1
|
Wang Y, Li N, Chen X, Zhao Y, Qu L, Cai D. Mechanistic insights into sevoflurane-induced hippocampal neuronal damage and cognitive dysfunction through the NEAT1/Nrf2 signaling axis in aged rats. Cell Biol Toxicol 2024; 41:13. [PMID: 39707048 PMCID: PMC11662051 DOI: 10.1007/s10565-024-09964-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 11/29/2024] [Indexed: 12/23/2024]
Abstract
The use of anesthetics during surgery can cause severe neurological damage and cognitive dysfunction in elderly patients. However, this health issue currently lacks corresponding therapeutic strategies. This research involved the utilization of single-cell RNA sequencing (scRNA-seq) and transcriptomic assessment to pinpoint crucial cell classifications and molecular pathways, as well as the lncRNA expression profiles, that undergo substantial alterations in aged rats experiencing sevoflurane-induced cognitive impairment. The results of our investigation pointed towards the enrichment of differentially expressed genes in neurons within the Nrf2/ARE signaling pathway, alongside an elevated expression of lncRNA NEAT1. Subsequently, by constructing a rat model to induce neuronal dysfunction with sevoflurane and performing experiments both in vivo and in vitro (including TUNEL staining, H&E staining, immunohistochemistry, immunofluorescence, and flow cytometry to assess apoptosis levels), we confirmed that NEAT1 inhibits the Nrf2/ARE/HO-1 pathway-related factors. Sevoflurane promotes oxidative stress and apoptosis in primary hippocampal neurons through the NEAT1/Nrf2/ARE/HO-1 axis. This study elucidates the molecular mechanism by which sevoflurane induces hippocampal neuronal damage and cognitive decline in elderly rats via the regulation of the lncRNA NEAT1/Nrf2 signaling axis. We discovered that upregulation of NEAT1 suppresses the Nrf2 signaling pathway, further inducing neuronal damage and cognitive dysfunction, furnishing an essential citation to grasp the molecular pathways involved in neuronal harm and devising corresponding treatment methodologies.
Collapse
Affiliation(s)
- Yiliang Wang
- Department of Anesthesiology, the First Hospital of China Medical University, Shenyang, 110001, Liaoning, People's Republic of China
- Department of Health Statistics, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, People's Republic of China
| | - Nu Li
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, People's Republic of China
| | - Xiaoyu Chen
- Department of Anesthesiology, General Hospital of Northern Theater Command, Shenyang, 110001, Liaoning, People's Republic of China
| | - Yue Zhao
- Department of Anesthesiology, Second Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, 110001, Liaoning, People's Republic of China
| | - Letian Qu
- Department of Geriatrics, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, People's Republic of China.
| | - Dasheng Cai
- Department of Anesthesiology, the First Hospital of China Medical University, Shenyang, 110001, Liaoning, People's Republic of China.
| |
Collapse
|
2
|
Yu Z, Luo F. The Role of Reactive Oxygen Species in Alzheimer's Disease: From Mechanism to Biomaterials Therapy. Adv Healthc Mater 2024; 13:e2304373. [PMID: 38508583 DOI: 10.1002/adhm.202304373] [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: 12/09/2023] [Revised: 03/13/2024] [Indexed: 03/22/2024]
Abstract
Alzheimer's disease (AD) is a chronic, insidious, and progressive neurodegenerative disease that remains a clinical challenge for society. The fully approved drug lecanemab exhibits the prospect of therapy against the pathological processes, while debatable adverse events conflict with the drug concentration required for the anticipated therapeutic effects. Reactive oxygen species (ROS) are involved in the pathological progression of AD, as has been demonstrated in much research regarding oxidative stress (OS). The contradiction between anticipated dosage and adverse event may be resolved through targeted transport by biomaterials and get therapeutic effects through pathological progression via regulation of ROS. Besides, biomaterials fix delivery issues by promoting the penetration of drugs across the blood-brain barrier (BBB), protecting the drug from peripheral degradation, and elevating bioavailability. The goal is to comprehensively understand the mechanisms of ROS in the progression of AD disease and the potential of ROS-related biomaterials in the treatment of AD. This review focuses on OS and its connection with AD and novel biomaterials in recent years against AD via OS to inspire novel biomaterial development. Revisiting these biomaterials and mechanisms associated with OS in AD via thorough investigations presents a considerable potential and bright future for improving effective interventions for AD.
Collapse
Affiliation(s)
- Zhuohang Yu
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Feng Luo
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
3
|
Zhang X, Tian X, Wang Y, Yan Y, Wang Y, Su M, Lv H, Li K, Hao X, Xing X, Song S. Application of lipopolysaccharide in establishing inflammatory models. Int J Biol Macromol 2024; 279:135371. [PMID: 39244120 DOI: 10.1016/j.ijbiomac.2024.135371] [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: 08/25/2024] [Accepted: 09/04/2024] [Indexed: 09/09/2024]
Abstract
Lipopolysaccharide (LPS), a unique component of the outer membrane of Gram-negative bacteria, possesses immune-activating properties. It induces an immune response by stimulating host cells to produce a lot of inflammatory cytokines with a thermogenic effect, which may cause an inflammatory response. In the past few decades, the structure and function of LPS and its mechanism leading to inflammation have been extensively analyzed. Since LPS can cause inflammation, it is often used to establish inflammation models. These models are crucial in the study of inflammatory diseases that pose a serious threat to human health. In addition, the non-pro-inflammatory effects of LPS under certain circumstances are also being studied widely. This review summarizes the methods by which LPS has been used to establish inflammatory models at the cellular and animal levels to study related diseases. It also introduces in detail the evaluation indicators necessary for the successful establishment of these models, providing a reference for future research.
Collapse
Affiliation(s)
- Xiao Zhang
- Marine College, Shandong University, Weihai, Shandong 264209, China.
| | - Xiao Tian
- Marine College, Shandong University, Weihai, Shandong 264209, China.
| | - Yan Wang
- Marine College, Shandong University, Weihai, Shandong 264209, China.
| | - Yong Yan
- JD Berry Agricultural Development Co., Ltd, Weihai, Shandong 264209, China.
| | - Yuan Wang
- Marine College, Shandong University, Weihai, Shandong 264209, China.
| | - Meicai Su
- Marine College, Shandong University, Weihai, Shandong 264209, China.
| | - Haifei Lv
- Marine College, Shandong University, Weihai, Shandong 264209, China.
| | - Kaitao Li
- Marine College, Shandong University, Weihai, Shandong 264209, China.
| | - Xiaobin Hao
- Marine College, Shandong University, Weihai, Shandong 264209, China.
| | - Xiang Xing
- Marine College, Shandong University, Weihai, Shandong 264209, China; Weihai Research Institute of Industrial Technology, Shandong University, Weihai 264209, China.
| | - Shuliang Song
- Marine College, Shandong University, Weihai, Shandong 264209, China; Weihai Research Institute of Industrial Technology, Shandong University, Weihai 264209, China.
| |
Collapse
|
4
|
Baghcheghi Y, Razazpour F, Seyedi F, Arefinia N, Hedayati-Moghadam M. Exploring the molecular mechanisms of PPARγ agonists in modulating memory impairment in neurodegenerative disorders. Mol Biol Rep 2024; 51:945. [PMID: 39215798 DOI: 10.1007/s11033-024-09850-6] [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: 05/06/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
Neurodegenerative diseases are characterized by progressive memory impairment and cognitive decline. This review aims to unravel the molecular mechanisms involved in the enhancement of memory function and mitigation of memory impairment through the activation of PPARγ agonists in neurodegenerative diseases. The findings suggest that PPARγ agonists modulate various molecular pathways involved in memory formation and maintenance. Activation of PPARγ enhances synaptic plasticity, promotes neuroprotection, suppresses neuroinflammation, attenuates oxidative stress, and regulates amyloid-beta metabolism. The comprehensive understanding of these molecular mechanisms would facilitate the development of novel therapeutic approaches targeting PPARγ to improve memory function and ultimately to alleviate the burden of neurodegenerative diseases. Further research, including clinical trials, is warranted to explore the efficacy, safety, and optimal use of specific PPARγ agonists as potential therapeutic agents in the treatment of memory impairments associated with neurodegenerative diseases.
Collapse
Affiliation(s)
- Yousef Baghcheghi
- Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, Iran
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Fateme Razazpour
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Fatemeh Seyedi
- Department of Anatomical Sciences, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Nasir Arefinia
- Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, Iran
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Mahdiyeh Hedayati-Moghadam
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran.
- Department of Physiology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran.
| |
Collapse
|
5
|
Ahmad F, Karan A, Sharma R, Sharma NS, Sundar V, Jayaraj R, Mukherjee S, DeCoster MA. Evolving therapeutic interventions for the management and treatment of Alzheimer's disease. Ageing Res Rev 2024; 95:102229. [PMID: 38364913 DOI: 10.1016/j.arr.2024.102229] [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: 09/17/2023] [Revised: 12/11/2023] [Accepted: 02/10/2024] [Indexed: 02/18/2024]
Abstract
Alzheimer's Disease (AD) patients experience diverse symptoms, including memory loss, cognitive impairment, behavioral abnormalities, mood changes, and mental issues. The fundamental objective of this review is to discuss novel therapeutic approaches, with special emphasis on recently approved marketed formulations for the treatment of AD, especially Aducanumab, the first FDA approved moiety that surpasses the blood-brain barrier (BBB) and reduces amyloid plaques in the brain, thereby reducing associated cognitive decline. However, it is still in the phase IV trial and is to be completed by 2030. Other drugs such as lecanemab are also under clinical trial and has recently been approved by the FDA and is also discussed here. In this review, we also focus on active and passive immunotherapy for AD as well as several vaccines, such as amyloid-beta epitope-based vaccines, amyloid-beta DNA vaccines, and stem cell therapy for AD, which are in clinical trials. Furthermore, ongoing pre-clinical trials associated with AD and other novel strategies such as curcumin-loaded nanoparticles, Crispr/ cas9, precision medicine, as well as some emerging therapies like anti-sense therapy are also highlighted. Additionally, we discuss some off-labeled drugs like non-steroidal anti-inflammatory drugs (NSAID), anti-diabetic drugs, and lithium, which can manage symptoms of AD and different non-pharmacological approaches are also covered which can help to manage AD. In summary, we have tried to cover all the therapeutic interventions which are available for the treatment and management of AD under sections approved, clinical phase, pre-clinical phase or futuristic interventions, off-labelled drugs, and non-pharmacological interventions for AD, offering positive findings and well as challenges that remain.
Collapse
Affiliation(s)
- Faizan Ahmad
- Department of Medical Elementology and Toxicology, Jamia Hamdard University, Delhi, India
| | - Anik Karan
- Department of Mechanical and Bioengineering, University of Kansas, Lawrence, KS, USA.
| | - Rashi Sharma
- Department of Biotechnology, Delhi Technological University, Bawana, Delhi, India
| | - Navatha Shree Sharma
- Department of Surgery Transplant, University of Nebraska Medical Centre, Omaha, NE, USA
| | - Vaishnavi Sundar
- Department of Internal Medicine, University of Nebraska Medical Centre, Omaha, NE, USA
| | - Richard Jayaraj
- Department of Paediatrics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, UAE
| | - Sudip Mukherjee
- Biomedical Engineering, Indian Institute of Technology- Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Mark A DeCoster
- Cellular Neuroscience Laboratory, Biomedical Engineering, College of Engineering and Science, Louisiana Tech University, Ruston, LA, USA; Cellular Neuroscience Laboratory, Institute for Micromanufacturing, College of Engineering and Science, Louisiana Tech University, Ruston, LA, USA.
| |
Collapse
|
6
|
Monsalvo-Maraver LA, Ovalle-Noguez EA, Nava-Osorio J, Maya-López M, Rangel-López E, Túnez I, Tinkov AA, Tizabi Y, Aschner M, Santamaría A. Interactions Between the Ubiquitin-Proteasome System, Nrf2, and the Cannabinoidome as Protective Strategies to Combat Neurodegeneration: Review on Experimental Evidence. Neurotox Res 2024; 42:18. [PMID: 38393521 PMCID: PMC10891226 DOI: 10.1007/s12640-024-00694-3] [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: 11/19/2023] [Revised: 01/13/2024] [Accepted: 02/04/2024] [Indexed: 02/25/2024]
Abstract
Neurodegenerative disorders are chronic brain diseases that affect humans worldwide. Although many different factors are thought to be involved in the pathogenesis of these disorders, alterations in several key elements such as the ubiquitin-proteasome system (UPS), the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway, and the endocannabinoid system (ECS or endocannabinoidome) have been implicated in their etiology. Impairment of these elements has been linked to the origin and progression of neurodegenerative disorders, while their potentiation is thought to promote neuronal survival and overall neuroprotection, as proved with several experimental models. These key neuroprotective pathways can interact and indirectly activate each other. In this review, we summarize the neuroprotective potential of the UPS, ECS, and Nrf2 signaling, both separately and combined, pinpointing their role as a potential therapeutic approach against several hallmarks of neurodegeneration.
Collapse
Affiliation(s)
- Luis Angel Monsalvo-Maraver
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Av. Universidad 3000, C.U. Coyoacán, 04510, Mexico City, Mexico.
| | - Enid A Ovalle-Noguez
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Av. Universidad 3000, C.U. Coyoacán, 04510, Mexico City, Mexico
| | - Jade Nava-Osorio
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Av. Universidad 3000, C.U. Coyoacán, 04510, Mexico City, Mexico
| | - Marisol Maya-López
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Av. Universidad 3000, C.U. Coyoacán, 04510, Mexico City, Mexico
- Doctorado en Ciencias Biológicas y de La Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | - Edgar Rangel-López
- Instituto Nacional de Neurología y Neurocirugía, S.S.A., Mexico City, Mexico
| | - Isaac Túnez
- Instituto de Investigaciones Biomédicas Maimonides de Córdoba (IMIBIC), Departamento de Bioquímica y Biología Molecular, Facultad de Medicina y Enfermería, Universidad de Córdoba, Red Española de Excelencia en Estimulación Cerebral (REDESTIM), Córdoba, Spain
| | - Alexey A Tinkov
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
- Yaroslavl State University, Yaroslavl, Russia
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Abel Santamaría
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Av. Universidad 3000, C.U. Coyoacán, 04510, Mexico City, Mexico.
| |
Collapse
|
7
|
Kamel GAM, Elariny HA. Pioglitazone attenuates tamoxifen-induced liver damage in rats via modulating Keap1/Nrf2/HO-1 and SIRT1/Notch1 signaling pathways: In-vivo investigations, and molecular docking analysis. Mol Biol Rep 2023; 50:10219-10233. [PMID: 37934372 PMCID: PMC10676319 DOI: 10.1007/s11033-023-08847-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 09/26/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND Tamoxifen (TAM) is a chemotherapeutic drug widely utilized to treat breast cancer. On the other hand, it exerts deleterious cellular effects in clinical applications as an antineoplastic agent, such as liver damage and cirrhosis. TAM-induced hepatic toxicity is mainly attributed to oxidative stress and inflammation. Pioglitazone (PIO), a peroxisome proliferator-activated receptor-gamma (PPAR-γ) agonist, is utilized to treat diabetes mellitus type-2. PIO has been reported to exert anti-inflammatory and antioxidant effects in different tissues. This research assessed the impact of PIO against TAM-induced hepatic intoxication. METHODS Rats received PIO (10 mg/kg) and TAM (45 mg/kg) orally for 10 days. RESULTS TAM increased aspartate aminotransferase (AST) and alanine aminotransferase (ALT), triggered several histopathological alterations, NF-κB p65, increased hepatic oxidative stress, and pro-inflammatory cytokines. PIO protects against TAM-induced liver dysfunction, reduced malondialdehyde (MDA), and pro-inflammatory markers along with improved hepatic antioxidants. Moreover, PIO, increased hepatic Bcl-2 expression while reducing Bax expression and caspase-3 levels. In addition, PIO decreased Keap-1, Notch1, and Hes-1 while upregulated HO-1, Nrf2, and SIRT1. Molecular docking showed the binding affinity of PIO for Keap-1, NF-κB, and SIRT1. CONCLUSION PIO mitigated TAM hepatotoxicity by decreasing apoptosis, inflammation, and oxidative stress. The protecting ability of PIO was accompanied by reducing Keap-1 and NF-κB and regulating Keap1/Nrf2/HO-1 and Sirt1/Notch1 signaling. A schematic diagram illustrating the protective effect of PIO against TAM hepatotoxicity. PIO prevented TAM-induced liver injury by regulating Nrf2/HO-1 and SIRT1/Notch1 signaling and mitigating oxidative stress, inflammation, and apoptosis.
Collapse
Affiliation(s)
- Gellan Alaa Mohamed Kamel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, P.N. 11754, Nasr City, Cairo, Egypt.
| | - Hemat A Elariny
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, P.N. 11754, Nasr City, Cairo, Egypt
| |
Collapse
|
8
|
Basagni F, Di Paolo ML, Cozza G, Dalla Via L, Fagiani F, Lanni C, Rosini M, Minarini A. Double Attack to Oxidative Stress in Neurodegenerative Disorders: MAO-B and Nrf2 as Elected Targets. Molecules 2023; 28:7424. [PMID: 37959843 PMCID: PMC10650714 DOI: 10.3390/molecules28217424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/30/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
Oxidative stress and neuroinflammation play a pivotal role in triggering the neurodegenerative pathological cascades which characterize neurodegenerative disorders, such as Alzheimer's and Parkinson's diseases. In search for potential efficient treatments for these pathologies, that are still considered unmet medical needs, we started from the promising properties of the antidiabetic drug pioglitazone, which has been repositioned as an MAO-B inhibitor, characterized by promising neuroprotective properties. Herein, with the aim to broaden its neuroprotective profile, we tried to enrich pioglitazone with direct and indirect antioxidant properties by hanging polyphenolic and electrophilic features that are able to trigger Nrf2 pathway and the resulting cytoprotective genes' transcription, as well as serve as radical scavengers. After a preliminary screening on MAO-B inhibitory properties, caffeic acid derivative 2 emerged as the best inhibitor for potency and selectivity over MAO-A, characterized by a reversible mechanism of inhibition. Furthermore, the same compound proved to activate Nrf2 pathway by potently increasing Nrf2 nuclear translocation and strongly reducing ROS content, both in physiological and stressed conditions. Although further biological investigations are required to fully clarify its neuroprotective properties, we were able to endow the pioglitazone scaffold with potent antioxidant properties, representing the starting point for potential future pioglitazone-based therapeutics for neurodegenerative disorders.
Collapse
Affiliation(s)
- Filippo Basagni
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy;
| | - Maria Luisa Di Paolo
- Department of Molecular Medicine, University of Padova, Via G. Colombo 3, 35131 Padova, Italy; (M.L.D.P.); (G.C.)
| | - Giorgio Cozza
- Department of Molecular Medicine, University of Padova, Via G. Colombo 3, 35131 Padova, Italy; (M.L.D.P.); (G.C.)
| | - Lisa Dalla Via
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy;
- Consorzio Interuniversitario Nazionale per la Scienza e Tecnologia dei Materiali (INSTM), 50121 Firenze, Italy
| | - Francesca Fagiani
- Department of Drug Sciences (Pharmacology Section), University of Pavia, V.le Taramelli 14, 27100 Pavia, Italy; (F.F.); (C.L.)
- Division of Neuroscience, IRCCS San Raffaele Hospital, Via Olgettina 60, 20132 Milan, Italy
| | - Cristina Lanni
- Department of Drug Sciences (Pharmacology Section), University of Pavia, V.le Taramelli 14, 27100 Pavia, Italy; (F.F.); (C.L.)
| | - Michela Rosini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy;
| | - Anna Minarini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy;
| |
Collapse
|
9
|
Vázquez-González D, Corona JC. Pioglitazone enhances brain mitochondrial biogenesis and phase II detoxification capacity in neonatal rats with 6-OHDA-induced unilateral striatal lesions. Front Neurosci 2023; 17:1186520. [PMID: 37575308 PMCID: PMC10416244 DOI: 10.3389/fnins.2023.1186520] [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: 03/16/2023] [Accepted: 07/17/2023] [Indexed: 08/15/2023] Open
Abstract
The psychostimulant methylphenidate (MPH) is the first-line pharmacological treatment for attention-deficit/hyperactivity disorder (ADHD), but has numerous adverse side effects. The PPARγ receptor agonist pioglitazone (PIO) is known to improve mitochondrial bioenergetics and antioxidant capacity, both of which may be deficient in ADHD, suggesting utility as an adjunct therapy. Here, we assessed the effects of PIO on ADHD-like symptoms, mitochondrial biogenesis and antioxidant pathways in multiple brain regions of neonate rats with unilateral striatal lesions induced by 6-hydroxydopamine (6-OHDA) as an experimental ADHD model. Unilateral striatal injection of 6-OHDA reduced ipsilateral dopaminergic innervation by 33% and increased locomotor activity. This locomotor hyperactivity was not altered by PIO treatment for 14 days. However, PIO increased the expression of proteins contributing to mitochondrial biogenesis in the striatum, hippocampus, cerebellum and prefrontal cortex of 6-OHDA-lesioned rats. In addition, PIO treatment enhanced the expression of the phase II transcription factor Nrf2 in the striatum, prefrontal cortex and cerebellum. In contrast, no change in the antioxidant enzyme catalase was observed in any of the brain regions analyzed. Thus, PIO may improve mitochondrial biogenesis and phase 2 detoxification in the ADHD brain. Further studies are required to determine if different dose regimens can exert more comprehensive therapeutic effects against ADHD neuropathology and behavior.
Collapse
Affiliation(s)
| | - Juan Carlos Corona
- Laboratory of Neurosciences, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| |
Collapse
|
10
|
Ye B, He D, Hu J, Yang S, Gao X, Cui M, Li Z, Wang H, Huang B, Fu S, Liu D. Notopterol inhibits LPS-induced inflammation in BV-2 cells via AKT/Nrf2/HO-1 signaling axis. Int Immunopharmacol 2023; 120:110334. [PMID: 37244113 DOI: 10.1016/j.intimp.2023.110334] [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: 01/30/2023] [Revised: 04/24/2023] [Accepted: 05/10/2023] [Indexed: 05/29/2023]
Abstract
Accumulating research has indicated that inordinate activation of microglia releases inflammatory cytokines, damages neurons, and causes neuroinflammation, which eventually could lead to neurodegenerative diseases such as Parkinson's disease and Huntington's disease, etc. Notopterol (NOT) has anti-inflammatory and anti-oxidant functions in boundary tissues, but the effects of NOT on neuroinflammation have not been covered. Therefore, this study attempts to investigate the effect of NOT on neuroinflammation and the underlying mechanisms. According to the findings, NOT dramatically decreased the expression of pro-inflammatory mediators (interleukin-6 (IL-6), inducible nitric-oxide synthase (iNOS), tumor necrosis factor-α (TNF-α), and Cyclooxygenase-2 (COX-2)) in LPS-exposed BV-2 cells. Western blot analysis revealed that NOT could promote the activation of AKT/Nrf2/HO-1 signaling pathway. Further studies have shown that anti-inflammatory property of NOT was inhibited by MK2206 (an AKT inhibitor), RA (an Nrf2 inhibitor), and SnPP IX (an HO-1 inhibitor). In addition, it was also discovered that NOT could weaken the damage of LPS to BV-2 cells and improve their survival rate. As a result, our results imply that NOT inhibits the inflammatory response of BV-2 cells through the AKT/Nrf2/HO-1 signaling axis and exerts a neuroprotective effect by inhibiting the activation of BV-2 cells.
Collapse
Affiliation(s)
- Bojian Ye
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Dewei He
- College of Animal Science, Jilin University, Changchun, China
| | - Jinping Hu
- College of Animal Science, Jilin University, Changchun, China.
| | - Shuo Yang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiyu Gao
- College of Animal Science, Jilin University, Changchun, China
| | - Mingchi Cui
- College of Veterinary Medicine, Jilin University, Changchun, China.
| | - Zhe Li
- College of Veterinary Medicine, Jilin University, Changchun, China.
| | - Hefei Wang
- College of Veterinary Medicine, Jilin University, Changchun, China.
| | - Bingxu Huang
- College of Animal Science, Jilin University, Changchun, China.
| | - Shoupeng Fu
- College of Veterinary Medicine, Jilin University, Changchun, China.
| | - Dianfeng Liu
- College of Animal Science, Jilin University, Changchun, China.
| |
Collapse
|
11
|
Araki W. Aβ Oligomer Toxicity-Reducing Therapy for the Prevention of Alzheimer's Disease: Importance of the Nrf2 and PPARγ Pathways. Cells 2023; 12:1386. [PMID: 37408220 DOI: 10.3390/cells12101386] [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: 03/29/2023] [Revised: 04/30/2023] [Accepted: 05/09/2023] [Indexed: 07/07/2023] Open
Abstract
Recent studies have revealed that soluble amyloid-β oligomers (AβOs) play a pathogenetic role in Alzheimer's disease (AD). Indeed, AβOs induce neurotoxic and synaptotoxic effects and are also critically involved in neuroinflammation. Oxidative stress appears to be a crucial event underlying these pathological effects of AβOs. From a therapeutic standpoint, new drugs for AD designed to remove AβOs or inhibit the formation of AβOs are currently being developed. However, it is also worth considering strategies for preventing AβO toxicity itself. In particular, small molecules with AβO toxicity-reducing activity have potential as drug candidates. Among such small molecules, those that can enhance Nrf2 and/or PPARγ activity can effectively inhibit AβO toxicity. In this review, I summarize studies on the small molecules that counteract AβO toxicity and are capable of activating Nrf2 and/or PPARγ. I also discuss how these interrelated pathways are involved in the mechanisms by which these small molecules prevent AβO-induced neurotoxicity and neuroinflammation. I propose that AβO toxicity-reducing therapy, designated ATR-T, could be a beneficial, complementary strategy for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Wataru Araki
- Department of Neurology and Neurological Science, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
- Memory Clinic Ochanomizu, Bunkyo-ku, Tokyo 113-8510, Japan
| |
Collapse
|
12
|
Tahamtan M, Aghaei I, Shabani M, Nazari A, Pooladvand V, Razavinasab M. Peroxisome proliferator-activated receptor-γ doesn't modify altered electrophysiological properties of the CA1 pyramidal neurons in a rat model of hepatic cirrhosis. Metab Brain Dis 2022; 37:2687-2697. [PMID: 35943675 DOI: 10.1007/s11011-022-01057-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 07/22/2022] [Indexed: 10/15/2022]
Abstract
Regarding the low quality of life due to the cognitive complications in the patients with hepatic cirrhosis (HC), the goal of this study was to examine the possible neuroprotective effect of pioglitazone (PIO) on the electrophysiological alterations of hippocampus, a major area of cognition, in the experimental model of bile duct ligation (BDL). We used adult male Wistar rats in the present study to perform BDL or sham surgery. Pioglitazone was administered in BDL rats two weeks after the surgery for the next continuous four weeks. The effects of pioglitazone on BDL-induced electrophysiological alterations of the CA1 pyramidal neurons in the hippocampus were evaluated by whole-cell patch clamp recordings. Our findings demonstrated that chronic administration of PIO could not reverse the electrophysiological changes in the CA1 pyramidal neurons of the hippocampus in BDL rats but could improve the hepatic dysfunction.Together, the results of this study suggest that PIO administration cannot counteract altered intrinsic properties of the hippocampal neurons which has been shown recently as an involved mechanism of the cognitive impairments in hepatic encephalopathy (HE).
Collapse
Affiliation(s)
- Mahshid Tahamtan
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Iraj Aghaei
- Neuroscience Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Shabani
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, 76198-13159, Kerman, Iran.
| | - Abbas Nazari
- Department of Biology, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - Vahid Pooladvand
- Biochemical Department, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Moazamehosadat Razavinasab
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, 76198-13159, Kerman, Iran.
| |
Collapse
|
13
|
Shi J, Hou J, Sun Y, Jia Z, Zhou Y, Wang C, Zhao H. Chaihujialonggumulitang shows psycho-cardiology therapeutic effect on acute myocardial infarction with comorbid anxiety by the activation of Nrf2/HO-1 pathway and suppression of oxidative stress and apoptosis. Biomed Pharmacother 2022; 153:113437. [PMID: 36076489 DOI: 10.1016/j.biopha.2022.113437] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/12/2022] [Accepted: 07/15/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Anxiety is a common comorbidity of cardiovascular diseases, which deteriorated cardiac function. Chaihujialonggumulitang (BFG) was reported to have antioxidant properties, alleviate myocardial ischemia injury and improve anxiety-like behavior. The Nuclear factor erythroid 2-related factor 2 (Nrf2) /heme oxygenase-1 (HO-1) pathway is the main mechanism to defend against oxidative stress, and improve cardiac function. This study was to investigate the possible mechanism of BFG in the treatment of psycho-cardiology. METHODS AMI with comorbid anxiety rat model was established by ligation of the left anterior descending coronary artery combined with uncertain empty bottle stimulation, followed by the administration of BFG (1 mL/100 g/d by gavage) or Dimethyl fumarate (DMF, 10 mg/kg/d by intraperitoneal injection) for 6 days. Echocardiography, myocardial injury markers, H&E, and Masson staining were employed to evaluate cardiac function. Behavioral tests and hippocampus neurotransmitters were applied to record anxiety-like behavior. We employed immunohistochemistry, RT-PCR, western blotting, and biochemical analysis to detect the protein and gene expression of Nrf2/HO-1 pathway-related factors, and oxidative stress and apoptosis parameters. RESULTS Rats in the AMI and complex groups showed cardiac function deterioration, as well as anxiety-like behavior. BFG improved echocardiography indicators, reduced myocardial injury markers, and attenuated myocardial pathological changes. BFG also ameliorated anxiety-like behaviors and elevated neurotransmitters levels. BFG promoted the activation of Nrf2/HO-1 pathway, increased antioxidant enzyme activities, reduced lipid peroxidation levels, and alleviated oxidative damage and apoptosis. DMF showed therapeutic effects and molecular mechanisms similar to BFG. CONCLUSION BFG may possess a psycho-cardiology therapeutic effect on AMI with comorbid anxiety by the activation of the Nrf2/HO-1 pathway and suppression of oxidative stress and apoptosis.
Collapse
Affiliation(s)
- Jinyu Shi
- Beijing University of Chinese Medicine, Beijing 100029, China; The DongFang Hospital of Beijing University of Chinese Medicine, Beijing 100078, China
| | - Jiqiu Hou
- The DongFang Hospital of Beijing University of Chinese Medicine, Beijing 100078, China
| | - Yize Sun
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zihao Jia
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yue Zhou
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Chao Wang
- The DongFang Hospital of Beijing University of Chinese Medicine, Beijing 100078, China.
| | - Haibin Zhao
- The DongFang Hospital of Beijing University of Chinese Medicine, Beijing 100078, China.
| |
Collapse
|
14
|
Lipopolysaccharide-Induced Model of Neuroinflammation: Mechanisms of Action, Research Application and Future Directions for Its Use. Molecules 2022; 27:molecules27175481. [PMID: 36080253 PMCID: PMC9457753 DOI: 10.3390/molecules27175481] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/23/2022] [Accepted: 08/23/2022] [Indexed: 11/19/2022] Open
Abstract
Despite advances in antimicrobial and anti-inflammatory therapies, inflammation and its consequences still remain a significant problem in medicine. Acute inflammatory responses are responsible for directly life-threating conditions such as septic shock; on the other hand, chronic inflammation can cause degeneration of body tissues leading to severe impairment of their function. Neuroinflammation is defined as an inflammatory response in the central nervous system involving microglia, astrocytes, and cytokines including chemokines. It is considered an important cause of neurodegerative diseases, such as Alzheimer’s disease, Parkinson’s disease and amyotrophic lateral sclerosis. Lipopolysaccharide (LPS) is a strong immunogenic particle present in the outer membrane of Gram-negative bacteria. It is a major triggering factor for the inflammatory cascade in response to a Gram-negative bacteria infection. The use of LPS as a strong pro-inflammatory agent is a well-known model of inflammation applied in both in vivo and in vitro studies. This review offers a summary of the pathogenesis associated with LPS exposure, especially in the field of neuroinflammation. Moreover, we analyzed different in vivo LPS models utilized in the area of neuroscience. This paper presents recent knowledge and is focused on new insights in the LPS experimental model.
Collapse
|
15
|
Zhang X, Liang S, Gao X, Huang H, Lao F, Dai X. Protective Effect of Chitosan Oligosaccharide against Hydrogen Peroxide-Mediated Oxidative Damage and Cell Apoptosis via Activating Nrf2/ARE Signaling Pathway. Neurotox Res 2021; 39:1708-1720. [PMID: 34622385 DOI: 10.1007/s12640-021-00419-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 09/07/2021] [Accepted: 09/17/2021] [Indexed: 12/11/2022]
Abstract
Chitosan oligosaccharide (COS), hydrolyzed and deacetylated from chitosan, has been reported to possess varieties of biological activities. Alzheimer's disease (AD) is a multifactorial progressive neurodegenerative disorder characterized by cognitive decline and memory loss, where oxidative stress was reported to be an overwhelming cause of the occurrence of AD. We have previously reported that COS could significantly decrease cell death, ROS generation, and lipid peroxidation, though the potential mechanism was yet to be determined. This study was designed to investigate the neuroprotective effect of COS against hydrogen peroxide (H2O2)-induced oxidative stress and apoptosis in neuronal SH-SY5Y cells. Our results indicated that COS could dose-dependently scavenge H2O2 in the cell-free systems. Accordingly, COS markedly decreased H2O2-induced cell apoptosis and intracellular ROS generation, while increased antioxidant capacity in SH-SY5Y cells. Further, COS significantly reduced the expression of Bax and upregulated Bcl-2. The mRNA and protein expression levels of nuclear Nrf2, heme oxygenase 1 (HO-1), and NAD(P)H: quinone oxidoreductase 1 (NQO1) were significantly increased upon COS treatment. Moreover, Nrf2-siRNA evidently reversed the promotive effect of COS on expression levels of HO-1 and NQO1, and ARE-driven transcriptional activity as determined by double-luciferase reporter gene assay. Besides, COS reversed H2O2-mediated increased phosphorylation of ERK1/2 and p38 MAPK. In conclusion, our findings indicate that COS could protect SH-SY5Y cells from oxidative damage and apoptosis via regulating Nrf2/ARE signaling pathway, which may provide new applications for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Xiaoxia Zhang
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing, 100191, China
| | - Shuang Liang
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing, 100191, China
| | - Xiaohan Gao
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing, 100191, China
| | - Hanchang Huang
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing, 100191, China
| | - Fengxue Lao
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing, 100191, China
| | - Xueling Dai
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing, 100191, China.
| |
Collapse
|
16
|
Alzoubi K, Khabour O, Alfaqih M, Tashtoush M, Al-Azzam S, Mhaidat N, Alrabadi N. The protective effects of pioglitazone against cognitive impairment caused by L-Methionine administration in a rat model. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 21:77-84. [PMID: 34370649 DOI: 10.2174/1871527320666210809122523] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 04/02/2021] [Accepted: 04/30/2021] [Indexed: 11/22/2022]
Abstract
PURPOSE Accumulating evidence indicates that elevated levels of methionine are associated with cognitive decline including loss of memory. The exact mechanisms behind this observation are not completely understood but could be related to an increase in oxidative stress markers in hippocampal tissues. The above increase in oxidative stress could be directly caused by an increase in the blood levels of methionine (hypermethioninemia) or one of its metabolites, such as homocysteine. Pioglitazone is a drug primarily used for the treatment of type 2 diabetes mellitus. Several reports showed that using pioglitazone protects against cognitive decline observed in Alzheimer's disease. Pioglitazone has antioxidant properties independent of its hypoglycemic effects. Taken together, we hypothesized that pioglitazone protects against memory loss triggered by elevated levels of methionine through lowering of oxidative stress in the hippocampus. METHOD To test this hypothesis, we used chronic administration of L-methionine in a rat model. Spatial learning and memory were evaluated in the model using a radial arm water maze (RAWM). The levels of several markers related to oxidative stress were measured in hippocampal tissues recovered from experimental rats. RESULTS Current results showed that administration of L-methionine was associated with a significant loss of short- and long-term memory and an increase in blood homocysteine levels. The above memory changes were associated with an increase in lipid peroxidation and a decrease in the activity of catalase and glutathione peroxidase antioxidant enzymes in the hippocampus. The combined treatment of pioglitazone with L-methionine protected rat model from memory loss. It also prevented changes observed in lipid peroxidation and changes in the activity of catalase and glutathione peroxidase enzymes. CONCLUSION Current findings indicate that pioglitazone is a viable therapeutic option that protects against cognitive changes observed upon administration of L-methionine.
Collapse
Affiliation(s)
- Karem Alzoubi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid-22110. Jordan
| | - Omar Khabour
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid. Jordan
| | - Mahmoud Alfaqih
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid-22110. Jordan
| | - Murad Tashtoush
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid-22110. Jordan
| | - Sayer Al-Azzam
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid-22110. Jordan
| | - Nizar Mhaidat
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid-22110. Jordan
| | - Nasr Alrabadi
- Department of Pharmacology, Faculty of Medicine, Jordan University of Science and Technology, Irbid-22110. Jordan
| |
Collapse
|
17
|
Hu Y, Tao R, Chen L, Xiong Y, Xue H, Hu L, Yan C, Xie X, Lin Z, Panayi AC, Mi B, Liu G. Exosomes derived from pioglitazone-pretreated MSCs accelerate diabetic wound healing through enhancing angiogenesis. J Nanobiotechnology 2021; 19:150. [PMID: 34020670 PMCID: PMC8139165 DOI: 10.1186/s12951-021-00894-5] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/11/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Enhanced angiogenesis can promote diabetic wound healing. Mesenchymal stem cells (MSCs)-derived exosomes, which are cell-free therapeutics, are promising candidates for the treatment of diabetic wound healing. The present study aimed to investigate the effect of exosomes derived from MSCs pretreated with pioglitazone (PGZ-Exos) on diabetic wound healing. RESULTS We isolated PGZ-Exos from the supernatants of pioglitazone-treated BMSCs and found that PGZ-Exos significantly promote the cell viability and proliferation of Human Umbilical Vein Vascular Endothelial Cells (HUVECs) injured by high glucose (HG). PGZ-Exos enhanced the biological functions of HUVECs, including migration, tube formation, wound repair and VEGF expression in vitro. In addition, PGZ-Exos promoted the protein expression of p-AKT, p-PI3K and p-eNOS and suppressed that of PTEN. LY294002 inhibited the biological function of HUVECs through inhibition of the PI3K/AKT/eNOS pathway. In vivo modeling in diabetic rat wounds showed that pioglitazone pretreatment enhanced the therapeutic efficacy of MSCs-derived exosomes and accelerated diabetic wound healing via enhanced angiogenesis. In addition, PGZ-Exos promoted collagen deposition, ECM remodeling and VEGF and CD31 expression, indicating adequate angiogenesis in diabetic wound healing. CONCLUSIONS PGZ-Exos accelerated diabetic wound healing by promoting the angiogenic function of HUVECs through activation of the PI3K/AKT/eNOS pathway. This offers a promising novel cell-free therapy for treating diabetic wound healing.
Collapse
Affiliation(s)
- Yiqiang Hu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Ranyang Tao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Lang Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Yuan Xiong
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Hang Xue
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Liangcong Hu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Chenchen Yan
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Xudong Xie
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Ze Lin
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Adriana C Panayi
- Department of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02215, USA
| | - Bobin Mi
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| | - Guohui Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| |
Collapse
|
18
|
Zhang F, Jin C, Wang X, Yan H, Tan H, Gao C. Dietary supplementation with pioglitazone hydrochloride and l-carnosine improves the growth performance, muscle fatty acid profiles and shelf life of yellow-feathered broiler chickens. ACTA ACUST UNITED AC 2020; 7:168-175. [PMID: 33997345 PMCID: PMC8110847 DOI: 10.1016/j.aninu.2020.05.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 05/05/2020] [Accepted: 05/15/2020] [Indexed: 12/19/2022]
Abstract
The present study aimed to investigate the effects of dietary pioglitazone hydrochloride (PGZ) and l-carnosine (LC) supplementation on the growth performance, meat quality, antioxidant status, and meat shelf life of yellow-feathered broiler chickens. Five hundred broiler chickens were randomly assigned into 4 experimental diets using a 2 × 2 factorial arrangement with 2 PGZ supplemental levels (0 and 15 mg/kg) and 2 LC supplemental levels (0 and 400 mg/kg) in basal diets for 28 d. The feed-to-gain ratio decreased whereas the average daily gain increased with PGZ supplementation. Greater dressing percentages, contents of intramuscular fat (IMF) in breast and thigh muscles, C18:3n-6, C18:1n-9 and monounsaturated fatty acid (MUFA) percentages of thigh muscle were observed with PGZ addition. Additionally, significant synergistic effects between PGZ and LC on the C18:1n-9 and MUFA contents were found. Supplementation with LC decreased drip loss, cooking loss and total volatile basic nitrogen, and increased the redness (a∗) value, the superoxide dismutase and glutathione peroxidase activities in thigh muscles. Moreover, the malondialdehyde content decreased when diets were supplemented with LC, and there was a synergistic effect between PGZ and LC. Additionally, the mRNA abundance of lipogenesis-related genes, such as peroxisome proliferator-activated receptor γ (PPARγ), PPARγ co-activator 1α and fatty acid-binding protein 3, increased with PGZ supplementation, and relevant antioxidation genes, such as nuclear factor erythroid-2-related factor 2 and superoxide dismutase 1, were enhanced with LC supplementation. In conclusion, the results indicated that the supplementation of PGZ and LC could improve the growth performance, antioxidant ability, IMF content, and meat shelf life of yellow-feathered broiler chickens.
Collapse
Affiliation(s)
- Fan Zhang
- College of Animal Science, South China Agricultural University, Guangdong Provincial Key Laboratory of Animal Nutrition Control, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, 510642, Guangdong, China
| | - Chenglong Jin
- College of Animal Science, South China Agricultural University, Guangdong Provincial Key Laboratory of Animal Nutrition Control, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, 510642, Guangdong, China
| | - Xiuqi Wang
- College of Animal Science, South China Agricultural University, Guangdong Provincial Key Laboratory of Animal Nutrition Control, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, 510642, Guangdong, China
| | - Huichao Yan
- College of Animal Science, South China Agricultural University, Guangdong Provincial Key Laboratory of Animal Nutrition Control, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, 510642, Guangdong, China
| | - Huize Tan
- WENS Foodstuff Group Co., Ltd, Yunfu, 527400, Guangdong, China
| | - Chunqi Gao
- College of Animal Science, South China Agricultural University, Guangdong Provincial Key Laboratory of Animal Nutrition Control, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, 510642, Guangdong, China
| |
Collapse
|
19
|
Ayaydin H, Akaltun İ, Koyuncu İ, Çelİk H, Kİrmİt A, Takatak H. High KEAP1, NRF2 and Low HO-1 Serum Levels in Children with Autism. ACTA ACUST UNITED AC 2020; 57:274-279. [PMID: 33354117 DOI: 10.29399/npa.24862] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 05/13/2020] [Indexed: 12/27/2022]
Abstract
Introduction The purpose of our study was to investigate heme oxygenase-1 (HO-1), nuclear factor erythroid-2-related factor 2 (NRF2), and kelch-like ECH-associated protein 1 (KEAP1) levels in children with autism spectrum disorder (ASD) and to reveal their association with the severity of autism. Methods This study measured serum HO-1, KEAP1, and NRF2 levels in 43 patients with ASD (aged 3-12 years) and in 41 age- and gender-matched healthy controls. ASD severity was rated using the Childhood Autism Rating Scale (CARS). HO-1, KEAP1, and NRF2 levels were determined in the biochemistry laboratory using the ELISA technique. Results HO-1 levels were significantly lower in patients aged 3-12 years compared to controls aged 3-12, while KEAP1 and NRF2 levels were significantly higher (p=0.020, p<0.001, and p=0.017, respectively). No correlation was determined between ASD severity on the basis of total CARS scores and HO-1, KEAP1 or NRF2 (p>0.05). Conclusion This study suggests that oxidative stress is higher in children with ASD and that HO-1 levels are insufficient to achieve oxidative balance.
Collapse
Affiliation(s)
- Hamza Ayaydin
- Harran University Faculty of Medicine, Department of Child and Adolescent Psychiatry, Şanlıurfa, Turkey
| | - İsmail Akaltun
- Gaziantep Dr. Ersin Arslan Training and Research Hospital, Department of Child and Adolescent Psychiatry, Gaziantep, Turkey
| | - İsmail Koyuncu
- Harran University Faculty of Medicine, Department of Biochemistry, Şanlıurfa, Turkey
| | - Hakim Çelİk
- Harran University Faculty of Medicine, Department of Physiology, Şanlıurfa, Turkey
| | - Adnan Kİrmİt
- Harran University Faculty of Medicine, Department of Biochemistry, Şanlıurfa, Turkey
| | - Hatice Takatak
- Harran University Faculty of Medicine, Department of Child and Adolescent Psychiatry, Şanlıurfa, Turkey
| |
Collapse
|
20
|
Fernández-Mendívil C, Luengo E, Trigo-Alonso P, García-Magro N, Negredo P, López MG. Protective role of microglial HO-1 blockade in aging: Implication of iron metabolism. Redox Biol 2020; 38:101789. [PMID: 33212416 PMCID: PMC7680814 DOI: 10.1016/j.redox.2020.101789] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/06/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
Heme oxygenase-1 (HO-1) is an inducible enzyme known for its anti-inflammatory, antioxidant and neuroprotective effects. However, increased expression of HO-1 during aging and age-related neurodegenerative diseases have been associated to neurotoxic ferric iron deposits. Being microglia responsible for the brain's innate immune response, the aim of this study was to understand the role of microglial HO-1 under inflammatory conditions in aged mice. For this purpose, aged wild type (WT) and LysMCreHmox1△△ (HMOX1M-KO) mice that lack HO-1 in microglial cells, were used. Aged WT mice showed higher basal expression levels of microglial HO-1 in the brain than adult mice. This increase was even higher when exposed to an inflammatory stimulus (LPS via i.p.) and was accompanied by alterations in different iron-related metabolism proteins, resulting in an increase of iron deposits, oxidative stress, ferroptosis and cognitive decline. Furthermore, microglia exhibited a primed phenotype and increased levels of inflammatory markers such as iNOS, p65, IL-1β, TNF-α, Caspase-1 and NLRP3. Interestingly, all these alterations were prevented in aged HMOX1M-KO and WT mice treated with the HO-1 inhibitor ZnPPIX. In order to determine the effects of microglial HO-1-dependent iron overload, aged WT mice were treated with the iron chelator deferoxamine (DFX). DFX caused major improvements in iron, inflammatory and behavioral alterations found in aged mice exposed to LPS. In conclusion, this study highlights how microglial HO-1 overexpression contributes to neurotoxic iron accumulation providing deleterious effects in aged mice exposed to an inflammatory insult. Microglial HO-1 increases with aging and under an acute inflammatory stimulus. LPS-dependent microglial HO-1 upregulation during aging leads to iron overload. Microglial HO-1-dependent iron accumulation leads to ferroptosis. HO-1-dependent iron alterations lead to neuroinflammation. HO-1 inhibitors/iron chelators reduce iron accumulation and neuroinflammation.
Collapse
Affiliation(s)
- Cristina Fernández-Mendívil
- Instituto Teófilo Hernando for Drug Discovery. Department of Pharmacology. School of Medicine. Universidad Autónoma Madrid. Madrid, Spain; Instituto de Investigación Sanitario (IIS-IP), Hospital Universitario de La Princesa, Madrid, Spain
| | - Enrique Luengo
- Instituto Teófilo Hernando for Drug Discovery. Department of Pharmacology. School of Medicine. Universidad Autónoma Madrid. Madrid, Spain; Instituto de Investigación Sanitario (IIS-IP), Hospital Universitario de La Princesa, Madrid, Spain
| | - Paula Trigo-Alonso
- Instituto Teófilo Hernando for Drug Discovery. Department of Pharmacology. School of Medicine. Universidad Autónoma Madrid. Madrid, Spain; Instituto de Investigación Sanitario (IIS-IP), Hospital Universitario de La Princesa, Madrid, Spain
| | - Nuria García-Magro
- Department of Anatomy, Histology and Neuroscience. School of Medicine. Universidad Autónoma de Madrid. Madrid, Spain
| | - Pilar Negredo
- Department of Anatomy, Histology and Neuroscience. School of Medicine. Universidad Autónoma de Madrid. Madrid, Spain
| | - Manuela G López
- Instituto Teófilo Hernando for Drug Discovery. Department of Pharmacology. School of Medicine. Universidad Autónoma Madrid. Madrid, Spain; Instituto de Investigación Sanitario (IIS-IP), Hospital Universitario de La Princesa, Madrid, Spain.
| |
Collapse
|
21
|
Sun W, Cheng Z, Chen H, Lin G, Chen H. Tetrahydropyrimidines, ZL-5015 Alleviated Lipopolysaccharide (LPS)-Induced Acute Pneumonia in Rats by Activating the NRF-2/HO-1 Pathway. MEDICAL SCIENCE MONITOR : INTERNATIONAL MEDICAL JOURNAL OF EXPERIMENTAL AND CLINICAL RESEARCH 2020; 26:e924482. [PMID: 32844782 PMCID: PMC8147033 DOI: 10.12659/msm.924482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Acute pneumonia is a severe inflammatory disease of the respiratory system. Drugs used to treat acute pneumonia often have strong side effects. Recent studies have shown that tetrahydropyrimidines, ZL-5015 has anti-inflammatory and antitumor effects. However, whether ZL-5015 can relieve symptoms of acute pneumonia is unclear. MATERIAL AND METHODS In this study, we used lipo-polysaccharide (LPS) to stimulate SD rats to simulate conditions of acute pneumonia. Diverse doses of ZL-5015 were used for treatment of these rats. After the rates were sacrificed, serum, lung tissue, and bronchoalveolar lavage fluid were collected for the next study. Hematoxylin-eosin (H&E) staining then was used to detect pathologic changes in lung tissues. Enzyme-linked immunosorbent assay was performed to assess levels of inflammatory factors in serum. Commercial kits were used to assess levels of reactive oxygen species (ROS) in bronchoalveolar lavage fluid. RESULTS Treatment of ZL-5015 relieved stenosis of the alveolar space and pulmonary edema. Furthermore, levels of inflammatory factors (TNF-alpha, IL-1ß and IL-18) in the lung tissues and serum were downregulated after treatment with ZL-5015. Production of ROS also was suppressed after application of ZL-5015. Moreover, inhibition of expression of NRF-2 and HO-1 was relieved after treatment with ZL-5015. The therapeutic effect of ZL-5015 showed a dose-response relationship. CONCLUSIONS ZL-5015 alleviated LPS-induced inflammatory injury and oxidative damage by activating the NRF-2/HO-1 pathway.
Collapse
Affiliation(s)
- Wei Sun
- Department of Emergency, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China (mainland)
| | - Zhou Cheng
- Department of Emergency, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China (mainland)
| | - Hanyan Chen
- Department of Emergency, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China (mainland)
| | - Guifen Lin
- Department of Emergency, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China (mainland)
| | - Hongxing Chen
- Department of Emergency, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China (mainland)
| |
Collapse
|
22
|
Effects of the probiotic formulation SLAB51 in in vitro and in vivo Parkinson's disease models. Aging (Albany NY) 2020; 12:4641-4659. [PMID: 32155131 PMCID: PMC7093198 DOI: 10.18632/aging.102927] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/04/2020] [Indexed: 12/20/2022]
Abstract
Parkinson is a common neurodegenerative disorder, characterized by motor and non-motor symptoms, including abnormalities in the gut function, which may appear before the motor sign. To date, there are treatments that can help relieve Parkinson’ disease (PD)-associated symptoms, but there is no cure to control the onset and progression of this disorder. Altered components of the gut could represent a key role in gut-brain axis, which is a bidirectional system between the central nervous system and the enteric nervous system. Diet can alter the microbiota composition, affecting gut-brain axis function. Gut microbiome restoration through selected probiotics’ administration has been reported. In this study, we investigated the effects of the novel formulation SLAB51 in PD. Our findings indicate that this probiotic formulation can counteract the detrimental effect of 6-OHDA in vitro and in vivo models of PD. The results suggest that SLAB51 can be a promising candidate for the prevention or as coadjuvant treatment of PD.
Collapse
|