ventricles. As the critical role in neurogenesis, the neural stem cells have the capacity to differentiate into various cells and to self-renew. This process is controlled through different methods. The mammalian target of rapamycin (mTOR) controls cellular growth, cell proliferation, apoptosis, and autophagy. The transcription factor Nrf2 (nuclear factor erythroid 2-related factor 2) is a major regulator of metabolism, protein quality control, and antioxidative defense, and is linked to neurogenesis. However, dysregulation in neurogenesis, mTOR, and Nrf2 activity have all been associated with neurodegenerative diseases such as Alzheimer’s, Huntington’s, and Parkinson’s. Understanding the role of these complexes in both neurogenesis and neurodegenerative disease could be necessary to develop future therapies. Here, we review both mTOR and Nrf2 complexes, their crosstalk and role in neurogenesis, and their implication in neurodegenerative diseases.
Collapse
Affiliation(s)
- Linda Ines Zoungrana
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (L.I.Z.); (M.K.-H.); (H.W.); (M.K.F.); (L.B.); (D.R.)
| | - Meredith Krause-Hauch
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (L.I.Z.); (M.K.-H.); (H.W.); (M.K.F.); (L.B.); (D.R.)
| | - Hao Wang
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (L.I.Z.); (M.K.-H.); (H.W.); (M.K.F.); (L.B.); (D.R.)
| | - Mohammad Kasim Fatmi
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (L.I.Z.); (M.K.-H.); (H.W.); (M.K.F.); (L.B.); (D.R.)
| | - Lauryn Bates
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (L.I.Z.); (M.K.-H.); (H.W.); (M.K.F.); (L.B.); (D.R.)
| | - Zehui Li
- Department of Medical Engineering, College of Engineering and Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (Z.L.); (P.K.)
| | - Parth Kulkarni
- Department of Medical Engineering, College of Engineering and Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (Z.L.); (P.K.)
| | - Di Ren
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (L.I.Z.); (M.K.-H.); (H.W.); (M.K.F.); (L.B.); (D.R.)
| | - Ji Li
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (L.I.Z.); (M.K.-H.); (H.W.); (M.K.F.); (L.B.); (D.R.)
- Correspondence: ; Tel.: +1-813-974-4917
| |
Collapse
|
34
|
Xu M, Li L, Liu H, Lu W, Ling X, Gong M. Rutaecarpine Attenuates Oxidative Stress-Induced Traumatic Brain Injury and Reduces Secondary Injury via the PGK1/KEAP1/NFR2 Signaling Pathway. Front Pharmacol 2022; 13:807125. [PMID: 35529443 PMCID: PMC9070303 DOI: 10.3389/fphar.2022.807125] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/22/2022] [Indexed: 01/17/2023] Open
Abstract
The oxidative stress response caused by traumatic brain injury (TBI) leads to secondary damage in the form of tissue damage and cell death. Nuclear transcription-related factor 2 (NRF2) is a key factor in the body against oxidative stress and has an important role in combating oxidative damage in TBI neurons. In the present study, we investigated whether rutaecarpine could activate the PGK1/KEAP1/NRF2 pathway to antagonize oxidative damage in TBI neurons. We performed controlled cortical impact (CCI) surgery on mice and taken H2O2 treatment on PC12 cells to construct TBI models. The results of western blot showed that the expression of PGK1, KEAP and NRF2 was regulated and accompanied by altered levels of oxidative stress, and the use of rutaecarpine in the TBI model mice significantly improved cognitive dysfunction, increased antioxidant capacity and reduced apoptosis in brain tissue. Similar antioxidant damage results were obtained using rutaecarpine in a PC12 cell model. Furthermore, through the use of the protein synthesis inhibitor CHX and the proteasome synthesis inhibitor MG-132, rutaecarpine was found to promote the expreesions of PGK1 and NRF2 by accelerating PGK1 ubiquitination to reduce PGK1 expression. Therefore, rutaecarpine may be a promising therapeutic agent for the treatment of TBI-related neuro-oxidative damage.
Collapse
Affiliation(s)
- Min Xu
- Department of Neurosurgery, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan, China
| | - Liu Li
- Collaborative Innovation Center of Jiangsu Province of Cancer Prevention and Treatment of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hua Liu
- Department of Neurosurgery, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Wei Lu
- Department of Neurosurgery, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan, China
| | - Xiaoyang Ling
- Department of Neurosurgery, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan, China
| | - Mingjie Gong
- Department of Neurosurgery, Changshu No.2 People’s Hospital, The Affiliated Changshu Hospital of Xuzhou Medical University, Changshu, China
- *Correspondence: Mingjie Gong,
| |
Collapse
|
35
|
The Interplay between Autophagy and Redox Signaling in Cardiovascular Diseases. Cells 2022; 11:cells11071203. [PMID: 35406767 PMCID: PMC8997791 DOI: 10.3390/cells11071203] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 12/20/2022] Open
Abstract
Reactive oxygen and nitrogen species produced at low levels under normal cellular metabolism act as important signal molecules. However, at increased production, they cause damage associated with oxidative stress, which can lead to the development of many diseases, such as cardiovascular, metabolic, neurodegenerative, diabetes, and cancer. The defense systems used to maintain normal redox homeostasis plays an important role in cellular responses to oxidative stress. The key players here are Nrf2-regulated redox signaling and autophagy. A tight interface has been described between these two processes under stress conditions and their role in oxidative stress-induced diseases progression. In this review, we focus on the role of Nrf2 as a key player in redox regulation in cell response to oxidative stress. We also summarize the current knowledge about the autophagy regulation and the role of redox signaling in this process. In line with the focus of our review, we describe in more detail information about the interplay between Nrf2 and autophagy pathways in myocardium and the role of these processes in cardiovascular disease development.
Collapse
|
36
|
PrP Sc Inhibition and Cellular Protection of DBL on a Prion-Infected Cultured Cell via Multiple Pathways. Mol Neurobiol 2022; 59:3310-3321. [PMID: 35303279 DOI: 10.1007/s12035-022-02729-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 01/01/2022] [Indexed: 10/18/2022]
Abstract
Prion diseases are kinds of fatal neurodegenerative diseases without effective therapeutic and prophylactic tools currently. In this study, the inhibition of PrPSc propagation and cellular protectivity of 3,4-dihydroxybenzalacetone (DBL), a small catechol-containing compound isolated and purified from the ethanol extract of Inonotus obliquus, upon a prion-infected cell line SMB-S15 were evaluated. Western blots showed that after incubation with 10 μM of DBL for 14 days, the level of PrPSc in SMB-S15 cells was significantly decreased. Meanwhile, the levels of ROS and hydrogen peroxide were decreased with a dose-dependent manner, whereas the levels of some antioxidant factors, such as HO-1, GCLC and GCLM, were significantly increased. The activities of total glutathione and SOD were up-regulated. DBL-treated SMB-S15 cells also showed the up-regulation of UPR-related proteins, including PERK, IRE1α, ATF6 and GRP78, and activation of autophagy system. Furthermore, the SIRT3 abnormalities caused by prion infection were relieved by DBL treatment. On the contrary, these comprehensive changes were not significantly noticed in the normal partner cell line SMB-PS under the same experimental condition. Those data indicate that treatment of DBL on prion-infected cells can reduce PrPSc level, activate UPR and autophagy system and meanwhile relieve intracellular oxidative stress, endoplasmic reticulum stress and mitochondrial dysfunction by raising the levels of multiple antioxidant factors. The PrPSc inhibition and protective effectiveness of DBL upon the prion-infected cells in vitro make it worthy of further study.
Collapse
|
37
|
Muhammad F, Liu Y, Zhou Y, Yang H, Li H. Antioxidative role of Traditional Chinese Medicine in Parkinson's disease. JOURNAL OF ETHNOPHARMACOLOGY 2022; 285:114821. [PMID: 34838943 DOI: 10.1016/j.jep.2021.114821] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/24/2021] [Accepted: 11/05/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Neuroprotective Traditional Chinese Medicine (TCM) has been practiced in alternative medicine from early days. TCM-derived neuroprotective compounds, such as Chrysin, Cannabidiol, Toonasinoids, and β-asaron, exert significant effectiveness's towards Parkinson's disease (PD). Further, these neuroprotective TCM showed antioxidative, anti-inflammatory, anti-tumor, anti-septic, analgesic properties. Recent research showed that the reduction in the reactive oxygen species (ROS) decreased the α-synuclein (α-syn) toxicity and enhanced the dopaminergic neuron regenerations, the main hallmarks of PD. Therefore, the neuroprotective effects of novel TCM due to its antiradical activities needed deep investigations. AIMS OF THE STUDY This review aims to enlighten the neuroprotective TCM and its components with their antioxidative properties to the scientific community for future research. METHOD The relevant information on the neuroprotective TCM was gathered from scientific databases (PubMed, Web of Science, Google Scholar, ScienceDirect, SciFinder, Wiley Online Library, ACS Publications, and CNKI). Information was also gained from MS and Ph.D. thesis, books, and online databases. The literature cited in this review dates from 2001 to June 2, 0201. RESULTS Novel therapies for PD are accessible, mostly rely on Rivastigmine and Donepezil, offers to slow down the progression of disease at an early stage but embraces lots of disadvantages. Researchers are trying to find a potential drug against PD, which is proficient at preventing or curing the disease progress, but still needed to be further identified. Oxidative insult and mitochondrial dysfunction are thought to be the main culprit of neurodegenerations. Reactive oxygen species (ROS) are the only causative agent in all interactions, leading to PD, from mitochondrial dysfunctions, α-syn aggregative toxicity, and DA neurons degenerations. It is evident from the redox balance, which seems an imperative therapeutic approach against PD and was necessary for the significant neuronal activities. CONCLUSION Our study is explaining the newly discovered TCM and their neuroprotective and antioxidative properties. But also bring up the possible treatment approaches against PD for future researchers.
Collapse
Affiliation(s)
- Fahim Muhammad
- College of Life Sciences, Lanzhou University, Lanzhou, China
| | - Yan Liu
- School of Pharmacy, Lanzhou University, Donggang West Road No. 199, Lanzhou, 730020, China
| | - Yongtao Zhou
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China; Clinical Center for Parkinson's Disease, Capital Medical University, Beijing, China
| | - Hui Yang
- Instiute of Biology Gansu Academy of Sciences, China.
| | - Hongyu Li
- College of Life Sciences, Lanzhou University, Lanzhou, China; School of Pharmacy, Lanzhou University, Donggang West Road No. 199, Lanzhou, 730020, China.
| |
Collapse
|
38
|
Davidson JM, Chung RS, Lee A. The converging roles of sequestosome-1/p62 in the molecular pathways of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Neurobiol Dis 2022; 166:105653. [PMID: 35143965 DOI: 10.1016/j.nbd.2022.105653] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 01/18/2022] [Accepted: 02/03/2022] [Indexed: 01/03/2023] Open
Abstract
Investigations into the pathogenetic mechanisms underlying amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) have provided significant insight into the disease. At the cellular level, ALS and FTD are classified as proteinopathies, which is motor neuron degeneration and death characterized by pathological protein aggregates or dysregulated proteostasis. At both the clinical and molecular level there are common signaling pathways dysregulated across the ALS and FTD spectrum (ALS/FTD). Sequestosome-1/p62 is a multifunctional scaffold protein with roles in several signaling pathways including proteostasis, protein degradation via the ubiquitin proteasome system and autophagy, the antioxidant response, inflammatory response, and apoptosis. Notably these pathways are dysregulated in ALS and FTD. Mutations in the functional domains of p62 provide links to the pathogenetic mechanisms of p62 and dyshomeostasis of p62 levels is noted in several types of ALS and FTD. We present here that the dysregulated ALS and FTD signaling pathways are linked, with p62 converging the molecular mechanisms. This review summarizes the current literature on the complex role of p62 in the pathogenesis across the ALS/FTD spectrum. The focus is on the underlying convergent molecular mechanisms of ALS and FTD-associated proteins and pathways that dysregulate p62 levels or are dysregulated by p62, with emphasis on how p62 is implicated across the ALS/FTD spectrum.
Collapse
Affiliation(s)
- Jennilee M Davidson
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 2 Technology Place, NSW 2109, Australia..
| | - Roger S Chung
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 2 Technology Place, NSW 2109, Australia..
| | - Albert Lee
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 2 Technology Place, NSW 2109, Australia..
| |
Collapse
|
39
|
Tomer S, Mu W, Suryawanshi G, Ng H, Wang L, Wennerberg W, Rezek V, Martin H, Chen I, Kitchen S, Zhen A. Cannabidiol modulates expression of type I IFN response genes and HIV infection in macrophages. Front Immunol 2022; 13:926696. [PMID: 36248834 PMCID: PMC9560767 DOI: 10.3389/fimmu.2022.926696] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 09/15/2022] [Indexed: 01/27/2023] Open
Abstract
Cannabis (Cannabis sativa) is a widely used drug in the United States and the frequency of cannabis use is particularly high among people living with HIV (PLWH). One key component of cannabis, the non-psychotropic (-)-cannabidiol (CBD) exerts a wide variety of biological actions, including anticonvulsive, analgesic, and anti-inflammatory effects. However, the exact mechanism of action through which CBD affects the immune cell signaling remains poorly understood. Here we report that CBD modulates type I interferon responses in human macrophages. Transcriptomics analysis shows that CBD treatment significantly attenuates cGAS-STING-mediated activation of type I Interferon response genes (ISGs) in monocytic THP-1 cells. We further showed that CBD treatment effectively attenuates 2'3-cGAMP stimulation of ISGs in both THP-1 cells and primary human macrophages. Interestingly, CBD significantly upregulates expression of autophagy receptor p62/SQSTM1. p62 is critical for autophagy-mediated degradation of stimulated STING. We observed that CBD treated THP-1 cells have elevated autophagy activity. Upon 2'3'-cGAMP stimulation, CBD treated cells have rapid downregulation of phosphorylated-STING, leading to attenuated expression of ISGs. The CBD attenuation of ISGs is reduced in autophagy deficient THP-1 cells, suggesting that the effects of CBD on ISGs is partially mediated by autophagy induction. Lastly, CBD decreases ISGs expression upon HIV infection in THP-1 cells and human primary macrophages, leading to increased HIV RNA expression 24 hours after infection. However, long term culture with CBD in infected primary macrophages reduced HIV viral spread, suggesting potential dichotomous roles of CBD in HIV replication. Our study highlights the immune modulatory effects of CBD and the needs for additional studies on its effect on viral infection and inflammation.
Collapse
Affiliation(s)
- Shallu Tomer
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Wenli Mu
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Gajendra Suryawanshi
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Hwee Ng
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Li Wang
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Wally Wennerberg
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Valerie Rezek
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Heather Martin
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Irvin Chen
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Scott Kitchen
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Anjie Zhen
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
- *Correspondence: Anjie Zhen,
| |
Collapse
|
40
|
Binyamin O, Frid K, Keller G, Saada A, Gabizon R. Comparing anti-aging hallmark activities of Metformin and Nano-PSO in a mouse model of genetic Creutzfeldt-Jakob Disease. Neurobiol Aging 2021; 110:77-87. [PMID: 34875507 DOI: 10.1016/j.neurobiolaging.2021.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 10/20/2021] [Accepted: 11/04/2021] [Indexed: 01/10/2023]
Abstract
Advanced age is the main risk factor for the manifestation of late onset neurodegenerative diseases. Metformin, an anti-diabetic drug, was shown to extend longevity, and to ameliorate the activity of recognized aging hallmarks. Here, we compared the clinical, pathologic and biochemical effects of Metformin to those of Nano-PSO (Granagard), a brain targeted anti-oxidant shown by us to delay disease advance in transgenic mice mimicking for genetic Creutzfeldt Jacob disease (CJD) linked to the E200KPrP mutation. We demonstrate that both Metformin and Nano-PSO reduced aging hallmarks activities such as activated AMPK, the main energy sensor of cells as well as Nrf2 and COX IV1, regulators of oxidation, and mitochondrial activity. Both compounds reduced inflammation and increased stem cells production, however did not decrease PrP accumulation. As opposed to Nano-PSO, Metformin neither delayed clinical disease advance in these mice nor reduced the accumulation of sulfated glycosaminoglycans, a pathologic feature of prion disease. We conclude that elevation of anti-aging markers may not be sufficient to delay the fatal advance of genetic CJD.
Collapse
Affiliation(s)
- Orli Binyamin
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah Medical Center, Jerusalem, Israel; Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Kati Frid
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah Medical Center, Jerusalem, Israel; Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Guy Keller
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah Medical Center, Jerusalem, Israel; Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ann Saada
- Department of Genetic and Metabolic Diseases, Hadassah Medical Center, Jerusalem Israel, Jerusalem, Israel; Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ruth Gabizon
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah Medical Center, Jerusalem, Israel; Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
41
|
Leite ADOF, Bento Torres Neto J, dos Reis RR, Sobral LL, de Souza ACP, Trévia N, de Oliveira RB, Lins NADA, Diniz DG, Diniz JAP, Vasconcelos PFDC, Anthony DC, Brites D, Picanço Diniz CW. Unwanted Exacerbation of the Immune Response in Neurodegenerative Disease: A Time to Review the Impact. Front Cell Neurosci 2021; 15:749595. [PMID: 34744633 PMCID: PMC8570167 DOI: 10.3389/fncel.2021.749595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/23/2021] [Indexed: 11/13/2022] Open
Abstract
The COVID-19 pandemic imposed a series of behavioral changes that resulted in increased social isolation and a more sedentary life for many across all age groups, but, above all, for the elderly population who are the most vulnerable to infections and chronic neurodegenerative diseases. Systemic inflammatory responses are known to accelerate neurodegenerative disease progression, which leads to permanent damage, loss of brain function, and the loss of autonomy for many aged people. During the COVID-19 pandemic, a spectrum of inflammatory responses was generated in affected individuals, and it is expected that the elderly patients with chronic neurodegenerative diseases who survived SARSCoV-2 infection, it will be found, sooner or later, that there is a worsening of their neurodegenerative conditions. Using mouse prion disease as a model for chronic neurodegeneration, we review the effects of social isolation, sedentary living, and viral infection on the disease progression with a focus on sickness behavior and on the responses of microglia and astrocytes. Focusing on aging, we discuss the cellular and molecular mechanisms related to immunosenescence in chronic neurodegenerative diseases and how infections may accelerate their progression.
Collapse
Affiliation(s)
- Amanda de Oliveira Ferreira Leite
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - João Bento Torres Neto
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - Renata Rodrigues dos Reis
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - Luciane Lobato Sobral
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - Aline Cristine Passos de Souza
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - Nonata Trévia
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - Roseane Borner de Oliveira
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - Nara Alves de Almeida Lins
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - Daniel Guerreiro Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém, Brazil
| | | | | | | | - Dora Brites
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Cristovam Wanderley Picanço Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| |
Collapse
|
42
|
Cai ZY, Fu MD, Liu K, Duan XC. Therapeutic effect of Keap1-Nrf2-ARE pathway-related drugs on age-related eye diseases through anti-oxidative stress. Int J Ophthalmol 2021; 14:1260-1273. [PMID: 34414093 DOI: 10.18240/ijo.2021.08.19] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 12/29/2020] [Indexed: 12/11/2022] Open
Abstract
Age-related eye diseases, including cataract, glaucoma, diabetic retinopathy (DR), and age-related macular degeneration (AMD), are the leading causes of vision loss in the world. Several studies have shown that the occurrence and development of these diseases have an important relationship with oxidative stress in the eye. The Keap1-Nrf2-ARE pathway is a classical pathway that resists oxidative stress and inflammation in the body. This pathway is also active in the development of age-related eye diseases. A variety of drugs have been shown to treat age-related eye diseases through the Keap1-Nrf2-ARE (Kelch-like ECH-Associating protein 1- nuclear factor erythroid 2 related factor 2-antioxidant response element) pathway. This review describes the role of oxidative stress in the development of age-related eye diseases, the function and regulation of the Keap1-Nrf2-ARE pathway, and the therapeutic effects of drugs associated with this pathway on age-related eye diseases.
Collapse
Affiliation(s)
- Zi-Yan Cai
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha 410011, Hunan Province, China
| | - Meng-Die Fu
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha 410011, Hunan Province, China
| | - Ke Liu
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha 410011, Hunan Province, China
| | - Xuan-Chu Duan
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha 410011, Hunan Province, China.,Department of Ophthalmology, Changsha Aier Eye Hospital, Changsha 410011, Hunan Province, China
| |
Collapse
|
43
|
Modulation of Nrf2 and NF-κB Signaling Pathways by Naturally Occurring Compounds in Relation to Cancer Prevention and Therapy. Are Combinations Better Than Single Compounds? Int J Mol Sci 2021; 22:ijms22158223. [PMID: 34360990 PMCID: PMC8348704 DOI: 10.3390/ijms22158223] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 12/14/2022] Open
Abstract
Nrf2 (nuclear factor erythroid 2-related factor 2) and NF-κB (nuclear factor–kappa B) signaling pathways play a central role in suppressing or inducing inflammation and angiogenesis processes. Therefore, they are involved in many steps of carcinogenesis through cooperation with multiple signaling molecules and pathways. Targeting both transcription factors simultaneously may be considered an equally important strategy for cancer chemoprevention and therapy. Several hundreds of phytochemicals, mainly edible plant and vegetable components, were shown to activate Nrf2 and mediate antioxidant response. A similar number of phytochemicals was revealed to affect NF-κB. While activation of Nrf2 and inhibition of NF-κB may protect normal cells against cancer initiation and promotion, enhanced expression and activation in cancer cells may lead to resistance to conventional chemo- or radiotherapy. Most phytochemicals, through different mechanisms, activate Nrf2, but others, such as luteolin, can act as inhibitors of both Nrf2 and NF-κB. Despite many experimental data confirming the above mechanisms currently, limited evidence exists demonstrating such activity in humans. Combinations of phytochemicals resembling that in a natural food matrix but allowing higher concentrations may improve their modulating effect on Nrf2 and NF-κB and ultimately cancer prevention and therapy. This review presents the current knowledge on the effect of selected phytochemicals and their combinations on Nrf2 and NF-κB activities in the above context.
Collapse
|
44
|
Natural compounds modulate the autophagy with potential implication of stroke. Acta Pharm Sin B 2021; 11:1708-1720. [PMID: 34386317 PMCID: PMC8343111 DOI: 10.1016/j.apsb.2020.10.018] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/12/2020] [Accepted: 08/21/2020] [Indexed: 12/12/2022] Open
Abstract
Stroke is considered a leading cause of mortality and neurological disability, which puts a huge burden on individuals and the community. To date, effective therapy for stroke has been limited by its complex pathological mechanisms. Autophagy refers to an intracellular degrading process with the involvement of lysosomes. Autophagy plays a critical role in maintaining the homeostasis and survival of cells by eliminating damaged or non-essential cellular constituents. Increasing evidence support that autophagy protects neuronal cells from ischemic injury. However, under certain circumstances, autophagy activation induces cell death and aggravates ischemic brain injury. Diverse naturally derived compounds have been found to modulate autophagy and exert neuroprotection against stroke. In the present work, we have reviewed recent advances in naturally derived compounds that regulate autophagy and discussed their potential application in stroke treatment.
Collapse
Key Words
- AD, Alzheimer's disease
- ALS, amyotrophic lateral sclerosis
- AMPK, 5′-adenosine monophosphate-activated protein kinase
- ATF6, activating transcription factor 6
- ATG, autophagy related genes
- Autophagy
- BCL-2, B-cell lymphoma 2
- BNIP3L, BCL2/adenovirus
- COPII, coat protein complex II
- Cerebral ischemia
- ER, endoplasmic reticulum
- FOXO, forkhead box O
- FUNDC1, FUN14 domain containing 1
- GPCR, G-protein coupled receptor
- HD, Huntington's disease
- IPC, ischemic preconditioning
- IRE1, inositol-requiring enzyme 1
- JNK, c-Jun N-terminal kinase
- LAMP, lysosomal-associated membrane protein
- LC3, light chain 3
- LKB1, liver kinase B1
- Lysosomal activation
- Mitochondria
- Mitophagy
- Natural compounds
- Neurological disorders
- Neuroprotection
- OGD/R, oxygen and glucose deprivation-reperfusion
- PD, Parkinson's disease
- PERK, protein kinase R (PKR)-like endoplasmic reticulum kinase
- PI3K, phosphatidylinositol 3-kinase
- ROS, reactive oxygen species
- SQSTM1, sequestosome 1
- TFEB, transcription factor EB
- TIGAR, TP53-induced glycolysis and apoptosis regulator
- ULK, Unc-51- like kinase
- Uro-A, urolithin A
- eIF2a, eukaryotic translation-initiation factor 2
- mTOR, mechanistic target of rapamycin
- ΔΨm, mitochondrial membrane potential
Collapse
|
45
|
Deficiency of optineurin enhances osteoclast differentiation by attenuating the NRF2-mediated antioxidant response. Exp Mol Med 2021; 53:667-680. [PMID: 33864025 PMCID: PMC8102640 DOI: 10.1038/s12276-021-00596-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 01/26/2021] [Accepted: 02/03/2021] [Indexed: 02/02/2023] Open
Abstract
Abnormally increased resorption contributes to bone degenerative diseases such as Paget's disease of bone (PDB) through unclear mechanisms. Recently, the optineurin (OPTN) gene has been implicated in PDB, and global OPTN knockout mice (Optn-/-) were shown to exhibit increased formation of osteoclasts (osteoclastogenesis). Growing evidence, including our own, has demonstrated that intracellular reactive oxygen species (ROS) stimulated by receptor activator of nuclear factor kappa-B ligand (RANKL) can act as signaling molecules to promote osteoclastogenesis. Here, we report that OPTN interacts with nuclear factor erythroid-derived factor 2-related factor 2 (NRF2), the master regulator of the antioxidant response, defining a pathway through which RANKL-induced ROS could be regulated for osteoclastogenesis. In this study, monocytes from Optn-/- and wild-type (Optn+/+) mice were utilized to differentiate into osteoclasts, and both qRT-PCR and tartrate-resistant acid phosphatase (TRAP) staining showed that the Optn-/- monocytes exhibited enhanced osteoclastogenesis compared to the Optn+/+ cells. CellROX® staining, qRT-PCR, and Western blotting indicated that OPTN deficiency reduced the basal expression of Nrf2, inhibited the expression of NRF2-responsive antioxidants, and increased basal and RANKL-induced intracellular ROS levels, leading to enhanced osteoclastogenesis. Coimmunoprecipitation (co-IP) showed direct interaction, and immunofluorescence staining showed perinuclear colocalization of the OPTN-NRF2 granular structures during differentiation. Finally, curcumin and the other NRF2 activators attenuated the hyperactive osteoclastogenesis induced by OPTN deficiency. Collectively, our findings reveal a novel OPTN-mediated mechanism for regulating the NRF2-mediated antioxidant response in osteoclasts and extend the therapeutic potential of OPTN in the aging process resulting from ROS-triggered oxidative stress, which is associated with PDB and many other degenerative diseases.
Collapse
|
46
|
Antioxidant and Anti-inflammatory Effect of Cannabidiol Contributes to the Decreased Lipid Peroxidation of Keratinocytes of Rat Skin Exposed to UV Radiation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021. [DOI: 10.1155/2021/6647222] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
There is a great need for compounds with antioxidant and anti-inflammatory properties for protection against UV radiation, which is the most prooxidative physical factor that skin cells are exposed to everyday. Therefore, the aim of the study was to evaluate the mechanism of phytocannabinoid-cannabidiol (CBD) action in vivo on lipid metabolism in keratinocytes of rat skin exposed to UVA/UVB radiation. Our results show that CBD protects keratinocytes against the effects of UVA/UVB radiation by reducing lipid peroxidation products: 4-HNE and 8-isoPGF2α. In addition, CBD significantly increases the level of endocannabinoids, such as anandamide, 2-arachidonylglycerol, and palmitoylethanolamide, and the activation of their receptors CB1/2 or TRPV1. The above changes are due to the protective effect of CBD against the UVA/UVB-induced decrease in the level/activity of superoxide dismutase and the components of the thioredoxin and glutathione systems. CBD also increases the in vivo transcriptional activity of Nrf2 and the expression of its Bach1 inhibitor as well as preventing the UVA/UVB-induced increase in the expression of Nrf2 activators p21, p62, p38, and KAP1 and proinflammatory factors such as NFκB and TNFα. By counteracting oxidative stress and changes in lipid structure in keratinocytes, CBD prevents cellular metabolic disturbances, protecting the epidermis against UV damage.
Collapse
|
47
|
Ahmad S, Khan A, Ali W, Jo MH, Park J, Ikram M, Kim MO. Fisetin Rescues the Mice Brains Against D-Galactose-Induced Oxidative Stress, Neuroinflammation and Memory Impairment. Front Pharmacol 2021; 12:612078. [PMID: 33716741 PMCID: PMC7947859 DOI: 10.3389/fphar.2021.612078] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/08/2021] [Indexed: 12/21/2022] Open
Abstract
Herein, we have evaluated the protective potentials of Fisetin against d-galactose-induced oxidative stress, neuroinflammation, and memory impairment in mice. d-galactose (D-gal) causes neurological impairment by inducing reactive oxygen species (ROS), neuroinflammation, and synaptic dysfunction, whereas fisetin (Fis) is a natural flavonoid having potential antioxidant effects, and has been used against different models of neurodegenerative diseases. Here, the normal mice were injected with D-gal (100 mg/kg/day for 60 days) and fisetin (20 mg/kg/day for 30 days). To elucidate the protective effects of fisetin against d-galactose induced oxidative stress-mediated neuroinflammation, we conducted western blotting, biochemical, behavioral, and immunofluorescence analyses. According to our findings, D-gal induced oxidative stress, neuroinflammation, synaptic dysfunctions, and cognitive impairment. Conversely, Fisetin prevented the D-gal-mediated ROS accumulation, by regulating the endogenous anti-oxidant mechanisms, such as Sirt1/Nrf2 signaling, suppressed the activated p-JNK/NF-kB pathway, and its downstream targets, such as inflammatory cytokines. Hence, our results together with the previous reports suggest that Fisetin may be beneficial in age-related neurological disorders.
Collapse
Affiliation(s)
- Sareer Ahmad
- Division of Life Science and Applied Life Science (BK 21 Plus), College of Natural Sciences, Gyeongsang National University, Jinju, South Korea
| | - Amjad Khan
- Division of Life Science and Applied Life Science (BK 21 Plus), College of Natural Sciences, Gyeongsang National University, Jinju, South Korea
| | - Waqar Ali
- Division of Life Science and Applied Life Science (BK 21 Plus), College of Natural Sciences, Gyeongsang National University, Jinju, South Korea
| | - Myeung Hoon Jo
- Division of Life Science and Applied Life Science (BK 21 Plus), College of Natural Sciences, Gyeongsang National University, Jinju, South Korea
| | - Junsung Park
- Division of Life Science and Applied Life Science (BK 21 Plus), College of Natural Sciences, Gyeongsang National University, Jinju, South Korea
| | - Muhammad Ikram
- Division of Life Science and Applied Life Science (BK 21 Plus), College of Natural Sciences, Gyeongsang National University, Jinju, South Korea
| | - Myeong Ok Kim
- Division of Life Science and Applied Life Science (BK 21 Plus), College of Natural Sciences, Gyeongsang National University, Jinju, South Korea
| |
Collapse
|
48
|
Bennett JP, Onyango IG. Energy, Entropy and Quantum Tunneling of Protons and Electrons in Brain Mitochondria: Relation to Mitochondrial Impairment in Aging-Related Human Brain Diseases and Therapeutic Measures. Biomedicines 2021; 9:225. [PMID: 33671585 PMCID: PMC7927033 DOI: 10.3390/biomedicines9020225] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/18/2021] [Accepted: 02/18/2021] [Indexed: 11/16/2022] Open
Abstract
Adult human brains consume a disproportionate amount of energy substrates (2-3% of body weight; 20-25% of total glucose and oxygen). Adenosine triphosphate (ATP) is a universal energy currency in brains and is produced by oxidative phosphorylation (OXPHOS) using ATP synthase, a nano-rotor powered by the proton gradient generated from proton-coupled electron transfer (PCET) in the multi-complex electron transport chain (ETC). ETC catalysis rates are reduced in brains from humans with neurodegenerative diseases (NDDs). Declines of ETC function in NDDs may result from combinations of nitrative stress (NS)-oxidative stress (OS) damage; mitochondrial and/or nuclear genomic mutations of ETC/OXPHOS genes; epigenetic modifications of ETC/OXPHOS genes; or defects in importation or assembly of ETC/OXPHOS proteins or complexes, respectively; or alterations in mitochondrial dynamics (fusion, fission, mitophagy). Substantial free energy is gained by direct O2-mediated oxidation of NADH. Traditional ETC mechanisms require separation between O2 and electrons flowing from NADH/FADH2 through the ETC. Quantum tunneling of electrons and much larger protons may facilitate this separation. Neuronal death may be viewed as a local increase in entropy requiring constant energy input to avoid. The ATP requirement of the brain may partially be used for avoidance of local entropy increase. Mitochondrial therapeutics seeks to correct deficiencies in ETC and OXPHOS.
Collapse
Affiliation(s)
| | - Isaac G. Onyango
- International Clinical Research Center, St. Anne’s University Hospital, CZ-65691 Brno, Czech Republic;
| |
Collapse
|
49
|
Casein Kinase 2-Interacting Protein-1 Alleviates High Glucose-Reduced Autophagy, Oxidative Stress, and Apoptosis in Retinal Pigment Epithelial Cells via Activating the p62/KEAP1/NRF2 Signaling Pathway. J Ophthalmol 2021; 2021:6694050. [PMID: 33628480 PMCID: PMC7892229 DOI: 10.1155/2021/6694050] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/18/2020] [Accepted: 01/05/2021] [Indexed: 01/11/2023] Open
Abstract
Background Casein kinase 2-interacting protein-1 (CKIP-1) has been proved to be associated with complications of diabetes. Diabetic retinopathy is a main diabetic complication which usually leads to blindness. The current study aims to investigate the role of CKIP-1 in high glucose-treated retinal pigment epithelial (RPE) cells which is a component of blood-retinal barriers. Methods The RPE cells, ARPE-19, are treated with high glucose to mimic the diabetic stimulation. CKIP-1 was overexpressed in ARPE-19 cells to evaluate its effects on autophagy, oxidative stress, and apoptosis induced by high glucose treatment, using Western blot, immunofluorescence, and flow cytometry assays, respectively. Results CKIP-1 was expressed at a lower level in high glucose-treated cells than in normal glucose cells. Overexpression of CKIP-1 enhanced the Nrf2 translocation to the nucleus. Furthermore, high glucose-induced autophagy, oxidative stress, and apoptosis were inhibited after overexpression of CKIP-1. Also, CKIP-1 regulates the p62/Keap1/Nrf2 signaling, which might be the potential mechanism in this model. Conclusion In conclusion, CKIP-1 may be a potential therapeutic target that protects RPE cells from injury and subsequent diabetic retinopathy induced by high glucose.
Collapse
|
50
|
Marmolejo-Martínez-Artesero S, Casas C, Romeo-Guitart D. Endogenous Mechanisms of Neuroprotection: To Boost or Not to Boost. Cells 2021; 10:cells10020370. [PMID: 33578870 PMCID: PMC7916582 DOI: 10.3390/cells10020370] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/05/2021] [Accepted: 02/05/2021] [Indexed: 12/11/2022] Open
Abstract
Postmitotic cells, like neurons, must live through a lifetime. For this reason, organisms/cells have evolved with self-repair mechanisms that allow them to have a long life. The discovery workflow of neuroprotectors during the last years has focused on blocking the pathophysiological mechanisms that lead to neuronal loss in neurodegeneration. Unfortunately, only a few strategies from these studies were able to slow down or prevent neurodegeneration. There is compelling evidence demonstrating that endorsing the self-healing mechanisms that organisms/cells endogenously have, commonly referred to as cellular resilience, can arm neurons and promote their self-healing. Although enhancing these mechanisms has not yet received sufficient attention, these pathways open up new therapeutic avenues to prevent neuronal death and ameliorate neurodegeneration. Here, we highlight the main endogenous mechanisms of protection and describe their role in promoting neuron survival during neurodegeneration.
Collapse
Affiliation(s)
- Sara Marmolejo-Martínez-Artesero
- Department of Cell Biology, Physiology and Immunology, Institut de Neurociències (INc), Universitat Autònoma de Barcelona (UAB), Bellaterra, 08193 Barcelona, Spain;
| | - Caty Casas
- Department of Cell Biology, Physiology and Immunology, Institut de Neurociències (INc), Universitat Autònoma de Barcelona (UAB), Bellaterra, 08193 Barcelona, Spain;
| | - David Romeo-Guitart
- Department of Cell Biology, Physiology and Immunology, Institut de Neurociències (INc), Universitat Autònoma de Barcelona (UAB), Bellaterra, 08193 Barcelona, Spain;
- Laboratory “Hormonal Regulation of Brain Development and Functions”—Team 8, Institut Necker Enfants-Malades (INEM), INSERM U1151, Université Paris Descartes, Sorbonne Paris Cité, 75015 Paris, France
- Correspondence: ; Tel.: +33-01-40-61-53-57
| |
Collapse
|