1
|
Lykkesfeldt J, Carr AC, Tveden-Nyborg P. The pharmacology of vitamin C. Pharmacol Rev 2025; 77:100043. [PMID: 39986139 DOI: 10.1016/j.pharmr.2025.100043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 01/14/2025] [Indexed: 02/24/2025] Open
Abstract
Ascorbic acid, the reduced form of vitamin C, is a ubiquitous small carbohydrate. Despite decades of focused research, new metabolic functions of this universal electron donor are still being discovered and add to the complexity of our view of vitamin C in human health. Although praised as an unsurpassed water-soluble antioxidant in plasma and cells, the most interesting functions of vitamin C seem to be its roles as specific electron donor in numerous biological reactions ranging from the well-known hydroxylation of proline to cofactor for the epigenetic master regulators ten-eleven translocation enzymes and Jumonji domain-containing histone-lysine demethylases. Some of these functions may have important implications for disease prevention and treatment and have spiked renewed interest in, eg, vitamin C's potential in cancer therapy. Moreover, some fundamental pharmacokinetic properties of vitamin C remain to be established including if other mechanisms than passive diffusion governs the efflux of ascorbate anions from the cell. Taken together, there still seems to be much to learn about the pharmacology of vitamin C and its role in health and disease. This review explores new avenues of vitamin C and integrates our present knowledge of its pharmacology. SIGNIFICANCE STATEMENT: Vitamin C is involved in multiple biological reactions of which most are essential to human health. Hundreds of millions of people are considered deficient in vitamin C according to accepted guidelines, but little is known about the long-term consequences. Although the complexity of vitamin C's physiology and pharmacology has been widely disregarded in clinical studies for decades, it seems clear that a deeper understanding of particularly its pharmacology holds the key to unravel and possibly exploit the potential of vitamin C in disease prevention and therapy.
Collapse
Affiliation(s)
- Jens Lykkesfeldt
- Section of Biomedicine, Department of Veterinary and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Anitra C Carr
- Nutrition in Medicine Research Group, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Pernille Tveden-Nyborg
- Section of Biomedicine, Department of Veterinary and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
2
|
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
|
3
|
Orywal K, Socha K, Iwaniuk P, Kaczyński P, Farhan JA, Zoń W, Łozowicka B, Perkowski M, Mroczko B. Vitamins in the Prevention and Support Therapy of Neurodegenerative Diseases. Int J Mol Sci 2025; 26:1333. [PMID: 39941101 PMCID: PMC11818229 DOI: 10.3390/ijms26031333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 01/31/2025] [Accepted: 02/03/2025] [Indexed: 02/16/2025] Open
Abstract
Neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS), which are a consequence of the progressive loss of neuronal function and structure, cause significant cognitive impairment. The incidence of these diseases in the world's population is constantly increasing as a result of an aging population. Although genetic and environmental factors are most often mentioned as the pathogenetic factors of these diseases, increasing evidence points to the important role of proper nutrition in the prevention and support of the treatment of these disorders. A healthy, balanced diet can mitigate the risks associated with the risk factors mentioned above and slow the progression of the disease by reducing oxidative stress and inflammation. Vitamins B, D, E, C, K, and A have been shown to support cognitive functions and protect the nervous system. This review demonstrates the importance of vitamins in preventing and supporting the therapy of neurodegenerative diseases. Information regarding the health-promoting properties of these vitamins must be effectively communicated to consumers seeking to protect their health, particularly in the context of neurodegenerative diseases. Consequently, this review also examines the authorized health claims under EU food law related to these vitamins, assessing their role in promoting awareness of the vitamins' potential benefits for neuroprotection and the management of neurodegenerative diseases.
Collapse
Affiliation(s)
- Karolina Orywal
- Department of Biochemical Diagnostics, Medical University of Bialystok, Waszyngtona 15A, 15-269 Bialystok, Poland;
| | - Katarzyna Socha
- Department of Bromatology, Medical University of Bialystok, Mickiewicza 2D, 15-222 Bialystok, Poland;
| | - Piotr Iwaniuk
- Institute of Plant Protection—National Research Institute, Chełmońskiego 22, 15-195 Bialystok, Poland; (P.I.); (P.K.); (B.Ł.)
| | - Piotr Kaczyński
- Institute of Plant Protection—National Research Institute, Chełmońskiego 22, 15-195 Bialystok, Poland; (P.I.); (P.K.); (B.Ł.)
| | - Jakub Ali Farhan
- Department of Public International Law and European Law, University of Białystok, Mickiewicza 1, 15-213 Białystok, Poland; (J.A.F.); (W.Z.); (M.P.)
| | - Wojciech Zoń
- Department of Public International Law and European Law, University of Białystok, Mickiewicza 1, 15-213 Białystok, Poland; (J.A.F.); (W.Z.); (M.P.)
| | - Bożena Łozowicka
- Institute of Plant Protection—National Research Institute, Chełmońskiego 22, 15-195 Bialystok, Poland; (P.I.); (P.K.); (B.Ł.)
| | - Maciej Perkowski
- Department of Public International Law and European Law, University of Białystok, Mickiewicza 1, 15-213 Białystok, Poland; (J.A.F.); (W.Z.); (M.P.)
| | - Barbara Mroczko
- Department of Biochemical Diagnostics, Medical University of Bialystok, Waszyngtona 15A, 15-269 Bialystok, Poland;
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, Waszyngtona 15A, 15-269 Bialystok, Poland
| |
Collapse
|
4
|
Farzi A, Teymoor Davani A, Seyed A, Salehi O, Mosallanezhad Z. The effect of eight weeks of aerobic training with vitamin C on some apoptotic markers in the hippocampus tissue of rats with Alzheimer's disease; an experimental study. Neurol Res 2025; 47:77-86. [PMID: 39754544 DOI: 10.1080/01616412.2024.2448624] [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: 05/18/2024] [Accepted: 12/24/2024] [Indexed: 01/06/2025]
Abstract
OBJECTIVES The aim of this study was to investigate the effect of eight weeks of aerobic training (AT) and vitamin C supplementation (VC) on apoptotic markers in hippocampus tissue of AD rats treated with trimethyltin (TMT). MATERIALS AND METHODS In this experimental study, 32 Sprague- Dawley rats (mean age: 14-18 months and mean weight 270-320 g) were treated with (10 mg/kg) TMT and divided into 4 groups including: 1) ADcontrol, 2) VC, 3) AT and 4) AT+VC groups. In order to investigate the effects of AD induction on research variables, 8 healthy rats selected as healthy control group (HC). Groups 3 and 4 trained for eight weeks, three sessions per week and each session lasted 15-48 minutes with an intensity of 10-24 m/min. Groups 2 and 4 received 4 mg/kg VC orally. One-way ANOVA with Tukey's post- hoc tests were used for statistical analysis of data (p ≤ 0.05). RESULTS The gene expression levels of Caspase 3, FasL, Cyt-C and AP-1 in the AT, VC and AT+VC groups were significantly lower than TMT group (p ≤ 0.05); Caspase 3, FasL and Cyt-C levels were significantly lower in the AT+VC group compare to VC and ET groups (p ≤ 0.05). CytC levels in AT group were significantly lower than VC group (p = 0.002). Also, AP-1 levels in AT+VC group were significantly lower than AT group (p = 0.01). CONCLUSIONS It seems that AT and VC, both alone and interactively, can probably induce their anti-apoptotic effects in the hippocampus tissue of rats with AD via a common signaling pathway.
Collapse
Affiliation(s)
- Azadeh Farzi
- Department of Sport Physiology, Behbahan Branch, Islamic Azad University, Behbehan, Iran
| | - Amin Teymoor Davani
- Department of Sport Physiology, Behbahan Branch, Islamic Azad University, Behbehan, Iran
| | - Asiye Seyed
- Department of Sport Physiology, Behbahan Branch, Islamic Azad University, Behbehan, Iran
| | - Omidreza Salehi
- Department of Physical Education and Sport Sciences, University of Kurdistan, Sanandaj, Iran
| | - Zahra Mosallanezhad
- Department of Sport Sciences, Zand Institute of Higher Education, Shiraz, Iran
| |
Collapse
|
5
|
Pekdemir B, Raposo A, Saraiva A, Lima MJ, Alsharari ZD, BinMowyna MN, Karav S. Mechanisms and Potential Benefits of Neuroprotective Agents in Neurological Health. Nutrients 2024; 16:4368. [PMID: 39770989 PMCID: PMC11677798 DOI: 10.3390/nu16244368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/12/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
The brain contains many interconnected and complex cellular and molecular mechanisms. Injury to the brain causes permanent dysfunctions in these mechanisms. So, it continues to be an area where surgical intervention cannot be performed except for the removal of tumors and the repair of some aneurysms. Some agents that can cross the blood-brain barrier and reach neurons show neuroprotective effects in the brain due to their anti-apoptotic, anti-inflammatory and antioxidant properties. In particular, some agents act by reducing or modulating the accumulation of protein aggregates in neurodegenerative diseases (Alzheimer's disease, Parkinson's disease, Huntington's disease, Amyotrophic lateral sclerosis, and prion disease) caused by protein accumulation. Substrate accumulation causes increased oxidative stress and stimulates the brain's immune cells, microglia, and astrocytes, to secrete proinflammatory cytokines. Long-term or chronic neuroinflammatory response triggers apoptosis. Brain damage is observed with neuronal apoptosis and brain functions are impaired. This situation negatively affects processes such as motor movements, memory, perception, and learning. Neuroprotective agents prevent apoptosis by modulating molecules that play a role in apoptosis. In addition, they can improve impaired brain functions by supporting neuroplasticity and neurogenesis. Due to the important roles that these agents play in central nervous system damage or neurodegenerative diseases, it is important to elucidate many mechanisms. This review provides an overview of the mechanisms of flavonoids, which constitute a large part of the agents with neuroprotective effects, as well as vitamins, neurotransmitters, hormones, amino acids, and their derivatives. It is thought that understanding these mechanisms will enable the development of new therapeutic agents and different treatment strategies.
Collapse
Affiliation(s)
- Burcu Pekdemir
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Turkey;
| | - António Raposo
- CBIOS (Research Center for Biosciences and Health Technologies), Universidade Lusófona de Humanidades e Tecnologias, Campo Grande 376, 1749-024 Lisboa, Portugal
| | - Ariana Saraiva
- Research in Veterinary Medicine (I-MVET), Faculty of Veterinary Medicine, Lisbon University Centre, Lusófona University, Campo Grande 376, 1749-024 Lisboa, Portugal;
| | - Maria João Lima
- CERNAS Research Centre, Polytechnic University of Viseu, 3504-510 Viseu, Portugal;
| | - Zayed D. Alsharari
- Department of Clinical Nutrition, Faculty of Applied Medical Sciences, University of Tabuk, P.O. Box 741, Tabuk 71491, Saudi Arabia;
| | - Mona N. BinMowyna
- College of Education, Shaqra University, Shaqra 11911, Saudi Arabia;
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Turkey;
| |
Collapse
|
6
|
Kaya K, Şahin Y, Demirel HH, Çiftçi O. Investigation of oxidative, inflammatory and apoptotic effects of favipiravir use alone and combined with vitamin C on brain tissue of elderly rats. Drug Chem Toxicol 2024; 47:640-648. [PMID: 37424396 DOI: 10.1080/01480545.2023.2233054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 06/09/2023] [Accepted: 06/23/2023] [Indexed: 07/11/2023]
Abstract
Favipiravir is a nucleoside analogue antiviral drug and inhibits the replication of many RNA viruses, especially influenza viruses. Favipiravir has also been used to treat patients with mild to moderate COVID-19 disease. However, various side effects, including neurological side effects, have been reported related to the use of favipiravir. Therefore, in this study, we aimed to investigate the possible effects of favipiravir alone or in combination with vitamin C on aged rats' brain tissue and the possible mechanisms of these effects. A total of 30 rats used in the study were randomly divided into 5 equal groups and the first group was kept as the control group. High-dose (100 mg/kg) or low-dose (20 mg/kg) favipiravir was administered alone or in combination with vitamin C (150 mg/kg) to other groups. Administration of both high and low doses of favipiravir significantly increased TBARS levels in brain tissue of aged rats. Similarly, both high and low doses of favipiravir led to significant increases in Bcl-2 and caspase-3 relative mRNA expression. However, only low dose favipiravir caused a significant increase in iNOS and IL-1β relative mRNA expression levels. Similar results were also observed in histopathological examinations. However, co-administration of vitamin C with favipiravir attenuated some of the adverse effects of favipiravir. In conclusion, in this study, it was shown that the use of favipiravir caused some adverse effects through oxidative, inflammatory and apoptotic processes in the brain tissue of aged rats, and the potential of vitamin C to alleviate these effects.
Collapse
Affiliation(s)
- Kürşat Kaya
- Department of Medical Biochemistry, Faculty of Medicine, Pamukkale University, Denizli, Türkiye
| | - Yasemin Şahin
- Department of Medical Pharmacology, Faculty of Medicine, Pamukkale University, Denizli, Türkiye
| | - Hasan Hüseyin Demirel
- Bayat Laborant & Veterinary Health Division, Afyon Kocatepe University, Afyon, Türkiye
| | - Osman Çiftçi
- Department of Medical Pharmacology, Faculty of Medicine, Pamukkale University, Denizli, Türkiye
| |
Collapse
|
7
|
Li Q, Tang X, Huang L, Wang T, Huang Y, Jiang S. Anti-allergic effect of vitamin C through inhibiting degranulation and regulating T H1/T H2 cell polarization. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:5955-5963. [PMID: 38415860 DOI: 10.1002/jsfa.13419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 01/26/2024] [Accepted: 02/26/2024] [Indexed: 02/29/2024]
Abstract
BACKGROUND Food allergy has become a global public health problem. This study aimed to explore the possible anti-allergic effect of vitamin C (VC). A rat basophilic leukemia (RBL)-2H3 cell degranulation model was used to assess the effect of VC on degranulation in vitro, and an ovalbumin (OVA)-induced BALB/c mouse allergy model was used to assess the anti-allergy effect of VC in vivo. RESULTS In vitro, VC significantly attenuated the release of β-hexosaminidase, tryptase and histamine, and also reduced cytokine production (interleukins 4 and 6, tumor necrosis factor α) significantly (P < 0.05), with the inhibitory effect demonstrating a positive correlation with VC dose. In vivo, compared with the OVA group, the levels of serum immunoglobulins E and G1 of the VC low-dose (VCL) group (50 mg kg-1) and high-dose (VCH) group (200 mg·kg-1) were significantly reduced (P < 0.05). Furthermore, the plasma histamine level was also significantly decreased (P < 0.05). Moreover, TH2 cell polarization in mice of the VCL and VCH groups was significantly inhibited (P < 0.05), promoting the TH1/TH2 cell polarization balance. Additionally, VC treatment enhanced the expression of CD80 (P < 0.05) in spleen and small intestine tissues, while significantly inhibiting the expression of CD86 (P < 0.05); notably, high-dose VC treatment was more effective. CONCLUSION VC exerted an anti-allergic effect through inhibiting degranulation and regulating TH1/TH2 cell polarization balance. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Qian Li
- College of Tourism and Culinary Science, Yangzhou University, Yangzhou, People's Republic of China
| | - Xinlei Tang
- College of Tourism and Culinary Science, Yangzhou University, Yangzhou, People's Republic of China
| | - Lu Huang
- College of Tourism and Culinary Science, Yangzhou University, Yangzhou, People's Republic of China
| | - Tao Wang
- College of Tourism and Culinary Science, Yangzhou University, Yangzhou, People's Republic of China
| | - Yutong Huang
- College of Tourism and Culinary Science, Yangzhou University, Yangzhou, People's Republic of China
| | - Songsong Jiang
- College of Tourism and Culinary Science, Yangzhou University, Yangzhou, People's Republic of China
- Key Laboratory of Chinese Cuisine Intangible Cultural Heritage Technology Inheritance, Ministry of Culture and Tourism, Yangzhou, People's Republic of China
| |
Collapse
|
8
|
Zheng H, Wu H, Wang D, Wang S, Ji D, Liu X, Gao G, Su X, Zhang Y, Ling Y. Research progress of prodrugs for the treatment of cerebral ischemia. Eur J Med Chem 2024; 272:116457. [PMID: 38704941 DOI: 10.1016/j.ejmech.2024.116457] [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: 03/21/2024] [Revised: 04/20/2024] [Accepted: 04/25/2024] [Indexed: 05/07/2024]
Abstract
It is well-known that pharmacotherapy plays a pivotal role in the treatment and prevention of cerebral ischemia. Nevertheless, existing drugs, including numerous natural products, encounter various challenges when applied in cerebral ischemia treatment. These challenges comprise poor brain absorption due to low blood-brain barrier (BBB) permeability, limited water solubility, inadequate bioavailability, poor stability, and rapid metabolism. To address these issues, researchers have turned to prodrug strategies, aiming to mitigate or eliminate the adverse properties of parent drug molecules. In vivo metabolism or enzymatic reactions convert prodrugs into active parent drugs, thereby augmenting BBB permeability, improving bioavailability and stability, and reducing toxicity to normal tissues, ultimately aiming to enhance treatment efficacy and safety. This comprehensive review delves into multiple effective prodrug strategies, providing a detailed description of representative prodrugs developed over the past two decades. It underscores the potential of prodrug approaches to improve the therapeutic outcomes of currently available drugs for cerebral ischemia. The publication of this review serves to enrich current research progress on prodrug strategies for the treatment and prevention of cerebral ischemia. Furthermore, it seeks to offer valuable insights for pharmaceutical chemists in this field, offer guidance for the development of drugs for cerebral ischemia, and provide patients with safer and more effective drug treatment options.
Collapse
Affiliation(s)
- Hongwei Zheng
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Hongmei Wu
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China; Department of Neurosurgery, Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Dezhi Wang
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China; Department of Neurosurgery, Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Sijia Wang
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China; Department of Neurosurgery, Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Dongliang Ji
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China; Department of Neurosurgery, Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Xiao Liu
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Ge Gao
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Xing Su
- Department of Neurosurgery, Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu, PR China.
| | - Yanan Zhang
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China.
| | - Yong Ling
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China.
| |
Collapse
|
9
|
Li B, Xu L, Wang Z, Shi Q, Cui Y, Fan W, Wu Q, Tong X, Yan H. Neutrophil Extracellular Traps Regulate Surgical Brain Injury by Activating the cGAS-STING Pathway. Cell Mol Neurobiol 2024; 44:36. [PMID: 38637346 PMCID: PMC11026279 DOI: 10.1007/s10571-024-01470-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 03/15/2024] [Indexed: 04/20/2024]
Abstract
Surgical brain injury (SBI), induced by neurosurgical procedures or instruments, has not attracted adequate attention. The pathophysiological process of SBI remains sparse compared to that of other central nervous system diseases thus far. Therefore, novel and effective therapies for SBI are urgently needed. In this study, we found that neutrophil extracellular traps (NETs) were present in the circulation and brain tissues of rats after SBI, which promoted neuroinflammation, cerebral edema, neuronal cell death, and aggravated neurological dysfunction. Inhibition of NETs formation by peptidylarginine deiminase (PAD) inhibitor or disruption of NETs with deoxyribonuclease I (DNase I) attenuated SBI-induced damages and improved the recovery of neurological function. We show that SBI triggered the activation of cyclic guanosine monophosphate-adenosine monophosphate synthase stimulator of interferon genes (cGAS-STING), and that inhibition of the cGAS-STING pathway could be beneficial. It is worth noting that DNase I markedly suppressed the activation of cGAS-STING, which was reversed by the cGAS product cyclic guanosine monophosphate-adenosine monophosphate (cGMP-AMP, cGAMP). Furthermore, the neuroprotective effect of DNase I in SBI was also abolished by cGAMP. NETs may participate in the pathophysiological regulation of SBI by acting through the cGAS-STING pathway. We also found that high-dose vitamin C administration could effectively inhibit the formation of NETs post-SBI. Thus, targeting NETs may provide a novel therapeutic strategy for SBI treatment, and high-dose vitamin C intervention may be a promising translational therapy with an excellent safety profile and low cost.
Collapse
Affiliation(s)
- Bingbing Li
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China
| | - Lixia Xu
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, 300350, China
| | - Zhengang Wang
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China
| | - Qi Shi
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China
| | - Yang Cui
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China
| | - Weijia Fan
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, 300350, China
| | - Qiaoli Wu
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, 300350, China
| | - Xiaoguang Tong
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China.
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, 300350, China.
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, 300350, China.
| | - Hua Yan
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China.
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, 300350, China.
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, 300350, China.
| |
Collapse
|
10
|
Li J, Xie F, Ma X. Advances in nanomedicines: a promising therapeutic strategy for ischemic cerebral stroke treatment. Nanomedicine (Lond) 2024; 19:811-835. [PMID: 38445614 DOI: 10.2217/nnm-2023-0266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024] Open
Abstract
Ischemic stroke, prevalent among the elderly, necessitates attention to reperfusion injury post treatment. Limited drug access to the brain, owing to the blood-brain barrier, restricts clinical applications. Identifying efficient drug carriers capable of penetrating this barrier is crucial. Blood-brain barrier transporters play a vital role in nutrient transport to the brain. Recently, nanoparticles emerged as drug carriers, enhancing drug permeability via surface-modified ligands. This article introduces the blood-brain barrier structure, elucidates reperfusion injury pathogenesis, compiles ischemic stroke treatment drugs, explores nanomaterials for drug encapsulation and emphasizes their advantages over conventional drugs. Utilizing nanoparticles as drug-delivery systems offers targeting and efficiency benefits absent in traditional drugs. The prospects for nanomedicine in stroke treatment are promising.
Collapse
Affiliation(s)
- Jun Li
- Faculty of Environment & Life, Beijing University of Technology, Beijing, 100124, PR China
- Beijing Molecular Hydrogen Research Center, Beijing, 100124, PR China
| | - Fei Xie
- Faculty of Environment & Life, Beijing University of Technology, Beijing, 100124, PR China
- Beijing Molecular Hydrogen Research Center, Beijing, 100124, PR China
| | - Xuemei Ma
- Faculty of Environment & Life, Beijing University of Technology, Beijing, 100124, PR China
- Beijing Molecular Hydrogen Research Center, Beijing, 100124, PR China
| |
Collapse
|
11
|
Scotti L, da Silva PR, Monteiro AFM, de Araújo RSA, do Nascimento VL, Monteiro KLC, de Aquino TM, Dos Santos Silva WF, da Silva Junior EF, Scotti MT, Mendonça Junior FJB. The Multitarget Action of Vitamins in the Ischemic Stroke. Curr Top Med Chem 2024; 24:2465-2488. [PMID: 39301898 DOI: 10.2174/0115680266316939240909070627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 08/08/2024] [Accepted: 08/19/2024] [Indexed: 09/22/2024]
Abstract
A stroke, also known as a cerebral hemorrhage, occurs when there is an interruption in the blood supply to a part of the brain, resulting in damage to brain cells. This issue is one of the leading causes of death in developed countries, currently killing about 5 million people annually. Individuals who survive ischemic stroke often face serious vision problems, paralysis, dementia, and other sequelae. The numerous efforts to prevent and/or treat stroke sequelae seem insufficient, which is concerning given the increasing global elderly population and the well-known association between aging and stroke risk. In this review, we aim to present and discuss the importance of vitamins in stroke prevention and/or incidence. Vitamins from diet or dietary supplements influence the body at various levels; they are a relevant factor but are reported only in isolated articles. This review reports and updates the multitarget role of vitamins involved in reducing stroke risk.
Collapse
Affiliation(s)
- Luciana Scotti
- Post-Graduate Program in Natural and Synthetic Bioactive Compounds, Federal University of Paraíba (UFPB), João Pessoa, Paraiba, Brazil
| | - Pablo Rayff da Silva
- Postgraduate Program in Dentistry, Health Sciences Center, Federal University of Paraíba, João Pessoa, Paraíba, Brazil
| | - Alex France M Monteiro
- Post-Graduate Program in Natural and Synthetic Bioactive Compounds, Federal University of Paraíba (UFPB), João Pessoa, Paraiba, Brazil
- Postgraduate Program in Chemistry, Department of Chemistry, Federal Rural University of Pernambuco, Campus I-Recife/PE, Brazil
| | | | - Vanessa Lima do Nascimento
- Research Group on Therapeutic Strategies - GPET, Institute of Chemistry and Biotechnology, Federal University of Alagoas (UFAL), Maceió, Brazil
| | - Kadja Luana Chagas Monteiro
- Research Group on Therapeutic Strategies - GPET, Institute of Chemistry and Biotechnology, Federal University of Alagoas (UFAL), Maceió, Brazil
| | - Thiago Mendonça de Aquino
- Research Group on Therapeutic Strategies - GPET, Institute of Chemistry and Biotechnology, Federal University of Alagoas (UFAL), Maceió, Brazil
| | - Wadja Feitosa Dos Santos Silva
- Research Group on Therapeutic Strategies - GPET, Institute of Chemistry and Biotechnology, Federal University of Alagoas (UFAL), Maceió, Brazil
| | - Edeildo Ferreira da Silva Junior
- Research Group on Therapeutic Strategies - GPET, Institute of Chemistry and Biotechnology, Federal University of Alagoas (UFAL), Maceió, Brazil
| | - Marcus T Scotti
- Post-Graduate Program in Natural and Synthetic Bioactive Compounds, Federal University of Paraíba (UFPB), João Pessoa, Paraiba, Brazil
| | - Francisco Jaime Bezerra Mendonça Junior
- Post-Graduate Program in Natural and Synthetic Bioactive Compounds, Federal University of Paraíba (UFPB), João Pessoa, Paraiba, Brazil
- Laboratory of Synthesys and Drug Delivery - LSVM, State University of Paraíba (UEPB), João Pessoa, Brazil
| |
Collapse
|
12
|
Feng S, Yang M, Liu S, He Y, Deng S, Gong Y. Oxidative stress as a bridge between age and stroke: A narrative review. JOURNAL OF INTENSIVE MEDICINE 2023; 3:313-319. [PMID: 38028635 PMCID: PMC10658045 DOI: 10.1016/j.jointm.2023.02.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 12/09/2022] [Accepted: 02/07/2023] [Indexed: 12/01/2023]
Abstract
Stroke is the third most common cause of death globally and a leading cause of disability. The cellular and molecular changes following stroke and causes of neuronal death are not fully understood, and there are few effective treatments currently available. A rapid increase in the levels of reactive oxygen species (ROS) post stroke can overwhelm antioxidant defenses and trigger a series of pathophysiologic events including the inflammatory response, blood-brain barrier (BBB) disruption, apoptosis, and autophagy, ultimately leading to neuron degeneration and apoptosis. It is thought that beyond a certain age, the ROS accumulation resulting from stroke increases the risk of morbidity and mortality. In the present review, we summarize the role of oxidative stress (OS) as a link between aging and stroke pathogenesis. We also discuss how antioxidants can play a beneficial role in the prevention and treatment of stroke by eliminating harmful ROS, delaying aging, and alleviating damage to neurons.
Collapse
Affiliation(s)
- Shengjie Feng
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Miaoxian Yang
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Shengpeng Liu
- Department of Pediatrics, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, Guangdong, 518020,China
| | - Yu He
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Shuixiang Deng
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ye Gong
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
13
|
Orellana-Urzúa S, Briones-Valdivieso C, Chichiarelli S, Saso L, Rodrigo R. Potential Role of Natural Antioxidants in Countering Reperfusion Injury in Acute Myocardial Infarction and Ischemic Stroke. Antioxidants (Basel) 2023; 12:1760. [PMID: 37760064 PMCID: PMC10525378 DOI: 10.3390/antiox12091760] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Stroke and acute myocardial infarction are leading causes of mortality worldwide. The latter accounts for approximately 9 million deaths annually. In turn, ischemic stroke is a significant contributor to adult physical disability globally. While reperfusion is crucial for tissue recovery, it can paradoxically exacerbate damage through oxidative stress (OS), inflammation, and cell death. Therefore, it is imperative to explore diverse approaches aimed at minimizing ischemia/reperfusion injury to enhance clinical outcomes. OS primarily arises from an excessive generation of reactive oxygen species (ROS) and/or decreased endogenous antioxidant potential. Natural antioxidant compounds can counteract the injury mechanisms linked to ROS. While promising preclinical results, based on monotherapies, account for protective effects against tissue injury by ROS, translating these models into human applications has yielded controversial evidence. However, since the wide spectrum of antioxidants having diverse chemical characteristics offers varied biological actions on cell signaling pathways, multitherapy has emerged as a valuable therapeutic resource. Moreover, the combination of antioxidants in multitherapy holds significant potential for synergistic effects. This study was designed with the aim of providing an updated overview of natural antioxidants suitable for preventing myocardial and cerebral ischemia/reperfusion injuries.
Collapse
Affiliation(s)
- Sofía Orellana-Urzúa
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380000, Chile;
| | | | - Silvia Chichiarelli
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University of Rome, 00185 Rome, Italy;
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Faculty of Pharmacy and Medicine, Sapienza University, P.le Aldo Moro 5, 00185 Rome, Italy;
| | - Ramón Rodrigo
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380000, Chile;
| |
Collapse
|
14
|
Yuan Y, Sheng P, Ma B, Xue B, Shen M, Zhang L, Li D, Hou J, Ren J, Liu J, Yan BC, Jiang Y. Elucidation of the mechanism of Yiqi Tongluo Granule against cerebral ischemia/reperfusion injury based on a combined strategy of network pharmacology, multi-omics and molecular biology. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 118:154934. [PMID: 37393828 DOI: 10.1016/j.phymed.2023.154934] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 06/10/2023] [Accepted: 06/19/2023] [Indexed: 07/04/2023]
Abstract
BACKGROUND Ischemic stroke is caused by local lesions of the central nervous system and is a severe cerebrovascular disease. A traditional Chinese medicine, Yiqi Tongluo Granule (YQTL), shows valuable therapeutic effects. However, the substances and mechanisms remain unclear. PURPOSE We combined network pharmacology, multi-omics, and molecular biology to elucidate the mechanisms by which YQTL protects against CIRI. STUDY DESIGN We innovatively created a combined strategy of network pharmacology, transcriptomics, proteomics and molecular biology to study the active ingredients and mechanisms of YQTL. We performed a network pharmacology study of active ingredients absorbed by the brain to explore the targets, biological processes and pathways of YQTL against CIRI. We also conducted further mechanistic analyses at the gene and protein levels using transcriptomics, proteomics, and molecular biology techniques. RESULTS YQTL significantly decreased the infarction volume percentage and improved the neurological function of mice with CIRI, inhibited hippocampal neuronal death, and suppressed apoptosis. Fifteen active ingredients of YQTL were detected in the brains of rats. Network pharmacology combined with multi-omics revealed that the 15 ingredients regulated 19 pathways via 82 targets. Further analysis suggested that YQTL protected against CIRI via the PI3K-Akt signaling pathway, MAPK signaling pathway, and cAMP signaling pathway. CONCLUSION We confirmed that YQTL protected against CIRI by inhibiting nerve cell apoptosis enhanced by the PI3K-Akt signaling pathway.
Collapse
Affiliation(s)
- Yue Yuan
- Institute for Chinese Materia Medica, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory of TCM Pharmacology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100730, China
| | - Peng Sheng
- Department of Neurology, Medical College, Institute of Translational Medicine, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225001, China
| | - Bo Ma
- Chinese Academy of Medical Sciences & Peking Union Medical College Institute of Materia Medica, Beijing 100730, China
| | - Bingjie Xue
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Mengmeng Shen
- Department of Neurology, Medical College, Institute of Translational Medicine, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225001, China
| | - Ling Zhang
- Department of Neurology, Medical College, Institute of Translational Medicine, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225001, China
| | - Dan Li
- Shineway Pharmaceutical Group Co., Ltd., Shijiazhuang 051430, China
| | - Jincai Hou
- Shineway Pharmaceutical Group Co., Ltd., Shijiazhuang 051430, China
| | - Junguo Ren
- Beijing Key Laboratory of TCM Pharmacology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100730, China
| | - Jianxun Liu
- Beijing Key Laboratory of TCM Pharmacology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100730, China.
| | - Bing Chun Yan
- Department of Neurology, Medical College, Institute of Translational Medicine, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225001, China.
| | - Yunyao Jiang
- Institute for Chinese Materia Medica, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
15
|
Atchie B, Jarvis S, Bellon R, Barton T, Disalvo L, Salottolo K, Bar-Or R, Bar-Or D. Oxidation‑reduction potential parameters worsen following intraarterial therapy in patients with reduced collateral circulation and middle cerebral artery occlusions. Exp Ther Med 2023; 25:295. [PMID: 37229324 PMCID: PMC10203750 DOI: 10.3892/etm.2023.11994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 03/24/2023] [Indexed: 05/27/2023] Open
Abstract
Collateral circulation is important for cerebral perfusion in acute ischemic strokes. Monitoring the oxidation-reduction potential (ORP) may be useful to assess collateral status or treatment efficacy. The objectives of the present study were to determine if the ORP was associated with collateral circulation status in middle cerebral artery (MCA) occlusions and to identify patterns in the ORP and the collateral circulation status among patients treated with intraarterial therapy (IAT) over time. The present pilot study was nested within a prospective cohort study measuring the ORP of the peripheral venous plasma of stroke patients. The population included in the present study were patients with MCA (M1/M2) occlusions. Two ORP parameters were examined: Static ORP (sORP; mV), indicating oxidative stress, and capacity ORP (cORP; µC), indicating antioxidant reserves. Collateral status was retrospectively graded using Miteff's system as good (grade 1) or reduced (grade 2/3). Comparisons were made between collateral status groups (reduced vs. good collaterals) in all patients, within a subset including only patients who received IAT, and between thrombolysis in cerebral infraction scale score (TICI) groups (0-2a vs. 2b/3). The Fisher's exact test, Student's t-test and Wilcoxon tests were used (α<0.20). The 19 patients were categorized based on their collaterals: Good collaterals (53%) and reduced collaterals (47%). The baseline characteristics were similar with the exception that the patients with good collaterals had a lower international normalized ratio (P=0.12) and were more likely to have a stroke on the left side (P=0.18) or to have a mismatch (P=0.05). The admission sORP values were comparable (169.5 vs. 164.2 mV; P=0.65), as was admission cORP (P=0.73). When considering only the patients who received IAT (n=12), admission sORP (P=0.69) and cORP (P=0.90) were also statistically similar. On day 2, after IAT, both groups experienced a worsening in ORP measures; however, the patients with good collaterals had a significantly lower sORP (169.4 vs. 203.5 mV; P=0.02) and a higher cORP (0.2 vs. 0.1 µC; P=0.002) compared with the patients with reduced collaterals. Neither sORP nor cORP were significantly different between TICI score groups on admission or on day 2. Upon discharge, patients with a TICI of 2b-3 had a significantly better sORP (P=0.03) and cORP (P=0.12) compared with those with a TICI of 0-2a. In conclusion, upon patient admission, the ORP parameters were not significantly different between the collateral circulation status groups for MCA occlusions. The ORP parameters worsened after IAT regardless of the collateral circulation status; however, after IAT, on day 2, patients with good collaterals experienced less oxidative stress (sORP) and had higher antioxidant reserves (cORP) than patients with reduced collaterals.
Collapse
Affiliation(s)
- Benjamin Atchie
- Department of Neuroradiology, Swedish Medical Center, Injury Outcomes Network (ION) Research, Englewood, CO 80113, USA
- Department of Neurointerventional Surgery, Radiology Imaging Associates, Injury Outcomes Network (ION) Research, Englewood, CO 80113, USA
| | - Stephanie Jarvis
- Department of Epidemiology, Injury Outcomes Network (ION) Research, Englewood, CO 80113, USA
| | - Richard Bellon
- Department of Neuroradiology, Swedish Medical Center, Injury Outcomes Network (ION) Research, Englewood, CO 80113, USA
- Department of Neurointerventional Surgery, Radiology Imaging Associates, Injury Outcomes Network (ION) Research, Englewood, CO 80113, USA
| | - Trevor Barton
- Department of Neurology, Swedish Medical Center, Injury Outcomes Network (ION) Research, Englewood, CO 80113, USA
| | - Lauren Disalvo
- Department of Neurology, Swedish Medical Center, Injury Outcomes Network (ION) Research, Englewood, CO 80113, USA
| | - Kristin Salottolo
- Department of Epidemiology, Injury Outcomes Network (ION) Research, Englewood, CO 80113, USA
| | - Raphael Bar-Or
- Department of Basic Science, Injury Outcomes Network (ION) Research, Englewood, CO 80113, USA
| | - David Bar-Or
- Department of Directors, Injury Outcomes Network (ION) Research, Englewood, CO 80113, USA
| |
Collapse
|
16
|
Daei S, Abbasalipourkabir R, Khajvand-Abedini M, Ziamajidi N. The Alleviative Efficacy of Vitamins A, C, and E Against Zinc Oxide Nanoparticles-Induced Hepatic Damage by Reducing Apoptosis in Rats. Biol Trace Elem Res 2023; 201:1252-1260. [PMID: 35364806 DOI: 10.1007/s12011-022-03218-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/23/2022] [Indexed: 02/07/2023]
Abstract
Nanoparticles are vastly exploited in today's technology. However, it is realized that exposure to high concentrations of nanoparticles (NPs) may have adverse effects on human health. According to previous reports, zinc oxide (ZnO) NPs cause toxic effects in tissues via inducing apoptosis. The current work was designed to evaluate possible protective activities of vitamins (Vits) A, C, and E against ZnO NPs-induced apoptosis in the liver of rats. To this aim, fifty-four adult male Wistar rats were randomly distributed into nine groups (n = 6 rats for each group), namely, Control1 (water), Control2 (olive oil), Vit A (1000 IU/kg), Vit C (200 mg/kg), Vit E (100 IU/kg), ZnO (200 mg/kg), ZnO + VitA, ZnO + VitC, and ZnO + VitE. To investigate apoptosis, the mRNA and protein expression of Bcl-2-associated X (Bax) and B-cell lymphoma protein 2 (Bcl-2) were examined by qRT-PCR and western blot techniques. The mRNA and protein expression of TNF-α as well as the activity of caspase 3,7 were also measured. The results revealed that ZnO NPs considerably enhance the ratio of Bax to Bcl-2 mRNA and protein expression as well as the activity of caspase 3,7 compared to the control group. Furthermore, the findings implied that the elevated level of TNF-α may link with ZnO NPs-mediated apoptosis in the liver of rats. More importantly, Vits A, C, and E exhibited ameliorative properties against apoptosis-inducing effects of ZnO NPs. Thus, administration of Vits A, C, and E may be effective in preventing liver damage and apoptosis caused by ZnO NPs.
Collapse
Affiliation(s)
- Sajedeh Daei
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Roghayeh Abbasalipourkabir
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Maryam Khajvand-Abedini
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Nasrin Ziamajidi
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
- Molecular Medicine Research Center, Hamadan University of Medical Science, Hamadan, Iran.
| |
Collapse
|
17
|
Ziamajidi N, Daei S, Khajvand-Abedini M, Abbasalipourkabir R, Nourian A. Vitamins A, C, and E Exert Anti-apoptotic Function in the Testis of Rats After Exposure to Zinc Oxide Nanoparticles. Chonnam Med J 2023; 59:48-53. [PMID: 36794239 PMCID: PMC9900230 DOI: 10.4068/cmj.2023.59.1.48] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/18/2022] [Accepted: 11/23/2022] [Indexed: 02/03/2023] Open
Abstract
Some reports emphasize that zinc oxide nanoparticles (ZnO NPs) are detrimental to the reproductive organs of animals. As such, this research aimed at exploring the apoptotic potential of ZnO NPs on testis along with the beneficial role of Vitamins (V) A, C, and E against ZnO NP-induced damage. To this aim, a population of 54 healthy, male Wistar rats were used in this work and then assigned into nine groups of 6 rats as G1: Control 1 (Water); G2: Control 2 (Olive oil); G3: VA (1000 IU/kg), G4: VC (200 mg/kg), G5: VE (100 IU/kg), G6: ZnO NPs exposed animals (200 mg/kg); and G7, 8 and 9: ZnO NPs-exposed animals that were pre-treated with either VA, C, or E. Apoptosis rates were estimated by measuring the level of apoptotic regulatory markers including Bcl-2-associated X (Bax) and B-cell lymphoma protein 2 (Bcl-2) using western blotting and qRT-PCR assays. The data indicated that ZnO NPs exposure elevates the level of Bax protein and gene expression, whereas the protein and gene expression of Bcl-2 was reduced. Further, the activation of caspase-3,7 occurred after exposure to ZnO NPs, while the above alterations were significantly alleviated in the rats that were co-treated with VA, C, or E and ZnO NPs relative to the rats in the ZnO NPs group. In summary, VA, C, and E exerted anti-apoptotic functions in the testis of rats following administration of ZnO NPs.
Collapse
Affiliation(s)
- Nasrin Ziamajidi
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamedan, Iran.,Molecular Medicine Research Center, Hamadan University of Medical Science, Hamedan, Iran
| | - Sajedeh Daei
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamedan, Iran
| | - Maryam Khajvand-Abedini
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamedan, Iran
| | - Roghayeh Abbasalipourkabir
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamedan, Iran
| | - Alireza Nourian
- Department of Pathobiology, Faculty of Veterinary Science, Bu-Ali Sina University, Hamedan, Iran
| |
Collapse
|
18
|
Aragón-González A, Shaw PJ, Ferraiuolo L. Blood-Brain Barrier Disruption and Its Involvement in Neurodevelopmental and Neurodegenerative Disorders. Int J Mol Sci 2022; 23:ijms232315271. [PMID: 36499600 PMCID: PMC9737531 DOI: 10.3390/ijms232315271] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
The blood-brain barrier (BBB) is a highly specialized and dynamic compartment which regulates the uptake of molecules and solutes from the blood. The relevance of the maintenance of a healthy BBB underpinning disease prevention as well as the main pathomechanisms affecting BBB function will be detailed in this review. Barrier disruption is a common aspect in both neurodegenerative diseases, such as amyotrophic lateral sclerosis, and neurodevelopmental diseases, including autism spectrum disorders. Throughout this review, conditions altering the BBB during the earliest and latest stages of life will be discussed, revealing common factors involved. Due to the barrier's role in protecting the brain from exogenous components and xenobiotics, drug delivery across the BBB is challenging. Potential therapies based on the BBB properties as molecular Trojan horses, among others, will be reviewed, as well as innovative treatments such as stem cell therapies. Additionally, due to the microbiome influence on the normal function of the brain, microflora modulation strategies will be discussed. Finally, future research directions are highlighted to address the current gaps in the literature, emphasizing the idea that common therapies for both neurodevelopmental and neurodegenerative pathologies exist.
Collapse
Affiliation(s)
- Ana Aragón-González
- Sheffield Institute for Translational Neuroscience, University of Sheffield, SITraN, 385a Glossop Road, Sheffield S10 2HQ, UK
- Facultad de Medicina, Universidad de Málaga, 29010 Málaga, Spain
| | - Pamela J. Shaw
- Sheffield Institute for Translational Neuroscience, University of Sheffield, SITraN, 385a Glossop Road, Sheffield S10 2HQ, UK
| | - Laura Ferraiuolo
- Sheffield Institute for Translational Neuroscience, University of Sheffield, SITraN, 385a Glossop Road, Sheffield S10 2HQ, UK
- Correspondence: ; Tel.: +44-(0)114-222-2257; Fax: +44-(0)114-222-2290
| |
Collapse
|
19
|
Morris-Blanco KC, Chokkalla AK, Kim T, Bhatula S, Bertogliat MJ, Gaillard AB, Vemuganti R. High-Dose Vitamin C Prevents Secondary Brain Damage After Stroke via Epigenetic Reprogramming of Neuroprotective Genes. Transl Stroke Res 2022; 13:1017-1036. [PMID: 35306630 PMCID: PMC9485293 DOI: 10.1007/s12975-022-01007-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/27/2022] [Accepted: 03/14/2022] [Indexed: 12/27/2022]
Abstract
Vitamin C has recently been identified as an epigenetic regulator by activating ten-eleven translocases (TETs), enzymes involved in generating DNA hydroxymethylcytosine (5hmC). Currently, we investigated whether high-dose vitamin C promotes neuroprotection through epigenetic modulation of 5hmC, if there are sex-specific differences in outcome, and the therapeutic potential of vitamin C in stroke-related comorbidities in adult mice. Post-stroke treatment with ascorbate (reduced form), but not dehydroascorbate (oxidized form), increased TET3 activity and 5hmC levels and reduced infarct following focal ischemia. Hydroxymethylation DNA immunoprecipitation sequencing showed that ascorbate increased 5hmC across the genome and specifically in promoters of several stroke pathophysiology-related genes, particularly anti-inflammatory genes. Ascorbate also decreased markers of oxidative stress, mitochondrial fragmentation, and apoptosis in cortical peri-infarct neurons and promoted motor and cognitive functional recovery in both sexes via TET3. Furthermore, post-stroke ascorbate treatment reduced infarct volume and improved motor function recovery in aged, hypertensive and diabetic male and female mice. Delayed ascorbate treatment at 6 h of reperfusion was still effective at reducing infarct volume and motor impairments in adult mice. Collectively, this study shows that post-stroke treatment with high-dose ascorbate protects the brain through epigenetic reprogramming and may function as a robust therapeutic against stroke injury.
Collapse
Affiliation(s)
- Kahlilia C Morris-Blanco
- Department of Neurological Surgery, University of Wisconsin-Madison, Mail code CSC-8660, 600 Highland Ave, Madison, WI, 53792, USA
- William S. Middleton Veterans Administration Hospital, Madison, WI, USA
| | - Anil K Chokkalla
- Department of Neurological Surgery, University of Wisconsin-Madison, Mail code CSC-8660, 600 Highland Ave, Madison, WI, 53792, USA
- Cellular and Molecular Pathology Program, University of Wisconsin-Madison, Madison, WI, USA
| | - TaeHee Kim
- Department of Neurological Surgery, University of Wisconsin-Madison, Mail code CSC-8660, 600 Highland Ave, Madison, WI, 53792, USA
| | - Saivenkateshkomal Bhatula
- Department of Neurological Surgery, University of Wisconsin-Madison, Mail code CSC-8660, 600 Highland Ave, Madison, WI, 53792, USA
- William S. Middleton Veterans Administration Hospital, Madison, WI, USA
| | - Mario J Bertogliat
- Department of Neurological Surgery, University of Wisconsin-Madison, Mail code CSC-8660, 600 Highland Ave, Madison, WI, 53792, USA
| | - Alexis B Gaillard
- Department of Neurological Surgery, University of Wisconsin-Madison, Mail code CSC-8660, 600 Highland Ave, Madison, WI, 53792, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin-Madison, Mail code CSC-8660, 600 Highland Ave, Madison, WI, 53792, USA.
- William S. Middleton Veterans Administration Hospital, Madison, WI, USA.
- Cellular and Molecular Pathology Program, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
20
|
Fyn Signaling in Ischemia-Reperfusion Injury: Potential and Therapeutic Implications. Mediators Inflamm 2022; 2022:9112127. [PMID: 36157893 PMCID: PMC9499810 DOI: 10.1155/2022/9112127] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/20/2022] [Accepted: 08/27/2022] [Indexed: 11/18/2022] Open
Abstract
Ischemic stroke caused by arterial occlusion is the most common type of stroke and is one of the leading causes of disability and death, with the incidence increasing each year. Fyn is a nonreceptor tyrosine kinase belonging to the Src family of kinases (SFKs), which is related to many normal and pathological processes of the nervous system, including neurodevelopment and disease progression. In recent years, more and more evidence suggests that Fyn may be closely related to cerebral ischemia-reperfusion, including energy metabolism disorders, excitatory neurotoxicity, intracellular calcium homeostasis, free radical production, and the activation of apoptotic genes. This paper reviews the role of Fyn in the pathological process of cerebral ischemia-reperfusion, including neuroexcitotoxicity and neuroinflammation, to explore how Fyn affects specific signal cascades and leads to cerebral ischemia-reperfusion injury. In addition, Fyn also promotes the production of superoxide and endogenous NO, so as to quickly react to produce peroxynitrite, which may also mediate cerebral ischemia-reperfusion injury, which is discussed in this paper. Finally, we revealed the treatment methods related to Fyn inhibitors and discussed its potential as a clinical treatment for ischemic stroke.
Collapse
|
21
|
Gallego I, Villate-Beitia I, Saenz-Del-Burgo L, Puras G, Pedraz JL. Therapeutic Opportunities and Delivery Strategies for Brain Revascularization in Stroke, Neurodegeneration, and Aging. Pharmacol Rev 2022; 74:439-461. [PMID: 35302047 DOI: 10.1124/pharmrev.121.000418] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 01/18/2022] [Accepted: 01/22/2022] [Indexed: 12/25/2022] Open
Abstract
Central nervous system (CNS) diseases, especially acute ischemic events and neurodegenerative disorders, constitute a public health problem with no effective treatments to allow a persistent solution. Failed therapies targeting neuronal recovery have revealed the multifactorial and intricate pathophysiology underlying such CNS disorders as ischemic stroke, Alzheimeŕs disease, amyotrophic lateral sclerosis, vascular Parkisonism, vascular dementia, and aging, in which cerebral microvasculature impairment seems to play a key role. In fact, a reduction in vessel density and cerebral blood flow occurs in these scenarios, contributing to neuronal dysfunction and leading to loss of cognitive function. In this review, we provide an overview of healthy brain microvasculature structure and function in health and the effect of the aforementioned cerebral CNS diseases. We discuss the emerging new therapeutic opportunities, and their delivery approaches, aimed at recovering brain vascularization in this context. SIGNIFICANCE STATEMENT: The lack of effective treatments, mainly focused on neuron recovery, has prompted the search of other therapies to treat cerebral central nervous system diseases. The disruption and degeneration of cerebral microvasculature has been evidenced in neurodegenerative diseases, stroke, and aging, constituting a potential target for restoring vascularization, neuronal functioning, and cognitive capacities by the development of therapeutic pro-angiogenic strategies.
Collapse
Affiliation(s)
- Idoia Gallego
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P); Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine, Institute of Health Carlos III, Madrid, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.); and Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.)
| | - Ilia Villate-Beitia
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P); Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine, Institute of Health Carlos III, Madrid, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.); and Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.)
| | - Laura Saenz-Del-Burgo
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P); Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine, Institute of Health Carlos III, Madrid, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.); and Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.)
| | - Gustavo Puras
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P); Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine, Institute of Health Carlos III, Madrid, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.); and Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.)
| | - José Luis Pedraz
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P); Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine, Institute of Health Carlos III, Madrid, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.); and Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.)
| |
Collapse
|
22
|
Ashok A, Andrabi SS, Mansoor S, Kuang Y, Kwon BK, Labhasetwar V. Antioxidant Therapy in Oxidative Stress-Induced Neurodegenerative Diseases: Role of Nanoparticle-Based Drug Delivery Systems in Clinical Translation. Antioxidants (Basel) 2022; 11:antiox11020408. [PMID: 35204290 PMCID: PMC8869281 DOI: 10.3390/antiox11020408] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/02/2022] [Accepted: 02/05/2022] [Indexed: 02/04/2023] Open
Abstract
Free radicals are formed as a part of normal metabolic activities but are neutralized by the endogenous antioxidants present in cells/tissue, thus maintaining the redox balance. This redox balance is disrupted in certain neuropathophysiological conditions, causing oxidative stress, which is implicated in several progressive neurodegenerative diseases. Following neuronal injury, secondary injury progression is also caused by excessive production of free radicals. Highly reactive free radicals, mainly the reactive oxygen species (ROS) and reactive nitrogen species (RNS), damage the cell membrane, proteins, and DNA, which triggers a self-propagating inflammatory cascade of degenerative events. Dysfunctional mitochondria under oxidative stress conditions are considered a key mediator in progressive neurodegeneration. Exogenous delivery of antioxidants holds promise to alleviate oxidative stress to regain the redox balance. In this regard, natural and synthetic antioxidants have been evaluated. Despite promising results in preclinical studies, clinical translation of antioxidants as a therapy to treat neurodegenerative diseases remains elusive. The issues could be their low bioavailability, instability, limited transport to the target tissue, and/or poor antioxidant capacity, requiring repeated and high dosing, which cannot be administered to humans because of dose-limiting toxicity. Our laboratory is investigating nanoparticle-mediated delivery of antioxidant enzymes to address some of the above issues. Apart from being endogenous, the main advantage of antioxidant enzymes is their catalytic mechanism of action; hence, they are significantly more effective at lower doses in detoxifying the deleterious effects of free radicals than nonenzymatic antioxidants. This review provides a comprehensive analysis of the potential of antioxidant therapy, challenges in their clinical translation, and the role nanoparticles/drug delivery systems could play in addressing these challenges.
Collapse
Affiliation(s)
- Anushruti Ashok
- Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (A.A.); (S.S.A.); (S.M.); (Y.K.)
| | - Syed Suhail Andrabi
- Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (A.A.); (S.S.A.); (S.M.); (Y.K.)
| | - Saffar Mansoor
- Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (A.A.); (S.S.A.); (S.M.); (Y.K.)
| | - Youzhi Kuang
- Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (A.A.); (S.S.A.); (S.M.); (Y.K.)
| | - Brian K. Kwon
- Department of Orthopaedics, Faculty of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada;
| | - Vinod Labhasetwar
- Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (A.A.); (S.S.A.); (S.M.); (Y.K.)
- Correspondence:
| |
Collapse
|
23
|
Natural compounds modulate the autophagy with potential implication of stroke. Acta Pharm Sin B 2021; 11:1708-1720. [PMID: 34386317 PMCID: PMC8343111 DOI: 10.1016/j.apsb.2020.10.018] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/12/2020] [Accepted: 08/21/2020] [Indexed: 12/12/2022] Open
Abstract
Stroke is considered a leading cause of mortality and neurological disability, which puts a huge burden on individuals and the community. To date, effective therapy for stroke has been limited by its complex pathological mechanisms. Autophagy refers to an intracellular degrading process with the involvement of lysosomes. Autophagy plays a critical role in maintaining the homeostasis and survival of cells by eliminating damaged or non-essential cellular constituents. Increasing evidence support that autophagy protects neuronal cells from ischemic injury. However, under certain circumstances, autophagy activation induces cell death and aggravates ischemic brain injury. Diverse naturally derived compounds have been found to modulate autophagy and exert neuroprotection against stroke. In the present work, we have reviewed recent advances in naturally derived compounds that regulate autophagy and discussed their potential application in stroke treatment.
Collapse
Key Words
- AD, Alzheimer's disease
- ALS, amyotrophic lateral sclerosis
- AMPK, 5′-adenosine monophosphate-activated protein kinase
- ATF6, activating transcription factor 6
- ATG, autophagy related genes
- Autophagy
- BCL-2, B-cell lymphoma 2
- BNIP3L, BCL2/adenovirus
- COPII, coat protein complex II
- Cerebral ischemia
- ER, endoplasmic reticulum
- FOXO, forkhead box O
- FUNDC1, FUN14 domain containing 1
- GPCR, G-protein coupled receptor
- HD, Huntington's disease
- IPC, ischemic preconditioning
- IRE1, inositol-requiring enzyme 1
- JNK, c-Jun N-terminal kinase
- LAMP, lysosomal-associated membrane protein
- LC3, light chain 3
- LKB1, liver kinase B1
- Lysosomal activation
- Mitochondria
- Mitophagy
- Natural compounds
- Neurological disorders
- Neuroprotection
- OGD/R, oxygen and glucose deprivation-reperfusion
- PD, Parkinson's disease
- PERK, protein kinase R (PKR)-like endoplasmic reticulum kinase
- PI3K, phosphatidylinositol 3-kinase
- ROS, reactive oxygen species
- SQSTM1, sequestosome 1
- TFEB, transcription factor EB
- TIGAR, TP53-induced glycolysis and apoptosis regulator
- ULK, Unc-51- like kinase
- Uro-A, urolithin A
- eIF2a, eukaryotic translation-initiation factor 2
- mTOR, mechanistic target of rapamycin
- ΔΨm, mitochondrial membrane potential
Collapse
|
24
|
Baek SJ, Hammock BD, Hwang IK, Li Q, Moustaid-Moussa N, Park Y, Safe S, Suh N, Yi SS, Zeldin DC, Zhong Q, Bradbury JA, Edin ML, Graves JP, Jung HY, Jung YH, Kim MB, Kim W, Lee J, Li H, Moon JS, Yoo ID, Yue Y, Lee JY, Han HJ. Natural Products in the Prevention of Metabolic Diseases: Lessons Learned from the 20th KAST Frontier Scientists Workshop. Nutrients 2021; 13:1881. [PMID: 34072678 PMCID: PMC8227583 DOI: 10.3390/nu13061881] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/20/2021] [Accepted: 05/20/2021] [Indexed: 12/29/2022] Open
Abstract
The incidence of metabolic and chronic diseases including cancer, obesity, inflammation-related diseases sharply increased in the 21st century. Major underlying causes for these diseases are inflammation and oxidative stress. Accordingly, natural products and their bioactive components are obvious therapeutic agents for these diseases, given their antioxidant and anti-inflammatory properties. Research in this area has been significantly expanded to include chemical identification of these compounds using advanced analytical techniques, determining their mechanism of action, food fortification and supplement development, and enhancing their bioavailability and bioactivity using nanotechnology. These timely topics were discussed at the 20th Frontier Scientists Workshop sponsored by the Korean Academy of Science and Technology, held at the University of Hawaii at Manoa on 23 November 2019. Scientists from South Korea and the U.S. shared their recent research under the overarching theme of Bioactive Compounds, Nanoparticles, and Disease Prevention. This review summarizes presentations at the workshop to provide current knowledge of the role of natural products in the prevention and treatment of metabolic diseases.
Collapse
Affiliation(s)
- Seung J. Baek
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (S.J.B.); (I.-K.H.); (H.-Y.J.); (Y.-H.J.); (W.K.); (J.L.)
| | - Bruce D. Hammock
- Department of Entomology, University of California, Davis, CA 95616, USA;
| | - In-Koo Hwang
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (S.J.B.); (I.-K.H.); (H.-Y.J.); (Y.-H.J.); (W.K.); (J.L.)
| | - Qingxiao Li
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI 96822, USA;
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences & Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA;
| | - Yeonhwa Park
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA; (Y.P.); (Y.Y.)
| | - Stephen Safe
- Department of Biochemistry & Biophysics, Texas A & M University, College Station, TX 77843, USA;
| | - Nanjoo Suh
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA;
| | - Sun-Shin Yi
- Department of Medical Sciences, Soonchunhyang University, Asan 31538, Korea; (S.-S.Y.); (J.-S.M.); (I.-D.Y.)
| | - Darryl C. Zeldin
- National Institutes of Environmental Health, National Institutes of Health, Research Triangle Park, NC 27709, USA; (D.C.Z.); (J.A.B.); (M.L.E.); (J.P.G.); (H.L.)
| | - Qixin Zhong
- Department of Food Sciences, University of Tennessee, Knoxville, TN 37996, USA;
| | - Jennifer Alyce Bradbury
- National Institutes of Environmental Health, National Institutes of Health, Research Triangle Park, NC 27709, USA; (D.C.Z.); (J.A.B.); (M.L.E.); (J.P.G.); (H.L.)
| | - Matthew L. Edin
- National Institutes of Environmental Health, National Institutes of Health, Research Triangle Park, NC 27709, USA; (D.C.Z.); (J.A.B.); (M.L.E.); (J.P.G.); (H.L.)
| | - Joan P. Graves
- National Institutes of Environmental Health, National Institutes of Health, Research Triangle Park, NC 27709, USA; (D.C.Z.); (J.A.B.); (M.L.E.); (J.P.G.); (H.L.)
| | - Hyo-Young Jung
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (S.J.B.); (I.-K.H.); (H.-Y.J.); (Y.-H.J.); (W.K.); (J.L.)
| | - Young-Hyun Jung
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (S.J.B.); (I.-K.H.); (H.-Y.J.); (Y.-H.J.); (W.K.); (J.L.)
| | - Mi-Bo Kim
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA;
| | - Woosuk Kim
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (S.J.B.); (I.-K.H.); (H.-Y.J.); (Y.-H.J.); (W.K.); (J.L.)
| | - Jaehak Lee
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (S.J.B.); (I.-K.H.); (H.-Y.J.); (Y.-H.J.); (W.K.); (J.L.)
| | - Hong Li
- National Institutes of Environmental Health, National Institutes of Health, Research Triangle Park, NC 27709, USA; (D.C.Z.); (J.A.B.); (M.L.E.); (J.P.G.); (H.L.)
| | - Jong-Seok Moon
- Department of Medical Sciences, Soonchunhyang University, Asan 31538, Korea; (S.-S.Y.); (J.-S.M.); (I.-D.Y.)
| | - Ik-Dong Yoo
- Department of Medical Sciences, Soonchunhyang University, Asan 31538, Korea; (S.-S.Y.); (J.-S.M.); (I.-D.Y.)
| | - Yiren Yue
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA; (Y.P.); (Y.Y.)
| | - Ji-Young Lee
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA;
| | - Ho-Jae Han
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (S.J.B.); (I.-K.H.); (H.-Y.J.); (Y.-H.J.); (W.K.); (J.L.)
| |
Collapse
|
25
|
Ji W, Liu K, Zhao G, Wu F, Jiang Y, Hou L, Zhang M, Mao L. Electrochemical Sensing of Ascorbate as an Index of Neuroprotection from Seizure Activity by Physical Exercise in Freely Moving Rats. ACS Sens 2021; 6:546-552. [PMID: 33346640 DOI: 10.1021/acssensors.0c02326] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Physical exercise (PE) has been drawing increasing attention to prevent and alleviate neural damage of brain diseases; however, in vivo sensing of the neuroprotection ability of PE remains a challenge. Here, we find that ascorbate can be used as a small molecular index for neuroprotective function of PE and the neuroprotection ability of PE can thus be in vivo monitored with an online electrochemical system (OECS) in freely moving animals. With the OECS as the sensing system, we find that the concentration of ascorbate in the microdialysate from the striatum increases greatly in kainic acid (KA)-induced seizure rats and reaches twice the basal level (i.e., 214.4 ± 32.7%, p < 0.001, n = 4) at a time point 90 min after KA microinjection. Such an increase of ascorbate is obviously attenuated (i.e., 153.6 ± 23.9% of the basal level, p < 0.05, n = 3) after PE, showing the neuroprotective activity of PE. This finding is believed to be significant in providing chemical insight into the neuroprotection ability of PE.
Collapse
Affiliation(s)
- Wenliang Ji
- Department of Chemistry, Renmin University of China, Beijing 100872, China
| | - Kun Liu
- Capital University of Physical Education and Sports, Beijing 100191, China
| | - Gang Zhao
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, the Chinese Academy of Sciences (CAS), Beijing 100190, China
| | - Fei Wu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, the Chinese Academy of Sciences (CAS), Beijing 100190, China
| | | | | | - Meining Zhang
- Department of Chemistry, Renmin University of China, Beijing 100872, China
| | - Lanqun Mao
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, the Chinese Academy of Sciences (CAS), Beijing 100190, China
| |
Collapse
|
26
|
Kılıç E, Çağlayan B, Caglar Beker M. Physiological and pharmacological roles of melatonin in the pathophysiological components of cellular injury after ischemic stroke. Turk J Med Sci 2020; 50:1655-1664. [PMID: 32962330 PMCID: PMC7672349 DOI: 10.3906/sag-2008-32] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 09/21/2020] [Indexed: 12/22/2022] Open
Abstract
Apart from its metabolic or physiological functions, melatonin has a potent cytoprotective activity in the physiological and pathological conditions. It is synthetized by the pineal gland and released into the blood circulation but particularly cerebrospinal fluid in a circadian manner. It can also easily diffuse through cellular membranes due its small size and lipophilic structure. Its cytoprotective activity has been linked to its potent free radical scavenger activity with the desirable characteristics of a clinically- reliable antioxidant. Melatonin detoxifies oxygen and nitrogen-based free radicals and oxidizing agents, including the highly toxic hydroxyl-and peroxynitrite radicals, initiating cellular damage. However, the cytoprotective activity of melatonin is complex and cannot be solely limited to its free radical scavenger activity. It regulates cellular signaling pathways through receptor– dependent and independent mechanisms. Most of these downstream molecules, such as PI3K/AKT pathway components, also contribute to the cytoprotective effects of melatonin. In this term, melatonin is a promising molecule for the treatment of neurodegenerative disorders, such as ischemic stroke, which melatonin reduces ischemic brain injury in animal models of ischemic stroke. It regulates also circadian rhythm proteins after ischemic stroke, playing roles in cellular survival. In this context, present article summarizes the possible role of melatonin in the pathophysiological events after ischemic stroke.
Collapse
Affiliation(s)
- Ertuğrul Kılıç
- Department of Physiology, School of Medicine, İstanbul Medipol University, İstanbul, Turkey,Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, İstanbul, Turkey
| | - Berrak Çağlayan
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, İstanbul, Turkey,Department of Medical Biology, International School of Medicine, İstanbul Medipol University, İstanbul, Turkey
| | - Mustafa Caglar Beker
- Department of Physiology, School of Medicine, İstanbul Medipol University, İstanbul, Turkey,Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, İstanbul, Turkey
| |
Collapse
|