1
|
Kocsis AE, Kucsápszky N, Santa-Maria AR, Hunyadi A, Deli MA, Walter FR. Much More than Nutrients: The Protective Effects of Nutraceuticals on the Blood-Brain Barrier in Diseases. Nutrients 2025; 17:766. [PMID: 40077636 PMCID: PMC11901837 DOI: 10.3390/nu17050766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
The dysfunction of the blood-brain barrier (BBB) is well described in several diseases, and is considered a pathological factor in many neurological disorders. This review summarizes the most important groups of natural compounds, including alkaloids, flavonoids, anthocyanidines, carotenoids, lipids, and vitamins that were investigated for their potential protective effects on brain endothelium. The brain penetration of these compounds and their interaction with BBB efflux transporters and solute carriers are discussed. The cerebrovascular endothelium is considered a therapeutic target for natural compounds in diseases. In preclinical studies modeling systemic and central nervous system diseases, nutraceuticals exerted beneficial effects on the BBB. In vivo, they decreased BBB permeability, brain edema, astrocyte swelling, and morphological changes in the vessel structure and basal lamina. At the level of brain endothelial cells, nutraceuticals increased cell survival and decreased apoptosis. From the general endothelial functions, decreased angiogenesis and increased levels of vasodilating agents were demonstrated. From the BBB functions, elevated barrier integrity by tightened intercellular junctions, and increased expression and activity of BBB transporters, such as efflux pumps, solute carriers, and metabolic enzymes, were shown. Nutraceuticals enhanced the antioxidative defense and exerted anti-inflammatory effects at the BBB. The most important signaling changes mediating the increased cell survival and BBB stability were the activation of the WNT, PI3K-AKT, and NRF2 pathways, and inhibition of the MAPK, JNK, ERK, and NF-κB pathways. Nutraceuticals represent a valuable source of new potentially therapeutic molecules to treat brain diseases by protecting the BBB.
Collapse
Affiliation(s)
- Anna E. Kocsis
- Biological Barriers Research Group, Institute of Biophysics, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary; (A.E.K.); (N.K.)
| | - Nóra Kucsápszky
- Biological Barriers Research Group, Institute of Biophysics, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary; (A.E.K.); (N.K.)
| | - Ana Raquel Santa-Maria
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Attila Hunyadi
- Institute of Pharmacognosy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary;
- Interdisciplinary Centre of Natural Products, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
- HUN-REN-SZTE Biologically Active Natural Products Research Group, Eötvös u. 6, H-6720 Szeged, Hungary
- Graduate Institute of Natural Products, Kaohsiung Medical University, Shih-Chuan 1st Rd. 100, Kaohsiung 807, Taiwan
| | - Mária A. Deli
- Biological Barriers Research Group, Institute of Biophysics, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary; (A.E.K.); (N.K.)
| | - Fruzsina R. Walter
- Biological Barriers Research Group, Institute of Biophysics, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary; (A.E.K.); (N.K.)
| |
Collapse
|
2
|
Mahakalakar N, Mohariya G, Taksande B, Kotagale N, Umekar M, Vinchurney M. "Nattokinase as a potential therapeutic agent for preventing blood-brain barrier dysfunction in neurodegenerative disorders". Brain Res 2025; 1849:149352. [PMID: 39592088 DOI: 10.1016/j.brainres.2024.149352] [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: 05/25/2024] [Revised: 10/11/2024] [Accepted: 11/23/2024] [Indexed: 11/28/2024]
Abstract
Neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis are characterized by progressive destruction of neurons and cognitive impairment, and thorough studies have provided evidence that these pathologies have a close relationship to the failure of the blood-brain barrier (BBB). Nattokinase (NK), a protease found in fermented soybeans, has been extensively studied because it displays powerful neuroprotective abilities, which is why current research was reviewed in the present article. It was concluded that there is enough evidence in preclinical studies using experimental animals that NK supplementation can alleviate the condition related to BBB dysfunction, reduce brain inflammation, and improve cognitive ability. Furthermore, the study of NK on the cardiovascular system leads to certain assumptions, which include the impact on vasculature function and the ability to manage blood flow, which is the key feature of BBB integrity. Such assumed mechanisms are fibrinolytic action, anti-inflammatory and antioxidant action, and endothelium function modulation. There are many positive research findings, and it seems that NK may serve as an effective opponent for BBB breakdown; however, a new research level should be taken to disclose the application and therapeutic use of NK in brain neurodegenerative disease.
Collapse
Affiliation(s)
- Nivedita Mahakalakar
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur (M.S.) 441 002, India
| | - Gunjan Mohariya
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur (M.S.) 441 002, India
| | - Brijesh Taksande
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur (M.S.) 441 002, India
| | - Nandkishor Kotagale
- Government College of Pharmacy (GCOP), Kathora Naka, V.M.V. Road, Amravati (M.S.) 444604, India
| | - Milind Umekar
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur (M.S.) 441 002, India
| | - Madhura Vinchurney
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur (M.S.) 441 002, India.
| |
Collapse
|
3
|
de Lima EP, Laurindo LF, Catharin VCS, Direito R, Tanaka M, Jasmin Santos German I, Lamas CB, Guiguer EL, Araújo AC, Fiorini AMR, Barbalho SM. Polyphenols, Alkaloids, and Terpenoids Against Neurodegeneration: Evaluating the Neuroprotective Effects of Phytocompounds Through a Comprehensive Review of the Current Evidence. Metabolites 2025; 15:124. [PMID: 39997749 PMCID: PMC11857241 DOI: 10.3390/metabo15020124] [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: 12/24/2024] [Revised: 02/07/2025] [Accepted: 02/09/2025] [Indexed: 02/26/2025] Open
Abstract
Neurodegenerative diseases comprise a group of chronic, usually age-related, disorders characterized by progressive neuronal loss, deformation of neuronal structure, or loss of neuronal function, leading to a substantially reduced quality of life. They remain a significant focus of scientific and clinical interest due to their increasing medical and social importance. Most neurodegenerative diseases present intracellular protein aggregation or their extracellular deposition (plaques), such as α-synuclein in Parkinson's disease and amyloid beta (Aβ)/tau aggregates in Alzheimer's. Conventional treatments for neurodegenerative conditions incur high costs and are related to the development of several adverse effects. In addition, many patients are irresponsive to them. For these reasons, there is a growing tendency to find new therapeutic approaches to help patients. This review intends to investigate some phytocompounds' effects on neurodegenerative diseases. These conditions are generally related to increased oxidative stress and inflammation, so phytocompounds can help prevent or treat neurodegenerative diseases. To achieve our aim to provide a critical assessment of the current literature about phytochemicals targeting neurodegeneration, we reviewed reputable databases, including PubMed, EMBASE, and COCHRANE, seeking clinical trials that utilized phytochemicals against neurodegenerative conditions. A few clinical trials investigated the effects of phytocompounds in humans, and after screening, 13 clinical trials were ultimately included following PRISMA guidelines. These compounds include polyphenols (flavonoids such as luteolin and quercetin, phenolic acids such as rosmarinic acid, ferulic acid, and caffeic acid, and other polyphenols like resveratrol), alkaloids (such as berberine, huperzine A, and caffeine), and terpenoids (such as ginkgolides and limonene). The gathered evidence underscores that quercetin, caffeine, ginkgolides, and other phytochemicals are primarily anti-inflammatory, antioxidant, and neuroprotective, counteracting neuroinflammation, neuronal oxidation, and synaptic dysfunctions, which are crucial aspects of neurodegenerative disease intervention in various included conditions, such as Alzheimer's and other dementias, depression, and neuropsychiatric disorders. In summary, they show that the use of these compounds is related to significant improvements in cognition, memory, disinhibition, irritability/lability, aberrant behavior, hallucinations, and mood disorders.
Collapse
Affiliation(s)
- Enzo Pereira de Lima
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Vitor Cavallari Strozze Catharin
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Rosa Direito
- Laboratory of Systems Integration Pharmacology, Clinical and Regulatory Science, Research Institute for Medicines, Universidade de Lisboa (iMed.ULisboa), Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Masaru Tanaka
- HUN-REN-SZTE Neuroscience Research Group, Danube Neuroscience Research Laboratory, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Tisza Lajos Krt. 113, H-6725 Szeged, Hungary
| | - Iris Jasmin Santos German
- Department of Biological Sciences (Anatomy), School of Dentistry of Bauru, University of São Paulo (FOB-USP), Alameda Doutor Octávio Pinheiro Brisolla, 9-75, Bauru 17012-901, São Paulo, Brazil
| | - Caroline Barbalho Lamas
- Department of Gerontology, School of Gerontology, Universidade Federal de São Carlos (UFSCar), São Carlos 13565-905, São Paulo, Brazil
| | - Elen Landgraf Guiguer
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Adriano Cressoni Araújo
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Adriana Maria Ragassi Fiorini
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Marília 17500-000, São Paulo, Brazil
| | - Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| |
Collapse
|
4
|
Zhang K, Hou B, Yan T, Qiao R, Qu P, Xu X, Zhang H. Identification of therapeutic target genes for age-related hearing loss through systematic genome-wide mendelian randomization of druggable genes. Exp Gerontol 2025; 200:112676. [PMID: 39778696 DOI: 10.1016/j.exger.2025.112676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/21/2024] [Accepted: 01/04/2025] [Indexed: 01/11/2025]
Abstract
BACKGROUND Age-related hearing loss (ARHL) is a common sensory disorder with significant public health implications. However, few effective treatment options are available. Mendelian randomization (MR) has been used to repurpose existing drugs and identify new therapeutic targets. Therefore, we performed a systematic genome-wide MR of drug-eligible individuals to explore potential therapeutic targets for ARHL. METHODS We obtained data on the expression quantitative trait locis (eQTLs) of druggable genes, which were then subjected to two-sample MR analyses and co-localisation analyses with data from the ARHL genome-wide association study to identify genes highly associated with ARHL. Additionally, we conducted phenome-wide research, enrichment analysis, protein network construction, drug prediction, and molecular docking to help develop more effective and targeted therapeutic treatments. RESULTS Overall, the MR analysis of eQTL data showed that 14 drug targets were significantly associated with ARHL. GO analysis of 14 potential targets revealed their primary involvement in biological processes such as the endoplasmic reticulum unfolded protein response, ER-nucleus signaling pathway, and fibroblast apoptotic process. Additionally, important cellular components include the Bcl-2 family of proteins and the endoplasmic reticulum lumen. After filtering using methods such as phenome-wide research, enrichment analysis, protein network construction, drug prediction, and molecular docking, six potentially druggable genes (BAK1, AMFR, LAMP3, STK17B, ACP5, and CD9) and six drugs (beclomethasone, propyl pyrazole triol, momelotinib, monoisoamyl-2,3-dimercaptosuccinate, pterostilbene, and naftidrofuryl) that may affect ARHL outcomes were finally identified. CONCLUSIONS Our findings identified 14 potential drug targets for ARHL. These findings offer promising leads for more effective treatments for ARHL and help determine the priority of drug development, potentially reducing costs.
Collapse
Affiliation(s)
- Kun Zhang
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, Shandong, China; NHC Key Laboratory of Otorhinolaryngology, Shandong University, Jinan, Shandong, China
| | - Bo Hou
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, Shandong, China; NHC Key Laboratory of Otorhinolaryngology, Shandong University, Jinan, Shandong, China
| | - Tao Yan
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, Shandong, China; NHC Key Laboratory of Otorhinolaryngology, Shandong University, Jinan, Shandong, China
| | - Ruru Qiao
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, Shandong, China; NHC Key Laboratory of Otorhinolaryngology, Shandong University, Jinan, Shandong, China
| | - Peng Qu
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China; Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Xinbo Xu
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, Shandong, China; NHC Key Laboratory of Otorhinolaryngology, Shandong University, Jinan, Shandong, China.
| | - Hanbing Zhang
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, Shandong, China; NHC Key Laboratory of Otorhinolaryngology, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
5
|
Anchimowicz J, Zielonka P, Jakiela S. Plant Secondary Metabolites as Modulators of Mitochondrial Health: An Overview of Their Anti-Oxidant, Anti-Apoptotic, and Mitophagic Mechanisms. Int J Mol Sci 2025; 26:380. [PMID: 39796234 PMCID: PMC11720160 DOI: 10.3390/ijms26010380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/29/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025] Open
Abstract
Plant secondary metabolites (PSMs) are a diverse group of bioactive compounds, including flavonoids, polyphenols, saponins, and terpenoids, which have been recognised for their critical role in modulating cellular functions. This review provides a comprehensive analysis of the effects of PSMs on mitochondrial health, with particular emphasis on their therapeutic potential. Emerging evidence shows that these metabolites improve mitochondrial function by reducing oxidative stress, promoting mitochondrial biogenesis, and regulating key processes such as apoptosis and mitophagy. Mitochondrial dysfunction, a hallmark of many pathologies, including neurodegenerative disorders, cardiovascular diseases, and metabolic syndrome, has been shown to benefit from the protective effects of PSMs. Recent studies show that PSMs can improve mitochondrial dynamics, stabilise mitochondrial membranes, and enhance bioenergetics, offering significant promise for the prevention and treatment of mitochondrial-related diseases. The molecular mechanisms underlying these effects, including modulation of key signalling pathways and direct interactions with mitochondrial proteins, are discussed. The integration of PSMs into therapeutic strategies is highlighted as a promising avenue for improving treatment efficacy while minimising the side effects commonly associated with synthetic drugs. This review also highlights the need for future research to elucidate the specific roles of individual PSMs and their synergistic interactions within complex plant matrices, which may further optimise their therapeutic utility. Overall, this work provides valuable insights into the complex role of PSMs in mitochondrial health and their potential as natural therapeutic agents targeting mitochondrial dysfunction.
Collapse
Affiliation(s)
| | | | - Slawomir Jakiela
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (J.A.); (P.Z.)
| |
Collapse
|
6
|
Fang F, Bao S, Chen D, Duan X, Zhao Y, Ma Y. Protective effects and mechanism of quercetin from Rhododendron dauricum against cerebral ischemia-reperfusion injury. Eur J Pharmacol 2024; 985:177126. [PMID: 39532226 DOI: 10.1016/j.ejphar.2024.177126] [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: 09/04/2024] [Revised: 10/18/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
METHODS This study seeks to identify the bioactive compounds within Rhododendron dauricum and explore potential mechanisms for treating cerebral I/R injury through a comprehensive analysis employing network pharmacology, complemented by experimental validation. RESULTS The core targets associated with quercetin in the treatment of cerebral I/R injury are TNF-α, IL-6, IL-1β, and AKT1. Notably, we propose for the first time that its mode of action primarily involves the inhibition of the TNF-α/RhoA/ROCK2 pathway. CONCLUSION Our findings reveal that quercetin emerges as a pivotal bioactive component of Rhododendron dauricum in the context of cerebral I/R injury treatment.
Collapse
Affiliation(s)
- Fang Fang
- Department of Pharmacy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Siwei Bao
- Department of Pharmacy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Danxia Chen
- Department of Pharmacy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Xiaofeng Duan
- Department of Pharmacy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yuefen Zhao
- Department of Pharmacy, Chinese medicine hospitals Changji Hui Autonomous Prefecture, Xinjiang, 831100, China.
| | - Yabin Ma
- Department of Pharmacy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
7
|
Gołąbek-Grenda A, Juzwa W, Kaczmarek M, Olejnik A. Resveratrol and Its Natural Analogs Mitigate Immune Dysregulation and Oxidative Imbalance in the Endometriosis Niche Simulated in a Co-Culture System of Endometriotic Cells and Macrophages. Nutrients 2024; 16:3483. [PMID: 39458478 PMCID: PMC11510005 DOI: 10.3390/nu16203483] [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: 08/21/2024] [Revised: 10/11/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Inflammation and immune cell dysfunction are critical facilitators of endometriosis pathophysiology. Macrophages are renowned for stimulating lesion growth, vascularization, innervation, and pain generation. By combining macrophages and endometriotic cells, we determined if resveratrol and its natural analogs can target the immune dysregulation and oxidative imbalance in endometriosis. Methods: After treatment with compounds (5, 10, 25 µM), we evaluated the expression of key inflammatory and oxidative stress markers, cytokines release, and ROS production by applying q-PCR, ELISA, Cytometric Beads Array, and multiplexed fluorogenic staining and flow cytometry analysis with bioimaging. Results: The results showed that endometriosis-related macrophages treated with stilbenes have impaired expression of pro-inflammatory markers (IL6, IL8, IL1B, TNF, CCL2, CXCL10, PTGS2). The effect of resveratrol, pterostilbene, and piceatannol was observed, especially in reducing IL1B, CCL2, and CXCL10 genes up to 3.5-, 5-, and 7.7-fold at 25 µM, respectively. Also, with piceatannol or polydatin exposure, the IL-6 decrease was noticeable. This study reported an antioxidant effect by reducing ROS-positive cells from 96% to 48% by pterostilbene. Results from flow cytometry correlated with the transcript activation of detoxification enzymes (SOD, GPX). Conclusions: Prospects for potential therapy based on regulating the immune microenvironment and reducing the accumulation of free radicals with stilbenes application were described in the article.
Collapse
Affiliation(s)
- Agata Gołąbek-Grenda
- Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, 48 Wojska Polskiego St., 60-627 Poznan, Poland; (A.G.-G.); (W.J.)
| | - Wojciech Juzwa
- Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, 48 Wojska Polskiego St., 60-627 Poznan, Poland; (A.G.-G.); (W.J.)
| | - Mariusz Kaczmarek
- Department of Cancer Immunology, Poznan University of Medical Sciences, Garbary 15 St., 61-866 Poznan, Poland;
- Gene Therapy Laboratory, Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Centre, Garbary 15 St., 61-866 Poznan, Poland
| | - Anna Olejnik
- Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, 48 Wojska Polskiego St., 60-627 Poznan, Poland; (A.G.-G.); (W.J.)
| |
Collapse
|
8
|
Begh MZA, Khan J, Zehravi M, Sweilam SH, Raja AD, Muthukumar A, Haque MA, Kar NR, Singh LP, Priya BD, Alshehri MA, Ahmad I, Kang S, Moon S, Park MN, Emran TB, Kim B. Targeting Neurological Disorders with Stilbenes: Bridging the Preclinical-Clinical Gap. Int J Biol Sci 2024; 20:5474-5494. [PMID: 39494329 PMCID: PMC11528462 DOI: 10.7150/ijbs.102032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 09/25/2024] [Indexed: 11/05/2024] Open
Abstract
Neurological disorders (NDs) encompass a range of debilitating conditions that affect the nervous system, including prevalent illnesses such as Alzheimer's disease, Parkinson's disease, and ischemic stroke. Despite significant ongoing studies, effective therapeutic strategies to halt or slow down the progression of these illnesses are still lacking. Stilbenes, a class of natural polyphenols, have shown potential as candidates for therapeutic strategies due to their capacity to protect the nervous system. Preclinical studies have provided strong evidence that stilbenes can regulate many cellular pathways implicated in neurodegeneration, with resveratrol being a well-studied compound that has shown the ability to reduce oxidative damage, promote neurogenesis, and enhance mitochondrial function - crucial for maintaining brain health. In preclinical animal models, initial research has also shown promise in additional substances such as piceatannol and pterostilbene. Furthermore, clinical studies have explored the therapeutic benefits of stilbenes in NDs. Despite promising results in preclinical research, the use of stilbenes in clinical trials is currently limited, with most studies focusing on resveratrol. Although several clinical studies have demonstrated the beneficial impact of resveratrol supplementation on brain health and degenerative consequences, other investigations have yielded ambiguous findings, underscoring the urgent need for more comprehensive and precisely planned clinical research. This study delves into the potential benefits of stilbenes as neuroprotective agents for NDs. It emphasizes the need for more clinical research to enhance our understanding of their therapeutic effectiveness in specific patient groups.
Collapse
Affiliation(s)
- Md. Zamshed Alam Begh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Jishan Khan
- Department of Pharmacy, International Islamic University Chittagong, Kumira, Chittagong 4318, Bangladesh
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy, College of Dentistry & Pharmacy, Buraydah Private Colleges, Buraydah 51418, Saudi Arabia
| | - Sherouk Hussein Sweilam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Egyptian Russian University, Cairo-Suez Road, Badr City, Cairo 11829, Egypt
| | - A. Dinesh Raja
- Department of Pharmaceutics, KMCH College of Pharmacy, Coimbatore, Tamil Nadu 641048, India
| | - A. Muthukumar
- Department of Pharmacology, The Oxford College of Pharmacy, Bengaluru, Karnataka 560068, India
| | - M Akiful Haque
- Department of Pharmaceutical Analysis, School of Pharmacy, Anurag University, Hyderabad, India
| | - Nihar Ranjan Kar
- Centurion University of Technology and Management, Gopalpur, Balasore 756044, Odisha, India
| | - Laliteshwar Pratap Singh
- Department of Pharmaceutical Chemistry, Narayan Institute of Pharmacy, Gopal Narayan Singh University, Sasaram 821305, India
| | - B. Dharani Priya
- Department of Pharmaceutics, KMCH College of Pharmacy, Coimbatore, Tamil Nadu 641048, India
| | - Mohammed Ali Alshehri
- Department of Biology, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha 61421, Saudi Arabia
| | - Sojin Kang
- Department of Pathology, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02453, Republic of Korea
| | - Seungjoon Moon
- Department of Pathology, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02453, Republic of Korea
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02453, Republic of Korea
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02453, Republic of Korea
| |
Collapse
|
9
|
Zou J, Zheng Z, Ye W, Jin M, Yang P, Little PJ, Wang J, Liu Z. Targeting the smooth muscle cell KEAP1-Nrf2-STING axis with pterostilbene attenuates abdominal aortic aneurysm. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155696. [PMID: 38763007 DOI: 10.1016/j.phymed.2024.155696] [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: 12/21/2023] [Revised: 04/27/2024] [Accepted: 04/29/2024] [Indexed: 05/21/2024]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a life-threatening aortic disease, and to date, there are currently no effective pharmacological treatments to address this condition. Activation of cytosolic DNA sensing adaptor stimulator of interferon genes (STING) signaling is a crucial mechanism in AAA formation. PURPOSE This study investigated pterostilbene (Pt), a naturally occurring polyphenol and resveratrol analogue, as a STING inhibitor for preventing AAA. METHODS We evaluated the effect of Pt on AAA formation in angiotensin II (AngII)-infused apolipoprotein E-deficient (ApoE-/-) mice. We used histological analysis, MMP activity measurement, western blot, and immunohistochemistry to detect AAA formation and development. We applied RNA sequencing, molecular docking, cellular thermal shift assay (CETSA) and functional studies to dissect the molecular mechanism of Pt-regulating KEAP1-Nrf2-STING signaling. We conditionally knocked down Nrf2 in vascular smooth muscle cells (VSMCs) in vivo to investigate its role in Pt-mediated protective effects on AAA. RESULTS Pt effectively blocked the formation of AAA in AngII-infused ApoE-/- mice. Whole transcriptome sequencing analysis revealed that nuclear factor erythroid 2-related factor 2 (Nrf2) and STING pathway in VSMCs were linked to the anti-AAA effects of pterostilbene. Mechanistically, Pt upregulated Nrf2 target genes (e.g., HO-1 and NQO1) through activation of the KEAP1/Nrf2 signaling, which restricted the immunostimulatory axis of mtDNA-STING-TBK1-NF-κB, thereby alleviating VSMC inflammation and preserving the VSMC contractile phenotype. Subsequently, molecular docking and CETSA revealed a binding mode between Pt and KEAP1/Nrf2. Intriguingly, the inhibitory effect of Pt on STING signaling and the protective role of Pt in AAA were largely abrogated by VSMC-specific Nrf2 knockdown in mice. CONCLUSION Collectively, naturally derived Pt shows promising efficacy for the treatment of AAA by targeting the KEAP1-Nrf2-STING axis in VSMCs.
Collapse
Affiliation(s)
- Jiami Zou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Zhihua Zheng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Weile Ye
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Mei Jin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Pinglian Yang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Peter J Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD 4102, Australia; Department of Pharmacy, Guangzhou Xinhua University, Guangzhou 510520, China
| | - Jiaojiao Wang
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong, 523808, China.
| | - Zhiping Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
10
|
Uchikawa H, Uekawa K, Hasegawa Y. Perivascular macrophages in cerebrovascular diseases. Exp Neurol 2024; 374:114680. [PMID: 38185314 DOI: 10.1016/j.expneurol.2024.114680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/10/2023] [Accepted: 01/02/2024] [Indexed: 01/09/2024]
Abstract
Cerebrovascular diseases are a major cause of stroke and dementia, both requiring long-term care. These diseases involve multiple pathophysiologies, with mitochondrial dysfunction being a crucial contributor to the initiation of inflammation, apoptosis, and oxidative stress, resulting in injuries to neurovascular units that include neuronal cell death, endothelial cell death, glial activation, and blood-brain barrier disruption. To maintain brain homeostasis against these pathogenic conditions, brain immune cells, including border-associated macrophages and microglia, play significant roles as brain innate immunity cells in the pathophysiology of cerebrovascular injury. Although microglia have long been recognized as significant contributors to neuroinflammation, attention has recently shifted to border-associated macrophages, such as perivascular macrophages (PVMs), which have been studied based on their crucial roles in the brain. These cells are strategically positioned around the walls of brain vessels, where they mainly perform critical functions, such as perivascular drainage, cerebrovascular flexibility, phagocytic activity, antigen presentation, activation of inflammatory responses, and preservation of blood-brain barrier integrity. Although PVMs act as scavenger and surveillant cells under normal conditions, these cells exert harmful effects under pathological conditions. PVMs detect mitochondrial dysfunction in injured cells and implement pathological changes to regulate brain homeostasis. Therefore, PVMs are promising as they play a significant role in mitochondrial dysfunction and, in turn, disrupt the homeostatic condition. Herein, we summarize the significant roles of PVMs in cerebrovascular diseases, especially ischemic and hemorrhagic stroke and dementia, mainly in correlation with inflammation. A better understanding of the biology and pathobiology of PVMs may lead to new insights on and therapeutic strategies for cerebrovascular diseases.
Collapse
Affiliation(s)
- Hiroki Uchikawa
- Department of Translational Neuroscience, Barrow Aneurysm and AVM Research Center, Barrow Neurological Institute, Phoenix, AZ, USA; Department of Neurosurgery, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Kumamoto, Japan
| | - Ken Uekawa
- Department of Neurosurgery, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Kumamoto, Japan
| | - Yu Hasegawa
- Department of Pharmaceutical Science, School of Pharmacy at Fukuoka, International University of Health and Welfare, Okawa, Fukuoka, Japan.
| |
Collapse
|
11
|
Li K, Lin M, Huang K, Han J, Wei L, Miao L, Chen H, Gong Q, Li X, Hu L. Therapeutic effect and mechanism of action of pterostilbene nano drugs in dry eye models. Exp Eye Res 2024; 241:109836. [PMID: 38387712 DOI: 10.1016/j.exer.2024.109836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 02/05/2024] [Accepted: 02/17/2024] [Indexed: 02/24/2024]
Abstract
Dry eye disease is a multifactorial dysfunction of the tear film and ocular surface, with etiology involving inflammation and oxidative stress on the ocular surface. Pterostilbene (PS) is a secondary metabolite extracted from plants, which possesses remarkable anti-inflammatory and antioxidant effects. However, its application is limited by light instability and very poor water solubility. We modified fat-soluble PS into a biparental pterostilbene-glutaric anhydride-arginine-glycine-aspartic acid (PS-GA-RGD) nanomedicine by prodrug ligation of functional peptides. The aim of this study was to explore the protective effect and potential mechanism of PS-GA-RGD on dry eye disease in vitro and in vivo. We demonstrated good long-term biocompatibility of PS-GA-RGD through rabbit eye stimulation test. Lipopolysaccharide (LPS) was used to induce murine macrophages RAW 264.7 to establish an inflammation and oxidative stress model. In this model, PS-GA-RGD effectively reduced the production of ROS and 8-OHdG, enhancing the expression of antioxidant factor Nrf2 and antioxidant enzyme heme oxygenase-1. In addition, the expression of NF-κB inflammatory pathway significantly increased in LPS-induced RAW 264.7 cells, while PS-GA-RGD could significantly reduce this pathway. Hypertonic saline was utilized to establish a hypertonic model of human corneal epithelial cells. PS-GA-RGD was found to significantly reduce the production of ROS and NLRP3 inflammasomes in this model, exhibiting superior efficacy compared to PS. Experimental dry eye animal models were co-induced with subcutaneous injection of scopolamine and an intelligently controlled environmental system. We demonstrated that PS-GA-RGD nano drugs can prevent and reduce corneal epithelial cell defects and apoptosis, protect conjunctival goblet cells, and have an excellent anti-inflammatory effect. Finally, we demonstrated that RGD sequence in PS-GA-RGD can enhance cellular uptake, corneal retention, and penetration, thereby increasing their bioavailability and efficacy by a cell uptake assay and rabbit corneal drug retention experiment. Overall, this study highlights the potential of PS-GA-RGD nanomedicines in the treatment of dry eyes.
Collapse
Affiliation(s)
- Kexin Li
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Meng Lin
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Kaiyan Huang
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jiaxin Han
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Linzhi Wei
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Lijie Miao
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Huijuan Chen
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Qianwen Gong
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| | - Xingyi Li
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| | - Liang Hu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
12
|
Di W, Zhao A, Li X, Chen J, Dai Y, Li J, Lei W, Yang Y, Lu H. Pterostilbene protects against H 2 O 2 -induced oxidative stress by regulating GAS6/Axl signaling in HL-1 cells. Cell Biochem Funct 2024; 42:e3956. [PMID: 38403920 DOI: 10.1002/cbf.3956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 02/27/2024]
Abstract
Pterostilbene (PTE, trans-3,5-dimethoxy-4'-hydroxystilbene), a natural plant polyphenol, possesses numerous pharmacological effects, including antioxidant, antidiabetic, antiatherosclerotic, and neuroprotective aspects. This study aims to investigate whether PTE plays a protective role against oxidative stress injury by GAS6/Axl signaling pathway in cardiomyocytes. Hydrogen peroxide (H2 O2 )-induced oxidative stress HL-1 cells were used as models. The mechanism by which PTE protected oxidative stress is investigated by combining cell viability, cell ROS levels, apoptosis assay, molecular docking, quantitative real-time PCR, and western blot analysis. GAS6 shRNA was performed to investigate the involvement of GAS6/Axl pathways in PTE's protective role. The results showed that PTE treatment improved the cell morphology and viability, and inhibited the apoptosis rate and ROS levels in H2 O2 -injured HL-1 cells. Particularly, PTE treatment upregulated the levels of GAS6, Axl, and markers related to oxidative stress, apoptosis, and mitochondrial function related. Molecular docking showed that PTE and GAS6 have good binding ability. Taken together, PTE plays a protective role against oxidative stress injury through inhibiting oxidative stress and apoptosis and improving mitochondrial function. Particularly, GAS6/Axl axis is the surprisingly prominent in the PTE-mediated pleiotropic effects.
Collapse
Affiliation(s)
- Wencheng Di
- Department of Cardiovascular Medicine, National Clinical Research Center for Infectious Diseases, Shenzhen, China
| | - Aizhen Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Xiaoru Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Junmin Chen
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
- Department of Cardiology, Affiliated Hospital, Yan'an University, Yan'an, China
| | - Yongbin Dai
- Department of Cardiovascular Medicine, National Clinical Research Center for Infectious Diseases, Shenzhen, China
| | - Jiawen Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Wangrui Lei
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Hongzhou Lu
- Department of Cardiovascular Medicine, National Clinical Research Center for Infectious Diseases, Shenzhen, China
| |
Collapse
|
13
|
Liu P, Tang W, Xiang K, Li G. Pterostilbene in the treatment of inflammatory and oncological diseases. Front Pharmacol 2024; 14:1323377. [PMID: 38259272 PMCID: PMC10800393 DOI: 10.3389/fphar.2023.1323377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
Pterostilbene (PTS), a naturally occurring analog of resveratrol (RSV), has garnered significant attention due to its potential therapeutic effects in treating inflammatory and oncological diseases. This comprehensive review elucidates the pharmacological properties, mechanisms of action, and therapeutic potential of PTS. Various studies indicate that PTS exhibits anti-inflammatory, antioxidant, and antitumour properties, potentially making it a promising candidate for clinical applications. Its influence on regulatory pathways like NF-κB and PI3K/Akt underscores its diverse strategies in addressing diseases. Additionally, PTS showcases a favorable pharmacokinetic profile with better oral bioavailability compared to other stilbenoids, thus enhancing its therapeutic potential. Given these findings, there is an increased interest in incorporating PTS into treatment regimens for inflammatory and cancer-related conditions. However, more extensive clinical trials are imperative to establish its safety and efficacy in diverse patient populations.
Collapse
Affiliation(s)
- Peijun Liu
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Weihua Tang
- Department of Radiology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Kali Xiang
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Guangcai Li
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| |
Collapse
|
14
|
Qu X, Zhang L, Wang L. Pterostilbene as a Therapeutic Alternative for Central Nervous System Disorders: A Review of the Current Status and Perspectives. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:14432-14457. [PMID: 37786984 DOI: 10.1021/acs.jafc.3c06238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
Neurological disorders are diverse, have complex causes, and often result in disability; yet, effective treatments remain scarce. The resveratrol derivative pterostilbene possesses numerous physiological activities that hold promise as a novel therapy for the central nervous system (CNS) disorders. This review aimed to summarize the protective mechanisms of pterostilbene in in vitro and in vivo models of CNS disorders and the pharmacokinetics and safety to assess its possible effects on CNS disorders. Available evidence supports the protective effects of pterostilbene in CNS disorders involving mechanisms such as antioxidant and anti-inflammatory activity, regulation of lipid metabolism and vascular smooth muscle cell proliferation, improvement of synaptic function and neurogenesis, induction of glioma cell cycle arrest, and inhibition of glioma cell migration and invasion. Studies have identified possible molecular targets and pathways for the protective actions of pterostilbene in CNS disorders including the AMPK/STAT3, Akt, NF-κB, MAPK, and ERK signaling pathways. The possible pharmacological effects and molecular pathways of pterostilbene in CNS disorders are critically discussed in this review. Future studies should aim to increase our understanding of pterostilbene in animal models and humans to further evaluate its role in CNS disorders and the detailed mechanisms.
Collapse
Affiliation(s)
- Xin Qu
- Department of Orthopedics, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, P.R. China
| | - Lijuan Zhang
- Departments of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang 110000, Liaoning, P.R. China
| | - Lin Wang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang 110000, Liaoning, P.R. China
| |
Collapse
|
15
|
Wang X, Zhang J, Wang S, Song Z, Sun H, Wu F, Lin X, Jin K, Jin X, Wang W, Lin Q, Wang F. Berberine modulates gut microbiota to attenuate cerebral ferroptosis induced by ischemia-reperfusion in mice. Eur J Pharmacol 2023:175782. [PMID: 37245860 DOI: 10.1016/j.ejphar.2023.175782] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 04/21/2023] [Accepted: 05/10/2023] [Indexed: 05/30/2023]
Abstract
Ferroptosis was reported to be involved in cerebral ischemia-reperfusion injury (CIRI), on which the effects of berberine (BBR) remain unclear. Moreover, based on the critical role of gut microbiota in pleiotropic actions of BBR, we hypothesized that BBR can suppress CIRI-induced ferroptosis by modulating the gut microbiota. In this study, the results showed that BBR obviously attenuated the behavioral deficits of CIRI mice, accompanied with the improved survival rate and neuron damages, as phenocopied by dirty cage experiment. The typical morphological changes in ferroptotic cells and biomarkers of ferroptosis were attenuated in BBR- and its fecal microbiota-treated mice, accompanied by reduced malondialdehyde and reactive oxygen species, and the increased glutathione (GSH). BBR was found to alter the gut microbiota of CIRI mice with decreased abundance of Muribaculaceae, Erysipelotrichaceae, Helicobacteraceae, Streptococcaceae and Tannerellaceae, but elevated Bacteroidaceae and Enterobacteriaceae. KEGG analysis based on the 16S rRNA results indicated that multiple metabolic pathways including ferroptosis and GSH metabolism, were altered by BBR. Oppositely, the antibiotics administration counteracted the protective properties of BBR. Summarily, this study revealed the therapeutic potential of BBR on CIRI via inhibiting neuronal ferroptosis, in which upregulated glutathione peroxidase 1 (GPX1) was possibly involved. Moreover, the BBR-modulated gut microbiota was shown to play the critical role in the underlying mechanism.
Collapse
Affiliation(s)
- Xinyu Wang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jiamin Zhang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Sisi Wang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Zhengyang Song
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Hongxia Sun
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Fangquan Wu
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Xiaohui Lin
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Keke Jin
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Xiaofeng Jin
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Wantie Wang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| | - Qiongqiong Lin
- Department of Pathology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Fangyan Wang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
16
|
Li B, Zhang B, Li Z, Li S, Li J, Wang A, Hou J, Xu J, Zhang R. Ginkgolide C attenuates cerebral ischemia/reperfusion-induced inflammatory impairments by suppressing CD40/NF-κB pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023; 312:116537. [PMID: 37094696 DOI: 10.1016/j.jep.2023.116537] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 04/06/2023] [Accepted: 04/19/2023] [Indexed: 05/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ginkgo biloba L. (Ginkgoaceae), a traditional Chinese medicine, has been applied for thousands of years for the treatment of cardio-cerebral vascular diseases in China. It is written in Compendium of Materia Medica that Ginkgo has the property of "dispersing poison", which is now referred to as anti-inflammatory and antioxidant. Ginkgolides are important active ingredients in Ginkgo biloba leaves and ginkgolide injection has been frequently applied in clinical practice for the treatment of ischemic stroke. However, few studies have explored the effect and mechanism of ginkgolide C (GC) with anti-inflammatory activity in cerebral ischemia/reperfusion injury (CI/RI). AIM OF THE STUDY The present study aimed to demonstrate whether GC was capable of attenuating CI/RI. Furthermore, the anti-inflammatory effect of GC in CI/RI was explored around the CD40/NF-κB pathway. MATERIALS AND METHODS In vivo, middle cerebral artery occlusion/reperfusion (MCAO/R) model was established in rats. The neuroprotective effect of GC was assessed by neurological scores, cerebral infarct rate, microvessel ultrastructure, blood-brain barrier (BBB) integrity, brain edema, neutrophil infiltration, and levels of TNF-α, IL-1β, IL-6, ICAM-1, VCAM-1, and iNOS. In vitro, rat brain microvessel endothelial cells (rBMECs) were preincubated in GC before hypoxia/reoxygenation (H/R) culture. The cell viability, levels of CD40, ICAM-1, MMP-9, TNF-α, IL-1β, and IL-6, and activation of NF-κB pathway were examined. In addition, the anti-inflammatory effect of GC was also investigated by silencing CD40 gene in rBMECs. RESULTS GC attenuated CI/RI as demonstrated by decreasing neurological scores, reducing cerebral infarct rate, improving microvessel ultrastructural features, ameliorating BBB disruption, attenuating brain edema, inhibiting MPO activity, and downregulating levels of TNF-α, IL-1β, IL-6, ICAM-1, VCAM-1, and iNOS. Coherently, in rBMECs exposed to H/R GC enhanced cell viability and downregulated levels of ICAM-1, MMP-9, TNF-α, IL-1β, and IL-6. Furthermore, GC suppressed CD40 overexpression and hindered translocation of NF-κB p65 from the cytosol to the nucleus, phosphorylation of IκB-α, and activation of IKK-β in H/R rBMECs. However, GC failed to protect rBMECs from H/R-induced inflammatory impairments and suppress activation of NF-κB pathway when CD40 gene was silenced. CONCLUSIONS GC attenuates cerebral ischemia/reperfusion-induced inflammatory impairments by suppressing CD40/NF-κB pathway, which may provide an available therapeutic drug for CI/RI.
Collapse
Affiliation(s)
- Bin Li
- Graduate Department, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China; Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Baoke Zhang
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Zhenyu Li
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Shasha Li
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Jun Li
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Aiwu Wang
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Jinling Hou
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Jiping Xu
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Rui Zhang
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| |
Collapse
|
17
|
Kongsui R, Jittiwat J. Ameliorative effects of 6‑gingerol in cerebral ischemia are mediated via the activation of antioxidant and anti‑inflammatory pathways. Biomed Rep 2023; 18:26. [PMID: 36909941 PMCID: PMC9996095 DOI: 10.3892/br.2023.1608] [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: 11/10/2022] [Accepted: 02/01/2023] [Indexed: 02/17/2023] Open
Abstract
Focal ischemia occurs when an embolus or thrombus occludes an artery, causing the rapid obstruction of cerebral blood flow. Although stroke represents a main cause of disability and mortality in developing countries, therapeutic approaches available for this condition remain very limited. The aim of the present study was to examine the effects of the phytochemical, 6-gingerol, on the brain infarct volume, neuronal loss and on the oxidative stress parameters, cyclooxygenase-2 (COX-2) and interleukin (IL)-6, in an animal model of focal ischemic stroke. Male Wistar rats, weighing 250-300 g, were divided into the following six groups: i) The control; ii) right middle cerebral artery occlusion (Rt.MCAO) + vehicle; iii) Rt.MCAO + piracetam; iv) Rt.MCAO + 6-gingerol (6-Gin) at 5 mg/kg body weight (BW); v) Rt.MCAO + 6-Gin at 10 mg/kg BW; and vi) the Rt.MCAO + 6-Gin at 20 mg/kg BW group. The rats in each group received the vehicle or piracetam or 6-gingerol intraperitoneally for 7 days following Rt.MCAO. The brain infarct volume, neuronal loss and alterations in antioxidant and anti-inflammatory levels were assessed in the cortex and hippocampus. The results revealed that the brain infarct volume, malondialdehyde level and the density ratio of COX-2 and IL-6 to β-actin were significantly decreased following treatment with 6-gingerol. In addition, neuronal density and superoxide dismutase activity in the cortex and hippocampus were increased. On the whole, the findings of the present study suggest that 6-gingerol exerts antioxidant and anti-inflammatory effects in vivo, which effectively ameliorate the brain damage induced by focal cerebral ischemic strok.
Collapse
Affiliation(s)
- Ratchaniporn Kongsui
- Division of Physiology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand.,The Unit of Excellence in Translational Neurosciences Initiative, University of Phayao, Phayao 56000, Thailand
| | - Jinatta Jittiwat
- Faculty of Medicine, Mahasarakham University, Mahasarakham 44000, Thailand
| |
Collapse
|
18
|
Chen X, Song QL, Li ZH, Ji R, Wang JY, Cao ML, Mu XF, Zhang Y, Guo DY, Yang J. Pterostilbene ameliorates oxidative damage and ferroptosis in human ovarian granulosa cells by regulating the Nrf2/HO-1 pathway. Arch Biochem Biophys 2023; 738:109561. [PMID: 36898621 DOI: 10.1016/j.abb.2023.109561] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 02/16/2023] [Accepted: 03/05/2023] [Indexed: 03/11/2023]
Abstract
The survival of ovarian granulosa cells is of great significance to the physiological maintenance of the ovary. Oxidative damage to the ovarian granulosa cells can lead to various diseases related to ovarian dysfunction. Pterostilbene exerts many pharmacological effects, such as anti-inflammatory and cardiovascular protective effects. Moreover, pterostilbene was shown to have antioxidant properties. This study aimed to investigate the effect and underlying mechanism of pterostilbene on oxidative damage in ovarian granulosa cells. Ovarian granulosa cell (OGC) lines COV434 and KGN were exposed to H2O2 to establish an oxidative damage model. After treatment with different concentrations of H2O2 or pterostilbene, the cell viability, mitochondrial membrane potential, oxidative stress, and iron levels were detected, and the expression of ferroptosis-related and Nrf2/HO-1 signaling pathway-related proteins were evaluated. Pterostilbene treatment could effectively improve cell viability, reduce oxidative stress, and inhibit ferroptosis stimulated by H2O2. More importantly, pterostilbene could up-regulate Nrf2 transcription by stimulating histone acetylation, and inhibition of Nrf2 signaling could reverse the therapeutic effect of pterostilbene. In conclusion, this research shows that pterostilbene protects human OGCs from oxidative stress and ferroptosis through the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Xin Chen
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei, China
| | - Qian Lin Song
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ze Hong Li
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei, China
| | - Rui Ji
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei, China
| | - Jia Yu Wang
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei, China
| | - Ming Liang Cao
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xue Feng Mu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yan Zhang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Duan Ying Guo
- Longgang District People's Hospital of Shenzhen, Shenzhen, China.
| | - Jing Yang
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei, China.
| |
Collapse
|
19
|
Xue Y, Gu M, Chen C, Yao Y, Li Y, Weng G, Gu Y. Apolipoprotein E mimetic peptide COG1410 alleviates blood‑brain barrier injury in a rat model of ischemic stroke. Mol Med Rep 2023; 27:85. [PMID: 36866740 PMCID: PMC10018278 DOI: 10.3892/mmr.2023.12972] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/15/2023] [Indexed: 03/04/2023] Open
Abstract
Blood‑brain barrier (BBB) damage is one of the main causes of poor outcomes and increased mortality rates following cerebral ischemia‑reperfusion injury. Apolipoprotein E (ApoE) and its mimetic peptide have been previously reported to exhibit potent neuroprotective properties in various central nervous system disease models. Therefore, the present study aimed to investigate the possible role of the ApoE mimetic peptide COG1410 in cerebral ischemia‑reperfusion injury and its potential underlying mechanism. Male SD rats were subjected to 2 h middle cerebral artery occlusion followed by 22 h reperfusion. Evans blue leakage and IgG extravasation assays results revealed that COG1410 treatment significantly reduced BBB permeability. In addition, in situ zymography and western blotting were used to prove that COG1410 was able to downregulate the activities of MMPs and upregulate the expression of occludin in the ischemic brain tissue samples. Subsequently, COG1410 was found to significantly reverse microglia activation while also suppressing inflammatory cytokine production, according to immunofluorescence signal of Iba‑1 and CD68 and protein expression of COX‑2. Consequently, this neuroprotective mechanism mediated by COG1410 was further tested using the BV2 cell line in vitro, which was exposed to oxygen glucose deprivation followed by reoxygenation. The mechanism of COG1410 was found to be mediated, as least partly, through the activation of triggering receptor expressed on myeloid cells 2. In conclusion, the data suggest that COG1410 can alleviate BBB injury and neuroinflammation following ischemic stroke.
Collapse
Affiliation(s)
- Yunwen Xue
- Clinical Research Center, Hainan Provincial Hospital of Traditional Chinese Medicine, Haikou, Hainan 570203, P.R. China
| | - Minhua Gu
- Clinical Research Center, Hainan Provincial Hospital of Traditional Chinese Medicine, Haikou, Hainan 570203, P.R. China
| | - Cuilan Chen
- Clinical Research Center, Hainan Provincial Hospital of Traditional Chinese Medicine, Haikou, Hainan 570203, P.R. China
| | - Yujian Yao
- Clinical Research Center, Hainan Provincial Hospital of Traditional Chinese Medicine, Haikou, Hainan 570203, P.R. China
| | - Yuzhen Li
- Clinical Research Center, Hainan Provincial Hospital of Traditional Chinese Medicine, Haikou, Hainan 570203, P.R. China
| | - Guohu Weng
- Hainan Clinical Research Center for Preventive Treatment of Diseases, Hainan Provincial Hospital of Traditional Chinese Medicine, Haikou, Hainan 570203, P.R. China
| | - Yong Gu
- Clinical Research Center, Hainan Provincial Hospital of Traditional Chinese Medicine, Haikou, Hainan 570203, P.R. China
| |
Collapse
|
20
|
Xu S, Li X, Li Y, Li X, Lv E, Zhang X, Shi Y, Wang Y. Neuroprotective effect of Dl-3-n-butylphthalide against ischemia-reperfusion injury is mediated by ferroptosis regulation via the SLC7A11/GSH/GPX4 pathway and the attenuation of blood-brain barrier disruption. Front Aging Neurosci 2023; 15:1028178. [PMID: 36909944 PMCID: PMC9995665 DOI: 10.3389/fnagi.2023.1028178] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 02/07/2023] [Indexed: 02/25/2023] Open
Abstract
Background Stroke is one of the most severe diseases worldwide, resulting in physical and mental problems. Dl-3-n-butylphthalide, a compound derived from celery seed, has been approved for treating ischemic stroke in China. No study has evaluated how Dl-3-n-butylphthalide affects the ferroptosis SLC7A11/GSH/GPX4 signal pathway and blood-brain barrier (BBB) PDGFRβ/PI3K/Akt signal pathways in the rat middle cerebral artery occlusion/reperfusion (MCAO/R) model of ischemic stroke. Methods Sprague-Dawley rats were used to develop the MCAO/R model. Our study used three incremental doses (10, 20, and 30) of Dl-3-n-butylphthalide injected intraperitoneally 24 h after MCAO/R surgery. The neuroprotective effect and success of the model were evaluated using the neurofunction score, brain water content determination, and triphenyl-tetrazolium chloride-determined infarction area changes. Pathological changes in the brain tissue and the degree of apoptosis were examined by hematoxylin and eosin, Nissl, and terminal deoxynucleotidyl transferase dUTP nick end labeling staining. In addition, pathway proteins and RNA expression levels were studied to verify the effects of Dl-3-n-butyphthalide on both pathways. At the same time, commercial kits were used to detect glutathione, reactive oxygen species, and malondialdehyde, to detect oxidative stress in brain tissues. Results The middle dose of Dl-3-n-butylphthalide not only improved MCAO-induced brain dysfunction and alleviated pathological damage, brain inflammatory response, oxidative stress, and apoptosis but also protected against ferroptosis and reduced BBB damage. These changes resulted in improved neurological function in the cerebral cortex. Conclusion We speculate that Dl-3-n-butylphthalide has a neuroprotective effect on focal cerebral ischemia/reperfusion, which may be mediated through ferroptosis-dependent SLC7A11/GSH/GPX4 signal pathway and PDGFRβ/PI3/Akt signal pathway.
Collapse
Affiliation(s)
- Shuangli Xu
- Emergency Department, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Xuewei Li
- Department of Rheumatology, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Yutian Li
- School of Pharmacy, Weifang Medical University, Weifang, Shandong, China
| | - Xiangling Li
- Department of Internal Medicine, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - E Lv
- Department of Histology and Embryology, Weifang Medical University, Weifang, Shandong, China
| | - Xiaojun Zhang
- Department II of Neurology, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Youkui Shi
- Emergency Department, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Yanqiang Wang
- Department of Rheumatology, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| |
Collapse
|
21
|
Ciccone L, Nencetti S, Marino M, Battocchio C, Iucci G, Venditti I, Marsotto M, Montalesi E, Socci S, Bargagna B, Orlandini E. Pterostilbene fluorescent probes as potential tools for targeting neurodegeneration in biological applications. J Enzyme Inhib Med Chem 2022; 37:1812-1820. [PMID: 35758192 PMCID: PMC9246042 DOI: 10.1080/14756366.2022.2091556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Several epidemiological studies suggest that a diet rich in fruit and vegetables reduces the incidence of neurodegenerative diseases. Resveratrol (Res) and its dimethylated metabolite, pterostibene (Ptb), have been largely studied for their neuroprotective action. The clinical use of Res is limited because of its rapid metabolism and its poor bioavailability. Ptb with two methoxy groups and one hydroxyl group has a good membrane permeability, metabolic stability and higher in vivo bioavailability in comparison with Res. The metabolism and pharmacokinetics of Ptb are still sparse, probably due to the lack of tools that allow following the Ptb destiny both in living cells and in vivo. In this contest, we propose two Ptb fluorescent derivatives where Ptb has been functionalised by benzofurazan and rhodamine-B-isothiocyanate, compounds 1 and 2, respectively. Here, we report the synthesis, the optical and structural characterisation of 1 and 2, and, their putative cytotoxicity in two different cell lines.
Collapse
Affiliation(s)
- Lidia Ciccone
- Department of Pharmacy, University of Pisa, Pisa, Italy.,CISUP - Centre for Instrumentation Sharing, University of Pisa, Pisa, Italy
| | - Susanna Nencetti
- Department of Pharmacy, University of Pisa, Pisa, Italy.,CISUP - Centre for Instrumentation Sharing, University of Pisa, Pisa, Italy
| | - Maria Marino
- Department of Science, University Roma Tre, Rome, Italy
| | | | | | - Iole Venditti
- Department of Science, University Roma Tre, Rome, Italy
| | | | | | - Simone Socci
- Department of Earth Science, University of Pisa, Pisa, Italy
| | | | - Elisabetta Orlandini
- Department of Earth Science, University of Pisa, Pisa, Italy.,Research Centre E. Piaggio, University of Pisa, Pisa, Italy
| |
Collapse
|
22
|
Uysal E, Dokur M, Kucukdurmaz F, Altınay S, Polat S, Batcıoglu K, Sezgın E, Sapmaz Erçakallı T, Yaylalı A, Yılmaztekin Y, Cetın Z, Saygılı İ, Barut O, Kazımoglu H, Maralcan G, Koc S, Guney T, Eser N, Sökücü M, Dokur SN. Targeting the PANoptosome with 3,4-Methylenedioxy-β-Nitrostyrene, Reduces PANoptosis and Protects the Kidney against Renal İschemia-Reperfusion Injury. J INVEST SURG 2022; 35:1824-1835. [PMID: 36170987 DOI: 10.1080/08941939.2022.2128117] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/23/2022] [Accepted: 09/17/2022] [Indexed: 10/14/2022]
Abstract
OBJECTIVES The objectives of this study were a) to investigate the effect of targeting the PANoptosome with 3,4-methylenedioxy-β-nitrostyrene (MNS) on PANoptosis in the Renal ischemia-reperfussion (RIR) model b) to investigate the kidney protective effect of MNS toward RIR injury. METHODS Thirty-two rats were divided into four groups randomly. The groups were assigned as Control, Sham, DMSO (dimethyl sulfoxide) and MNS groups. The rats in the MNS group were intraperitoneally given 20 mg/kg of MNS 30 minutes before reperfusion. 2% DMSO solvent that dissolves MNS were given to the rats in DMSO group. Left nephrectomy was performed on the rats under anesthesia at the 6th hour after reperfusion. Glutathione peroxidase (GPx), malondialdehyde (MDA), catalase (CAT), superoxide dismutase (SOD) and 8-Okso-2'-deoksiguanozin (8-OHdG) levels were measured. Immunohistochemical analysis, electron microscopic and histological examinations were carried out in the tissues. RESULTS Total tubular injury score was lower in the MNS group (p < 0.001). Caspase-3, Gasdermin D and MLK (Mixed Lineage Kinase Domain Like Pseudokinase) expressions were considerably decreased in the MNS group (p < 0.001). Apoptotic index (AI) was found to be low in the MNS group (p < 0.001). CAT and SOD levels were higher in the MNS Group (p = 0.006, p = 0.0004, respectively). GPx, MDA, and 8-OH-dG levels were similar (p > 0.05) in all groups. MNS considerably improved the tissue structure, based on the electron microscopic analysis. CONCLUSIONS Our results suggested that MNS administrated before the reperfusion reduces pyroptosis, apoptosis and necroptosis. These findings suggest that MNS significantly protects the kidney against RIR injury by reducing PANoptosis as a result of specific inhibition of Nod-like receptor pyrin domain-containing 3 (NLRP 3), one of the PANoptosome proteins.
Collapse
Affiliation(s)
- Erdal Uysal
- Department of General Surgery, Sanko University School of Medicine, Gaziantep, Turkey
| | - Mehmet Dokur
- Department of Emergency Medicine, Biruni University Faculty of Medicine, İstanbul, Turkey
| | - Faruk Kucukdurmaz
- Department of Urology, Sanko University School of Medicine, Gaziantep, Turkey
| | - Serdar Altınay
- Deparment of Pathology, Bakırköy Dr Sadi Konuk Health Aplication and Research Center, University of Health Sciences, School of Medicine, İstanbul, Turkey
| | - Sait Polat
- Department of Histology and Embryology, Çukurova University, Faculty of Medicine, Adana, Turkey
| | - Kadir Batcıoglu
- Department of Biochemistry Malatya, Inonu University Faculty of Pharmacy, Battalgazi, Turkey
| | - Efe Sezgın
- Izmir Yuksek Teknoloji Enstitusu, Laboratory of Nutrigenomics and Epidemiology, Food Engineering, Izmir Institute of Technology, İzmir, Turkey
| | - Tuğçe Sapmaz Erçakallı
- Department of Histology and Embryology, Çukurova University, Faculty of Medicine, Adana, Turkey
| | - Aslı Yaylalı
- Faculty of Medicine, Department of Histology and Embryology and IVF Center, Kahramanmaraş Sütçü İmam University, Kahramanmaraş, Turkey
| | - Yakup Yılmaztekin
- Faculty of Pharmacy Department of Biochemistry, Inonu University, Malatya, Turkey
| | - Zafer Cetın
- Department of Medical Biology, Sanko University School of Medicine, Gaziantep, Turkey
| | - İlker Saygılı
- Department of Biochemistry, Sanko University School of Medicine, Gaziantep, Turkey
| | - Osman Barut
- Department of Urology, Kahramanmaras Sutcu Imam University Faculty of Medicine, Kahramanmaras, Turkey
| | - Hatem Kazımoglu
- Department of Urology, Sanko University School of Medicine, Gaziantep, Turkey
| | - Gokturk Maralcan
- Department of General Surgery, Sanko University School of Medicine, Gaziantep, Turkey
| | - Suna Koc
- Department of Anesthesiology and Reanimation, Biruni University Faculty of Medicine, Istanbul, Turkey
| | - Turkan Guney
- Department of Medical Biochemistry, Beykent University, Faculty of Medicine, Istanbul, Turkey
| | - Nadire Eser
- Department of Pharmacology, Kahramanmaraş Sütçü İmam University, Faculty of Medicine, Kahramanmaraş, Turkey
| | - Mehmet Sökücü
- Department of Patology, Sanko University School of Medicine, Gaziantep, Turkey
| | - Sema Nur Dokur
- Faculty of Medicine, Biruni University, Istanbul, Turkey
| |
Collapse
|
23
|
Zhang L, Sui S, Wang S, Sun J. Neuroprotective Effect of Corosolic Acid Against Cerebral Ischemia-Reperfusion Injury in Experimental Rats. J Oleo Sci 2022; 71:1501-1510. [PMID: 36089398 DOI: 10.5650/jos.ess22130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Several therapeutic approaches were also urgently needed as ischaemic stroke was one of the most common brain disorders. Many phytochemicals have recently been discovered for the advancement of lead-like libraries that are concentrated on the peripheral and central nervous systems. Science does not yet understand how these drugs work, nor do they comprehend their in vivo characteristics. We investigated the potential benefits of corosolic acid (CA) in the treatment of brain injury caused by ischemia/reperfusion (I/R) in adult male Sprague-Dawley rats. Injury occurs after a 2-hour transient occlusion of the posterior cerebral artery and subsequent reperfusion (after 20 hours). Furthermore, the experiment assessed the size of the infarct, the amount of brain water present, as well as the neurofunctional conditions in rats. In the study, several markers of inflammation and cytokines associated with brain injury were measured. The Elisa kit was used in this study to measure the mRNA expression of interleukin-6 (IL-6), interleukin-10 (IL-10), interleukin 1β, TNF-α (tumor necrosis factor), cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2), and nitrous oxide (NO). The CA treatment significantly reduced brain water content, brain infarction volume, neurological scores, and Evans blue leakage (p < 0.001 and p < 0.001). Experimental rats were treated with CA after a significantly reduced level of anti-inflammatory, pro-inflammatory, and oxidative stress mediators was noted in their body tissues and serum (p < 0.001). By suppressing inflammatory responses in rats, CA demonstrated anti-inflammatory and neuroprotective properties.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Neurology, Central Hospital Affiliated to Shandong First Medical University
| | - Songtao Sui
- Department of Neurosurgery, Qingdao West Coast New Area Central Hospital
| | - Si Wang
- Department of Neurology, Central Hospital Affiliated to Shandong First Medical University
| | - Jinbo Sun
- Department of Neurology, Central Hospital Affiliated to Shandong First Medical University
| |
Collapse
|
24
|
New Insights into Dietary Pterostilbene: Sources, Metabolism, and Health Promotion Effects. Molecules 2022; 27:molecules27196316. [PMID: 36234852 PMCID: PMC9571692 DOI: 10.3390/molecules27196316] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/17/2022] [Accepted: 09/21/2022] [Indexed: 11/17/2022] Open
Abstract
Pterostilbene (PTS), a compound most abundantly found in blueberries, is a natural analog of resveratrol. Several plant species, such as peanuts and grapes, produce PTS. While resveratrol has been extensively studied for its antioxidant properties, recent evidence also points out the diverse therapeutic potential of PTS. Several studies have identified the robust pharmacodynamic features of PTS, including better intestinal absorption and elevated hepatic stability than resveratrol. Indeed, due to its higher bioavailability paired with reduced toxicity compared to other stilbenes, PTS has become an attractive drug candidate for the treatment of several disease conditions, including diabetes, cancer, cardiovascular disease, neurodegenerative disorders, and aging. This review article provides an extensive summary of the nutraceutical potential of PTS in various disease conditions while discussing the crucial mechanistic pathways implicated. In particular, we share insights from our studies about the Nrf2-mediated effect of PTS in diabetes and associated complications. Moreover, we elucidate the important sources of PTS and discuss in detail its pharmacokinetics and the range of formulations and routes of administration used across experimental studies and human clinical trials. Furthermore, this review also summarizes the strategies successfully used to improve dietary availability and the bio-accessibility of PTS.
Collapse
|
25
|
Tian Q, Yin H, Li J, Jiang J, Ren B, Liu J. Neuroprotective, Anti-inflammatory Effect of Furanochrome, Visnagin Against Middle Cerebral Ischemia-Induced Rat Model. Appl Biochem Biotechnol 2022; 194:5767-5780. [PMID: 35819694 DOI: 10.1007/s12010-022-04009-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2022] [Indexed: 11/28/2022]
Abstract
In recent years, the medical field had significantly progressed to a greater extent which was evidenced with increased life expectancy and decreased mortality rate. Due to the growth of medical field, numerous communicable diseases are prevented and eradicated, whereas the non-communicable disease incidence has been increased globally. One such non-communicable disease which threatens the global population is stroke. Stroke tends to be the second leading cause of death and disability in older population. In lower- and middle-income countries, increased incidence rate of stroke was also evidenced in younger population which is alarming. Lifestyle changes, poor physical activity, stress, consumption of alcohol, oral contraception, and smoking tend to be the causative agents of stroke. Since thrombus formation is the major pathology of stroke, drugs were targeted to thrombolysis. Currently thrombolytic, antiplatelet, and anticoagulant therapies were given for the stroke patients. But the recovery rate of stroke patients with available drugs is very slow. Hence, it is a need of today to discover a drug with increased recovery rate and decreased or nil side effects. Phytochemicals are the best options to treat such non-communicable chronic diseases. Visnagin is one such compound which is used to regulate blood pressure, treat kidney stones, tumors of bile duct, renal colic, and whooping cough. It possesses anti-inflammatory, neuroprotective, and cardioprotective properties; it was also proven to treat epileptic seizures. In this study, the anti-ischemic effect of a furanochrome visnagin was assessed in in vivo rat model. Middle cerebral ischemic/reperfusion was induced in healthy male Sprague Dawley rats and treated with different concentrations of visnagin. The neuroprotective effect of visnagin against cerebral ischemia-induced rats was assessed by analyzing the neurological score, brain edema, infract volume, and Evans blue leakage. The anti-inflammatory property of visnagin was assessed by quantifying proinflammatory cytokines in serum and brain tissues of cerebral ischemia-induced rats. Prostaglandin E-2, COX-2, and NFκ-β were estimated to assess the anti-ischemic effect of visnagin. Histopathological analysis with H&E staining was performed to confirm the neuroprotective effect of visnagin against cerebral ischemia. Our results authentically confirm that visnagin has prevented the inflammation in brain region of cerebral ischemia-induced rats. The neurological scoring and the quantification of PGE-2, COX-2, and NFκ-β prove the anti-ischemic effect of visnagin. Furthermore, the histopathological analysis of hippocampal region provides evidence to the neuroprotective effect of visnagin against cerebral ischemia. Overall, our study confirms visnagin as a potent alternative drug to treat stroke.
Collapse
Affiliation(s)
- Qiangyuan Tian
- Department of Cerebrovascular Diseases Center, Linyi Traditional Chinese Medical Hospital, No. 211, Jiefang Road, Lanshan District, Linyi City, 276003, Shandong Province, China.
| | - Hua Yin
- Department of Cerebrovascular Diseases Center, Linyi Traditional Chinese Medical Hospital, No. 211, Jiefang Road, Lanshan District, Linyi City, 276003, Shandong Province, China
| | - Jisen Li
- Department of Cerebrovascular Diseases Center, Linyi Traditional Chinese Medical Hospital, No. 211, Jiefang Road, Lanshan District, Linyi City, 276003, Shandong Province, China
| | - Jinggong Jiang
- Department of Cerebrovascular Diseases Center, Linyi Traditional Chinese Medical Hospital, No. 211, Jiefang Road, Lanshan District, Linyi City, 276003, Shandong Province, China
| | - Binbin Ren
- Department of Cerebrovascular Diseases Center, Linyi Traditional Chinese Medical Hospital, No. 211, Jiefang Road, Lanshan District, Linyi City, 276003, Shandong Province, China
| | - Junhui Liu
- Department of Cerebrovascular Diseases Center, Linyi Traditional Chinese Medical Hospital, No. 211, Jiefang Road, Lanshan District, Linyi City, 276003, Shandong Province, China
| |
Collapse
|
26
|
Polyphenols for the Treatment of Ischemic Stroke: New Applications and Insights. Molecules 2022; 27:molecules27134181. [PMID: 35807426 PMCID: PMC9268254 DOI: 10.3390/molecules27134181] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 06/23/2022] [Accepted: 06/28/2022] [Indexed: 11/16/2022] Open
Abstract
Ischemic stroke (IS) is a leading cause of death and disability worldwide. Currently, the main therapeutic strategy involves the use of intravenous thrombolysis to restore cerebral blood flow to prevent the transition of the penumbra to the infarct core. However, due to various limitations and complications, including the narrow time window in which this approach is effective, less than 10% of patients benefit from such therapy. Thus, there is an urgent need for alternative therapeutic strategies, with neuroprotection against the ischemic cascade response after IS being one of the most promising options. In the past few decades, polyphenolic compounds have shown great potential in animal models of IS because of their high biocompatibility and ability to target multiple ischemic cascade signaling pathways, although low bioavailability is an issue that limits the applications of several polyphenols. Here, we review the pathophysiological changes following cerebral ischemia and summarize the research progress regarding the applications of polyphenolic compounds in the treatment of IS over the past 5 years. Furthermore, we discuss several potential strategies for improving the bioavailability of polyphenolic compounds as well as some essential issues that remain to be addressed for the translation of the related therapies to the clinic.
Collapse
|
27
|
Li X, Li S, Ma C, Li T, Yang L. Preparation of baicalin-loaded ligand-modified nanoparticles for nose-to-brain delivery for neuroprotection in cerebral ischemia. Drug Deliv 2022; 29:1282-1298. [PMID: 35467483 PMCID: PMC9045769 DOI: 10.1080/10717544.2022.2064564] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Neuroprotection in cerebral ischemia (CI) has received increasing attention. However, efficient delivery of therapeutic agents to the brain remains a major challenge due to the complex environment of the brain. Nose-to-brain-based delivery is a promising approach. Here, we optimized a nanocarrier formulation of neuroprotective agents that can be used for nose-to-brain delivery by obtaining RVG29 peptide-modified polyethylene glycol–polylactic acid-co-glycolic acid nanoparticles (PEG–PLGA RNPs) that have physicochemical properties that lead to stable and sustained drug release and thereby improve the bioavailability of neuroprotective agents. The brain-targeting ability of PEG–PLGA RNPs administered through nasal inhalation was verified in a rat model of CI. It was found that delivery to the whole brain can be achieved with little delivery to the peripheral circulation. Baicalin (BA) was selected as the neuroprotective agent for delivery. After intranasal administration of BA–PEG–PLGA RNPs, the neurological dysfunction of rats with ischemic brain injury was significantly alleviated, the cerebral infarction area was reduced, and nerve trauma and swelling were relieved. Furthermore, it was demonstrated that the neuroprotective effects of BA in a rat model of CI may be mediated by inhibition of inflammation and alleviation of oxidative stress. The immunohistochemical results obtained after treatment with nanoparticles loaded with BA showed that Nrf2/HO-1 was activated in the area in which ischemic brain damage had occurred and that its expression was significantly higher in the group treated with BA–PEG–PLGA RNPs than in the other groups. The ELISA results showed that the levels of IL-1β, IL-6, and TNF-α were abnormally increased in the serum of rats with cerebral ischemia. After treatment with BA-loaded nanoparticles, IL-1β, IL-6, and TNF-α levels decreased significantly. Oxidative stress was alleviated; the levels of glutathione and superoxide dismutase increased; and the levels of reactive oxygen species and malondialdehyde decreased, in animals to which BA–PEG–PLGA RNPs were delivered by intranasal inhalation. In conclusion, BA–PEG–PLGA RNPs can effectively deliver BA to rats and thereby exert neuroprotective effects against CI.
Collapse
Affiliation(s)
- Xinxin Li
- College of Chinese Medicine, Changchun University of Chinese Medicine, ChangChun, China
| | - Shuling Li
- Affiliated Hospital of Changchun University of Chinese Medicine, ChangChun, China
| | - Chun Ma
- Affiliated Hospital of Changchun University of Chinese Medicine, ChangChun, China
| | - Tieshu Li
- College of Chinese Medicine, Changchun University of Chinese Medicine, ChangChun, China
| | - Lihua Yang
- Affiliated Hospital of Changchun University of Chinese Medicine, ChangChun, China
| |
Collapse
|
28
|
Zhu L, Lu F, Zhang X, Liu S, Mu P. SIRT1 Is Involved in the Neuroprotection of Pterostilbene Against Amyloid β 25-35-Induced Cognitive Deficits in Mice. Front Pharmacol 2022; 13:877098. [PMID: 35496289 PMCID: PMC9047953 DOI: 10.3389/fphar.2022.877098] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by amyloid-β (Aβ) deposits and neurofibrillary tangles. Pterostilbene (PTE), a bioactive component mainly in blueberries, is found to have neuroprotective properties. However, the specific underlying mechanisms of PTE in protecting AD remain unclear. Herein, we explored its effects on Aβ25-35-induced neuronal damage in vivo and in vitro and further compared the roles with its structural analog resveratrol (RES) in improving learning-memory deficits. We found that intragastric administration of PTE (40 mg/kg) displayed more effective neuroprotection on Aβ25-35-induced cognitive dysfunction assessed using the novel object test, Y-maze test, and Morris water maze test. Then, we found that PTE improved neuronal plasticity and alleviated neuronal loss both in vivo and in vitro. Additionally, PTE upregulated the expression of sirtuin-1 (SIRT1) and nuclear factor erythroid 2-related factor 2 (Nrf2) and the level of superoxide dismutase (SOD), and inhibited mitochondria-dependent apoptosis in the Aβ25-35-treated group. However, SIRT1 inhibitor EX527 reversed the neuroprotection and induced a drop in mitochondrial membrane potential in PTE-treated primary cortical neurons. Our data suggest that PTE's enhancing learning-memory ability and improving neuroplasticity might be related to inhibiting mitochondria-dependent apoptosis via the antioxidant effect regulated by SIRT1/Nrf2 in AD.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Biochemistry and Molecular Biology, Shenyang Medical College, Shenyang, China
| | - Fangjin Lu
- Department of Pharmacology, Shenyang Medical College, Shenyang, China
| | - Xiaoran Zhang
- Department of Biochemistry and Molecular Biology, Shenyang Medical College, Shenyang, China
| | - Siyuan Liu
- Department of Biochemistry and Molecular Biology, Shenyang Medical College, Shenyang, China
| | - Ping Mu
- Department of Physiology, Shenyang Medical College, Shenyang, China
| |
Collapse
|