1
|
Khan WU, Salman M, Ali M, Majid H, Yar MS, Akhtar M, Parvez S, Najmi AK. Neuroprotective Effects of Sulforaphane in a rat model of Alzheimer's Disease induced by Aβ (1-42) peptides. Neurochem Int 2024; 179:105839. [PMID: 39173832 DOI: 10.1016/j.neuint.2024.105839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/13/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
The intricate nature of Alzheimer's disease (AD) has presented significant hurdles in the development of effective interventions. Sulforaphane (SFN) is of interest due to its antioxidative, anti-inflammatory, and neuroprotective properties, which could address various aspects of AD pathology. This study explores the potential of SFN in a rat model of AD induced by Aβ (1-42) peptides. AD symptoms were triggered in rats by injecting Aβ (1-42) peptides directly into their cerebral ventricles. SFN (10 mg/kg and 20 mg/kg), Trigonelline (10 mg/kg), and Pioglitazone (10 mg/kg) were administered in Aβ (1-42) treated animals. Behavioral assessments were performed using the Novel Object Recognition tests. Various biochemical parameters, such as soluble Aβ (1-42), IRS-S312, GSK-3β, TNF-α, acetylcholinesterase, nitrite levels, lipid peroxidation, and reduced glutathione activity, were quantified using ELISA kits and spectrophotometric assays. Histopathological analyses included Hematoxylin and Eosin, Crystal Violet, Congo red, and IRS-1 Immunohistochemistry staining. Quantification was performed to assess neuronal loss and Aβ plaque burden. The novelty of this study lies in its comprehensive evaluation of SFN's impact on multiple AD-related pathways at dual doses. The Novel Object Recognition test revealed that SFN, especially at higher doses, improved memory deficits induced by Aβ (1-42). Biochemically, SFN reduced hippocampal Aβ levels, IRS-S312, GSK-3β, TNF-α, and acetylcholinesterase activity, while increasing glutathione levels, all in a dose-dependent manner. Histopathological analyses further confirmed SFN's protective role against Aβ-induced neuronal damage, amyloidosis, and changes in insulin signaling. These results highlight SFN's potential as a multifaceted therapeutic agent for AD, offering a promising avenue for treatment due to its antioxidative, anti-inflammatory, and neuroprotective properties. The inclusion of combination treatments with Trigonelline and Pioglitazone alongside SFN offers insights into potential synergistic effects, which could pave the way for developing combination therapies for AD.
Collapse
Affiliation(s)
- Wasi Uzzaman Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Mohd Salman
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Mubashshir Ali
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Haya Majid
- Department of Translational and Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - M Shahar Yar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Mohd Akhtar
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Suhel Parvez
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
2
|
Calabrese V, Osakabe N, Siracusa R, Modafferi S, Di Paola R, Cuzzocrea S, Jacob UM, Fritsch T, Abdelhameed AS, Rashan L, Wenzel U, Franceschi C, Calabrese EJ. Transgenerational hormesis in healthy aging and antiaging medicine from bench to clinics: Role of food components. Mech Ageing Dev 2024; 220:111960. [PMID: 38971236 DOI: 10.1016/j.mad.2024.111960] [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: 04/27/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/08/2024]
Abstract
Neurodegenerative diseases have multifactorial pathogenesis, mainly involving neuroinflammatory processes. Finding drugs able to treat these diseases, expecially because for most of these diseases there are no effective drugs, and the current drugs cause undesired side effects, represent a crucial point. Most in vivo and in vitro studies have been concentrated on various aspects related to neurons (e.g. neuroprotection), however, there has not been focus on the prevention of early stages involving glial cell activation and neuroinflammation. Recently, it has been demonstrated that nutritional phytochemicals including polyphenols, the main active constituents of the Mediterranean diet, maintain redox balance and neuroprotection through the activation of hormetic vitagene pathway. Recent lipidomics data from our laboratory indicate mushrooms as strong nutritional neuronutrients with strongly activity against neuroinflammation in Meniere' diseaseas, a model of cochleovestibular neural degeneration, as well as in animal model of traumatic brain injury, or rotenone induced parkinson's disease. Moreover, Hidrox®, an aqueous extract of olive containing hydroxytyrosol, and Boswellia, acting as Nrf2 activators, promote resilience by enhancing the redox potential, and thus, regulate through hormetic mechanisms, cellular stress response mechanisms., Thus, modulation of cellular stress pathways, in particular vitagenes system, may be an innovative approach for therapeutic intervention in neurodegenerative disorders.
Collapse
Affiliation(s)
- Vittorio Calabrese
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
| | - Naomi Osakabe
- Department of Bioscience and Engineering, Shibaura Institute Technology, Tokyo, Japan.
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina 98166, Italy
| | - Sergio Modafferi
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Rosanna Di Paola
- Department of Veterinary Sciences, University of Messina, Messina 98168, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina 98166, Italy
| | | | | | - Ali S Abdelhameed
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Luay Rashan
- Biodiversity Unit, Dhofar University, Salalah, Oman
| | - Uwe Wenzel
- Institut für Ernährungswissenschaft, Justus Liebig Universitat Giessen, Germany
| | | | - Edward J Calabrese
- Department of Environmental Health Sciences, Morrill I, N344, University of Massachusetts, Amherst, MA 01003, USA
| |
Collapse
|
3
|
Cascajosa-Lira A, Prieto AI, Pichardo S, Jos A, Cameán AM. Protective effects of sulforaphane against toxic substances and contaminants: A systematic review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155731. [PMID: 38824824 DOI: 10.1016/j.phymed.2024.155731] [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: 02/29/2024] [Revised: 05/03/2024] [Accepted: 05/10/2024] [Indexed: 06/04/2024]
Abstract
BACKGROUND Sulforaphane (SFN) is a dietary isothiocyanate, derived from glucoraphanin, present in cruciferous vegetables belonging to the Brassica genus. It is a biologically active phytochemical that acts as a nuclear factor erythroid 2-related factor 2 (Nrf2) inducer. Thus, it has been reported to have multiple protective functions including anticancer responses and protection against a toxic agent's action. PURPOSE The present work systematically reviewed and synthesised the protective properties of sulforaphane against a toxic agent. This review reveals the mechanism of the action of SFN in each organ or system. METHODS The PRISMA guideline was followed in this sequence: researched literature, organised retrieved documents, abstracted relevant information, assessed study quality and bias, synthesised data, and prepared a comprehensive report. Searches were conducted on Science Direct and PubMed using the keywords "Sulforaphane" AND ("protective effects" OR "protection against"). RESULTS Reports showed that liver and the nervous system are the target organs on which attention was focused, and this might be due to the key role of oxidative stress in liver and neurodegenerative diseases. However, protective activities have also been demonstrated in the lungs, heart, immune system, kidneys, and endocrine system. SFN exerts its protective effects by activating the Nrf2 pathway, which enhances antioxidant defenses and reduces oxidative stress. It also suppresses inflammation by decreasing interleukin production. Moreover, SFN inhibits apoptosis by preventing caspase 3 cleavage and increasing Bcl2 levels. Overall, SFN demonstrates multifaceted mechanisms to counteract the adverse effects of toxic agents. CONCLUSION SFN has potential clinical applications as a chemoprotective agent. Nevertheless, more studies are necessary to set the safe doses of SFN in humans.
Collapse
Affiliation(s)
- Antonio Cascajosa-Lira
- Área de Toxicología, Facultad de Farmacia, Universidad de Sevilla, Profesor García González n 2, 41012 Seville, España.
| | - Ana I Prieto
- Área de Toxicología, Facultad de Farmacia, Universidad de Sevilla, Profesor García González n 2, 41012 Seville, España.
| | - Silvia Pichardo
- Área de Toxicología, Facultad de Farmacia, Universidad de Sevilla, Profesor García González n 2, 41012 Seville, España.
| | - Angeles Jos
- Área de Toxicología, Facultad de Farmacia, Universidad de Sevilla, Profesor García González n 2, 41012 Seville, España.
| | - Ana M Cameán
- Área de Toxicología, Facultad de Farmacia, Universidad de Sevilla, Profesor García González n 2, 41012 Seville, España.
| |
Collapse
|
4
|
Jin Y, Li X, Wei C, Yuan Q. Effects of exercise-targeted hippocampal PDE-4 methylation on synaptic plasticity and spatial learning/memory impairments in D-galactose-induced aging rats. Exp Brain Res 2024; 242:309-320. [PMID: 38052997 PMCID: PMC10805951 DOI: 10.1007/s00221-023-06749-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 11/15/2023] [Indexed: 12/07/2023]
Abstract
Physical exercise reduces the effects of aging and cognitive decline by improving synaptic plasticity and spatial learning. However, the underlying neurobiological mechanisms are unclear. A total of 45 Male SPF Sprague-Dawley rats were acclimatized and then allocated into three groups, 15 in each group: the saline control (DC) group, D-gal-induced aging (DA) group, and D-gal-induced aging + exercise (DE) group. Six weeks of intraperitoneal injections of D-gal at a concentration of 100 mg/kg body weight/d was injected to establish model of aging in the DA and DE groups. Morris water maze test was implemented to evaluate the hippocampus related cognition. SOD activity and MDA was tested to assess the aging in all groups. H&E and Nissl staining was used to observe the histopathological changes of hippocampal neurons in aging rats. Quantitative real-time polymerase chain reaction, western blotting and immunofluorescence staining techniques were used to investigate the expression of synaptic genes and proteins in the hippocampus. Massarray methylation system was employed to measure the PDE-4 gene methylation level in rat hippocampal tissues. Our results demonstrated that exercise intervention improves cognitive function in D-gal-induced aging rats. The methylation of CpG sites in PDE-4 in the hippocampus was significantly increased. The physical exercise significantly increased PDE-4 gene methylation and effectively decreased PDE-4 gene and protein expression. These beneficial behavioral and morphological effects were attributed to PDE-4 methylation, which was activated cAMP/PKA/CREB pathway and improved synaptic plasticity. Exercise induced PDE-4 methylation is key mechanism underpinning the amelioration of learning/memory impairment, suggesting the potential efficacy of physical exercise training in delaying brain aging.
Collapse
Affiliation(s)
- Yu Jin
- School of Sport Medicine and Health, Chengdu Sport University, Chengdu, 610041, China
| | - Xue Li
- School of Sport Medicine and Health, Chengdu Sport University, Chengdu, 610041, China.
| | - Changling Wei
- School of Sport Medicine and Health, Chengdu Sport University, Chengdu, 610041, China
| | - Qiongjia Yuan
- School of Sport Medicine and Health, Chengdu Sport University, Chengdu, 610041, China
| |
Collapse
|
5
|
Yang J, He L, Dai S, Zheng H, Cui X, Ou J, Zhang X. Therapeutic efficacy of sulforaphane in autism spectrum disorders and its association with gut microbiota: animal model and human longitudinal studies. Front Nutr 2024; 10:1294057. [PMID: 38260076 PMCID: PMC10800504 DOI: 10.3389/fnut.2023.1294057] [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: 09/14/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024] Open
Abstract
Introduction Sulforaphane (SFN) has been found to alleviate complications linked with several diseases by regulating gut microbiota (GM), while the effect of GM on SFN for autism spectrum disorders (ASD) has not been studied. Therefore, this study aimed to investigate the relationship between the effects of SFN on childhood ASD and GM through animal model and human studies. Methods We evaluated the therapeutic effects of SFN on maternal immune activation (MIA) induced ASD-like rat model and pediatric autism patients using three-chamber social test and OSU Autism Rating Scale-DSM-IV (OARS-4), respectively, with parallel GM analysis using 16SrRNA sequencing. Results SFN significantly improved the sniffing times of ASD-like rats in the three-chamber test. For human participants, the average verbal or non-verbal communication (OSU-CO) scores of SFN group had changed significantly at the 12-wk endpoint. SFN was safe and no serious side effects after taking. GM changes were similar for both ASD-like rats and ASD patients, such as consistent changes in order Bacillales, family Staphylococcaceae and genus Staphylococcus. Although the gut microbiota composition was significantly altered in SFN-treated ASD-like rats, the alteration of GM was not evident in ASD patients after 12 weeks of SFN treatment. However, in the network analysis, we found 25 taxa correlated with rats' social behavior, 8 of which were associated with SFN treatment in ASD-like rats, For ASD patients, we found 35 GM abundance alterations correlated with improvements in ASD symptoms after SFN treatment. Moreover, family Pasteurellaceae and genus Haemophilus were found to be associated with SFN administration in the network analyses in both ASD-like rats and ASD patients. Discussion These findings suggest that SFN could provide a novel avenue for preventing and treating ASD, and its therapeutic effects might be related to gut microbiota.
Collapse
Affiliation(s)
| | | | | | | | | | - Jianjun Ou
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xiaojie Zhang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
6
|
Otoo RA, Allen AR. Sulforaphane's Multifaceted Potential: From Neuroprotection to Anticancer Action. Molecules 2023; 28:6902. [PMID: 37836745 PMCID: PMC10574530 DOI: 10.3390/molecules28196902] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/16/2023] [Accepted: 08/30/2023] [Indexed: 10/15/2023] Open
Abstract
Sulforaphane (SFN) is a naturally occurring compound found in cruciferous vegetables such as broccoli and cauliflower. It has been widely studied for its potential as a neuroprotective and anticancer agent. This review aims to critically evaluate the current evidence supporting the neuroprotective and anticancer effects of SFN and the potential mechanisms through which it exerts these effects. SFN has been shown to exert neuroprotective effects through the activation of the Nrf2 pathway, the modulation of neuroinflammation, and epigenetic mechanisms. In cancer treatment, SFN has demonstrated the ability to selectively induce cell death in cancer cells, inhibit histone deacetylase, and sensitize cancer cells to chemotherapy. SFN has also shown chemoprotective properties through inhibiting phase I metabolizing enzymes, modulating phase II xenobiotic-metabolizing enzymes, and targeting cancer stem cells. In addition to its potential as a therapeutic agent for neurological disorders and cancer treatment, SFN has shown promise as a potential treatment for cerebral ischemic injury and intracranial hemorrhage. Finally, the ongoing and completed clinical trials on SFN suggest potential therapeutic benefits, but more research is needed to establish its effectiveness. Overall, SFN holds significant promise as a natural compound with diverse therapeutic applications.
Collapse
Affiliation(s)
- Raymond A. Otoo
- Division of Radiation Health, University of Arkansas for Medical Sciences, 4301 West Markham, Suite 441B-2, Little Rock, AR 72205, USA;
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, 4301 West Markham, Suite 441B-2, Little Rock, AR 72205, USA
- Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, 4301 West Markham, Suite 441B-2, Little Rock, AR 72205, USA
| | - Antiño R. Allen
- Division of Radiation Health, University of Arkansas for Medical Sciences, 4301 West Markham, Suite 441B-2, Little Rock, AR 72205, USA;
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, 4301 West Markham, Suite 441B-2, Little Rock, AR 72205, USA
- Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, 4301 West Markham, Suite 441B-2, Little Rock, AR 72205, USA
| |
Collapse
|
7
|
Khan F, Joshi A, Devkota HP, Subramaniyan V, Kumarasamy V, Arora J. Dietary glucosinolates derived isothiocyanates: chemical properties, metabolism and their potential in prevention of Alzheimer's disease. Front Pharmacol 2023; 14:1214881. [PMID: 37554984 PMCID: PMC10404612 DOI: 10.3389/fphar.2023.1214881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 07/04/2023] [Indexed: 08/10/2023] Open
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia affecting millions of people worldwide. It is a progressive, irreversible, and incurable neurodegenerative disorder that disrupts the synaptic communication between millions of neurons, resulting in neuronal death and functional loss due to the abnormal accumulation of two naturally occurring proteins, amyloid β (Aβ) and tau. According to the 2018 World Alzheimer's Report, there is no single case of an Alzheimer's survivor; even 1 in 3 people die from Alzheimer's disease, and it is a growing epidemic across the globe fruits and vegetables rich in glucosinolates (GLCs), the precursors of isothiocyanates (ITCs), have long been known for their pharmacological properties and recently attracted increased interest for the possible prevention and treatment of neurodegenerative diseases. Epidemiological evidence from systematic research findings and clinical trials suggests that nutritional and functional dietary isothiocyanates interfere with the molecular cascades of Alzheimer's disease pathogenesis and prevent neurons from functional loss. The aim of this review is to explore the role of glucosinolates derived isothiocyanates in various molecular mechanisms involved in the progression of Alzheimer's disease and their potential in the prevention and treatment of Alzheimer's disease. It also covers the chemical diversity of isothiocyanates and their detailed mechanisms of action as reported by various in vitro and in vivo studies. Further clinical studies are necessary to evaluate their pharmacokinetic parameters and effectiveness in humans.
Collapse
Affiliation(s)
- Farhana Khan
- Laboratory of Bio-Molecular Technology, Department of Botany, Mohanlal Sukhadia University, Udaipur, Rajasthan, India
| | - Abhishek Joshi
- Laboratory of Bio-Molecular Technology, Department of Botany, Mohanlal Sukhadia University, Udaipur, Rajasthan, India
| | - Hari Prasad Devkota
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Vetriselvan Subramaniyan
- Department of Pharmacology, Center for Transdisciplinary Research, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Vinoth Kumarasamy
- Department of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Jaya Arora
- Laboratory of Bio-Molecular Technology, Department of Botany, Mohanlal Sukhadia University, Udaipur, Rajasthan, India
| |
Collapse
|
8
|
Liu X, Tang AL, Chen J, Gao N, Zhang G, Xiao C. RIPK1 in the inflammatory response and sepsis: Recent advances, drug discovery and beyond. Front Immunol 2023; 14:1114103. [PMID: 37090690 PMCID: PMC10113447 DOI: 10.3389/fimmu.2023.1114103] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/24/2023] [Indexed: 04/25/2023] Open
Abstract
Cytokine storms are an important mechanism of sepsis. TNF-α is an important cytokine. As a regulator of TNF superfamily receptors, RIPK1 not only serves as the basis of the scaffold structure in complex I to promote the activation of the NF-κB and MAPK pathways but also represents an important protein in complex II to promote programmed cell death. Ubiquitination of RIPK1 is an important regulatory function that determines the activation of cellular inflammatory pathways or the activation of death pathways. In this paper, we introduce the regulation of RIPK1, RIPK1 PANoptosome's role in Inflammatory and sepsis, and perspectives.
Collapse
Affiliation(s)
- Xiaoyu Liu
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - A-Ling Tang
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Jie Chen
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nan Gao
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guoqiang Zhang
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
| | - Cheng Xiao
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
9
|
Liu S, Deng S, Ding Y, Flores JJ, Zhang X, Jia X, Hu X, Peng J, Zuo G, Zhang JH, Gong Y, Tang J. Secukinumab attenuates neuroinflammation and neurobehavior defect via PKCβ/ERK/NF-κB pathway in a rat model of GMH. Exp Neurol 2023; 360:114276. [PMID: 36402169 DOI: 10.1016/j.expneurol.2022.114276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 11/05/2022] [Accepted: 11/11/2022] [Indexed: 11/18/2022]
Abstract
AIMS Germinal matrix hemorrhage (GMH) is a disastrous clinical event for newborns. Neuroinflammation plays an important role in the development of neurological deficits after GMH. The purpose of this study is to investigate the anti-inflammatory role of secukinumab after GMH and its underlying mechanisms involving PKCβ/ERK/NF-κB signaling pathway. METHODS A total of 154 Sprague-Dawley P7 rat pups were used. GMH was induced by intraparenchymal injection of bacterial collagenase. Secukinumab was administered intranasally post-GMH. PKCβ activator PMA and p-ERK activator Ceramide C6 were administered intracerebroventricularly at 24 h prior to GMH induction, respectively. Neurobehavioral tests, western blot and immunohistochemistry were used to evaluate the efficacy of Secukinumab in both short-term and long-term studies. RESULTS Endogenous IL-17A, IL-17RA, PKCβ and p-ERK were increased after GMH. Secukinumab treatment improved short- and long-term neurological outcomes, reduced the synthesis of MPO and Iba-1 in the perihematoma area, and inhibited the synthesis of proinflammatory factors, such as NF-κB, IL-1β, TNF-α and IL-6. Additionally, PMA and ceramide C6 abolished the beneficial effects of Secukinumab. CONCLUSION Secukinumab treatment suppressed neuroinflammation and attenuated neurological deficits after GMH, which was mediated through the downregulation of the PKCβ/ERK/NF-κB pathway. Secukinumab treatment may provide a promising therapeutic strategy for GMH patients.
Collapse
Affiliation(s)
- Shengpeng Liu
- Department of Pediatrics, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, China; Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; Department of Pediatrics, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Guangdong, China
| | - Shuixiang Deng
- Department of Critical Care Medicine, HuaShan Hospital, Fudan University, Shanghai 200040, China; Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Yan Ding
- Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Jerry J Flores
- Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Xiaoli Zhang
- Department of Pediatrics, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, China; Department of Pediatrics, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Guangdong, China
| | - Xiaojing Jia
- Department of Pediatrics, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, China; Department of Pediatrics, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Guangdong, China
| | - Xiao Hu
- Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Jun Peng
- Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Gang Zuo
- Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Ye Gong
- Department of Critical Care Medicine, HuaShan Hospital, Fudan University, Shanghai 200040, China; Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Jiping Tang
- Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| |
Collapse
|
10
|
Zhang J, Zha Y, Jiao Y, Li Y, Zhang S. Protective role of cezanne in doxorubicin-induced cardiotoxicity by inhibiting autophagy, apoptosis and oxidative stress. Toxicology 2023; 485:153426. [PMID: 36639017 DOI: 10.1016/j.tox.2023.153426] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/30/2022] [Accepted: 01/09/2023] [Indexed: 01/11/2023]
Abstract
Doxorubicin (DOX) is frequently used in clinical practice for its broad-spectrum effects. However, its benefit is limited by a series of complications, including excessive apoptosis and autophagy of cardiomyocytes, overproduction of reactive oxygen species (ROS) and high level of oxidative stress. As a new protein, OTU domain-containing 7B (OTUD7B), also called Cezanne, has been reported to regulate many pathological processes. However, whether it plays a role in DOX-induced cardiotoxicity is still unclear. We discovered that the Cezanne level was significantly increased in DOX-treated neonatal rat cardiomyocytes (NRCMs) and C57BL/6 J mice hearts. In vitro, the knockdown of Cezanne with adenovirus in NRCMs significantly worsened DOX-induced apoptosis, autophagy and oxidative stress, while Cezanne overexpression showed opposite results. In vivo, the overexpression of Cezanne using cardiomyocyte-targeted adeno-associated virus 9 (AAV9) significantly reduced cardiomyocyte apoptosis, autophagy and oxidative stress level when C57BL/6 J mice were subjected to DOX. Mechanistically, the overexpression of Cezanne significantly reversed the in-activation of the PI3K/AKT/mTOR pathway induced by DOX, while the inhibitors of this pathway abolished the effect of Cezanne, suggesting that the PI3K/AKT/mTOR pathway plays a role in the protective function of Cezanne. These findings indicate that Cezanne could ameliorate DOX-induced cardiotoxicity by attenuating the apoptosis and autophagy of cardiomyocytes and decreasing the level of oxidative stress.
Collapse
Affiliation(s)
- Jiayan Zhang
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Yafang Zha
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Yuheng Jiao
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Yanyan Li
- Department of Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai 200092, China
| | - Song Zhang
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China.
| |
Collapse
|
11
|
Song H, Wang YH, Zhou HY, Cui KM. Sulforaphane alleviates LPS-induced inflammatory injury in ARPE-19 cells by repressing the PWRN2/NF-kB pathway. Immunopharmacol Immunotoxicol 2022; 44:868-876. [PMID: 35766158 DOI: 10.1080/08923973.2022.2090954] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Age-related macular degeneration (AMD) is the leading cause of blindness in the elderly population and its pathogenesis has been associated with inflammatory damage to retinal pigment epithelial (RPE) cells. Here, we explored the ability of sulforaphane to protect ARPE-19 cells from lipopolysaccharide (LPS)-induced inflammatory injury and elucidated the underlying molecular mechanism. METHODS Cell viability, apoptosis, inflammation, PWRN2 expression, nuclear transcription factor-kappa B (NF-kB) activity, and the interaction between PWRN2 and the IkBa protein were assessed in RPE cells under- or over-expressing PWRN2 that had been treated with LPS and sulforaphane. RESULTS Overexpression of PWRN2 in LPS-treated cells promoted NF-kB activation by interacting with IkBa, thus reducing cell viability. In contrast, PWRN2 downregulation repressed LPS-induced NF-kB activation and apoptosis in RPE cells. Similarly, sulforaphane downregulated PWRN2 and inhibited NF-kB activation in a concentration-dependent manner. Conversely, PWRN2 overexpression or NF-kB upregulation weakened the anti-inflammatory effects of sulforaphane. CONCLUSION Our results suggest that sulforaphane protects RPE cells from LPS-induced inflammatory injury by suppressing the PWRN2/NF-kB pathway.
Collapse
Affiliation(s)
- Hui Song
- Eye Center, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, P.R. China
| | - Ying-Hao Wang
- Eye Center, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, P.R. China
| | - Hai-Yan Zhou
- Eye Center, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, P.R. China
| | - Kun-Ming Cui
- Eye Center, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, P.R. China
| |
Collapse
|
12
|
Xie W, Tian S, Yang J, Cai S, Jin S, Zhou T, Wu Y, Chen Z, Ji Y, Cui J. OTUD7B deubiquitinates SQSTM1/p62 and promotes IRF3 degradation to regulate antiviral immunity. Autophagy 2022; 18:2288-2302. [PMID: 35100065 PMCID: PMC9542415 DOI: 10.1080/15548627.2022.2026098] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Deubiquitination plays an important role in the regulation of the crosstalk between macroautophagy/autophagy and innate immune signaling, yet its regulatory mechanisms are not fully understood. Here we identify the deubiquitinase OTUD7B as a negative regulator of antiviral immunity by targeting IRF3 (interferon regulatory factor 3) for selective autophagic degradation. Mechanistically, OTUD7B interacts with IRF3, and activates IRF3-associated cargo receptor SQSTM1/p62 (sequestosome 1) by removing its K63-linked poly-ubiquitin chains at lysine 7 (K7) to enhance SQSTM1 oligomerization. Moreover, viral infection increased the expression of OTUD7B, which forms a negative feedback loop by promoting IRF3 degradation to balance type I interferon (IFN) signaling. Taken together, our study reveals a specific role of OTUD7B in mediating the activation of cargo receptors in a substrate-dependent manner, which could be a potential target against excessive immune responses.Abbreviations: Baf A1: bafilomycin A1; CGAS: cyclic GMP-AMP synthase; DDX58/RIG-I: DExD/H-box helicase 58; DSS: dextran sodium sulfate; DUBs: deubiquitinating enzymes; GFP: green fluorescent protein; IFN: interferon; IKKi: IKBKB/IkappaB kinase inhibitor; IRF3: interferon regulatory factor 3; ISGs: interferon-stimulated genes; MAVS: mitochondrial antiviral signaling protein; MOI: multiplicity of infection; PAMPs: pathogen-associated molecular patterns; SeV: Sendai virus; siRNA: small interfering RNA; SQSTM1/p62: sequestosome 1; STING1: stimulator of interferon response cGAMP interactor 1; TBK1: TANK binding kinase 1; Ub: ubiquitin; WT: wild-type; VSV: vesicular stomatitis virus.
Collapse
Affiliation(s)
- Weihong Xie
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China,Huizhou Municipal Central Hospital, Huizhou, P.R.China
| | - Shuo Tian
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
| | - Jiahui Yang
- Huizhou Municipal Central Hospital, Huizhou, P.R.China
| | - Sihui Cai
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
| | - Shouheng Jin
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
| | - Tao Zhou
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
| | - Yaoxing Wu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
| | - Zhiyun Chen
- Huizhou Municipal Central Hospital, Huizhou, P.R.China
| | - Yanqin Ji
- Huizhou Municipal Central Hospital, Huizhou, P.R.China,CONTACT Yanqin Ji Huizhou Municipal Central Hospital, Huizhou, Guangdong, 516001, P.R.China
| | - Jun Cui
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China,Jun Cui; School of Life Sciences, Sun Yat-sen University, 132 Waihuan East Road, Guangdong 510006, Guangdong, P.R.China
| |
Collapse
|
13
|
Ding MR, Qu YJ, Hu B, An HM. Signal pathways in the treatment of Alzheimer's disease with traditional Chinese medicine. Biomed Pharmacother 2022; 152:113208. [PMID: 35660246 DOI: 10.1016/j.biopha.2022.113208] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 11/28/2022] Open
Abstract
AIM OF THE REVIEW This study aimed to reveal the classical signal pathways and important potential targets of traditional Chinese medicine (TCM) for treating Alzheimer's disease (AD), and provide support for further investigation on TCM and its active ingredients. MATERIALS AND METHODS Literature survey was conducted using PubMed, Web of Science, Google Scholar, CNKI, and other databases, with "Alzheimer's disease," "traditional Chinese medicine," "medicinal herb," "Chinese herb," and "natural plant" as the primary keywords. RESULTS TCM could modulate signal pathways related to AD pathological progression, including NF-κB, Nrf2, JAK/STAT, ubiquitin-proteasome pathway, autophagy-lysosome pathway-related AMPK/mTOR, GSK-3/mTOR, and PI3K/Akt/mTOR, as well as SIRT1 and PPARα pathway. It could regulate crosstalk between pathways through a multitarget, thus maintaining chronic inflammatory interaction balance, inhibiting oxidative stress damage, regulating ubiquitin-proteasome system function, modulating autophagy, and eventually improving cognitive impairment in patients with AD. CONCLUSION TCM could be multilevel, multitargeted, and multifaceted to prevent and treat AD. In-depth research on the prevention and treatment of AD with TCM could provide new ideas for exploring the pathogenesis of AD and developing new anti-AD drugs.
Collapse
Affiliation(s)
- Min-Rui Ding
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yan-Jie Qu
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Bing Hu
- Institute of Traditional Chinese Medicine in Oncology, Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Hong-Mei An
- Department of Science & Technology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| |
Collapse
|
14
|
Zhou J, Ni W, Ling Y, Lv X, Niu D, Zeng Y, Qiu Y, Si Y, Wang Z, Hu J. Human neural stem cells secretome inhibits lipopolysaccharide-induced neuroinflammation through modulating microglia polarization by activating PPAR-γ. Stem Cells Dev 2022; 31:369-382. [PMID: 35481777 DOI: 10.1089/scd.2022.0081] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Neuroinflammation is one of the typical events in multiple neurodegenerative diseases, whereas microglia are the critical participants in the pathogenesis of neuroinflammation. Several studies suggest that neural stem cells (NSCs) present immunomodulatory benefits due to their paracrine products, which contain mounting trophic factors. In the current study, the anti-inflammatory effects of neural stem cells secretome (NSC-S) on lipopolysaccharide (LPS)-induced neuroinflammatory models were evaluated in vivo and the underlying mechanism was further investigated in vitro. It was revealed that NSC-S significantly attenuated the severity of LPS-induced behaviour disorders and inflammatory response in mice. In vitro studies found that NSC-S significantly promoted the polarization of microglia from proinflammatory M1 to anti-inflammatory M2 phenotype, and reduced the production of proinflammatory cytokines while elevated anti-inflammatory cytokines in BV2 cells. NSC-S promoted peroxisome proliferator-activated receptor gamma (PPAR-γ) pathway activation. However, these effects of NSC-S were abrogated by PPAR-γ inhibitor GW9662. Notably, the fatty acid binding protein 5 (FABP5) in NSC-S may mediate PPAR-γ activation and inflammation remission. In summary, NSC-S promotes the regression of LPS-induced microglia-mediated inflammation through the PPAR-γ pathway. This function might be achieved via FABP5.
Collapse
Affiliation(s)
- Jiqin Zhou
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, , Jiangsu, China;
| | - Wei Ni
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, , Jiangsu, China;
| | - Yating Ling
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, , Jiangsu, China;
| | - Xiaorui Lv
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, , Jiangsu, China;
| | - Dongdong Niu
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, , Jiangsu, China;
| | - Yu Zeng
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, , Jiangsu, China;
| | - Yun Qiu
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, , Jiangsu, China;
| | - Yu Si
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, , Jiangsu, China;
| | - Ziyu Wang
- Health Clinical Laboratories, Health BioMed Co.,Ltd, Ningbo, Zhejiang, China;
| | - Jiabo Hu
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, 301, , Jiangsu, China, 212013;
| |
Collapse
|
15
|
Dana AH, Alejandro SP. Role of sulforaphane in endoplasmic reticulum homeostasis through regulation of the antioxidant response. Life Sci 2022; 299:120554. [PMID: 35452639 DOI: 10.1016/j.lfs.2022.120554] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/10/2022] [Accepted: 04/11/2022] [Indexed: 02/09/2023]
Abstract
Nowadays, the nutraceutical agent sulforaphane (SFN) shows great versatility in turning on different cellular responses. Mainly, this isothiocyanate acts as a master regulator of cellular homeostasis due to its antioxidant response and cytoplasmic, mitochondrial, and endoplasmic reticulum (ER) protein modulation. Even more, SFN acts as an effective strategy to counteract oxidative stress, apoptosis, and ER stress, among others as seen in different injury models. Particularly, ER stress is buffered by the unfolded protein response (UPR) activation, which is the first instance in orchestrating the recovery of ER function. Interestingly, different studies highlight a close interrelationship between ER stress and oxidative stress, two events driven by the accumulation of reactive oxygen species (ROS). This response inevitably perpetuates itself and acts as a vicious cycle that triggers the development of different pathologies, such as cardiovascular diseases, neurodegenerative diseases, and others. Accordingly, it is vital to target ER stress and oxidative stress to increase the effectiveness of clinical therapies used to treat these diseases. Therefore, our study is focused on the role of SFN in preserving cellular homeostasis balance by regulating the ER stress response through the Nrf2-modulated antioxidant pathway.
Collapse
Affiliation(s)
- Arana-Hidalgo Dana
- Department of Cardiovascular Biomedicine, National Institute of Cardiology Ignacio Chávez, Mexico City, Mexico
| | - Silva-Palacios Alejandro
- Department of Cardiovascular Biomedicine, National Institute of Cardiology Ignacio Chávez, Mexico City, Mexico.
| |
Collapse
|